WO2024232668A1 - Anti-ptgfrn monoclonal antibody and use thereof - Google Patents
Anti-ptgfrn monoclonal antibody and use thereof Download PDFInfo
- Publication number
- WO2024232668A1 WO2024232668A1 PCT/KR2024/006210 KR2024006210W WO2024232668A1 WO 2024232668 A1 WO2024232668 A1 WO 2024232668A1 KR 2024006210 W KR2024006210 W KR 2024006210W WO 2024232668 A1 WO2024232668 A1 WO 2024232668A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cancer
- ptgfrn
- cells
- cancer cells
- blood
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
Definitions
- the present invention relates to an anti-PTGFRN monoclonal antibody for determining the level of blood cancer cells, treating or preventing cancer, predicting the prognosis of cancer metastasis, and screening for anticancer drugs.
- Hepatocellular carcinoma accounts for more than 90% of liver cancers and is the third leading cause of cancer-related mortality.
- the main treatments for HCC include surgical resection, local resection for early-stage HCC, or liver transplantation.
- most patients with early-stage HCC are already diagnosed with advanced disease, and survival is limited to palliative treatment such as transarterial chemoembolization, systemic treatment with tyrosine kinase inhibitors, and selective internal radiotherapy.
- HCC has an exceptionally high recurrence rate, and the treatment effect is not satisfactory.
- the diagnosis and monitoring of HCC mainly depend on serum biomarker detection, pathological examination, and image analysis.
- AFP a common serum marker
- DCP des-gamma-carboxy-prothrombin
- Circulating tumor cells are cancer cells found in the peripheral blood of cancer patients, and they are cancer cells that move through the blood. Metastasis can be the direct cause of most cancer-related deaths, and circulating tumor cells are the seeds of this metastasis phenomenon and are cells that spread cancer. They form homo- and hetero-clusters to evade immune cells and internal and external inhibitory signals to metastasize, and their expression level is related to the survival rate of cancer. Since an increase in the number of circulating tumor cells in the peripheral blood indicates that metastasis is progressing in cancer patients, a method has been developed to use circulating tumor cells as a cancer metastasis diagnostic marker. In addition, circulating tumor cells are associated with a poor prognosis for cancer patients.
- blood cancer cells have been separated and studied using a cell search system using EpCAM, a microfluidics system based on cell size and physical characteristics, and a flow cytometer.
- EpCAM EpCAM
- a microfluidics system based on cell size and physical characteristics
- a flow cytometer a flow cytometer
- EpCAM Epithelial Cell Adhesion Molecule
- ASGPR asialoglycoprotein receptor
- the proportion of 85 hepatocellular cancer patients with more than 1 circulating cancer cell per 5 ml of blood was approximately 81%, or 69 cases.
- interest in EpCAM-positive blood cancer cells exhibiting stem cell characteristics has emerged in the field of hepatocellular carcinoma, based on research results showing that EpCAM is a marker of hepatocellular carcinoma stem cells that induce metastasis of hepatocellular carcinoma or induce recurrence of treatment.
- the rate of detecting more than 1 circulating cancer cell per 7.5 ml of blood in 50 hepatocellular carcinoma patients using an EpCAM-based circulating cancer cell detection method was only 28%, or 14 cases.
- the EpCAM-based circulating cancer cell detection method did not have a detection rate exceeding 50%. This is because EpCAM expression decreases due to epithelial-mesenchymal transition that occurs during the metastatic process of cancer, and is a fundamental limitation of the EpCAM-based circulating cancer cell detection method.
- Epithelial-mesenchymal transition refers to the entire process by which adherent epithelial cancer cells transition into mesenchymal cancer cells with motility and invasiveness.
- epithelial cell markers may be down-regulated. Therefore, if circulating cancer cells are detected solely based on epithelial cell markers, most circulating cancer cells may not be detected.
- EpCAM is used as a marker to detect circulating cancer cells, only 50-70% of all circulating cancer cells can be detected in lung cancer, breast cancer, cervical cancer, and nasopharyngeal cancer, and in the case of liver cancer, only 25% of the circulating cancer cells are detected.
- the results of analyzing the circulating cancer cells with mixed and mesenchymal markers in hepatocellular carcinoma showed that patients with more circulating cancer cells had stronger invasive ability, and these patients showed higher clinical stages.
- the results of analyzing the circulating cancer cells and circulating cancer cell clusters collected from the peripheral blood of 214 hepatocellular carcinoma patients showed that the circulating cancer cell clusters were closely related to the overall survival (OS) or progression-free survival (PFS) and showed a poor prognosis.
- OS overall survival
- PFS progression-free survival
- EMT electrospray
- mesenchymal cells are known to have a hybrid epithelial/mesenchymal state or a partial phenotype that has both epithelial and mesenchymal characteristics, and they are known to have a tendency to form clusters and have immune-evading abilities to evade the host's immune system. Therefore, in order to detect all circulating cancer cells in a patient, it is necessary to discover markers that can detect not only epithelial circulating cancer cells, but also circulating cancer cells showing an EMT phenotype, circulating cancer cells showing partial EMT, and entirely new types of circulating cancer cells.
- PTGFRN Prostaglandin F2 Receptor Negative Regulator, EWI-F, CD9P-1, CD315) is a type I transmembrane protein of the immunoglobulin superfamily that negatively affects the Prostaglandin F2 Receptor.
- PTGFRN is a component of the Tetraspanin Enriched Microdomain (TEM), which consists of tetraspanins, integrins, signaling enzymes, and proteoglycans, and interacts with tetraspanin proteins such as CD9 and CD81.
- TEM Tetraspanin Enriched Microdomain
- PTGFRN is expressed in large quantities as a structural protein in exosomes, and its separation function through exosomes is being studied.
- PTGFRN is involved in the regeneration of muscle cells, the growth of glioma, and metastasis.
- PTGFRN is a molecule that is up-regulated in many cancer cells, including gliomas, and is known to be overexpressed in glioblastoma multiforme (GBM), a malignant brain tumor, where it promotes cell growth and radioresistance through PI3K-AKT signaling.
- GBM glioblastoma multiforme
- the present inventors have prepared 70 kinds of monoclonal antibodies that bind to the surface of human embryonic stem cells, and one of them, 63-D7, binds well to the surface of various human cancer cells including human embryonic stem cells, embryonic cancer cells, and liver cancer cells, and does not bind to normal cells such as human peripheral blood mononuclear cells (PBMC), fetal lung fibroblasts (MRC5), and normal hepatocytes.
- PBMC peripheral blood mononuclear cells
- MRC5 fetal lung fibroblasts
- PTGFRN recognized by 63-D7 was highly co-expressed with B7-H3 (57.6%), MVP (31.9%), TGF ⁇ R1 (51.8%), PD-L1 (37.8%), PD-L2 (17.5%), and CD47 (38.1%), which are associated with drug resistance and immune evasion in circulating cancer cells, showing that the expression of PTGFRN is closely related to metastasis or recurrence, and suggesting that these molecules can induce immune evasion of cancer cells.
- 63-D7 is an antibody that induces internalization in liver cancer cells and pancreatic cancer cells, and in fact, through these properties, we suggest that 63-D7 can be developed as a cancer treatment agent in the future by inducing liver cancer cell death using an Antibody-Drug Conjugate (ADC).
- ADC Antibody-Drug Conjugate
- the present invention aims to provide a use of PTGFRN as a surface molecular marker of cancer cells.
- the present invention aims to provide a monoclonal antibody 63-D7 that specifically binds to PTGFRN.
- the present invention aims to provide the use of monoclonal antibody 63-D7 as an agonistic antibody against PTGFRN that increases mobility and invasiveness in cancer cells.
- the present invention aims to provide a use of PTGFRN as a surface molecular marker of blood cancer cells.
- Another object of the present invention is to provide a use of the monoclonal antibody 63-D7 as a substance that specifically binds to PTGFRN to isolate, purify or detect PTGFRN protein.
- An anti-PTGFRN monoclonal antibody comprising a heavy chain variable region comprising HCDR1 consisting of the amino acid sequence of SEQ ID NO: 3, HCDR2 consisting of the amino acid sequence of SEQ ID NO: 4, and HCDR3 consisting of the amino acid sequence of SEQ ID NO: 5; and a light chain variable region comprising LCDR1 consisting of the amino acid sequence of SEQ ID NO: 6, LCDR2 consisting of the amino acid sequence of SEQ ID NO: 7, and LCDR3 consisting of the amino acid sequence of SEQ ID NO: 8.
- the heavy chain variable region is an anti-PTGFRN monoclonal antibody comprising the amino acid sequence of sequence number 1.
- An antibody-drug conjugate comprising a drug conjugated to any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
- a pharmaceutical composition for treating or preventing cancer comprising any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
- the cancer is selected from the group consisting of brain and spinal tumors, head and neck cancer, lung cancer, breast cancer, thymoma, mesothelioma, esophageal cancer, stomach cancer, colon cancer, liver cancer, thyroid cancer, pancreatic cancer, biliary tract cancer, kidney cancer, bladder cancer, prostate cancer, testicular cancer, germ cell tumor, ovarian cancer, cervical cancer, endometrial cancer, lymphoma, acute leukemia, chronic leukemia, multiple myeloma, sarcoma, malignant melanoma, neuroblastoma, glioblastoma, and skin cancer, a pharmaceutical composition for treating or preventing cancer.
- composition for detecting PTGFRN comprising any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
- a method for providing information for diagnosing cancer comprising the step of treating a sample isolated from an individual with any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
- a method for providing information necessary for determining the level of cancer cells in blood comprising the step of treating a sample isolated from an individual with any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
- a method for providing information necessary for determining the level of blood cancer cells wherein the sample is selected from the group consisting of blood, plasma, bone marrow fluid, lymph, saliva, tears, urine, mucous membrane fluid, and amniotic fluid in the above 10.
- a method for providing information necessary for determining the level of cancer cells in blood further comprising the step of treating a sample with a substance that specifically binds to EpCAM in the above 10.
- the blood cancer cells are blood cancer cells that have undergone epithelial-mesenchymal transition or are undergoing epithelial-mesenchymal transition.
- a method for providing information necessary for determining the level of blood cancer cells wherein the blood cancer cells are not epithelial cells or mesenchymal cells.
- the blood cancer cells are blood cancer cells in the blood of a patient with liver cancer, lung cancer, colon cancer, thyroid cancer, pancreatic cancer, malignant melanoma, breast cancer, neuroblastoma or glioblastoma, a method for providing information necessary for determining the level of blood cancer cells.
- a method for providing information necessary for determining the level of blood cancer cells wherein blood cancer cells in the above 10 more express B7-H3 (CD276) protein.
- a method for providing information necessary for predicting the prognosis of cancer metastasis comprising the step of detecting PTGFRN by treating a sample isolated from an individual with any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
- a method for providing information necessary for predicting the prognosis of cancer metastasis further comprising a step of providing information that if the level of detected PTGFRN is higher than that of the control group, the prognosis of cancer metastasis is worse than that of the control group.
- a method for screening the efficacy of an anticancer drug comprising the step of detecting PTGFRN by treating a sample isolated from a patient administered an anticancer drug with any one of the anti-PTGFRN monoclonal antibodies 1 to 3 above.
- a method for screening the efficacy of an anticancer drug further comprising a step of predicting that the anticancer drug has a better efficacy in inhibiting cancer metastasis than the control group if the level of PTGFRN detected in the above 20 is lower than that of the control group.
- composition for detecting cancer cells in blood comprising a substance that binds to PTGFRN.
- the anti-PTGFRN monoclonal antibody 63-D7 of the present invention can be used for determining the level of blood cancer cells, treating or preventing cancer, predicting the prognosis of cancer metastasis, and screening for anticancer drugs.
- the anti-PTGFRN monoclonal antibody of the present invention can specifically bind to PTGFRN on the surface of various cancer cells, including liver cancer.
- anti-PTGFRN monoclonal antibody of the present invention various antibody-drug conjugates can be developed and used as anticancer agents.
- the anti-PTGFRN monoclonal antibody of the present invention stimulates PTGFRN to increase the mobility and invasiveness of cancer cells
- the anti-PTGFRN monoclonal antibody of the present invention can be used to prevent and inhibit the cancer growth of primary cancer by promoting the migration of primary cancer cells.
- the anti-PTGFRN monoclonal antibody of the present invention can detect blood cancer cells that are neither epithelial nor mesenchymal.
- the anti-PTGFRN monoclonal antibody of the present invention can partially detect circulating cancer cells that have undergone epithelial-mesenchymal transition.
- the anti-PTGFRN monoclonal antibody of the present invention can detect blood cancer cells with high efficiency in liver cancer patients.
- Blood cancer cells detected by the anti-PTGFRN monoclonal antibody of the present invention are measured at a higher level in patients with secondary metastatic cancer than in patients with primary cancer, so that the stage of cancer can be determined through the anti-PTGFRN monoclonal antibody of the present invention.
- the anti-PTGFRN monoclonal antibody of the present invention can be internalized into cancer cells, thereby effectively delivering the drug into cancer cells.
- the present invention also provides PTGFRN as a surface marker of blood cancer cells.
- Anti-PTGFRN monoclonal antibodies can be used for the isolation, purification, and detection of PTGFRN protein through highly specific binding to PTGFRN protein.
- the anti-PTGFRN monoclonal antibody binds to epithelial-mesenchymal transition circulating cancer cells and partially epithelial-mesenchymal circulating cancer cells that express PTGFRN on their surface, thereby enabling the detection of previously undetectable circulating cancer cells.
- Anti-PTGFRN monoclonal antibodies may be useful in the diagnosis of metastatic cancer by detecting cancer cells in the blood.
- Fig. 1a is a graph showing the results of flow cytometry, showing the degree of binding of antibodies 246-D7, 247-B9 and monoclonal antibody 63-D7 of the present invention to human embryonic stem cell H9, two types of liver cancer cells (Huh7, HepG2), two types of lung cancer cells (A549, NCI-H358), and peripheral blood mononuclear cells (PBMC), respectively.
- Fig. 1b is a table showing the graph of Fig. 1a.
- Fig. 1b is a table showing the graph of Fig. 1a.
- FIG. 1c is a result of immunocytochemical staining, in order to confirm the detection of cancer cells in the blood, double staining was performed using monoclonal antibodies 63-D7, 246-D7, 247-B9 and CD45 antibody labeled with biotin in the blood of eight liver cancer patients and one normal person, respectively, and the nuclei were stained with DAPI, and a Merge of these was illustrated.
- Figure 1d shows the results of Figure 1c in a table.
- Figure 2 shows the results of flow cytometry, which shows that the monoclonal antibody 63-D7 of the present invention strongly binds to various cancer cells (SK-Hep1, A549, NCI-H358, NCI-H460, NCI-H1703, NCI-H23, Colo205, HCT116, SNU790, 8505C, BxPC3, A375, MCF-7, MDA-MB-435, SH-SY5Y, U87-MG) including human pluripotent stem cells (H9), human embryonic carcinoma cells (NT2/D1), and liver cancer cells (Huh7, HepG2, SNU387, SNU449) and human embryonic kidney cells (HEK293FT), but weakly binds to bone marrow-derived mesenchymal stem cells (BM-MSC) and human dermal fibroblasts (HDF), and human peripheral blood fibroblasts (PBS).
- BM-MSC bone marrow-derived mesenchymal stem cells
- HDF human dermal
- PBMCs blood mononuclear cells
- MRC5 human fetal lung fibroblasts
- DPCs dental pulp cells
- normal hepatocytes normal hepatocytes.
- the solid line is monoclonal antibody 63-D7 and the shaded background is a negative control containing only the secondary antibody.
- Figure 3a shows the results of separating proteins immunoprecipitated using monoclonal antibody 63-D7 from cell lysates of NT-2/D1 human embryonic cancer stem cells (NT-2/D1-biotin) whose cell surface was labeled with biotin, by 10% SDS-PAGE, Western blotting, transferring to a nitrocellulose membrane, and analyzing by reaction with streptavidin-HRP (SA-HRP).
- SA-HRP streptavidin-HRP
- Figure 3b shows the results of separating proteins immunoprecipitated in the same manner as in Figure 3a by 10% SDS-PAGE, and staining the polyacrylamide gel with PageBlue.
- 1X means that 3 mg of cell lysate was used, and 3x means that three times that amount was used.
- the protein within the dotted box in Fig. 3b was extracted and subjected to LC-MS/MS.
- Fig. 3c shows the result of analyzing the protein recovered after immunoprecipitation with monoclonal antibody 63-D7 in Fig. 3b by LC-MS/MS, and the part of the amino acid sequence of the 63-D7 antigen that matches the amino acid sequence of the PTGFRN protein is underlined.
- Figure 4a shows the results of Western blotting using mouse ⁇ -PTGFRN antibody after immunoprecipitation of proteins and a negative control protein without antibody (No Ab) using mouse anti-PTGFRN monoclonal antibody ( ⁇ -PTGFRN) and mouse 63-D7 monoclonal antibody from biotin-labeled human non-small cell lung cancer A549 cell lysates, respectively, separated by 10% SDS-PAGE.
- Figure 4b shows the results of analysis using streptavidin-HRP (SA-HRP) after removing the nitrocellulose membrane of Figure 4a with a signal removing solution, confirming that PTRFRN is a cell surface molecule recognized by 63-D7.
- SA-HRP streptavidin-HRP
- Figure 4c shows the results of immunoprecipitation and Western blotting of the cell lysate using a known mouse anti-FLAG monoclonal antibody ( ⁇ -FLAG), a known mouse anti-PTGFRN monoclonal antibody ( ⁇ -PTGFRN), and monoclonal antibody 63-D7 to reconfirm that the antigen of monoclonal antibody 63-D7 is PTGFRN.
- WB stands for Western blot.
- Figure 5a shows the results of a PTGFRN knockdown experiment performed in hepatoma cell lines Huh7 and SNU449.
- the expression of PTGFRN mRNA was analyzed by qPCR in hepatoma cells treated with a negative control siRNA (siCon) and two types of siRNA against PTGFRN (siPTGFRN#1 and siPTGFRN#2), respectively, to perform knockdown.
- Figure 5b shows the results of flow cytometry to determine the extent of binding of mouse PTGFRN monoclonal antibody ( ⁇ -PTGFRN) to PTGFRN on the surface of knocked-down Huh7 and SNU449 cells.
- siCon negative control siRNA
- siPTGFRN#1 and siPTGFRN#2 two types of siRNA against PTGFRN
- FIG. 5c is a graph statistically analyzing Figure 5b.
- Figure 5d shows the results of measuring cancer cell clonogenic survival in PTGFRN knockdown Huh7 and SNU449, and the surviving cell clones 8 days after cell inoculation were stained with crystal violet.
- Figure 5e is a graph statistically analyzing Figure 5d, and * indicates a p value of p ⁇ 0.05.
- Figure 6a shows the results of a PTGFRN knockdown experiment performed in hepatoma cell lines Huh7 and SNU449.
- the human embryonic carcinoma cell line HEK293FT was transfected with a negative control scrambled shRNA (shScramble) or two shRNAs for PTGFRN (shPTGFRN#1, shPTGFRN#2), and the lentivirus was recovered and treated to Huh7 and SNU449 hepatoma cells to perform knockdown.
- PTGFRN protein expression was confirmed by Western blotting.
- Figure 6b shows the results of analyzing the decrease in PTGFRN expression in Huh7 and SNU449 hepatoma cells using a flow cytometer.
- Figure 6c is a graph analyzing the statistical data of Figure 6b.
- Figure 6d shows the results of measuring cancer cell clonogenic survival in SNU449 knocked down with shPTGFRN#1. The cell clones that survived 8 days after cell inoculation were stained with crystal violet.
- shScramble is a control.
- Figure 6e is the result of statistical analysis of Figure 6d, where *** indicates p ⁇ 0.005 versus the control group (shScramble).
- Figure 6f is the result of measuring cancer cell clonogenic survival in SNU449 knocked down with shPTGFRN#2.
- Figure 6g is the result of statistical analysis of Figure 6f, where *** indicates p ⁇ 0.005.
- Figure 7a is a photograph of the results of measuring the degree of migration of liver cancer cells using a negative control shRNA (shScramble) or two shRNAs against PTGFRN (shPTGFRN#1, shPTGFRN#2).
- PTGFRN knockdown liver cancer cells Huh7 and SNU449 were dispensed into a 24-well transwell chamber and cultured for two days. The cells that had migrated to the lower chamber were stained with crystal violet and taken.
- Figure 7b is a graph that statistically processed the experiment in Figure 7a after repeating it three times. *** indicates p ⁇ 0.001, ** indicates p ⁇ 0.01, and * indicates p ⁇ 0.05.
- Figure 7c is a photograph of cells that had invaded the lower layer after being stained with crystal violet after being seeded on a Matrigel-coated transwell chamber and cultured for two days, measuring the invasiveness of cancer cells using a negative control shRNA (shscramble) or shRNA against two types of PTGFRN (shPTGFRN#1, shPTGFRN#2).
- Figure 7d is a graph that is the result of statistical processing after repeating the experiment in Figure 7c three times. *** indicates p ⁇ 0.001, ** indicates p ⁇ 0.01, and * indicates p ⁇ 0.05.
- Figure 8a is a photograph of attached liver cancer cells observed under a microscope after knockdown was performed by treating liver cancer cells with a negative control scrambled shRNA (shScramble) or two types of shRNA against PTGFRN (shPTGFRN#1, shPTGFRN#2) in the liver cancer cell line Huh7, and the degree of cell attachment was analyzed by reacting the PTGFRN knockdown liver cancer cells to a 12-well cell culture plate coated with Matrigel, CollagenI, CollagenIV, and gelatin.
- Figure 8b is a graph showing the degree of cell binding statistically processed by analyzing the microscope photographs using Image J after repeating the experiment in Figure 8a.
- Figure 8c is a photograph of attached liver cancer cells observed under a microscope after knockdown was performed by treating liver cancer cells with a negative control scrambled shRNA (shScramble) or two shRNAs against PTGFRN (shPTGFRN#1, shPTGFRN#2) in the liver cancer cell line SNU449, and the degree of cell attachment was analyzed by reacting the PTGFRN knockdown liver cancer cells to a 12-well cell culture plate coated with Matrigel, CollagenI, CollagenIV, and gelatin.
- shScramble negative control scrambled shRNA
- shPTGFRN#1, shPTGFRN#2 shRNAs against PTGFRN
- Figure 8d is a graph showing the degree of cell binding statistically processed by analyzing the microscope photographs using Image J after repeating the experiment in Figure 8c. *** indicates p ⁇ 0.001, ** indicates p ⁇ 0.01, * indicates p ⁇ 0.05, and ns indicates no statistical significance.
- Figure 9a shows the results of measuring the immune evasion ability of PTGFRN knockdown liver cancer cells against NK cells.
- SNU449 liver cancer cells were treated with negative control scrambled shRNA (shscramble) or shPTGFRN#2 to perform knockdown, and the PTGFRN knockdown liver cancer cells were co-cultured with NK92, and the remaining SNU449 cells were stained with crystal violet. 1:1, 2.5:1, and 5:1 represent the ratios (E:T ratios) of NK92, which is an effector cell, and SNU449, which is a target cell.
- Figure 9b is a graph showing the results of quantifying the absorbance at OD540 after melting the cells in Figure 9a.
- Figure 9c is a graph showing the degree of cell death by expressing the OD540 results measured in Figure 9b as an E:T ratio.
- Figure 10 shows the results of Western blot analysis of the changes in the expression of B7-H3, FAK, Src proteins, and phosphorylated p-FAK (Y397), p-Src (Y416) proteins in the protein lysates of PTGFRN knockdown hepatoma cells Huh7 and SNU449, which were treated with a negative control shRNA (shScramble) or two types of shRNA against PTGFRN (shPTGFRN#1, shPTGFRN#2). GAPDH is the control, and the numbers are quantified using Image J.
- Figure 11a shows the results of immunoprecipitation of the lysate obtained after co-transfection of HEK293FT cells with pcDNA3.1(+)PTGFRN-FLAG vector and pcDNA3.1(+)PTGFRN-myc vector to confirm the cis interaction of the same PTGFRN in one cell with mouse anti-FLAG, rabbit anti-myc, and 63-D7, and Western blotting with anti-FLAG and anti-myc antibodies. No Ab is the control, and the arrow indicates the immunoprecipitated PTGFRN.
- Figure 11b shows the results of mixing the lysates of HEK293FT cells transfected with pcDNA3.1(+)PTGFRN-FLAG vector and pcDNA3.1(+)PTGFRN-myc vector, respectively, and immunoprecipitating them with mouse anti-FLAG, rabbit anti-myc, and 63-D7 to confirm trans-interactions between different cells, and performing Western blotting with anti-FLAG and anti-myc antibodies. No Ab is the control, and the arrow indicates immunoprecipitated PTGFRN.
- Figure 11c shows the results of Western blotting with ⁇ -PTGFRN after immunoprecipitation with mouse anti-PTGFRN monoclonal antibody ( ⁇ -PTGFRN), mouse anti-63-D7 monoclonal antibody, mouse anti-SLC3A2, and mouse anti-B7-H3, respectively, in hepatoma cell SNU449, showing that PTGFRN is immunoprecipitated by SLC3A2 (CD98hc) and B7-H3.
- ⁇ -PTGFRN mouse anti-PTGFRN monoclonal antibody
- mouse anti-63-D7 monoclonal antibody mouse anti-SLC3A2
- mouse anti-B7-H3 mouse anti-B7-H3
- Figure 11d shows the results of co-transfecting HEK293FT cells with pcDNA3.1(+)PTGFRN-myc vector and pcDNA3.1(+)B7-H3-FLAG vector, followed by immunoprecipitation with anti-FLAG, anti-B7-H3, anti-myc, and 63-D7, and detection with anti-myc and anti-FLAG antibodies, confirming that they interact with each other.
- HC stands for immunoglobulin heavy chain.
- WB stands for western blot.
- Figures 12a and 12b show the results of measuring cell adhesion on a Materigel-coated plate after sorting 63-D7 positive and negative cells with 63-D7 antibody and magnetic beads in liver cancer cell lines Huh7 and HepG2, respectively.
- Figure 12c shows the results of flow cytometry analysis to compare the cancer cell binding ability of 63-D7 with that of CD133, CD44, and EpCAM, which are cancer stem cell positive markers, when they are adherent cells, after culturing Huh7 as tumor cells to increase cancer stem cell potential.
- Figure 12d is a graph statistically analyzing Figure 12c. *** indicates p ⁇ 0.005.
- FIG. 12e is a graph showing the results of measuring the clonogenic survival of 63-D7 positive and negative cancer cells in the liver cancer cell line Huh7 sorted with 63-D7, showing the cancer stemness of the cell clones that survived 8 days after inoculation, stained with crystal violet.
- Fig. 12f is a graph showing the statistical results of Fig. 12e. *** indicates p ⁇ 0.005 compared to the control group.
- Fig. 12g is a graph showing the results of measuring the clonogenic survival of 63-D7 positive and negative cancer cells in the liver cancer cell line HepG2 sorted with 63-D7.
- Fig. 12h is a graph showing the statistical results of Fig. 12g. *** indicates p ⁇ 0.005 compared to the control group.
- Figures 13a and 13b show the results of analyzing the internalization of monoclonal antibody 63-D7 into human liver cancer cells Huh7, HepG2, SNU449 and human pancreatic cancer cells SNU213 and BxPC3 by flow cytometry.
- the solid line is the monoclonal antibody and 63-D7 reacted at 4°C
- the dotted line is the 63-D7 antibody reacted at 37°C after the reaction at 4°C
- the shaded background includes only the secondary antibody.
- Figure 13c is a graph comparing the degree of attachment of 63-D7 to the cell surface of each cancer cell by statistically processing it as the relative average fluorescence intensity by repeating the same experiment as Figure 13a three times.
- Figure 13d is a graph comparing the degree of attachment of 63-D7 to the cell surface of each cancer cell by statistically processing the relative average fluorescence intensity by repeating the same experiment as Figure 13b three times. **** indicates p ⁇ 0.001, *** indicates p ⁇ 0.005, ** indicates p ⁇ 0.01, and * indicates p ⁇ 0.05.
- Figures 14a and 14b show the results of measuring the cell viability of Huh7 cells treated with 63-D7 or mouse IgG isotype antibody. After Huh7 cells were treated with 0 nM to 100 nM of 63-D7 or mouse isotype antibody, respectively, Figure 14a shows that they were treated with ⁇ -mFc-CL-DMDM and Figure 14b shows that they were treated with ⁇ -mFc-CL-MMAF for 48 hours, and the cell viability was measured using CCK-8.
- * indicates p ⁇ 0.05
- ** indicates p ⁇ 0.01
- **** indicates p ⁇ 0.001.
- Fig. 15a is a drawing showing a method for detecting blood cancer cells recognized by the PTGFRN-specific monoclonal antibody 63-D7 of the present invention in the blood of a liver cancer patient, and shows a method for removing CD45 positive cells and staining the remaining cells with Dylight488-conjugated 63-D7 and another marker (antibody).
- Fig. 15b is a graph showing the recovery rate measured by counting the Huh7 cells recovered after removing CD45 positive cells as shown in 15a by adding 0, 10, 30, 50, and 100 Huh7 cells to PBMC.
- Fig. 15c shows the blood cancer cells recovered as shown in Fig. 15a, stained with Dylight649-conjugated anti-mouse IgG and Dylight 488-conjugated 63-D7, and DAPI that stains the nucleus, and Merge that merges them.
- FIGS. 16A to 16C are confocal microscopy images showing blood cancer cells double-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention and anti-MVP, anti-HSA, anti-PanCK, anti-E-cadherin, anti-EpCAM, 63-D7, anti-Vimentin, anti-Twist, and anti-ZEB1 antibodies in the blood of a liver cancer patient, and DAPI that stains the nucleus and Merge that merges them.
- Figure 17 is a graph comparing the number of all blood cancer cells (63-D7+ cells/ml) recognized by the monoclonal antibody 63-D7 of the present invention in the blood of liver cancer patients (53 primary HCC and 42 metastatic HCC) and normal people and hepatitis patients (26 non-neoplastic) through a comparative test (Kruskal-Wallis Test) among the three groups (p value ⁇ 0.0001). It shows that the p-values for each group are all ⁇ 0.0001, indicating that there is significance in the differences among all three groups.
- FIG. 18a is a confocal microscope image showing circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-EpCAM, and anti-Vimentin antibodies in the blood of a liver cancer patient, together with DAPI, which stains the nucleus, and a Merge, which merges them.
- BF stands for bright field.
- 18b is a confocal microscope image showing circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-MVP, and anti-B7-H3 antibodies in the blood of a liver cancer patient, together with DAPI, which stains the nucleus, and a Merge, which merges them.
- BF stands for bright field.
- BF stands for bright field.
- Figure 19 is a confocal microscope image showing blood cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-TGF ⁇ R1, and anti-B7-H3 antibodies in the blood of a liver cancer patient, along with DAPI that stains the nucleus and a Merge that combines them.
- BF stands for bright field.
- FIG. 20a is a confocal microscope image of circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-ULBP1, and anti-B7-H3 antibodies in the blood of a liver cancer patient, together with DAPI, which stains the nucleus, and a Merge, which merges them.
- BF stands for bright field.
- 20b is a confocal microscope image of circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-MICA/B, and anti-B7-H3 antibodies in the blood of a liver cancer patient, together with DAPI, which stains the nucleus, and a Merge, which merges them.
- BF stands for bright field.
- FIG. 21a is a confocal microscope image showing circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-PD-L1, and anti-B7-H3 antibodies in the blood of a liver cancer patient, together with DAPI that stains the nucleus and a Merge that merges them.
- BF stands for bright field.
- FIG. 21b is a confocal microscope image showing circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-PD-L2, and anti-B7-H3 antibodies in the blood of a liver cancer patient, together with DAPI that stains the nucleus and a Merge that merges them.
- BF stands for bright field.
- Figure 22 is a confocal microscope image showing blood cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-CD47, and anti-B7-H3 antibodies in the blood of a liver cancer patient, along with DAPI that stains the nucleus and a Merge that combines them.
- BF stands for bright field.
- Figure 23 shows the base sequence and amino acid sequence of the heavy chain gene variable region of the monoclonal antibody 63-D7, with the CDR (Complementarity Determining Region) that binds to the antigen indicated in bold.
- Figure 24 shows the base sequence and amino acid sequence of the light chain gene variable region of the monoclonal antibody 63-D7, with the CDR (Complementarity Determining Region) that binds to the antigen indicated in bold.
- the present invention provides an anti-PTGFRN monoclonal antibody for determining the level of blood cancer cells, treating or preventing cancer, predicting the prognosis of cancer metastasis, and screening for anticancer drugs.
- the present invention provides an anti-PTGFRN monoclonal antibody comprising a heavy chain variable region comprising HCDR1 consisting of the amino acid sequence of SEQ ID NO: 3, HCDR2 consisting of the amino acid sequence of SEQ ID NO: 4, and HCDR3 consisting of the amino acid sequence of SEQ ID NO: 5; and a light chain variable region comprising LCDR1 consisting of the amino acid sequence of SEQ ID NO: 6, LCDR2 consisting of the amino acid sequence of SEQ ID NO: 7, and LCDR3 consisting of the amino acid sequence of SEQ ID NO: 8.
- antibody as used herein includes a complete antibody and any antigen-binding fragment (i.e., "antigen-binding portion") or single chain thereof.
- An antibody refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains joined to each other by disulfide bonds, or an antigen-binding portion thereof.
- Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
- the heavy chain constant region is comprised of three domains, CH1, CH2, and CH3.
- Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
- the light chain constant region is comprised of one domain, CL.
- the VH and VL regions can be further subdivided into hypervariable regions, called complementarity determining regions (CDRs), interspersed with more conserved regions, called framework regions (FRs).
- CDRs complementarity determining regions
- Each VH and VL is composed of three CDRs and four FRs, namely FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4, arranged in the following order from amino terminus to carboxy terminus.
- the variable regions of the heavy and light chains comprise a binding domain that interacts with an antigen.
- the constant regions of the antibody can mediate binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
- the CDRs included in the VH region are called heavy chain complementary determining region 1 (HCDR1), HCDR2, and HCDR3, and the CDRs included in the VL region are called light chain complementary determining region 1 (LCDR1), LCDR2, and LCDR3.
- antibody portion refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., PTGFRN). It has been determined that the antigen binding function of an antibody can also be exerted by fragments of full-length antibodies.
- an antigen e.g., PTGFRN
- binding fragments encompassed by the term "antigen-binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of one arm of an antibody; (v) a dAb fragment consisting of a VH domain [Ward et al., (1989) Nature 341:544-546]; and (vi) an isolated complementarity determining region (CDR) or (vii) a combination of two or more isolated CDRs, which may, in some cases, be linked by a synthetic linker.
- CDR complementarity determining region
- the two domains of the Fv fragment namely the VL and VH
- they can be joined by a synthetic linker, which can be prepared by recombinant methods, into a single protein chain in which the VL and VH regions pair to form a monovalent molecule (called single-chain Fv (scFv)).
- single-chain Fv single-chain Fv
- Such single-chain antibodies are also included in the term "antigen-binding portion" of an antibody.
- antibody fragments can be obtained by conventional techniques known to those skilled in the art, and these fragments are screened for utility in the same manner as complete antibodies.
- the antigen-binding portion can be prepared by recombinant DNA techniques, or by enzymatic or chemical cleavage of complete immunoglobulins.
- the antibody herein may be a monoclonal antibody or a polyclonal antibody.
- the antibody may be produced using various techniques known in the art for producing humanized antibodies.
- monoclonal antibody refers to an antibody that exhibits a single binding specificity and exhibits affinity for a particular epitope.
- PTGFRN-specific monoclonal antibodies can be efficiently produced in virtually unlimited quantities in a highly purified form. PTGFRN-specific monoclonal antibodies can specifically bind to a specific epitope of PTGFRN.
- the antibodies herein that bind to PTGFRN can be produced by viral or oncogene transformation of B cells fused to immortalized cells obtained from a nonhumanized animal having a genome comprising a heavy chain transgene and a light chain transgene, phage display techniques using a library of human antibody genes, somatic cell hybridization techniques, etc.
- Methods for producing hybridomas in animal systems, including immunization protocols and isolation and fusion techniques of immunized spleen cells, for producing monoclonal antibodies are well known in the art.
- Antibodies or antigen-binding fragments thereof can be labeled with radionuclides, fluorescors, enzymes, etc.
- epitopes refers to a portion of an antigen to which an immunoglobulin or antibody specifically binds.
- Epitopes may be formed from adjacent amino acids or from non-adjacent amino acids that are juxtaposed by tertiary folding of the protein. Epitopes formed from adjacent amino acids are generally retained upon exposure to denaturing solvents, whereas epitopes formed by tertiary folding are generally lost upon treatment with denaturing solvents.
- Epitopes typically comprise at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acids in a unique spatial conformation.
- the epitope (or antigenic determinant site) functions as a conformational epitope in that no additional increase in affinity for the epitope/antigenic determinant site is observed even when it is within a larger amino acid segment (e.g., a protein having a three-dimensional structure).
- isotype refers to the class of antibodies (e.g., IgM or IgG1) encoded by the heavy chain constant region genes.
- the monoclonal antibodies of the invention are of the IgG1 isotype.
- the present invention produces a monoclonal antibody that binds to the surface of human embryonic stem cells, and searches for an antigen of monoclonal antibody 63-D7, which binds to the surface of human embryonic stem cells, embryonic cancer cells, and various cancer cells, but does not bind to normal cells such as human peripheral mononuclear cells or human hepatocytes.
- the 63-D7 monoclonal antibody recognizes the cell surface protein PTGFRN (Prostaglandin F2 Receptor Negative Regulator, CD315).
- PTGFRN Prostaglandin F2 Receptor Negative Regulator, CD315
- PTGFRN recognized by the 63-D7 monoclonal antibody is expressed in cell lines such as liver cancer, lung cancer, colon cancer, pancreatic cancer, and thyroid cancer.
- the results of an experiment on 95 patients with hepatocellular carcinoma confirmed that the 63-D7 monoclonal antibody can detect circulating cancer cells with an efficiency of 97%. Since the 63-D7 antibody is co-expressed with Vimentin, an EMT marker, at a ratio of about 53% in primary HCC and about 28% in secondary HCC, it was confirmed that the 63-D7 antibody can be used to detect circulating cancer cells showing an EMT phenotype. In addition, circulating cancer cells recognized by 63-D7 can also detect intermediate circulating cancer cells that do not express either EpCAM or Vimentin, and specifically, it was confirmed that expression was found at a ratio of about 46% in primary HCC and about 72% in secondary HCC.
- the 63-D7 antibody is a new marker that can detect both intermediate phenotype and EMT phenotype circulating cancer cells that could not be identified by existing EpCAM marker-based circulating cancer cell diagnostic technology.
- PTGFRN Prostaglandin F2 Receptor Negative Regulator, EWI-F, CD9P-1, CD315) is a type I transmembrane protein of the immunoglobulin superfamily, which negatively affects the Prostaglandin F2 Receptor.
- PTGFRN includes a full-length sequence, a fragment that performs an equivalent function, or a polypeptide having a continuous amino acid sequence of PTGFRN (e.g., the PTGFRN amino acid sequence disclosed in FIG. 3c).
- PTGFRN may be, but is not limited to, a PTGFRN derived from a vertebrate, more specifically, a human or mouse.
- the present invention provides an anti-PTGFRN monoclonal antibody, wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 1.
- the present invention may be an antibody comprising a single chain variable region (scFv) comprising an amino acid sequence that is at least 80%, 85%, 90%, 95%, 98%, 99% or more identical to the heavy chain variable region amino acid sequence (SEQ ID NO: 1) produced by the anti-PTGFRN monoclonal antibody clone 63-D7 of the present invention.
- scFv single chain variable region
- the present invention provides an anti-PTGFRN monoclonal antibody, wherein the light chain variable region comprises the amino acid sequence of SEQ ID NO: 2.
- the anti-PTGFRN monoclonal antibody of the present invention may be an antibody comprising a single chain variable region (scFv) comprising an amino acid sequence that is at least 80%, 85%, 90%, 95%, 98%, 99% or more identical to the light chain variable region amino acid sequence (SEQ ID NO: 2) produced by clone 63-D7.
- scFv single chain variable region
- the present invention provides an antibody-drug conjugate (ADC) in which a drug is conjugated to the PTGFRN monoclonal antibody of the present invention.
- ADC antibody-drug conjugate
- the monoclonal antibody of the present invention can promote cellular internalization of PTGFRN in various cancer cells, and is therefore suitable as an antibody used in antibody-drug conjugates.
- the drug can be selected by those skilled in the art according to the purpose without limitation in type.
- it can be a known anticancer drug, but is not limited thereto.
- the method of conjugation can be selected without limitation by a method known in the art as long as it can bring the antibody and the drug into contact.
- the present invention provides a pharmaceutical composition for treating or preventing cancer comprising the anti-PTGFRN monoclonal antibody of the present invention.
- the present invention provides a method for treating, preventing or diagnosing cancer comprising the anti-PTGFRN monoclonal antibody of the present invention.
- cancer may be any one selected from the group consisting of brain and spinal tumors, head and neck cancer, lung cancer, breast cancer, thymoma, mesothelioma, esophageal cancer, stomach cancer, colon cancer, liver cancer, thyroid cancer, pancreatic cancer, biliary tract cancer, kidney cancer, bladder cancer, prostate cancer, testicular cancer, germ cell tumor, ovarian cancer, cervical cancer, endometrial cancer, lymphoma, acute leukemia, chronic leukemia, multiple myeloma, sarcoma, malignant melanoma, neuroblastoma, glioblastoma, and skin cancer, but is not limited thereto.
- Metastatic cancer is cancer that has spread from one organ to another, including the lymph nodes.
- the cancer present in the organ where the cancer has spread is called the primary cancer.
- metastases can be brain metastases, bone metastases, liver metastases, or lung metastases.
- Metastatic cancer can be a malignant tumor.
- a malignant tumor can be a brain or spinal tumor, head and neck cancer, lung cancer, breast cancer, thymoma, mesothelioma, esophageal cancer, stomach cancer, colon cancer, liver cancer, pancreatic cancer, biliary tract cancer, kidney cancer, bladder cancer, prostate cancer, testicular cancer, germ cell tumor, ovarian cancer, cervical cancer, endometrial cancer, lymphoma, acute leukemia, chronic leukemia, multiple myeloma, sarcoma, malignant melanoma, skin cancer, or a combination of these.
- the monoclonal antibody of the present invention is an agonistic antibody for the cancer, and can stimulate PTGFRN on the surface of cancer cells to promote the movement and invasiveness of cancer cells.
- the monoclonal antibody can promote the movement of cancer cells in primary cancer, thereby preventing the occurrence of early primary cancer or inhibiting its growth.
- the present invention provides a hybridoma producing the above anti-PTGFRN monoclonal antibody.
- a hybridoma may include a hybridoma cell or a hybridoma cell line.
- the hybridoma is a cell created by artificially fusing two different types of cells, and refers to a cell or cell line in which two or more homologous or heterologous cells are fused using a substance that causes cell fusion, such as polyethylene glycol (PEG) or a certain type of virus, thereby integrating different functions of each cell into a single cell.
- PEG polyethylene glycol
- a hybridoma that secretes a monoclonal antibody can be cultured in large quantities in vitro or in vivo.
- the present invention provides a composition for detecting PTGFRN comprising the anti-PTGFRN monoclonal antibody of the present invention.
- the above antigen-antibody complex can be detected using a detection label.
- the detection label can be selected from enzymes, fluorescent substances, ligands, luminescent substances, microparticles, redox molecules, or radioisotopes, but is not particularly limited thereto.
- the present invention provides a method for providing information for diagnosing cancer, comprising the step of treating a separated sample with an anti-PTGFRN monoclonal antibody of the present invention.
- an entity may be a mammal, including a human.
- a biological sample refers to a sample obtained from a living organism.
- the biological sample may be, for example, blood, plasma, bone marrow fluid, lymph, saliva, tears, urine, mucous membrane fluid, amniotic fluid, or a combination thereof.
- the substance that specifically binds to PTGFRN may be an anti-PTGFRN antibody or an antigen-binding fragment thereof or an aptamer.
- An aptamer refers to an oligonucleic acid or peptide that binds to a target molecule.
- the present invention provides a method for providing information necessary for determining the level of blood cancer cells, comprising the step of treating a sample isolated from an individual with an anti-PTGFRN monoclonal antibody of the present invention.
- it can provide information necessary for diagnosing metastatic cancer, including the steps of contacting a biological sample isolated from an individual with the anti-PTGFRN monoclonal antibody of the present invention to bind blood cancer cells in the sample to the anti-PTGFRN monoclonal antibody of the present invention; detecting the blood cancer cells from the reaction mixture; and determining, if the blood cancer cells are detected, that the individual has or is likely to have metastatic cancer.
- the anti-PTGFRN monoclonal antibody of the present invention can specifically bind to PTGFRN, PTGFRN can be detected, and furthermore, PTGFRN can be separated or purified using the composition.
- PTGFRN By contacting the anti-PTGFRN monoclonal antibody of the present invention with a sample and detecting the formation of an antigen-antibody complex, the PTGFRN protein can be separated, identified, or detected.
- the antigen-antibody complex refers to a combination of PTGFRN and a monoclonal antibody that recognizes it for the purpose of confirming the presence or absence of PTGFRN in a sample.
- the antigen-antibody complex can be detected using a detection label.
- the label can be selected from an enzyme, a fluorescent substance, a ligand, a luminescent substance, a microparticle, a redox molecule, or a radioisotope, but is not particularly limited thereto.
- antigen-antibody complexes can be detected using a colorimetric method, an electrochemical method, a fluorescence method, a luminometry method, a particle counting method, a visual assessment, or a scintillation counting method.
- a colorimetric method an electrochemical method, a fluorescence method, a luminometry method, a particle counting method, a visual assessment, or a scintillation counting method.
- it can be detected by flow cytometry, immunocytochemistry, radioimmunoassay (RIA), enzyme-linked immunosorbent assay (ELISA), Western blotting, immunoprecipitation assay, immunodiffusion assay, complement fixation assay, protein chip, etc.
- immune precipitation and immunoblotting which are useful for recovering a small amount of target protein.
- an antigen-antibody complex can be detected using an enzyme-linked immunosorbent assay (ELISA).
- the ELISA includes various ELISA methods, such as a direct ELISA using a labeled antibody that recognizes an antigen attached to a solid support, an indirect ELISA using a labeled antibody that recognizes a capture antibody in a complex of antibodies that recognize the antigen attached to the solid support, a direct sandwich ELISA using another labeled antibody that recognizes an antigen in a complex of an antibody and an antigen attached to the solid support, and an indirect sandwich ELISA using a labeled secondary antibody that recognizes the antibody after reacting with another antibody that recognizes the antigen in a complex of an antibody and an antigen attached to the solid support.
- Circulating tumor cells are rare tumor cells that exist in the blood and circulate in the body after undergoing a tumor invasion process. Circulating tumor cells are known to be a factor involved in cancer metastasis and recurrence. Circulating tumor cells may be circulating tumor cells that have undergone epithelial-mesenchymal transition.
- Epithelial-mesenchymal transition is the process by which epithelial cells lose cell polarity and cell-to-cell adhesion and transform into mesenchymal cells that are mobile and invasive. Epithelial-mesenchymal transition is essential for many developmental processes, including mesoderm formation and neural tube formation, and is known to occur in wound healing, organ fibrosis, and in the initiation of metastasis during cancer progression.
- the circulating cancer cells may be circulating cancer cells that have undergone epithelial-mesenchymal transition, circulating cancer cells that have not undergone epithelial-mesenchymal transition, or circulating cancer cells that are undergoing epithelial-mesenchymal transition, and are preferably, but not limited to, circulating cancer cells that have undergone epithelial-mesenchymal transition or are undergoing epithelial-mesenchymal transition.
- Cancer cells in the blood may be neither epithelial nor mesenchymal cells.
- detection of blood cancer cells comprises separating a complex in which blood cancer cells and a substance that specifically binds to PTGFRN are combined from a biological sample.
- the detection method comprises electron microscopy, microfiltration, centrifugation, microfluidics, immunostaining, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA), flow cytometry, fluorescense activated cell sorting (FACS), or a combination thereof.
- the detection method may be polymerase chain reaction (PCR), electrophoresis, northern blotting, western blotting, or a combination thereof.
- PTGFRN was confirmed to be expressed in liver cancer, lung cancer, colon cancer, pancreatic cancer, and thyroid cancer cell lines, and 97% of hepatocellular carcinoma patients were detected with circulating cancer cells.
