WO2025018697A1 - Sample treatment method for cancer diagnosis, and method for providing information about cancer diagnosis by using same - Google Patents
Sample treatment method for cancer diagnosis, and method for providing information about cancer diagnosis by using same Download PDFInfo
- Publication number
- WO2025018697A1 WO2025018697A1 PCT/KR2024/009914 KR2024009914W WO2025018697A1 WO 2025018697 A1 WO2025018697 A1 WO 2025018697A1 KR 2024009914 W KR2024009914 W KR 2024009914W WO 2025018697 A1 WO2025018697 A1 WO 2025018697A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cancer
- pstat3
- antibody
- diagnosis
- sample
- 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
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N1/00—Sampling; Preparing specimens for investigation
- G01N1/28—Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
- G01N1/30—Staining; Impregnating ; Fixation; Dehydration; Multistep processes for preparing samples of tissue, cell or nucleic acid material and the like for analysis
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N1/00—Sampling; Preparing specimens for investigation
- G01N1/28—Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
- G01N1/36—Embedding or analogous mounting of samples
-
- 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
-
- 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
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N1/00—Sampling; Preparing specimens for investigation
- G01N1/28—Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
- G01N1/30—Staining; Impregnating ; Fixation; Dehydration; Multistep processes for preparing samples of tissue, cell or nucleic acid material and the like for analysis
- G01N2001/302—Stain compositions
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2440/00—Post-translational modifications [PTMs] in chemical analysis of biological material
- G01N2440/14—Post-translational modifications [PTMs] in chemical analysis of biological material phosphorylation
Definitions
- the present invention relates to a method for processing a sample for cancer diagnosis and a method for providing information on cancer diagnosis using the same.
- cancer Normally, cells divide, grow, and die to maintain a balance in cell number through cellular regulatory functions. However, if a change occurs in the genes of a cell for various reasons, the cell changes abnormally, matures incompletely, and proliferates excessively. This is called cancer.
- the present invention aims to provide a method for processing a blood sample separated from an individual and information that can diagnose cancer early, with high accuracy, and in a short period of time using the same.
- the present invention aims to provide a method for processing a sample for cancer diagnosis.
- the purpose of the present invention is to provide a method for providing information for cancer diagnosis using the above sample processing method.
- a method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment comprising the steps of: treating a sample separated from an organism with a fixing reagent to fix phosphorylated STAT3 present in cells of the sample; perforating the cell membrane of the cells; and detecting the fixed phosphorylated STAT3.
- a method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment wherein the sample separated from the subject in 1 above is unprocessed blood.
- a method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment wherein the fixing reagent in 1 above is formaldehyde or paraformaldehyde.
- a method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment wherein the cancer in the above 1 is any one selected from the group consisting of lung cancer, bile duct cancer, bladder cancer, kidney cancer, liver cancer, stomach cancer, and urinary tract cancer.
- a method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment wherein in the above 1, the fixed reagent is processed before the phosphorylated STAT3 in the blood is dephosphorylated.
- a method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment comprising: a step of treating a sample separated from an object with a fixing reagent to fix phosphorylated STAT3 present in cells of the sample; a step of perforating the cell membrane of the cell; a step of detecting the fixed phosphorylated STAT3; and a step of distinguishing between cancer patients and normal people based on information about the detected phosphorylated STAT3.
- a method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment wherein the sample separated from the subject in 6 above is untreated blood.
- a method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment wherein the fixing reagent in 6 above is formaldehyde or paraformaldehyde.
- a method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment wherein the cancer in the above 6 is any one selected from the group consisting of lung cancer, bile duct cancer, bladder cancer, kidney cancer, liver cancer, stomach cancer, and urinary tract cancer.
- a method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment wherein in the above 6, the fixed reagent is treated before the phosphorylated STAT3 in the blood is dephosphorylated.
- a method for providing information for predicting responsiveness to an immune checkpoint inhibitor comprising the step of measuring the expression level of fixed phosphorylated STAT3 in a sample isolated from an entity.
- a method for providing information for predicting responsiveness to an immune checkpoint inhibitor wherein the sample separated from the subject in the above 11 is untreated blood.
- a method for providing information for predicting responsiveness to an immune checkpoint inhibitor further comprising a step of predicting that responsiveness to an immune checkpoint inhibitor will be better than that of a control group if the expression level of the measured phosphorylated STAT3 is 20 to 60% in the above 11.
- the immune checkpoint inhibitor is any one selected from the group consisting of anti-PD-1 antibody, anti-PD-L1 antibody, anti-CTLA4 antibody, anti-PD-L2 antibody, LTF2 regulatory antibody, anti-LAG3 antibody, anti-A2aR antibody, anti-TIGIT antibody, anti-TIM-3 antibody, anti-B7-H3 antibody, anti-B7-H4 antibody, anti-VISTA antibody, anti-CD47 antibody, anti-BTLA antibody, anti-KIR antibody, anti-IDO antibody, cisplatin, and combinations thereof.
- the fixed phosphorylated STAT3 is obtained by treating the sample with a fixing reagent to fix phosphorylated STAT3 present in the cells of the sample, and a method for providing information for predicting responsiveness to an immune checkpoint inhibitor.
- sample processing method and information providing method of the present invention not only can cancer be diagnosed at an early stage, but also whether cancer has recurred after cancer surgery or treatment can be diagnosed.
- cancer can be diagnosed from a small amount of sample in a very short period of time.
- the information providing method for diagnosis of the present invention can diagnose cancer with higher sensitivity compared to existing diagnostic methods.
- the method for providing information for predicting responsiveness to an immune checkpoint inhibitor of the present invention can predict that responsiveness to an immune checkpoint inhibitor will be best at a specific phosphorylated pSTAT3 expression level.
- Figures 1a and 1b show that there is almost no expression of pSTAT3 in blood cells of healthy people when confirmed using a flow cytometer.
- pSTAT3 means phosphorylated STAT3.
- Figures 1c to 1e show that in the case of pSTAT3, which is expressed by IL-6, one of the cytokines that induces phosphorylation of STAT3, the expression decreases rapidly over time.
- Figures 2a and 2b show the expression of pSTAT3 in the unstimulated basal state in blood cells from healthy people and non-small cell lung cancer patients using flow cytometry.
- pSTAT3 was hardly expressed in the unstimulated basal state, some of the non-small cell lung cancer patients (patient 4) had high expression, and others (patient 1) had expression similar to that of healthy people.
- FIG. 2c is a schematic diagram of an experiment in which peripheral blood mononuclear cells (PBMCs) were separated from blood from non-small cell lung cancer patients and left at room temperature for different periods of time before isolation.
- PBMCs peripheral blood mononuclear cells
- Figures 2d and 2e confirm that the expression level of pSTAT3 in the unstimulated basal state rapidly decreases as the time left at room temperature after blood is separated from the subject increases.
- Figure 3a shows the results of examining the time left at room temperature for less than 1 hour before isolating peripheral blood mononuclear cells after blood collection, and confirming that pSTAT3 expression in the unstimulated basal state was observed in all non-small cell lung cancer patients compared to healthy people.
- Figures 3b to 3f show that the unstimulated basal pSTAT3 expression in non-small cell lung cancer patients is constant and independent of cancer stage, tumor histology, metastasis, and EGFR mutation. In particular, it was confirmed that the level of unstimulated basal pSTAT3 expression differs significantly between healthy people and cancer patients from stage 1 onwards.
- Figure 4a shows which cells among the immune cells of non-small cell lung cancer patients express pSTAT3 in the unstimulated basal state. It was confirmed that pSTAT3 is hardly expressed in B cells, NK cells, and monocytes, but is expressed in the unstimulated basal state in CD4 T cells and CD8 T cells.
- Figures 4b and 4c show the results confirming that the CD27+CD45RA+ group had the highest expression of pSTAT3 in the unstimulated basal state in both CD4 T cells and CD8 T cells.
- Figures 5a and 5b show the expression of pSTAT3 in the unstimulated basal state before and after surgery in non-small cell lung cancer patients to determine whether the unstimulated basal pSTAT3 detected in cancer patients is due to cancer, and if so, whether the unstimulated basal pSTAT3 decreases or disappears after cancer surgery. It was confirmed that the expression of pSTAT3 in the unstimulated basal state decreases in the entire immune cell population in the blood after surgery.
- Figures 5c and 5d show the expression of pSTAT3 in CD27+CD45RA+ CD4 T cells in the unstimulated baseline before and after surgery in patients with non-small cell lung cancer.
- Figure 6a shows the unstimulated basal state of pSTAT3 in whole peripheral blood cells from various cancer types.
- Figure 6b shows pSTAT3 in the unstimulated basal state in CD27+CD45RA+ CD4 T cells in various cancer types.
- Figure 6c is a numerical representation of the results of Figure 6a.
- Figure 6d is a numerical representation of the results of Figure 6b.
- Figure 7a shows the results of confirming pSTAT3 expression after stimulating blood immune cells of a healthy person using various types of cytokines (10 ng/ml).
- Figure 7b shows the results of confirming pSTAT3 in various immune cells in the unstimulated basal state of the patient and pSTAT3 induced by cytokines (IL-6, IL-21, IFN- ⁇ , IL-10 or no stimulation (NS); 0.5 ng/ml) of a healthy person.
- cytokines IL-6, IL-21, IFN- ⁇ , IL-10 or no stimulation (NS); 0.5 ng/ml
- Figure 7c is the result of principal component analysis based on the values obtained in Figure 7b.
- Figures 7d to 7f show the results of confirming pSTAT3 expression in each immune cell after stimulating PBMCs of healthy people with IL-6 (0.5 ng/ml).
- Figure 7d shows pSTAT3 expression in each immune cell in total PBMCs
- Figures 7e and 7f show pSTAT3 expression in CD4 T cells and CD8 T cells, respectively.
- Figure 8a is a schematic diagram illustrating an experimental method for finding factors that induce pSTAT3 expression in the unstimulated basal state using PBMCs and serum from non-small cell lung cancer (NSCLC) patients.
- NSCLC non-small cell lung cancer
- Figure 8b shows the results of measuring the concentration of inflammatory cytokines (IL-1 ⁇ , IL-6, IL-8, IL-10, IL-12p70, TNF) in the serum of NSCLC patients using BD Cytometric Bead Array.
- IL-1 ⁇ , IL-6, IL-8, IL-10, IL-12p70, TNF inflammatory cytokines
- Figures 8c to 8e show the results of co-culturing the serum of NSCLC patients with PBMCs of healthy people and measuring the expression of pSTAT3 after 1 hour.
- Figure 8c shows the expression of pSTAT3 in each immune cell in total PBMCs
- Figures 8d and 8e show the expression of pSTAT3 in CD4 T cells and CD8 T cells, respectively.
- Serum-induced pSTAT3 was confirmed in CD4 T cells and CD8 T cells, and was the highest in the CD27+CD45RA+ group.
- Figure 8f shows the proportional relationship between the serum IL-6 concentration of NSCLC patients and the serum-induced pSTAT3 of CD27+CD45RA+ CD4 T cells (CD4 Tn) measured after co-culture of the same serum with PBMCs from healthy people.