- an EMT marker in about 53% of primary HCCs and about 28% of secondary HCCs, it was confirmed to be useful for detecting circulating cancer cells showing an EMT phenotype and can be used for diagnosing metastatic cancer.
- circulating cancer cells recognized by 63-D7 were simultaneously expressed in about 46% of primary HCCs and about 72% of secondary HCCs, even in intermediate circulating cancer cells that did not express either EpCAM or Vimentin, so it can be usefully used for detecting circulating cancer cells by complementing the shortcomings of existing EpCAM marker-based circulating cancer cell diagnosis technology.
- the blood cancer cells may be derived from a primary cancer selected from the group consisting of brain and spinal cancer, head and neck cancer, lung cancer, breast cancer, thymoma, mesothelioma, esophageal cancer, stomach cancer, colon cancer, liver cancer, pancreatic cancer, biliary tract cancer, kidney cancer, bladder cancer, prostate cancer, testicular cancer, germ cell tumor, ovarian cancer, cervical cancer, endometrial cancer, lymphoma, acute leukemia, chronic leukemia, multiple myeloma, sarcoma, malignant melanoma, and skin cancer.
- a primary cancer selected from the group consisting of brain and spinal cancer, head and neck cancer, lung cancer, breast cancer, thymoma, mesothelioma, esophageal cancer, stomach cancer, colon cancer, liver cancer, pancreatic cancer, biliary tract cancer, kidney cancer, bladder cancer, prostate cancer, testicular cancer, germ cell tumor, ovarian cancer, cervical
- the present invention provides a method for providing information necessary for determining the level of cancer cells in blood, which further includes a step of treating a sample isolated from an individual with a substance that specifically binds to EpCAM.
- the present invention provides a method for providing information necessary for determining the level of cancer cells in blood, wherein the substance that specifically binds to EpCAM is an anti-EpCAM antibody or an antigen-binding fragment thereof.
- EpCAM is a transmembrane glycoprotein that mediates Ca 2+ -dependent homotypic cell-cell adhesion in epithelial cells. EpCAM is known to be involved in cell signaling, migration, proliferation, and differentiation. EpCAM can be human or mouse EpCAM. For example, EpCAM can be a polypeptide having an amino acid sequence of GenBank Accession No. NP_002345. For example, EpCAM can be a polypeptide encoded by a nucleotide sequence of GenBank Accession No. NM_002354. The substance that specifically binds to EpCAM can be an anti-EpCAM antibody or an antigen-binding fragment thereof. The antibody or antigen-binding fragment thereof is as described above.
- the blood cancer cells may be blood cancer cells undergoing epithelial-mesenchymal transition or undergoing epithelial-mesenchymal transition.
- the blood cancer cells may be in a hybrid E/M state.
- the blood cancer cells may be neither epithelial cells nor mesenchymal cells.
- the blood cancer cells may be blood cancer cells in the blood of a patient with liver cancer, lung cancer, colon cancer, thyroid cancer, pancreatic cancer, malignant melanoma, breast cancer, neuroblastoma, or glioblastoma.
- the blood cancer cells can further express B7-H3 (CD276) protein.
- the present invention provides a method for providing information necessary for predicting the prognosis of cancer metastasis, comprising the step of detecting PTGFRN by treating a sample isolated from an individual with the anti-PTGFRN monoclonal antibody of the present invention.
- the present invention provides a method for providing information necessary for predicting the prognosis of cancer metastasis, further comprising a step of providing information that if the level of detected PTGFRN is higher than that of the control group, the prognosis of cancer metastasis is worse than that of the control group.
- the present invention provides a method for screening the efficacy of an anticancer drug, comprising the step of detecting PTGFRN by treating a sample isolated from a patient administered an anticancer drug with an anti-PTGFRN monoclonal antibody of the present invention.
- the present invention provides a method for screening the efficacy of an anticancer drug, further comprising a step of predicting that the anticancer drug will have a better efficacy in inhibiting cancer metastasis compared to a control group if the level of detected PTGFRN is lower than that of a control group.
- the present invention provides a composition for detecting cancer cells in blood, comprising a substance that binds to PTGFRN.
- the present invention provides a composition for isolating, purifying, detecting and detecting the concentration of PTGFRN protein, which comprises a substance that binds to PTGFRN.
- composition for detecting blood cancer cells of the present invention may further include a substance that specifically binds to epithelial cell adhesion molecule (EpCAM).
- EpCAM epithelial cell adhesion molecule
- the substance binding to PTGFRN may be an anti-PTGFRN antibody or an aptamer.
- the anti-PTGFRN antibody may be a monoclonal antibody or a polyclonal antibody.
- the circulating cancer cells may be, but are not limited to, circulating cancer cells in the blood of a patient with liver cancer, lung cancer, colon cancer, thyroid cancer, pancreatic cancer, melanoma, breast cancer, neuroblastoma, or glioblastoma.
- the circulating cancer cells may be circulating cancer cells that have undergone epithelial-mesenchymal transition or are undergoing epithelial-mesenchymal transition.
- the circulating cancer cells may be in a hybrid E/M state.
- the substance binding to PTGFRN may be an anti-PTGFRN antibody of the present invention.
- the composition for detecting blood cancer cells in blood of the present invention may be included in the form of a kit.
- the kit of the present invention when the kit of the present invention is applied to an immunoassay, the kit of the present invention may optionally include a secondary antibody and a substrate of a label.
- the kit according to the present invention may be manufactured into a plurality of separate packages or compartments containing the above-mentioned reagent components.
- the composition for detecting blood cancer cells of the present invention may be included in the form of a microarray.
- the monoclonal antibody is used as a hybridizable array element and is immobilized on a substrate.
- a preferred substrate is a suitable rigid or semi-rigid support, such as a membrane, a filter, a chip, a slide, a wafer, a fiber, a magnetic bead or a non-magnetic bead, a gel, a tubing, a plate, a polymer, a microparticle, and a capillary.
- the hybridizable array element is arranged and immobilized on the substrate, and such immobilization may be performed by a chemical bonding method or a covalent bonding method such as UV.
- the hybridizable array element may be bonded to a glass surface modified to include an epoxy compound or an aldehyde group, and may also be bonded to a polylysine-coated surface by UV.
- the hybridizable array element may be bonded to the substrate through a linker (e.g., ethylene glycol oligomer and diamine).
- the present invention provides a composition for monitoring cancer metastasis comprising a substance that binds to PTGFRN.
- the substance is not limited as long as it can measure the level of PTGFRN.
- Human embryonic stem cells H9 were purchased from the Wicell Research Institute and cultured according to the provided protocol.
- the culture medium contained DMEM/F12 (Invitrogen, Seoul, Korea), 20% Knockout SR (Invitrogen), 0.1 mM ⁇ -mercaptoethanol (Sigma, St Luis, MO, USA), 2 mM glutamine (Invitrogen), 0.1 mM nonessential amino acids (Invitrogen), 100 U/ml penicillin G (Sigma), 100 ⁇ g/ml streptomycin (Sigma), and 4 ng/ml bFGF (PeproTech, Rocky Hill, NJ).
- Human embryonic carcinoma NT-2/D1 cells were purchased from ATCC (Manassas, VA, USA) and cultured according to the provided protocol.
- Human hepatocarcinoma Human hepatocarcinoma (Huh7, HepG2, SNU387, SNU449), non-small cell lung cancer (A549, NCI-H358, NCI-H460, NCI-H1703, NCI-H23), and thyroid cancer (SNU-790) cells were purchased from Korea Cell Line Bank (KCLB, Seoul, Korea).
- Human normal lung fetal fibroblast (MRC5), colon cancer (Colo205), and pancreatic cancer (BxPC3) cells were purchased from ATCC, and human hepatocytes were purchased from Termo Fisher Scientific (Waltham, USA) and cultured according to the provided protocol.
- PBMCs Human peripheral blood mononuclear cells
- Human embryonic stem cells (H9) and human peripheral mononuclear cells (PBMC) were also used in the analysis.
- PBA 1% bovine serum albumin, 0.02% NaN 3 in PBS, pH 7.4
- the antibodies were reacted for 30 minutes at 4 °C, respectively.
- the primary antibody and the corresponding anti-mouse IgG-FITC were reacted for another 30 minutes at 4 °C.
- the antibody reaction was analyzed for propidium iodide (PI)-negative cells using FACS Calibur and Cell Quest software (BD sciences).
- 63-D7, 246-D7, and 247-B9 antibodies that bound to human embryonic stem cells (H9), liver cancer cell lines (Huh7, HepG2), and lung cancer cell lines (A549) but not to PBMCs were selected (Fig. 1a, Fig. 1b).
- 246-D7 and 247-B9 bound to the epithelial lung cancer cell line NCI-H358, but 63-D7 did not bind.
- 63-D7, 246-D7, and 247-B9 antibodies were purified by protein G-agarose column chromatography, and the purified antibodies were biotinylated according to the protocol provided using the DSB-XTM Biotin Protein Labeling Kit (Molecular Probes, Seoul, Korea).
- PBMCs were isolated from the blood of one healthy adult and eight patients with hepatocellular carcinoma (HCC) using the same method as in Example 1-2.
- the recovered PBMC cells were divided into 2 to 4 cells depending on the number of cells and attached to poly-L-lysine-coated slides.
- the prepared slides were treated with 4% paraformaldehyde (PFA) for 10 minutes at room temperature to fix the cells.
- PFA paraformaldehyde
- PBS pH 7.4 containing 1 mM Ca 2+ and 0.5 mM Mg 2+ was always used in all PBS used for cell staining.
- the cells were blocked with blocking solution (10% horse serum, 0.1% BSA, PBS, pH 7.4) for 1 hour. Then, the plate was reacted with mouse antibody CD45 antibody (BD Biosciences, Seoul Korea) at room temperature, blocked from light, for 1 hour, followed by anti-mouse IgG-Alexa488 (Invitrogen, 1:3000) at room temperature, blocked from light, for another 1 hour. After washing with PBS (pH 7.4), 5 ⁇ g of 63-D7-biotin, 246-D7-biotin, and 247-B9-biotin antibodies were added respectively to stain the antigens recognized by each antibody and reacted for 12 hours at room temperature, blocked from light, 4°C.
- blocking solution 10% horse serum, 0.1% BSA, PBS, pH 7.4
- Example 3 Analysis of 63-D7 binding to various cells using a flow cytometer
- various cancer cells including embryonic cancer (NT2/D1), metastatic liver cancer (SNU387, SNU449), lung cancer (NCI-H460, NCI-H1703, NCI-H23), colon cancer (Colo205, HCT116), thyroid cancer (SNU790), and pancreatic cancer (BxPC3), as well as human embryonic kidney cells (HEK293FT), bone marrow-derived mesenchymal stem cells (BM-MSC), human dermal fibroblasts (HDF), and human peripheral blood (PB) cells.
- HEK293FT human embryonic kidney cells
- BM-MSC bone marrow-derived mesenchymal stem cells
- HDF human dermal fibroblasts
- PB peripheral blood
- PBMCs primary mononuclear cells
- MRC5 normal lung fetal fibroblasts
- DPCs dental pulp cells
- hepatocytes normal hepatocytes. Specifically, cells were detached by treating with 0.05% trypsin, washed with phosphate-buffered saline (PBS), and filtered using a 40 ⁇ m strainer (BD Biosciences) to separate single cells. 5 ⁇ 10 5 cells were mixed with PBA (1% bovine serum albumin, 0.02% NaN 3 in PBS, Ph 7.4), and then reacted with 63-D7 antibody at 4 °C for 30 min.
- PBA 1% bovine serum albumin, 0.02% NaN 3 in PBS, Ph 7.4
- the primary antibody and corresponding anti-mouse IgG-FITC (BD Biosciences) were reacted for another 30 min at 4 °C.
- the antibody reaction to propidium iodide (PI)-negative cells was analyzed using FACS Calibur and Cell Quest software (BD sciences).
- PI propidium iodide
- the 63-D7 antibody binds to all cancer cells including hepatoma cells, but does not bind to epithelial lung cancer cells H358.
- it does not bind to normal cells such as PBMC, dental pulp cells (DPC), and MRC5, and does not bind to normal hepatocytes, demonstrating cancer cell-specific binding (Fig. 2).
- the experimental results are shown in Fig. 2 and Table 1 below.
- NT-2/D1 cancer cells that bind well were washed twice with PBS (pH 7.4), and a solution of NHS-Sulfo-LC-biotin (Pierce) dissolved in PBS (pH 8.0) was added and reacted at 4°C for 30 minutes, and then washed twice with PBS (pH 8.0).
- lysis buffer 25 mM Tris-HCl, pH 7.5, 250 mM NaCl, 5 mM EDTA, 1% Nonidet P-40, 2 ⁇ g/ml aprotinin, 100 ⁇ g/ml PMSF, 5 ⁇ g/ml leupeptin, 1 mM NaF, 1 mM Na 3 VO 4 ), centrifuged at 12,000 rpm for 40 minutes to remove nuclei, and stored at -70°C until use to prepare a protein solution.
- lysis buffer 25 mM Tris-HCl, pH 7.5, 250 mM NaCl, 5 mM EDTA, 1% Nonidet P-40, 2 ⁇ g/ml aprotinin, 100 ⁇ g/ml PMSF, 5 ⁇ g/ml leupeptin, 1 mM NaF, 1 mM Na 3 VO 4 .
- protein-G-plus agarose (Merck millipore, Darmstadt, Germany) was added to the prepared protein solution.
- 20 ⁇ l of protein-G-agarose was added to the cell extract of about 1 ⁇ 10 7 cells to remove molecules that bind nonspecifically, and the mixture was reacted at 4 °C for 2 hours.
- the molecules that bind to protein-G-plus agarose were removed through centrifugation, and the remaining supernatant was collected and used to immunoprecipitate molecules that bind to 63-D7.
- the membrane was blocked with 5% skim milk powder for 2 hours at room temperature, washed three times with PBST [phosphate buffer solution (PBS), pH 7.4 containing 0.05% Tween 20], and the Western membrane was reacted with streptavidin-HRP (Streptavidin-HRP, GE healthcare) at room temperature for 1 hour to bind to the surface antigen immunoprecipitated by 63-D7 that was biotin-labeled. After washing with PBST, the bound protein was confirmed using an ECL detection kit (GE healthcare). As shown in Fig. 3a, in NT-2/D1 cells, the 63-D7 antibody recognizes and immunoprecipitates a surface antigen labeled with biotin with a size of approximately 130 kDa (Fig. 3a).
- the polyacrylamide gel containing the protein immunoprecipitated by 63-D7 was stained with PageBlue Protein Staining Solution (Thermo Fischer Scientific) according to the supplier's protocol (Fig. 3b).
- the protein band stained at the 130 kDa position which was assumed to be the protein immunoprecipitated by 63-D7, was excised from the gel and subjected to LC-MS/MS (Liquid Chromatography with Tandem Mass Spectrometry) analysis (ProteomeTech, Seoul, Korea).
- the ProFound search engine (http:/129.85.19.192/profound_bin/WebProFound.exe) developed by Rockefeller University was used to identify proteins from the analyzed mass spectrum.
- the antigen protein recognized by 63-D7 was confirmed to be Homo sapiens Prostaglandin F2 receptor negative regulator (PTGFRN) (Fig. 3c).
- the underlined amino acids in Figure 3c actually represent the peptides identified by mass spectrometry, showing that 71 amino acids are identical to PTGFRN.
- antigen protein of monoclonal antibody 63-D7 is PTGFRN
- immunoprecipitation, Western blotting, gene knockdown by RNA interference, and flow cytometry were performed using commercially available antibodies, gene expression vectors, siRNA, and shRNA against PTGFRN.
- a mouse anti-PTGFRN monoclonal antibody ( ⁇ -PTGFRN, R&D system) was purchased and used for immunoprecipitation and Western blotting.
- a cell lysate (150 ⁇ g) was prepared from A549 cells labeled with biotin as described in Example 4, and the lysate was immunoprecipitated with the monoclonal antibody 63-D7 (5 ⁇ g) and ⁇ -PTGFRN (2.5 ⁇ g) as described above.
- the membrane was reacted with gentle shaking in Striping solution (100 mM 2ME, 2% SDS, 62.5 mM Tris-HCl, pH6.7) at 50 °C for 30 min, washed twice with the above PBST for 10 min, and blocked with 5% skim milk powder for 1 h at room temperature. After washing three times with 0.1% PBST, streptavidin-HRP (SA-HRP, 1:5000) was added and reacted at room temperature for 1 h. Then, it was washed three times with PBST, and the biotin-labeled protein was confirmed with an ECL detection kit.
- Striping solution 100 mM 2ME, 2% SDS, 62.5 mM Tris-HCl, pH6.7
- PTGFRN with a size of approximately 130 kDa identical to cell surface PTGFRN, was detected in the immunoprecipitate of 63-D7 and ⁇ -PTGFRN (Fig. 4b). These results show that 63-D7 binds to cell surface PTGFRN.
- PTGFRN gene expression vector pcDNA3.1(+)-PTGFRN-DYK (GeneScript, Piscataway, NJ, USA) was purchased, and recombinant PTGFRN tagged with a FLAG tag was overexpressed in human embryonic kidney cells HEK293FT.
- the cell lysate was immunoprecipitated and analyzed by Western blotting using mouse anti-FLAG monoclonal antibody ( ⁇ -FLAG) (Invitrogen), mouse anti-PTGFRN monoclonal antibody ( ⁇ -PTGFRN) (AbCAM), and 63-D7.
- HEK293FT human embryonic kidney cells HEK293FT
- PEI polyetherimide
- the cell lysate was immunoprecipitated with 63-D7, anti-FLAG antibody, and ⁇ -PTGFRN as described above, and the eluted protein and the negative control protein without antibody (No Ab) were separated using 10% SDS-PAGE and transferred to a nitrocellulose membrane.
- the membrane was blocked with 5% skim milk powder for 1 hour at room temperature. After washing three times with 0.1% TBST, the known mouse anti-FLAG antibody, or ⁇ -PTGFRN, 63-D7 was reacted at 4 °C for 16 hours. After washing three times with 0.1% TBST, the membrane was further reacted with anti-mouse IgG-HRP (1:10,000; Millipore) for 1 hour at room temperature.
- PTGFRN the antigen of antibody 63-D7
- two kinds of siRNA (Bioneer, Daejeon, Korea) targeting PTGFRN were purchased and transiently knocked down in hepatocellular carcinoma cell lines Huh7 and SNU449.
- siRNA-lipofectamine RNAiMAX (Invitrogen) was each diluted in TOM (Welgene, Gyeongsan, Korea) and incubated for 5 min at room temperature. Then, each siRNA dilution and RNAiMAX dilution were mixed and reacted for 20 min at room temperature. The siRNA-lipofectamine mixture was added to each cell medium and reacted for 24 hours.
- qPCR was performed using primers targeting PTGFRN and GAPDH and Power SYBR Green PCR Master Mix (applied biosystem), and GAPDH was used as a reference gene.
- the specific sequences of the primers used are shown in Table 2 below.
- the knocked-down cancer cells showed a reduction efficiency of PTGFRN mRNA of 85-88% and 71-72% in Huh7 and SNU449, respectively, compared to the negative control group (Fig. 5a).
- Fig. 5a when flow cytometry analysis was performed using cells with PTGFRN knocked-down as in Example 3, it was observed that the binding of 63-D7 antibody was reduced by 51-75% and 71-78% in Huh7 and SNU449, respectively, indicating that PTGFRN expression was successfully knocked down (Fig. 5b, Fig. 5c).
- a clonogenic survival assay was performed using the PTGFRN knockdown Huh7 cells and SNU449 cells described above.
- the PTGFRN knockdown Huh7 cells prepared through the above experiment were detached with 0.05% Trypsin-EDTA (Welgene, Gyeongsan, Korea) and neutralized with cell culture medium containing 10% fetal bovine serum (Corning).
- the cells were passed through a 40 ⁇ m strainer (SPL, Pocheon, Korea) to prepare single cells. 2.0 ⁇ 103 cells per well were plated in a 6-well plate, and the cells were allowed to disperse and attach as single cells.
- Cells inserted with the target gene were selected for one week with RPMI-1640 medium containing 10% fetal bovine serum (Corning) and 2 ⁇ g/ml puromycin (Gibco). After selection, the cells were cultured in RPMI-1640 medium containing 10% fetal bovine serum (Corning) and 100 ng/ml doxycycline (Sigma) to induce knockdown, and then cultured for 72 h. The cultured cells were detached with 0.05% Trypsin-EDTA (Welgene), neutralized with cell culture medium containing 10% fetal bovine serum (Corning), and then harvested.
- RPMI-1640 medium containing 10% fetal bovine serum (Corning) and 2 ⁇ g/ml puromycin (Gibco). After selection, the cells were cultured in RPMI-1640 medium containing 10% fetal bovine serum (Corning) and 100 ng/ml doxycycline (Sigma) to induce knockdown, and then cultured for 72 h
- PTGFRN protein expression in the harvested cells was analyzed by Western blot, and PTGFRN expression was observed to be reduced by 57-84% and 88-89% in Huh7 and SNU449, respectively (Fig. 6a).
- PTGFRN#1, shPTGFRN#2 two types of shPTGFRN
- PTGFRN#1 reduced by 90% (Fig. 6d, Fig. 6e) and shPTGFRN#2 reduced by 47% (Fig. 6f, Fig. 6g).
- transwell experiments were performed using Huh7 and SNU449 cells in which PTGFRN was knocked down with the above shPTGFRN#1 and shPTGFRN#2.
- the PTGFRN knocked down Huh7 cells prepared through the above experiment were detached with 0.05% Trypsin-EDTA (Welgene), neutralized with cell culture medium containing 10% fetal bovine serum (Corning), and then passed through a 40 ⁇ m strainer (SPL) to prepare single cells.
- Trypsin-EDTA Trypsin-EDTA
- SPL 40 ⁇ m strainer
- RPMI-1640 solution containing 250 ⁇ g/ml Matrigel was coated on transwells for 2 hours at 37°C, and the same experiment as above was performed. Invasiveness was reduced by 82-93% in PTGFRN knockdown Huh7 cells and by 57-91% in SNU449 cells (Fig. 7c, 7d).
- PTGFRN deficiency reduced motility and invasiveness in both Huh7 and SNU449 cells, and these results indicate that PTGFRN is an important cell surface molecule that promotes migration and invasiveness, which are important for metastasis in hepatocellular carcinoma cells.
- PTGFRN knockdown liver cancer cells Huh7 and SNU449 bind well to various extracellular matrices (ECM).
- ECM extracellular matrices
- 200 ⁇ g/ml Matrigel, 20 ⁇ g/ml CollagenI, 20 ⁇ g/ml CollagenIV, and 0.1% Gelatin were coated and cultured in an incubator at 37°C with 5% CO2 and 95% air for 2 hours.
- PTGFRN knockdown liver cancer cells Huh7 and SNU449 were prepared, diluted to a concentration of 5.0 X 104 cells/ml, and then dispensed onto plates that were washed twice with PBS (pH 7.4) and cultured for 20-60 minutes.
- PTGFRN knockdown SNU449 cells manufactured through the above experiment were detached with 0.05% Trypsin-EDTA (Welgene, Gyeongsan, Korea) and neutralized with cell culture medium containing 10% fetal bovine serum (Corning) and 200 ng/ml doxycycline (Sigma). Then, 2.0 X 104 cells were dispensed per well in a 12-well plate and cultured in an incubator at 37°C and 5% CO2 for one day.
- NK92 effector cells
- target cells shPTGFRN-SNU449
- a medium containing 400 U/ml IL2 (Peprotech), 10% fetal bovine serum (Corning), and 200 ng/ml doxycycline (Sigma) dispensed, and cultured for 2 days at 37°C, 5% CO2 .
- floating NK92 cells were washed and removed, and the SNU449 target cells that remained attached to the bottom and survived were stained with 0.5% (w/v) crystal violet solution.
- NK cytotoxicity (%) ⁇ 1 - (target OD value/No NK OD value) ⁇ *100
- PTGFRN acts as an immune checkpoint molecule in cancer cells to suppress apoptosis by NK cells, thereby providing cancer cells with the ability to evade the host's immunity.
- the knocked-down cells were harvested, washed twice with phosphate buffer (Ph 7.4), and lysed using RIPA lysis buffer (50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1% NP-40, 0.1% SDS, 0.5% Deoxycholate). The cells were centrifuged at 4°C, 12,000 rpm for 30 minutes, and the supernatant was mixed with 5X sample buffer to prepare a protein sample. Afterwards, they were separated on an 8-10% SDS-PAGE gel and transferred to a nitrocellulose membrane for Western blotting.
- RIPA lysis buffer 50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1% NP-40, 0.1% SDS, 0.5% Deoxycholate.
- the cells were centrifuged at 4°C, 12,000 rpm for 30 minutes, and the supernatant was mixed with 5X sample buffer to prepare a protein sample. Afterwards, they were separated on an 8-10% SDS
- Blocking was performed using 5% skim milk powder, and each target antibody, rabbit anti-PTGFRN polyclonal antibody (abcam, 97567), rabbit anti-B7-H3 polyclonal antibody (sinobiological, 11188-RP02), rabbit anti-FAK polyclonal antibody (Cell Signaling Technology, 3285), rabbit anti-p-FAK polyclonal antibody (CST, 3283), rabbit anti-Src monoclonal antibody (CST, 2109), rabbit anti-p-Src monoclonal antibody (CST, 6943), rabbit anti-GAPDH polyclonal antibody (CSB, PA00025A0Rb), was mixed in 5% BSA and reacted at 4°C for 16 hours.
- rabbit anti-PTGFRN polyclonal antibody (abcam, 97567)
- rabbit anti-B7-H3 polyclonal antibody sinobiological, 11188-RP02
- rabbit anti-FAK polyclonal antibody Cell Signaling Technology, 3285
- PTGFRN regulates the migration and invasiveness of cancer cells through the FAK signaling pathway.
- PTGFRN expressed in the cell membrane contributes to forming clusters between cells
- PTGFRN expressed in circulating cancer cells will help in the growth or survival and immune evasion of cancer cells.
- 2.5 X 10 5 cells were seeded per well in a 6-well plate, and the next day, 12 ⁇ g of PEI, pcDNA3.1(+)PTGFRN-FLAG vector, and pcDNA3.1(+)-PTGFRN-myc vector were diluted together in TOM (Welgene) to 4 ⁇ g each, and reacted at room temperature for 5 minutes.
- the diluted PEI solution and the diluted vector solution were mixed and reacted at room temperature for 20 minutes, and then treated together in one HEK293FT cell and cultured for 24 hours in an incubator with 37°C and 5% CO 2 .
- pcDNA3.1(+)PTGFRN-FLAG vector and pcDNA3.1(+)-PTGFRN-myc vector were prepared respectively and reacted separately in two HEK293FT cell wells, and after 24 hours, the culture medium was replaced with new one, cultured for another 48 hours, and cell lysates were prepared by treating cell lysis buffer in the same manner as in Example 4.
- each cell lysate was prepared using BCA (Thermo), and each lysate was immunoprecipitated using mouse anti-FLAG monoclonal antibody, rabbit anti-myc tag monoclonal antibody, and mouse 63-D7 antibody, and then analyzed by Western blotting with anti-FLAG monoclonal antibody and rabbit anti-myc tag monoclonal antibody.
- SNU449 cell lysates were prepared as in Example 4, and then immunoprecipitated using mouse anti-PTGFRN monoclonal antibody, mouse 63-D7 monoclonal antibody, mouse anti-SLC3A2 monoclonal antibody, and mouse anti-B7-H3 monoclonal antibody.
- Western blotting was performed using mouse anti-PTGFRN monoclonal antibody ( ⁇ -PTGFRN). As a result, it was confirmed that PTGFRN binds to SLC3A2 (CD98hc) and B7-H3 (CD276) (Fig. 11c).
- B7-H3 is known to be an immune checkpoint regulator in cancer cells
- HEK293FT cells were seeded at 2.5 X 10 5 cells per well in a 6-well plate.
- 12 ⁇ g of PEI, PTGFRN vector with myc tag (pcDNA3.1(+)PTGFRN-myc) and PTGFRN vector with FLAG tag (pcDNA3.1(+)-B7-H3-FLAG) were diluted together with 4 ⁇ g each in TOM (Welgene, Gyeongsan, Korea) and reacted at room temperature for 5 minutes.
- the diluted PEI solution and the diluted vector solution were mixed and reacted at room temperature for 20 minutes, treated to transfect HEK293FT cells, and cultured in an incubator with 5% CO 2 at 37°C for 24 hours. After 24 hours, the culture medium was replaced with new medium, and after culturing for another 48 hours, the cell lysate was prepared as in Example 4. Using BCA (Thermo Fischer Scientific), 50 ⁇ g of each cell lysate was prepared, and then immunoprecipitated with anti-FLAG, anti-B7-H3, anti-myc, and 63-D7. The immunoprecipitates were detected with anti-myc and anti-FLAG antibodies.
- BCA Thermo Fischer Scientific
- PTGFRN was detected in the immunoprecipitate by anti-FLAG antibody, and B7-H3 was detected in the immunoprecipitate by anti-myc antibody. Therefore, it can be seen that PTGFRN and B7-H3 physically bind and interact with each other (Fig. 11d). These results suggest that PTGFRN is involved in immune regulation through B7-H3, which is known as an immune checkpoint regulatory molecule.
- Example 7 Analysis of adhesion, cancer stemness, and clonogenicity of 63-D7 positive liver cancer cells
- Huh7 and HepG2 cells were sorted into 63-D7 positive and negative using magnetic beads with 63-D7 antibody.
- 63-D7-biotin antibody was separated using Neutravidin-conjugated magnetic beads (Thermo Fishcer Scientific) according to the manufacturer's protocol. Cells attached to the antibody were separated as 63-D7(+), and non-attached cells were prepared as 63-D(-). Next, 12-well plates were coated with Matrigel and blocked in serum-free DMEM/F12 medium containing 5% BSA at 37°C for at least 1 hour. 63-D7(+), 63-D7(-) hepatoma cells were seeded at 105 cells per well and reacted at 37°C for 6 hours. To remove non-attached cells, the plates were washed with PBS (pH 7.4).
- Adherent cells were stained with 0.5% crystal violet (Sigma-Aldrich) in 2% ethanol for 30 min at room temperature, and the stained cells were dissolved in 0.1% SDS and measured for absorbance at 570 nm.
- 63-D7(+) Huh7 cells showed a 43% increase in adhesion compared to 63-D7(-) Huh7 cells
- 63-D7(+) HepG2 cells showed a 245% increase in adhesion compared to 63-D7(-) HepG2 cells, suggesting that 63-D7-positive hepatoma cells have significantly better adhesion to the extracellular matrix than do negative hepatoma cells (Fig. 12a, Fig. 12b).
- tumorspheres that induce cancer stemness were cultured.
- attached and growing Huh7 liver cancer cells were detached with trypsin-EDTA (Welgene, Daegu, Korea), filtered through a 40 ⁇ m strainer, and centrifuged at 1500 rpm for 3 minutes at room temperature.
- the cells were seeded at 2.1 x 10 3 cells/cm 2 on very low adherent plates (Corning, SEOUL, KOREA) and cultured in DMEM/F12 medium (Corning) supplemented with 20 ng/ml fibroblast growth factor 2 (FGF2, R&D systems, Seoul, Korea), 20 ng/ml epidermal growth factor (EGF, PeproTech, Seoul, Korea), and 1 x B27 supplement (Life Technologies). The medium was changed every 3 days, and the culture was performed for at least 9 days.
- the expression of CD133, CD44, and EpCAM known as representative liver cancer stem cell markers, and PTGFRN, a 63-D7 antigen, was analyzed by flow cytometry in the same manner as in Example 3.
- the cancer stem cell-positive markers CD133, CD44, and EpCAM increased by about 33%, 43%, and 59%, respectively, compared to the adherent cells, and at this time, the binding of 63-D7 also increased by 61% (Fig. 12c, Fig. 12d).
- Example 6-2 To further analyze whether 63-D7-positive liver cancer cells show liver cancer stem cell properties, a clonogenic survival experiment showing cancer stem cell properties was performed as in Example 6-2 using liver cancer cells sorted with 63-D7 in the same manner as above. As a result, it was observed that clonogenicity increased by 492% in Huh7 (Fig. 12e, Fig. 12f) and by 56% in HepG2 (Fig. 12g, Fig. 12h). These results suggest that 63-D7-positive liver cancer cells show remarkable cancer stem cell properties compared to negative liver cancer cells and have cancer stem cell properties with excellent clonogenic survival ability.
- Example 8 Internalization analysis after 63-D7 treatment in liver and pancreatic cancer cells
- Cells were detached with 0.05% Trypsin-EDTA (Welgene) and neutralized with cell culture medium containing 10% fetal bovine serum (FBS; VWR, PA, USA), and then passed through a 40 ⁇ m strainer to prepare single cells. Approximately 1 ⁇ 10 5 cells per ml of each single cell were mixed with PBA (1% bovine serum albumin, 0.02% NaN 3 in PBS) and reacted with antibody 63-D7 at 4 °C for 30 min. To induce antibody internalization, cells were washed once with PBA, and then cells were suspended in 100 ⁇ l of culture medium and reacted for 30 min at 37 °C to allow cell membrane internalization.
- PBA 1% bovine serum albumin, 0.02% NaN 3 in PBS
- 63-D7 antibody bound to the surface of cancer cells at 4°C is internalized into the cells by cell activity at 37°C, thereby reducing binding on the surface, suggesting that 63-D7 is an antibody that can be applied to the development of antibody therapeutics using ADC.
- Example 8 it was confirmed that the 63-D7 antibody having a mouse IgG1 constant region bound to the surface of liver cancer cells and was internalized into the cells at a temperature of 37°C. Based on the above characteristics, it was assumed that when an anticancer drug is conjugated to the 63-D7 antibody, the drug delivered via the 63-D7 antibody would exhibit cytotoxicity in cancer cells, and an experiment was performed to confirm this.
- ⁇ -mFc-CL-DMDM AM-102DD, Moradec, USA
- ⁇ -mFc-CL-MMAF AM-102AF, Moradec
- Huh7 cells were dispensed into each well of a 96-well plate (SPL) using RPMI-1640 (Biowest, France) medium containing 10% FBS, and then cultured for 24 hours. The next day, the existing culture medium was removed, and 63-D7 antibody or mouse IgG isotype control (Invitrogen, 31903) diluted in the culture medium at various concentrations was dispensed into each well at 100 nM, 10 nM, 1 nM, 0.1 nM, and 0.01 nM, and the antibody was allowed to bind to the cell surface for 10 minutes at 37°C.
- the drug-conjugated secondary antibody at a concentration of 12.7 nM was diluted in the culture medium, the primary antibody was removed, and the antibody was dispensed into each well. After culturing the cells for 48 hours under 5% CO2 conditions at 37°C, Cell Counting Kit-8 (CCK-8, K1018, APExBIO, USA) was used to determine the cell viability. 10 ⁇ l of CCK-8 was added to each well, reacted for 3 hours, and the absorbance at OD450nm was measured using a plate reader.
- 63-D7 antibody is internalized into liver cancer cells, and based on this characteristic, it can exhibit anticancer effects by effectively delivering drugs into liver cancer cells expressing PTGFRN.
- Example 10 Analysis of 63-D7 antigen in blood cancer cells from liver cancer patients
- PTGFRN promotes survival, migration, invasion, cell adhesion, and immune evasion of hepatocellular carcinoma (HCC) cells, which are biological characteristics of circulating cancer cells that are important for metastasis (Figs. 5 to 9). Therefore, we hypothesized that PTGFRN could serve as a surface marker to detect circulating cancer cells in HCC patients, and indeed, we confirmed this possibility in preliminary experiments to detect circulating cancer cells (Figs. 1b and 1c).
- peripheral blood mononuclear cells PBMCs
- PBMCs peripheral blood mononuclear cells
- Enrichment of circulating cancer cells by depletion of CD45-positive cells from these leukocyte lineages was performed using a Human CD45 depletion kit (EasySep®, Stem Cells Technologies, Vancouver, BC, Canada) according to the provided protocol.
- EasySep® Stem Cells Technologies, Vancouver, BC, Canada
- anti-CD45 antibody was added to the cell suspension and reacted at room temperature for 15 minutes, then dextran-coated magnetic nanoparticles were reacted with the cells at room temperature for 10 minutes, and the cell suspension was placed in an EasySep® big easy magnet (StemCell Technologies, Vancouver, Canada) at room temperature for 10 minutes.
- the unbound cell fraction was then transferred to a clean tube and collected, and the collected cells were centrifuged at 3,560 ⁇ g for 5 minutes, resuspended in 400 ⁇ l of RPMI1640 medium, divided into eight fractions, and finally seeded onto glass slides coated with 0.1 ⁇ g/mL of poly-L-lysine.
- the cells were reacted at room temperature for 2-4 hours to induce spontaneous binding of the cells to the glass slides. Unbound cells were washed with PBS (pH 7.4) before fixation.
- the bound cells were fixed in 3.7% paraformaldehyde (PFA) and stored in the refrigerator before analysis by confocal microscopy.
- PFA paraformaldehyde
- CD45-depleted cells were fixed with 3.7% PFA, blocked with 10% normal horse serum, and then reacted with Dylight 649-conjugated anti-mouse IgG (Vector laboratories) and Dylight 488-conjugated 63-D7. After washing with PBS, nuclei were stained with DAPI (4,6-diamidino-2-phenylindole). Fluorescent signals were detected with a Leica TCS SP5 confocal microscope (Leica Microsystems, Seoul, Korea). As a result, all 63-D7-positive cells were confirmed to be CD45-negative (lower panel of Fig. 15c, Table 3). The expression profiles of EMT markers in 63-D7+ nucleated CTCs of HCC patients by double immunofluorescence staining are specifically shown in Table 3 below.
- HCC liver cancer patients
- HCC liver cancer patients
- Normal normal people
- hepatitis patients are shown in Table 4 below.
- HCC M 69 3.75 11 21 14 1.90 2 HCC F 63 1.13 10 15 11 1.50 3 HCC F 62 1.05 9.4 28 22 2.98 4 HCC M 65 3.2 10 61 46 6.10 5 HCC M 64 2.35 10.6 44 31 4.15 6 HCC M 54 2.43 10 3 2 0.30 7 HCC M 68 3.54 10 38 29 3.80 8 HCC M 64 2 10.2 14 10 1.37 9 HCC M 73 1.68 10 33 25 3.30 10 HCC M 54 2.08 8 15 14 1.88 11 HCC F 60 1.48 10.1 22 16 2.18 12 HCC F 62 2.23 10.4 26 19 2.50 13 HCC F 45 3.63 9.4 24 19 2.55 14 HCC M 60 1.87 10.1 6 5 0.59 15 HCC M 72 1.5 10.2 25 18 2.45 16 HCC M 45 1.45 8 20 19 2.50 17 HCC M 68 1.375 9 28
- 63-D7-positive circulating cancer cells were heterogeneous, ranging from 18 ⁇ m to 40 ⁇ m, and they had hyperchromatic nuclei and a high nuclear-to-cytoplasmic ratio (Figs. 16a to 16c).
- 63-D7-positive circulating cancer cells were detected in all HCC patients, and 92 of 95 patients (approximately 97%) showed a higher cell number than that of normal adults (Fig. 17, Table 4) (p ⁇ 0.0001).
- the number of cells isolated from HCC patients ranged from 0.1 to 50.72 per ml, and the average was 6.13 per ml (Table 4). Since 63-D7-positive circulating cancer cells were not found in patients with chronic hepatitis and cirrhosis (Table 4), it shows that they are circulating cancer cells caused by cancer development, not inflammatory cells caused by hepatitis, etc.
- CD45-depleted cells were fixed with 3.7% PFA, blocked with 10% normal horse serum, and then reacted with anti-MVP (Aviva systems, San Diego, CA, USA), anti-CD44 (BD bioscience, Seoul, Korea), anti-CD90 (BD bioscience), Alexa 555-conjugated anti-EpCAM (BD Biosciences), Alexa 555-conjugated anti-E-cadherin (Cell Signaling Technology, Beverly, MA, USA), and anti-HSA (Novus, Litteleton, CO, USA), followed by additional reaction with Dylight 650-conjugated anti-rabbit IgG (Thermo Fischer Scientific), and finally Dylight 488-conjugated 63-D7, and stained as in Example 10-1.
- anti-MVP Aviva systems, San Diego, CA, USA
- anti-CD44 BD bioscience, Seoul, Korea
- anti-CD90 BD bioscience
- Alexa 555-conjugated anti-EpCAM BD Biosciences
- CD45-depleted cells were fixed and permeabilized with 0.1% Triton X-100 before blocking and reacted with anti-vimentin (Santa Cruz Biotechnology), anti-Twist1 (AbCAM, Cambridge, UK), anti-ZEB1 (AbCAM), or phycoerythrin-conjugated anti-panCK antibodies (BD biosciences, San Jose, CA, USA).
- the cells were incubated with Dylight 650-conjugated anti-rabbit IgG (Thermo Fischer Scientific) depending on the conjugation status of the primary antibody and finally reacted with Dylight 488-conjugated 63-D7, and the fluorescence signals were detected and analyzed by a Leica TCS SP5 confocal microscope (Leica Microsystems).
- MVP cell surface major vault protein
- 63-D7/HSA staining approximately 25% (50/203) of the 63-D7-positive circulating cancer cells were HSA-positive in both primary and secondary HCC patients (Fig. 16a, Table 3).
- 63-D7/E-cadherin staining all circulating cancer cells were E-cadherin-negative (0/18) in all primary HCC patients (Fig. 16b, Table 3).
- 63-D7/panCK (pan-cytokeratin) staining all circulating cancer cells were panCK-negative in both primary (0/104) and secondary HCC patients (0/24) (Fig. 16a, Table 3).
- 63-D7/EpCAM staining approximately 15% (67/442) of 63-D7-positive cells were EpCAM-positive in both primary and secondary HCC patients (Fig. 16b, Table 3).
- epithelial markers such as PanCK or E-cadherin are not expressed in PTGFRN-positive circulating cancer cells recognized by 63-D7, regardless of whether they are primary or secondary metastatic/recurrent cancers. This suggests that 63-D7-positive circulating cancer cells are at least not epithelial circulating cancer cells.
- 63-D7-positive circulating cancer cells exhibit a mesenchymal phenotype
- representative mesenchymal markers Vimentin, Twist, and ZEB1 were observed in primary HCC patients.
- Approximately 24% (43/180) of 63-D7-positive circulating cancer cells were vimentin-positive (Fig. 16c, Table 3).
- Twist (3.2%, 6/189) and ZEB1 (5%, 3/60) positivity were also observed in small fractions in 63-D7-positive circulating cancer cells (Fig. 16c, Table 3).
- 63-D7-positive circulating cancer cells exhibit a significantly predominant mesenchymal phenotype when comparing the proportions of epithelial and mesenchymal phenotypes, it can be seen that in most cases, they are a partial intermediate-stage cell population in which the distinction between epithelial and mesenchymal phenotypes is unclear.
- triple fluorescence staining for 63-D7/EpCAM/vimentin was performed using CD45-depleted circulating cancer cells, using the epithelial representative marker EpCAM and the mesenchymal representative marker Vimentin (Fig. 18a, Table 6). The results of the triple fluorescence staining are specifically shown in Table 6 below.
- CD45-depleted cells were fixed and permeabilized with 0.1% Triton X-100 before blocking and reacted first with anti-Vimentin antibody (Santa Cruz Biotechnology). Cells were then reacted with Dylight 650-conjugated anti-rabbit IgG (Thermo Scientific), Alexa 555-conjugated anti-EpCAM (Cell Signaling Technology), and Dylight 488-conjugated 63-D7, washing cells four times with PBS containing Ca 2+ and Mg 2+ between each step. Nuclei were stained with DAPI, and fluorescence signals were detected by confocal microscopy (Leica Microsystems).
- the 63-D7 + EpCAM + Vimentin + triple-positive circulating cancer cells were 9.1% (47/518) in primary HCC patients, whereas 0.3% (3/933) in secondary HCC patients.
- MVP-positive circulating cancer cells 51.3% were MVP-single positive in MVP/EpCAM/Vimentin triple staining (Lee, Joh et al. 2017), so we performed MVP/EpCAM/Vimentin triple staining using the same method.
- MVP single-positive circulating cancer cells were classified into primary HCC and secondary HCC, MVP single-positive circulating cancer cells were 53.5% in primary HCC patients and 50.3% in secondary HCC patients (Table 6).
- MVP single-positive circulating cancer cells were found in all secondary HCC patients (9/9 patients, 100%) (Table 6).
- MVP 50.3%) or 63-D7 (72.3%) single-positive circulating cancer cells were EpCAM-negative and Vimentin-negative and were predominantly present in all secondary HCC patients. Therefore, PTGFRN recognized by 63-D7 is a novel marker representing most circulating cancer cells in patients with HCC that are neither epithelial nor mesenchymal, and it can be seen as an excellent diagnostic marker observed in all patients, especially in patients with secondary metastatic/recurrent HCC.
- the cells were then reacted with Dylight 650-conjugated anti-rabbit B7-H3 (Sinobiological, Beijing, China) and Dylight 488-conjugated 63-D7 antibody. Between each step, cells were washed four times with PBS containing Ca 2+ and Mg 2+ . Nuclei were stained with 4,6-diamidino-2-phenylindole (DAPI) and fluorescence signals were detected with a Leica TCS SP5 confocal microscope. The expression profiles of EMT and immune checkpoint markers in nucleated CTCs from secondary HCC patients by triple immunofluorescence staining are shown in Table 7 below.
- EMT provides cancer cells with metastatic potential, immunosuppressive potential, and cancer stemness.
- EMT can induce the expression of immunosuppressive molecules such as PD-L1 and CD47 in cancer stem cells and circulating cancer cells.
- immunosuppressive molecules such as PD-L1 and CD47 in cancer stem cells and circulating cancer cells.
- Approximately 53% of 63-D7-positive circulating cancer cells were EMT phenotype circulating cancer cells in primary HCC patients, but the proportion of EMT phenotype circulating cancer cells decreased to 27.6% in secondary HCC patients (Table 6). Instead, most 63-D7-positive circulating cancer cells (72.3%) were partial/intermediate phenotype circulating cancer cells in secondary HCC patients (Table 6).
- TGF ⁇ /TGF ⁇ R1 signaling exerts multiple immunosuppressive effects in CSCs.
- TGF ⁇ is also required for the suppression of NK cell function.
- TGF ⁇ /TGF ⁇ R1 signaling provides the high metastasis-initiating ability of breast CSCs.
- TGF ⁇ 1 signaling also promotes T cell-mediated tumor evasion in colorectal cancer through increased B7-H3 expression. Therefore, although the number of EMT phenotype CSCs was reduced in PTGFRN-positive CSCs from secondary HCC patients, we investigated to what extent TGF ⁇ R1 expression was positive in 63-D7-positive CSCs.
- circulating cancer cells were simultaneously triple-stained with anti-ULBP1/63-D7/anti-B7-H3 antibodies or anti-MICA/B/63-D7/anti-B7-H3 antibodies (Fig. 20a, Fig. 20b, Table 7).
- ULBP1 and MICA/B are known as ligands for the activating receptor NKG2D of NK and CD8 T cells, and are molecules that promote immunity.
- ULBP1/63-D7/B7-H3 staining 4.1% (9/222) of total PTGFRN-positive circulating tumor cells were ULBP1-positive in secondary HCC patients, whereas 50.5% (112/222) of total PTGFRN-positive circulating tumor cells were B7-H3-positive.
- MICA/B/63-D7/B7-H3 staining all patients (18/18) had MICA/B-positive circulating tumor cells.
- PD-L1 and PD-L2 are well-known ligands for the immunosuppressive receptor PD-1 on NK and CD8 T cells. Therefore, circulating cancer cells were simultaneously triple-stained with anti-PD-L1/63-D7/anti-B7-H3 antibodies or anti-PD-L2/63-D7/anti-B7-H3 antibodies, respectively (Fig. 21a, Fig. 21b, Table 7).
- PD-L1/63-D7/B7-H3 staining 37.8% (197/521) of the total PTGFRN-positive circulating cancer cells in patients with secondary hepatocellular carcinoma were PD-L1 positive, and 55.7% (290/521) of the total PTGFRN were B7-H3 positive.
- PD-L2/63-D7/B7-H3 staining 17.5% (69/395) of the total PTGFRN-positive circulating cancer cells in patients with secondary hepatocellular carcinoma were PD-L2 positive, and 56.2% (222/395) of the total PTGFRN were B7-H3 positive (Fig. 21a, Fig. 21b, Table 7). Meanwhile, all patients (22/22) had PD-L1 or PD-L2 positive circulating cancer cells.
- CD47 is a “don’t eat me” signal for macrophages and inhibits phagocytic innate immune surveillance.
- CD47/63-D7/B7-H3 staining 38.1% (115/302) of the total PTGFRN-positive CBCTs in patients with secondary HCC were CD47-positive, and 51.7% (156/302) were B7-H3-positive (Fig. 22 , Table 7 ). And all patients (14/14) had CD47-positive CBCTs.
- immunostimulatory receptors such as ULBP1 (4.1%) and MICA/B (34.7%) were detected in PTGFRN-positive circulating cancer cells
- immune checkpoint regulatory molecules such as TFG ⁇ R1 (51.8%), PD-L1 (37.8%), PD-L2 (17.5%), CD47 (38.1%), and B7-H3 (50.5–65.3%) were more widely detected in PTGFRN-positive circulating cancer cells from secondary HCC patients.
- B7-H3 was predominantly expressed in all PTGFRN-positive circulating cancer cells (average 57.6%).
- PTGFRN is responsible for immune evasion of circulating cancer cells in secondary HCC patients, and its expression is closely associated with immunosuppressive receptors and plays a role in immune evasion.
- PTGFRN recognized by 63-D7 antibody in circulating cancer cells may serve as an excellent diagnostic marker for indicating the stage or progression of liver cancer.
- Vigorously growing hybridoma 63-D7 cells (5 ⁇ 10 6 cells) were harvested by centrifugation, washed twice with cold PBS, and total RNA was extracted using RNAiso plus reagent (TaKaRa, Otsu, Japan) according to the manufacturer's protocol. The A260 of total RNA was measured using Nanodrop to quantify the amount of RNA.
- 1 unit of DNase I (TaKaRa) per 1 ⁇ g of total RNA was added and reacted at 37 °C for 30 minutes to remove residual DNA.
- 1 ⁇ l of 50 mM EDTA was added and reacted at 65 °C for 10 minutes to inactivate DNase I and denature total RNA.
- a reverse transcription polymerase chain reaction mixture was prepared using total RNA and Prime Script RT reagent Kit (TaKaRa), and cDNA was synthesized according to the manufacturer's protocol.
- a 5'-ggt gtc gac GGA TAC AGT TGG TGC AGC ATC-3' (SEQ ID NO: 23) oligonucleotide primer corresponding to the kappa chain constant region and a 5'MK 5'-cgg aag ctt GAY ATT GTG MTS ACM CAR WCT MCA-3' (SEQ ID NO: 24) oligonucleotide primer corresponding to the N-terminus of the kappa chain variable region were used.
- an EcoRI restriction site was provided at the end of the 5'-primer for the heavy chain and a SalI restriction site was provided at the end of the 3'-primer.
- a HindIII restriction site was provided at the end of the 5'-primer and a SalI restriction site was provided at the end of the 3'-primer.
- amplified DNA was obtained at a position corresponding to approximately 400 bp, which is estimated to be a DNA fragment corresponding to the heavy chain variable region, and approximately 390 bp, which is estimated to be a DNA fragment corresponding to the light chain variable region.
- the heavy chain of the polymerase chain reaction product was treated with EcoRI and SalI, and the light chain was treated with HindIII and SalI, and then developed on a 1.0% (w/v) agarose gel.
- DNA corresponding to approximately 400 bp and 390 bp was isolated using FavorPrep GEL PCR Purification Kit (Favorgen, Pingtung, Taiwan).
- the vector pBluescript KS+ for cloning the heavy chain gene was treated with EcoRI and SalI
- the vector pBluescript KS+ for cloning the light chain gene was treated with HindIII and SalI, and then isolated using FavorPrep GEL PCR Purification Kit.
- the heavy chain corresponded to subgroup IIB (Fig. 23) and the light chain corresponded to subgroup V (Fig. 24).
- the antigen binding sites, CDR 1, 2, and 3 are indicated in bold for each sequence.
- the results of comparing the amino acid sequences of the antibodies using BLAST confirmed that 63-D7 is a new antibody that was not previously known.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Biomedical Technology (AREA)
- Hematology (AREA)
- Urology & Nephrology (AREA)
- Biochemistry (AREA)
- Microbiology (AREA)
- Organic Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Biotechnology (AREA)
- Epidemiology (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Analytical Chemistry (AREA)
- Physics & Mathematics (AREA)
- Food Science & Technology (AREA)
- Cell Biology (AREA)
- Oncology (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Genetics & Genomics (AREA)
- Biophysics (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Hospice & Palliative Care (AREA)
- Mycology (AREA)
- Peptides Or Proteins (AREA)
Abstract
Description
본 발명은 혈중 암세포 수준 판단, 암 치료 또는 예방, 암 전이 예후 예측 및 항암제 스크리닝을 위한 항 PTGFRN 단일클론항체에 관한 것이다.The present invention relates to an anti-PTGFRN monoclonal antibody for determining the level of blood cancer cells, treating or preventing cancer, predicting the prognosis of cancer metastasis, and screening for anticancer drugs.
간세포암(Hepatocellular carcinoma, HCC)은 간암의 90% 이상을 차지하며, 암으로 인한 사망률 가운데 세번째를 차지한다. 간세포암의 주요 치료법은 외과적 절제, 초기 간세포암에만 적용되는 국소 절제 또는 간 이식 등이 있다. 그러나 초기 간세포암 진단 당시 대부분의 환자는 이미 진행된 병기로 발견되어 경동맥 화학색전술과 같은 완화적 치료, tyrosine kinase inhibitor를 이용한 전신 치료, 선택적인 내부 방사선 치료를 통해서만 생존이 유한하게 연장된다. 하지만 간세포암은 재발률이 유난히 높아 치료 효과가 만족스럽지 못하다. 현재 간세포암의 진단 및 모니터링은 주로 혈청 바이오마커 검출, 병리학적 검사 및 영상 분석에 의존하지만 일반적인 혈청 마커로 쓰는 AFP는 58-66% 정도로 진단 성능이 좋지 않으며, AFP를 또 다른 마커인 des-gamma-carboxy-prothrombin(DCP)와 조합하여 진단할 때도 민감도는 82% 정도에 불과하다. 그리고 영상 및 병리 검사는 진단 정확도 및 민감도에 한계가 있어 간세포암의 진단 및 모니터링을 위한 더 나은 접근 방식이 필요하다.Hepatocellular carcinoma (HCC) accounts for more than 90% of liver cancers and is the third leading cause of cancer-related mortality. The main treatments for HCC include surgical resection, local resection for early-stage HCC, or liver transplantation. However, most patients with early-stage HCC are already diagnosed with advanced disease, and survival is limited to palliative treatment such as transarterial chemoembolization, systemic treatment with tyrosine kinase inhibitors, and selective internal radiotherapy. However, HCC has an exceptionally high recurrence rate, and the treatment effect is not satisfactory. Currently, the diagnosis and monitoring of HCC mainly depend on serum biomarker detection, pathological examination, and image analysis. However, the diagnostic performance of AFP, a common serum marker, is poor at around 58-66%, and the sensitivity of AFP in combination with another marker, des-gamma-carboxy-prothrombin (DCP), is only around 82% when diagnosed. Moreover, imaging and pathology tests have limitations in diagnostic accuracy and sensitivity, so better approaches are needed for the diagnosis and monitoring of HCC.
혈중 암세포(Circulating tumor cell, CTC)란 암환자의 말초 혈액에서 발견되는 암세포로, 혈액을 통해 이동하는 암세포를 말한다. 전이는 대다수의 암 관련 사망의 직접적인 원인이 될 수 있고, 혈중 암세포는 이런 전이현상에 씨앗이 되는 것으로 암을 퍼트리는 세포이다. 이들은 homo-, hetero-클러스터를 이루어 면역세포로부터 회피하고 내외부의 억제 신호를 회피하여 전이를 하며, 이들의 발현 정도는 암의 생존율과 연관이 있다. 말초 혈액에서 혈중 암세포의 수가 증가하는 것은 암 환자에서 전이가 진행되는 것을 의미하므로, 혈중 암세포를 암 전이 진단 마커로 사용할 수 있는 방법이 개발되었다. 또한 혈중 암세포는 암 환자의 좋지 않은 예후와 관련되어 있다. 혈중 암세포의 검출은 조직검사와 달리 액상 조직검사(Liquid biopsy)를 통해 간단한 방법으로 암의 진단이 가능하므로, 암 진단의 새로운 대안으로 제시되고 있다. 그리고 동물 모델을 통해 혈중 암세포는 암 초기부터 원발암에서 혈액을 통해 전파된다는 것이 알려졌고, 간세포암에서도 전이가 관찰되지 않는 초기 단계부터 혈중 암세포는 검출된다. 따라서 혈중 암세포 진단을 통한 간세포암의 조기발견 방법 및 관리 방법이 제안되었다. 하지만 혈중 암세포는 그 수가 매우 적어 실제로 혈액에서 효율적으로 분리해내기 쉽지 않다. 또한 다양한 암 종에서 각기 다른 혈중 암세포가 나와서 이동할 것으로 추정되므로, 크기가 다양하고 수가 매우 적은 혈중 암세포를 정확히 검출 및 분리해내기는 쉽지 않다. 현재까지 혈중 암세포들은 EpCAM을 이용한 Cell search system, 세포 크기 및 물리적 특성에 기인한 microfluidics system, 유세포 분석기를 통해 분리하여 연구하고 있지만, 혈중 암세포들의 다양성으로 인해 백혈구와 비슷한 크기의 혈중 암세포들도 존재하고, 물리적인 특성으로 분리할 수 없는 세포들도 존재하므로, 간세포암의 특성을 반영하는 표면 바이오 마커를 이용한 혈중 암세포 분리가 중요하다.Circulating tumor cells (CTCs) are cancer cells found in the peripheral blood of cancer patients, and they are cancer cells that move through the blood. Metastasis can be the direct cause of most cancer-related deaths, and circulating tumor cells are the seeds of this metastasis phenomenon and are cells that spread cancer. They form homo- and hetero-clusters to evade immune cells and internal and external inhibitory signals to metastasize, and their expression level is related to the survival rate of cancer. Since an increase in the number of circulating tumor cells in the peripheral blood indicates that metastasis is progressing in cancer patients, a method has been developed to use circulating tumor cells as a cancer metastasis diagnostic marker. In addition, circulating tumor cells are associated with a poor prognosis for cancer patients. Unlike tissue examinations, detection of circulating tumor cells is a simple method for diagnosing cancer through liquid biopsy, and is therefore being presented as a new alternative for cancer diagnosis. And through animal models, it has been known that blood cancer cells are transmitted from the primary cancer through blood from the early stage of cancer, and blood cancer cells are detected from the early stage when metastasis is not observed in hepatocellular carcinoma. Therefore, early detection and management methods of hepatocellular carcinoma through blood cancer cell diagnosis have been proposed. However, blood cancer cells are very small in number, so it is not easy to effectively separate them from the blood. In addition, it is estimated that different blood cancer cells will come out and move from various types of cancer, so it is not easy to accurately detect and separate blood cancer cells that are of various sizes and very small in number. To date, blood cancer cells have been separated and studied using a cell search system using EpCAM, a microfluidics system based on cell size and physical characteristics, and a flow cytometer. However, due to the diversity of blood cancer cells, there are blood cancer cells similar in size to leukocytes, and there are cells that cannot be separated due to physical characteristics, so it is important to separate blood cancer cells using surface biomarkers that reflect the characteristics of hepatocellular carcinoma.
다양한 암의 혈중 암세포를 분리할 수 있는 마커로 현재 미국 FDA 승인을 받은 마커로는 상피성 세포 유래(epithelial origin)인 Epithelial Cell Adhesion Molecule(EpCAM)이 있다. 그러나 간은 상피성 유래 기관임에도 대다수의 간세포나 간세포암이 EpCAM 음성이며, 실제로 면역조직화학적 방법으로 염색 시 간세포암 조직의 15-30% 정도만이 EpCAM 양성으로 나타난다. 따라서 간세포암에서 EpCAM과 다른 마커로, 아시알로페투인(asialofetuin)을 사용하여 여기에 결합하는 아시알로글리코프로테인 수용체(asialoglycoprotein receptor, ASGPR) 양성 혈중 암세포를 검출하는 방법이 사용되었는데, 간세포암 환자 85명에서 5 ml 혈액당 1개 이상 혈중 암세포가 검출되는 비율은 69명으로 약 81% 정도였다. 하지만 ASGPR로 검출된 혈중 암세포 수와 간세포암 전이 사이에 병기와 관련된 뚜렷한 연관성은 없었다. 한편, EpCAM이 간세포암의 전이를 유도하거나 치료의 재발을 유도하는 간암줄기세포의 마커라는 연구 결과를 통해, 줄기세포 특성을 나타내는 EpCAM 양성 혈중 암세포에 대한 관심이 간세포암 분야에서 대두되었다.Among markers that can isolate circulating cancer cells of various cancers, the one currently approved by the US FDA is the Epithelial Cell Adhesion Molecule (EpCAM), which is of epithelial origin. However, although the liver is an epithelial-derived organ, the majority of hepatocytes and hepatocellular cancer are EpCAM-negative, and in fact, only about 15-30% of hepatocellular cancer tissues are EpCAM-positive when stained with an immunohistochemical method. Therefore, a method of detecting circulating cancer cells positive for the asialoglycoprotein receptor (ASGPR) that binds to asialofetuin, a marker other than EpCAM, was used in hepatocellular cancer. The proportion of 85 hepatocellular cancer patients with more than 1 circulating cancer cell per 5 ml of blood was approximately 81%, or 69 cases. However, there was no clear correlation between the number of circulating cancer cells detected by ASGPR and hepatocellular cancer metastasis related to the stage. Meanwhile, interest in EpCAM-positive blood cancer cells exhibiting stem cell characteristics has emerged in the field of hepatocellular carcinoma, based on research results showing that EpCAM is a marker of hepatocellular carcinoma stem cells that induce metastasis of hepatocellular carcinoma or induce recurrence of treatment.
중국의 Fudan 대학은 수술 전 간세포암 환자 123명 중 7.5 ml 혈액당 EpCAM 양성 혈중 암세포가 2개 이상 검출되는 환자는 51명으로 41%에 불과했지만, 수술 후 재발되는 비율은 혈중 암세포 2개 이하 환자들과 비교했을 때 87.5% 대 15.5% 로 월등히 높았다. 다른 연구에서도 간세포암 환자 59명 중 7.5 ml 혈액당 EpCAM 양성 혈중 암세포 1개 이상 검출되는 환자는 18명으로 31%에 불과했지만, EpCAM 양성 혈중 암세포가 중간 이상 병기에서 관찰되며 생존율과 밀접한 관계가 있음을 밝혀냈다. 또 다른 연구에서도 EpCAM 기반 혈중 암세포 검출 방법을 사용하여 간세포암 환자 50명에서 7.5 ml 혈액당 혈중 암세포가 1개 이상 검출되는 비율은 14명으로 28%에 불과했으며, 전반적으로 EpCAM 기반 혈중 암세포 검출방법은 검출 비율이 50%가 넘지 않음을 알 수 있는데, 이는 암이 전이과정에서 일어나는 상피-중간엽 전이 등으로 인해 EpCAM 발현이 감소하기 때문이며, EpCAM 기반 혈중 암세포 검출 방법의 근본적인 한계이다.Fudan University in China reported that among 123 patients with hepatocellular carcinoma before surgery, only 51 patients (41%) had more than 2 EpCAM-positive circulating cancer cells per 7.5 ml of blood, but the rate of recurrence after surgery was significantly higher (87.5% vs. 15.5%) compared to patients with 2 or fewer circulating cancer cells. In another study, only 18 patients (31%) had more than 1 EpCAM-positive circulating cancer cells per 7.5 ml of blood among 59 patients with hepatocellular carcinoma, but EpCAM-positive circulating cancer cells were observed in intermediate or higher stages and were closely related to the survival rate. In another study, the rate of detecting more than 1 circulating cancer cell per 7.5 ml of blood in 50 hepatocellular carcinoma patients using an EpCAM-based circulating cancer cell detection method was only 28%, or 14 cases. Overall, the EpCAM-based circulating cancer cell detection method did not have a detection rate exceeding 50%. This is because EpCAM expression decreases due to epithelial-mesenchymal transition that occurs during the metastatic process of cancer, and is a fundamental limitation of the EpCAM-based circulating cancer cell detection method.
상피-중간엽 전이(Epitherial-Mesenchimal Transition, EMT)란, 부착 성을 가지는 상피성(epithelial) 암세포가 이동성과 침윤성을 가지는 중간엽성(mesenchymal) 암세포로 전이(transition)되는 과정 전체를 말한다. EMT 과정을 거칠 때, 상피성 세포 마커들은 하향 조절(down-regulation) 될 수 있으므로, 상피성 세포 마커에만 의존하여 혈중 암세포를 검출하는 경우, 대부분의 혈중 암세포가 검출되지 않을 수 있다. 실제로 EpCAM을 마커로 사용하여 혈중 암세포를 검출할 경우 폐암, 유방암, 자궁경부암, 비인두암 등에서는 전체 혈중 암세포 중에서 50-70% 정도만 검출할 수 있고, 간암의 경우에는 검출되는 혈중 암세포가 전체의 25%에 불과하다. 간암에서 혼합 및 중간엽성 마커를 가진 혈중 암세포를 검출하여 분석한 결과, 혈중 암세포가 많을 경우 더 강력한 침습 능력을 가지며 이러한 환자는 높은 임상 단계를 나타냈다. 또한 간세포암 환자 214명의 말초 혈액에서 혈중 암세포 및 혈중 암세포 클러스터를 수집하여 분석한 결과, 혈중 암세포 클러스터는 전반적 생존율(overall survival, OS)이나 무진행생존율(progression-free survival, PFS) 밀접하게 연관되어 있고 불량한 예후를 보여줬다는 결과도 발표되었다.Epithelial-mesenchymal transition (EMT) refers to the entire process by which adherent epithelial cancer cells transition into mesenchymal cancer cells with motility and invasiveness. During the EMT process, epithelial cell markers may be down-regulated. Therefore, if circulating cancer cells are detected solely based on epithelial cell markers, most circulating cancer cells may not be detected. In fact, when EpCAM is used as a marker to detect circulating cancer cells, only 50-70% of all circulating cancer cells can be detected in lung cancer, breast cancer, cervical cancer, and nasopharyngeal cancer, and in the case of liver cancer, only 25% of the circulating cancer cells are detected. The results of analyzing the circulating cancer cells with mixed and mesenchymal markers in hepatocellular carcinoma showed that patients with more circulating cancer cells had stronger invasive ability, and these patients showed higher clinical stages. In addition, the results of analyzing the circulating cancer cells and circulating cancer cell clusters collected from the peripheral blood of 214 hepatocellular carcinoma patients showed that the circulating cancer cell clusters were closely related to the overall survival (OS) or progression-free survival (PFS) and showed a poor prognosis.
한편 최근에는 EMT는 가역적이고 일시적일 수 있고, 세포가 완전한 EMT를 통해 중간엽성 세포로 전환되기보다 부분적인 EMT가 일어난다는 연구 결과가 발표되고 있다. 실제로 대부분의 혈중 암세포는 상피성과 중간엽성을 모두 가지는 중간 형태의 하이브리드 E/M 상태(hybrid epithelial/mesenchymal state) 또는 부분적인 표현형(phenotype)을 가지는 상태로 알려져 있으며, 이들은 클러스터를 이루는 성질이 있고 면역회피능력을 가져 숙주의 면역시스템을 회피할 수 있다고 알려져 있다. 따라서 환자의 모든 혈중 암세포를 검출해내기 위해서는, 상피성 혈중 암세포뿐만 아니라, EMT 표현형을 나타내는 혈중 암세포, 부분적인 EMT를 나타내는 혈중 암세포, 그리고 전혀 새로운 형태의 혈중 암세포를 모두 검출할 수 있는 마커를 발굴할 필요가 있다.Meanwhile, recent studies have shown that EMT can be reversible and temporary, and that partial EMT occurs rather than cells transforming into mesenchymal cells through complete EMT. In fact, most circulating cancer cells are known to have a hybrid epithelial/mesenchymal state or a partial phenotype that has both epithelial and mesenchymal characteristics, and they are known to have a tendency to form clusters and have immune-evading abilities to evade the host's immune system. Therefore, in order to detect all circulating cancer cells in a patient, it is necessary to discover markers that can detect not only epithelial circulating cancer cells, but also circulating cancer cells showing an EMT phenotype, circulating cancer cells showing partial EMT, and entirely new types of circulating cancer cells.
PTGFRN(Prostaglandin F2 Receptor Negative Regulator, EWI-F, CD9P-1, CD315)은 Immunoglobulin superfamily로 Ⅰ형 막관통 단백질(transmembrane protein)으로, Prostaglandin F2 Receptor에 음성적으로 작용한다. PTGFRN은 tetraspanin, integrin, 신호전달 효소 및 proteoglycan 등으로 이루어진 Tetraspanin Enriched Microdomain(TEM)의 구성요소이며, CD9 및 CD81과 같은 tetraspanin 단백질과 상호작용을 한다. PTGFRN은 엑소좀에서 구조단백질로서 많이 발현되어 있어, 이를 통한 엑소좀의 분리 기능이 연구되고 있다. 이외에도 PTGFRN의 발현은 근육세포의 재생, 신경교종의 성장, 전이 등에 관여한다. PTGFRN은 신경아교종을 포함하는 많은 암세포에서 상향 조절(up-regulation)되는 분자로서, 악성 뇌종양인 교모세포종(glioblastoma multiforme, GBM)에서 과발현 되어 PI3K-AKT 신호전달을 통해 세포 성장과 방사선 저항성을 촉진한다고 알려져 있다.PTGFRN (Prostaglandin F2 Receptor Negative Regulator, EWI-F, CD9P-1, CD315) is a type I transmembrane protein of the immunoglobulin superfamily that negatively affects the Prostaglandin F2 Receptor. PTGFRN is a component of the Tetraspanin Enriched Microdomain (TEM), which consists of tetraspanins, integrins, signaling enzymes, and proteoglycans, and interacts with tetraspanin proteins such as CD9 and CD81. PTGFRN is expressed in large quantities as a structural protein in exosomes, and its separation function through exosomes is being studied. In addition, the expression of PTGFRN is involved in the regeneration of muscle cells, the growth of glioma, and metastasis. PTGFRN is a molecule that is up-regulated in many cancer cells, including gliomas, and is known to be overexpressed in glioblastoma multiforme (GBM), a malignant brain tumor, where it promotes cell growth and radioresistance through PI3K-AKT signaling.
본 발명자들은 인간배아줄기세포의 표면에 결합하는 70종의 단일클론항체를 제조하였으며 이들 중 하나인 63-D7는 인간배아줄기세포와 배아암세포뿐만 아니라 간암세포를 포함한 다양한 인간 암세포 표면에 잘 결합하였으며, 정상세포인 인간 말초 혈액 단핵구 세포(peripheral blood mononuclear cells, PBMC), 태아 폐섬유아세포(fetal lung fibroblast, MRC5) 및 정상 간세포에는 결합하지 않았다. 이 발명에서 63-D7항체가 간암 환자에서 혈중 암세포를 인식한다는 것을 알아낸 후, 그 표적 분자가 PTGFRN임을 밝혔다. 63-D7은 간암, 폐암, 대장암, 갑상선암 및 췌장암세포에서 PTGFRN을 인식하였다. 간암세포를 이용한 생존능, invasion, migration 실험에서 PTGFRN 유전자 발현을 억제하면 암세포의 클론성 생존, 이동, 침입을 억제하여 결론적으로 암의 전이를 억제할 수 있음을 확인하였다. 또한 간암 환자 95명을 대상으로 한 분석에서 63-D7 항체는 혈중 암세포를 92명(97%, 92/95)에서 검출할 수 있으며, 전이 또는 재발하는 환자의 경우에는 그 검출할 수 있는 비율이 더욱 증가함을 관찰하였다. 그리고 63-D7이 인식하는 PTGFRN은 혈중 암세포에서 약물 저항성 및 면역 회피와 관련된 B7-H3 (57.6%), MVP (31.9%), TGFβR1 (51.8%), PD-L1 (37.8%), PD-L2 (17.5%) 및 CD47 (38.1%)와 높은 비율로 함께 발현하여, PTGFRN의 발현이 전이나 재발과 밀접하게 연관됨을 보여주었으며, 이들 분자들을 통해 암세포의 면역회피를 유도할 수 있음을 제시한다. 또한 63-D7이 간암세포와 췌장암세포에서 내부화를 유도하는 항체임을 밝혔으며, 실제로 이런 특성을 통해 Antibody-Drug Conjugate(ADC)를 이용하여 간암세포 사멸을 유도함으로써 63-D7를 향후 암 치료제로 개발할 수 있음을 제시한다. 또한 63-D7을 처리한 간암세포에서 간암세포의 침윤성이 증가함을 확인할 수 있었으므로 63-D7 항체 자체는 간암세포의 이동과 침윤성을 촉진하는 아고니스틱 항체(agonistic antibody)이다. 이러한 성질을 활용하여 63-D7 항체를 원발암에서 암세포의 이동을 촉진하여 초기 원발암 발생을 예방하거나 원발암 성장을 방해하는데 응용할 수 있을 것으로 기대된다.The present inventors have prepared 70 kinds of monoclonal antibodies that bind to the surface of human embryonic stem cells, and one of them, 63-D7, binds well to the surface of various human cancer cells including human embryonic stem cells, embryonic cancer cells, and liver cancer cells, and does not bind to normal cells such as human peripheral blood mononuclear cells (PBMC), fetal lung fibroblasts (MRC5), and normal hepatocytes. In this invention, after finding that the 63-D7 antibody recognizes blood cancer cells in liver cancer patients, it was revealed that its target molecule is PTGFRN. 63-D7 recognized PTGFRN in liver cancer, lung cancer, colon cancer, thyroid cancer, and pancreatic cancer cells. In experiments on viability, invasion, and migration using liver cancer cells, it was confirmed that inhibition of PTGFRN gene expression inhibits the clonal survival, migration, and invasion of cancer cells, and consequently inhibits cancer metastasis. In addition, in an analysis of 95 patients with liver cancer, the 63-D7 antibody could detect circulating cancer cells in 92 patients (97%, 92/95), and the detectable rate further increased in patients with metastasis or recurrence. In addition, PTGFRN recognized by 63-D7 was highly co-expressed with B7-H3 (57.6%), MVP (31.9%), TGFβR1 (51.8%), PD-L1 (37.8%), PD-L2 (17.5%), and CD47 (38.1%), which are associated with drug resistance and immune evasion in circulating cancer cells, showing that the expression of PTGFRN is closely related to metastasis or recurrence, and suggesting that these molecules can induce immune evasion of cancer cells. In addition, we revealed that 63-D7 is an antibody that induces internalization in liver cancer cells and pancreatic cancer cells, and in fact, through these properties, we suggest that 63-D7 can be developed as a cancer treatment agent in the future by inducing liver cancer cell death using an Antibody-Drug Conjugate (ADC). In addition, we were able to confirm that the invasiveness of liver cancer cells treated with 63-D7 increased, so the 63-D7 antibody itself is an agonistic antibody that promotes the migration and invasiveness of liver cancer cells. By utilizing these properties, it is expected that the 63-D7 antibody can be applied to promote the migration of cancer cells in primary cancers to prevent the occurrence of early primary cancers or to interfere with the growth of primary cancers.
본 발명은 암세포의 표면 분자 마커로서 PTGFRN의 용도를 제공하는 것을 목적으로 한다.The present invention aims to provide a use of PTGFRN as a surface molecular marker of cancer cells.
본 발명은 PTGFRN에 특이적으로 결합하는 단일클론항체 63-D7을 제공하는 것을 목적으로 한다.The present invention aims to provide a monoclonal antibody 63-D7 that specifically binds to PTGFRN.
본 발명은 단일클론항체 63-D7의 암세포에서 이동성 및 침윤성을 증가시키는 PTGFRN에 대한 아고니스틱 항체로서의 용도를 제공하는 것을 목적으로 한다.The present invention aims to provide the use of monoclonal antibody 63-D7 as an agonistic antibody against PTGFRN that increases mobility and invasiveness in cancer cells.
본 발명은 혈중 암세포의 표면 분자 마커로서 PTGFRN의 용도를 제공하는 것을 목적으로 한다.The present invention aims to provide a use of PTGFRN as a surface molecular marker of blood cancer cells.
본 발명의 다른 목적은 PTGFRN에 특이적으로 결합하는 물질로서 단일클론항체 63-D7를 이용하여 PTGFRN 단백질을 분리, 정제 또는 검출하는 상기 단일클론항체의 용도를 제공하는 것이다.Another object of the present invention is to provide a use of the monoclonal antibody 63-D7 as a substance that specifically binds to PTGFRN to isolate, purify or detect PTGFRN protein.
1. 서열번호 3의 아미노산 서열로 이루어진 HCDR1, 서열번호 4의 아미노산 서열로 이루어진 HCDR2 및 서열번호 5의 아미노산 서열로 이루어진 HCDR3을 포함하는 중쇄 가변영역; 및 서열번호 6의 아미노산 서열로 이루어진 LCDR1, 서열번호 7의 아미노산 서열로 이루어진 LCDR2 및 서열번호 8의 아미노산 서열로 이루어진 LCDR3을 포함하는 경쇄 가변영역을 포함하는 항 PTGFRN 단일클론항체.1. An anti-PTGFRN monoclonal antibody comprising a heavy chain variable region comprising HCDR1 consisting of the amino acid sequence of SEQ ID NO: 3, HCDR2 consisting of the amino acid sequence of SEQ ID NO: 4, and HCDR3 consisting of the amino acid sequence of SEQ ID NO: 5; and a light chain variable region comprising LCDR1 consisting of the amino acid sequence of SEQ ID NO: 6, LCDR2 consisting of the amino acid sequence of SEQ ID NO: 7, and LCDR3 consisting of the amino acid sequence of SEQ ID NO: 8.
2. 위 1에 있어서, 중쇄 가변영역은 서열번호 1의 아미노산 서열을 포함하는 항 PTGFRN 단일클론항체.2. In the above 1, the heavy chain variable region is an anti-PTGFRN monoclonal antibody comprising the amino acid sequence of
3. 청구항 1에 있어서, 경쇄 가변영역은 서열번호 2의 아미노산 서열을 포함하는 항 PTGFRN 단일클론항체.3. An anti-PTGFRN monoclonal antibody according to
4. 위 1 내지 3 중 어느 하나의 항 PTGFRN 단일클론항체에 약물이 접합된 항체-약물 컨쥬게이트.4. An antibody-drug conjugate comprising a drug conjugated to any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
5. 위 1 내지 3 중 어느 하나의 항 PTGFRN 단일클론항체를 포함하는 암의 치료 또는 예방용 약학 조성물.5. A pharmaceutical composition for treating or preventing cancer comprising any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
6. 위 5에 있어서, 암은 뇌척수종양, 두경부암, 폐암, 유방암, 흉선종, 중피종, 식도암, 위암, 대장암, 간암, 갑상선암, 췌장암, 담도암, 신장암, 방광암, 전립선암, 고환암, 생식세포종, 난소암, 자궁 경부암, 자궁 내막암, 림프종, 급성 백혈병, 만성 백혈병, 다발성 골수종, 육종, 악성 흑색종, 신경모세포종, 교모세포종 및 피부암으로 이루어진 군에서 선택되는, 암의 치료 또는 예방용 약학 조성물.6. In the above 5, the cancer is selected from the group consisting of brain and spinal tumors, head and neck cancer, lung cancer, breast cancer, thymoma, mesothelioma, esophageal cancer, stomach cancer, colon cancer, liver cancer, thyroid cancer, pancreatic cancer, biliary tract cancer, kidney cancer, bladder cancer, prostate cancer, testicular cancer, germ cell tumor, ovarian cancer, cervical cancer, endometrial cancer, lymphoma, acute leukemia, chronic leukemia, multiple myeloma, sarcoma, malignant melanoma, neuroblastoma, glioblastoma, and skin cancer, a pharmaceutical composition for treating or preventing cancer.
7. 위 1 내지 3 중 어느 하나의 항 PTGFRN 단일클론항체를 생산하는 하이브리도마.7. A hybridoma producing any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
8. 위 1 내지 3 중 어느 하나의 항 PTGFRN 단일클론항체를 포함하는 PTGFRN 검출용 조성물.8. A composition for detecting PTGFRN comprising any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
9. 개체로부터 분리된 시료에 위 1 내지 3 중 어느 하나의 항 PTGFRN 단일클론항체를 처리하는 단계를 포함하는 암의 진단을 위한 정보 제공 방법.9. A method for providing information for diagnosing cancer, comprising the step of treating a sample isolated from an individual with any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
10. 개체로부터 분리된 시료에 위 1 내지 3 중 어느 하나의 항 PTGFRN 단일클론항체를 처리하는 단계를 포함하는 혈중 암세포 수준 판단에 필요한 정보 제공 방법.10. A method for providing information necessary for determining the level of cancer cells in blood, comprising the step of treating a sample isolated from an individual with any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
11. 위 10에 있어서, 시료는 혈액, 혈장, 골수액, 림프액, 타액, 누액, 뇨, 점막액, 양수로 이루어진 군에서 선택되는, 혈중 암세포 수준 판단에 필요한 정보 제공 방법.11. A method for providing information necessary for determining the level of blood cancer cells, wherein the sample is selected from the group consisting of blood, plasma, bone marrow fluid, lymph, saliva, tears, urine, mucous membrane fluid, and amniotic fluid in the above 10.
12. 위 10에 있어서, 시료에 EpCAM에 특이적으로 결합하는 물질을 처리하는 단계를 더 포함하는, 혈중 암세포 수준 판단에 필요한 정보 제공 방법.12. A method for providing information necessary for determining the level of cancer cells in blood, further comprising the step of treating a sample with a substance that specifically binds to EpCAM in the above 10.
13. 위 12에 있어서, EpCAM에 특이적으로 결합하는 물질은 항-EpCAM 항체 또는 이의 항원 결합 단편인, 혈중 암세포 수준 판단에 필요한 정보 제공 방법.13. A method for providing information necessary for determining the level of cancer cells in blood, wherein in the above 12, the substance that specifically binds to EpCAM is an anti-EpCAM antibody or an antigen-binding fragment thereof.
14. 위 10에 있어서, 혈중 암세포는 상피-중간엽 전이되거나 상피-중간엽 전이 진행 중인 혈중 암세포인, 혈중 암세포 수준 판단에 필요한 정보 제공 방법.14. In the above 10, a method for providing information necessary for determining the level of blood cancer cells, wherein the blood cancer cells are blood cancer cells that have undergone epithelial-mesenchymal transition or are undergoing epithelial-mesenchymal transition.
15. 위 10에 있어서, 혈중 암세포는 상피성 세포 또는 중간엽성 세포가 아닌, 혈중 암세포 수준 판단에 필요한 정보 제공 방법.15. In the above 10, a method for providing information necessary for determining the level of blood cancer cells, wherein the blood cancer cells are not epithelial cells or mesenchymal cells.
16. 위 10에 있어서, 혈중 암세포는 간암, 폐암, 대장암, 감상선암, 췌장암, 악성 흑색종, 유방암, 신경모세포종 또는 교모세포종 환자의 혈액 내 혈중 암세포인, 혈중 암세포 수준 판단에 필요한 정보 제공 방법.16. In the above 10, the blood cancer cells are blood cancer cells in the blood of a patient with liver cancer, lung cancer, colon cancer, thyroid cancer, pancreatic cancer, malignant melanoma, breast cancer, neuroblastoma or glioblastoma, a method for providing information necessary for determining the level of blood cancer cells.
17. 위 10에 있어서, 혈중 암세포는 B7-H3(CD276) 단백질을 더 발현하는, 혈중 암세포 수준 판단에 필요한 정보 제공 방법.17. A method for providing information necessary for determining the level of blood cancer cells, wherein blood cancer cells in the above 10 more express B7-H3 (CD276) protein.
18. 개체로부터 분리된 시료에 위 1 내지 3 중 어느 하나의 항 PTGFRN 단일클론항체를 처리하여 PTGFRN을 검출하는 단계를 포함하는 암 전이 예후 예측에 필요한 정보 제공 방법.18. A method for providing information necessary for predicting the prognosis of cancer metastasis, comprising the step of detecting PTGFRN by treating a sample isolated from an individual with any one of the above 1 to 3 anti-PTGFRN monoclonal antibodies.
19. 위 18에 있어서, 검출된 PTGFRN의 수준이 대조군 대비 높으면 대조군 대비 암 전이 예후가 더 나쁜 것이라는 정보를 제공하는 단계를 더 포함하는, 암 전이 예후 예측에 필요한 정보 제공 방법.19. A method for providing information necessary for predicting the prognosis of cancer metastasis, further comprising a step of providing information that if the level of detected PTGFRN is higher than that of the control group, the prognosis of cancer metastasis is worse than that of the control group.
20. 항암 약물을 투여 받은 환자로부터 분리된 시료에 위 1 내지 3 중 어느 하나의 항 PTGFRN 단일클론항체를 처리하여 PTGFRN을 검출하는 단계를 포함하는 항암 약물의 효능 스크리닝 방법.20. A method for screening the efficacy of an anticancer drug, comprising the step of detecting PTGFRN by treating a sample isolated from a patient administered an anticancer drug with any one of the anti-PTGFRN
21. 위 20에 있어서, 검출된 PTGFRN의 수준이 대조군 대비 낮으면 대조군 대비 상기 항암 약물의 암 전이 억제 효능이 더 우수할 것으로 예측하는 단계를 더 포함하는, 항암 약물의 효능 스크리닝 방법.21. A method for screening the efficacy of an anticancer drug, further comprising a step of predicting that the anticancer drug has a better efficacy in inhibiting cancer metastasis than the control group if the level of PTGFRN detected in the above 20 is lower than that of the control group.
22. PTGFRN에 결합하는 물질을 포함하는 혈중 암세포 검출용 조성물.22. A composition for detecting cancer cells in blood, comprising a substance that binds to PTGFRN.
본 발명의 항 PTGFRN 단일클론항체 63-D7은 혈중 암세포 수준 판단, 암 치료 또는 예방, 암 전이 예후 예측 및 항암제 스크리닝에 사용될 수 있다.The anti-PTGFRN monoclonal antibody 63-D7 of the present invention can be used for determining the level of blood cancer cells, treating or preventing cancer, predicting the prognosis of cancer metastasis, and screening for anticancer drugs.
본 발명의 항 PTGFRN 단일클론항체는 간암을 포함하는 다양한 암세포 표면의 PTGFRN에 특이적으로 결합할 수 있다.The anti-PTGFRN monoclonal antibody of the present invention can specifically bind to PTGFRN on the surface of various cancer cells, including liver cancer.
본 발명의 항 PTGFRN 단일클론항체를 이용하여, 다양한 항체-약물 컨쥬게이트를 개발하여 항암제로 사용할 수 있다.Using the anti-PTGFRN monoclonal antibody of the present invention, various antibody-drug conjugates can be developed and used as anticancer agents.
본 발명의 항 PTGFRN 단일클론항체가 PTGFRN를 자극하여 암세포의 이동성 및 침윤성을 증가시킨다는 점을 활용하여, 본 발명의 항 PTGFRN 단일클론항체를 원발암 세포의 이동을 촉진함으로써 원발암의 암 성장을 예방하고 억제하는 데 사용할 수 있다.By utilizing the fact that the anti-PTGFRN monoclonal antibody of the present invention stimulates PTGFRN to increase the mobility and invasiveness of cancer cells, the anti-PTGFRN monoclonal antibody of the present invention can be used to prevent and inhibit the cancer growth of primary cancer by promoting the migration of primary cancer cells.
본 발명의 항 PTGFRN 단일클론항체는 상피성도 중간엽성도 가지지 않는 혈중 암세포를 검출할 수 있다.The anti-PTGFRN monoclonal antibody of the present invention can detect blood cancer cells that are neither epithelial nor mesenchymal.
본 발명의 항 PTGFRN 단일클론항체는 부분적으로 상피-중간엽 전이된 혈중 암세포를 검출할 수 있다.The anti-PTGFRN monoclonal antibody of the present invention can partially detect circulating cancer cells that have undergone epithelial-mesenchymal transition.
본 발명의 항 PTGFRN 단일클론항체는 간암 환자에서 높은 효율로 혈중 암세포를 검출할 수 있다.The anti-PTGFRN monoclonal antibody of the present invention can detect blood cancer cells with high efficiency in liver cancer patients.
본 발명의 항 PTGFRN 단일클론항체에 의해 검출된 혈중 암세포는 1차 암환자에서보다 2차 전이성 암 환자에게서 더 높게 측정되어, 본 발명의 항 PTGFRN 단일클론항체를 통해 암의 병기를 판단할 수 있다.Blood cancer cells detected by the anti-PTGFRN monoclonal antibody of the present invention are measured at a higher level in patients with secondary metastatic cancer than in patients with primary cancer, so that the stage of cancer can be determined through the anti-PTGFRN monoclonal antibody of the present invention.
본 발명의 항 PTGFRN 단일클론항체는 암 세포에 내부화 될 수 있어, 암 세포 내부로 약물을 효과적으로 전달할 수 있다.The anti-PTGFRN monoclonal antibody of the present invention can be internalized into cancer cells, thereby effectively delivering the drug into cancer cells.
본 발명은 또한 혈중 암세포의 표면 마커로서 PTGFRN를 제공한다.The present invention also provides PTGFRN as a surface marker of blood cancer cells.
항 PTGFRN 단일클론항체는 PTGFRN 단백질과의 높은 특이적 결합을 통해 PTGFRN 단백질의 분리, 정제 및 검출에 사용될 수 있다.Anti-PTGFRN monoclonal antibodies can be used for the isolation, purification, and detection of PTGFRN protein through highly specific binding to PTGFRN protein.
항 PTGFRN 단일클론항체는 PTGFRN를 표면에 발현하는 상피-중간엽 전이된 혈중 암세포와 부분적 상피-중갑엽성 혈중 암세포에 결합하므로 기존에는 검출할 수 없었던 혈중 암세포를 검출할 수 있다.The anti-PTGFRN monoclonal antibody binds to epithelial-mesenchymal transition circulating cancer cells and partially epithelial-mesenchymal circulating cancer cells that express PTGFRN on their surface, thereby enabling the detection of previously undetectable circulating cancer cells.
항 PTGFRN 단일클론항체는 혈중 암세포 검출을 통해 전이성 암 진단에 유용하게 사용될 수 있다.Anti-PTGFRN monoclonal antibodies may be useful in the diagnosis of metastatic cancer by detecting cancer cells in the blood.
도 1a는 유세포 분석(flow cytometry) 결과를 나타낸 그래프로서, 항체 246-D7, 247-B9와 본 발명의 단일클론항체 63-D7가 각각 인간배아줄기세포 H9, 2종의 간암세포(Huh7, HepG2), 2종의 폐암세포(A549, NCI-H358), 말초혈액단핵구세포(PBMC)와 결합하는 정도를 보여준다. 도 1b는 도 1a의 그래프를 표로 나타낸 것이다. 도 1c는 면역세포화학염색 결과로서, 혈중 암세포 검출 여부 확인을 위해 간암환자 8명과 정상인 1명의 혈액에서 각각 바이오틴(biotin)으로 표지한 단일클론항체 63-D7, 246-D7, 247-B9와 CD45 항체를 사용해 이중염색하였고, 핵을 DAPI로 염색하였으며, 이들을 병합한 Merge를 도시하였다. 도 1d는 도 1c의 결과를 표로 나타낸 것이다.Fig. 1a is a graph showing the results of flow cytometry, showing the degree of binding of antibodies 246-D7, 247-B9 and monoclonal antibody 63-D7 of the present invention to human embryonic stem cell H9, two types of liver cancer cells (Huh7, HepG2), two types of lung cancer cells (A549, NCI-H358), and peripheral blood mononuclear cells (PBMC), respectively. Fig. 1b is a table showing the graph of Fig. 1a. Fig. 1c is a result of immunocytochemical staining, in order to confirm the detection of cancer cells in the blood, double staining was performed using monoclonal antibodies 63-D7, 246-D7, 247-B9 and CD45 antibody labeled with biotin in the blood of eight liver cancer patients and one normal person, respectively, and the nuclei were stained with DAPI, and a Merge of these was illustrated. Figure 1d shows the results of Figure 1c in a table.
도 2는 유세포 분석(flow cytometry)의 결과로서, 본 발명의 단일클론항체 63-D7가 인간만능줄기세포(H9), 인간 배아암종세포(NT2/D1) 및 간암세포(Huh7, HepG2, SNU387, SNU449)를 포함한 다양한 암세포(SK-Hep1, A549, NCI-H358, NCI-H460, NCI-H1703, NCI-H23, Colo205, HCT116, SNU790, 8505C, BxPC3, A375, MCF-7, MDA-MB-435, SH-SY5Y, U87-MG)와 인간 배아 신장세포(HEK293FT)에는 강하게 결합하지만, 골수-유래 중간엽 줄기세포(BM-MSC)와 인간 피부 섬유아세포(HDF)에는 미약하게 결합하고, 인간 말초 혈액 단핵구 세포(PBMC), 인간 태아 폐 섬유아세포(MRC5), 치수세포(DPC) 및 정상 간세포(Hepatocyte)에는 결합하지 않음을 보여준다. 이 때, 실선은 단일클론항체 63-D7이고 음영 바탕은 2차 항체만 포함한 음성 대조군이다.Figure 2 shows the results of flow cytometry, which shows that the monoclonal antibody 63-D7 of the present invention strongly binds to various cancer cells (SK-Hep1, A549, NCI-H358, NCI-H460, NCI-H1703, NCI-H23, Colo205, HCT116, SNU790, 8505C, BxPC3, A375, MCF-7, MDA-MB-435, SH-SY5Y, U87-MG) including human pluripotent stem cells (H9), human embryonic carcinoma cells (NT2/D1), and liver cancer cells (Huh7, HepG2, SNU387, SNU449) and human embryonic kidney cells (HEK293FT), but weakly binds to bone marrow-derived mesenchymal stem cells (BM-MSC) and human dermal fibroblasts (HDF), and human peripheral blood fibroblasts (PBS). It shows no binding to blood mononuclear cells (PBMCs), human fetal lung fibroblasts (MRC5), dental pulp cells (DPCs), and normal hepatocytes. Here, the solid line is monoclonal antibody 63-D7 and the shaded background is a negative control containing only the secondary antibody.
도 3a는 세포 표면을 바이오틴으로 표지한 NT-2/D1 인간 배아암줄기세포(NT-2/D1-biotin)의 세포 용출액에서 단일클론항체 63-D7을 사용하여 면역 침강시킨 단백질을 10% SDS-PAGE를 통해 분리하고, 웨스턴 블롯팅 하여 니트로셀룰로스 막으로 옮긴 후, 스트렙타비딘-HRP(SA-HRP)로 반응시켜 분석한 결과이다. 항체를 넣지 않고 동일 실험을 진행한 것(No Ab)을 음성 대조군(Con)으로 하였다. 화살표는 63-D7에 의해 면역 침강된 단백질을 나타낸다. 도 3b는 도 3a와 동일하게 면역 침강한 단백질을 10% SDS-PAGE를 통해 분리한 후, 폴리아크릴아마이드 젤을 PageBlue로 염색한 결과이다. 1X는 3 mg 세포 용출액을 사용한 것이고 3x는 그것의 3배 양을 사용한 것이다. 이후 도 3b의 점선 상자 내부의 단백질을 오려내어 LC-MS/MS를 수행하였다. 도 3c는 도 3b에서 단일클론항체 63-D7로 면역 침강한 후 회수한 단백질을 LC-MS/MS로 분석한 결과로, 63-D7 항원의 아미노산 서열 중 PTGFRN 단백질의 아미노산 서열과 일치하는 부분을 밑줄로 표시하였다.Figure 3a shows the results of separating proteins immunoprecipitated using monoclonal antibody 63-D7 from cell lysates of NT-2/D1 human embryonic cancer stem cells (NT-2/D1-biotin) whose cell surface was labeled with biotin, by 10% SDS-PAGE, Western blotting, transferring to a nitrocellulose membrane, and analyzing by reaction with streptavidin-HRP (SA-HRP). The same experiment performed without adding antibody (No Ab) was used as a negative control (Con). The arrow indicates the protein immunoprecipitated by 63-D7. Figure 3b shows the results of separating proteins immunoprecipitated in the same manner as in Figure 3a by 10% SDS-PAGE, and staining the polyacrylamide gel with PageBlue. 1X means that 3 mg of cell lysate was used, and 3x means that three times that amount was used. Afterwards, the protein within the dotted box in Fig. 3b was extracted and subjected to LC-MS/MS. Fig. 3c shows the result of analyzing the protein recovered after immunoprecipitation with monoclonal antibody 63-D7 in Fig. 3b by LC-MS/MS, and the part of the amino acid sequence of the 63-D7 antigen that matches the amino acid sequence of the PTGFRN protein is underlined.