- Figures 8g and 8h show that when serum from NSCLC patients was co-cultured with PBMCs from healthy people, serum-induced pSTAT3 was not detected at all when IL-6 signaling was blocked by treatment with anti-IL-6 or anti-IL-6R antibodies.
- Figure 8i shows the proportional relationship between the unstimulated basal pSTAT3 expression of CD4 Tn obtained from PBMCs of NSCLC patients and the serum-induced pSTAT3 of CD4 Tn obtained previously.
- Figure 8j shows the proportional relationship between the unstimulated basal pSTAT3 expression of CD4 Tn obtained from PBMCs of NSCLC patients and the serum IL-6 concentration obtained previously.
- Figure 8k shows the results of dividing NSCLC patients into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to the level of pSTAT3 expression in the unstimulated basal state in each group.
- Figure 8l shows the results of dividing NSCLC patients into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to the level of pSTAT3 expression in the unstimulated basal state, and showing the serum IL-6 concentrations in each group.
- Figure 9a shows the correlation between the expression of pSTAT3 in the unstimulated basal state of CD27+CD45RA+ CD4 T cells (CD4 Tn) analyzed in PBMCs of NSCLC patients and the expression of CD206 in TAMs (CD14+CD11b+HLA-DR+CD11c+) analyzed in TILs.
- Figures 9b and 9c show the results of comparing the expression levels of CD206 in TAMs in each group when NSCLC patients were divided into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to the expression levels of pSTAT3 in the unstimulated basal state.
- Figure 9d shows the correlation between the expression of pSTAT3 in the unstimulated basal state of CD27+CD45RA+ CD4 T cells (CD4 Tn) analyzed in PBMCs of NSCLC patients and the proportion of CD103+CD39+ cells in CD8+ TIL.
- Figures 9e and 9f show the results comparing the ratio of CD103+CD39+ cells in CD8+ TILs in the pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi patient groups.
- Figure 10a shows the results of dividing stage IV NSCLC patients who received pembrolizumab monotherapy or combination therapy (pemetrexed + cisplatin) as first-line chemotherapy into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to the level of pSTAT3 expression in the unstimulated basal state of CD27+CD45RA+ CD4 T cells (CD4 Tn).
- pembrolizumab monotherapy or combination therapy pemetrexed + cisplatin
- Figure 10b compares the response after 3 months of immunotherapy in each patient group when pSTAT3 ex vivo -lo and pSTAT3 ex vivo -int were grouped together.
- PD indicates progressive disease
- SD indicates stable disease
- PR indicates partial response.
- Figure 10c shows a comparison of the responses after 3 months of immunotherapy of pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi when pSTAT3 ex vivo -lo and pSTAT3 ex vivo -int were divided into separate patient groups.
- PD indicates progressive disease
- SD indicates stable disease
- PR indicates partial response.
- the present invention provides a method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment, comprising the steps of treating a sample separated from an individual with a fixing reagent to fix phosphorylated STAT3 present in cells of the sample; the step of perforating the cell membrane of the cells; and the step of detecting the fixed phosphorylated STAT3.
- the subject means an animal that can develop cancer, or an animal from which information for diagnosing cancer is to be provided.
- the animal may be a mammal, and preferably may be a dog, cat, or human, but is not limited thereto.
- the sample may be a biological sample taken from any body fluid or tissue of the subject, such as blood, saliva, urine, tissue, cells, lymph, bone marrow fluid, spinal fluid, brain extract, synovial fluid or tissue fluid.
- the sample may be blood.
- Blood is unprocessed blood, and the term "unprocessed blood” refers to blood collected from a patient without being treated with any additional stimulating factors such as cytokines. That is, the method for processing a sample of the present invention detects phosphorylated STAT3 in blood separated from an individual without being treated with any stimulating factors.
- the blood may preferably be human peripheral blood, but is not limited thereto.
- Fixation reagent refers to any reagent capable of fixing the phosphorylation state of a protein, and may be formaldehyde or paraformaldehyde. Products available include, but are not limited to, BD Cytofix TM Fixation Buffer and eBioscience TM IC Fixation Buffer.
- Phosphorylated STAT3 refers to a state in which a phosphate group is attached to the STAT3 protein (Signal Transducer and activator of transcription 3), specifically, it refers to a state in which a phosphate group is attached to a tyrosine residue of the STAT3 protein.
- pSTAT3 in unstimulated basal state refers to pSTAT3 detected using the blood processing method of the present invention without treating a separate stimulating factor such as a cytokine.
- pSTAT3 in unstimulated basal state can be used interchangeably with "ex-vivo pSTAT3".
- the step of perforating the cell membrane is a step of permeabilizing the cell to detect STAT3 within the cell.
- the step of imposing permeability may use any method known in the art, and specifically may be treating a permeabilization reagent.
- the permeabilization reagent refers to a reagent that helps facilitate the detection of phosphorylated STAT3 within the cell, and may be methanol, ethanol, and BD Permeabilizing solution 3, but is not limited thereto.
- Detection can be performed using any method known in the art that can identify the phosphorylated STAT3 protein.
- detection can be performed using antibodies, detection can be performed using protein staining, detection can be performed using mass spectrometry, etc.
- detection can be performed using flow cytometry, ELISA, Western blot, immunoprecipitation assay, complement fixation assay, and can be performed using an ELISA kit or a kit for implementing Western blot, immunoprecipitation assay, complement fixation assay, or flow cytometry.
- the cancer may preferably be any one selected from the group consisting of lung cancer, bile duct cancer, bladder cancer, kidney cancer, liver cancer, stomach cancer, and urinary tract cancer, but is not limited thereto.
- Early diagnosis is the diagnosis of a disease condition when the disease has not progressed much. In the case of early diagnosis of cancer, it may be the diagnosis of cancer at a low stage. For example, it may be the diagnosis of cancer at stage 1.
- Diagnosing recurrence after treatment can mean diagnosing or monitoring whether cancer comes back after surgery or treatment.
- the fixing reagent may be treated before the phosphorylated STAT3 in the blood is dephosphorylated.
- the dephosphorylation is the removal of a phosphate group from an organic compound by hydrolysis, and may be the removal of a phosphate group attached to a STAT3 protein.
- the inventors of the present invention have confirmed that when phosphorylated STAT3 is present in blood separated from an individual, dephosphorylation of the phosphorylated STAT3 occurs after a certain period of time. Accordingly, the method for treating blood of the present invention fixes phosphorylated STAT3 with a fixing reagent before the dephosphorylation occurs, and detects it.
- the present invention can provide a method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment, comprising the steps of treating a fixing reagent on a sample separated from an individual to fix phosphorylated STAT3 present in cells of the sample; the step of perforating the cell membrane of the cell; the step of detecting the fixed phosphorylated STAT3; and the step of distinguishing between cancer patients and normal people based on information about the detected phosphorylated STAT3.
- the present invention detects pSTAT3 in blood separated from an individual, and since pSTAT3 is detected in cancer patients, while pSTAT3 is hardly detected in normal people, when pSTAT3 is detected, information can be provided that allows for early diagnosis of a cancer patient.
- the present invention provides a method for providing information for predicting responsiveness to an immune checkpoint inhibitor, comprising the step of measuring the expression level of fixed phosphorylated STAT3 in a sample isolated from an individual.
- the present invention provides a method for providing information for predicting responsiveness to an immune checkpoint inhibitor, wherein the sample separated from the subject is untreated blood.
- the present invention provides a method for providing information for predicting responsiveness to an immune checkpoint inhibitor, further comprising a step of predicting that responsiveness to an immune checkpoint inhibitor will be good compared to a control group if the expression level of the measured phosphorylated STAT3 is 20 to 60%.
- the expression level of the above phosphorylated STAT3 refers to the expression level in CD4 T cells in the sample, and represents the ratio of CD4 T cells expressing phosphorylated STAT3 among the total CD4 T cells in the sample.
- the above-mentioned expression amount of phosphorylated STAT3 refers to the expression amount in CD27+CD45RA+ CD4 T cells in the sample, and refers to the ratio of CD27+CD45RA+ CD4 T cells expressing phosphorylated STAT3 among the total CD27+CD45RA+ CD4 T cells in the sample.
- it may be the ratio of CD27+CD45RA+ CD4 T cells expressing phosphorylated STAT3 among CD27+CD45RA+ CD4 T cells in blood, but is not limited thereto.
- control group may be, for example, a group in which the measured expression level of phosphorylated STAT3 is less than 20% or a group in which the expression level is greater than 60%.
- the immune checkpoint inhibitor may be any one selected from the group consisting of anti-PD-1 antibody, anti-PD-L1 antibody, anti-CTLA4 antibody, anti-PD-L2 antibody, LTF2 regulatory antibody, anti-LAG3 antibody, anti-A2aR antibody, anti-TIGIT antibody, anti-TIM-3 antibody, anti-B7-H3 antibody, anti-B7-H4 antibody, anti-VISTA antibody, anti-CD47 antibody, anti-BTLA antibody, anti-KIR antibody, anti-IDO antibody, cisplatin, and combinations thereof.
- the immune checkpoint inhibitor is Pembrolizumab or a combination of Pembrolizumab and Cisplatin.
- the present invention provides a method for providing information for predicting responsiveness to an immune checkpoint inhibitor, wherein the fixed phosphorylated STAT3 is obtained by treating the sample with a fixing reagent to fix phosphorylated STAT3 present in the cells of the sample.
- Untreated PBMCs or cytokine-treated PBMCs were fixed with fixation reagent (BD IC fixation buffer) at 4°C for 20 minutes. After permeabilization with 99.8% methanol at -20°C for 30 minutes, the samples were stained with the required fluorescent conjugated antibodies. The stained samples were analyzed by flow cytometry. All pSTAT3 assays of the present invention were analyzed by the above method.
- pSTAT3 in the unstimulated basal state was confirmed by flow cytometry in total PBMCs of healthy individuals.
- the control group (isotype control) used the corresponding isotype antibody instead of anti-STAT3 (pY705) antibody (Fig. 1a).
- the expression of pSTAT3 in the unstimulated basal state was almost absent in total PBMCs of several healthy individuals (Fig. 1b).
- Example 4 Determination of pSTAT3 expression in unstimulated basal states between healthy individuals and non-small cell lung cancer patients
- Example 5 Confirmation of pSTAT3 expression level according to storage time after blood collection
- PBMCs peripheral blood mononuclear cells
- Fig. 2c peripheral blood mononuclear cells
- Example 6 Comparison of pSTAT3 expression in unstimulated basal state, with blood collected at room temperature left for less than 1 hour
- PBMCs were isolated, and unstimulated basal pSTAT3 was analyzed in total PBMCs.
- NSCLC patients were also divided and analyzed according to stage, tumor histology, metastasis, and EGFR mutation.
- unstimulated basal pSTAT3 expression was confirmed in all NSCLC patients compared to healthy people (Fig. 3a).
- the unstimulated basal pSTAT3 expression of NSCLC patients was constant regardless of cancer stage, tumor histology, metastasis, and EGFR mutation, and in particular, it was confirmed that the values were clearly different from those of healthy people starting from Stage 1 cancer patients (Figs. 3b to 3f).