도 4a는 바이오틴 표지한 인간 비소세포암 A549 세포 용출액에서 각각 생쥐 항-PTGFRN 단일클론항체(α-PTGFRN) 및 생쥐 63-D7 단일클론항체를 사용하여 면역 침강한 단백질 및 항체를 넣지 않은 음성 대조군 단백질(No Ab)를 10% SDS-PAGE로 분리한 후 생쥐 α-PTGFRN 항체를 사용해 웨스턴 블롯팅을 수행한 결과이다. 도 4b는 도 4a의 니트로셀룰로스 막을 신호 제거 용액으로 제거한 후 스트렙타비딘-HRP(SA-HRP)을 사용해 분석한 결과로 PTRFRN이 63-D7에 의해 인식되는 세포표면분자임을 확인하였다. 도 4c는 단일클론항체 63-D7의 항원이 PTGFRN임을 재확인하기 위하여 PTGFRN-FLAG 단백질을 인간 배아 신장세포 HEK293FT에서 과발현 시킨 후 세포 용출액을 공지의 생쥐 항-FLAG 단일클론항체(α-FLAG), 공지의 생쥐 항-PTGFRN 단일클론항체(α-PTGFRN) 및 단일클론항체 63-D7으로 면역 침강 및 웨스턴 블롯을 수행한 결과로서, PTRFRN이 63-D7에 의해 인식되는 세포표면분자임을 검증하였다. WB는 웨스턴 블롯을 의미한다.Figure 4a shows the results of Western blotting using mouse α-PTGFRN antibody after immunoprecipitation of proteins and a negative control protein without antibody (No Ab) using mouse anti-PTGFRN monoclonal antibody (α-PTGFRN) and mouse 63-D7 monoclonal antibody from biotin-labeled human non-small cell lung cancer A549 cell lysates, respectively, separated by 10% SDS-PAGE. Figure 4b shows the results of analysis using streptavidin-HRP (SA-HRP) after removing the nitrocellulose membrane of Figure 4a with a signal removing solution, confirming that PTRFRN is a cell surface molecule recognized by 63-D7. Figure 4c shows the results of immunoprecipitation and Western blotting of the cell lysate using a known mouse anti-FLAG monoclonal antibody (α-FLAG), a known mouse anti-PTGFRN monoclonal antibody (α-PTGFRN), and monoclonal antibody 63-D7 to reconfirm that the antigen of monoclonal antibody 63-D7 is PTGFRN. WB stands for Western blot.
도 5a는 간암세포주 Huh7 및 SNU449에서 PTGFRN 녹다운(Knockdown) 실험을 수행한 결과로, 음성 대조군 siRNA(siCon) 및 PTGFRN에 대한 두 종의 siRNA(siPTGFRN#1 및 siPTGFRN#2)를 각각 처리하여 녹다운을 수행한 간암세포에서 PTGFRN mRNA의 발현을 qPCR로 분석하였다. 도 5b는 녹다운한 Huh7 및 SNU449 세포 표면의 PTGFRN을 분석하기 위해 생쥐 PTGFRN 단일클론항체 (α-PTGFRN)가 결합하는 정도를 유세포분석으로 확인한 결과이다. 실선은 단일클론항체이고 음영 바탕은 2차 항체만 포함한 음성 대조군(isotype) 자료이다. 도 5c는 도 5b를 통계적으로 분석한 그래프이다. 도 5d는 PTGFRN 녹다운한 Huh7 및 SNU449에서 암세포 클론원성 생존을 측정한 결과로, 세포 접종 후 8일째 생존한 세포 클론을 크리스탈 바이올렛으로 염색한 것이다. 도 5e는 5d를 통계적으로 분석한 그래프로, *은 p<0.05인 p value를 나타낸다.Figure 5a shows the results of a PTGFRN knockdown experiment performed in hepatoma cell lines Huh7 and SNU449. The expression of PTGFRN mRNA was analyzed by qPCR in hepatoma cells treated with a negative control siRNA (siCon) and two types of siRNA against PTGFRN (
도 6a는 간암세포주 Huh7 및 SNU449에서 PTGFRN 녹다운 실험을 수행한 결과로, 음성대조군 scrambled shRNA(shScramble) 또는 두 종의 PTGFRN에 대한 shRNA(shPTGFRN#1, shPTGFRN#2)를 인간 배아 암세포 HEK293FT 트랜스펙션 한 후 렌티바이러스를 회수하여 Huh7 및 SNU449 간암세포에 처리하여 녹다운을 수행하고, PTGFRN 단백질 발현을 웨스턴 블롯팅으로 확인하였다. 도 6b는 Huh7 및 SNU449 간암세포에서 PTGFRN 발현이 감소한 것을 유세포분석기로 분석한 결과이다. 도 6c는 도 6b를 통계 분석한 그래프이다. 도 6d는 shPTGFRN#1으로 녹다운한 SNU449에서 암세포 클론원성 생존을 측정한 결과로, 세포 접종 후 8일째 생존한 세포 클론을 크리스탈 바이올렛으로 염색하였으며, shScramble은 대조군이다. 도 6e는 6d을 통계 분석한 결과로서, ***는 대조군(shScramble)에 대해 p<0.005 인 것을 나타낸다. 도 6f는 shPTGFRN#2으로 녹다운한 SNU449에서 암세포 클론원성 생존을 측정한 결과이다. 도 6g는 도 6f의 통계 분석한 결과로서, ***는 p<0.005인 것을 나타낸다.Figure 6a shows the results of a PTGFRN knockdown experiment performed in hepatoma cell lines Huh7 and SNU449. The human embryonic carcinoma cell line HEK293FT was transfected with a negative control scrambled shRNA (shScramble) or two shRNAs for PTGFRN (
도 7a는 음성 대조군 shRNA(shScramble) 또는 두 종의 PTGFRN에 대한 shRNA(shPTGFRN#1, shPTGFRN#2)를 이용하여 간암세포의 이동 정도를 측정한 결과로, PTGFRN 녹다운 간암세포 Huh7 및 SNU449를 24-well 트랜스웰 챔버에 분주한 후 이틀간 배양하여 아래의 챔버로 이동한 세포를 크리스탈 바이올렛으로 염색하고 찍은 사진이다. 도7b는 도7a 실험을 3회 반복하여 통계 처리한 그래프이며, ***은 p<0.001, **은 p<0.01, *은 p<0.05인 것을 나타낸다. 도 7c는 음성 대조군 shRNA(shscramble) 또는 두 종의 PTGFRN에 대한 shRNA(shPTGFRN#1, shPTGFRN#2)를 이용하여 암세포의 침입성을 측정한 결과로, PTGFRN 녹다운 간암세포 Huh7 및 SNU449를 Matrigel이 코팅된 트랜스웰 챔버에 분주한 후 이틀간 배양하여 아래층으로 침입한 세포를 크리스탈 바이올렛으로 염색하고 찍은 사진이다. 도 7d는 도 7c 실험을 3번 반복하여 통계처리한 그래프이며, ***은 p<0.001, **은 p<0.01, *은 p<0.05인 것을 나타낸다.Figure 7a is a photograph of the results of measuring the degree of migration of liver cancer cells using a negative control shRNA (shScramble) or two shRNAs against PTGFRN (
도 8a는 간암세포주 Huh7에서 음성대조군 scrambled shRNA(shScramble) 또는 두 종의 PTGFRN에 대한 shRNA(shPTGFRN#1, shPTGFRN#2)를 간암세포에 처리하여 녹다운을 수행하고, PTGFRN 녹다운된 간암세포를 마트리겔(Matrigel), CollagenⅠ, CollagenⅣ, 젤라틴(Gelatin)이 코팅된 12-well 세포배양 플레이트에 반응시켜 세포의 부착 정도를 분석한 결과로, 부착한 간암세포를 현미경으로 관찰한 사진이다. 도 8b는 도 8a 실험을 반복 수행한 후 현미경 사진을 Image J로 분석하여 세포의 결합 정도를 통계 처리한 그래프이다. ***은 p<0.001, **은 p<0.01, *은 p<0.05인 것을 나타내며, ns는 통계적 유의성이 없음을 나타낸다. 도 8c는 간암세포주 SNU449에서 음성대조군 scrambled shRNA(shScramble) 또는 두 종의 PTGFRN에 대한 shRNA(shPTGFRN#1, shPTGFRN#2)를 간암세포에 처리하여 녹다운을 수행하고, PTGFRN 녹다운된 간암세포를 마트리겔(Matrigel), CollagenⅠ, CollagenⅣ, 젤라틴(Gelatin)이 코팅된 12-well 세포배양 플레이트에 반응시켜 세포의 부착 정도를 분석한 결과로, 부착한 간암세포를 현미경으로 관찰한 사진이다. 도 8d는 도 8c 실험을 반복 수행한 후 현미경 사진을 Image J로 분석하여 세포의 결합 정도를 통계 처리한 그래프이다. ***은 p<0.001, **은 p<0.01, *은 p<0.05인 것을 나타내며, ns는 통계적 유의성이 없음을 나타낸다.Figure 8a is a photograph of attached liver cancer cells observed under a microscope after knockdown was performed by treating liver cancer cells with a negative control scrambled shRNA (shScramble) or two types of shRNA against PTGFRN (
도 9a는 PTGFRN 녹다운 간암세포의 NK세포에 대한 면역회피능력을 측정한 결과로, SNU449에서 음성대조군 scrambled shRNA(shscramble) 또는 shPTGFRN#2를 간암세포에 처리하여 녹다운을 수행하고, PTGFRN 녹다운 간암세포를 NK92와 공동 배양 후 사멸되지 않고 남은 SNU449세포를 크리스탈 바이올렛으로 염색한 것이다. 1:1, 2.5:1, 5:1은 이펙터 세포(effector cell)인 NK92와 타겟 세포(target cell)인 SNU449의 비율(E:T 비율)을 나타낸다. 도 9b는 도 9a의 세포를 녹인 후 OD540에서 흡광도를 측정하여 정량화 한 것을 그래프로 나타낸 것이다. 도 9c는 도 9b에서 측정한 OD540 결과를 E:T 비율로 표시하여 세포사멸 정도를 제시한 그래프이다Figure 9a shows the results of measuring the immune evasion ability of PTGFRN knockdown liver cancer cells against NK cells. SNU449 liver cancer cells were treated with negative control scrambled shRNA (shscramble) or
도 10은 음성 대조군 shRNA(shScramble) 또는 두 종의 PTGFRN에 대한 shRNA(shPTGFRN#1, shPTGFRN#2)를 이용하여 PTGFRN 녹다운 간암세포 Huh7 및 SNU449의 단백질 용출액을 통해 PTGFRN 감소에 따른 세포의 B7-H3, FAK, Src의 단백질 및 인산화된 p-FAK(Y397), p-Src(Y416) 단백질의 발현 변화를 웨스턴 블롯으로 분석한 결과이고, GAPDH는 대조군이며, 숫자는 image J로 정량한 것이다.Figure 10 shows the results of Western blot analysis of the changes in the expression of B7-H3, FAK, Src proteins, and phosphorylated p-FAK (Y397), p-Src (Y416) proteins in the protein lysates of PTGFRN knockdown hepatoma cells Huh7 and SNU449, which were treated with a negative control shRNA (shScramble) or two types of shRNA against PTGFRN (
도 11a는 HEK293FT 세포에 pcDNA3.1(+)PTGFRN-FLAG 벡터와 pcDNA3.1(+)PTGFRN-myc 벡터를 co-transfection 시킨 후 용출액을 얻어 한 세포내에서 같은 PTGFRN의 cis 상호작용 확인하기 위해 생쥐 항-FLAG, 토끼 항-myc, 63-D7로 면역침강하고, anti-FLAG, anti-myc 항체로 웨스턴 블롯을 수행한 결과이다. No Ab는 대조군이며 화살표는 면역 침강된 PTGFRN을 의미한다. 도 11b는 HEK293FT 세포에 pcDNA3.1(+)PTGFRN-FLAG 벡터와 pcDNA3.1(+)PTGFRN-myc 벡터를 각각 transfection 시킨 후 각각의 용출액을 섞어서 다른 세포 사이 trans 상호작용을 확인하기 위해 생쥐 항-FLAG, 토끼 항-myc, 63-D7로 면역침강하고, anti-FLAG, anti-myc 항체로 웨스턴 블롯을 수행한 결과이다. No Ab는 대조군이며 화살표는 면역 침강된 PTGFRN을 의미한다. 도 11c는 간암세포 SNU449에서 각각 생쥐 항-PTGFRN 단일클론항체(α-PTGFRN), 생쥐 항-63-D7 단일클론항체, 생쥐 항-SLC3A2, 생쥐 항-B7-H3로 면역 침강시킨 후 α-PTGFRN로 웨스턴 블롯팅 한 결과로, SLC3A2(CD98hc)과 B7-H3에 의해 PTGFRN이 면역 침강되는 것을 보여준다. 도 11d는 B7-H3와 PTGFRN의 상호작용을 확인하기 위해, HEK293FT 세포에 pcDNA3.1(+)PTGFRN-myc 벡터와 pcDNA3.1(+)B7-H3-FLAG 벡터를 co-transfection시킨 다음, 항-FLAG, 항-B7-H3, 항-myc, 63-D7로 면역 침강하고 항-myc, 항-FLAG 항체로 검출한 결과 서로 상호 작용함을 확인한 결과이다. HC는 면역글로뷸린(immunoglobulin) 중쇄를 의미한다. 도 11a 내지 도 11d에서 WB는 웨스턴 블롯을 의미한다.Figure 11a shows the results of immunoprecipitation of the lysate obtained after co-transfection of HEK293FT cells with pcDNA3.1(+)PTGFRN-FLAG vector and pcDNA3.1(+)PTGFRN-myc vector to confirm the cis interaction of the same PTGFRN in one cell with mouse anti-FLAG, rabbit anti-myc, and 63-D7, and Western blotting with anti-FLAG and anti-myc antibodies. No Ab is the control, and the arrow indicates the immunoprecipitated PTGFRN. Figure 11b shows the results of mixing the lysates of HEK293FT cells transfected with pcDNA3.1(+)PTGFRN-FLAG vector and pcDNA3.1(+)PTGFRN-myc vector, respectively, and immunoprecipitating them with mouse anti-FLAG, rabbit anti-myc, and 63-D7 to confirm trans-interactions between different cells, and performing Western blotting with anti-FLAG and anti-myc antibodies. No Ab is the control, and the arrow indicates immunoprecipitated PTGFRN. Figure 11c shows the results of Western blotting with α-PTGFRN after immunoprecipitation with mouse anti-PTGFRN monoclonal antibody (α-PTGFRN), mouse anti-63-D7 monoclonal antibody, mouse anti-SLC3A2, and mouse anti-B7-H3, respectively, in hepatoma cell SNU449, showing that PTGFRN is immunoprecipitated by SLC3A2 (CD98hc) and B7-H3. Figure 11d shows the results of co-transfecting HEK293FT cells with pcDNA3.1(+)PTGFRN-myc vector and pcDNA3.1(+)B7-H3-FLAG vector, followed by immunoprecipitation with anti-FLAG, anti-B7-H3, anti-myc, and 63-D7, and detection with anti-myc and anti-FLAG antibodies, confirming that they interact with each other. HC stands for immunoglobulin heavy chain. In Figures 11a to 11d, WB stands for western blot.
도 12a 및 12b는 간암세포주 각각 Huh7 및 HepG2에서 63-D7 항체와 마그네틱 비드로 63-D7 양성 및 음성 세포를 sorting 한 후, Materigel-coated plate에서 세포 부착성을 측정한 결과이다. 도 12c는 Huh7를 종양구 배양하여 암줄기세포성을 증가시킨 후, 63-D7의 암세포 결합력을 암줄기세포 양성 마커인 CD133, CD44 및 EpCAM이 부착성 세포일때와 비교하여 유세포분석으로 표시한 결과이다. 도 12d는 도 12c를 통계적으로 분석한 그래프이다. ***은 p<0.005인 것을 나타낸다. 도 12e는 63-D7으로 sorting 한 간암세포주 Huh7에서 63-D7 양성 및 음성 암세포의 암줄기세포성을 보여주는 클론원성 생존을 측정한 결과로, 세포를 접종 후 8일째 생존한 세포 클론을 크리스탈 바이올렛으로 염색한 그림이다. 도 12f는 도 12e의 결과를 통계 처리한 그림이다. ***은 대조군에 대해 p<0.005인 것을 나타낸다. 도 12g는 63-D7으로 sorting 한 간암세포주 HepG2에서 63-D7 양성 및 음성 암세포의 클론원성 생존을 측정한 결과이다. 도 12h는 도 12g의 결과를 통계 처리한 그림이다. ***은 대조군에 대해 p<0.005인 것을 나타낸다.Figures 12a and 12b show the results of measuring cell adhesion on a Materigel-coated plate after sorting 63-D7 positive and negative cells with 63-D7 antibody and magnetic beads in liver cancer cell lines Huh7 and HepG2, respectively. Figure 12c shows the results of flow cytometry analysis to compare the cancer cell binding ability of 63-D7 with that of CD133, CD44, and EpCAM, which are cancer stem cell positive markers, when they are adherent cells, after culturing Huh7 as tumor cells to increase cancer stem cell potential. Figure 12d is a graph statistically analyzing Figure 12c. *** indicates p<0.005. Fig. 12e is a graph showing the results of measuring the clonogenic survival of 63-D7 positive and negative cancer cells in the liver cancer cell line Huh7 sorted with 63-D7, showing the cancer stemness of the cell clones that survived 8 days after inoculation, stained with crystal violet. Fig. 12f is a graph showing the statistical results of Fig. 12e. *** indicates p<0.005 compared to the control group. Fig. 12g is a graph showing the results of measuring the clonogenic survival of 63-D7 positive and negative cancer cells in the liver cancer cell line HepG2 sorted with 63-D7. Fig. 12h is a graph showing the statistical results of Fig. 12g. *** indicates p<0.005 compared to the control group.
도 13a 및 도 13b는 유세포 분석을 통해 단일클론항체 63-D7가 인간 간암세포 Huh7, HepG2, SNU449와 인간 췌장암세포 SNU213, BxPC3에 내부화(internalization)되는 것을 분석한 결과이다. 도 13a 및 도 13b에서 실선은 4℃에서 반응한 단일클론항체이고, 63-D7, 점선은 4℃에서의 반응 후 37℃에서 반응한 63-D7 항체이며, 음영 바탕은 2차 항체만 포함한 것이다. 도 13c는 도 13a와 동일한 실험을 3회 반복하여 각 암세포의 세포 표면에서 63-D7의 부착 정도를 상대적인 평균 형광세기로 통계적으로 처리하여 비교한 그래프이다. ****은 p<0.001, ***은 p<0.005, **은 p<0.01, *은 p<0.05인 것을 나타낸다. 도 13d는 도 13b와 동일한 실험을 3회 반복하여 각 암세포의 세포 표면에서 63-D7의 부착 정도를 상대적인 평균 형광세기로 통계적으로 처리하여 비교한 그래프이다. ****은 p<0.001, ***은 p<0.005, **은 p<0.01, *은 p<0.05인 것을 나타낸다.Figures 13a and 13b show the results of analyzing the internalization of monoclonal antibody 63-D7 into human liver cancer cells Huh7, HepG2, SNU449 and human pancreatic cancer cells SNU213 and BxPC3 by flow cytometry. In Figures 13a and 13b, the solid line is the monoclonal antibody and 63-D7 reacted at 4°C, the dotted line is the 63-D7 antibody reacted at 37°C after the reaction at 4°C, and the shaded background includes only the secondary antibody. Figure 13c is a graph comparing the degree of attachment of 63-D7 to the cell surface of each cancer cell by statistically processing it as the relative average fluorescence intensity by repeating the same experiment as Figure 13a three times. **** indicates p<0.001, *** indicates p<0.005, ** indicates p<0.01, and * indicates p<0.05. Figure 13d is a graph comparing the degree of attachment of 63-D7 to the cell surface of each cancer cell by statistically processing the relative average fluorescence intensity by repeating the same experiment as Figure 13b three times. **** indicates p<0.001, *** indicates p<0.005, ** indicates p<0.01, and * indicates p<0.05.
도 14a 및 도 14b는 63-D7 또는 생쥐 IgG isotype항체가 처리된 Huh7 세포의 세포 생존율을 측정한 결과이다. Huh7 세포에 63-D7 또는 생쥐 isotype항체를 각각 0 nM 내지 100 nM 처리한 후, 도 14a는 α-mFc-CL-DMDM를, 도 14b는 α-mFc-CL-MMAF를 48시간 동안 처리하여 세포 생존율은 CCK-8을 이용해 측정한 것이다. 도 14a 및 도 14b에서 *는 p<0.05, **는 p<0.01, ****는 p<0.001를 나타낸다.Figures 14a and 14b show the results of measuring the cell viability of Huh7 cells treated with 63-D7 or mouse IgG isotype antibody. After Huh7 cells were treated with 0 nM to 100 nM of 63-D7 or mouse isotype antibody, respectively, Figure 14a shows that they were treated with α-mFc-CL-DMDM and Figure 14b shows that they were treated with α-mFc-CL-MMAF for 48 hours, and the cell viability was measured using CCK-8. In Figures 14a and 14b, * indicates p<0.05, ** indicates p<0.01, and **** indicates p<0.001.
도 15a은 간암 환자 혈액에서 본 발명의 PTGFRN 특이적인 단일클론항체 63-D7에 의해 인식된 혈중 암세포를 검출하는 방법을 나타낸 그림으로, CD45 양성 세포들을 제거하고 남은 세포를 Dylight488 접합된 63-D7과 다른 마커(항체)로 염색하는 방법을 나타낸 것이다. 도 15b는 PBMC에 Huh7세포를 0, 10, 30, 50, 100개씩 넣고 15a에 도시된 방법과 같이 CD45 양성 세포 제거 후 회수된 Huh7세포를 세어 회수율을 측정한 그래프이다. 도 15c는 도 15a에 도시된 방법과 같이 회수한 혈중 암세포를 Dylight649 접합된 항-마우스 IgG 및 Dylight 488 접합된 63-D7과 함께 염색을 수행한 것이며, 핵을 염색한 DAPI, 그리고 이들을 병합한 Merge를 도시한 것이다.Fig. 15a is a drawing showing a method for detecting blood cancer cells recognized by the PTGFRN-specific monoclonal antibody 63-D7 of the present invention in the blood of a liver cancer patient, and shows a method for removing CD45 positive cells and staining the remaining cells with Dylight488-conjugated 63-D7 and another marker (antibody). Fig. 15b is a graph showing the recovery rate measured by counting the Huh7 cells recovered after removing CD45 positive cells as shown in 15a by adding 0, 10, 30, 50, and 100 Huh7 cells to PBMC. Fig. 15c shows the blood cancer cells recovered as shown in Fig. 15a, stained with Dylight649-conjugated anti-mouse IgG and Dylight 488-conjugated 63-D7, and DAPI that stains the nucleus, and Merge that merges them.
도 16a 내지 도 16c는 간암 환자 혈액에서 본 발명의 PTGFRN 특이적인 단일클론항체 63-D7과 항-MVP, 항-HSA, 항-PanCK, 항-E-cadherin, 항-EpCAM, 63-D7, 항-Vimentin, 항-Twist, 항-ZEB1 항체가 이중 염색된 혈중 암세포를 도시한 공초점 현미경 이미지이며, 핵을 염색한 DAPI, 그리고 이들을 병합한 Merge를 도시한 것이다.FIGS. 16A to 16C are confocal microscopy images showing blood cancer cells double-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention and anti-MVP, anti-HSA, anti-PanCK, anti-E-cadherin, anti-EpCAM, 63-D7, anti-Vimentin, anti-Twist, and anti-ZEB1 antibodies in the blood of a liver cancer patient, and DAPI that stains the nucleus and Merge that merges them.
도 17은 간암 환자 혈액(primary HCC 53명 및 metastatic HCC 42명)과 정상인 및 간염환자(non-neoplastic 26명) 혈액에서 본 발명의 단일클론항체 63-D7에 의해 인식된 모든 혈중 암세포 수(63-D7+ cells/ml)를 세 그룹 간 비교 검정(Kruskal-Wallis Test)을 통해 비교한 그래프로(p value <0.0001), 각 그룹 간에 p-value가 모두 <0.0001으로 세 그룹 간 차이에서 모두 유의성이 있음을 도시한 것이다.Figure 17 is a graph comparing the number of all blood cancer cells (63-D7+ cells/ml) recognized by the monoclonal antibody 63-D7 of the present invention in the blood of liver cancer patients (53 primary HCC and 42 metastatic HCC) and normal people and hepatitis patients (26 non-neoplastic) through a comparative test (Kruskal-Wallis Test) among the three groups (p value <0.0001). It shows that the p-values for each group are all <0.0001, indicating that there is significance in the differences among all three groups.
도 18a는 간암 환자 혈액에서 본 발명의 PTGFRN 특이적인 단일클론항체 63-D7, 항-EpCAM, 항-Vimentin 항체로 삼중 염색된 혈중 암세포를 도시한 공초점 현미경 이미지이며, 핵을 염색한 DAPI, 그리고 이들을 병합한 Merge를 함께 도시한 것이다. BF는 명시야(bright field)를 의미한다. 도 18b는 간암 환자 혈액에서 본 발명의 PTGFRN 특이적인 단일클론항체 63-D7, 항-MVP, 항-B7-H3 항체로 삼중 염색된 혈중 암세포를 도시한 공초점 현미경 이미지이며, 핵을 염색한 DAPI, 그리고 이들을 병합한 Merge를 함께 도시한 것이다. BF는 bright field를 말하며 명시야를 의미한다. BF는 명시야(bright field)를 의미한다.FIG. 18a is a confocal microscope image showing circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-EpCAM, and anti-Vimentin antibodies in the blood of a liver cancer patient, together with DAPI, which stains the nucleus, and a Merge, which merges them. BF stands for bright field. FIG. 18b is a confocal microscope image showing circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-MVP, and anti-B7-H3 antibodies in the blood of a liver cancer patient, together with DAPI, which stains the nucleus, and a Merge, which merges them. BF stands for bright field. BF stands for bright field.
도 19는 간암 환자 혈액에서 본 발명의 PTGFRN 특이적인 단일클론항체 63-D7, 항-TGFβR1, 항-B7-H3 항체로 삼중 염색된 혈중 암세포를 도시한 공초점 현미경 이미지이며, 핵을 염색한 DAPI, 그리고 이들을 병합한 Merge를 함께 도시한 것이다. BF는 명시야(bright field)를 의미한다.Figure 19 is a confocal microscope image showing blood cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-TGFβR1, and anti-B7-H3 antibodies in the blood of a liver cancer patient, along with DAPI that stains the nucleus and a Merge that combines them. BF stands for bright field.
도 20a는 간암 환자 혈액에서 본 발명의 PTGFRN 특이적인 단일클론항체 63-D7, 항-ULBP1, 항-B7-H3 항체로 삼중 염색된 혈중 암세포를 도시한 공초점 현미경 이미지이며, 핵을 염색한 DAPI, 그리고 이들을 병합한 Merge를 함께 도시한 것이다. BF는 명시야(bright field)를 의미한다. 도 20b는 간암 환자 혈액에서 본 발명의 PTGFRN 특이적인 단일클론항체 63-D7, 항-MICA/B, 항-B7-H3 항체로 삼중 염색된 혈중 암세포를 도시한 공초점 현미경 이미지이며, 핵을 염색한 DAPI, 그리고 이들을 병합한 Merge를 함께 도시한 것이다. BF는 명시야(bright field)를 의미한다.FIG. 20a is a confocal microscope image of circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-ULBP1, and anti-B7-H3 antibodies in the blood of a liver cancer patient, together with DAPI, which stains the nucleus, and a Merge, which merges them. BF stands for bright field. FIG. 20b is a confocal microscope image of circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-MICA/B, and anti-B7-H3 antibodies in the blood of a liver cancer patient, together with DAPI, which stains the nucleus, and a Merge, which merges them. BF stands for bright field.
도 21a는 간암 환자 혈액에서 본 발명의 PTGFRN 특이적인 단일클론항체 63-D7, 항-PD-L1, 항-B7-H3 항체로 삼중 염색된 혈중 암세포를 도시한 공초점 현미경 이미지이며, 핵을 염색한 DAPI, 그리고 이들을 병합한 Merge를 함께 도시한 것이다. BF는 명시야(bright field)를 의미한다. 도 21b는 간암 환자 혈액에서 본 발명의 PTGFRN 특이적인 단일클론항체 63-D7, 항-PD-L2, 항-B7-H3 항체로 삼중 염색된 혈중 암세포를 도시한 공초점 현미경 이미지이며, 핵을 염색한 DAPI, 그리고 이들을 병합한 Merge를 함께 도시한 것이다. BF는 명시야(bright field)를 의미한다.FIG. 21a is a confocal microscope image showing circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-PD-L1, and anti-B7-H3 antibodies in the blood of a liver cancer patient, together with DAPI that stains the nucleus and a Merge that merges them. BF stands for bright field. FIG. 21b is a confocal microscope image showing circulating cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-PD-L2, and anti-B7-H3 antibodies in the blood of a liver cancer patient, together with DAPI that stains the nucleus and a Merge that merges them. BF stands for bright field.
도 22는 간암 환자 혈액에서 본 발명의 PTGFRN 특이적인 단일클론항체 63-D7, 항-CD47, 항-B7-H3 항체로 삼중 염색된 혈중 암세포를 도시한 공초점 현미경 이미지이며, 핵을 염색한 DAPI, 그리고 이들을 병합한 Merge를 함께 도시한 것이다. BF는 명시야(bright field)를 의미한다.Figure 22 is a confocal microscope image showing blood cancer cells triple-stained with the PTGFRN-specific monoclonal antibody 63-D7 of the present invention, anti-CD47, and anti-B7-H3 antibodies in the blood of a liver cancer patient, along with DAPI that stains the nucleus and a Merge that combines them. BF stands for bright field.
도 23은 단일클론항체 63-D7 중쇄 유전자 가변영역의 염기서열과 아미노산 서열을 표시한 것으로 항원과 결합하는 CDR(Complementarity Determining Region)를 진한 글씨로 표시한 그림이다. Figure 23 shows the base sequence and amino acid sequence of the heavy chain gene variable region of the monoclonal antibody 63-D7, with the CDR (Complementarity Determining Region) that binds to the antigen indicated in bold.
도 24는 단일클론항체 63-D7 경쇄 유전자 가변영역의 염기서열과 아미노산 서열을 표시한 것으로 항원과 결합하는 CDR(Complementarity Determining Region)를 진한 글씨로 표시한 그림이다.Figure 24 shows the base sequence and amino acid sequence of the light chain gene variable region of the monoclonal antibody 63-D7, with the CDR (Complementarity Determining Region) that binds to the antigen indicated in bold.
본 발명은 혈중 암세포 수준 판단, 암 치료 또는 예방, 암 전이 예후 예측 및 항암제 스크리닝을 위한 항 PTGFRN 단일클론항체를 제공한다.The present invention provides an anti-PTGFRN monoclonal antibody for determining the level of blood cancer cells, treating or preventing cancer, predicting the prognosis of cancer metastasis, and screening for anticancer drugs.
본 발명은 서열번호 3의 아미노산 서열로 이루어진 HCDR1, 서열번호 4의 아미노산 서열로 이루어진 HCDR2 및 서열번호 5의 아미노산 서열로 이루어진 HCDR3을 포함하는 중쇄 가변영역; 및 서열번호 6의 아미노산 서열로 이루어진 LCDR1, 서열번호 7의 아미노산 서열로 이루어진 LCDR2 및 서열번호 8의 아미노산 서열로 이루어진 LCDR3을 포함하는 경쇄 가변영역을 포함하는 항 PTGFRN 단일클론항체를 제공한다.The present invention provides an anti-PTGFRN monoclonal antibody comprising a heavy chain variable region comprising HCDR1 consisting of the amino acid sequence of SEQ ID NO: 3, HCDR2 consisting of the amino acid sequence of SEQ ID NO: 4, and HCDR3 consisting of the amino acid sequence of SEQ ID NO: 5; and a light chain variable region comprising LCDR1 consisting of the amino acid sequence of SEQ ID NO: 6, LCDR2 consisting of the amino acid sequence of SEQ ID NO: 7, and LCDR3 consisting of the amino acid sequence of SEQ ID NO: 8.
본 명세서에서 "항체"라는 용어는 완전한 항체 및 이의 임의의 항원 결합 단편(즉, "항원 결합 부위") 또는 단일쇄를 포함한다. 항체란 다이설파이드 결합에 의해 서로 연결된 적어도 2개의 중쇄(H) 및 2개의 경쇄(L)를 포함하는 당단백질, 또는 그 항원 결합 부분을 말한다.The term "antibody" as used herein includes a complete antibody and any antigen-binding fragment (i.e., "antigen-binding portion") or single chain thereof. An antibody refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains joined to each other by disulfide bonds, or an antigen-binding portion thereof.
각각의 중쇄는 중쇄 가변 영역(본 명세서에서 VH로 약칭함) 및 중쇄 불변 영역으로 이루어진다. 상기 중쇄 불변 영역은 3개의 도메인, 즉 CH1, CH2 및 CH3으로 이루어진다. 각각의 경쇄는 경쇄 가변 영역(본 명세서에서 VL로 약칭함) 및 경쇄 불변 영역으로 이루어진다. 상기 경쇄 불변 영역은 1개의 도메인, 즉 CL로 이루어진다. 상기 VH 영역 및 VL 영역은 골격구조 영역(FR)이라 불리는 더 보존된 영역이 산재되어 있는 상보성 결정 영역(CDR)이라 불리는 과가변 영역으로 추가로 세분될 수 있다.Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2, and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into hypervariable regions, called complementarity determining regions (CDRs), interspersed with more conserved regions, called framework regions (FRs).
각각의 VH 및 VL은 아미노 말단에서 카복시 말단으로 하기 순서로 배열되는 3개의 CDR 및 4개의 FR, 즉 FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4로 이루어진다. 상기 중쇄 및 경쇄의 가변 영역은 항원과 상호작용하는 결합 도메인을 포함한다. 상기 항체의 불변 영역은 숙주 조직 또는 인자[면역계의 각종 세포(예를 들어, 이펙터 세포) 및 전형적 보체계의 제1 성분(Clq)을 포함함]에 대한 면역글로불린의 결합을 매개할 수 있다.Each VH and VL is composed of three CDRs and four FRs, namely FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4, arranged in the following order from amino terminus to carboxy terminus. The variable regions of the heavy and light chains comprise a binding domain that interacts with an antigen. The constant regions of the antibody can mediate binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
VH 영역에 포함되는 CDR은 중쇄 상보성 결정영역1((heavy chain complementary determine region 1, HCDR1), HCDR2 및 HCDR3라 하고, VL 영역에 포함되는 CDR은 경쇄 상보성 결정영역 1(light chain complementary determine region 1, LCDR1), LCDR2 및 LCDR3라 한다.The CDRs included in the VH region are called heavy chain complementary determining region 1 (HCDR1), HCDR2, and HCDR3, and the CDRs included in the VL region are called light chain complementary determining region 1 (LCDR1), LCDR2, and LCDR3.
본 명세서에서 항체의 "항원 결합 부위"(또는 간단히 "항체 일부분")라는 용어는 항원(예를 들어, PTGFRN)에 특이적으로 결합하는 능력을 보유하는 항체의 하나 이상의 단편을 의미한다. 항체의 항원 결합 기능은 전체 길이 항체의 단편에 의해서도 발휘될 수 있는 것으로 확인되었다.The term "antigen binding portion" of an antibody (or simply "antibody portion") as used herein refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., PTGFRN). It has been determined that the antigen binding function of an antibody can also be exerted by fragments of full-length antibodies.
항체의 "항원 결합 부분"이란 용어에 포함되는 결합 단편의 예로는 (i) Fab 단편, 즉 VL, VH, CL 및 CH1 도메인으로 구성되는 1가 단편; (ii) F(ab')2 단편, 즉 힌지 영역에서 다이설파이드 가교에 의해 연결된 2개의 Fab 단편을 포함하는 2가 단편; (iii) VH 및 CH1 도메인으로 구성되는 Fd 단편; (iv) 항체의 한쪽 팔의 VL 및 VH 도메인으로 구성되는 Fv 단편; (v) VH 도메인으로 구성되는 dAb 단편[Ward et al., (1989) Nature 341:544-546]; 및 (vi) 분리된 상보성 결정 영역(CDR) 또는 (vii) 경우에 따라 합성 링커에 의해 연결될 수 있는 2개 이상의 분리된 CDR의 조합을 포함한다. 또한, Fv 단편의 2개의 도메인, 즉 VL 및 VH는 별개의 유전자에 의해 코딩되지만, 재조합법을 이용하여, VL 영역과 VH 영역이 쌍을 이루어 1가 분자[단일쇄 Fv(scFv)라 함]를 형성한 단일 단백질쇄로 만들 수 있는 합성 링커에 의해 이들을 연결할 수 있다. 이러한 단일쇄 항체도 또한 항체의 "항원 결합 부위"이라는 용어에도 포함되는 것으로 한다. 이러한 항체 단편은 당업자에게 알려진 통상의 기법을 이용하여 얻을 수 있으며, 이 단편을 완전한 항체와 동일한 방식으로 유용성이 있는지에 대해 스크리닝한다. 항원 결합 부분은 재조합 DNA 기법, 또는 완전한 면역글로불린의 효소적 또는 화학적 절단에 의해 제조할 수 있다.Examples of binding fragments encompassed by the term "antigen-binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of one arm of an antibody; (v) a dAb fragment consisting of a VH domain [Ward et al., (1989) Nature 341:544-546]; and (vi) an isolated complementarity determining region (CDR) or (vii) a combination of two or more isolated CDRs, which may, in some cases, be linked by a synthetic linker. Also, although the two domains of the Fv fragment, namely the VL and VH, are encoded by separate genes, they can be joined by a synthetic linker, which can be prepared by recombinant methods, into a single protein chain in which the VL and VH regions pair to form a monovalent molecule (called single-chain Fv (scFv)). Such single-chain antibodies are also included in the term "antigen-binding portion" of an antibody. Such antibody fragments can be obtained by conventional techniques known to those skilled in the art, and these fragments are screened for utility in the same manner as complete antibodies. The antigen-binding portion can be prepared by recombinant DNA techniques, or by enzymatic or chemical cleavage of complete immunoglobulins.
본 명세서에서 항체는 단일클론항체 또는 다클론 항체일 수 있다. 항체는 당업계에 공지된 인간화 항체의 제조를 위한 다양한 기법을 이용하여 제조할 수 있다.The antibody herein may be a monoclonal antibody or a polyclonal antibody. The antibody may be produced using various techniques known in the art for producing humanized antibodies.
본 명세서에서 "단일클론항체"라는 용어는 단일 결합 특이성을 나타내고 특정 에피토프에 대해 친화성을 나타내는 항체를 말한다.The term "monoclonal antibody" as used herein refers to an antibody that exhibits a single binding specificity and exhibits affinity for a particular epitope.
본 명세서에서 PTGFRN 특이적인 단일클론항체는 효율적으로 사실상 무한한 양으로 매우 정제된 형태로 제조될 수 있다. PTGFRN 특이적인 단일클론항체는 PTGFRN의 특정 에피토프에 특이적으로 결합할 수 있다.In the present disclosure, PTGFRN-specific monoclonal antibodies can be efficiently produced in virtually unlimited quantities in a highly purified form. PTGFRN-specific monoclonal antibodies can specifically bind to a specific epitope of PTGFRN.
본 명세서에서 PTGFRN에 결합하는 항체로, 중쇄 트랜스진 및 경쇄 트랜스진을 포함하는 게놈을 갖는 비인간화 동물로부터 얻은 불멸화된 세포에 융합된 B세포의 바이러스 또는 암 유전자 형질전환, 인간 항체 유전자의 라이브러리를 이용한 파지 디스플레이 기법, 체세포 하이브리드화 기법 등을 사용하여 제조할 수 있다. 단일클론항체를 생산하기 위한, 면역화 프로토콜 및 면역화된 비장 세포의 단리 및 융합 기법을 비롯한 동물계에서의 하이브리도마 생산 방법은 당업계에 잘 알려져 있다.The antibodies herein that bind to PTGFRN can be produced by viral or oncogene transformation of B cells fused to immortalized cells obtained from a nonhumanized animal having a genome comprising a heavy chain transgene and a light chain transgene, phage display techniques using a library of human antibody genes, somatic cell hybridization techniques, etc. Methods for producing hybridomas in animal systems, including immunization protocols and isolation and fusion techniques of immunized spleen cells, for producing monoclonal antibodies are well known in the art.
항체 또는 이의 항원 결합 단편은 방사종(radionuclides), 형광원(fluorescors) 및 효소(enzymes) 등에 의해 표지화될 수 있다.Antibodies or antigen-binding fragments thereof can be labeled with radionuclides, fluorescors, enzymes, etc.
본 명세서에서 "에피토프" 또는 "항원 결정 부위"라는 용어는 면역글로불린 또는 항체가 특이적으로 결합하는 항원의 한 부위를 말한다. 에피토프는 인접된 아미노산으로부터 형성될 수도 있고 단백질의 3차 폴딩에 의해 병치된 비인접 아미노산으로부터 형성될 수도 있다. 인접된 아미노산으로부터 형성된 에피토프는 일반적으로 변성 용매에 노출되어도 유지되는 반면, 3차 폴딩에 의해 형성된 에피토프는 변성 용매로 처리시 일반적으로 상실된다. 에피토프는 전형적으로 독특한 공간적 입체구조로 적어도 3개, 4개, 5개, 6개, 7개, 8개, 9개, 10개, 11개, 12개, 13개, 14개 또는 15개의 아미노산을 포함한다. 에피토프의 공간적 입체구조를 결정하는 방법으로는 당업계에 공지된 기법 및 본 명세서에 기재된 기법, 예를 들어 x선 결정학적 분석 및 2차원 핵 자기 공명을 들 수 있다. 이와 관련하여, 예를 들어 문헌[Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G. E. Morris, Ed. (1996)]을 참조할 수 있다. 바람직한 실시형태에서, 상기 에피토프(또는 항원 결정 부위)는, 더 큰 아미노산 분절(예를 들어, 3차원 구조를 갖는 단백질)에 내에 있을 때에도 에피토프/항원 결정 부위에 대한 친화성의 추가적인 증가가 관찰되지 않는다는 점에서 입체 에피토프로서 기능한다.The term "epitope" or "antigenic determining portion" as used herein refers to a portion of an antigen to which an immunoglobulin or antibody specifically binds. Epitopes may be formed from adjacent amino acids or from non-adjacent amino acids that are juxtaposed by tertiary folding of the protein. Epitopes formed from adjacent amino acids are generally retained upon exposure to denaturing solvents, whereas epitopes formed by tertiary folding are generally lost upon treatment with denaturing solvents. Epitopes typically comprise at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acids in a unique spatial conformation. Methods for determining the spatial conformation of an epitope include techniques known in the art and techniques described herein, such as x-ray crystallography and two-dimensional nuclear magnetic resonance. In this regard, reference may be made, for example, to the literature [Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G. E. Morris, Ed. (1996)]. In a preferred embodiment, the epitope (or antigenic determinant site) functions as a conformational epitope in that no additional increase in affinity for the epitope/antigenic determinant site is observed even when it is within a larger amino acid segment (e.g., a protein having a three-dimensional structure).
본 명세서에서 사용되는 "단위형(isotype)"이란 용어는 중쇄 불변 영역 유전자에 의해 코딩되는 항체 클래스(예를 들어, IgM 또는 IgG1)를 말한다. 일 실시형태에서, 본 발명의 단일클론항체는 IgG1 단위형이다.The term "isotype" as used herein refers to the class of antibodies (e.g., IgM or IgG1) encoded by the heavy chain constant region genes. In one embodiment, the monoclonal antibodies of the invention are of the IgG1 isotype.
본 발명은 인간 배아줄기세포 세포 표면에 결합하는 단일클론항체를 제조하고, 제조한 항체 중 인간 배아줄기세포, 배아암세포 및 다양한 암세포 표면에 결합하면서, 정상세포인 인간 말초 단핵구 세포 또는 인간 간세포에는 결합하지 않는 단일클론항체 63-D7의 항원을 탐색하였다.The present invention produces a monoclonal antibody that binds to the surface of human embryonic stem cells, and searches for an antigen of monoclonal antibody 63-D7, which binds to the surface of human embryonic stem cells, embryonic cancer cells, and various cancer cells, but does not bind to normal cells such as human peripheral mononuclear cells or human hepatocytes.
본 발명에서 63-D7 단일클론항체가 세포표면 단백질 PTGFRN(Prostaglandin F2 Receptor Negative Regulator, CD315)를 인식함을 밝혔다. 또한, 63-D7 단일클론항체가 인식하는 PTGFRN는 간암, 폐암, 대장암, 췌장암 및 갑상선암 등의 세포주에서 발현됨을 확인하였다.In the present invention, it was revealed that the 63-D7 monoclonal antibody recognizes the cell surface protein PTGFRN (Prostaglandin F2 Receptor Negative Regulator, CD315). In addition, it was confirmed that PTGFRN recognized by the 63-D7 monoclonal antibody is expressed in cell lines such as liver cancer, lung cancer, colon cancer, pancreatic cancer, and thyroid cancer.
본 발명에서 간세포암 환자 95명 대상으로 실험한 결과 63-D7 단일클론항체가 97%의 효율로 혈중 암세포를 검출할 수 있음을 확인하였다. 63-D7 항체는 EMT 마커인 Vimentin과 1차 HCC에서 약 53%, 2차 HCC에서 약 28%의 비율로 함께 발현되므로, 63-D7 항체를 EMT 표현형을 나타내는 혈중 암세포 검출에 사용할 수 있음을 확인하였다. 또한 63-D7이 인식하는 혈중 암세포는 EpCAM과 Vimentin 모두를 발현하지 않는 중간 형태의 혈중 암세포도 검출할 수 있으며, 구체적으로 1차 HCC에서 약 46%, 2차 HCC에서 약 72%의 비율로 발현이 확인되었다.In the present invention, the results of an experiment on 95 patients with hepatocellular carcinoma confirmed that the 63-D7 monoclonal antibody can detect circulating cancer cells with an efficiency of 97%. Since the 63-D7 antibody is co-expressed with Vimentin, an EMT marker, at a ratio of about 53% in primary HCC and about 28% in secondary HCC, it was confirmed that the 63-D7 antibody can be used to detect circulating cancer cells showing an EMT phenotype. In addition, circulating cancer cells recognized by 63-D7 can also detect intermediate circulating cancer cells that do not express either EpCAM or Vimentin, and specifically, it was confirmed that expression was found at a ratio of about 46% in primary HCC and about 72% in secondary HCC.
따라서, 63-D7 항체는 기존의 EpCAM 마커 기반 혈중 암세포 진단 기술로는 확인할 수 없던 중간 형태 표현형 및 EMT 표현형의 혈중 암세포를 모두 검출해낼 수 있는 새로운 마커임을 알 수 있다.Therefore, it can be seen that the 63-D7 antibody is a new marker that can detect both intermediate phenotype and EMT phenotype circulating cancer cells that could not be identified by existing EpCAM marker-based circulating cancer cell diagnostic technology.
PTGFRN(Prostaglandin F2 Receptor Negative Regulator, EWI-F, CD9P-1, CD315)은 Immunoglobulin superfamily로 Ⅰ형 막관통 단백질(transmembrane protein)으로, Prostaglandin F2 Receptor에 음성적으로 작용한다. 본 명세서에서 PTGFRN은 전장 서열, 동등한 기능을 수행하는 단편 또는 PTGFRN의 연속된 아미노산 서열(예컨대, 도 3c에 개시된 PTGFRN 아미노산 서열)을 갖는 폴리펩타이드를 포함한다.PTGFRN (Prostaglandin F2 Receptor Negative Regulator, EWI-F, CD9P-1, CD315) is a type I transmembrane protein of the immunoglobulin superfamily, which negatively affects the Prostaglandin F2 Receptor. In the present specification, PTGFRN includes a full-length sequence, a fragment that performs an equivalent function, or a polypeptide having a continuous amino acid sequence of PTGFRN (e.g., the PTGFRN amino acid sequence disclosed in FIG. 3c).
PTGFRN 은 척추동물, 더 구체적으로, 인간 또는 마우스 유래 PTGFRN 일 수 있으나, 이에 특별히 제한하는 것은 아니다.PTGFRN may be, but is not limited to, a PTGFRN derived from a vertebrate, more specifically, a human or mouse.
본 발명은 중쇄 가변영역은 서열번호 1의 아미노산 서열을 포함하는 항 PTGFRN 단일클론항체를 제공한다.The present invention provides an anti-PTGFRN monoclonal antibody, wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 1.
본 발명의 항 PTGFRN 단일클론항체 클론 63-D7에 의해 생성된 중쇄 가변 영역 아미노산 서열(서열번호 1)과 적어도 80%, 85%, 90%, 95%, 98%, 99% 또는 그 이상 동일한 아미노산 서열을 포함하는 단일쇄 가변영역(scFv)을 포함하는 항체일 수 있다.The present invention may be an antibody comprising a single chain variable region (scFv) comprising an amino acid sequence that is at least 80%, 85%, 90%, 95%, 98%, 99% or more identical to the heavy chain variable region amino acid sequence (SEQ ID NO: 1) produced by the anti-PTGFRN monoclonal antibody clone 63-D7 of the present invention.
본 발명은 경쇄 가변영역은 서열번호 2의 아미노산 서열을 포함하는 항 PTGFRN 단일클론항체를 제공한다.The present invention provides an anti-PTGFRN monoclonal antibody, wherein the light chain variable region comprises the amino acid sequence of SEQ ID NO: 2.
본 발명의 항 PTGFRN 단일클론항체는 클론 63-D7에 의해 생성된 경쇄 가변 영역 아미노산 서열(서열번호 2)과 적어도 80%, 85%, 90%, 95%, 98%, 99% 또는 그 이상 동일한 아미노산 서열을 포함하는 단일쇄 가변영역(scFv)을 포함하는 항체일 수 있다.The anti-PTGFRN monoclonal antibody of the present invention may be an antibody comprising a single chain variable region (scFv) comprising an amino acid sequence that is at least 80%, 85%, 90%, 95%, 98%, 99% or more identical to the light chain variable region amino acid sequence (SEQ ID NO: 2) produced by clone 63-D7.
본 발명은 본 발명의 PTGFRN 단일클론항체에 약물이 접합된 항체-약물 컨쥬게이트(antibody-drug conjugate, ADC)를 제공한다.The present invention provides an antibody-drug conjugate (ADC) in which a drug is conjugated to the PTGFRN monoclonal antibody of the present invention.
본 발명의 단일클론항체는 다양한 암세포에서 PTGFRN의 세포 내부화를 촉진할 수 있으므로, 항체-약물 컨쥬게이트에 사용되는 항체로써 적합하다.The monoclonal antibody of the present invention can promote cellular internalization of PTGFRN in various cancer cells, and is therefore suitable as an antibody used in antibody-drug conjugates.
항체-약물 컨쥬게이트에서 약물은 종류에 제한 없이 목적에 따라 당업자에 의해 선택될 수 있다. 예를 들어, 공지의 항암 약물일 수 있으나, 이에 제한되는 것은 아니다. 또한, 상기 컨쥬게이트하는 방식은 항체와 약물을 접할시킬 수 있는 것이라면 당업계 공지의 방법으로 제한 없이 선택될 수 있다.In the antibody-drug conjugate, the drug can be selected by those skilled in the art according to the purpose without limitation in type. For example, it can be a known anticancer drug, but is not limited thereto. In addition, the method of conjugation can be selected without limitation by a method known in the art as long as it can bring the antibody and the drug into contact.
본 발명은 본 발명의 항 PTGFRN 단일클론항체를 포함하는 암의 치료 또는 예방용 약학 조성물을 제공한다. The present invention provides a pharmaceutical composition for treating or preventing cancer comprising the anti-PTGFRN monoclonal antibody of the present invention.
본 발명은 본 발명의 항 PTGFRN 단일클론항체를 포함하는 암의 치료, 예방 또는 진단 방법을 제공한다.The present invention provides a method for treating, preventing or diagnosing cancer comprising the anti-PTGFRN monoclonal antibody of the present invention.
본 명세서에서 암은 뇌척수종양, 두경부암, 폐암, 유방암, 흉선종, 중피종, 식도암, 위암, 대장암, 간암, 갑상선암, 췌장암, 담도암, 신장암, 방광암, 전립선암, 고환암, 생식세포종, 난소암, 자궁 경부암, 자궁 내막암, 림프종, 급성 백혈병, 만성 백혈병, 다발성 골수종, 육종, 악성 흑색종, 신경모세포종, 교모세포종 및 피부암으로 이루어진 군에서 선택되는 어느 하나일 수 있으나, 이에 제한되는 것은 아니다.In the present specification, cancer may be any one selected from the group consisting of brain and spinal tumors, head and neck cancer, lung cancer, breast cancer, thymoma, mesothelioma, esophageal cancer, stomach cancer, colon cancer, liver cancer, thyroid cancer, pancreatic cancer, biliary tract cancer, kidney cancer, bladder cancer, prostate cancer, testicular cancer, germ cell tumor, ovarian cancer, cervical cancer, endometrial cancer, lymphoma, acute leukemia, chronic leukemia, multiple myeloma, sarcoma, malignant melanoma, neuroblastoma, glioblastoma, and skin cancer, but is not limited thereto.
전이성 암은 암이 발병한 하나의 기관에서 림프절을 포함한 다른 기관으로 전이되는 암이다. 암이 발병한 기관에 존재하는 암은 원발암(primary cancer)이라 한다. 전이는 전이되는 조직에 따라 뇌 전이, 뼈 전이, 간 전이, 또는 폐 전이일 수 있다. 전이성 암은 악성 종양일 수 있다. 악성 종양은 뇌척수종양, 두경부암, 폐암, 유방암, 흉선종, 중피종, 식도암, 위암, 대장암, 간암, 췌장암, 담도암, 신장암, 방광암, 전립선암, 고환암, 생식세포종, 난소암, 자궁 경부암, 자궁 내막암, 림프종, 급성 백혈병, 만성 백혈병, 다발성 골수종, 육종, 악성 흑색종, 피부암, 또는 이들의 조합일 수 있다.Metastatic cancer is cancer that has spread from one organ to another, including the lymph nodes. The cancer present in the organ where the cancer has spread is called the primary cancer. Depending on the tissue to which it has spread, metastases can be brain metastases, bone metastases, liver metastases, or lung metastases. Metastatic cancer can be a malignant tumor. A malignant tumor can be a brain or spinal tumor, head and neck cancer, lung cancer, breast cancer, thymoma, mesothelioma, esophageal cancer, stomach cancer, colon cancer, liver cancer, pancreatic cancer, biliary tract cancer, kidney cancer, bladder cancer, prostate cancer, testicular cancer, germ cell tumor, ovarian cancer, cervical cancer, endometrial cancer, lymphoma, acute leukemia, chronic leukemia, multiple myeloma, sarcoma, malignant melanoma, skin cancer, or a combination of these.
본 발명의 단일클론항체는 상기 암의 아고니스틱 항체(agonistic antibody)로서, 암세포 표면의 PTGFRN를 자극하여 암세포의 이동과 침윤성을 촉진시킬 수 있다. 또한, 상기 단일클론항체는 원발암에서 암세포의 이동을 촉진하여 초기 원발암의 발생을 예방하거나 성장을 방해할 수 있다.The monoclonal antibody of the present invention is an agonistic antibody for the cancer, and can stimulate PTGFRN on the surface of cancer cells to promote the movement and invasiveness of cancer cells. In addition, the monoclonal antibody can promote the movement of cancer cells in primary cancer, thereby preventing the occurrence of early primary cancer or inhibiting its growth.
본 발명은 상기 항 PTGFRN 단일클론항체를 생산하는 하이브리도마를 제공한다.The present invention provides a hybridoma producing the above anti-PTGFRN monoclonal antibody.
본 명세서에서 하이브리도마는 하이브리도마 세포 또는 하이브리도마 세포주를 포함하는 것일 수 있다. 상기 하이브리도마는 2개의 다른 종류의 세포를 인공적으로 융합시켜 만든 세포로, 폴리에틸렌글리콜(Polyethylene glycol, PEG) 등 세포융합을 일으키게 하는 물질이나 어떤 종의 바이러스를 사용하여 둘 이상의 동종 세포나 이종세포가 융합되어, 각기 다른 세포가 갖는 다른 기능을 하나의 세포 속에 통합시킨 세포 또는 세포주를 의미한다. 단일클론항체를 분비하는 하이브리도마는 이를 시험관 내에서 또는 생체 내에서 대량으로 배양할 수 있다.In this specification, a hybridoma may include a hybridoma cell or a hybridoma cell line. The hybridoma is a cell created by artificially fusing two different types of cells, and refers to a cell or cell line in which two or more homologous or heterologous cells are fused using a substance that causes cell fusion, such as polyethylene glycol (PEG) or a certain type of virus, thereby integrating different functions of each cell into a single cell. A hybridoma that secretes a monoclonal antibody can be cultured in large quantities in vitro or in vivo.
본 발명은 본 발명의 항 PTGFRN 단일클론항체를 포함하는 PTGFRN 검출용 조성물을 제공한다.The present invention provides a composition for detecting PTGFRN comprising the anti-PTGFRN monoclonal antibody of the present invention.
상기 항원-항체 복합체는 검출 표지체(detection label)를 이용하여 검출할 수 있다. 예컨대, 효소, 형광물질, 리간드, 발광물질, 미세입자(microparticle), 레독스 분자 또는 방사선동위원소 등에서 선택될 수 있으며, 이에 특별히 제한하는 것은 아니다.The above antigen-antibody complex can be detected using a detection label. For example, the detection label can be selected from enzymes, fluorescent substances, ligands, luminescent substances, microparticles, redox molecules, or radioisotopes, but is not particularly limited thereto.
본 발명은 분리된 시료에 본 발명의 항 PTGFRN 단일클론항체를 처리하는 단계를 포함하는 암의 진단을 위한 정보 제공 방법을 제공한다.The present invention provides a method for providing information for diagnosing cancer, comprising the step of treating a separated sample with an anti-PTGFRN monoclonal antibody of the present invention.
본 명세서에서 개체는 인간을 포함하는 포유동물일 수 있다.In this specification, an entity may be a mammal, including a human.
본 명세서에서 생물학적 시료는 생물로부터 수득된 시료를 말한다. 생물학적 시료는 예를 들면 혈액, 혈장, 골수액, 림프액, 타액, 누액, 뇨, 점막액, 양수, 또는 이들의 조합일 수 있다.In this specification, a biological sample refers to a sample obtained from a living organism. The biological sample may be, for example, blood, plasma, bone marrow fluid, lymph, saliva, tears, urine, mucous membrane fluid, amniotic fluid, or a combination thereof.
본 명세서에서 PTGFRN에 특이적으로 결합하는 물질은 항-PTGFRN 항체 또는 이의 항원 결합 단편 또는 앱타머일 수 있다. 앱타머는 표적 분자에 결합하는 올리고핵산 또는 펩타이드를 말한다.In the present specification, the substance that specifically binds to PTGFRN may be an anti-PTGFRN antibody or an antigen-binding fragment thereof or an aptamer. An aptamer refers to an oligonucleic acid or peptide that binds to a target molecule.
본 발명은 개체로부터 분리된 시료에 본 발명의 항 PTGFRN 단일클론항체를 처리하는 단계를 포함하는 혈중 암세포 수준 판단에 필요한 정보 제공 방법을 제공한다.The present invention provides a method for providing information necessary for determining the level of blood cancer cells, comprising the step of treating a sample isolated from an individual with an anti-PTGFRN monoclonal antibody of the present invention.
예를 들어, 개체로부터 분리된 생물학적 시료와 본 발명의 항 PTGFRN 단일클론항체을 접촉시켜 상기 시료 중 혈중 암세포와 본 발명의 항 PTGFRN 단일클론항체를 결합시키는 단계; 반응 혼합물로부터 상기 혈중 암세포를 검출하는 단계; 및 상기 혈중 암세포가 검출된 경우 상기 개체를 전이성 암에 걸렸거나 걸릴 확률이 높은 것으로 판별하는 단계를 포함하는 전이성 암 진단에 필요한 정보를 제공할 수 있다.For example, it can provide information necessary for diagnosing metastatic cancer, including the steps of contacting a biological sample isolated from an individual with the anti-PTGFRN monoclonal antibody of the present invention to bind blood cancer cells in the sample to the anti-PTGFRN monoclonal antibody of the present invention; detecting the blood cancer cells from the reaction mixture; and determining, if the blood cancer cells are detected, that the individual has or is likely to have metastatic cancer.
본 발명의 항 PTGFRN 단일클론항체는 PTGFRN에 특이적으로 결합할 수 있으므로, PTGFRN을 검출할 수 있고, 또한, 상기 조성물을 이용하여 PTGFRN의 분리 또는 정제가 가능하다. 본 발명의 항 PTGFRN 단일클론항체를 시료와 접촉시켜 항원-항체 복합체 형성을 검출함으로써 PTGFRN 단백질을 분리, 정체 또는 검출할 수 있다.Since the anti-PTGFRN monoclonal antibody of the present invention can specifically bind to PTGFRN, PTGFRN can be detected, and furthermore, PTGFRN can be separated or purified using the composition. By contacting the anti-PTGFRN monoclonal antibody of the present invention with a sample and detecting the formation of an antigen-antibody complex, the PTGFRN protein can be separated, identified, or detected.
항원-항체 복합체란 시료 중의 PTGFRN의 존재 또는 부재를 확인하기 위한 상기 PTGFRN와 이를 인지하는 단일클론항체의 결합물을 의미한다. 항원-항체 복합체는 검출 표지체(detection label)를 이용하여 검출할 수 있다. 예컨대, 효소, 형광물질, 리간드, 발광물질, 미세입자(microparticle), 레독스 분자 또는 방사선동위원소 등에서 선택될 수 있으며, 이에 특별히 제한하는 것은 아니다.The antigen-antibody complex refers to a combination of PTGFRN and a monoclonal antibody that recognizes it for the purpose of confirming the presence or absence of PTGFRN in a sample. The antigen-antibody complex can be detected using a detection label. For example, the label can be selected from an enzyme, a fluorescent substance, a ligand, a luminescent substance, a microparticle, a redox molecule, or a radioisotope, but is not particularly limited thereto.
항원-항체 복합체의 형성은 비색법(colorimetric method), 전기화학법(electrochemical method), 형광법(fluorimetric method), 발광법(luminometry), 입자계수법(particle counting method), 육안측정법(visual assessment), 또는 섬광계수법(scintillation counting method) 등을 사용하여 검출할 수 있다. 예컨대, 유세포 분석법(flow cytometry), 면역세포화학법, 방사능면역분석법(RIA), 효소면역측정법(ELISA), 웨스턴 블롯팅(Western Blotting), 면역침강 분석법(Immunoprecipitation Assay), 면역확산 분석법(Immunodiffusion assay), 보체 고정 분석법(Complement Fixation Assay), 단백질 칩(protein chip) 등에 의해 검출할 수 있다. 보다 구체적으로, 소량의 목적 단백질을 회수하는데 유용한 면역침강법(immune precipitation) 및 면역블롯팅을 통해 검출할 수 있다.The formation of antigen-antibody complexes can be detected using a colorimetric method, an electrochemical method, a fluorescence method, a luminometry method, a particle counting method, a visual assessment, or a scintillation counting method. For example, it can be detected by flow cytometry, immunocytochemistry, radioimmunoassay (RIA), enzyme-linked immunosorbent assay (ELISA), Western blotting, immunoprecipitation assay, immunodiffusion assay, complement fixation assay, protein chip, etc. More specifically, it can be detected by immune precipitation and immunoblotting, which are useful for recovering a small amount of target protein.
구체적으로, 항원-항체 복합체를 효소면역측정법(ELISA, enzyme-linked immunosorbent assay)를 이용하여 검출할 수 있다. 상기 ELISA는 고체 지지체에 부착된 항원을 인지하는 표지된 항체를 이용하는 직접적 ELISA, 고체 지지체에 부착된 항원을 인지하는 항체의 복합체에서 포획 항체를 인지를 하는 표지된 항체를 이용하는 간접적 ELISA, 고체 지지체에 부착된 항체와 항원의 복합체에서 항원을 인지하는 표지된 또 다른 항체를 이용하는 직접적 샌드위치 ELISA, 고체 지지체에 부착된 항체와 항원의 복합체에서 항원을 인지하는 또 다른 항체와 반응시킨 후 이 항체를 인지하는 표지된 2차 항체를 이용하는 간접적 샌드위치 ELISA 등 다양한 ELISA 방법을 포함한다.Specifically, an antigen-antibody complex can be detected using an enzyme-linked immunosorbent assay (ELISA). The ELISA includes various ELISA methods, such as a direct ELISA using a labeled antibody that recognizes an antigen attached to a solid support, an indirect ELISA using a labeled antibody that recognizes a capture antibody in a complex of antibodies that recognize the antigen attached to the solid support, a direct sandwich ELISA using another labeled antibody that recognizes an antigen in a complex of an antibody and an antigen attached to the solid support, and an indirect sandwich ELISA using a labeled secondary antibody that recognizes the antibody after reacting with another antibody that recognizes the antigen in a complex of an antibody and an antigen attached to the solid support.
혈중 암세포(Circulating tumor cell, CTC)는 종양 침윤(tumor invasion) 과정을 거쳐 혈액 중에 존재하여 체내를 순환하는 희소의 종양세포이다. 혈중 암세포는 암의 전이 및 재발에 관여하는 인자로 알려져 있다. 혈중 암세포는 상피-중간엽 전이된 혈중 암세포일 수 있다.Circulating tumor cells (CTCs) are rare tumor cells that exist in the blood and circulate in the body after undergoing a tumor invasion process. Circulating tumor cells are known to be a factor involved in cancer metastasis and recurrence. Circulating tumor cells may be circulating tumor cells that have undergone epithelial-mesenchymal transition.
상피-중간엽 전이(epithelial-mesenchymal transition, EMT)는 상피 세포가 세포 극성 및 세포간 부착을 상실하고 중간엽 세포로 전환되어 이동성 및 침윤성 세포가 되는 과정이다. 상피-중간엽 전이는 중배엽 형성 및 신경관 형성을 포함하는 많은 발달 과정에 필수적인 과정이고, 상처 치료, 장기 섬유증(organ fibrosis), 및 암 진행 중 전이의 개시에 일어나는 것으로 알려져 있다.Epithelial-mesenchymal transition (EMT) is the process by which epithelial cells lose cell polarity and cell-to-cell adhesion and transform into mesenchymal cells that are mobile and invasive. Epithelial-mesenchymal transition is essential for many developmental processes, including mesoderm formation and neural tube formation, and is known to occur in wound healing, organ fibrosis, and in the initiation of metastasis during cancer progression.
혈중 암세포는 상피-중간엽 전이된 혈중 암세포, 상피-중간엽 전이되지 않은 혈중 암세포 또는 상피-중간엽 전이 진행 중인 혈중 암세포일 수 있고, 바람직하게는 상피-중간엽 전이되거나 상피-중간엽 전이 진행 중인 혈중 암세포일 수 있으나, 이에 제한되는 것은 아니다.The circulating cancer cells may be circulating cancer cells that have undergone epithelial-mesenchymal transition, circulating cancer cells that have not undergone epithelial-mesenchymal transition, or circulating cancer cells that are undergoing epithelial-mesenchymal transition, and are preferably, but not limited to, circulating cancer cells that have undergone epithelial-mesenchymal transition or are undergoing epithelial-mesenchymal transition.
혈중 암세포는 상피성 세포(epithelial cell)도 또는 중간엽성 세포(mesenchymal cell)도 아닌 세포일 수 있다.Cancer cells in the blood may be neither epithelial nor mesenchymal cells.
본 명세서에서 혈중 암세포의 검출은 생물학적 시료로부터 혈중 암세포와 PTGFRN에 특이적으로 결합하는 물질이 결합한 복합체를 분리하는 것을 포함한다. 검출 방법은 전자 현미경 관찰, 미세여과, 원심분리, 미세유동, 면역염색(immunostaining), 면역침강(immunoprecipitation), ELISA(enzyme-linked immunosorbent assay), 유세포 분석(flow cytometry), FACS(fluorescense activated cell sorting), 또는 이들의 조합을 포함한다. 또한, 검출 방법은 폴리머라제 연쇄 반응(polymerase chain reaction: PCR), 전기 영동, 노던 블롯팅, 웨스턴 블롯팅, 또는 이들의 조합일 수 있다In the present specification, detection of blood cancer cells comprises separating a complex in which blood cancer cells and a substance that specifically binds to PTGFRN are combined from a biological sample. The detection method comprises electron microscopy, microfiltration, centrifugation, microfluidics, immunostaining, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA), flow cytometry, fluorescense activated cell sorting (FACS), or a combination thereof. In addition, the detection method may be polymerase chain reaction (PCR), electrophoresis, northern blotting, western blotting, or a combination thereof.
또한, 본 발명의 일 구체 예에 따르면, PTGFRN은 간암, 폐암, 대장암, 췌장암, 갑상선암 세포주에서 발현이 확인되며, 간세포암 환자의 97%의 비율로 혈중 암세포가 검출되었다, 동시에, 또한 EMT 마커인 Vimentin과 1차 HCC에서는 약 53%, 2차 HCC에서는 약 28% 동시에 발현이 확인되므로, EMT 표현형을 나타내는 혈중 암세포 검출에 유용하며 전이성 암 진단에 사용할 수 있음을 확인하였다. 또한 63-D7이 인식하는 혈중 암세포는 EpCAM과 Vimentin 모두를 발현하지 않은 중간형태 혈중 암세포에도 1차 HCC에서는 약 46%, 2차 HCC에서는 약 72% 동시에 발현이 확인되어 기존의 EpCAM 마커 기반 혈중 암세포 진단 기술의 단점을 보완하여 혈중 암세포 검출에 유용하게 사용할 수 있다.In addition, according to one specific example of the present invention, PTGFRN was confirmed to be expressed in liver cancer, lung cancer, colon cancer, pancreatic cancer, and thyroid cancer cell lines, and 97% of hepatocellular carcinoma patients were detected with circulating cancer cells. At the same time, since it was confirmed to be simultaneously expressed with Vimentin, an EMT marker, in about 53% of primary HCCs and about 28% of secondary HCCs, it was confirmed to be useful for detecting circulating cancer cells showing an EMT phenotype and can be used for diagnosing metastatic cancer. In addition, circulating cancer cells recognized by 63-D7 were simultaneously expressed in about 46% of primary HCCs and about 72% of secondary HCCs, even in intermediate circulating cancer cells that did not express either EpCAM or Vimentin, so it can be usefully used for detecting circulating cancer cells by complementing the shortcomings of existing EpCAM marker-based circulating cancer cell diagnosis technology.
혈중 암세포는 뇌척수종양, 두경부암, 폐암, 유방암, 흉선종, 중피종, 식도암, 위암, 대장암, 간암, 췌장암, 담도암, 신장암, 방광암, 전립선암, 고환암, 생식세포종, 난소암, 자궁 경부암, 자궁 내막암, 림프종, 급성 백혈병, 만성 백혈병, 다발성 골수종, 육종, 악성 흑색종 및 피부암으로 이루어진 군에서 선택되는 원발성 암 유래일 수 있다.The blood cancer cells may be derived from a primary cancer selected from the group consisting of brain and spinal cancer, head and neck cancer, lung cancer, breast cancer, thymoma, mesothelioma, esophageal cancer, stomach cancer, colon cancer, liver cancer, pancreatic cancer, biliary tract cancer, kidney cancer, bladder cancer, prostate cancer, testicular cancer, germ cell tumor, ovarian cancer, cervical cancer, endometrial cancer, lymphoma, acute leukemia, chronic leukemia, multiple myeloma, sarcoma, malignant melanoma, and skin cancer.
본 발명은 개체로부터 분리된 시료에 EpCAM에 특이적으로 결합하는 물질을 처리하는 단계를 더 포함하는 혈중 암세포 수준 판단에 필요한 정보 제공 방법을 제공한다. 본 발명은 상기 EpCAM에 특이적으로 결합하는 물질은 항-EpCAM 항체 또는 이의 항원 결합 단편인 혈중 암세포 수준 판단에 필요한 정보 제공 방법을 제공한다.The present invention provides a method for providing information necessary for determining the level of cancer cells in blood, which further includes a step of treating a sample isolated from an individual with a substance that specifically binds to EpCAM. The present invention provides a method for providing information necessary for determining the level of cancer cells in blood, wherein the substance that specifically binds to EpCAM is an anti-EpCAM antibody or an antigen-binding fragment thereof.
EpCAM은 상피성 세포에서 Ca2+ 의존성 동형(homotypic) 세포간 부착을 매개하는 막관통 당단백질이다. EpCAM은 세포 신호 전달, 이동, 증식, 및 분화에 관련된 것으로 알려져 있다. EpCAM은 인간 또는 마우스 EpCAM일 수 있다. 예를 들면, EpCAM은 GenBank Accession No. NP_002345의 아미노산 서열을 갖는 폴리펩타이드일 수 있다. 예를 들면, EpCAM은 GenBank Accession No. NM_002354의 뉴클레오타이드 서열에 의해 암호화되는 폴리펩타이드일 수 있다. EpCAM에 특이적으로 결합하는 물질은 항-EpCAM 항체 또는 그의 항원 결합 단편일 수 있다. 항체 또는 이의 항원 결합 단편은 전술한 바와 같다.EpCAM is a transmembrane glycoprotein that mediates Ca 2+ -dependent homotypic cell-cell adhesion in epithelial cells. EpCAM is known to be involved in cell signaling, migration, proliferation, and differentiation. EpCAM can be human or mouse EpCAM. For example, EpCAM can be a polypeptide having an amino acid sequence of GenBank Accession No. NP_002345. For example, EpCAM can be a polypeptide encoded by a nucleotide sequence of GenBank Accession No. NM_002354. The substance that specifically binds to EpCAM can be an anti-EpCAM antibody or an antigen-binding fragment thereof. The antibody or antigen-binding fragment thereof is as described above.
본 발명의 혈중 암세포 수준 판단에 필요한 정보 제공 방법에서 혈중 암세포는 상피-중간엽 전이되거나 상피-중간엽 전이 진행 중인 혈중 암세포일 수 있다.In the method for providing information necessary for determining the level of blood cancer cells of the present invention, the blood cancer cells may be blood cancer cells undergoing epithelial-mesenchymal transition or undergoing epithelial-mesenchymal transition.
본 발명의 혈중 암세포 수준 판단에 필요한 정보 제공 방법에서 혈중 암세포는 하이브리드 E/M 상태일 수 있다.In the method for providing information necessary for determining the level of blood cancer cells of the present invention, the blood cancer cells may be in a hybrid E/M state.
본 발명의 혈중 암세포 수준 판단에 필요한 정보 제공 방법에서 혈중 암세포는 상피성 세포도 중간엽성 세포도 아닐 수 있다.In the method for providing information necessary for determining the level of blood cancer cells of the present invention, the blood cancer cells may be neither epithelial cells nor mesenchymal cells.
본 발명의 혈중 암세포 수준 판단에 필요한 정보 제공 방법에서 혈중 암세포는 간암, 폐암, 대장암, 감상선암, 췌장암, 악성 흑색종, 유방암, 신경모세포종 또는 교모세포종 환자의 혈액 내 혈중 암세포일 수 있다.In the method for providing information necessary for determining the level of blood cancer cells of the present invention, the blood cancer cells may be blood cancer cells in the blood of a patient with liver cancer, lung cancer, colon cancer, thyroid cancer, pancreatic cancer, malignant melanoma, breast cancer, neuroblastoma, or glioblastoma.
본 발명의 혈중 암세포 수준 판단에 필요한 정보 제공 방법에서 혈중 암세포는 B7-H3(CD276) 단백질을 더 발현할 수 있다.In the method for providing information necessary for determining the level of blood cancer cells of the present invention, the blood cancer cells can further express B7-H3 (CD276) protein.
본 발명은 개체로부터 분리된 시료에 본 발명의 항 PTGFRN 단일클론항체를 처리하여 PTGFRN을 검출하는 단계를 포함하는 암 전이 예후 예측에 필요한 정보 제공 방법을 제공한다.The present invention provides a method for providing information necessary for predicting the prognosis of cancer metastasis, comprising the step of detecting PTGFRN by treating a sample isolated from an individual with the anti-PTGFRN monoclonal antibody of the present invention.
본 발명은 검출된 PTGFRN의 수준이 대조군 대비 높으면 대조군 대비 암 전이 예후가 더 나쁜 것이라는 정보를 제공하는 단계를 더 포함하는 암 전이 예후 예측에 필요한 정보 제공 방법을 제공한다.The present invention provides a method for providing information necessary for predicting the prognosis of cancer metastasis, further comprising a step of providing information that if the level of detected PTGFRN is higher than that of the control group, the prognosis of cancer metastasis is worse than that of the control group.
본 발명은 항암 약물을 투여 받은 환자로부터 분리된 시료에 본 발명의 항 PTGFRN 단일클론항체를 처리하여 PTGFRN을 검출하는 단계를 포함하는 항암 약물의 효능 스크리닝 방법을 제공한다.The present invention provides a method for screening the efficacy of an anticancer drug, comprising the step of detecting PTGFRN by treating a sample isolated from a patient administered an anticancer drug with an anti-PTGFRN monoclonal antibody of the present invention.
본 발명은 검출된 PTGFRN의 수준이 대조군 대비 낮으면 대조군 대비 상기 항암 약물의 암 전이 억제 효능이 더 우수할 것으로 예측하는 단계를 더 포함하는 항암 약물의 효능 스크리닝 방법을 제공한다.The present invention provides a method for screening the efficacy of an anticancer drug, further comprising a step of predicting that the anticancer drug will have a better efficacy in inhibiting cancer metastasis compared to a control group if the level of detected PTGFRN is lower than that of a control group.
본 발명은 PTGFRN에 결합하는 물질을 포함하는 혈중 암세포 검출용 조성물을 제공한다.The present invention provides a composition for detecting cancer cells in blood, comprising a substance that binds to PTGFRN.
본 발명은 PTGFRN에 결합하는 물질을 포함하는 PTGFRN 단백질 분리, 정제, 검출 및 농도 검출용 조성물을 제공한다.The present invention provides a composition for isolating, purifying, detecting and detecting the concentration of PTGFRN protein, which comprises a substance that binds to PTGFRN.
본 발명의 혈중 암세포 검출용 조성물은 상피 세포 접착 분자(epithelial cell adhesion molecule, EpCAM)에 특이적으로 결합하는 물질을 더 포함할 수 있다.The composition for detecting blood cancer cells of the present invention may further include a substance that specifically binds to epithelial cell adhesion molecule (EpCAM).
본 발명의 혈중 암세포 검출용 조성물에서 PTGFRN에 결합하는 물질은 항 PTFGRN 항체 또는 앱타머 일 수 있다. 항 PTFGRN 항체는 단일클론항체 또는 다클론항체일 수 있다.In the composition for detecting blood cancer cells of the present invention, the substance binding to PTGFRN may be an anti-PTGFRN antibody or an aptamer. The anti-PTGFRN antibody may be a monoclonal antibody or a polyclonal antibody.
혈중 암세포는 간암, 폐암, 대장암, 갑상선암, 췌장암, 흑색종, 유방암, 신경모세포종 또는 교모세포종 환자의 혈액 내 혈중 암세포일 수 있으나, 이에 제한되는 것은 아니다. 혈중 암세포는 상피-중간엽 전이되거나 상피-중간엽 전이 진행 중인 혈중 암세포일 수 있다. 혈중 암세포는 하이브리드 E/M 상태일 수 있다.The circulating cancer cells may be, but are not limited to, circulating cancer cells in the blood of a patient with liver cancer, lung cancer, colon cancer, thyroid cancer, pancreatic cancer, melanoma, breast cancer, neuroblastoma, or glioblastoma. The circulating cancer cells may be circulating cancer cells that have undergone epithelial-mesenchymal transition or are undergoing epithelial-mesenchymal transition. The circulating cancer cells may be in a hybrid E/M state.
본 발명의 혈중 암세포 검출용 조성물에서 PTGFRN에 결합하는 물질은 본 발명의 항 PTFGRN 항체일 수 있다.In the composition for detecting blood cancer cells of the present invention, the substance binding to PTGFRN may be an anti-PTGFRN antibody of the present invention.
본 발명의 혈중 혈중암세포 검출용 조성물은 키트 형태로 포함될 수 있다. 예컨대, 본 발명의 키트가 면역 분석에 적용되는 경우, 본 발명의 키트는 선택적으로, 이차항체 및 표지의 기질을 포함할 수 있다. 나아가, 본 발명에 따른 키트는 상기한 시약 성분을 포함하는 다수의 별도 패키징 또는 컴파트먼트로 제작될 수 있다. The composition for detecting blood cancer cells in blood of the present invention may be included in the form of a kit. For example, when the kit of the present invention is applied to an immunoassay, the kit of the present invention may optionally include a secondary antibody and a substrate of a label. Furthermore, the kit according to the present invention may be manufactured into a plurality of separate packages or compartments containing the above-mentioned reagent components.
본 발명의 혈중암세포 검출용 조성물은 마이크로어레이의 형태로 포함될 수 있다. 마이크로어레이에 있어서, 상기 단일클론항체는 혼성화 어레이 요소(hybridizable array element)로서 이용되며, 기질(substrate) 상에 고정화된다. 바람직한 기질은 적합한 견고성 또는 반-견고성 지지체로서, 예컨대, 막, 필터, 칩, 슬라이드, 웨이퍼, 파이버, 자기성 비드 또는 비자기성 비드, 겔, 튜빙, 플레이트, 고분자, 미소입자 및 모세관을 포함할 수 있다. 상기 혼성화 어레이 요소는 상기 기질 상에 배열되고 고정화되며, 이와 같은 고정화는 화학적 결합 방법 또는 UV와 같은 공유 결합적 방법에 의해 수행될 수 있다. 예를 들어, 상기 혼성화 어레이 요소는 에폭시 화합물 또는 알데히드기를 포함하도록 변형된 글래스 표면에 결합될 수 있고, 또한 폴리라이신 코팅 표면에서 UV에 의해 결합될 수 있다. 또한, 상기 혼성화 어레이 요소는 링커(예: 에틸렌 글리콜 올리고머 및 디아민)를 통해 기질에 결합될 수 있다.The composition for detecting blood cancer cells of the present invention may be included in the form of a microarray. In the microarray, the monoclonal antibody is used as a hybridizable array element and is immobilized on a substrate. A preferred substrate is a suitable rigid or semi-rigid support, such as a membrane, a filter, a chip, a slide, a wafer, a fiber, a magnetic bead or a non-magnetic bead, a gel, a tubing, a plate, a polymer, a microparticle, and a capillary. The hybridizable array element is arranged and immobilized on the substrate, and such immobilization may be performed by a chemical bonding method or a covalent bonding method such as UV. For example, the hybridizable array element may be bonded to a glass surface modified to include an epoxy compound or an aldehyde group, and may also be bonded to a polylysine-coated surface by UV. In addition, the hybridizable array element may be bonded to the substrate through a linker (e.g., ethylene glycol oligomer and diamine).
본 발명은 PTGFRN에 결합하는 물질을 포함하는 암 전이 모니터링용 조성물을 제공한다. 상기 물질은 PTGFRN의 수준을 측정할 수 있는 것이라면 제한되지 않는다.The present invention provides a composition for monitoring cancer metastasis comprising a substance that binds to PTGFRN. The substance is not limited as long as it can measure the level of PTGFRN.
실시예 1. 암세포주 배양 및 연구 윤리Example 1. Cancer cell line culture and research ethics
1-1. 암세포주 배양1-1. Cancer cell line culture
인간 배아줄기세포 H9은 위스콘신대학 연구소(Wicell Research institute)에서 구입하여 제공된 프로토콜에 따라 배양하였다. 배양에 사용된 배지는 DMEM/F12(Invitrogen, Seoul, Korea), 20%의 녹아웃(Knockout) SR(Invitrogen), 0.1 mM의 β-머캅토에탄올(mercaptoethanol)(Sigma, St Luis, MO, USA), 2 mM의 글루타민(glutamine)(Invitrogen), 0.1 mM의 비필수 아미노산(Invitrogen), 100 U/㎖의 페니실린(penicillin) G(Sigma), 100㎍/㎖의 스트렙토마이신(streptomycin)(Sigma), 4 ng/㎖의 bFGF(PeproTech, Rocky Hill, NJ)을 섞어 만들었다. 인간배아암세포 NT-2/D1는 ATCC(ATCC, Manassas, VA, USA)에서 구입하고 제공된 프로토콜에 따라 배양하였다. 인간 간암(Huh7, HepG2, SNU387, SNU449), 비소세포폐암(A549, NCI-H358, NCI-H460, NCI-H1703, NCI-H23), 갑상선암(SNU-790)는 Korea Cell Line Bank(KCLB, 서울, 한국)에서 구입하였다. 인간 정상 폐 태아 섬유아세포(MRC5), 대장암(Colo205), 췌장암(BxPC3)는 ATCC에서 구입하고, 인간 간세포는 Termo Fisher Scientifc(Waltham, USA)에서 구입해서 제공된 프로토콜에 따라 배양하였다.Human embryonic stem cells H9 were purchased from the Wicell Research Institute and cultured according to the provided protocol. The culture medium contained DMEM/F12 (Invitrogen, Seoul, Korea), 20% Knockout SR (Invitrogen), 0.1 mM β-mercaptoethanol (Sigma, St Luis, MO, USA), 2 mM glutamine (Invitrogen), 0.1 mM nonessential amino acids (Invitrogen), 100 U/㎖ penicillin G (Sigma), 100 ㎍/㎖ streptomycin (Sigma), and 4 ng/㎖ bFGF (PeproTech, Rocky Hill, NJ). Human embryonic carcinoma NT-2/D1 cells were purchased from ATCC (Manassas, VA, USA) and cultured according to the provided protocol. Human hepatocarcinoma (Huh7, HepG2, SNU387, SNU449), non-small cell lung cancer (A549, NCI-H358, NCI-H460, NCI-H1703, NCI-H23), and thyroid cancer (SNU-790) cells were purchased from Korea Cell Line Bank (KCLB, Seoul, Korea). Human normal lung fetal fibroblast (MRC5), colon cancer (Colo205), and pancreatic cancer (BxPC3) cells were purchased from ATCC, and human hepatocytes were purchased from Termo Fisher Scientific (Waltham, USA) and cultured according to the provided protocol.
1-2. 연구 윤리 및 환자의 동의1-2. Research ethics and patient consent
인간 말초 혈액 단핵구 세포(peripheral blood mononuclear cell, PBMC)는 Ficoll-Paque Plus method (GE Healthcare, Seoul, Korea)에서 제공된 방법을 이용하여 농배 구배 원심력(Ficoll-Paque gradient centrifugation)을 통해 기증자로부터 분리하였다. 간암 환자의 혈액 채취는 서울 삼성병원 외과에서 간암 절제를 시행하는 간암 환자로부터 동의서를 받은 후, 헤파린(heparin) 코팅된 튜브로 수집하였고, 모든 과정은 서울삼성병원 IRB를 거쳐 승인되었으며, 선의로 세종대학교에 전달되어 세종대학교에서 분석하였다. 암 진행 단계 분석은 암에 관한 미국암연합위원회(American Joint Committee on Cancer (AJCC, 2010))가 지정한 기준을 사용하였다.Human peripheral blood mononuclear cells (PBMCs) were isolated from donors by Ficoll-Paque gradient centrifugation using the method provided by the Ficoll-Paque Plus method (GE Healthcare, Seoul, Korea). Blood samples from liver cancer patients undergoing liver resection in the Department of Surgery, Samsung Hospital, Seoul were collected into heparin-coated tubes after obtaining informed consent. All procedures were approved by the IRB of Seoul Samsung Hospital and were transferred to Sejong University in good faith for analysis there. Cancer staging was analyzed using the criteria specified by the American Joint Committee on Cancer (AJCC, 2010).
실시예 2. 63-D7 항체의 선발Example 2. Selection of 63-D7 antibody
이전 연구(Choi HS, et al., Cell Tissue Res. vol. 333, pp. 197-206, 2008)에서 제조한 인간배아줄기세포에 특이적인 항체 70종 중에서, 간암세포 두 종(Huh7, HepG2)과 폐암세포 두 종(A549, NCI-H358)에 대한 결합 정도를 관찰하기 위해 유세포 분석(Flow cytometry)를 수행하였다. 구체적으로 Huh7, HepG2, A549, NCI-H358 암세포를 0.05% 트립신(trypsin)을 처리하여 세포를 떼어내고 PBS(pH 7.4)로 세척한 후, 단일세포로 분리하기 위해 40 ㎛ 스트레이너(strainer, BD Biosciences)를 이용하여 필터하였다. 이때 인간배아줄기세포(H9), 인간말초단핵구세포(PBMC)도 함께 분석에 사용하였다. 세포 5×105 cell을 PBA(1% bovine serum albumin, 0.02% NaN3 in PBS, pH7.4)에 섞은 후, 항체들을 각각 4 ℃에서 30분간 반응시켰다. PBA로 2번 세척한 후, 1차 항체와 상응하는 항-마우스 IgG-FITC(BD Biosciences)를 4 ℃에서 30분간 더 반응시켰다. PBA로 2번 세척한 후, FACS Calibur와 Cell Quest software(BD sciences)를 이용하여 프로피디움 요오드화물(propidium iodide, PI)-음성 세포에 대해서 항체 반응 여부를 분석하였다. 그 결과로 인간배아줄기세포(H9), 간암세포주(Huh7, HepG2), 폐암세포주(A549)에 결합하고 PBMC에는 결합하지 않은 63-D7, 246-D7, 247-B9 항체를 선발하였다 (도 1a, 도 1b). 이 중에서 246-D7과 247-B9은 상피성 폐암세포주인 NCI-H358에 결합하지만 63-D7은 결합하지 않았다. 항체들의 특성을 분석하기위해서 63-D7, 246-D7, 247-B9 항체를 프로테인 G-아가로스 컬럼 크로마토그래피로 정제하고 정제한 항체들을 각각 DSB-XTM Biotin Protein Labeling Kit (Molecular Probes, Seoul, Korea)를 사용하여 제공한 프로토콜에 따라 biotin을 붙였다.In order to observe the degree of binding to two liver cancer cells (Huh7, HepG2) and two lung cancer cells (A549, NCI-H358) among the 70 antibodies specific for human embryonic stem cells produced in a previous study (Choi HS, et al., Cell Tissue Res. vol. 333, pp. 197-206, 2008), flow cytometry was performed. Specifically, Huh7, HepG2, A549, and NCI-H358 cancer cells were treated with 0.05% trypsin to detach the cells, washed with PBS (pH 7.4), and then filtered using a 40 μm strainer (BD Biosciences) to separate them into single cells. Human embryonic stem cells (H9) and human peripheral mononuclear cells (PBMC) were also used in the analysis. After mixing 5 × 10 5 cells with PBA (1% bovine serum albumin, 0.02% NaN 3 in PBS, pH 7.4), the antibodies were reacted for 30 minutes at 4 °C, respectively. After washing twice with PBA, the primary antibody and the corresponding anti-mouse IgG-FITC (BD Biosciences) were reacted for another 30 minutes at 4 °C. After washing twice with PBA, the antibody reaction was analyzed for propidium iodide (PI)-negative cells using FACS Calibur and Cell Quest software (BD sciences). As a result, 63-D7, 246-D7, and 247-B9 antibodies that bound to human embryonic stem cells (H9), liver cancer cell lines (Huh7, HepG2), and lung cancer cell lines (A549) but not to PBMCs were selected (Fig. 1a, Fig. 1b). Among these, 246-D7 and 247-B9 bound to the epithelial lung cancer cell line NCI-H358, but 63-D7 did not bind. To analyze the characteristics of the antibodies, 63-D7, 246-D7, and 247-B9 antibodies were purified by protein G-agarose column chromatography, and the purified antibodies were biotinylated according to the protocol provided using the DSB-XTM Biotin Protein Labeling Kit (Molecular Probes, Seoul, Korea).
세 가지 항체가 간암 환자의 혈액에서 혈중 암세포를 검출할 수 있는지 탐색하기 위하여, 건강한 성인 1명과 간암(hepatocellular carcinoma, HCC) 환자 8명으로부터 실시예 1-2과 같은 방법으로 혈액에서 PBMC를 분리하였다. 회수된 PBMC 세포는 폴리-L-라이신이 코팅된 슬라이드에 세포 수에 따라 2~4개에 나누어 붙였다. 준비된 슬라이드는 4% 파라포름알데하이드(paraformaldehyde, PFA)로 상온에서 10분간 처리하여 세포를 고정시켰다. 세포염색에서 사용하는 모든 PBS에서는 세포 손실을 방지하기 위하여 항상 1mM Ca2+, 0.5mM Mg2+을 포함한 PBS(pH 7.4)를 사용하였다. PBS로 세척한 후, 블로킹 용액(10% 말 혈청, 0.1% BSA, PBS, pH 7.4)으로 1시간 동안 세포를 블로킹하였다. 그리고, 마우스 항체인 CD45 항체(BD Biosciences, Seoul Korea)로 실온에서 빛을 차단한 채 1시간 동안 반응시키고, 이어서 anti-mouse IgG-Alexa488(Invitrogen, 1:3000)을 실온에서 빛을 차단한 채 1시간 더 반응시켰다. PBS(pH 7.4)로 세척한 후, 각 항체들이 인식하는 항원을 염색하기 위해 5 ㎍의 63-D7-biotin, 246-D7-biotin, 247-B9-biotin 항체를 각각 넣고 4 ℃ 실온에서 빛을 차단한 채 12시간 동안 반응시켰다. PBS로 세척한 후, Cy3-conjugated streptavidin (Vector Laboratories, BURLINGAME, CA, USA, 1:3000)으로 실온에서 빛을 차단한 채 1시간 동안 반응시켰다. PBS로 세척한 후, DAPI(4,6-diamidino- 2-phenylindole)를 이용하여 핵을 염색하였다. 혈액에서 회수한 세포에서 항체들에 양성인 혈중 암세포를 셀 때, 혈액 때문에 생기는 비특이적인 결합을 제거하기 위해 DAPI-양성인 세포만 세어 그 수를 계산하였다. 그 결과, 정상인의 혈액에서는 검출되지 않지만 대부분의 간암환자 혈액에서는 항체 양성인 세포가 검출되었으며 특히 63-D7 항체는 8명 환자중에서 6명에게서 총 51개 혈중 암세포를 검출할 수 있어 추가 분석이 필요한 항체로 선정하였다 (도 1c, 도 1d).To investigate whether three antibodies can detect circulating cancer cells in the blood of patients with liver cancer, PBMCs were isolated from the blood of one healthy adult and eight patients with hepatocellular carcinoma (HCC) using the same method as in Example 1-2. The recovered PBMC cells were divided into 2 to 4 cells depending on the number of cells and attached to poly-L-lysine-coated slides. The prepared slides were treated with 4% paraformaldehyde (PFA) for 10 minutes at room temperature to fix the cells. To prevent cell loss, PBS (pH 7.4) containing 1 mM Ca 2+ and 0.5 mM Mg 2+ was always used in all PBS used for cell staining. After washing with PBS, the cells were blocked with blocking solution (10% horse serum, 0.1% BSA, PBS, pH 7.4) for 1 hour. Then, the plate was reacted with mouse antibody CD45 antibody (BD Biosciences, Seoul Korea) at room temperature, blocked from light, for 1 hour, followed by anti-mouse IgG-Alexa488 (Invitrogen, 1:3000) at room temperature, blocked from light, for another 1 hour. After washing with PBS (pH 7.4), 5 μg of 63-D7-biotin, 246-D7-biotin, and 247-B9-biotin antibodies were added respectively to stain the antigens recognized by each antibody and reacted for 12 hours at room temperature, blocked from light, 4°C. After washing with PBS, the plate was reacted with Cy3-conjugated streptavidin (Vector Laboratories, BURLINGAME, CA, USA, 1:3000) at room temperature, blocked from light, for 1 hour. After washing with PBS, nuclei were stained with DAPI (4,6-diamidino-2-phenylindole). When counting circulating cancer cells positive for antibodies in cells recovered from blood, only DAPI-positive cells were counted to eliminate nonspecific binding caused by blood. As a result, antibody-positive cells were detected in the blood of most liver cancer patients, but not in the blood of normal people. In particular, the 63-D7 antibody was selected as an antibody requiring further analysis because it could detect a total of 51 circulating cancer cells in 6 out of 8 patients (Fig. 1c, Fig. 1d).