- Example 7 Identification of immune cells expressing pSTAT3 in the unstimulated basal state
- CD4 T cells and CD8 T cells of NSCLC patients were classified into four subsets according to the presence of CD27 and CD45RA, and pSTAT3 in the unstimulated basal state was compared and analyzed in each subset. As a result, it was confirmed that the CD27+CD45RA+ group had the highest expression of pSTAT3 in the unstimulated basal state in both CD4 T cells and CD8 T cells (Fig. 4b, 4c).
- Example 8 Confirmation of pSTAT3 detection in unstimulated baseline before and after surgery in patients with non-small cell lung cancer
- PBMCs Blood of patients scheduled to undergo surgical removal of cancer (Before OP) was collected, and PBMCs were isolated and stored. When the same patients revisited the hospital (between 2 and 8 months after surgery; After OP), blood was collected once more, and PBMCs were isolated and stored. The stored PBMCs were thawed all at once, and pSTAT3 in the unstimulated basal state was analyzed. Before OP included the blood of all patients, but postoperative blood was divided into 2 to 4 months, 4 to 6 months, and 6 to 8 months depending on the time of revisit. As a result, it was confirmed that the expression of pSTAT3 in the unstimulated basal state was reduced in the entire immune cell population in the blood after surgery (Fig. 5a, 5b), and it was also confirmed that the expression of pSTAT3 in the unstimulated basal state was reduced in CD27+CD45RA+ CD4 T cells (Fig. 5c, 5d).
- Example 9 Identification of pSTAT3 in unstimulated basal state in various cancer types
- PBMCs from the blood of patients with lung (specifically non-small cell lung cancer), skin (specifically melanoma), bile duct (specifically gallbladder), bladder (specifically bladder cancer), kidney (specifically renal cell carcinoma), liver (specifically hepatocellular carcinoma), stomach (specifically advanced gastric cancer), and urothelial (specifically urothelial cancer) cancers and analyzed pSTAT3 in the unstimulated basal state.
- lung specifically non-small cell lung cancer
- skin specifically melanoma
- bile duct specifically gallbladder
- bladder specifically bladder cancer
- kidney specifically renal cell carcinoma
- liver specifically hepatocellular carcinoma
- stomach specifically advanced gastric cancer
- urothelial specifically urothelial cancer
- Example 10 Stimulation of blood immune cells in healthy individuals using cytokines
- PBMCs from healthy people were treated with various cytokines (10 ng/ml) and cultured in a CO 2 incubator at 37°C for 30 minutes, and then pSTAT3 was analyzed. As a result, it was confirmed that the cytokines that induce pSTAT3 are IL-6, IL-10, IL-21, and IFN-b (Fig. 7a).
- pSTAT3 in the unstimulated basal state of patients and pSTAT3 induced by cytokines (IL-6, IL-21, IFN- ⁇ , IL-10, or no stimulation (NS); 0.5 ng/ml) of healthy people were identified and compared in various immune cells.
- IL-6 cytokines
- IL-21 IL-21
- IFN- ⁇ IL-10
- NS no stimulation
- pSTAT3 expressed in each immune cell was confirmed.
- pSTAT3 was induced only in CD4 T cells and CD8 T cells in PBMCs from healthy people treated with IL-6, and among them, pSTAT3 expression was highest in the CD27+CD45RA+ group (Figs. 7d to 7f).
- Example 11 Identification of pSTAT3 inducers in unstimulated basal state in NSCLC patients using plasma
- the concentration of IL-6 in the serum of NSCLC patients was confirmed to be significantly higher than that of other inflammatory cytokines (Fig. 8b).
- serum-induced pSTAT3 was highly expressed in T cells, especially in the CD27+CD45RA+ group (Figs. 8c to 8e).
- This serum-induced pSTAT3 was directly proportional to the concentration of IL-6 in the serum (Fig. 8f), and when the IL-6 signaling was disrupted by adding anti-IL-6 or anti-IL-6R antibodies during co-culture of serum and PBMCs, the serum-induced pSTAT3 observed previously was not observed at all (Figs. 8g, 8h).
- the expression level of pSTAT3 in the unstimulated basal state analyzed in PBMCs of NSCLC patients, serum-induced pSTAT3, and serum IL-6 concentration showed a proportional relationship (Figs. 8i, 8j).
- the analysis of pSTAT3 in the unstimulated basal state showed superior sensitivity compared to the analysis of serum IL-6 concentration. That is, when NSCLC patients were divided into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to the level of pSTAT3 expression in the unstimulated basal state (Fig. 8k), pSTAT3 ex vivo -lo and pSTAT3 ex vivo -int could not be distinguished based on serum IL-6 concentration alone (Fig. 8l).
- Example 12 Correlation between unstimulated baseline pSTAT3 and tumor-infiltrating immune cell differentiation in NSCLC patients
- PBMCs and tumor-infiltrating lymphocytes (TILs) from NSCLC patients were examined.
- CD206+% TAMs were higher in the pSTAT3 ex vivo -int and pSTAT3 ex vivo -hi patient groups than in the pSTAT3 ex vivo -lo (Figs. 9b, 9c).
- Example 13 Correlation between baseline pSTAT3 and response to immune checkpoint inhibitors in NSCLC patients
- pSTAT3 ex vivo -lo When the patients were divided into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to pSTAT3 in the unstimulated basal state (Fig. 10a), pSTAT3 ex vivo -hi, in which a high level of IL-6 in the serum was expected to be detected based on the results of the previous Example 11, showed a lower proportion of patients who showed a partial response (PR) compared to other patients (pSTAT3 ex vivo -lo/int) (Fig. 10b).
- PR partial response
- the proportion of PR patients was particularly high in the pSTAT3 ex vivo -int group, and the pSTAT3 ex vivo -lo group was not as responsive as the pSTAT3 ex vivo -hi group (Fig. 10c). In other words, it was confirmed that the highest responsiveness to immune checkpoint inhibitors can be predicted in the patient group in which the basal pSTAT3 level in the blood was approximately 20 to 60% of unstimulated level.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Chemical & Material Sciences (AREA)
- Molecular Biology (AREA)
- Biomedical Technology (AREA)
- Biochemistry (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- General Health & Medical Sciences (AREA)
- Analytical Chemistry (AREA)
- Physics & Mathematics (AREA)
- Urology & Nephrology (AREA)
- Hematology (AREA)
- Food Science & Technology (AREA)
- Medicinal Chemistry (AREA)
- Biotechnology (AREA)
- Microbiology (AREA)
- Cell Biology (AREA)
- Oncology (AREA)
- Hospice & Palliative Care (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Description
본 발명은 암 진단을 위한 시료 처리 방법 및 이를 이용한 암 진단에 대한 정보 제공 방법에 관한 것이다. The present invention relates to a method for processing a sample for cancer diagnosis and a method for providing information on cancer diagnosis using the same.
정상적으로는 세포는 세포 내 조절기능에 의해 분열하며 성장하고 죽어 없어지기도 하여 세포수의 균형을 유지하는데, 여러가지 이유로 인해 세포의 유전자에 변화가 일어나면 비정상적으로 세포가 변하여 불완전하게 성숙하고, 과다하게 증식하게 되는데 이를 암(cancer)이라고 한다.Normally, cells divide, grow, and die to maintain a balance in cell number through cellular regulatory functions. However, if a change occurs in the genes of a cell for various reasons, the cell changes abnormally, matures incompletely, and proliferates excessively. This is called cancer.
최근 암 환자의 액체 생검을 이용한 다양한 암 진단 방법이 연구 및 개발되는 추세이다. 이 중 환자의 혈액 또는 혈장 내의 circulating tumor DNA (ctDNA) 검출하는 방법이 대표적이다. 다만, ctDNA 분석 방법이 정확성 측면에서 비교적 효용성이 높은 방법이기는 하나 시간, 비용 및 접근성에 상당한 제약이 따르며, 민감도 증폭 문제가 존재한다. 따라서, 본 발명은 개체로부터 분리된 혈액 시료를 처리하는 방법 및 이를 이용하여 암을 조기에 높은 정확도와 단 시간 내에 진단할 수 있는 정보를 제공하고자 하였다. Recently, various cancer diagnosis methods using liquid biopsy of cancer patients are being studied and developed. Among these, a method of detecting circulating tumor DNA (ctDNA) in the patient's blood or plasma is representative. However, although the ctDNA analysis method is a relatively useful method in terms of accuracy, it has significant limitations in time, cost, and accessibility, and there is a problem of sensitivity amplification. Therefore, the present invention aims to provide a method for processing a blood sample separated from an individual and information that can diagnose cancer early, with high accuracy, and in a short period of time using the same.
본 발명은 암 진단을 위한 시료 처리 방법을 제공하는 것을 목적으로 한다. The present invention aims to provide a method for processing a sample for cancer diagnosis.
본 발명은 상기 시료 처리 방법을 이용하여 암 진단을 위한 정보 제공 방법을 제공하는 것을 목적으로 한다. The purpose of the present invention is to provide a method for providing information for cancer diagnosis using the above sample processing method.
1. 개체로부터 분리된 시료에 고정 시약을 처리하여 상기 시료의 세포 내에 존재하는 인산화된 STAT3를 고정시키는 단계; 상기 세포의 세포막을 천공하는 단계; 및 상기 고정된 인산화된 STAT3를 검출하는 단계를 포함하는, 암의 조기 진단 또는 치료 후 재발 여부 진단을 위한 시료의 처리 방법.1. A method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment, comprising the steps of: treating a sample separated from an organism with a fixing reagent to fix phosphorylated STAT3 present in cells of the sample; perforating the cell membrane of the cells; and detecting the fixed phosphorylated STAT3.
2. 위 1에 있어서, 상기 개체로부터 분리된 시료는 무처리 혈액인, 암의 조기 진단 또는 치료 후 재발 여부 진단을 위한 시료의 처리 방법.2. A method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment, wherein the sample separated from the subject in 1 above is unprocessed blood.
3. 위 1에 있어서, 상기 고정 시약은 포름알데하이드 또는 파라포름알데하이드인, 암의 조기 진단 또는 치료 후 재발 여부 진단을 위한 시료의 처리 방법.3. A method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment, wherein the fixing reagent in 1 above is formaldehyde or paraformaldehyde.
4. 위 1에 있어서, 상기 암은 폐암, 담관암, 방광암, 신장암, 간암, 위암 및 요로암으로 이루어진 군에서 선택되는 어느 하나인, 암의 조기 진단 또는 치료 후 재발 여부 진단을 위한 시료의 처리 방법.4. A method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment, wherein the cancer in the above 1 is any one selected from the group consisting of lung cancer, bile duct cancer, bladder cancer, kidney cancer, liver cancer, stomach cancer, and urinary tract cancer.
5. 위 1에 있어서, 상기 고정 시약은 상기 혈액의 인산화된 STAT3가 탈인산화되기 전에 처리되는 것인, 암 조기 진단 또는 치료 후 재발 여부 진단을 위한 시료의 처리 방법.5. A method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment, wherein in the above 1, the fixed reagent is processed before the phosphorylated STAT3 in the blood is dephosphorylated.