실시예 3. 유세포 분석기를 통한 다양한 세포에 대한 63-D7 결합 분석Example 3. Analysis of 63-D7 binding to various cells using a flow cytometer
단일클론항체 63-D7가 배아줄기세포(H9), 간암(Huh7, HepG2), 폐암(A549, NCI-H358) 외에도, 다양한 암세포와 정상세포에 결합하는지 확인하기 위하여 배아암(NT2/D1), 전이성 간암(SNU387, SNU449), 폐암 (NCI-H460, NCI-H1703, NCI-H23), 대장암(Colo205, HCT116), 갑상선암 (SNU790), 췌장암(BxPC3)을 포함하는 다양한 암세포(SK-Hep1, 8505C, A375, MCF-7, MDA-MB-435, SH-SY5Y, U87-MG)와 인간 배아 신장세포(HEK293FT), 골수-유래 중간엽 줄기세포(BM-MSC), 인간 피부 섬유아세포(HDF), 인간 말초 혈액 단핵구 세포(PBMC), 정상 폐 태아 섬유아세포로 알려진 MRC5, 치수세포(DPC) 및 정상 간세포(Hepatocyte)를 사용하여 유세포 분석(Flow cytometry)을 수행하였다. 구체적으로 세포를 0.05% 트립신을 처리해서 세포를 떼어내고 PBS(pH 7.4)로 세척한 후, 단일세포로 분리하기 위해 40 ㎛ 스트레이너(strainer, BD Biosciences)를 이용하여 필터하였다. 세포 5×105 cell을 PBA(1% bovine serum albumin, 0.02% NaN3 in PBS, Ph 7.4)에 섞은 후, 63-D7 항체를 4 ℃에서 30분간 반응시켰다. PBA로 2번 세척한 후, 1차 항체와 상응하는 항-마우스 IgG-FITC(BD Biosciences)를 4 ℃에서 30분간 더 반응시켰다. PBA로 2번 세척한 후, FACS Calibur와 Cell Quest software(BD sciences)를 이용하여 프로피디움 요오드화물(propidium iodide, PI)-음성 세포에 대해서 항체 반응 여부를 분석하였다. 도 2에 도시된 바와 같이, 63-D7 항체는 간암세포를 포함한 다른 모든 암세포에 결합하지만 상피성 폐암세포 H358에는 결합하지 않는다. 그리고 정상세포인 PBMC, 치수세포 (DPC) 및 MRC5에는 결합하지 않고 정상 간세포에도 결합하지 않는 것을 볼 수 있어 암세포 특이적인 결합력을 보여준다(도 2). 실험 결과는 도 2와 하기의 표 1에 나타나 있다.To determine whether monoclonal antibody 63-D7 binds to various cancer cells and normal cells in addition to embryonic stem cells (H9), liver cancer (Huh7, HepG2), and lung cancer (A549, NCI-H358), various cancer cells (SK-Hep1, 8505C, A375, MCF-7, MDA-MB-435, SH-SY5Y, U87-MG) including embryonic cancer (NT2/D1), metastatic liver cancer (SNU387, SNU449), lung cancer (NCI-H460, NCI-H1703, NCI-H23), colon cancer (Colo205, HCT116), thyroid cancer (SNU790), and pancreatic cancer (BxPC3), as well as human embryonic kidney cells (HEK293FT), bone marrow-derived mesenchymal stem cells (BM-MSC), human dermal fibroblasts (HDF), and human peripheral blood (PB) cells. Flow cytometry was performed using primary mononuclear cells (PBMCs), normal lung fetal fibroblasts (MRC5), dental pulp cells (DPCs), and normal hepatocytes. Specifically, cells were detached by treating with 0.05% trypsin, washed with phosphate-buffered saline (PBS), and filtered using a 40 μm strainer (BD Biosciences) to separate single cells. 5 × 10 5 cells were mixed with PBA (1% bovine serum albumin, 0.02% NaN 3 in PBS, Ph 7.4), and then reacted with 63-D7 antibody at 4 °C for 30 min. After washing twice with PBA, the primary antibody and corresponding anti-mouse IgG-FITC (BD Biosciences) were reacted for another 30 min at 4 °C. After washing twice with PBA, the antibody reaction to propidium iodide (PI)-negative cells was analyzed using FACS Calibur and Cell Quest software (BD sciences). As shown in Fig. 2, the 63-D7 antibody binds to all cancer cells including hepatoma cells, but does not bind to epithelial lung cancer cells H358. In addition, it does not bind to normal cells such as PBMC, dental pulp cells (DPC), and MRC5, and does not bind to normal hepatocytes, demonstrating cancer cell-specific binding (Fig. 2). The experimental results are shown in Fig. 2 and Table 1 below.
실시예 4. 항체 63-D7이 결합하는 항원 분리Example 4. Isolation of antigen bound by antibody 63-D7
63-D7 항체가 인식하는 세포 표면 항원을 분석하기 위해 세포 표면 단백질만을 바이오틴으로 표지한 후, 63-D7로 침전시켜 분리하는 면역침강법(Immunoprecipitation)을 시행하였다. 잘 결합하는 NT-2/D1 암세포를 PBS(pH 7.4)로 2회 세척한 후, NHS-Sulfo-LC-biotin(Pierce)를 PBS(pH 8.0)에 녹인 용액을 넣어 4 ℃에서 30분간 반응시킨 후 PBS(pH 8.0)으로 2회 세척하였다. 이후 용해 완충액(25 mM Tris-HCl, pH 7.5, 250 mM NaCl, 5 mM EDTA, 1% Nonidet P-40, 2 ㎍/ml aprotinin, 100 ㎍/ml PMSF, 5 ㎍/ml leupeptin, 1 mM NaF, 1 mM Na3VO4)을 이용하여 4 ℃에서 30분간 반응시킨 후, 12,000 rpm 속도로 40분간 원심분리해서 핵을 제거하고 사용하기 전까지 -70 ℃에서 보관하여 단백질 용액을 준비하였다. 준비된 단백질 용액에 단백질-G-플러스 아가로스(Merck millipore, Darmstadt, Germany)를 가한 후, 비특이적으로 결합하는 분자들을 제거하기 위해 약 1×107 cell의 세포 추출물에 20 ㎕ 단백질-G-아가로스를 넣고 4 ℃에서 2시간 동안 반응시킨 후 원심분리를 통해 단백질-G-플러스 아가로스에 결합하는 분자들은 제거하고, 나머지 상층액을 회수하여 63-D7에 결합하는 분자들을 면역침강 하는 데 사용하였다. 63-D7에 의해 인식되는 표면항원을 면역침강 하기 위해서 회수한 상층액에 항체 5 ㎍을 넣고 4 ℃에서 12시간 동안 반응시킨 후, 20 ㎕ 단백질-G-플러스 아가로스를 넣고 4 ℃에서 3시간 동안 더 반응시켰다. 면역침강된 면역 혼합체를 용해 용액을 이용해서 5회 세척하였다. 이후 항체에 결합된 항원을 용출(elution)하기 위해 5×샘플 완충액를 넣고, 100 ℃에서 10분간 끓인 후 10% SDS-PAGE를 이용하여 전개하고, 니트로셀룰로스 막으로 웨스턴 블롯팅을 하였다. 이 막을 5% 탈지분유를 이용하여 실온에서 2시간 동안 블로킹 한 후, PBST[0.05%의 트윈 20을 첨가한 인산 완충액(PBS), pH 7.4]로 3번 세척하고, 웨스턴 막을 스트렙타비딘-HRP(Streptavidin-HRP, GE healthcare)로 실온에서 1시간 반응시켜 바이오틴 표지되어 63-D7에 의해 면역침강된 표면항원에 결합하도록 하였다. 이후 PBST로 세척한 후, 결합된 단백질을 ECL 검출 키트(GE healthcare)로 확인하였다. 도 3a에 나타난 바와 같이, NT-2/D1 세포에서 63-D7 항체는 약 130 kDa 크기의 바이오틴에 의해 표지된 표면항원을 인식하여 면역침강 시킴을 알 수 있다(도 3a).To analyze the cell surface antigen recognized by the 63-D7 antibody, an immunoprecipitation method was performed in which only the cell surface proteins were labeled with biotin and then precipitated with 63-D7 to isolate them. NT-2/D1 cancer cells that bind well were washed twice with PBS (pH 7.4), and a solution of NHS-Sulfo-LC-biotin (Pierce) dissolved in PBS (pH 8.0) was added and reacted at 4°C for 30 minutes, and then washed twice with PBS (pH 8.0). Afterwards, the solution was incubated at 4°C for 30 minutes using lysis buffer (25 mM Tris-HCl, pH 7.5, 250 mM NaCl, 5 mM EDTA, 1% Nonidet P-40, 2 μg/ml aprotinin, 100 μg/ml PMSF, 5 μg/ml leupeptin, 1 mM NaF, 1 mM Na 3 VO 4 ), centrifuged at 12,000 rpm for 40 minutes to remove nuclei, and stored at -70°C until use to prepare a protein solution. After adding protein-G-plus agarose (Merck millipore, Darmstadt, Germany) to the prepared protein solution, 20 μl of protein-G-agarose was added to the cell extract of about 1 × 10 7 cells to remove molecules that bind nonspecifically, and the mixture was reacted at 4 °C for 2 hours. The molecules that bind to protein-G-plus agarose were removed through centrifugation, and the remaining supernatant was collected and used to immunoprecipitate molecules that bind to 63-D7. To immunoprecipitate the surface antigen recognized by 63-D7, 5 μg of antibody was added to the collected supernatant and reacted at 4 °C for 12 hours, and then 20 μl of protein-G-plus agarose was added and reacted at 4 °C for 3 hours. The immunoprecipitated immune mixture was washed five times using a dissolution solution. After that, 5× sample buffer was added to elute the antigen bound to the antibody, boiled at 100 °C for 10 minutes, developed using 10% SDS-PAGE, and Western blotting was performed with a nitrocellulose membrane. The membrane was blocked with 5% skim milk powder for 2 hours at room temperature, washed three times with PBST [phosphate buffer solution (PBS), pH 7.4 containing 0.05% Tween 20], and the Western membrane was reacted with streptavidin-HRP (Streptavidin-HRP, GE healthcare) at room temperature for 1 hour to bind to the surface antigen immunoprecipitated by 63-D7 that was biotin-labeled. After washing with PBST, the bound protein was confirmed using an ECL detection kit (GE healthcare). As shown in Fig. 3a, in NT-2/D1 cells, the 63-D7 antibody recognizes and immunoprecipitates a surface antigen labeled with biotin with a size of approximately 130 kDa (Fig. 3a).
실시예 5. 항체 63-D7에 의해 면역침강된 항원의 동정 및 검증Example 5. Identification and validation of antigen immunoprecipitated by antibody 63-D7
5-1. 항체 63-D7에 의해 면역침강된 항원의 동정5-1. Identification of antigen immunoprecipitated by antibody 63-D7
63-D7에 의해 인식되는 항원을 동정하기 위해, 63-D7에 의해 면역침강된 단백질을 포함하는 폴리아크릴아마이드 젤을 PageBlue Protein Staining Solution(Thermo Fischer Scientific)으로 공급자의 프로토콜에 따라 염색하였다(도 3b). 젤에서 63-D7에 의해 면역침강된 단백질로 추정되는 130 kDa 위치에 염색된 단백질 밴드를 잘라 LC-MS/MS(Liquid Chromatography with Tandem Mass Spectrometry) 분석을 의뢰하였다(ProteomeTech, Seoul, Korea). 분석이 완료된 매스 스펙트럼으로부터 단백질 동정을 위하여 록펠러(Rockefeller) 대학에서 개발한 ProFound 검색 엔진(http:/129.85.19.192/profound_bin/ WebProFound.exe)을 이용하였다. 그 결과, 63-D7이 인식하는 항원 단백질이 Homo sapiens Prostaglandin F2 receptor negative regulator(PTGFRN)임을 확인하였다(도 3c). 도 3c의 밑줄 친 아미노산은 실제로 질량분석기를 통해 확인한 펩타이드를 표시한 것으로, PTGFRN와 71개의 아미노산이 일치함을 보여준다.To identify the antigen recognized by 63-D7, the polyacrylamide gel containing the protein immunoprecipitated by 63-D7 was stained with PageBlue Protein Staining Solution (Thermo Fischer Scientific) according to the supplier's protocol (Fig. 3b). The protein band stained at the 130 kDa position, which was assumed to be the protein immunoprecipitated by 63-D7, was excised from the gel and subjected to LC-MS/MS (Liquid Chromatography with Tandem Mass Spectrometry) analysis (ProteomeTech, Seoul, Korea). The ProFound search engine (http:/129.85.19.192/profound_bin/WebProFound.exe) developed by Rockefeller University was used to identify proteins from the analyzed mass spectrum. As a result, the antigen protein recognized by 63-D7 was confirmed to be Homo sapiens Prostaglandin F2 receptor negative regulator (PTGFRN) (Fig. 3c). The underlined amino acids in Figure 3c actually represent the peptides identified by mass spectrometry, showing that 71 amino acids are identical to PTGFRN.
5-2. 항체 63-D7에 의해 면역침강된 항원의 검증5-2. Verification of antigen immunoprecipitated by antibody 63-D7
단일클론항체 63-D7의 항원 단백질이 PTGFRN인지 확인하기 위해, 상업적으로 판매되고 있는 PTGFRN에 대한 항체, 유전자 발현 벡터, siRNA 및 shRNA를 이용하여 면역침강, 웨스턴 블롯팅, RNA 간섭작용에 의한 유전자 녹다운 및 유세포 분석을 수행하였다.To confirm that the antigen protein of monoclonal antibody 63-D7 is PTGFRN, immunoprecipitation, Western blotting, gene knockdown by RNA interference, and flow cytometry were performed using commercially available antibodies, gene expression vectors, siRNA, and shRNA against PTGFRN.
단일클론항체 63-D7이 면역침강시킨 단백질이 PTGFRN인지 확인하기 위해, 생쥐 항-PTGFRN 단일클론항체(α-PTGFRN, R&D system)를 구입하여 면역침강법과 웨스턴 블롯팅을 수행하는 데 사용하였다. 상기 실시예 4에서 서술한 방법과 같이 바이오틴으로 표지한 A549 세포에서 세포용출액(150 μg)을 준비하고, 이 용출액을 상기 서술한 바와 동일하게 단일클론항체 63-D7(5 μg)과 α-PTGFRN(2.5 μg)으로 면역침강하였다. 이후, 항체를 넣지 않은 음성 대조군 단백질(No Ab)과 용출된 단백질을 10% SDS-PAGE를 통해 분리하고, 니트로셀룰로스 막으로 옮겨 공지의 생쥐 α-PTGFRN 항체로 웨스턴 블롯팅을 수행하였다. 그 결과, 63-D7 및 α-PTGFRN에서 면역침강된 약 130kDa PTGFRN이 동시에 검출되어, 이들이 PTGFRN을 인식하여 면역침강시킬 수 있음을 확인하였다(도 4a). 이 막을 Striping solution (100 mM 2ME, 2% SDS, 62.5 mM Tris-HCl, pH6.7)에서 50 ℃에서 30분간 살살 흔들며 반응시키고, 10분간 상기 PBST로 2회 세척하고 5% 탈지분유를 이용하여 실온에서 1시간 동안 블로킹하였다. 0.1% PBST로 3번 세척한 후, 스트렙타비딘-HRP(SA-HRP, 1:5000)를 넣고 실온에서 1시간 반응시켰다. 그리고 PBST로 3번 세척하고, 바이오틴이 표지된 단백질을 ECL 검출 키트로 확인하였다. 그 결과, 63-D7, α-PTGFRN의 면역침강체에서 세포 표면 PTGFRN과 동일하게 약 130 kDa 크기의 PTGFRN이 검출되었다(도 4b). 이러한 결과는 63-D7이 세포 표면 PTGFRN에 결합한다는 것을 보여준다.To confirm whether the protein immunoprecipitated by the monoclonal antibody 63-D7 was PTGFRN, a mouse anti-PTGFRN monoclonal antibody (α-PTGFRN, R&D system) was purchased and used for immunoprecipitation and Western blotting. A cell lysate (150 μg) was prepared from A549 cells labeled with biotin as described in Example 4, and the lysate was immunoprecipitated with the monoclonal antibody 63-D7 (5 μg) and α-PTGFRN (2.5 μg) as described above. Thereafter, the eluted protein and the negative control protein without antibody (No Ab) were separated through 10% SDS-PAGE, transferred to a nitrocellulose membrane, and Western blotting was performed with a known mouse α-PTGFRN antibody. As a result, approximately 130 kDa PTGFRN immunoprecipitated from 63-D7 and α-PTGFRN was detected simultaneously, confirming that they could recognize and immunoprecipitate PTGFRN (Fig. 4a). The membrane was reacted with gentle shaking in Striping solution (100 mM 2ME, 2% SDS, 62.5 mM Tris-HCl, pH6.7) at 50 ℃ for 30 min, washed twice with the above PBST for 10 min, and blocked with 5% skim milk powder for 1 h at room temperature. After washing three times with 0.1% PBST, streptavidin-HRP (SA-HRP, 1:5000) was added and reacted at room temperature for 1 h. Then, it was washed three times with PBST, and the biotin-labeled protein was confirmed with an ECL detection kit. As a result, PTGFRN with a size of approximately 130 kDa, identical to cell surface PTGFRN, was detected in the immunoprecipitate of 63-D7 and α-PTGFRN (Fig. 4b). These results show that 63-D7 binds to cell surface PTGFRN.
항체 63-D7가 PTGFRN을 직접 인식하여 면역침강시키는 것인지 간접 인식하여 침강시키는 것인지 구별하기 위한 실험을 수행하였다. PTGFRN 유전자 발현 벡터 pcDNA3.1(+)-PTGFRN-DYK(GeneScript, Piscataway, NJ, USA)를 구입하여 인간 배아 신장 세포 HEK293FT에서 FLAG tag가 달린 재조합 PTGFRN를 과발현 시킨 후, 이 세포 용해물을 마우스 항-FLAG 단일클론항체(α-FLAG)(Invitrogen), 생쥐 항-PTGFRN 단일클론항체(α-PTGFRN)(AbCAM), 63-D7를 사용해 면역 침강과 웨스턴 블롯을 수행하였다. 인간 배아 신장 세포 HEK293FT에서 PTGFRN을 과발현하기 위해, 우선 6-웰의 세포배양용 플레이트에 웰당 2.5 X 105개 HEK293FT 세포를 24시간 배양하고, 상기 pcDNA3.1(+)-PTGFRN-DYK 벡터 DNA 2 μg을 PEI(Polyetherimide) 6 μg(DNA:PEI=1:3)과 섞어 상온에서 20분간 반응한 혼합물을 세포 배양 배지에 첨가해 트랜스펙션(Transfection) 했다. 이후 72시간 동안 추가 배양한 세포로부터 상기 실시예 4에서 서술한 용해 방법과 같이 세포 용해완충액을 이용하여 세포 용해물을 준비하였다. 이 세포 용해물을 상기 서술한 바와 동일하게 63-D7과 항-FLAG 항체, α-PTGFRN로 면역 침강 후 항체를 넣지 않은 음성 대조군 단백질(No Ab)과 용출된 단백질을 10% SDS-PAGE를 이용하여 분리한 후 니트로셀룰로스 막으로 옮겼다. 이 막은 5% 탈지분유를 이용하여 실온에서 1시간 동안 블로킹을 하였다. 0.1% TBST로 3번 세척한 후, 공지의 마우스 항-FLAG 항체, 또는 α-PTGFRN, 63-D7를 4 ℃에서 16시간 반응시켰다. 0.1% TBST로 3번 세척한 후, 항-마우스 IgG-HRP(1:10,000; Millipore)를 실온에서 1시간 동안 더 반응시켰다. 0.1% TBST로 3번 세척한 후, ECL 검출 키트로 확인하였다(도 4c). 그 결과, 항-FLAG, α-PTGFRN, 63-D7에 의해 면역 침강된 각각의 단백질 모두 항-FLAG (도 4c 위), α-PTGFRN (도 4c 중간), 63-D7 (도 4c 아래)에 의해 인식됨을 관찰할 수 있었다. 이러한 결과로부터 본 발명의 단일클론항체 63-D7이 인간 배아암세포주 NT-2/D1의 표면에 존재하는 PTGFRN 단백질을 직접 인식하는 항체임이 검증되었다.An experiment was performed to distinguish whether antibody 63-D7 directly recognizes and immunoprecipitates PTGFRN or indirectly recognizes and precipitates it. PTGFRN gene expression vector pcDNA3.1(+)-PTGFRN-DYK (GeneScript, Piscataway, NJ, USA) was purchased, and recombinant PTGFRN tagged with a FLAG tag was overexpressed in human embryonic kidney cells HEK293FT. The cell lysate was immunoprecipitated and analyzed by Western blotting using mouse anti-FLAG monoclonal antibody (α-FLAG) (Invitrogen), mouse anti-PTGFRN monoclonal antibody (α-PTGFRN) (AbCAM), and 63-D7. To overexpress PTGFRN in human embryonic kidney cells HEK293FT, first, 2.5 X 10 5 HEK293FT cells per well in a 6-well cell culture plate were cultured for 24 hours, 2 μg of the pcDNA3.1(+)-PTGFRN-DYK vector DNA was mixed with 6 μg of polyetherimide (PEI) (DNA:PEI = 1:3), and the mixture was reacted at room temperature for 20 minutes, added to the cell culture medium, and transfection was performed. After additional culture for 72 hours, a cell lysate was prepared from the cells using a cell lysis buffer as described in Example 4. The cell lysate was immunoprecipitated with 63-D7, anti-FLAG antibody, and α-PTGFRN as described above, and the eluted protein and the negative control protein without antibody (No Ab) were separated using 10% SDS-PAGE and transferred to a nitrocellulose membrane. The membrane was blocked with 5% skim milk powder for 1 hour at room temperature. After washing three times with 0.1% TBST, the known mouse anti-FLAG antibody, or α-PTGFRN, 63-D7 was reacted at 4 °C for 16 hours. After washing three times with 0.1% TBST, the membrane was further reacted with anti-mouse IgG-HRP (1:10,000; Millipore) for 1 hour at room temperature. After washing three times with 0.1% TBST, the membrane was confirmed using an ECL detection kit (Fig. 4c). As a result, it was observed that each protein immunoprecipitated by anti-FLAG, α-PTGFRN, and 63-D7 was recognized by anti-FLAG (Fig. 4c top), α-PTGFRN (Fig. 4c middle), and 63-D7 (Fig. 4c bottom). From these results, it was verified that the monoclonal antibody 63-D7 of the present invention is an antibody that directly recognizes the PTGFRN protein present on the surface of the human embryonic carcinoma cell line NT-2/D1.
실시예 6. 간암세포에서 PTGFRN의 기능 분석Example 6. Functional analysis of PTGFRN in liver cancer cells
6-1. 간암세포주에서 siRNA를 이용한 녹다운(knockdown)6-1. Knockdown using siRNA in liver cancer cell lines
항체 63-D7의 항원인 PTGFRN이 간암세포의 성장에 어떻게 영향을 미치는지 알아보기 위해, 간암세포주 Huh7과 SNU449에서 PTGFRN을 타겟으로 하는 두 종의 siRNA(Bioneer, Daejeon, Korea)를 구매하여 일시적인 녹다운을 유도하였다. 6웰 플레이트에 8.0 x 105 cells/well로 세포를 분주한 후 다음날 100 nM의 음성 대조군(negative control) siRNA(siCon)(Genolution, Seoul, Korea)와 PTGFRN에 대한 두 종의 siRNA(siPTGFRN#1, siPTGFRN #2)(표 2), Lipofectamine RNAiMAX (Invitrogen)를 각각 TOM(Welgene, Gyeongsan, Korea)에 희석하고 5분간 상온에 두었다. 이후, 각각의 siRNA 희석액과 RNAiMAX 희석액을 섞어 20분간 상온에서 반응시켰다. siRNA-lipofectamine 혼합액을 각 세포의 배지를 교체한 후 넣어주고 24시간 동안 반응시켰다. 24시간이 지난 시점에서 배지를 새로운 배양 배지로 교체하고 48시간을 추가적으로 배양하였다. 트랜스펙션 후 72시간이 지난 시점에서 세포를 0.05% 트립신-EDTA(Welgene, Gyeongsan, Korea)로 떼어낸 후 10%의 Fetal bovine serum(Corning)을 포함하는 배양배지로 중화시키고, PBS(pH 7.4)를 이용해 두 번 세척하였다. 이후 3.0 x 105개의 세포를 RNAiso(Takara)를 이용하여 제조사의 매뉴얼에 따라 RNA를 분리한 후, PrimeScript(Takara)를 이용하여 제조사의 매뉴얼에 따라 cDNA를 합성하였다. 이후, PTGFRN을 타겟으로 하는 프라이머와 GAPDH를 타겟으로 하는 프라이머, Power SYBR Green PCR Master Mix(applied biosystem)을 이용하여 qPCR을 진행했으며 GAPDH를 레퍼런스 유전자로 사용하였다. 사용한 프라이머의 구체적인 서열은 하기 표 2에 나타나 있다.To investigate how PTGFRN, the antigen of antibody 63-D7, affects the growth of hepatocellular carcinoma cells, two kinds of siRNA (Bioneer, Daejeon, Korea) targeting PTGFRN were purchased and transiently knocked down in hepatocellular carcinoma cell lines Huh7 and SNU449. After seeding the cells at 8.0 × 10 5 cells/well in a 6-well plate, 100 nM of negative control siRNA (siCon) (Genolution, Seoul, Korea) and two kinds of siRNA against PTGFRN (
/SupplierReference
/Supplier
(서열번호 9)
Reverse: 5'-TTTCATTGGGACTGGAGAGG-3'
(서열번호 10)Forward: 5'-ACAACAGCTGGGTGAAAGC-3'
(sequence number 9)
Reverse: 5'-TTTCATTGGGACTGGAGAGG-3'
(sequence number 10)
(서열번호 11)
Reverse: 5'-GAGGTCCACCACCCTGTTGC-3'
(서열번호 12)Forward: 5'-ACAGCGACACCCACTCCTCC-3'
(Sequence number 11)
Reverse: 5'-GAGGTCCACCACCCTGTTGC-3'
(sequence number 12)
(서열번호 13)5'-UUCUCCGAACGUGUCACGUtt-3'
(Sequence number 13)
(서열번호 14)5'-CUCCUACAUUUACUGGUUtt-3'
(Sequence number 14)
(서열번호 15)5'-GUCCCAAUGAAACGAAGUAtt-3'
(Sequence number 15)
(서열번호 16)5'-ctagcGCCTTTGATGTGTCCTGGTTTTtactagtAAACCAGGACACATCAAAGGCTTTTTg-3'
(Sequence number 16)
(서열번호 17)5'-aattcAAAAAAGCCTTTGATGTGTCCTGGTTTTactagtaAAACCAGGACACATCAAAGGCg-3'
(Sequence number 17)
(서열번호 18)5'-ctagcCCTATTGAGATAGACTTCCAAtactagtTTGGAAGTCTATCTCAATAGGTTTTTTg-3'
(Sequence number 18)
(서열번호 19)5'-aattcAAAAAACCTATTGAGATAGACTTCCAAactagtaTTGGAAGTCTATCTCAATAGGg-3'
(Sequence number 19)
그 결과, 녹다운한 암세포에서는 음성대조군에 비해 Huh7과 SNU449에서 85-88% 과 71-72%의 PTGFRN mRNA의 감소 효율을 나타내었다 (도 5a). 또한 PTGFRN이 녹다운 된 세포를 이용하여 상기 실시예 3과 같이 유세포 분석(flow cytometry)을 진행한 결과, PTGFRN을 녹다운한 세포에서는 63-D7 항체의 결합이 Huh7과 SNU449 각각에서 51-75% 과 71-78% 정도 감소하는 것을 관찰할 수 있어 PTGFRN 발현이 성공적으로 녹다운 되었음을 알 수 있다 (도 5b, 도 5c).As a result, the knocked-down cancer cells showed a reduction efficiency of PTGFRN mRNA of 85-88% and 71-72% in Huh7 and SNU449, respectively, compared to the negative control group (Fig. 5a). In addition, when flow cytometry analysis was performed using cells with PTGFRN knocked-down as in Example 3, it was observed that the binding of 63-D7 antibody was reduced by 51-75% and 71-78% in Huh7 and SNU449, respectively, indicating that PTGFRN expression was successfully knocked down (Fig. 5b, Fig. 5c).
6-2. siRNA 및 shRNA를 이용한 PTGFRN 녹다운 간암세포에서 암세포 클론원성 생존분석6-2. Clonogenic survival analysis of cancer cells in PTGFRN knockdown liver cancer cells using siRNA and shRNA
다음으로, 간암세포의 생존 및 증식에서 PTGFRN의 역할을 조사하기 위해, 상기의 PTGFRN 녹다운 Huh7 세포와 SNU449 세포를 사용하여 클론원성 생존 분석(clonogenic survival assay)을 수행하였다. 상기의 실험을 통해 제조한 PTGFRN 녹다운 Huh7 세포를 0.05% Trypsin-EDTA(Welgene, Gyeongsan, Korea)로 떼어내고 10% fetal bovine serum(Corning)을 포함한 세포 배양 배지로 중화한 후, 세포를 40 μm 스트레이너(SPL, Pocheon, Korea)에 통과시켜 단일세포로 준비하였다. 6웰 플레이트에 웰당 2.0 X 103개씩 깔아 세포들이 흩어져서 단일세포로 부착하도록 하고 14일 동안 37 ℃, 5% CO2 조건에서 배양한 후, 형성된 콜로니(colony)를 0.5%(w/v)크리스탈 바이올렛 용액으로 염색하고 전체 콜로니의 수를 분석하였다. PTGFRN 녹다운 Huh7 세포와 SNU449 세포에서 클론원성 생존 분석을 한 결과, 각각 41-43% 및 29-35% 감소하는 것을 볼 수 있다(도 5d, 도 5e). 이러한 결과는 PTGFRN이 저밀도의 스트레스 하에서 간암세포 생존을 향상시키고 증식을 촉진하는 중요한 세포 표면 분자임을 보여준다.Next, to investigate the role of PTGFRN in the survival and proliferation of liver cancer cells, a clonogenic survival assay was performed using the PTGFRN knockdown Huh7 cells and SNU449 cells described above. The PTGFRN knockdown Huh7 cells prepared through the above experiment were detached with 0.05% Trypsin-EDTA (Welgene, Gyeongsan, Korea) and neutralized with cell culture medium containing 10% fetal bovine serum (Corning). The cells were passed through a 40 μm strainer (SPL, Pocheon, Korea) to prepare single cells. 2.0 × 103 cells per well were plated in a 6-well plate, and the cells were allowed to disperse and attach as single cells. After culturing for 14 days at 37°C under 5% CO2 conditions, the formed colonies were stained with 0.5% (w/v) crystal violet solution, and the total number of colonies was analyzed. Clonogenic survival assays in PTGFRN knockdown Huh7 cells and SNU449 cells showed 41-43% and 29-35% decreases, respectively (Fig. 5d, Fig. 5e). These results demonstrate that PTGFRN is an important cell surface molecule that enhances hepatoma cell survival and promotes proliferation under low-density stress.
항체 63-D7의 항원인 PTGFRN이 간암세포의 클론원성 생존에 장기적으로 어떻게 영향을 미치는지 분석하기 위해, 지속적으로 PTGFRN 유전자를 표적하는 shRNA를 이용하여 간암세포주에서 지속적인 유전자 녹다운을 진행하였다. 음성 타겟 scramble과 PTGFRN을 타겟으로 하는 shRNA 서열#1과 #2 (TRCN0000057448, TRCN00000057452)를 EZ-tet-PLKO벡터(Addgene, 85996)에 삽입하여 인간 배아신장세포 HEK293FT세포에 psPAX2, pMD2G와 함께 트랜스펙션 하였다(표 2). 트랜스펙션 후 약 48시간, 72시간의 세포배양액을 회수하여 간암세포주 Huh7, SNU449에 감염시켰다. 일주일간 10% Fetal bovine serum(Corning) 및 2 μg/ml의 puromycin(Gibco)이 포함된 RPMI-1640 medium으로 타겟 유전자가 삽입된 세포를 선별하였다. 선별이 끝난 후의 세포를 10% Fetal Bovine Serum(Corning)과 100ng/ml의 doxycycline(Sigma)이 포함된 RPMI-1640배지로 배양하여 녹다운을 유도한 후, 72시간 배양하였다. 배양된 세포는 0.05% Trypsin-EDTA(Welgene)로 떼어내고 10% fetal bovine serum(Corning)을 포함하는 세포 배양 배지로 중화한 후 수확하였다. 수확한 세포에서 PTGFRN 단백질 발현을 웨스턴 블롯으로 분석한 결과, Huh7과 SNU449 각각에서 57-84% 과 88-89%로 PTGFRN발현이 감소함을 관찰하였다(도 6a). 같은 세포를 상기 실시예 3의 유세포 분석방법으로 63-D7의 결합을 분석한 결과 두 종의 shPTGFRN(shPTGFRN#1, shPTGFRN#2)에 의해 간암세포 Huh7 및 SNU449 모두에서 세포표면 PTGFRN발현이 각각 44-49% 및 52-61%로 녹다운이 잘 되었음을 확인할 수 있었다(도 6b, 도 6c). 이 중에서 SNU449세포를 사용하여 상기와 동일하게 클론원성 생존분석을 수행하였으며, 그 결과, shPTGFRN#1에 의해서는 90% 감소하고 (도 6d, 도 6e) shPTGFRN#2에 의해서는 47% 감소하는 것을 관찰하였다 (도 6f, 도 6g). 이러한 결과는 PTGFRN이 간암세포에서 지속적으로 암줄기세포성을 제공하며, 간암세포 생존을 향상시키고 증식을 촉진하는 중요한 세포 표면 분자라는 것을 보여준다.To analyze how PTGFRN, the antigen of antibody 63-D7, affects the clonogenic survival of liver cancer cells in the long term, continuous gene knockdown was performed in liver cancer cell lines using shRNA targeting the PTGFRN gene. The negative target scramble and
6-3. PTGFRN 녹다운 간암 세포주에서 암세포 이동 및 침윤성 분석6-3. Analysis of cancer cell migration and invasion in PTGFRN knockdown liver cancer cell lines
다음으로 간암세포의 이동 및 전이에 있어 PTGFRN의 역할을 조사하기 위해, 상기의 shPTGFRN#1, shPTGFRN#2로 PTGFRN을 녹다운 한 Huh7과 SNU449 세포를 사용하여 트랜스웰 실험을 수행하였다. 상기의 실험을 통해 제조한 PTGFRN 녹다운 Huh7 세포를 0.05% Trypsin-EDTA(Welgene)로 떼어내고 10% fetal bovine serum (Corning)을 포함하는 세포 배양 배지로 중화한 후, 세포를 40 μm 스트레이너(SPL)에 통과시켜 단일 세포로 준비하였다.Next, to investigate the role of PTGFRN in the migration and metastasis of liver cancer cells, transwell experiments were performed using Huh7 and SNU449 cells in which PTGFRN was knocked down with the
세포의 이동성을 평가하기위해, 3.0 x 104의 세포를 RPMI1640 배지에 희석한 후 트랜스웰 챔버에 넣어 주고, 아래에 10% Fetal Bovine Serum (Corning)이 포함된 RPMI-1640배지를 chemoattractant로 이용해 이동을 유도하였다. 이후 5% CO2, 95% air가 공급되는 37 ℃ 배양기에서 37시간 동안 배양한 후 PBS(pH 7.4)로 두 번 세척하고, 이동하지 않은 세포를 면봉으로 제거한 다음 4% PFA로 고정하였다. 고정된 세포는 0.5%의 크리스탈 바이올렛 용액으로 염색하였으며 3번 반복하여 통계적으로 나타냈다. 그 결과 PTGFRN 녹다운 된 Huh7세포 경우 72%, SNU449 세포의 경우 74-85% 이동성이 감소하였다(도 7a, 도 7b).To evaluate cell motility, 3.0 x 10 4 cells were diluted in RPMI1640 medium and placed into a transwell chamber, and RPMI-1640 medium containing 10% Fetal Bovine Serum (Corning) was used as a chemoattractant to induce migration. After culturing for 37 h in a 37 ℃ incubator supplied with 5% CO 2 and 95% air, the cells were washed twice with PBS (pH 7.4), and non-migrated cells were removed with a cotton swab and fixed with 4% PFA. The fixed cells were stained with 0.5% crystal violet solution, which was repeated three times and presented statistically. As a result, migration was reduced by 72% in PTGFRN knockdown Huh7 cells and 74-85% in SNU449 cells (Fig. 7a, Fig. 7b).
간암세포의 전이를 위한 침윤성을 알아보기 위해 250 μg/ml의 Matrigel이 희석된 RPMI-1640 용액 0.1ml를 트랜스웰에 37 ℃에서 2시간 동안 코팅한 후 위 실험과 동일하게 수행하였다. PTGFRN 녹다운 된 Huh7 세포의 경우 82-93%, SNU449 세포의 경우 57-91% 침윤성이 감소하였다(도 7c, 도 7d).To investigate the invasiveness for metastasis of liver cancer cells, 0.1 ml of RPMI-1640 solution containing 250 μg/ml Matrigel was coated on transwells for 2 hours at 37°C, and the same experiment as above was performed. Invasiveness was reduced by 82-93% in PTGFRN knockdown Huh7 cells and by 57-91% in SNU449 cells (Fig. 7c, 7d).
이와 같이, PTGFRN의 결핍은 Huh7 및 SNU449 세포 모두에서 이동성과 침윤성을 감소시켰으며 이러한 결과는 PTGFRN이 간암세포에서 전이에 중요한 이동과 침윤성을 촉진하는 중요한 세포 표면 분자라는 것을 보여준다.Thus, PTGFRN deficiency reduced motility and invasiveness in both Huh7 and SNU449 cells, and these results indicate that PTGFRN is an important cell surface molecule that promotes migration and invasiveness, which are important for metastasis in hepatocellular carcinoma cells.
6-4. PTGFRN 녹다운 간암세포의 세포 확산 분석6-4. Cell proliferation analysis of PTGFRN knockdown liver cancer cells
PTGFRN이 녹다운된 간암세포 Huh7과 SNU449가 여러 세포외기질(Extracellular Matrix, ECM)에 잘 결합하는지 분석하기 위해서, 세포 확산 분석(Cell Spreading assay) 실험을 수행하였다. 세포 확산 실험을 위해서 200 μg/ml의 Matrigel, 20 μg/ml의 CollagenⅠ, 20 μg/ml의 Collagen Ⅳ, 0.1%의 Gelatin을 코팅한 후 2시간 동안 5% CO2, 95% air가 공급되는 37 ℃ 배양기에서 배양하였다. 이후 PTGFRN 녹다운 간암세포 Huh7과 SNU449를 준비하여 5.0 X 104 cell/ml 농도로 희석한 후, PBS(pH 7.4)로 2회 세척한 플레이트에 분주하여 20-60분간 배양하였다. 이후 4% Paraformaldehyde로 상온에서 10분간 고정하고, PBS(pH 7.4)로 세척한 후 세포의 사진을 현미경으로 관찰하였다. 현미경으로 관찰한 세포의 사진을 Image J 프로그램을 이용하여 분석하였다. Huh7 세포 분석에서 shScramble에서는 세포가 바닥에 붙어 퍼진 모양의 세포(spread cell)가 많지만, 두 종의 shPTGFRN에서는 세포가 바닥에 잘 붙지 못하여 세포 모양이 동글동글하게 떠있는 것(unspread cell)이 많은 것을 볼 수 있다(도 8a, 도 8b). 동일한 실험을 PTGFRN 녹다운 SNU449 세포에서는 수행한 결과 세포외기질에 대한 결합이 감소함을 알 수 있다(도 8c, 도 8d). 같은 실험을 3회 반복한 결과를 통계적으로 분석해보면, Huh7에서는 shPTGFRN 종류에 상관없이 녹다운의 경우에는 Matrigel을 제외한 Collagen I, Collagen IV 및 Gelatin 세포외기질에서 간암세포의 세포확산능이 감소하는 것을 관찰하였다(도 8b). SNU449에서는 shPTGFRN#2 녹다운 경우 Matrigel에 대한 세포확산능에서는 차이가 없지만, 나머지 세 종류의 세포외기질에 대해서는 세포확산능이 감소하는 것을 관찰할 수 있다(도 8d). 이러한 결과는 PTGFRN이 간암세포의 대부분의 세포외기질에 대한 부착 및 확산에 중요함을 보여준다.To analyze whether PTGFRN knockdown liver cancer cells Huh7 and SNU449 bind well to various extracellular matrices (ECM), a cell spreading assay experiment was performed. For the cell spreading experiment, 200 μg/ml Matrigel, 20 μg/ml CollagenⅠ, 20 μg/ml CollagenⅣ, and 0.1% Gelatin were coated and cultured in an incubator at 37℃ with 5% CO2 and 95% air for 2 hours. After that, PTGFRN knockdown liver cancer cells Huh7 and SNU449 were prepared, diluted to a concentration of 5.0 X 104 cells/ml, and then dispensed onto plates that were washed twice with PBS (pH 7.4) and cultured for 20-60 minutes. After fixing with 4% paraformaldehyde for 10 minutes at room temperature, the cells were washed with PBS (pH 7.4) and observed under a microscope. The microscopic cell photographs were analyzed using the Image J program. In the Huh7 cell analysis, in shScramble, there were many spread cells that were attached to the bottom, but in the two types of shPTGFRN, there were many unspread cells that were round and floating because the cells did not attach well to the bottom (Fig. 8a, Fig. 8b). When the same experiment was performed on PTGFRN knockdown SNU449 cells, it was found that the binding to the extracellular matrix was reduced (Fig. 8c, Fig. 8d). When the results of repeating the same experiment three times were statistically analyzed, in Huh7, regardless of the type of shPTGFRN, the cell proliferation ability of liver cancer cells was observed to decrease in Collagen I, Collagen IV, and Gelatin extracellular matrices, except for Matrigel, in the case of knockdown (Fig. 8b). In SNU449, there was no difference in cell proliferation ability on Matrigel when
6-5. PTGFRN 녹다운 간암세포의 NK 세포에 대한 면역회피능력 분석6-5. Analysis of the immune evasion ability of PTGFRN knockdown liver cancer cells against NK cells
암세포는 면역세포의 세포독성을 회피할 때 잘 자랄 수 있기 때문에, 대부분의 진행성 암세포는 면역 체크포인트 분자(immune checkpoint molecule)를 발현하여 자신을 공격하는 면역세포들의 공격을 피할 수 있는 능력을 가진다. 간암세포에서 발현하는 PTGFRN 분자가 면역회피에 중요한 역할을 하는 면역 체크포인트 분자로서 역할을 하는지 확인하기 위하여, 상기의 shScramble 및 shPTGFRN#2를 이용해 만든 PTGFRN 녹다운 SNU449 세포와 NK 세포주인 NK92를 공동 배양할 때 세포독성 정도를 분석하였다.Since cancer cells can grow well when they evade the cytotoxicity of immune cells, most advanced cancer cells have the ability to avoid attacks by immune cells attacking them by expressing immune checkpoint molecules. To confirm whether the PTGFRN molecule expressed in liver cancer cells serves as an immune checkpoint molecule that plays an important role in immune evasion, the degree of cytotoxicity was analyzed when PTGFRN knockdown SNU449 cells generated using the above shScramble and
상기의 실험을 통해 제조한 PTGFRN 녹다운 SNU449 세포를 0.05% Trypsin-EDTA(Welgene, Gyeongsan, Korea)로 떼어내고 10% fetal bovine serum(Corning), 200 ng/ml 독시사이클린(doxycycline)(Sigma)을 포함하는 세포 배양 배지로 중화한 후, 12웰 플레이트에 웰당 2.0 X 104개씩의 세포를 분주하여 37 ℃, 5% CO2 조건의 인큐베이터에서 하루 배양하였다. 하루 뒤, 400 U/ml IL2(Peprotech), 10% fetal bovine serum(Corning), 200 ng/ml 독시사이클린(sigma)을 포함한 배지에 이펙터 세포(effector cell, NK92):타겟 세포(target cell, shPTGFRN-SNU449)의 비율(E:T 비율)을 각각 1:1, 2.5:1 및 5:1로 하여 섞은 후 분주하고, 37 ℃, 5% CO2 조건에서 2일 동안 배양하였다. 배양 후에 부유하는 NK92 세포를 세척하여 제거하고 바닥에 붙어 생존한 SNU449 타겟 세포를 0.5%(w/v) 크리스탈 바이올렛 용액으로 염색하였다. 플레이트를 건조한 후, 크리스탈 바이올렛을 50% 에탄올, 0.1M sodium citrate 용액에 녹여 540nm에서 흡광도(OD540)를 측정하였다. 상대적인 OD540을 비교했을 때, shPTGFRN#2는 낮은 클론원성 생존능으로 인해 shScramble에 비해 약 50% 더 낮은 OD540을 보여주고 있으며, NK92와 공동 배양시에는 추가적으로 다시 약 50% 더 낮은 OD540을 보여준다(도 9a, 도 9b). 이러한 결과는 NK세포에 의한 간암세포의 사멸이 PTGFRN이 결핍될 때 더욱 증가한다는 것을 보여준다. 다음으로 E:T 비율을 1:1, 2.5:1 및 5:1로 하여 NK세포의 수를 증가시켰을 때 간암세포의 사멸 정도를 다음 수식을 이용해 분석하였다. PTGFRN knockdown SNU449 cells manufactured through the above experiment were detached with 0.05% Trypsin-EDTA (Welgene, Gyeongsan, Korea) and neutralized with cell culture medium containing 10% fetal bovine serum (Corning) and 200 ng/ml doxycycline (Sigma). Then, 2.0 X 104 cells were dispensed per well in a 12-well plate and cultured in an incubator at 37°C and 5% CO2 for one day. One day later, the effector cells (NK92):target cells (shPTGFRN-SNU449) were mixed at an E:T ratio of 1:1, 2.5:1, and 5:1 in a medium containing 400 U/ml IL2 (Peprotech), 10% fetal bovine serum (Corning), and 200 ng/ml doxycycline (Sigma), dispensed, and cultured for 2 days at 37°C, 5% CO2 . After culture, floating NK92 cells were washed and removed, and the SNU449 target cells that remained attached to the bottom and survived were stained with 0.5% (w/v) crystal violet solution. After drying the plate, the crystal violet was dissolved in 50% ethanol, 0.1 M sodium citrate solution, and the absorbance (OD540) was measured at 540 nm. When comparing the relative OD540,
[수식][formula]
NK cytotoxicity (%) = {1 - (target OD value/No NK OD value)}*100NK cytotoxicity (%) = {1 - (target OD value/No NK OD value)}*100
그 결과, NK92 세포 수가 증가할수록 간암세포의 사멸 정도가 증가하는 것을 볼 수 있다(도 9c). 이러한 결과는 PTGFRN이 암세포에서 면역 체크포인트 분자로 작용해 NK세포에 의한 세포사멸을 억제함으로써 암세포에 숙주의 면역을 회피하는 능력을 제공하는 것으로 보인다.As a result, it can be seen that as the number of NK92 cells increases, the degree of apoptosis of liver cancer cells increases (Fig. 9c). These results suggest that PTGFRN acts as an immune checkpoint molecule in cancer cells to suppress apoptosis by NK cells, thereby providing cancer cells with the ability to evade the host's immunity.
6-6. 간암에서 PTGFRN의 결핍에 따른 신호전달 물질의 변화 분석6-6. Analysis of changes in signaling substances due to PTGFRN deficiency in liver cancer
상기 실시예 6-3과 같이 녹다운 된 세포를 이용하여 암의 전이에 있어 중요한 주요 신호전달물질인 FAK 및 Src와 면역관문 분자로 알려진 B7-H3의 발현을 확인하였다.As in Example 6-3 above, the expression of FAK and Src, which are key signaling molecules important for cancer metastasis, and B7-H3, known as an immune checkpoint molecule, was confirmed using the knocked-down cells.
녹다운 된 세포를 회수하여 인산완충용액(Ph 7.4)로 두 번 세척한 후, RIPA lysis buffer(50mM Tris-HCl, pH 7.5, 150 mM NaCl, 1% NP-40, 0.1% SDS, 0.5% Deoxycholate)를 이용해 세포를 용해시켰다. 이후 4 ℃, 12000 rpm에서 30분간 원심분리하고, 상등액을 5X 샘플 완충액과 섞어 단백질 샘플을 준비하였다. 이후 8-10%의 SDS-PAGE 겔에서 분리하고 니트로셀룰로스 막으로 옮기는 웨스턴 블롯팅을 진행하였다. 5%의 탈지분유를 이용하여 블로킹하고, 각각의 타겟 항체인 토끼 항 PTGFRN 다클론항체(abcam, 97567), 토끼 항 B7-H3 다클론항체(sinobiological, 11188-RP02), 토끼 항 FAK 다클론항체(Cell Signaling Technology,3285), 토끼 항 p-FAK 다클론항체(CST,3283), 토끼 항 Src 단일클론항체(CST,2109), 토끼 항 p-Src 단일클론항체(CST, 6943), 토끼 항 GAPDH 다클론항체(CSB, PA00025A0Rb)를 5%의 BSA에 섞어 4 ℃에서 16시간 동안 반응시켰다. 0.1% TBST(Tris-Buffered Saline, 0.1% tween)로 10분간 3번의 세척 후, 항 토끼 IgG-HRP(1:12,000)와 실온에서 한시간 동안 반응시킨 후, 타겟 단백질을 ECL 검출 키트(Advansta)로 확인하였다(도 10).The knocked-down cells were harvested, washed twice with phosphate buffer (Ph 7.4), and lysed using RIPA lysis buffer (50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1% NP-40, 0.1% SDS, 0.5% Deoxycholate). The cells were centrifuged at 4°C, 12,000 rpm for 30 minutes, and the supernatant was mixed with 5X sample buffer to prepare a protein sample. Afterwards, they were separated on an 8-10% SDS-PAGE gel and transferred to a nitrocellulose membrane for Western blotting. Blocking was performed using 5% skim milk powder, and each target antibody, rabbit anti-PTGFRN polyclonal antibody (abcam, 97567), rabbit anti-B7-H3 polyclonal antibody (sinobiological, 11188-RP02), rabbit anti-FAK polyclonal antibody (Cell Signaling Technology, 3285), rabbit anti-p-FAK polyclonal antibody (CST, 3283), rabbit anti-Src monoclonal antibody (CST, 2109), rabbit anti-p-Src monoclonal antibody (CST, 6943), rabbit anti-GAPDH polyclonal antibody (CSB, PA00025A0Rb), was mixed in 5% BSA and reacted at 4°C for 16 hours. After washing three times with 0.1% TBST (Tris-Buffered Saline, 0.1% tween) for 10 minutes each, the sample was reacted with anti-rabbit IgG-HRP (1:12,000) at room temperature for one hour, and the target protein was confirmed using an ECL detection kit (Advansta) (Fig. 10).
그 결과, PTGFRN의 감소는 면역관문 조절 분자인 B7-H3의 발현을 감소시키고, FAK 신호전달 경로인 p-FAK 및 p-Src의 발현을 감소시켰다. 이러한 결과로부터 PTGFRN은 FAK 신호전달경로를 통해 암세포의 이동 및 침윤성을 조절한다는 것을 알 수 있다.As a result, the decrease in PTGFRN decreased the expression of B7-H3, an immune checkpoint regulatory molecule, and decreased the expression of p-FAK and p-Src, FAK signaling pathways. These results suggest that PTGFRN regulates the migration and invasiveness of cancer cells through the FAK signaling pathway.
6-7. 세포막에서의 PTGFRN 사이의 cis 및 trans 결합 측정6-7. Measurement of cis and trans binding between PTGFRN in the cell membrane
세포막에서 발현하는 PTGFRN이 세포 사이의 cluster를 이루는데 기여한다면, 혈중 암세포에 발현되는 PTGFRN이 암세포의 성장이나 생존 면역회피성에 도움을 줄 것이다. 이를 관찰하기 위해, 6웰 플레이트에 웰당 2.5 X 105개의 세포를 분주한 후, 다음 날 12 μg의 PEI, pcDNA3.1(+)PTGFRN-FLAG 벡터와 pcDNA3.1(+)-PTGFRN-myc 벡터를 함께 각각 4 μg씩 되도록 TOM(Welgene)에 희석한 후 실온에서 5분간 반응시켰다. 이후, PEI를 희석한 용액과 벡터를 희석한 용액을 섞어 실온에서 20분간 반응시킨 후, 한 HEK293FT 세포에 함께 처리하고 24시간을 37 ℃, 5% CO2의 배양기에서 배양하였다. 또한, pcDNA3.1(+)PTGFRN-FLAG 벡터와 pcDNA3.1(+)-PTGFRN-myc 벡터를 각각 준비해서 두개의 HEK293FT 세포 웰에 따로 반응시킨 후, 24시간이 지난 시점에서, 새로운 배양 배지로 교체하여 48시간을 더 배양하고, 실시예4와 같은 방법으로 세포 용해 완충액을 처리하여 세포 용출액을 준비하였다. BCA(Thermo)를 이용하여 각각의 세포 용출액을 50 μg씩 준비하고, 각 용출액을 생쥐 항-FLAG 단일클론항체, 토끼 항-myc tag 단일클론항체, 생쥐 63-D7 항체를 이용하여 면역침강시킨 후 항-FLAG 단일클론항체, 토끼 항-myc tag 단일클론항체로 웨스턴 블롯팅하여 분석하였다.If PTGFRN expressed in the cell membrane contributes to forming clusters between cells, PTGFRN expressed in circulating cancer cells will help in the growth or survival and immune evasion of cancer cells. To observe this, 2.5 X 10 5 cells were seeded per well in a 6-well plate, and the next day, 12 μg of PEI, pcDNA3.1(+)PTGFRN-FLAG vector, and pcDNA3.1(+)-PTGFRN-myc vector were diluted together in TOM (Welgene) to 4 μg each, and reacted at room temperature for 5 minutes. After that, the diluted PEI solution and the diluted vector solution were mixed and reacted at room temperature for 20 minutes, and then treated together in one HEK293FT cell and cultured for 24 hours in an incubator with 37°C and 5% CO 2 . Also, pcDNA3.1(+)PTGFRN-FLAG vector and pcDNA3.1(+)-PTGFRN-myc vector were prepared respectively and reacted separately in two HEK293FT cell wells, and after 24 hours, the culture medium was replaced with new one, cultured for another 48 hours, and cell lysates were prepared by treating cell lysis buffer in the same manner as in Example 4. 50 μg of each cell lysate was prepared using BCA (Thermo), and each lysate was immunoprecipitated using mouse anti-FLAG monoclonal antibody, rabbit anti-myc tag monoclonal antibody, and mouse 63-D7 antibody, and then analyzed by Western blotting with anti-FLAG monoclonal antibody and rabbit anti-myc tag monoclonal antibody.
그 결과, PTGFRN을 함께 발현시킨 세포 용출액에서는 FLAG과 myc tag이 모두 검출되지만 (도 11a), 각각을 단일로 트랜스펙션 한 후 섞어서 면역침강을 진행했을 때는 항-myc항체에 면역침강된 PTGFRN에서는 FLAG-tag된 PTGFRN이 검출되지 않고 항-FLAG항체에 면역침강된 PTGFRN에서는 myc-tag된 PTGFRN이 검출되지 않았다 (도 11b). 따라서 이를 통해 PTGFRN이 세포막에서 trans- 보다는 cis-형태로 서로 상호작용함을 확인할 수 있다.As a result, both FLAG and myc tags were detected in the cell lysate co-expressing PTGFRN (Fig. 11a), but when they were transfected individually and then mixed to perform immunoprecipitation, FLAG-tagged PTGFRN was not detected in PTGFRN immunoprecipitated with anti-myc antibody, and myc-tagged PTGFRN was not detected in PTGFRN immunoprecipitated with anti-FLAG antibody (Fig. 11b). Therefore, this confirms that PTGFRN interacts with each other in a cis- rather than a trans- form on the cell membrane.
6-8. 세포막에서의 PTGFRN 결합물질 분석6-8. Analysis of PTGFRN binding substances in the cell membrane
세포막에서 PTGFRN과 결합을 하는 다른 단백질들을 찾기 위해, 상기 실시예 4와 같이 SNU449 세포 용출액을 준비한 후, 생쥐 항-PTGFRN 단일클론항체, 생쥐 63-D7 단일클론항체, 생쥐 항-SLC3A2 단일클론항체, 생쥐 항-B7-H3 단일클론항체를 이용하여 면역침강시킨 후, 생쥐 항 PTGFRN 단일클론항체(α-PTGFRN)를 이용하여 웨스턴 블롯팅을 진행한 결과 PTGFRN은 SLC3A2(CD98hc)와 B7-H3 (CD276) 과 결합함을 확인하였다(도 11c).To find other proteins that bind to PTGFRN in the cell membrane, SNU449 cell lysates were prepared as in Example 4, and then immunoprecipitated using mouse anti-PTGFRN monoclonal antibody, mouse 63-D7 monoclonal antibody, mouse anti-SLC3A2 monoclonal antibody, and mouse anti-B7-H3 monoclonal antibody. Western blotting was performed using mouse anti-PTGFRN monoclonal antibody (α-PTGFRN). As a result, it was confirmed that PTGFRN binds to SLC3A2 (CD98hc) and B7-H3 (CD276) (Fig. 11c).
B7-H3는 암세포에서의 면역관문 조절 물질로 알려져 있으므로, B7-H3와 PTGFRN의 상호작용을 다시 한번 검증하기 위해서 6웰 플레이트에 HEK293FT 세포를 웰당 2.5 X 105의 세포 수로 분주한 후, 다음날 12 μg의 PEI, myc tag을 가진 PTGFRN 벡터(pcDNA3.1(+)PTGFRN-myc)와 FLAG-tag을 가진 PTGFRN 벡터(pcDNA3.1(+)-B7-H3-FLAG)를 함께 각각 4 μg씩 되도록 TOM(Welgene, Gyeongsan, Korea)에 희석한 후 실온에서 5분간 반응시켰다. 이후, PEI를 희석한 용액과 상기의 벡터를 희석한 용액을 섞어 실온에서 20분 반응시키고, HEK293FT세포에 처리하여 트랜스펙션 하고 24시간동안 37 ℃, 5% CO2의 배양기에서 배양하였다. 24시간이 지난 후 새로운 배양배지로 교체하고 48시간을 더 배양한 후 실시예 4와 같이 세포 용출액을 준비하였다. BCA(Thermo Fischer Scientific)를 이용하여 각각의 세포 용출액을 50μg씩 준비한 후, 항-FLAG, 항-B7-H3, 항-myc, 63-D7로 면역침강하였다. 면역침강물을 항-myc, 항-FLAG 항체로 검출한 결과 항-FLAG 항체에 의한 면역침강물에서 PTGFRN이 검출되고 항-myc항체에 의한 면역침강물에서 B7-H3가 검출되었으므로 PTGFRN과 B7-H3가 서로 물리적으로 결합하여 상호작용함을 알 수 있다(도 11d). 이러한 결과는 면역관문 조절 분자로 알려진 B7-H3를 통해서 PTGFRN이 면역조절에 관여할 것을 제시한다.Since B7-H3 is known to be an immune checkpoint regulator in cancer cells, in order to verify the interaction between B7-H3 and PTGFRN once again, HEK293FT cells were seeded at 2.5 X 10 5 cells per well in a 6-well plate. The next day, 12 μg of PEI, PTGFRN vector with myc tag (pcDNA3.1(+)PTGFRN-myc) and PTGFRN vector with FLAG tag (pcDNA3.1(+)-B7-H3-FLAG) were diluted together with 4 μg each in TOM (Welgene, Gyeongsan, Korea) and reacted at room temperature for 5 minutes. After that, the diluted PEI solution and the diluted vector solution were mixed and reacted at room temperature for 20 minutes, treated to transfect HEK293FT cells, and cultured in an incubator with 5% CO 2 at 37°C for 24 hours. After 24 hours, the culture medium was replaced with new medium, and after culturing for another 48 hours, the cell lysate was prepared as in Example 4. Using BCA (Thermo Fischer Scientific), 50 μg of each cell lysate was prepared, and then immunoprecipitated with anti-FLAG, anti-B7-H3, anti-myc, and 63-D7. The immunoprecipitates were detected with anti-myc and anti-FLAG antibodies. PTGFRN was detected in the immunoprecipitate by anti-FLAG antibody, and B7-H3 was detected in the immunoprecipitate by anti-myc antibody. Therefore, it can be seen that PTGFRN and B7-H3 physically bind and interact with each other (Fig. 11d). These results suggest that PTGFRN is involved in immune regulation through B7-H3, which is known as an immune checkpoint regulatory molecule.
실시예7. 63-D7 양성 간암세포의 부착성, 암줄기세포성 및 클론원성 분석Example 7. Analysis of adhesion, cancer stemness, and clonogenicity of 63-D7 positive liver cancer cells
63-D7의 항원 PTGFRN이 간암세포의 부착에 어떻게 영향을 미치는지 분석하기 위해서, 63-D7항체로 Huh7과 HepG2 세포를 63-D7 양성과 음성으로 마그네틱 비드를 사용하여 sorting하였다.To analyze how the antigen PTGFRN of 63-D7 affects the adhesion of hepatoma cells, Huh7 and HepG2 cells were sorted into 63-D7 positive and negative using magnetic beads with 63-D7 antibody.
Sorting을 하기 위해서 63-D7-biotin 항체를 Neutravidin-접합된 마그네틱 비드(Thermo Fishcer Scientific)를 사용하여 제조사의 프로토콜에 따라 분리하였다. 항체에 붙은 세포는 63-D7(+), 붙지 않은 세포는 63-D(-)로 분리하여 준비하였다. 다음으로 Matrigel로 12웰 플레이트를 코팅하고, 최소 1시간 동안 5% BSA를 포함하는 무혈청 DMEM/F12 배지에서 37 ℃에서 블로킹하였다. 63-D7 (+), 63-D7(-) 간암세포를 웰당 105개 세포로 접종하고 37 ℃에서 6시간 동안 반응하였다. 부착되지 않은 세포를 제거하기 위해, 플레이트를 PBS(pH 7.4)로 세척하였다. 부착 세포를 실온에서 30분간 2% 에탄올 중 0.5% 크리스탈 바이올렛(Sigma-Aldrich)으로 염색하고, 염색된 세포는 0.1% SDS에 용해한 후 570 nm에서 흡광도를 측정하였다. 그 결과, 63-D7(+) Huh7 세포는 63-D7(-) Huh7 세포에 비해 43% 부착력이 증가하였고, 63-D7(+) HepG2 세포는 63-D7(-) HepG2 세포에 비해 245% 부착력이 증가하여, 63-D7 양성 간암세포가 음성 간암세포보다 세포외기질에 대한 부착력이 현저히 우수함을 제시한다 (도 12a, 도 12b).For sorting, 63-D7-biotin antibody was separated using Neutravidin-conjugated magnetic beads (Thermo Fishcer Scientific) according to the manufacturer's protocol. Cells attached to the antibody were separated as 63-D7(+), and non-attached cells were prepared as 63-D(-). Next, 12-well plates were coated with Matrigel and blocked in serum-free DMEM/F12 medium containing 5% BSA at 37°C for at least 1 hour. 63-D7(+), 63-D7(-) hepatoma cells were seeded at 105 cells per well and reacted at 37°C for 6 hours. To remove non-attached cells, the plates were washed with PBS (pH 7.4). Adherent cells were stained with 0.5% crystal violet (Sigma-Aldrich) in 2% ethanol for 30 min at room temperature, and the stained cells were dissolved in 0.1% SDS and measured for absorbance at 570 nm. As a result, 63-D7(+) Huh7 cells showed a 43% increase in adhesion compared to 63-D7(-) Huh7 cells, and 63-D7(+) HepG2 cells showed a 245% increase in adhesion compared to 63-D7(-) HepG2 cells, suggesting that 63-D7-positive hepatoma cells have significantly better adhesion to the extracellular matrix than do negative hepatoma cells (Fig. 12a, Fig. 12b).
한편 63-D7 항원 양성 암세포가 암줄기세포성(Cancer stemness)을 가져서 암의 재발과 약물저항성을 유발하는데 연관되는지 관찰하기 위하여 암줄기세포성이 증가하도록 유도하는 종양구(tumorsphere)를 배양하였다. 종양구를 생성하기 위해서 부착하여 자라는 Huh7 간암세포를 트립신-EDTA (Welgene, 대구, 한국)로 분리하고 40 μm 스트레이너를 통해 여과한 후 실온에서 3분 동안 1500 rpm으로 원심 분리하였다. 이 세포를 부착성이 아주 낮은 플레이트(Corning, SEOUL, KOREA)에 2.1 x 103 세포/cm2로 접종하고 20 ng/ml 섬유아세포 성장인자(FGF2, R&D시스템, 서울, 한국), 20 ng/ml 표피상장인자(EGF, PeproTech, 서울, 한국) 및 1 x B27 보충제(Life Technologies)가 보충된 DMEM/F12 배지(Corning)에서 배양하였다. 배지는 3일마다 교체하였으며 최소 9일 이상 배양하였다. 대표적인 간암줄기세포 마커로 알려진 CD133, CD44 및 EpCAM과, 63-D7 항원인 PTGFRN 분자의 발현 여부를 상기 실시예 3과 같은 방법으로 유세포 분석법을 통해 분석하였다. 그 결과 암줄기세포성이 증가한 종양구 배양에서 암줄기세포 양성 마커들인 CD133, CD44, EpCAM이 부착성 세포일때와 비교할 때, 각각 약 33%, 43%, 59%로 증가하고 이때 63-D7의 결합도 61%로 함께 증가하는 것을 볼 수 있다 (도 12c, 도 12d). 이러한 결과는 63-D7 항원인 PTGFRN가 새로운 간암줄기세포 마커로 작용한다는 것을 제시한다.Meanwhile, to observe whether 63-D7 antigen-positive cancer cells have cancer stemness and are related to cancer recurrence and drug resistance, tumorspheres that induce cancer stemness were cultured. To generate tumorspheres, attached and growing Huh7 liver cancer cells were detached with trypsin-EDTA (Welgene, Daegu, Korea), filtered through a 40 μm strainer, and centrifuged at 1500 rpm for 3 minutes at room temperature. The cells were seeded at 2.1 x 10 3 cells/cm 2 on very low adherent plates (Corning, SEOUL, KOREA) and cultured in DMEM/F12 medium (Corning) supplemented with 20 ng/ml fibroblast growth factor 2 (FGF2, R&D systems, Seoul, Korea), 20 ng/ml epidermal growth factor (EGF, PeproTech, Seoul, Korea), and 1 x B27 supplement (Life Technologies). The medium was changed every 3 days, and the culture was performed for at least 9 days. The expression of CD133, CD44, and EpCAM, known as representative liver cancer stem cell markers, and PTGFRN, a 63-D7 antigen, was analyzed by flow cytometry in the same manner as in Example 3. As a result, in the tumor cell culture with increased cancer stem cell properties, the cancer stem cell-positive markers CD133, CD44, and EpCAM increased by about 33%, 43%, and 59%, respectively, compared to the adherent cells, and at this time, the binding of 63-D7 also increased by 61% (Fig. 12c, Fig. 12d). These results suggest that PTGFRN, a 63-D7 antigen, acts as a new liver cancer stem cell marker.
63-D7 양성 간암세포가 간암줄기세포성을 보여주는지 추가로 분석하기 위하여 위와 동일하게 63-D7으로 sorting한 간암세포를 사용하여, 실시예 6-2과 같이 암줄기세포성을 보여주는 클론원성 생존실험을 수행하였다. 그 결과, Huh7에서는 492%(도 12e, 도 12f), HepG2에서는 56% 클론원성이 증가하는 것을 관찰하였다(도 12g, 도 12h). 이런 결과는 63-D7 양성 간암세포가 음성 간암세포에 비해 현저한 암줄기세포성을 보이며 클론원성 생존능력이 우수한 암줄기세포 성질을 가진다는 것을 제시한다.To further analyze whether 63-D7-positive liver cancer cells show liver cancer stem cell properties, a clonogenic survival experiment showing cancer stem cell properties was performed as in Example 6-2 using liver cancer cells sorted with 63-D7 in the same manner as above. As a result, it was observed that clonogenicity increased by 492% in Huh7 (Fig. 12e, Fig. 12f) and by 56% in HepG2 (Fig. 12g, Fig. 12h). These results suggest that 63-D7-positive liver cancer cells show remarkable cancer stem cell properties compared to negative liver cancer cells and have cancer stem cell properties with excellent clonogenic survival ability.
실시예 8. 간암 및 췌장암 세포에서 63-D7처리 후 내부화 분석Example 8. Internalization analysis after 63-D7 treatment in liver and pancreatic cancer cells
항체를 처리했을 때, 항체가 암세포의 내부로 내부화 (internalization)되는 것은 항체-약물 컨쥬게이트(antibody-drug conjugate, ADC)를 통한 항암 항체 치료제 개발에서 핵심 전제조건이다. 단일클론항체 63-D7의 각종 암세포에 대한 세포막 내부화(internalization)를 확인하기 위해, 유세포 분석(Flow Cytometry)를 수행하였다.When treated with antibodies, internalization of antibodies into cancer cells is a key prerequisite for the development of anticancer antibody therapeutics using antibody-drug conjugates (ADCs). To confirm the internalization of monoclonal antibody 63-D7 into various cancer cells, flow cytometry analysis was performed.
세포를 0.05% Trypsin-EDTA(Welgene)로 떼어내고 10% fetal bovine serum(FBS; VWR, PA, USA)을 포함한 세포 배양 배지로 중화한 후, 세포를 40 μm 스트레이너에 통과시켜 단일세포로 준비하였다. 각 단일세포를 ml당 약 1 x 105개씩 PBA(1% bovine serum albumin, 0.02% NaN3 in PBS)에 섞은 후, 항체 63-D7을 4 ℃에서 30분간 반응시켰다. 항체의 세포 내부화를 유도하기 위해 PBA로 세포를 1회 세척한 후, 세포막 내부화가 일어날 수 있도록 세포를 100 μl의 배양 배지에 현탁하여 30분간 37 ℃에서 반응시켰다. PBA로 1번 세척한 후, 1차 항체와 상응하는 항-마우스 IgG-Alexa flour 488(Invitrogen)를 4 ℃에서 20분간 더 반응시켰다. PBA로 2번 세척한 후, FACSCalibur와 Cell Quest 소프트웨어(BD sciences)를 이용하여 PI(propidium iodide)-음성 세포에 대해서 항체 반응 여부를 분석하였다. 4 ℃에서 세포 표면에 결합한 단일클론항체 63-D7의 형광세기와 37 ℃에서 반응했을 때 63-D7의 형광세기의 정도를 유세포분석으로 비교할 때, 인간 간암세포 Huh7, HepG2, SNU387, SNU449와 인간 췌장암 세포인 BxPC3, SNU213에서 단일클론항체 63-D7결합이 뚜렷이 감소한 것을 볼 수 있다(도 13a, 도 13b). 이는 4 ℃에서 암세포 표면에 결합한 63-D7항체가 37 ℃에서 세포 활성에 의해 세포 내부로 내부화 되어 표면에서의 결합이 감소하는 것을 의미하며, 63-D7이 ADC를 활용한 항체 치료제 개발에 응용할 수 있는 항체라는 것을 제시한다.Cells were detached with 0.05% Trypsin-EDTA (Welgene) and neutralized with cell culture medium containing 10% fetal bovine serum (FBS; VWR, PA, USA), and then passed through a 40 μm strainer to prepare single cells. Approximately 1 × 10 5 cells per ml of each single cell were mixed with PBA (1% bovine serum albumin, 0.02% NaN 3 in PBS) and reacted with antibody 63-D7 at 4 °C for 30 min. To induce antibody internalization, cells were washed once with PBA, and then cells were suspended in 100 μl of culture medium and reacted for 30 min at 37 °C to allow cell membrane internalization. After washing once with PBA, anti-mouse IgG-Alexa flour 488 (Invitrogen) corresponding to the primary antibody was further reacted at 4 °C for 20 min. After washing twice with PBA, antibody reactivity was analyzed for PI (propidium iodide)-negative cells using FACSCalibur and Cell Quest software (BD sciences). When the fluorescence intensity of monoclonal antibody 63-D7 bound to the cell surface at 4 °C was compared by flow cytometry to the degree of fluorescence intensity of 63-D7 when reacted at 37 °C, it could be seen that the binding of monoclonal antibody 63-D7 was markedly reduced in human hepatoma cells Huh7, HepG2, SNU387, SNU449 and human pancreatic cancer cells BxPC3, SNU213 (Fig. 13a, Fig. 13b). This means that the 63-D7 antibody bound to the surface of cancer cells at 4°C is internalized into the cells by cell activity at 37°C, thereby reducing binding on the surface, suggesting that 63-D7 is an antibody that can be applied to the development of antibody therapeutics using ADC.
실시예 9. 약물 접합 63-D7 항체의 간암세포에서 세포 독성Example 9. Cytotoxicity of drug-conjugated 63-D7 antibody in hepatoma cells
앞선 실시예 8을 통해 생쥐 IgG1 불변영역을 가진 63-D7 항체가 간암세포 표면에 결합한 후 37 ℃ 온도 조건에서 세포에 내부화 되는 것을 확인하였다. 위 특성을 바탕으로 63-D7 항체에 항암 약물이 접합되어 있을 때, 63-D7 항체를 매개로 전달된 약물이 암세포 내에서 세포 독성을 나타낼 것으로 가정하고 이를 확인하기 위한 실험을 수행하였다.Through the above Example 8, it was confirmed that the 63-D7 antibody having a mouse IgG1 constant region bound to the surface of liver cancer cells and was internalized into the cells at a temperature of 37°C. Based on the above characteristics, it was assumed that when an anticancer drug is conjugated to the 63-D7 antibody, the drug delivered via the 63-D7 antibody would exhibit cytotoxicity in cancer cells, and an experiment was performed to confirm this.
실험에서 약물이 접합된 mouse IgG Fc 영역 특이적인 2차 항체[anti-mouse IgG(Fc Specific) antibody-drug conjugates(ADCs)]를 사용하였다. 구체적으로는 DNA Alkylation agent인 duocarmycin이 접합된 α-mFc-CL-DMDM(AM-102DD, Moradec, 미국) 항체와 tubulin polymerization inhibitor인 monomethyl auristatin이 접합된 α-mFc-CL-MMAF(AM-102AF, Moradec) 항체를 이용하였다. 먼저 96 well plate(SPL)의 각 웰에 5,000개의 Huh7 세포를 10% FBS가 포함된 RPMI-1640(Biowest, 프랑스) 배지를 이용하여 분주 후 24시간 동안 배양하였다. 다음 날 기존의 배양 배지를 제거한 다음, 여러 농도로 배양 배지에 희석된 63-D7 항체 혹은 mouse IgG isotype control (Invitrogen, 31903)을 100 nM, 10 nM, 1 nM, 0.1 nM, 0.01 nM로 각 웰에 분주 후 37℃에서 10분간 항체를 세포 표면에 결합시켰다. 이후, 12.7nM의 농도의 약물이 접합된 2차 항체를 배양 배지에 희석한다음 1차 항체를 제거하고 각 웰에 분주하였다. 세포를 48시간 동안 37 ℃, 5% CO2 조건에서 배양한 후, 세포 생존율을 알아보기 위해 Cell Counting Kit-8(CCK-8, K1018, APExBIO, 미국)을 사용하였다. 각 웰당 10 μl의 CCK-8을 더해 3시간 동안 반응시키고 plate reader를 이용하여 OD450nm에서의 흡광도를 측정하였다.In the experiment, drug-conjugated mouse IgG Fc region-specific secondary antibodies [anti-mouse IgG (Fc Specific) antibody-drug conjugates (ADCs)] were used. Specifically, α-mFc-CL-DMDM (AM-102DD, Moradec, USA) antibody conjugated with duocarmycin, a DNA alkylation agent, and α-mFc-CL-MMAF (AM-102AF, Moradec) antibody conjugated with monomethyl auristatin, a tubulin polymerization inhibitor, were used. First, 5,000 Huh7 cells were dispensed into each well of a 96-well plate (SPL) using RPMI-1640 (Biowest, France) medium containing 10% FBS, and then cultured for 24 hours. The next day, the existing culture medium was removed, and 63-D7 antibody or mouse IgG isotype control (Invitrogen, 31903) diluted in the culture medium at various concentrations was dispensed into each well at 100 nM, 10 nM, 1 nM, 0.1 nM, and 0.01 nM, and the antibody was allowed to bind to the cell surface for 10 minutes at 37℃. After that, the drug-conjugated secondary antibody at a concentration of 12.7 nM was diluted in the culture medium, the primary antibody was removed, and the antibody was dispensed into each well. After culturing the cells for 48 hours under 5% CO2 conditions at 37℃, Cell Counting Kit-8 (CCK-8, K1018, APExBIO, USA) was used to determine the cell viability. 10 μl of CCK-8 was added to each well, reacted for 3 hours, and the absorbance at OD450nm was measured using a plate reader.
그 결과, α-mFc-CL-DMDM ADC와 함께 100 nM의 63-D7항체를 처리한 경우 세포 생존율이 40%, 10 nM 63-D7 항체를 처리한 경우 20% 감소한 것을 확인하였다(도 14a). α-mFc-CL-MMAF를 사용하여 동일한 실험을 진행한 경우에는 100 nM에서 40%, 10nM에서 20%까지 간암 세포의 생존율이 감소하였다(도 14b). 그러나 생쥐 유래 isotype 항체를 처리한 세포의 생존율은 큰 변화를 보이지 않았다. 이는 실험에서 사용한 약물이 접합된 2차 항체가 생쥐의 Fc 영역에 특이적으로 결합하며, 생쥐 항체인 63-D7과 함께 세포 내부로 들어가 세포 독성을 유발하는 것을 증명한 것이다.As a result, it was confirmed that the cell viability decreased by 40% when 100 nM 63-D7 antibody was treated with α-mFc-CL-DMDM ADC, and by 20% when 10 nM 63-D7 antibody was treated (Fig. 14a). When the same experiment was performed using α-mFc-CL-MMAF, the viability of liver cancer cells decreased by 40% at 100 nM and by 20% at 10 nM (Fig. 14b). However, the viability of cells treated with mouse-derived isotype antibody did not show a significant change. This proves that the drug-conjugated secondary antibody used in the experiment specifically binds to the mouse Fc region and enters the cell together with the mouse antibody 63-D7 to induce cytotoxicity.
따라서 63-D7 항체가 간암세포에 내부화 되며, 이러한 특성을 바탕으로 PTGFRN를 발현하는 간암세포 내부로 약물을 효과적으로 전달함으로써 항암 효과를 나타낼 수 있음을 알 수 있다. 이러한 결과는 63-D7 항체를 사용한 ADC 전략을 이용하여 간암세포와 췌장암 세포의 세포사멸을 유도할 수 있다는 것을 제시한다.Therefore, it can be seen that 63-D7 antibody is internalized into liver cancer cells, and based on this characteristic, it can exhibit anticancer effects by effectively delivering drugs into liver cancer cells expressing PTGFRN. These results suggest that the ADC strategy using 63-D7 antibody can be used to induce apoptosis of liver cancer cells and pancreatic cancer cells.
실시예 10. 간암환자 혈중 암세포에서 63-D7 항원 분석Example 10. Analysis of 63-D7 antigen in blood cancer cells from liver cancer patients
10-1. 면역세포화학적 방법에 의한 간암환자 혈액에서 63-D7 양성 혈중 암세포의 검출10-1. Detection of 63-D7 positive blood cancer cells in the blood of liver cancer patients by immunocytochemical method
PTGFRN이 전이에 중요한 혈중 암세포의 생물학적 특성인 간세포암(Hepatocellular carcinoma, HCC) 세포의 생존, 이동, 침입, 세포부착 및 면역회피를 촉진시킴을 확인하였다(도 5 내지 도 9). 따라서 우리는 PTGFRN가 HCC 환자에서 혈중 암세포를 감지하는 표면 마커 역할을 할 수 있다고 가정했으며, 실제로 혈중 암세포를 검출하는 예비실험에서도 그 가능성을 확인하였다(도 1b, 도 1c).We confirmed that PTGFRN promotes survival, migration, invasion, cell adhesion, and immune evasion of hepatocellular carcinoma (HCC) cells, which are biological characteristics of circulating cancer cells that are important for metastasis (Figs. 5 to 9). Therefore, we hypothesized that PTGFRN could serve as a surface marker to detect circulating cancer cells in HCC patients, and indeed, we confirmed this possibility in preliminary experiments to detect circulating cancer cells (Figs. 1b and 1c).
HCC 환자에서 혈중 암세포를 효율적으로 검출하기 위해 우리는 적혈구를 제거한 다음, CD45 제거 프로토콜에 의해 CD45 양성 세포를 추가로 제거하고 남은 세포를 분석하는 전략을 선택했다(도 15c). 구체적으로, 먼저 간암환자 혈액을 수집 후 6시간 이내에 말초 혈액 단핵 세포(PBMC)를 Ficoll-Paque Plus(GE Healthcare, Seoul, Korea) 구배 원심분리에 의해 건강한 기증자, 간염환자, 또는 HCC 환자의 말초혈액으로부터 분리하였다. PBMC를 0.5% 소 혈청 알부민(BSA) 및 2 mM EDTA를 함유하는 PBS로 세척하고 이 완충액에 1×108 세포/mL의 농도로 재현탁시켰다. 이 백혈구 계열 세포에서 CD45 양성 세포의 제거에 의한 혈중 암세포의 농축은 제공된 프로토콜에 따라 Human CD45 제거 키트(EasySep®, Stem Cells Technologies, Vancouver, BC, Canada)를 사용하여 수행하였다. 먼저 항-CD45 항체를 세포 현탁액에 첨가하여 실온에서 15분간 반응시키고, 덱스트란으로 코팅된 자성 나노 입자를 실온에서 10분간 세포와 함께 반응시킨 후 세포 현탁액을 실온에서 10분 동안 EasySep® big easy magnet(StemCell Technologies, Vancouver, Canada)에 넣었다. 이후, 결합되지 않은 세포 분획을 깨끗한 튜브로 옮겨 회수하고, 회수된 세포를 3,560 xg에서 5분간 원심분리하고, 400 ㎕의 RPMI1640 배지에 재현탁하고, 8개의 분획으로 나누고, 최종적으로 0.1 ㎍/ml의 폴리-L-리신으로 코팅된 유리 슬라이드에 접종하였다. 유리 슬라이드에 대한 세포의 자발적인 결합을 유도하기 위해, 세포를 실온에서 2-4시간 동안 반응시켰다. 결합되지 않은 세포는 고정 전에 PBS(pH 7.4)로 세척하였다. 결합된 세포를 3.7% 파라포름알데히드(PFA)에 고정하고 공초점 현미경으로 분석하기 전에 냉장고에 보관했다.To efficiently detect circulating cancer cells in HCC patients, we chose a strategy to remove red blood cells, then further remove CD45-positive cells by a CD45 depletion protocol, and analyze the remaining cells (Fig. 15c). Specifically, peripheral blood mononuclear cells (PBMCs) were isolated from the peripheral blood of healthy donors, hepatitis patients, or HCC patients by Ficoll-Paque Plus (GE Healthcare, Seoul, Korea) gradient centrifugation within 6 h after collecting the blood of HCC patients. PBMCs were washed with PBS containing 0.5% bovine serum albumin (BSA) and 2 mM EDTA and resuspended in this buffer at a concentration of 1 × 10 8 cells/mL. Enrichment of circulating cancer cells by depletion of CD45-positive cells from these leukocyte lineages was performed using a Human CD45 depletion kit (EasySep®, Stem Cells Technologies, Vancouver, BC, Canada) according to the provided protocol. First, anti-CD45 antibody was added to the cell suspension and reacted at room temperature for 15 minutes, then dextran-coated magnetic nanoparticles were reacted with the cells at room temperature for 10 minutes, and the cell suspension was placed in an EasySep® big easy magnet (StemCell Technologies, Vancouver, Canada) at room temperature for 10 minutes. The unbound cell fraction was then transferred to a clean tube and collected, and the collected cells were centrifuged at 3,560 × g for 5 minutes, resuspended in 400 ㎕ of RPMI1640 medium, divided into eight fractions, and finally seeded onto glass slides coated with 0.1 ㎍/mL of poly-L-lysine. The cells were reacted at room temperature for 2-4 hours to induce spontaneous binding of the cells to the glass slides. Unbound cells were washed with PBS (pH 7.4) before fixation. The bound cells were fixed in 3.7% paraformaldehyde (PFA) and stored in the refrigerator before analysis by confocal microscopy.
CD45 양성 백혈구 제거 방법의 민감도와 특이성을 테스트하기 위해, 증가하는 수의 Huh7 세포를 PBMC에 첨가하고, 상기의 CD45 제거 프로토콜을 적용하여 CD45 양성인 세포를 제거하고 남은 63-D7 양성인 Huh7 세포의 수를 10% 정상 말 혈청으로 블로킹한 다음 측정하였다. 이때 Dylight488-conjugation Kit(Vector laboratories, Burlingame, CA, USA)를 사용하여 63-D7항체를 표지한 Dylight488-접합된 63-D7을 면역세포화학적 방법으로 Huh7를 염색하여 측정하였다. 탐지된 Huh7 세포 수 대 스파이킹 된 Huh7 세포 수의 선형 회귀는 0.9801의 R2를 산출하였다. Huh7 세포의 평균 회수율은 약 72%로, 이러한 방법을 통해 70% 이상의 세포를 검출할 수 있다는 것을 보여준다(도 15b).To test the sensitivity and specificity of the CD45-positive leukocyte depletion method, increasing numbers of Huh7 cells were added to PBMCs, and the CD45 depletion protocol described above was applied to remove CD45-positive cells. The number of remaining 63-D7-positive Huh7 cells was measured after blocking with 10% normal horse serum. At this time, Dylight488-conjugated 63-D7 labeled with 63-D7 antibody was stained for Huh7 by immunocytochemical method using Dylight488-conjugation Kit (Vector laboratories, Burlingame, CA, USA). Linear regression of the number of Huh7 cells detected versus the number of spiked Huh7 cells yielded an R2 of 0.9801. The average recovery of Huh7 cells was approximately 72%, showing that more than 70% of cells can be detected by this method (Fig. 15b).
CD45가 잘 제거되었는지 확인하기 위한 63-D7 및 CD45의 이중 염색을 위해서, CD45-고갈된 세포를 3.7% PFA로 고정하고, 10% 정상 말 혈청으로 블로킹한 다음 Dylight 649-접합된 항-마우스 IgG(Vector laboratories) 및 Dylight 488-접합된 63-D7로 반응시켰다. PBS로 세척한 후, DAPI(4,6-diamidino- 2-phenylindole)를 이용하여 핵을 염색하였다. 형광 신호는 Leica TCS SP5 공초점 현미경(Leica Microsystems, 서울, 한국)으로 검출하였다. 그 결과 63-D7 양성인 세포는 모두 CD45 음성임을 확인하였다(도 15c의 아래 패널, 표 3). 이중 면역 형광 염색에 의한 HCC 환자의 63-D7+ 유핵 CTC에서 EMT 마커의 발현 프로필은 하기 표 3에 구체적으로 나타나 있다.For double staining of 63-D7 and CD45 to confirm whether CD45 was well removed, CD45-depleted cells were fixed with 3.7% PFA, blocked with 10% normal horse serum, and then reacted with Dylight 649-conjugated anti-mouse IgG (Vector laboratories) and Dylight 488-conjugated 63-D7. After washing with PBS, nuclei were stained with DAPI (4,6-diamidino-2-phenylindole). Fluorescent signals were detected with a Leica TCS SP5 confocal microscope (Leica Microsystems, Seoul, Korea). As a result, all 63-D7-positive cells were confirmed to be CD45-negative (lower panel of Fig. 15c, Table 3). The expression profiles of EMT markers in 63-D7+ nucleated CTCs of HCC patients by double immunofluorescence staining are specifically shown in Table 3 below.
다음으로, 건강한 지원자 21명과 HCC 환자 95명(1차 HCC 환자 53명 및 2차 HCC 환자 42명)의 혈액 샘플을 상기 기술한 방법과 동일하게 CD45 양성 세포를 제거하고 남은 세포를 슬라이드에 붙여서 공초점 현미경으로 분석하였다. 간암환자(HCC, HCC(meta)), 정상인(Normal) 및 간염환자(의 혈액에서 63-D7 양성인 모든 혈중 암세포의 수를 하기 표 4에 나타냈다.Next, blood samples from 21 healthy volunteers and 95 HCC patients (53 primary HCC patients and 42 secondary HCC patients) were collected by the same method as described above to remove CD45-positive cells, and the remaining cells were attached to slides and analyzed by confocal microscopy. The numbers of all 63-D7-positive circulating cancer cells in the blood of liver cancer patients (HCC, HCC (meta)), normal people (Normal), and hepatitis patients are shown in Table 4 below.
정상 성인 21명 중 5명에서 몇 개의 63-D7 양성 세포가 검출되었지만 측정된 세포 수는 매우 드물었다(≤ 0.39 cells/ml)(표 4). HCC 환자의 혈액 세포를 63-D7 및 기타 항체로 염색한 경우, 63-D7 음성이면서 DAPI 양성 세포는 13.7%로 63-D7으로 검출되지 않는 혈중 암세포도 약 14% 존재하였다 (도 15c 위 패널, 표 3). HCC 환자로부터 분리된 세포를 63-D7 및 CD45 항체로 염색했을 때, 모든 63-D7 양성 세포는 CD45 음성이었으며(도 15c 아래 패널, 표 3), 이는 63-D7이 CD45 음성이면서 DAPI 양성 혈중 암세포의 약 86%를 검출해낼 수 있음을 제시한다. 그리고 63-D7 양성 혈중 암세포의 크기는 18 μm에서 40 μm 사이로 이질적이었고, 그들은 과염색성 핵과 높은 핵 대 세포질 비율을 가졌다(도 16a 내지 도 16c). 63-D7-양성 혈중 암세포는 모든 HCC 환자에서 검출되었고, 95명의 환자 중 92명(약 97%)이 정상 성인보다 높은 세포 수를 보였다(도 17, 표 4)(p<0.0001). HCC 환자로부터 분리된 세포 수는 ml당 0.1~ 50.72개 범위였으며 평균은 ml당 6.13개였다(표 4). 63-D7 양성 혈중 암세포는 만성 간염(Chronic hepatitis) 및 간경변증(Cirrhosis) 환자에서는 발견되지 않으므로(표 4), 간염 등으로 인한 염증성 세포가 아니라 암 발생으로 인한 혈중 암세포임을 보여준다.Although a few 63-D7-positive cells were detected in 5 of 21 normal adults, the measured cell numbers were very rare (≤ 0.39 cells/ml) (Table 4). When blood cells from HCC patients were stained with 63-D7 and other antibodies, 13.7% of the cells were 63-D7-negative and DAPI-positive, indicating that about 14% of the circulating cancer cells were not detected by 63-D7 (Fig. 15c upper panel, Table 3). When cells isolated from HCC patients were stained with 63-D7 and CD45 antibodies, all of the 63-D7-positive cells were CD45-negative (Fig. 15c lower panel, Table 3), suggesting that 63-D7 can detect about 86% of CD45-negative and DAPI-positive circulating cancer cells. And the size of 63-D7-positive circulating cancer cells was heterogeneous, ranging from 18 μm to 40 μm, and they had hyperchromatic nuclei and a high nuclear-to-cytoplasmic ratio (Figs. 16a to 16c). 63-D7-positive circulating cancer cells were detected in all HCC patients, and 92 of 95 patients (approximately 97%) showed a higher cell number than that of normal adults (Fig. 17, Table 4) (p<0.0001). The number of cells isolated from HCC patients ranged from 0.1 to 50.72 per ml, and the average was 6.13 per ml (Table 4). Since 63-D7-positive circulating cancer cells were not found in patients with chronic hepatitis and cirrhosis (Table 4), it shows that they are circulating cancer cells caused by cancer development, not inflammatory cells caused by hepatitis, etc.
63-D7과 임상 병리학적 변수들 사이의 연관성을 하기 표 5에 나타냈다.The associations between 63-D7 and clinical pathological variables are shown in Table 5.
(n=53)(n=53)
<2.45<2.45
(n=26)(n=26)
(n=27)(n=27)
53명의 1차 원발성 간암 환자에 대한 임상병리학적 분석에서 63-D7 양성 혈중 암세포의 수와 진행성 종양 병기 그룹 사이에서 미세혈관 침입, 병리학적 단계 및 임상적 단계에서 통계적으로 유의한 상관관계가 있고, 63-D7 양성 암세포 경우 미세혈관침입이 일어나고 병리학적 및 임상학적 단계에서 더 나쁜 단계로 진입한 환자라는 것을 말해준다(표 5).In a clinicopathological analysis of 53 patients with primary hepatocellular carcinoma, there was a statistically significant correlation between the number of 63-D7-positive circulating cancer cells and the advanced tumor stage group, with microvascular invasion, pathological stage, and clinical stage, indicating that patients with 63-D7-positive cancer cells had microvascular invasion and advanced to worse pathological and clinical stages (Table 5).
이는 63-D7 양성 혈중 암세포 수가 클수록 병리학적 및 임상학적으로 나쁜 상태라는 것을 의미한다. 환자 집단을 1차 원발성 간세포암종과 2차 전이성 및 재발성 간세포암종으로 나누었을 때, 2차 전이성 및 재발성 간세포암종 환자군에서 1차 원발성 간세포암종에 비해 63-D7 양성 혈중 암세포의 세포수가 더 증가하였다(도 17, 표 4)(p<0.0001). 따라서 63-D7 양성 혈중 암세포의 수(정상 vs 1차 HCC = ≤0.39 vs 2.54 cells/ml)는 1차 HCC 환자를 진단할 수 있는 우수한 진단 마커이며(표 4), 전이되거나 재발되면 더욱 그 수(평균 10.7 cells/ml)(표 4)가 증가하는 질병 진행과 밀접한 관련이 있는 우수한 진단 마커임을 보여준다.This means that the higher the number of 63-D7-positive circulating cancer cells, the worse the pathological and clinical condition. When the patient group was divided into primary HCC and secondary metastatic and recurrent HCC, the number of 63-D7-positive circulating cancer cells was more increased in the secondary metastatic and recurrent HCC patient group than in the primary HCC (Fig. 17, Table 4) (p<0.0001). Therefore, the number of 63-D7-positive circulating cancer cells (normal vs. primary HCC = ≤0.39 vs. 2.54 cells/ml) is an excellent diagnostic marker for diagnosing primary HCC patients (Table 4), and it is an excellent diagnostic marker closely related to disease progression, with the number further increasing in metastasis or recurrence (average 10.7 cells/ml) (Table 4).
10-2. 이중염색에 의한 63-D7 양성 혈중 암세포의 특성 분석10-2. Characteristics of 63-D7 positive blood cancer cells by double staining
63-D7 양성 혈중 암세포의 특성을 추가적으로 분석하기 위해, 세포를 63-D7 및 다양한 항체로 동시에 염색했다.To further characterize the 63-D7-positive blood cancer cells, cells were simultaneously stained with 63-D7 and various antibodies.
혈중 암세포를 63-D7 및 다양한 표면 마커들을 이용해 이중염색을 하기 위해서는, CD45 고갈 세포를 3.7% PFA로 고정하고, 10% 정상 말 혈청으로 블로킹한 다음 항-MVP(Aviva systems, San Diego, CA, USA), 항-CD44 (BD bioscience, Seoul, Korea), 항-CD90(BD bioscience), Alexa 555-접합된 항-EpCAM(BD Biosciences), Alexa 555-접합된 항-E-cadherin(Cell Signaling Technology, Beverly, MA, USA), 및 항-HSA(Novus, Litteleton, 콜로라도, 미국)를 각각 세포와 반응시키고, Dylight 650-접합된 항-토끼 IgG(Thermo Fischer Scientific)로 추가 반응시키고, 최종적으로 Dylight 488-접합된 63-D7으로 반응시켜 상기 실시예 10-1과 같이 염색하였다. 또한 Dylight 488-접합된 63-D7과 세포 내부 마커의 이중 염색을 위해서는, CD45-고갈된 세포를 고정하고 블로킹하기 전에 0.1% Triton X-100으로 투과시키고 항-vimentin(Santa Cruz Biotechnology), 항-Twist1(AbCAM, 케임브릿지, 영국), 항-ZEB1(AbCAM), 또는 Phycoerythrin 접합된 항-panCK 항체(BD biosciences, San Jose, CA, USA)를 반응시켰다. 이후 세포를 1차 항체의 접합 상태에 따라 Dylight 650-접합된 항-토끼 IgG(Thermo Fischer Scientific)와 함께 배양하고 최종적으로 Dylight 488-접합된 63-D7와 함께 반응시켜 형광 신호는 Leica TCS SP5 공초점 현미경 (Leica Microsystems)으로 검출하여 분석하였다.To double stain circulating cancer cells with 63-D7 and various surface markers, CD45-depleted cells were fixed with 3.7% PFA, blocked with 10% normal horse serum, and then reacted with anti-MVP (Aviva systems, San Diego, CA, USA), anti-CD44 (BD bioscience, Seoul, Korea), anti-CD90 (BD bioscience), Alexa 555-conjugated anti-EpCAM (BD Biosciences), Alexa 555-conjugated anti-E-cadherin (Cell Signaling Technology, Beverly, MA, USA), and anti-HSA (Novus, Litteleton, CO, USA), followed by additional reaction with Dylight 650-conjugated anti-rabbit IgG (Thermo Fischer Scientific), and finally Dylight 488-conjugated 63-D7, and stained as in Example 10-1. Also, for double staining of Dylight 488-conjugated 63-D7 and intracellular markers, CD45-depleted cells were fixed and permeabilized with 0.1% Triton X-100 before blocking and reacted with anti-vimentin (Santa Cruz Biotechnology), anti-Twist1 (AbCAM, Cambridge, UK), anti-ZEB1 (AbCAM), or phycoerythrin-conjugated anti-panCK antibodies (BD biosciences, San Jose, CA, USA). Then, the cells were incubated with Dylight 650-conjugated anti-rabbit IgG (Thermo Fischer Scientific) depending on the conjugation status of the primary antibody and finally reacted with Dylight 488-conjugated 63-D7, and the fluorescence signals were detected and analyzed by a Leica TCS SP5 confocal microscope (Leica Microsystems).