6. 개체로부터 분리된 시료에 고정 시약을 처리하여 상기 시료의 세포 내에 존재하는 인산화된 STAT3를 고정시키는 단계; 상기 세포의 세포막을 천공하는 단계; 상기 고정된 인산화된 STAT3를 검출하는 단계; 및 상기 검출된 인산화된 STAT3에 대한 정보를 기초로 암 환자와 정상인을 구분하는 단계를 포함하는, 암의 조기 진단 또는 치료 후 재발 여부 진단을 위한 정보제공 방법.6. A method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment, comprising: a step of treating a sample separated from an object with a fixing reagent to fix phosphorylated STAT3 present in cells of the sample; a step of perforating the cell membrane of the cell; a step of detecting the fixed phosphorylated STAT3; and a step of distinguishing between cancer patients and normal people based on information about the detected phosphorylated STAT3.
7. 위 6에 있어서, 상기 개체로부터 분리된 시료는 무처리 혈액인, 암의 조기 진단 또는 치료 후 재발 여부 진단을 위한 정보제공 방법.7. A method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment, wherein the sample separated from the subject in 6 above is untreated blood.
8. 위 6에 있어서, 상기 고정 시약은 포름알데하이드 또는 파라포름알데하이드인, 암의 조기 진단 또는 치료 후 재발 여부 진단을 위한 정보제공 방법.8. A method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment, wherein the fixing reagent in 6 above is formaldehyde or paraformaldehyde.
9. 위 6에 있어서, 상기 암은 폐암, 담관암, 방광암, 신장암, 간암, 위암 및 요로암으로 이루어진 군에서 선택되는 어느 하나인, 암의 조기 진단 또는 치료 후 재발 여부 진단을 위한 정보제공 방법.9. A method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment, wherein the cancer in the above 6 is any one selected from the group consisting of lung cancer, bile duct cancer, bladder cancer, kidney cancer, liver cancer, stomach cancer, and urinary tract cancer.
10. 위 6에 있어서, 상기 고정 시약은 상기 혈액의 인산화된 STAT3가 탈인산화되기 전에 처리되는 것인, 암의 조기 진단 또는 치료 후 재발 여부 진단을 위한 정보제공 방법.10. A method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment, wherein in the above 6, the fixed reagent is treated before the phosphorylated STAT3 in the blood is dephosphorylated.
11. 개체로부터 분리된 시료의 고정된 인산화된 STAT3의 발현량을 측정하는 단계를 포함하는 면역관문억제제에 대한 반응성 예측을 위한 정보 제공 방법.11. A method for providing information for predicting responsiveness to an immune checkpoint inhibitor, comprising the step of measuring the expression level of fixed phosphorylated STAT3 in a sample isolated from an entity.
12. 위 11에 있어서, 상기 개체로부터 분리된 시료는 무처리 혈액인, 면역관문억제제에 대한 반응성 예측을 위한 정보 제공 방법.12. A method for providing information for predicting responsiveness to an immune checkpoint inhibitor, wherein the sample separated from the subject in the above 11 is untreated blood.
13. 위 11에 있어서, 상기 측정된 인산화된 STAT3의 발현량이 20 내지 60%이면 대조군 대비 면역관문억제제에 대한 반응성이 좋을 것으로 예측하는 단계를 더 포함하는, 면역관문억제제에 대한 반응성 예측을 위한 정보 제공 방법.13. A method for providing information for predicting responsiveness to an immune checkpoint inhibitor, further comprising a step of predicting that responsiveness to an immune checkpoint inhibitor will be better than that of a control group if the expression level of the measured phosphorylated STAT3 is 20 to 60% in the above 11.
14. 위 11에 있어서, 상기 면역관문억제제는 항-PD-1 항체, 항-PD-L1 항체, 항-CTLA4 항체, 항 PD-L2 항체, LTF2 조절 항체, 항-LAG3 항체, 항-A2aR 항체, 항-TIGIT 항체, 항-TIM-3 항체, 항-B7-H3 항체, 항-B7-H4 항체, 항-VISTA 항체, 항-CD47 항체, 항-BTLA 항체, 항-KIR 항체, 항-IDO 항체, 시스플라틴(cisplatin) 및 이의 조합으로 이루어진 군으로부터 선택되는 어느 하나인, 면역관문억제제에 대한 반응성 예측을 위한 정보 제공 방법.14. A method for providing information for predicting responsiveness to an immune checkpoint inhibitor, wherein in the above 11, the immune checkpoint inhibitor is any one selected from the group consisting of anti-PD-1 antibody, anti-PD-L1 antibody, anti-CTLA4 antibody, anti-PD-L2 antibody, LTF2 regulatory antibody, anti-LAG3 antibody, anti-A2aR antibody, anti-TIGIT antibody, anti-TIM-3 antibody, anti-B7-H3 antibody, anti-B7-H4 antibody, anti-VISTA antibody, anti-CD47 antibody, anti-BTLA antibody, anti-KIR antibody, anti-IDO antibody, cisplatin, and combinations thereof.
15. 위 11에 있어서, 상기 고정된 인산화된 STAT3는 상기 시료에 고정 시약을 처리하여 상기 시료의 세포 내에 존재하는 인산화된 STAT3를 고정시켜 얻어진, 면역관문억제제에 대한 반응성 예측을 위한 정보 제공 방법.15. In the above 11, the fixed phosphorylated STAT3 is obtained by treating the sample with a fixing reagent to fix phosphorylated STAT3 present in the cells of the sample, and a method for providing information for predicting responsiveness to an immune checkpoint inhibitor.
본 발명의 시료 처리 방법 및 정보 제공 방법을 이용하여 조기에 암을 진단할 수 있을 뿐 아니라, 암 수술 또는 치료 후 암의 재발 여부를 진단할 수 있다. Using the sample processing method and information providing method of the present invention, not only can cancer be diagnosed at an early stage, but also whether cancer has recurred after cancer surgery or treatment can be diagnosed.
본 발명의 시료 처리 방법 및 정보 제공 방법을 이용하여 암을 높은 정확도로 진단할 수 있다. Using the sample processing method and information providing method of the present invention, cancer can be diagnosed with high accuracy.
본 발명의 시료 처리 방법 및 정보 제공 방법을 이용하여 소량의 시료로부터 매우 짧은 시간 내에 암을 진단할 수 있다. Using the sample processing method and information providing method of the present invention, cancer can be diagnosed from a small amount of sample in a very short period of time.
본 발명의 진단을 위한 정보 제공 방법은 기존의 진단 방법 대비 높은 민감도로 암을 진단할 수 있다.The information providing method for diagnosis of the present invention can diagnose cancer with higher sensitivity compared to existing diagnostic methods.
본 발명의 면역관문억제제에 대한 반응성 예측을 위한 정보 제공 방법은, 특정한 인산화된 pSTAT3 발현량에서 면역관문억제제에 대한 반응성이 가장 좋을 것임을 예측할 수 있다.The method for providing information for predicting responsiveness to an immune checkpoint inhibitor of the present invention can predict that responsiveness to an immune checkpoint inhibitor will be best at a specific phosphorylated pSTAT3 expression level.
도 1a 및 1b는 유세포 분석기를 이용하여 확인했을 때, 건강한 사람의 혈액 내 세포에서 pSTAT3의 발현이 거의 없음을 확인한 것이다. (pSTAT3는 인산화된 STAT3를 의미한다.)Figures 1a and 1b show that there is almost no expression of pSTAT3 in blood cells of healthy people when confirmed using a flow cytometer. (pSTAT3 means phosphorylated STAT3.)
도 1c 내지 1e는 STAT3의 인산화를 유도하는 사이토카인 중 하나인 IL-6에 의해 발현되는 pSTAT3의 경우, 시간이 지남에 따라 급격하게 그 발현이 감소하는 것을 확인한 것이다.Figures 1c to 1e show that in the case of pSTAT3, which is expressed by IL-6, one of the cytokines that induces phosphorylation of STAT3, the expression decreases rapidly over time.
도 2a 및 2b는 유세포 분석기를 이용하여 건강한 사람들과 비소세포폐암 환자들의 혈액 내 세포에서 무 자극 기저상태의 pSTAT3 발현을 확인한 것으로, 건강한 사람들에게서는 무 자극 기저상태의 pSTAT3가 거의 발현되지 않았으며, 비소세포폐암환자들 중 일부(patient 4)는 높게 발현되고, 또 다른 일부(patient 1)는 건강한 사람과 비슷하게 발현된 것을 확인한 것이다.Figures 2a and 2b show the expression of pSTAT3 in the unstimulated basal state in blood cells from healthy people and non-small cell lung cancer patients using flow cytometry. In healthy people, pSTAT3 was hardly expressed in the unstimulated basal state, some of the non-small cell lung cancer patients (patient 4) had high expression, and others (patient 1) had expression similar to that of healthy people.
도 2c는 비소세포폐암 환자로부터 분리된 혈액에서 말초혈액단핵구세포(peripheral blood mononuclear cell, PBMC)를 분리하기 전에 상온에 방치된 시간을 다르게 한 실험의 모식도를 나타낸 것이다. Figure 2c is a schematic diagram of an experiment in which peripheral blood mononuclear cells (PBMCs) were separated from blood from non-small cell lung cancer patients and left at room temperature for different periods of time before isolation.
도 2d 및 2e는 혈액이 개체로부터 분리된 후 상온에서 방치된 시간이 길어질수록 무 자극 기저상태의 pSTAT3의 발현량이 급격하게 감소한 것을 확인한 것이다. Figures 2d and 2e confirm that the expression level of pSTAT3 in the unstimulated basal state rapidly decreases as the time left at room temperature after blood is separated from the subject increases.
도 3a는 혈액 채취 후 말초혈액단핵구세포를 분리하기 전 상온에 방치된 시간을 1시간 이내로 하여 확인해본 결과, 건강한 사람과 비교하였을 때 모든 비소세포폐암 환자들에서 무 자극 기저상태의 pSTAT3 발현을 확인해볼 수 있었다.Figure 3a shows the results of examining the time left at room temperature for less than 1 hour before isolating peripheral blood mononuclear cells after blood collection, and confirming that pSTAT3 expression in the unstimulated basal state was observed in all non-small cell lung cancer patients compared to healthy people.
도 3b 내지 3f는 비소세포폐암 환자들의 무 자극 기저상태의 pSTAT3 발현이 암의 단계, 종양 조직학, 전이 및 EGFR 변이와는 무관하며 일정하게 나타나는 것을 확인한 것이다. 특히 암의 1기(stage 1)부터 건강한 사람과 암 환자 간 무 자극 기저 상태의 pSTAT3 발현 정도가 확연하게 차이나는 것을 확인하였다.Figures 3b to 3f show that the unstimulated basal pSTAT3 expression in non-small cell lung cancer patients is constant and independent of cancer stage, tumor histology, metastasis, and EGFR mutation. In particular, it was confirmed that the level of unstimulated basal pSTAT3 expression differs significantly between healthy people and cancer patients from
도 4a는 비소세포폐암 환자들의 면역세포들 중 어떤 세포들이 무 자극 기저상태의 pSTAT3을 발현하는지 확인해본 것으로, B cell, NK cell 및 Monocyte에서는 거의 발현되지 않고, CD4 T cell 및 CD8 T cell에서 무 자극 기저상태의 pSTAT3을 발현되는 것을 확인하였다.Figure 4a shows which cells among the immune cells of non-small cell lung cancer patients express pSTAT3 in the unstimulated basal state. It was confirmed that pSTAT3 is hardly expressed in B cells, NK cells, and monocytes, but is expressed in the unstimulated basal state in CD4 T cells and CD8 T cells.
도 4b 및 4c는 CD4 T cell 및 CD8 T cell 모두에서 CD27+CD45RA+인 그룹이 무 자극 기저상태의 pSTAT3의 발현이 가장 높음을 확인한 결과이다.Figures 4b and 4c show the results confirming that the CD27+CD45RA+ group had the highest expression of pSTAT3 in the unstimulated basal state in both CD4 T cells and CD8 T cells.
도 5a 및 5b는 암 환자에서 검출되는 무 자극 기저상태의 pSTAT3이 암으로 인한 것인지, 그렇다면 암 수술 후 무 자극 기저상태의 pSTAT3가 감소 혹은 없어지는지를 확인하기 위해, 비소세포폐암 환자들의 수술 전, 후에 무 자극 기저상태의 pSTAT3 발현을 확인한 것이다. 수술 이후 혈중 전체 면역 세포군에서 무 자극 기저상태의 pSTAT3 발현이 감소되는 것을 확인할 수 있었다.Figures 5a and 5b show the expression of pSTAT3 in the unstimulated basal state before and after surgery in non-small cell lung cancer patients to determine whether the unstimulated basal pSTAT3 detected in cancer patients is due to cancer, and if so, whether the unstimulated basal pSTAT3 decreases or disappears after cancer surgery. It was confirmed that the expression of pSTAT3 in the unstimulated basal state decreases in the entire immune cell population in the blood after surgery.
도 5c 및 5d는 비소세포폐암 환자들의 수술 전, 후의 무 자극 기저상태의 pSTAT3 발현을 CD27+CD45RA+ CD4 T Cells에서 확인한 것이다.Figures 5c and 5d show the expression of pSTAT3 in CD27+CD45RA+ CD4 T cells in the unstimulated baseline before and after surgery in patients with non-small cell lung cancer.
도 6a는 다양한 암 종에서 전체 말초혈액 세포에서의 무 자극 기저상태의 pSTAT3를 확인한 것이다.Figure 6a shows the unstimulated basal state of pSTAT3 in whole peripheral blood cells from various cancer types.
도 6b는 다양한 암 종에서 CD27+CD45RA+ CD4 T cells에서의 무 자극 기저상태의 pSTAT3을 확인한 것이다.Figure 6b shows pSTAT3 in the unstimulated basal state in CD27+CD45RA+ CD4 T cells in various cancer types.
도 6c는 상기 도 6a의 결과를 수치화 하여 나타낸 것이다.Figure 6c is a numerical representation of the results of Figure 6a.
도 6d는 상기 도 6b의 결과를 수치화 하여 나타낸 것이다.Figure 6d is a numerical representation of the results of Figure 6b.
도 7a는 여러 종류의 사이토카인(10ng/ml)을 사용하여 건강한 사람의 혈중 면역세포를 자극시킨 후 pSTAT3 발현을 확인한 결과이다.Figure 7a shows the results of confirming pSTAT3 expression after stimulating blood immune cells of a healthy person using various types of cytokines (10 ng/ml).
도 7b는 환자의 무 자극 기저상태의 pSTAT3 및 건강한 사람의 사이토카인(IL-6, IL-21, IFN-β, IL-10 또는 자극 없음(NS); 0.5 ng/ml)으로 유도된 pSTAT3를 다양한 면역 세포에서 확인한 결과이다.Figure 7b shows the results of confirming pSTAT3 in various immune cells in the unstimulated basal state of the patient and pSTAT3 induced by cytokines (IL-6, IL-21, IFN-β, IL-10 or no stimulation (NS); 0.5 ng/ml) of a healthy person.
도 7c는 도7b에서 얻은 값을 기반으로 주성분 분석을 한 결과이다.Figure 7c is the result of principal component analysis based on the values obtained in Figure 7b.
도 7d 내지 7f는 건강한 사람의 PBMC에 IL-6(0.5 ng/ml)로 자극을 준 후, 각 면역 세포에서 발현하는 pSTAT3를 확인한 결과이다. 도 7d는 Total PBMC에서 각 면역 세포의 pSTAT3 발현, 도 7e 및 7f는 각각 CD4 T cell 및 CD8 T cell에서 pSTAT3 발현을 확인한 것이다.Figures 7d to 7f show the results of confirming pSTAT3 expression in each immune cell after stimulating PBMCs of healthy people with IL-6 (0.5 ng/ml). Figure 7d shows pSTAT3 expression in each immune cell in total PBMCs, and Figures 7e and 7f show pSTAT3 expression in CD4 T cells and CD8 T cells, respectively.
도 8a는 비소세포폐암(NSCLC) 환자의 PBMC와 혈청을 통해 무 자극 기저상태의 pSTAT3 발현을 유도하는 인자를 찾는 실험 방법을 모식도로 나타낸 것이다.Figure 8a is a schematic diagram illustrating an experimental method for finding factors that induce pSTAT3 expression in the unstimulated basal state using PBMCs and serum from non-small cell lung cancer (NSCLC) patients.
도 8b는 NSCLC 환자의 혈청에서 BD Cytometric Bead Array를 사용해 염증성 사이토카인(IL-1β, IL-6, IL-8, IL-10, IL-12p70, TNF)의 농도를 측정한 결과이다.Figure 8b shows the results of measuring the concentration of inflammatory cytokines (IL-1β, IL-6, IL-8, IL-10, IL-12p70, TNF) in the serum of NSCLC patients using BD Cytometric Bead Array.
도 8c 내지 8e는 NSCLC 환자의 혈청과 건강한 사람의 PBMC를 공동 배양하고 1시간 후의 pSTAT3 발현을 측정한 결과이다. 도 8c는 Total PBMC에서 각 면역 세포의 pSTAT3 발현, 도 8d 및 8e는 각각 CD4 T cell 및 CD8 T cell에서 pSTAT3 발현을 확인한 것이다. 혈청 유도 pSTAT3는 CD4 T cell 및 CD8 T cell에서 확인 가능했으며 CD27+CD45RA+ 그룹에서 가장 높았다.Figures 8c to 8e show the results of co-culturing the serum of NSCLC patients with PBMCs of healthy people and measuring the expression of pSTAT3 after 1 hour. Figure 8c shows the expression of pSTAT3 in each immune cell in total PBMCs, and Figures 8d and 8e show the expression of pSTAT3 in CD4 T cells and CD8 T cells, respectively. Serum-induced pSTAT3 was confirmed in CD4 T cells and CD8 T cells, and was the highest in the CD27+CD45RA+ group.
도 8f는 NSCLC 환자의 혈청 내 IL-6 농도와, 동일한 혈청을 건강한 사람의 PBMC와 공동 배양한 후 측정한 CD27+CD45RA+ CD4 T cell(CD4 Tn)의 혈청 유도 pSTAT3 간 비례 관계를 나타낸 것이다.Figure 8f shows the proportional relationship between the serum IL-6 concentration of NSCLC patients and the serum-induced pSTAT3 of CD27+CD45RA+ CD4 T cells (CD4 Tn) measured after co-culture of the same serum with PBMCs from healthy people.
도 8g 및 8h는 NSCLC 환자의 혈청과 건강한 사람의 PBMC를 공동 배양할 때, anti-IL-6 또는 anti-IL-6R 항체를 처리하여 IL-6 신호전달을 방해한 경우, 혈청 유도 pSTAT3가 전혀 검출되지 않는 것을 보여준 결과이다.Figures 8g and 8h show that when serum from NSCLC patients was co-cultured with PBMCs from healthy people, serum-induced pSTAT3 was not detected at all when IL-6 signaling was blocked by treatment with anti-IL-6 or anti-IL-6R antibodies.
도 8i는 NSCLC 환자의 PBMC에서 얻은 CD4 Tn의 무 자극 기저상태의 pSTAT3 발현과, 앞서 얻은 CD4 Tn의 혈청 유도 pSTAT3과의 비례 관계를 나타낸 결과이다.Figure 8i shows the proportional relationship between the unstimulated basal pSTAT3 expression of CD4 Tn obtained from PBMCs of NSCLC patients and the serum-induced pSTAT3 of CD4 Tn obtained previously.
도 8j는 NSCLC 환자의 PBMC에서 얻은 CD4 Tn의 무 자극 기저상태의 pSTAT3 발현과, 앞서 얻은 혈청 내 IL-6 농도와의 비례 관계를 나타낸 결과이다.Figure 8j shows the proportional relationship between the unstimulated basal pSTAT3 expression of CD4 Tn obtained from PBMCs of NSCLC patients and the serum IL-6 concentration obtained previously.
도 8k는 NSCLC 환자를 무 자극 기저상태의 pSTAT3 발현 정도 따라 pSTAT3ex vivo-lo, pSTAT3ex vivo-int 및 pSTAT3ex vivo-hi로 나누었을 때, 각 그룹의 무 자극 기저상태의 pSTAT3 발현량을 나타낸 결과이다.Figure 8k shows the results of dividing NSCLC patients into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to the level of pSTAT3 expression in the unstimulated basal state in each group.
도 8l는 NSCLC 환자를 무 자극 기저상태의 pSTAT3 발현 정도에 따라 pSTAT3ex vivo-lo, pSTAT3ex vivo-int 및 pSTAT3ex vivo-hi로 나누었을 때, 각 그룹의 혈청 내 IL-6 농도를 나타낸 결과이다.Figure 8l shows the results of dividing NSCLC patients into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to the level of pSTAT3 expression in the unstimulated basal state, and showing the serum IL-6 concentrations in each group.
도 9a는 NSCLC 환자의 PBMC에서 분석한 CD27+CD45RA+ CD4 T cell(CD4 Tn)의 무 자극 기저상태의 pSTAT3의 발현과, TIL에서 분석한 TAM(CD14+CD11b+HLA-DR+CD11c+)의 CD206 발현 간 상관관계를 나타낸 결과이다. Figure 9a shows the correlation between the expression of pSTAT3 in the unstimulated basal state of CD27+CD45RA+ CD4 T cells (CD4 Tn) analyzed in PBMCs of NSCLC patients and the expression of CD206 in TAMs (CD14+CD11b+HLA-DR+CD11c+) analyzed in TILs.
도 9b 및 9c는 NSCLC 환자를 무 자극 기저상태의 pSTAT3의 발현정도에 따라 pSTAT3ex vivo-lo, pSTAT3ex vivo-int 및 pSTAT3ex vivo-hi로 나누었을 때, 각 그룹에서 TAM 내 CD206 발현 정도를 비교한 결과이다.Figures 9b and 9c show the results of comparing the expression levels of CD206 in TAMs in each group when NSCLC patients were divided into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to the expression levels of pSTAT3 in the unstimulated basal state.