우리는 이전 연구에서 세포 표면 MVP(major vault protein)가 HCC 환자의 혈중 암세포에서 약 90%에서 검출되고, 전이성 암세포에서 더욱 증가하는 현상을 발견하였다(Lee, et al., Sci Rep. vol. 7, 13201, 2017). 이에, 먼저 혈중 암세포를 63-D7과 MVP 항체로 동시에 염색해본 결과 63-D7 양성 혈중 암세포의 약 28%(100/363)가 1차 원발성 HCC 환자에서 MVP 양성이었고, 63-D7양성 혈중 암세포의 약 24%(35/147)가 2차 HCC 환자에서 양성이었다 (도 16a, 표 3). 63-D7/HSA 염색에서는 63-D7 양성 혈중 암세포의 약 25%(50/203)가 1차 및 2차 HCC 환자 모두에서 HSA 양성이었다 (도 16a, 표 3). 63-D7/E-cadherin 염색에서 모든 혈중 암세포는 1차 HCC 환자에서 모두에서 E-cadherin 음성(0/18)이였다(도 16b, 표 3). 63-D7/panCK(pan-cytokeratin) 염색에서 모든 혈중 암세포는 1차(0/104) 및 2차 HCC 환자(0/24) 모두에서 panCK 음성이었다(도 16a, 표 3). 63-D7/EpCAM 염색에서 63-D7 양성 세포의 약 15%(67/442)가 1차 및 2차 HCC 환자 모두에서 EpCAM 양성이었다(도 16b, 표 3). 이와 같이, 상피성 마커로 잘 알려진 PanCK이나 E-cadherin은 63-D7이 인식하는 PTGFRN 양성 혈중 암세포에서는 1차 원발암이든 2차 전이성/재발성 암이든 발현하지 않음을 알 수 있어 63-D7 양성 혈중 암세포는 최소한 상피성 혈중 암세포가 아님을 알 수 있다.In our previous study, we found that cell surface major vault protein (MVP) was detected in approximately 90% of circulating cancer cells from HCC patients and further increased in metastatic cancer cells (Lee, et al., Sci Rep. vol. 7, 13201, 2017). Therefore, when circulating cancer cells were simultaneously stained with 63-D7 and MVP antibodies, approximately 28% (100/363) of 63-D7-positive circulating cancer cells were MVP-positive in primary HCC patients, and approximately 24% (35/147) of 63-D7-positive circulating cancer cells were MVP-positive in secondary HCC patients (Fig. 16a, Table 3). In 63-D7/HSA staining, approximately 25% (50/203) of the 63-D7-positive circulating cancer cells were HSA-positive in both primary and secondary HCC patients (Fig. 16a, Table 3). In 63-D7/E-cadherin staining, all circulating cancer cells were E-cadherin-negative (0/18) in all primary HCC patients (Fig. 16b, Table 3). In 63-D7/panCK (pan-cytokeratin) staining, all circulating cancer cells were panCK-negative in both primary (0/104) and secondary HCC patients (0/24) (Fig. 16a, Table 3). In 63-D7/EpCAM staining, approximately 15% (67/442) of 63-D7-positive cells were EpCAM-positive in both primary and secondary HCC patients (Fig. 16b, Table 3). Thus, epithelial markers such as PanCK or E-cadherin are not expressed in PTGFRN-positive circulating cancer cells recognized by 63-D7, regardless of whether they are primary or secondary metastatic/recurrent cancers. This suggests that 63-D7-positive circulating cancer cells are at least not epithelial circulating cancer cells.
따라서, 63-D7 양성 혈중 암세포가 중간엽 표현형을 나타내는지를 알아보기 위해, 원발성 간세포암종 환자에서 대표적인 중간엽 마커 Vimentin, Twist 및 ZEB1를 1차 HCC 환자에서 관찰하였다. 63-D7 양성 혈중 암세포의 약 24%(43/180)는 vimentin 양성이였다(도 16c, 표 3). 그리고 63-D7 양성 혈중 암세포에서 Twist(3.2%, 6/189) 및 ZEB1(5%, 3/60) 양성도 작은 분획이었지만 관찰되었다(도 16c, 표 3). 따라서 63-D7 양성 혈중 암세포는 상피성 표현형과 중간엽 표현형 비중을 비교할 때는 중간엽 표현형이 현저히 우세하지만, 대부분의 경우 상피성과 중간엽성 구분이 불분명한 부분적 중간단계 세포 집단임을 알 수 있다.Therefore, to determine whether 63-D7-positive circulating cancer cells exhibit a mesenchymal phenotype, representative mesenchymal markers Vimentin, Twist, and ZEB1 were observed in primary HCC patients. Approximately 24% (43/180) of 63-D7-positive circulating cancer cells were vimentin-positive (Fig. 16c, Table 3). In addition, Twist (3.2%, 6/189) and ZEB1 (5%, 3/60) positivity were also observed in small fractions in 63-D7-positive circulating cancer cells (Fig. 16c, Table 3). Therefore, although 63-D7-positive circulating cancer cells exhibit a significantly predominant mesenchymal phenotype when comparing the proportions of epithelial and mesenchymal phenotypes, it can be seen that in most cases, they are a partial intermediate-stage cell population in which the distinction between epithelial and mesenchymal phenotypes is unclear.
10-3. 삼중염색에 의한 63-D7 양성 혈중 암세포의 특성 분석10-3. Characteristics of 63-D7 positive blood cancer cells by triple staining
1차 및 2차 HCC 환자의 63-D7 양성 혈중 암세포에서 상피성 표현형 마커와 중간엽 표현형 마커의 발현 양상을 좀 더 자세히 분석하기 위해, CD45가 고갈된 혈중 암세포를 사용하여 상피성 대표 마커인 EpCAM과 중간엽성 대표마커인 Vimentin을 사용하여 63-D7/EpCAM/vimentin에 대한 삼중 형광 염색을 수행했다(도 18a, 표 6). 삼중 형광 염색의 결과는 하기의 표 6에 구체적으로 나타냈다.To further analyze the expression patterns of epithelial and mesenchymal phenotype markers in 63-D7-positive circulating cancer cells from primary and secondary HCC patients, triple fluorescence staining for 63-D7/EpCAM/vimentin was performed using CD45-depleted circulating cancer cells, using the epithelial representative marker EpCAM and the mesenchymal representative marker Vimentin (Fig. 18a, Table 6). The results of the triple fluorescence staining are specifically shown in Table 6 below.
63-D7, Vimentin 및 EpCAM의 삼중 염색을 위해서는 CD45 고갈된 세포를 고정하고 블로킹하기 전에 0.1% Triton X-100으로 투과시키고 항-Vimentin 항체(Santa Cruz Biotechnology)로 먼저 반응시켰다. 그런 다음 세포를 Dylight 650-접합된 항 토끼 IgG(Thermo Scientific), Alexa 555-접합된 항-EpCAM(Cell Signaling Technology) 및 Dylight 488-접합된 63-D7과 함께 반응시키고 각 단계 사이에 Ca2+ 및 Mg2+를 포함하는 PBS로 세포를 4회 세척했다. 핵은 DAPI로 염색하고 형광 신호는 공초점 현미경 (Leica Microsystems)으로 검출하였다.For triple staining of 63-D7, Vimentin, and EpCAM, CD45-depleted cells were fixed and permeabilized with 0.1% Triton X-100 before blocking and reacted first with anti-Vimentin antibody (Santa Cruz Biotechnology). Cells were then reacted with Dylight 650-conjugated anti-rabbit IgG (Thermo Scientific), Alexa 555-conjugated anti-EpCAM (Cell Signaling Technology), and Dylight 488-conjugated 63-D7, washing cells four times with PBS containing Ca 2+ and Mg 2+ between each step. Nuclei were stained with DAPI, and fluorescence signals were detected by confocal microscopy (Leica Microsystems).
그 결과 63-D7+EpCAM+Vimentin+인 삼중 양성 혈중 암세포는 1차 HCC 환자에서 9.1%(47/518)인 반면, 2차 HCC 환자에서는 0.3%(3/933)였다. 상피성 세포를 대표하는 63-D7+EpCAM+Vimentin- 혈중 암세포는 1차 HCC 환자에서 0.6%(3/518)인 반면 2차 HCC 환자에서는 없었다(0/933). 중간엽성 세포를 대표하는 63-D7+EpCAM-Vimentin+ 혈중 암세포는 1차 HCC 환자에서 44.2%(229/518)인 반면 2차 HCC 환자에서는 27.3%(255/933)였다(도 18a, 표 6).As a result, the 63-D7 + EpCAM + Vimentin + triple-positive circulating cancer cells were 9.1% (47/518) in primary HCC patients, whereas 0.3% (3/933) in secondary HCC patients. The 63-D7 + EpCAM + Vimentin - circulating cancer cells, representing epithelial cells, were 0.6% (3/518) in primary HCC patients, whereas there were none (0/933) in secondary HCC patients. The 63-D7 + EpCAM - Vimentin + circulating cancer cells, representing mesenchymal cells, were 44.2% (229/518) in primary HCC patients, whereas 27.3% (255/933) in secondary HCC patients (Fig. 18a, Table 6).
따라서, 1차 원발성 간암에서 63-D7 양성 혈중 암세포의 0.6%는 상피성 표현형인 반면, 53.3%(9.1%+44.2%)는 반중간엽(EpCAM 및 vimentin 둘 다 양성) 및 중간엽성(vimentin 양성) 표현형이었다. 흥미롭게도, EpCAM도 Vimentin도 없는 부분적 중간형 표현형(partial or intermediate)인 63-D7 단일 양성 혈중 암세포는 1차 HCC 환자에서 46.1%이고 2차 HCC 환자에서는 무려 72.3% (675/933)이었다. 또한, 이러한 중간형 표현형은 모든 조사한 2차 HCC 환자(30/30 환자)에서 관찰되어, 63-D7만 발현하는 중간형 표현형은 전이 재발하는 간암환자의 핵심적인 혈중 암세포 마커임을 알 수 있다(표 6).Thus, 0.6% of 63-D7-positive circulating cancer cells in primary HCC were of the epithelial phenotype, whereas 53.3% (9.1% + 44.2%) were of the semimesenchymal (both EpCAM and vimentin positive) and mesenchymal (vimentin positive) phenotypes. Interestingly, 63-D7 single-positive circulating cancer cells with a partial or intermediate phenotype without EpCAM or Vimentin were 46.1% in primary HCC patients and a whopping 72.3% (675/933) in secondary HCC patients. In addition, this intermediate phenotype was observed in all investigated secondary HCC patients (30/30 patients), suggesting that the intermediate phenotype expressing only 63-D7 is a key circulating cancer cell marker in patients with recurrent metastatic HCC (Table 6).
이전 연구에서 MVP/EpCAM/Vimentin 삼중염색에서 MVP-양성 혈중 암세포의 51.3%가 MVP-단일 양성이었기 때문에 (Lee, Joh et al. 2017), 우리는 MVP/EpCAM/Vimentin 삼중염색을 동일한 방법으로 수행하였다. MVP 단일 양성 혈중 암세포을 1차 원발성 간암과 2차 간암으로 분류하였을 때 MVP 단일 양성 혈중 암세포는 원발성 간세포암 환자에서 53.5%, 이차성 간세포암종 환자에서 50.3%였다(표 6). 그리고 MVP 단일 양성 혈중 암세포는 모든 이차 HCC 환자(9/9 환자, 100%)에서 발견되었다(표 6).In a previous study, 51.3% of MVP-positive circulating cancer cells were MVP-single positive in MVP/EpCAM/Vimentin triple staining (Lee, Joh et al. 2017), so we performed MVP/EpCAM/Vimentin triple staining using the same method. When MVP single-positive circulating cancer cells were classified into primary HCC and secondary HCC, MVP single-positive circulating cancer cells were 53.5% in primary HCC patients and 50.3% in secondary HCC patients (Table 6). In addition, MVP single-positive circulating cancer cells were found in all secondary HCC patients (9/9 patients, 100%) (Table 6).
따라서, MVP(50.3%) 또는 63-D7(72.3%) 단일 양성 혈중 암세포는 EpCAM-음성 및 Vimentin-음성이며 모든 2차 HCC 환자에서 우세하게 나타났다. 따라서 63-D7이 인식하는 PTGFRN은 간암환자에서 상피성도 중간엽성도 뚜렷하게 보이지 않는 대부분의 혈중 암세포를 대변하는 새로운 마커이며, 특히 2차 전이성/재발성 간암환자에서는 모든 환자에서 관찰되는 우수한 진단 마커임을 알 수 있다.Therefore, MVP (50.3%) or 63-D7 (72.3%) single-positive circulating cancer cells were EpCAM-negative and Vimentin-negative and were predominantly present in all secondary HCC patients. Therefore, PTGFRN recognized by 63-D7 is a novel marker representing most circulating cancer cells in patients with HCC that are neither epithelial nor mesenchymal, and it can be seen as an excellent diagnostic marker observed in all patients, especially in patients with secondary metastatic/recurrent HCC.
10-4. 63-D7 양성 혈중 암세포에서 면역 관문 분자의 분석10-4. Analysis of immune checkpoint molecules in 63-D7 positive blood cancer cells
수많은 연구에서 HCC를 포함한 다양한 암에서 B7-H3 발현과 불량한 임상결과 사이에 강한 상관관계가 있음이 확립되어 있다. 우리는 PTGFRN이 HCC 세포에서 B7-H3와 상호작용한다는 것을 실험을 통해 확인했으므로(도 11c, 도 11d), 혈중 암세포에서 PTGFRN, B7-H3 및 기타 면역 조절 분자의 발현을 삼중염색으로 분석했다.Numerous studies have established a strong correlation between B7-H3 expression and poor clinical outcomes in various cancers, including HCC. Since we experimentally confirmed that PTGFRN interacts with B7-H3 in HCC cells (Fig. 11c, Fig. 11d), we analyzed the expression of PTGFRN, B7-H3, and other immune-regulatory molecules in circulating cancer cells by triple staining.
63-D7, B7-H3 및 면역 조절 분자의 삼중 염색을 위해 세포를 폴리-L-리신 코팅된 유리에 부착한 후 CD45가 고갈된 세포를 고정하고 블로킹하고 항-폴리 토끼 MVP항체(Aviva bioscience), ULBP1(Abcam), MICA/B(Bioss) 및 TGFβR1(Atlas)로 반응시켰다. 다음으로 Alexa 568 접합된 항-토끼 IgG(Thermo Fischer Scientific) 또는 Alexa 594-접합된 마우스 단일클론 PD-L1(Biolegend), PD-L2(Biolegend) 또는 피코에리트린-접합된 CD47(Biolegend)과 함께 반응시켰다. 그 세포를 다시 Dylight 650-접합된 항-토끼 B7-H3(Sinobiological, Beijing, China)와 Dylight 488-접합된 63-D7항체와 함께 반응시켰다. 각 단계 사이에 Ca2+ 및 Mg2+를 포함하는 PBS로 세포를 4회 세척했다. 핵은 4,6-diamidino-2-phenylindole(DAPI)로 염색하고 형광 신호는 Leica TCS SP5 공초점 현미경으로 검출하였다. 삼중 면역 형광 염색에 의한 2차 HCC 환자의 유핵 CTC에서 EMT 및 면역 체크포인트 마커의 발현 프로필을 하기의 표 7에 나타냈다.For triple staining of 63-D7, B7-H3, and immune-regulatory molecules, cells were adhered to poly-L-lysine-coated glasses, then CD45-depleted cells were fixed, blocked, and reacted with anti-poly rabbit MVP antibody (Aviva bioscience), ULBP1 (Abcam), MICA/B (Bioss), and TGFβR1 (Atlas). They were then reacted with Alexa 568-conjugated anti-rabbit IgG (Thermo Fischer Scientific) or Alexa 594-conjugated mouse monoclonal PD-L1 (Biolegend), PD-L2 (Biolegend), or phycoerythrin-conjugated CD47 (Biolegend). The cells were then reacted with Dylight 650-conjugated anti-rabbit B7-H3 (Sinobiological, Beijing, China) and Dylight 488-conjugated 63-D7 antibody. Between each step, cells were washed four times with PBS containing Ca 2+ and Mg 2+ . Nuclei were stained with 4,6-diamidino-2-phenylindole (DAPI) and fluorescence signals were detected with a Leica TCS SP5 confocal microscope. The expression profiles of EMT and immune checkpoint markers in nucleated CTCs from secondary HCC patients by triple immunofluorescence staining are shown in Table 7 below.
(% marker/CTCs)(% marker/CTCs)
or B7-H3+/total PTGFRN+
MVP + /total PTGFRN +
or B7-H3 + /total PTGFRN +
or B7-H3+/total PTGFRN+
TGFβR1 + /total PTGFRN +
or B7-H3 + /total PTGFRN +
or B7-H3+/total PTGFRN+
ULBP1 + /total PTGFRN +
or B7-H3 + /total PTGFRN +
or B7-H3+/total PTGFRN+
MICA/B + /total PTGFRN +
or B7-H3 + /total PTGFRN +
or B7-H3+/total PTGFRN+
PD-L1 + /total PTGFRN +
or B7-H3 + /total PTGFRN +
or B7-H3+/total PTGFRN+
PD-L2 + /total PTGFRN +
or B7-H3 + /total PTGFRN +
or B7-H3+/total PTGFRN+
CD47 + /total PTGFRN +
or B7-H3 + /total PTGFRN +
이전 연구에서 우리는 MVP가 스트레스가 많은 환경에서 HCC 세포의 생존, 증식, 이동 및 침입을 담당한다고 보고했다. 또한 B7-H3는 MVP와 상호작용하며 유방암 세포에서 암 줄기세포 농축을 촉진한다고 알려졌다. 따라서, 2차 간암 환자에서 간암 전이 및 재발 시의 PTGFRN 양성 혈중 암세포에 대해 얼마나 많은 혈중 암세포가 MVP 양성인지 알아보기 위해 혈중 암세포를 63-D7, 항-B7-H3 및 항-MVP 항체로 동시에 염색하였다(도 18b, 표 7). 총 PTGFRN 양성 혈중 암세포의 약 32%(111/348)가 MVP 양성이었고, 총 PTGFRN 양성 혈중 암세포의 약 62%(217/348)가 B7-H3 양성이었다. 따라서, PTGFRN 양성 혈중 암세포의 일부는 2차 HCC 환자의 말초 혈액에서 세포 생존, 증식, 이동 및 침입에서 B7-H3 및 MVP의 역할과 연관될 가능성이 높다.In our previous study, we reported that MVP is responsible for the survival, proliferation, migration, and invasion of HCC cells in a stressful environment. In addition, B7-H3 has been known to interact with MVP and promote cancer stem cell enrichment in breast cancer cells. Therefore, to determine how many PTGFRN-positive circulating cancer cells were MVP-positive in patients with secondary HCC, we simultaneously stained circulating cancer cells with 63-D7, anti-B7-H3, and anti-MVP antibodies (Fig. 18b, Table 7). About 32% (111/348) of the total PTGFRN-positive circulating cancer cells were MVP-positive, and about 62% (217/348) of the total PTGFRN-positive circulating cancer cells were B7-H3-positive. Therefore, a subset of PTGFRN-positive circulating cancer cells may likely be associated with the roles of B7-H3 and MVP in cell survival, proliferation, migration, and invasion in the peripheral blood of secondary HCC patients.
최근 연구에 따르면 EMT는 암세포에 전이성, 면역억제 가능성 및 암 줄기세포성를 제공한다. 또한 EMT는 암줄기세포 및 혈중 암세포에서 PD-L1 및 CD47과 같은 면역억제 분자의 발현을 유도할 수 있다. 63-D7 양성 혈중 암세포의 약 53%가 1차 원발성 간암 환자에서 EMT 표현형 혈중 암세포였지만, 2차 간암 환자에서는 EMT 표현형 혈중 암세포의 비율이 27.6%로 감소했다(표 6). 대신, 대부분의 63-D7 양성 혈중 암세포(72.3%)는 2차 간암 환자에서 부분/중간 표현형의 혈중 암세포였다(표 6). 최근 연구는 부분/중간 표현형 암 세포가 면역세포 세포독성을 피하고 면역 세포를 면역 억제 표현형으로 분극화 함으로써 종양면역 탈출을 매개한다고 제안한다. 따라서 부분/중간 표현형을 가진 63-D7 양성 혈중 암세포는 2차 간암 환자에서 중요한 역할을 할 것으로 예상된다.Recent studies have shown that EMT provides cancer cells with metastatic potential, immunosuppressive potential, and cancer stemness. In addition, EMT can induce the expression of immunosuppressive molecules such as PD-L1 and CD47 in cancer stem cells and circulating cancer cells. Approximately 53% of 63-D7-positive circulating cancer cells were EMT phenotype circulating cancer cells in primary HCC patients, but the proportion of EMT phenotype circulating cancer cells decreased to 27.6% in secondary HCC patients (Table 6). Instead, most 63-D7-positive circulating cancer cells (72.3%) were partial/intermediate phenotype circulating cancer cells in secondary HCC patients (Table 6). Recent studies suggest that partial/intermediate phenotype cancer cells mediate tumor immune escape by avoiding immune cell cytotoxicity and polarizing immune cells toward an immunosuppressive phenotype. Therefore, 63-D7-positive circulating cancer cells with partial/intermediate phenotype are expected to play an important role in patients with secondary liver cancer.
TGFβ/TGFβR1 신호는 암줄기세포에서 다중 면역억제 효과를 나타낸다. TGFβ는 또한 NK 세포 기능의 억제에 필요하다. 또한, TGFβ/TGFβR1 신호는 유방 암줄기세포의 높은 전이 개시 능력을 제공한다. TGFβ1 신호전달은 또한 B7-H3 발현 증가를 통해 결장직장암에서 T 세포 매개 종양 회피를 촉진한다. 따라서 2차 간암 환자의 PTGFRN 양성 혈중 암세포에서 EMT 표현형 혈중 암세포의 수가 감소하였지만, 63-D7 양성 혈중 암세포에서 TGFβR1의 발현이 어느 정도 양성인지를 조사하였다. 혈중 암세포가 항-TGFβR1, 63-D7 및 항-B7-H3 항체로 염색되었을 때, 모든 2차 HCC 환자가 TGFβR1 양성 혈중 암세포를 보유했다(도 19, 표 7). 세부 분석 결과 전체 PTGFRN 양성 혈중 암세포의 51.8%(249/481)가 TGFβR1 양성이었고, 전체 PTGFRN 양성 혈중 암세포의 58%(279/481)가 B7-H3 양성인 것으로 밝혀져, PTGFRN 양성 혈중 암세포는 TGFβR1- 및 B7-H3-매개 면역 억제 표현형을 가질 것으로 보인다.TGFβ/TGFβR1 signaling exerts multiple immunosuppressive effects in CSCs. TGFβ is also required for the suppression of NK cell function. In addition, TGFβ/TGFβR1 signaling provides the high metastasis-initiating ability of breast CSCs. TGFβ1 signaling also promotes T cell-mediated tumor evasion in colorectal cancer through increased B7-H3 expression. Therefore, although the number of EMT phenotype CSCs was reduced in PTGFRN-positive CSCs from secondary HCC patients, we investigated to what extent TGFβR1 expression was positive in 63-D7-positive CSCs. When CSCs were stained with anti-TGFβR1, 63-D7, and anti-B7-H3 antibodies, all secondary HCC patients had TGFβR1-positive CSCs (Fig. 19, Table 7). Detailed analysis revealed that 51.8% (249/481) of all PTGFRN-positive circulating cancer cells were TGFβR1-positive, and 58% (279/481) of all PTGFRN-positive circulating cancer cells were B7-H3-positive, suggesting that PTGFRN-positive circulating cancer cells likely have a TGFβR1- and B7-H3-mediated immunosuppressive phenotype.
PTGFRN-양성 혈중 암세포가 2차 HCC 환자에서 숙주 면역 반응과 어떻게 연관되는지 추가로 조사하기 위해, 혈중 암세포를 항-ULBP1/63-D7/항-B7-H3 항체 또는 항-MICA/B/63-D7/항-B7-H3항체로 동시에 삼중염색 했다(도 20a, 도 20b, 표 7).To further investigate how PTGFRN-positive circulating cancer cells are associated with the host immune response in patients with secondary HCC, circulating cancer cells were simultaneously triple-stained with anti-ULBP1/63-D7/anti-B7-H3 antibodies or anti-MICA/B/63-D7/anti-B7-H3 antibodies (Fig. 20a, Fig. 20b, Table 7).
ULBP1 및 MICA/B는 NK 및 CD8 T 세포의 활성화 수용체 NKG2D에 대한 리간드로, 면역을 촉진하는 분자로 알려져 있다. ULBP1/63-D7/B7-H3 염색에서, 총 PTGFRN-양성 혈중 암세포의 4.1%(9/222)가 2차 HCC 환자에서 ULBP1-양성인 반면, 총 PTGFRN-양성 혈중 암세포의 50.5%(112/222)는 B7-H3 양성 이였다. MICA/B/63-D7/B7-H3 염색에서 모든 환자(18/18)는 MICA/B 양성 혈중 암세포를 보유했다. 2차 HCC 환자에서 전체 PTGFRN 양성 혈중 암세포의 34.7%(142/409)가 MICA/B 양성이었고, 전체 PTGFRN 양성 혈중 암세포의 65.3%(267/409)가 B7-H3 양성이었다. 이런 결과는 2차 혈중 암세포가 면역활성 분자인 ULBP1(4.1%)이나 MICA/B(34.7%)를 일부 발현하지만, 동시에 대부분 면역관문 조절분자로 알려진 B7-H3(65.3%)를 더 우세하게 발현한다는 것을 제시한다.ULBP1 and MICA/B are known as ligands for the activating receptor NKG2D of NK and CD8 T cells, and are molecules that promote immunity. In ULBP1/63-D7/B7-H3 staining, 4.1% (9/222) of total PTGFRN-positive circulating tumor cells were ULBP1-positive in secondary HCC patients, whereas 50.5% (112/222) of total PTGFRN-positive circulating tumor cells were B7-H3-positive. In MICA/B/63-D7/B7-H3 staining, all patients (18/18) had MICA/B-positive circulating tumor cells. In patients with secondary HCC, 34.7% (142/409) of total PTGFRN-positive circulating cancer cells were MICA/B-positive, and 65.3% (267/409) of total PTGFRN-positive circulating cancer cells were B7-H3-positive. These results suggest that secondary circulating cancer cells express some immunoactivating molecules, such as ULBP1 (4.1%) or MICA/B (34.7%), but at the same time, most of them preferentially express B7-H3 (65.3%), which is known as an immune checkpoint regulatory molecule.
PD-L1과 PD-L2는 NK 및 CD8 T 세포의 면역 억제 수용체 PD-1에 대한 잘 알려진 리간드이다. 따라서 혈중 암세포를 항 PD-L1/63-D7/항 B7-H3 항체 또는 항 PD-L2/63-D7/항 B7-H3 항체로 동시에 각각 삼중염색하였다(도 21a, 도 21b, 표 7).PD-L1 and PD-L2 are well-known ligands for the immunosuppressive receptor PD-1 on NK and CD8 T cells. Therefore, circulating cancer cells were simultaneously triple-stained with anti-PD-L1/63-D7/anti-B7-H3 antibodies or anti-PD-L2/63-D7/anti-B7-H3 antibodies, respectively (Fig. 21a, Fig. 21b, Table 7).
PD-L1/63-D7/B7-H3 염색에서 2차 간암 환자에서 전체 PTGFRN 양성 혈중 암세포의 37.8%(197/521)가 PD-L1 양성이었고, 전체 PTGFRN의 55.7%(290/521)가 B7-H3 양성이었다. PD-L2/63-D7/B7-H3 염색에서는 2차 간암 환자에서 전체 PTGFRN 양성 혈중 암세포의 17.5%(69/395)가 PD-L2 양성이었고, 전체 PTGFRN의 56.2%(222/395)가 B7-H3 양성이었다(도 21a, 도 21b, 표7). 한편 모든 환자(22/22)는 PD-L1 또는 PD-L2 양성 혈중 암세포를 보유하고 있었다.In PD-L1/63-D7/B7-H3 staining, 37.8% (197/521) of the total PTGFRN-positive circulating cancer cells in patients with secondary hepatocellular carcinoma were PD-L1 positive, and 55.7% (290/521) of the total PTGFRN were B7-H3 positive. In PD-L2/63-D7/B7-H3 staining, 17.5% (69/395) of the total PTGFRN-positive circulating cancer cells in patients with secondary hepatocellular carcinoma were PD-L2 positive, and 56.2% (222/395) of the total PTGFRN were B7-H3 positive (Fig. 21a, Fig. 21b, Table 7). Meanwhile, all patients (22/22) had PD-L1 or PD-L2 positive circulating cancer cells.
마지막으로, 우리는 PTGFRN 양성 혈중 암세포가 CD47과 관련이 있는지 조사했는데, 이는 CD47이 대식세포에 대해 저항하는 "don’t eat me" 신호이고 식세포 선천면역 감시를 억제하기 때문이다(Matlung, Szilagyi et al. 2017). CD47/63-D7/B7-H3 염색에서 2차 간암 환자의 경우 총 PTGFRN 양성 혈중암의 38.1%(115/302)가 CD47 양성이었고, 51.7%(156/302)가 B7-H3 양성이었다(도 22, 표 7). 그리고 모든 환자(14/14)들은 CD47 양성 혈중 암세포를 가지고 있었다.Finally, we investigated whether PTGFRN-positive CBCTs were associated with CD47, because CD47 is a “don’t eat me” signal for macrophages and inhibits phagocytic innate immune surveillance (Matlung, Szilagyi et al. 2017). In CD47/63-D7/B7-H3 staining, 38.1% (115/302) of the total PTGFRN-positive CBCTs in patients with secondary HCC were CD47-positive, and 51.7% (156/302) were B7-H3-positive (Fig. 22 , Table 7 ). And all patients (14/14) had CD47-positive CBCTs.
이와 같이 ULBP1(4.1%) 및 MICA/B(34.7%)와 같은 면역자극 수용체가 PTGFRN 양성 혈중 암세포에서 검출되었지만, TFGβR1(51.8%), PD-L1(37.8%), PD-L2(17.5%), CD47(38.1%) 및 B7-H3 (50.5-65.3%)과 같은 면역 관문 조절 분자가 2차 HCC 환자의 PTGFRN 양성 혈중 암세포에서 더욱 널리 검출되었다. 특히, B7-H3는 PTGFRN-양성 혈중 암세포에서 모두 우세하게(평균 57.6%) 발현되었다. 따라서 이러한 결과는 PTGFRN이 2차 간암 환자에서 혈중 암세포의 면역 회피를 담당하고, 면역 억제 수용체와 밀접하게 연관되어 발현되며 면역회피 역할을 수행할 것임을 제시한다. 이러한 결과는 혈중 암세포에서 63-D7 항체에 의해 인식되는 PTGFRN은 간암 병기나 진행을 표현하는 우수한 진단 마커로 작용할 수 있음을 제시한다.Although immunostimulatory receptors such as ULBP1 (4.1%) and MICA/B (34.7%) were detected in PTGFRN-positive circulating cancer cells, immune checkpoint regulatory molecules such as TFGβR1 (51.8%), PD-L1 (37.8%), PD-L2 (17.5%), CD47 (38.1%), and B7-H3 (50.5–65.3%) were more widely detected in PTGFRN-positive circulating cancer cells from secondary HCC patients. In particular, B7-H3 was predominantly expressed in all PTGFRN-positive circulating cancer cells (average 57.6%). Therefore, these results suggest that PTGFRN is responsible for immune evasion of circulating cancer cells in secondary HCC patients, and its expression is closely associated with immunosuppressive receptors and plays a role in immune evasion. These results suggest that PTGFRN recognized by 63-D7 antibody in circulating cancer cells may serve as an excellent diagnostic marker for indicating the stage or progression of liver cancer.
실시예 11. 단일클론항체 63-D7 유전자 및 아미노산 분석Example 11. Analysis of monoclonal antibody 63-D7 gene and amino acid
왕성하게 자라는 하이브리도마 63-D7 세포 5 X 106개를 원심 분리하여 수확한 후, 차가운 PBS로 2회 세척하고 RNAiso plus reagent(TaKaRa, Otsu, Japan)으로 제조사의 프로토콜에 따라 전체 RNA을 추출하였다. 전체 RNA의 A260을 Nanodrop을 통하여 측정하여 RNA 양을 정량 하였다. 전체 RNA 1 μg당 1 unit의 DNaseⅠ(TaKaRa)을 넣고 37 ℃에서 30분 동안 반응시켜 잔여의 DNA를 제거한 후, 50 mM EDTA 1 μl를 넣고 65 ℃에서 10분 동안 반응시켜 DNaseⅠ를 불활성화하고 전체 RNA를 변성시켰다. 전체 RNA와 Prime Script RT reagent Kit(TaKaRa)를 사용해 역전사중합효소 연쇄반응혼합액을 만들고, 제조사의 프로토콜에 따라 cDNA을 합성하였다. 합성된 cDNA로 중쇄 클로닝을 하기 위해서는 IgG1 불변영역에 해당하는 중합효소연쇄반응 프라이머인 5'-gga gtc gac ATA GAC AGA TGG GGG TGT CGT TTT GGC-3'(서열번호 20)인 올리고뉴클레오타이드 25 pmole과, 중쇄 항체 가변영역 N말단에 해당하는 프라이머인 염기서열 5'MH1 5'-ctt ccg gaa ttc SAR GTN MAG CTG SAG SAG TC-3'(서열번호 21) 올리고뉴클레오타이드 25pmole 및 5'MH2 5'-ctt ccg gaa ttc SAR GTN MAG CTG SAG SAG TCW GG-3'(서열번호 22) 올리고뉴클레오타이드 25pmole을 넣어 연쇄 중합반응혼합액을 만들었다. 경쇄 클로닝을 위해서는 카파 사슬 불변영역에 해당하는 프라이머인 5'-ggt gtc gac GGA TAC AGT TGG TGC AGC ATC-3'(서열번호 23) 올리고뉴클레오타이드와, 카파 사슬 가변영역 N 말단에 해당하는 프라이머인 5'MK 5'-cgg aag ctt GAY ATT GTG MTS ACM CAR WCT MCA-3'(서열번호 24) 올리고뉴클레오타이드을 사용하였다. 중합효소연쇄반응 생산물의 효율적인 클로닝을 위하여 중쇄의 경우는 5'-프라이머 말단에 EcoRI, 3'-프라이머 말단에 SalI 제한효소 자리를 부여하였다. 경쇄의 경우는 5'-프라이머 말단에 HindIII, 3'-프라이머 말단에 SalI 제한효소 자리를 부여하였다. 중쇄 또는 경쇄의 중합효소반응 프라이머와 1 unit의 i-pfu DNA Polymerase(iNtRON Biotechnology, Seoul, Korea)를 각각 섞은 후, 먼저 95 ℃에서 5분간 1회 반응하고, 95 ℃에서 1분, 35 ℃에서 1분, 72 ℃에서 1분을 한 cycle로 하여 30회 반응시켰다. 그 결과 중쇄 가변영역에 해당하는 DNA 절편으로 추정되는 길이인 약 400 bp, 경쇄 가변영역 해당되는 DNA 절편으로 추정되는 길이인 약 390 bp에 해당되는 위치에서 증폭된 DNA를 얻을 수 있었다.Vigorously growing hybridoma 63-D7 cells (5 × 10 6 cells) were harvested by centrifugation, washed twice with cold PBS, and total RNA was extracted using RNAiso plus reagent (TaKaRa, Otsu, Japan) according to the manufacturer's protocol. The A260 of total RNA was measured using Nanodrop to quantify the amount of RNA. 1 unit of DNase I (TaKaRa) per 1 μg of total RNA was added and reacted at 37 °C for 30 minutes to remove residual DNA. 1 μl of 50 mM EDTA was added and reacted at 65 °C for 10 minutes to inactivate DNase I and denature total RNA. A reverse transcription polymerase chain reaction mixture was prepared using total RNA and Prime Script RT reagent Kit (TaKaRa), and cDNA was synthesized according to the manufacturer's protocol. In order to perform heavy chain cloning using the synthesized cDNA, 25 pmole of an oligonucleotide having the polymerase chain reaction primer corresponding to the IgG1 constant region, 5'-gga gtc gac ATA GAC AGA TGG GGG TGT CGT TTT GGC-3' (SEQ ID NO: 20), and 25 pmole of an oligonucleotide having the base sequence 5'MH1 5'-ctt ccg gaa ttc SAR GTN MAG CTG SAG SAG TC-3' (SEQ ID NO: 21) and 25 pmole of an oligonucleotide 5'MH2 5'-ctt ccg gaa ttc SAR GTN MAG CTG SAG SAG TCW GG-3' (SEQ ID NO: 22) corresponding to the N-terminus of the heavy chain antibody variable region were added to prepare a chain polymerization reaction mixture. For light chain cloning, a 5'-ggt gtc gac GGA TAC AGT TGG TGC AGC ATC-3' (SEQ ID NO: 23) oligonucleotide primer corresponding to the kappa chain constant region and a 5'MK 5'-cgg aag ctt GAY ATT GTG MTS ACM CAR WCT MCA-3' (SEQ ID NO: 24) oligonucleotide primer corresponding to the N-terminus of the kappa chain variable region were used. For efficient cloning of polymerase chain reaction products, an EcoRI restriction site was provided at the end of the 5'-primer for the heavy chain and a SalI restriction site was provided at the end of the 3'-primer. For the light chain, a HindIII restriction site was provided at the end of the 5'-primer and a SalI restriction site was provided at the end of the 3'-primer. After mixing the heavy chain or light chain polymerase reaction primers and 1 unit of i-pfu DNA Polymerase (iNtRON Biotechnology, Seoul, Korea) respectively, the reaction was performed once at 95 °C for 5 minutes, and then 30 times at 95 °C for 1 minute, 35 °C for 1 minute, and 72 °C for 1 minute. As a result, amplified DNA was obtained at a position corresponding to approximately 400 bp, which is estimated to be a DNA fragment corresponding to the heavy chain variable region, and approximately 390 bp, which is estimated to be a DNA fragment corresponding to the light chain variable region.
증폭한 63-D7 유전자를 클로닝 하기 위하여, 먼저 중합효소연쇄반응 산물에 대해 중쇄는 EcoRI 및 SalI으로 처리하고, 경쇄는 HindIII 및 SalI으로 처리한 후, 1.0 %(w/v) 아가로스 젤에 전개시켜 FavorPrep GEL PCR Purification Kit(Favorgen, Pingtung, Taiwan)로 약 400 bp와 390 bp에 해당하는 DNA를 분리하였다. 중쇄유전자를 클로닝 할 벡터 pBluescript KS+를 EcoRI과 SalI으로 처리하고 경쇄유전자를 클로닝 할 벡터 pBluescript KS+를 HindIII와 SalI으로 처리한 다음, FavorPrep GEL PCR Purification Kit로 분리하였다. 이 두 DNA를 T4 DNA 연결효소(New England Biolab, Massachusetts, U.S.A)로 연결하고 대장균 DH5α에 CaCl2 방법으로 형질 전환하였다. 항체 유전자들의 DNA 염기서열분석을 위하여 상기의 여러 클론들을 100 ㎍/㎖의 암피실린이 함유된 3ml의 LB 배지에서 밤새 배양한 후 DNA-Spin plasmid mini prep kit(Intron, 한국)를 사용하여, 제조사의 프로토콜에 따라 플라스미드 DNA를 분리하고, 중쇄의 경우 약 400bp 크기의 DNA 삽입물을 가진 대장균 클론, 경쇄의 경우 약 390bp 크기의 DNA 삽입물을 가진 대장균 클론들을 선발하였다. 각각의 DNA 삽입물의 염기서열을 Nucleotide sequencing(Bionics, Seoul, Korea)을 통해 확인하였다.To clone the amplified 63-D7 gene, first, the heavy chain of the polymerase chain reaction product was treated with EcoRI and SalI, and the light chain was treated with HindIII and SalI, and then developed on a 1.0% (w/v) agarose gel. DNA corresponding to approximately 400 bp and 390 bp was isolated using FavorPrep GEL PCR Purification Kit (Favorgen, Pingtung, Taiwan). The vector pBluescript KS+ for cloning the heavy chain gene was treated with EcoRI and SalI, and the vector pBluescript KS+ for cloning the light chain gene was treated with HindIII and SalI, and then isolated using FavorPrep GEL PCR Purification Kit. These two DNAs were ligated with T4 DNA ligase (New England Biolab, Massachusetts, USA) and transformed into E. coli DH5α using the CaCl 2 method. For DNA sequence analysis of antibody genes, the above-mentioned multiple clones were cultured overnight in 3 ml of LB medium containing 100 ㎍/㎖ of ampicillin. Plasmid DNA was isolated using a DNA-Spin plasmid mini prep kit (Intron, Korea) according to the manufacturer's protocol. For the heavy chain, E. coli clones with DNA inserts of approximately 400 bp in size were selected, and for the light chain, E. coli clones with DNA inserts of approximately 390 bp in size were selected. The base sequence of each DNA insert was confirmed by nucleotide sequencing (Bionics, Seoul, Korea).
중쇄 및 경쇄 DNA의 염기서열을 아미노산으로 번역한 후 항체 구조에 따른 항원인식결정 부위를 Kabat numbering을 통해 정리한 결과, 중쇄는 subgroup ⅡB에 해당하고(도 23) 경쇄는 subgroup V(도 24)에 해당하였다. 각 서열에 항원 결합 부위인 CDR 1, 2, 3를 진한 글씨로 표시하였다. 항체의 아미노산 서열을 BLAST로 비교해본 결과 63-D7은 기존에 알려지지 않은 새로운 항체임을 확인하였다.After translating the base sequences of the heavy and light chain DNA into amino acids and organizing the antigen recognition determining sites according to the antibody structure using Kabat numbering, the heavy chain corresponded to subgroup ⅡB (Fig. 23) and the light chain corresponded to subgroup V (Fig. 24). The antigen binding sites,
Claims (22)
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| KR20230059919 | 2023-05-09 | ||
| KR10-2023-0059919 | 2023-05-09 | ||
| KR1020240060780A KR20240163543A (en) | 2023-05-09 | 2024-05-08 | Anti-ptgfrn monoclonal antibody and uses therof |
| KR10-2024-0060780 | 2024-05-08 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024232668A1 true WO2024232668A1 (en) | 2024-11-14 |
Family
ID=93430779
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/KR2024/006210 Pending WO2024232668A1 (en) | 2023-05-09 | 2024-05-09 | Anti-ptgfrn monoclonal antibody and use thereof |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024232668A1 (en) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20190365914A1 (en) * | 2013-07-11 | 2019-12-05 | Universite Francois Rabelais | Novel antibody-drug conjugates and the use of same in therapy |
| KR20200071740A (en) * | 2017-09-28 | 2020-06-19 | 임팩트-바이오 리미티드. | Universal platform for the production of inhibitory chimeric antigen receptors (ICAR) |
| US20210188972A1 (en) * | 2017-10-13 | 2021-06-24 | A&G Pharmaceutical, Inc. | Monoclonal antibodies and conjugates against prostaglandin f2 receptor inhibitor and uses thereof |
-
2024
- 2024-05-09 WO PCT/KR2024/006210 patent/WO2024232668A1/en active Pending
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20190365914A1 (en) * | 2013-07-11 | 2019-12-05 | Universite Francois Rabelais | Novel antibody-drug conjugates and the use of same in therapy |
| KR20200071740A (en) * | 2017-09-28 | 2020-06-19 | 임팩트-바이오 리미티드. | Universal platform for the production of inhibitory chimeric antigen receptors (ICAR) |
| US20210188972A1 (en) * | 2017-10-13 | 2021-06-24 | A&G Pharmaceutical, Inc. | Monoclonal antibodies and conjugates against prostaglandin f2 receptor inhibitor and uses thereof |
Non-Patent Citations (2)
| Title |
|---|
| LIN DANFENG, SHEN LESANG, LUO MENG, ZHANG KUN, LI JINFAN, YANG QI, ZHU FANGFANG, ZHOU DAN, ZHENG SHU, CHEN YIDING, ZHOU JIAOJIAO: "Circulating tumor cells: biology and clinical significance", SIGNAL TRANSDUCTION AND TARGETED THERAPY, vol. 6, no. 1, 1 December 2021 (2021-12-01), XP055954502, DOI: 10.1038/s41392-021-00817-8 * |
| MARQUEZ JORGE, DONG JIANPING, DONG CHUN, TIAN CHANGSHENG, SERRERO GINETTE: "Identification of Prostaglandin F2 Receptor Negative Regulator (PTGFRN) as an internalizable target in cancer cells for antibody-drug conjugate development", PLOS ONE, PUBLIC LIBRARY OF SCIENCE, US, vol. 16, no. 1, 27 January 2021 (2021-01-27), US , pages e0246197, XP093235777, ISSN: 1932-6203, DOI: 10.1371/journal.pone.0246197 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| CN106661110B (en) | anti-MUC 1 antibodies or antigen-binding fragments thereof and uses thereof | |
| WO2013187556A1 (en) | Novel antibody specific for clec14a and uses thereof | |
| US12227570B2 (en) | Anti-CEACAM6 antibodies and methods of use | |
| WO2017010744A1 (en) | Antigenic composition for detecting auto-antibody with specific response to exosomal protein eif3a, and method for diagnosing liver cancer using antigenic composition | |
| WO2012112013A2 (en) | A marker comprising anti-ck8/18 complex autoantibody and its use for diagnosing cancer | |
| WO2022231032A1 (en) | Anti-cntn4-specific antibodies and use thereof | |
| WO2015130115A1 (en) | Novel antibody specific for tspan8 and uses thereof | |
| WO2021091319A1 (en) | Glypican-3-specific modified aptamer and use thereof | |
| WO2024155098A1 (en) | Regulatory t cell surface antigen and antibody that specifically binds thereto | |
| WO2024232668A1 (en) | Anti-ptgfrn monoclonal antibody and use thereof | |
| WO2022124765A1 (en) | Gpc3-specific antibody and use thereof | |
| WO2022244908A1 (en) | Anti-bcam antibody or antigen-binding fragment thereof | |
| WO2012165926A2 (en) | Marker for diagnosing liver cancer containing anti-atic autoantibody, and composition for diagnosing liver cancer containing antigen thereof | |
| WO2017171373A2 (en) | Composition for suppressing resistance to egfr-targeting agent | |
| Ren et al. | The RNA/DNA‐binding protein PSF relocates to cell membrane and contributes cells' sensitivity to antitumor drug, doxorubicin | |
| KR20240163543A (en) | Anti-ptgfrn monoclonal antibody and uses therof | |
| WO2022131889A1 (en) | Use of taci protein | |
| WO2011118994A9 (en) | Nlk as a marker for diagnosis of liver cancer and as a therapeutic agent thereof | |
| CN116333137A (en) | A kind of CD103 antibody and its application | |
| JP5939855B2 (en) | Transferrin receptor antibody | |
| WO2024151091A1 (en) | Diagnostic composition and pharmaceutical composition comprising biomarker of tregs in tumor | |
| CN119613546B (en) | IL-10 antibodies and uses thereof | |
| Lim et al. | A monoclonal antibody recognizing CD98 on human embryonic stem cells shows anti-tumor activity in hepatocellular carcinoma xenografts | |
| WO2025264059A1 (en) | Antibody specifically binding to lrrc15 and use thereof | |
| WO2019022274A1 (en) | Antibody specifically binding to pauf protein, and use thereof |
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: 24803722 Country of ref document: EP Kind code of ref document: A1 |