도 9d는 NSCLC 환자의 PBMC에서 분석한 CD27+CD45RA+ CD4 T cell(CD4 Tn)의 무 자극 기저상태의 pSTAT3의 발현과, CD8+ TIL 내 CD103+CD39+ cell의 비율 간 상관관계를 나타낸 결과이다.Figure 9d shows the correlation between the expression of pSTAT3 in the unstimulated basal state of CD27+CD45RA+ CD4 T cells (CD4 Tn) analyzed in PBMCs of NSCLC patients and the proportion of CD103+CD39+ cells in CD8+ TIL.
도 9e 및 9f는 pSTAT3ex vivo-lo, pSTAT3ex vivo-int, 및 pSTAT3ex vivo-hi 환자군의 CD8+ TIL 내 CD103+CD39+ cell의 비율을 비교한 결과이다.Figures 9e and 9f show the results comparing the ratio of CD103+CD39+ cells in CD8+ TILs in the pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi patient groups.
도 10a는 Pembrolizumab 단독요법 또는 병용요법(Pemetrexed + cisplatin)을 항암 1차 치료로 받은 4기 NSCLC 환자들을 대상으로, CD27+CD45RA+ CD4 T cell(CD4 Tn)의 무 자극 기저상태의 pSTAT3 발현 정도에 따라 pSTAT3ex vivo-lo, pSTAT3ex vivo-int, 및 pSTAT3ex vivo-hi로 나누었을 때, 각 그룹의 무 자극 기저상태의 pSTAT3 발현을 나타낸 결과이다.Figure 10a shows the results of dividing stage IV NSCLC patients who received pembrolizumab monotherapy or combination therapy (pemetrexed + cisplatin) as first-line chemotherapy into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to the level of pSTAT3 expression in the unstimulated basal state of CD27+CD45RA+ CD4 T cells (CD4 Tn).
도 10b는, pSTAT3ex vivo-lo와 pSTAT3ex vivo-int를 하나의 그룹으로 하였을 때 각 환자군에서 면역치료 3개월 후의 반응을 비교한 것이다. PD는 진행 병변(progressive disease), SD는 안정 병변(stable disease), PR은 부분 관해(partial response)를 의미한다.Figure 10b compares the response after 3 months of immunotherapy in each patient group when pSTAT3 ex vivo -lo and pSTAT3 ex vivo -int were grouped together. PD indicates progressive disease, SD indicates stable disease, and PR indicates partial response.
도 10c는 pSTAT3ex vivo-lo와 pSTAT3ex vivo-int를 별도 환자군으로 나누었을 때, pSTAT3ex vivo-lo, pSTAT3ex vivo-int 및 pSTAT3ex vivo-hi의 면역치료 3개월 후 반응을 비교한 것이다. PD는 진행 병변(progressive disease), SD는 안정 병변(stable disease), PR은 부분 관해(partial response)를 의미한다.Figure 10c shows a comparison of the responses after 3 months of immunotherapy of pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi when pSTAT3 ex vivo -lo and pSTAT3 ex vivo -int were divided into separate patient groups. PD indicates progressive disease, SD indicates stable disease, and PR indicates partial response.
이하, 본 발명을 상세하게 설명한다. Hereinafter, the present invention will be described in detail.
본 발명은 개체로부터 분리된 시료에 고정 시약을 처리하여 상기 시료의 세포 내에 존재하는 인산화된 STAT3를 고정시키는 단계; 상기 세포의 세포막을 천공하는 단계; 및 상기 고정된 인산화된 STAT3를 검출하는 단계를 포함하는, 암의 조기 진단 또는 치료 후 재발 여부 진단을 위한 시료의 처리 방법을 제공한다.The present invention provides a method for processing a sample for early diagnosis of cancer or diagnosis of recurrence after treatment, comprising the steps of treating a sample separated from an individual with a fixing reagent to fix phosphorylated STAT3 present in cells of the sample; the step of perforating the cell membrane of the cells; and the step of detecting the fixed phosphorylated STAT3.
개체는 암이 발병될 수 있는 동물, 그 외 암의 진단을 위한 정보를 제공받고자 하는 동물을 의미한다. 상기 동물은 포유류일 수 있고, 바람직하게는 개, 고양이 또는 인간일 수 있으나, 이에 제한되는 것은 아니다. The subject means an animal that can develop cancer, or an animal from which information for diagnosing cancer is to be provided. The animal may be a mammal, and preferably may be a dog, cat, or human, but is not limited thereto.
시료는 개체의 모든 체액 또는 조직에서 채취한 생체 시료일 수 있으며, 이는 혈액, 타액, 소변, 조직, 세포, 림프, 골수 액, 척수액, 뇌추출액, 관절액 또는 세포 조직액일 수 있다. 바람직하게 시료는 혈액일 수 있다. The sample may be a biological sample taken from any body fluid or tissue of the subject, such as blood, saliva, urine, tissue, cells, lymph, bone marrow fluid, spinal fluid, brain extract, synovial fluid or tissue fluid. Preferably, the sample may be blood.
혈액은 무처리 혈액으로, 용어 "무처리 혈액"은 사이토카인 등 별도의 자극인자를 처리하지 않은, 환자로부터 채취한 그대로의 혈액이다. 즉, 본 발명의 시료의 처리 방법은 개체로부터 분리된 혈액에 아무 자극인자도 처리도 하지 않은 상태에서 인산화된 STAT3를 검출하는 것이다. 상기 혈액은 바람직하게는 인간 말초 혈액일 수 있으나, 이에 제한되는 것은 아니다.Blood is unprocessed blood, and the term "unprocessed blood" refers to blood collected from a patient without being treated with any additional stimulating factors such as cytokines. That is, the method for processing a sample of the present invention detects phosphorylated STAT3 in blood separated from an individual without being treated with any stimulating factors. The blood may preferably be human peripheral blood, but is not limited thereto.
고정 시약은 단백질의 인산화 상태를 고정시킬 수 있는 모든 시약을 의미하는 것으로, 포름알데히드 또는 파라포름알데히드일 수 있으며, 이는 BD CytofixTM Fixation Buffer, eBioscienceTM IC Fixation Buffer 등의 제품을 이용할 수 있으나, 이에 제한되는 것은 아니다. Fixation reagent refers to any reagent capable of fixing the phosphorylation state of a protein, and may be formaldehyde or paraformaldehyde. Products available include, but are not limited to, BD Cytofix TM Fixation Buffer and eBioscience TM IC Fixation Buffer.
인산화된 STAT3(pSTAT3)는 STAT3 단백질(Signal Transducer and activator of transcription 3)에 인산기가 붙어있는 상태를 말하며, 구체적으로는 STAT3 단백질의 타이로신 잔기에 인산기가 붙어있는 상태를 의미한다.Phosphorylated STAT3 (pSTAT3) refers to a state in which a phosphate group is attached to the STAT3 protein (Signal Transducer and activator of transcription 3), specifically, it refers to a state in which a phosphate group is attached to a tyrosine residue of the STAT3 protein.
본 명세서에서 사용되는 용어 "무 자극 기저 상태의 pSTAT3"는 사이토카인 등의 별도의 자극인자를 처리하지 않는, 본 발명의 혈액 처리 방법을 이용하여 검출되는 pSTAT3를 의미한다. 용어 "무 자극 기저상태의 pSTAT3"는 "ex-vivo pSTAT3"와 상호교환적으로 사용될 수 있다. The term "pSTAT3 in unstimulated basal state" as used herein refers to pSTAT3 detected using the blood processing method of the present invention without treating a separate stimulating factor such as a cytokine. The term "pSTAT3 in unstimulated basal state" can be used interchangeably with "ex-vivo pSTAT3".
세포막을 천공하는 단계는 세포 내의 STAT3를 검출하기 위해 세포에 투과성을 부여하는 단계이다. 투과성을 부과하는 단계는 당업계에 공지된 모든 방법을 사용할 수 있으며, 구체적으로는 투과성 부과(permeabilization) 시약을 처리하는 것일 수 있다. 투과성 부과 시약은 세포 내부의 인산화된 STAT3의 검출을 용이하게 할 수 있도록 도와주는 시약을 의미하며, 이는 메탄올, 에탄올일 수 있으며, BD Permeabilizing solution 3를 이용할 수 있으나, 이에 제한되는 것은 아니다.The step of perforating the cell membrane is a step of permeabilizing the cell to detect STAT3 within the cell. The step of imposing permeability may use any method known in the art, and specifically may be treating a permeabilization reagent. The permeabilization reagent refers to a reagent that helps facilitate the detection of phosphorylated STAT3 within the cell, and may be methanol, ethanol, and
검출은 상기 인산화된 STAT3 단백질을 확인할 수 있는 당 업계의 공지된 모든 방법을 사용할 수 있다. 예를 들어, 이는 항체를 이용해서 검출하는 것, 단백질 염색법을 이용해서 검출하는 것, 질량 분석을 통해 검출하는 것 등일 수 있다. 예를 들어, 검출은 유세포 분석, ELISA, 웨스턴 블롯, 면역침전분석법, 보체 고정분석법을 이용하는 것일 수 있으며, ELISA 키트 또는 웨스턴 블롯, 면역침전분석법, 보체 고정법, 또는 유세포분석 등을 구현하기 위한 키트를 이용하는 것일 수 있다. Detection can be performed using any method known in the art that can identify the phosphorylated STAT3 protein. For example, detection can be performed using antibodies, detection can be performed using protein staining, detection can be performed using mass spectrometry, etc. For example, detection can be performed using flow cytometry, ELISA, Western blot, immunoprecipitation assay, complement fixation assay, and can be performed using an ELISA kit or a kit for implementing Western blot, immunoprecipitation assay, complement fixation assay, or flow cytometry.
암은 바람직하게는 폐암, 담관암, 방광암, 신장암, 간암, 위암, 및 요로암으로 이루어지는 군에서 선택되는 어느 하나일 수 있으나, 이에 제한되지는 않는다. The cancer may preferably be any one selected from the group consisting of lung cancer, bile duct cancer, bladder cancer, kidney cancer, liver cancer, stomach cancer, and urinary tract cancer, but is not limited thereto.
조기 진단은 병기가 많이 진행되지 않은 상태에서 병 상태를 판단하는 것으로, 암의 조기 진단의 경우, 암이 병기가 낮은 상태의 암을 진단하는 것일 수 있다. 예를 들어, 병기 1기의 암을 진단하는 것일 수 있다.Early diagnosis is the diagnosis of a disease condition when the disease has not progressed much. In the case of early diagnosis of cancer, it may be the diagnosis of cancer at a low stage. For example, it may be the diagnosis of cancer at
치료 후 재발 여부 진단은 암의 수술 또는 치료 후에 다시 암이 재발하는지 진단 또는 모니터링하는 것을 의미할 수 있다. Diagnosing recurrence after treatment can mean diagnosing or monitoring whether cancer comes back after surgery or treatment.
고정 시약은 상기 혈액의 인산화된 STAT3가 탈인산화되기 전에 처리되는 것일 수 있다. 상기 탈인산화(dephosphorylation)는 유기화합물로부터 인산기가 가수분해로 제거되는 것이며, STAT3 단백질에 붙어있던 인산기가 제거되는 것일 수 있다. 본 발명자는 개체로부터 분리된 혈액에서 인산화된 STAT3가 있는 경우, 일정 시간이 지나면 상기 인산화된 STAT3의 탈인산화가 일어남을 확인한 바, 본 발명의 혈액의 처리 방법은 상기 탈인산화가 일어나기 전에 고정 시약으로 인산화된 STAT3를 고정하여 이를 검출하는 것이다. The fixing reagent may be treated before the phosphorylated STAT3 in the blood is dephosphorylated. The dephosphorylation is the removal of a phosphate group from an organic compound by hydrolysis, and may be the removal of a phosphate group attached to a STAT3 protein. The inventors of the present invention have confirmed that when phosphorylated STAT3 is present in blood separated from an individual, dephosphorylation of the phosphorylated STAT3 occurs after a certain period of time. Accordingly, the method for treating blood of the present invention fixes phosphorylated STAT3 with a fixing reagent before the dephosphorylation occurs, and detects it.
본 발명은 개체로부터 분리된 시료에 고정 시약을 처리하여 상기 시료의 세포 내에 존재하는 인산화된 STAT3를 고정시키는 단계; 상기 세포의 세포막을 천공하는 단계; 상기 고정된 인산화된 STAT3를 검출하는 단계; 및 상기 검출된 인산화된 STAT3에 대한 정보를 기초로 암 환자와 정상인을 구분하는 단계를 포함하는, 암의 조기 진단 또는 치료 후 재발 여부 진단을 위한 정보제공 방법을 제공할 수 있다.The present invention can provide a method for providing information for early diagnosis of cancer or diagnosis of recurrence after treatment, comprising the steps of treating a fixing reagent on a sample separated from an individual to fix phosphorylated STAT3 present in cells of the sample; the step of perforating the cell membrane of the cell; the step of detecting the fixed phosphorylated STAT3; and the step of distinguishing between cancer patients and normal people based on information about the detected phosphorylated STAT3.
상기 개체, 시료, 혈액, 고정 시약, 인산화된 STAT3, 검출, 암, 조기진단, 치료 후 재발 여부 진단에 대해서는 상기 설명한 바와 같다.The above objects, samples, blood, fixed reagents, phosphorylated STAT3, detection, cancer, early diagnosis, and diagnosis of recurrence after treatment are as described above.
본 발명은 개체로부터 분리된 혈액의 pSTAT3을 검출함으로써, 암 환자의 경우 pSTAT3가 검출되는 반면, 정상인에게서는 pSTAT3가 거의 검출되지 않아, pSTAT3가 검출되면 암 환자라고 조기에 진단할 수 있는 정보를 제공할 수 있다. The present invention detects pSTAT3 in blood separated from an individual, and since pSTAT3 is detected in cancer patients, while pSTAT3 is hardly detected in normal people, when pSTAT3 is detected, information can be provided that allows for early diagnosis of a cancer patient.
본 발명은 개체로부터 분리된 시료의 고정된 인산화된 STAT3의 발현량을 측정하는 단계를 포함하는 면역관문억제제에 대한 반응성 예측을 위한 정보 제공 방법을 제공한다.The present invention provides a method for providing information for predicting responsiveness to an immune checkpoint inhibitor, comprising the step of measuring the expression level of fixed phosphorylated STAT3 in a sample isolated from an individual.
본 발명은 상기 개체로부터 분리된 시료는 무처리 혈액인 면역관문억제제에 대한 반응성 예측을 위한 정보 제공 방법을 제공한다.The present invention provides a method for providing information for predicting responsiveness to an immune checkpoint inhibitor, wherein the sample separated from the subject is untreated blood.
본 발명은 상기 측정된 인산화된 STAT3의 발현량이 20 내지 60%이면 대조군 대비 면역관문억제제에 대한 반응성이 좋을 것으로 예측하는 단계를 더 포함하는 면역관문억제제에 대한 반응성 예측을 위한 정보 제공 방법을 제공한다.The present invention provides a method for providing information for predicting responsiveness to an immune checkpoint inhibitor, further comprising a step of predicting that responsiveness to an immune checkpoint inhibitor will be good compared to a control group if the expression level of the measured phosphorylated STAT3 is 20 to 60%.
상기 인산화된 STAT3의 발현량은 시료 내 CD4 T cell에서의 발현량을 의미하는 것으로, 시료 내 전체 CD4 T cell 중 인산화된 STAT3를 발현하는 CD4 T cell의 비율을 의미한다.The expression level of the above phosphorylated STAT3 refers to the expression level in CD4 T cells in the sample, and represents the ratio of CD4 T cells expressing phosphorylated STAT3 among the total CD4 T cells in the sample.
상기 인산화된 STAT3의 발현량은 시료 내 CD27+CD45RA+ CD4 T cell에서의 발현량을 의미하는 것으로, 시료 내 전체 CD27+CD45RA+ CD4 T cell 중 인산화된 STAT3를 발현하는 CD27+CD45RA+ CD4 T cell의 비율을 의미한다. 바람직하게는 혈액 내 CD27+CD45RA+ CD4 T cell 중 인산화된 STAT3를 발현하는 CD27+CD45RA+ CD4 T cell의 비율일 수 있으나, 이에 제한되는 것은 아니다.The above-mentioned expression amount of phosphorylated STAT3 refers to the expression amount in CD27+CD45RA+ CD4 T cells in the sample, and refers to the ratio of CD27+CD45RA+ CD4 T cells expressing phosphorylated STAT3 among the total CD27+CD45RA+ CD4 T cells in the sample. Preferably, it may be the ratio of CD27+CD45RA+ CD4 T cells expressing phosphorylated STAT3 among CD27+CD45RA+ CD4 T cells in blood, but is not limited thereto.
본 발명의 면역관문억제제에 대한 반응성 예측을 위한 정보 제공 방법에서, 대조군은 예를 들어 상기 측정된 인산화된 STAT3의 발현량이 20% 미만인 그룹 또는 60% 초과인 그룹일 수 있다.In the method for providing information for predicting responsiveness to an immune checkpoint inhibitor of the present invention, the control group may be, for example, a group in which the measured expression level of phosphorylated STAT3 is less than 20% or a group in which the expression level is greater than 60%.
본원 명세서에서 면역관문억제제는 항-PD-1 항체, 항-PD-L1 항체, 항-CTLA4 항체, 항 PD-L2 항체, LTF2 조절 항체, 항-LAG3 항체, 항-A2aR 항체, 항-TIGIT 항체, 항-TIM-3 항체, 항-B7-H3 항체, 항-B7-H4 항체, 항-VISTA 항체, 항-CD47 항체, 항-BTLA 항체, 항-KIR 항체, 항-IDO 항체, 시스플라틴(cisplatin) 및 이의 조합으로 이루어진 군으로부터 선택되는 어느 하나일 수 있다.In the present specification, the immune checkpoint inhibitor may be any one selected from the group consisting of anti-PD-1 antibody, anti-PD-L1 antibody, anti-CTLA4 antibody, anti-PD-L2 antibody, LTF2 regulatory antibody, anti-LAG3 antibody, anti-A2aR antibody, anti-TIGIT antibody, anti-TIM-3 antibody, anti-B7-H3 antibody, anti-B7-H4 antibody, anti-VISTA antibody, anti-CD47 antibody, anti-BTLA antibody, anti-KIR antibody, anti-IDO antibody, cisplatin, and combinations thereof.
일 실시예에서, 면역관문억제제는 펨브롤리주맙(Pembrolizumab) 또는 펨브롤리주맙(Pembrolizumab) 및 시스플라틴(Cisplatin)의 조합이다.In one embodiment, the immune checkpoint inhibitor is Pembrolizumab or a combination of Pembrolizumab and Cisplatin.
본 발명은 상기 고정된 인산화된 STAT3는 상기 시료에 고정 시약을 처리하여 상기 시료의 세포 내에 존재하는 인산화된 STAT3를 고정시켜 얻어진 면역관문억제제에 대한 반응성 예측을 위한 정보 제공 방법을 제공한다.The present invention provides a method for providing information for predicting responsiveness to an immune checkpoint inhibitor, wherein the fixed phosphorylated STAT3 is obtained by treating the sample with a fixing reagent to fix phosphorylated STAT3 present in the cells of the sample.
개체, 시료, 혈액, 고정 시약, 인산화된 STAT3, 검출에 대해서는 위에서 설명한 바와 같다.The subjects, samples, blood, fixatives, phosphorylated STAT3, and detection were as described above.
이하, 본 발명을 구체적으로 설명하기 위해 실시예를 들어 상세하게 설명하기로 한다. Hereinafter, the present invention will be described in detail by way of examples in order to specifically explain the present invention.
실시예 1: pSTAT3 검출Example 1: Detection of pSTAT3
자극인자가 처리되지 않은 무처리 PBMC 또는 사이토카인이 처리된 PBMC를 고정 시약(BD IC fixation buffer)를 사용하여 4℃에서 20 분간 고정했다. 그 후 99.8 % 메탄올을 사용하여 -20℃에서 30 분간 투과성 부여를 한 뒤, 필요한 형광이 부착된 항체(fluorescent conjugated antibodies)를 사용하여 염색한다. 염색된 샘플은 유세포 분석(flow cytometry)를 통하여 분석되었다. 본 발명의 모든 pSTAT3 분석은 위 방식을 통해 분석되었다. Untreated PBMCs or cytokine-treated PBMCs were fixed with fixation reagent (BD IC fixation buffer) at 4°C for 20 minutes. After permeabilization with 99.8% methanol at -20°C for 30 minutes, the samples were stained with the required fluorescent conjugated antibodies. The stained samples were analyzed by flow cytometry. All pSTAT3 assays of the present invention were analyzed by the above method.
실시예 2: 건강한 사람의 혈액 내 pSTAT3 발현 확인Example 2: Confirmation of pSTAT3 expression in the blood of healthy people
건강한 사람의 total PBMC에서 무 자극 기저상태의 pSTAT3를 유세포 분석으로 확인한 것이다. 대조군(isotype control)은 anti-STAT3 (pY705) 항체 대신 대응하는 isotype 항체를 사용하였다(도 1a). 여러 건강한 사람들의 total PBMC에서 무 자극 기저상태의 pSTAT3의 발현이 거의 없음을 확인하였다(도 1b). The expression of pSTAT3 in the unstimulated basal state was confirmed by flow cytometry in total PBMCs of healthy individuals. The control group (isotype control) used the corresponding isotype antibody instead of anti-STAT3 (pY705) antibody (Fig. 1a). The expression of pSTAT3 in the unstimulated basal state was almost absent in total PBMCs of several healthy individuals (Fig. 1b).
실시예 3: IL-6에 의한 pSTAT3의 발현 확인Example 3: Confirmation of pSTAT3 expression by IL-6
-80℃ PBMC를 37℃에서 급속 해동한다. 해동된 PBMC 중 일부는 "After thaw" 샘플로써 바로 고정 및 투과성 부여가 되었다. 나머지는 세척 과정 (Washing steps)을 통해 배양에 적합한 상태로 만든 후 37℃ CO2 incubator에서 IL-6 (0.2ng/ml)과 함께 배양되었다. 30분 뒤 일부는 "IL6 stimulation" 샘플로써 바로 고정 및 투과성 부여가 되었다. 나머지는 세척 과정을 겪은 후 37℃ CO2 incubator에서 다시 배양되었다. 그 후 30분, 1시간, 2시간 뒤마다 일부 샘플을 꺼내 각각 "30 min rest", "1h rest", 그리고 "2h rest" 샘플로써 고정 및 투과성 부여가 되었다. 이 모든 작업이 끝난 뒤 모든 샘플들은 pSTAT3 염색 및 분석을 진행하였다. 그 결과, STAT3의 인산화를 유도하는 사이토카인 중 하나인 IL-6에 의해 발현되는 pSTAT3의 경우, 시간이 지남에 따라 급격하게 그 발현이 감소하는 것을 확인하였다(도 1c 내지 도 1e).-80℃ PBMC were rapidly thawed at 37℃. Some of the thawed PBMC were immediately fixed and permeabilized as "after thaw" samples. The rest were made suitable for culture through washing steps and then cultured with IL-6 (0.2 ng/ml) in a 37℃ CO 2 incubator. After 30 minutes, some were immediately fixed and permeabilized as "IL6 stimulation" samples. The rest were washed and cultured again in a 37℃ CO 2 incubator. After 30 minutes, 1 hour, and 2 hours, some samples were taken out and fixed and permeabilized as "30 min rest", "1 h rest", and "2 h rest" samples, respectively. After all of these tasks, all samples were stained and analyzed with pSTAT3. As a result, in the case of pSTAT3, which is expressed by IL-6, one of the cytokines that induces phosphorylation of STAT3, it was confirmed that its expression rapidly decreased over time (Figures 1c to 1e).
실시예 4: 건강한 사람과 비소세포 폐암 환자 간의 무 자극 기저상태의 pSTAT3 발현 확인Example 4: Determination of pSTAT3 expression in unstimulated basal states between healthy individuals and non-small cell lung cancer patients
건강한 사람과 비소세포폐암(NSCLC) 환자의 total PBMC에서 유세포 분석을 이용하여 무 자극 기저상태의 pSTAT3를 확인하였다. 건강한 사람들에게서는 무 자극 기저상태의 pSTAT3가 거의 발현되지 않았으며, 비소세포폐암환자들 중 일부는 높게 발현되고, 또 다른 일부는 건강한 사람과 비슷하게 발현된 것을 확인하였다(도 2a, 2b). We used flow cytometry to determine the basal level of pSTAT3 in total PBMCs from healthy subjects and non-small cell lung cancer (NSCLC) patients. In healthy subjects, pSTAT3 was barely expressed in the basal state, while in some NSCLC patients, it was highly expressed and in others, it was expressed similarly to healthy subjects (Fig. 2a, 2b).
실시예 5: 혈액 채취 후 보관시간에 따른 pSTAT3의 발현량 확인Example 5: Confirmation of pSTAT3 expression level according to storage time after blood collection
비소세포폐암 환자로부터 분리된 혈액에서 말초혈액단핵구(PBMC)를 분리하기 전에 상온에 방치된 시간을 다르게 하여 실험을 진행했다(도 2c). NSCLC 환자의 혈액을 채혈한 뒤 상온에서 1시간을 방치하고 그 중 일부 (Stored 1h 샘플)에서 PBMC를 분리 및 보관하였다. 나머지는 2시간을 더 방치하고 그 중 일부(Stored 3h 샘플)에서 PBMC를 분리 및 보관하였다. 나머지는 2시간을 더 방치하고(Stored 5h 샘플) PBMC를 분리 및 보관하였다. 보관된 PBMC들은 동시에 pSTAT3 염색 및 분석이 진행되었다. 그 결과, 혈액이 개체로부터 분리된 후 상온에서 방치된 시간이 길어질수록 무 자극 기저상태의 pSTAT3의 발현량이 급격하게 감소하는 것을 확인했다(도 2d, 2e).The experiments were conducted by varying the time of storage at room temperature before isolating peripheral blood mononuclear cells (PBMCs) from blood isolated from patients with non-small cell lung cancer (Fig. 2c). After collecting blood from a patient with NSCLC, PBMCs were isolated and stored from some of them (Stored 1h samples) for 1 hour. The remainder were stored for another 2 hours, and PBMCs were isolated and stored from some of them (Stored 3h samples). The remainder were stored for another 2 hours (Stored 5h samples) and PBMCs were isolated and stored. The stored PBMCs were simultaneously stained and analyzed for pSTAT3. As a result, it was confirmed that the expression level of pSTAT3 in the unstimulated basal state decreased rapidly as the time of storage at room temperature after blood was separated from the subject increased (Figs. 2d, 2e).
실시예 6: 채취된 혈액의 상온 방치 시간을 1시간 이내로 한, 무 자극 기저상태의 pSTAT3의 발현 비교 Example 6: Comparison of pSTAT3 expression in unstimulated basal state, with blood collected at room temperature left for less than 1 hour
건강한 사람과 NSCLC 환자의 혈액을 채혈한지 1시간 이내에 PBMC를 분리하였고 total PBMC에서 무 자극 기저상태의 pSTAT3가 분석되었다. NSCLC환자들은 stage, tumor histology, metastasis, EGFR mutation에 따라 나뉘어져 분석되기도 하였다. 그 결과, 건강한 사람과 비교하였을 때, 모든 비소세포폐암 환자들에게서 무 자극 기저상태의 pSTAT3 발현을 확인할 수 있었다(도 3a). 또한, 비소세포폐암 환자들의 무 자극 기저상태의 pSTAT3 발현이 암의 단계, 종양 조직학, 전이, EGFR 변이와는 무관하고 일정하게 나타나는 것을 확인하였으며, 특히 1기(Stage 1)의 암환자부터 건강한 사람과 그 값이 확연하게 차이나는 것을 확인했다(도 3b 내지 3f).Within 1 hour of collecting blood from healthy people and NSCLC patients, PBMCs were isolated, and unstimulated basal pSTAT3 was analyzed in total PBMCs. NSCLC patients were also divided and analyzed according to stage, tumor histology, metastasis, and EGFR mutation. As a result, unstimulated basal pSTAT3 expression was confirmed in all NSCLC patients compared to healthy people (Fig. 3a). In addition, it was confirmed that the unstimulated basal pSTAT3 expression of NSCLC patients was constant regardless of cancer stage, tumor histology, metastasis, and EGFR mutation, and in particular, it was confirmed that the values were clearly different from those of healthy people starting from
실시예 7: 무 자극 기저상태의 pSTAT3를 발현하는 면역세포 확인Example 7: Identification of immune cells expressing pSTAT3 in the unstimulated basal state
NSCLC 환자의 혈액의 Monocyte(CD14+), T cells(CD3+), B cells(CD19+), NK cells(CD56+CD3-)에서 무 자극 기저상태의 pSTAT3를 분석하였다. 그 결과, B cell, NK cell, Monocyte에서는 거의 발현되지 않고, T cell들이 무 자극 기저상태의 pSTAT3을 발현하는 것을 확인하였다(도 4a).We analyzed pSTAT3 in the unstimulated basal state in Monocytes (CD14+), T cells (CD3+), B cells (CD19+), and NK cells (CD56+CD3-) of the blood of NSCLC patients. As a result, we confirmed that pSTAT3 was hardly expressed in B cells, NK cells, and Monocytes, and that T cells expressed pSTAT3 in the unstimulated basal state (Fig. 4a).
NSCLC 환자의 CD4 T cell과 CD8 T cell은 CD27과 CD45RA 존재에 따라 4가지 subset으로 분류하였고, 각 subset에서 무 자극 기저상태의 pSTAT3를 비교 분석하였다. 그 결과, CD4 T cell 및 CD8 T cell 모두에서 CD27+CD45RA+인 그룹이 무 자극 기저상태의 pSTAT3의 발현이 가장 높은 것을 확인하였다(도 4b, 4c).CD4 T cells and CD8 T cells of NSCLC patients were classified into four subsets according to the presence of CD27 and CD45RA, and pSTAT3 in the unstimulated basal state was compared and analyzed in each subset. As a result, it was confirmed that the CD27+CD45RA+ group had the highest expression of pSTAT3 in the unstimulated basal state in both CD4 T cells and CD8 T cells (Fig. 4b, 4c).
실시예 8: 비소세포폐암 환자들의 수술 전과 수술 후의 무 자극 기저상태의 pSTAT3 검출 확인Example 8: Confirmation of pSTAT3 detection in unstimulated baseline before and after surgery in patients with non-small cell lung cancer
암 환자에서 검출되는 무 자극 기저상태의 pSTAT3가 암으로 인한 것인지 그렇다면 암 수술 후 무 자극 기저상태의 pSTAT3가 감소 혹은 없어지는지 확인하기 위해, 비소세포폐암 환자들의 수술 전, 후의 무 자극 기저상태의 pSTAT3 발현을 확인하였다. To determine whether the unstimulated basal pSTAT3 detected in cancer patients is due to cancer and, if so, whether the unstimulated basal pSTAT3 is reduced or eliminated after cancer surgery, we examined the unstimulated basal pSTAT3 expression before and after surgery in non-small cell lung cancer patients.
외과적 수술을 통해 암의 제거가 예정된 환자들의 혈액(Before OP)을 채취하여 PBMC를 분리 및 보관하였다. 같은 환자들이 병원을 재 방문하였을 때(수술 후 2개월~8개월 사이; After OP), 혈액을 한번 더 채취하여 PBMC를 분리 및 보관하였다. 보관된 PBMC들은 한번에 녹여져 무 자극 기저상태의 pSTAT3가 분석되었다. Before OP에는 모든 환자들의 혈액이 포함되어 있으나 수술 후 혈액은 재 방문 시기에 따라 2~4개월 후, 4~6개월 후, 6~8개월 후로 나누기도 하였다. 그 결과, 수술 이후 혈중 전체 면역 세포군에서 무 자극 기저상태의 pSTAT3 발현이 감소되는 것을 확인하였으며(도 5a, 5b), CD27+CD45RA+ CD4 T cell에서도 무 자극 기저상태의 pSTAT3의 발현이 감소되는 것을 확인하였다(도 5c, 5d).Blood of patients scheduled to undergo surgical removal of cancer (Before OP) was collected, and PBMCs were isolated and stored. When the same patients revisited the hospital (between 2 and 8 months after surgery; After OP), blood was collected once more, and PBMCs were isolated and stored. The stored PBMCs were thawed all at once, and pSTAT3 in the unstimulated basal state was analyzed. Before OP included the blood of all patients, but postoperative blood was divided into 2 to 4 months, 4 to 6 months, and 6 to 8 months depending on the time of revisit. As a result, it was confirmed that the expression of pSTAT3 in the unstimulated basal state was reduced in the entire immune cell population in the blood after surgery (Fig. 5a, 5b), and it was also confirmed that the expression of pSTAT3 in the unstimulated basal state was reduced in CD27+CD45RA+ CD4 T cells (Fig. 5c, 5d).
실시예 9: 다양한 암 종에서 무 자극 기저상태의 pSTAT3 확인Example 9: Identification of pSTAT3 in unstimulated basal state in various cancer types
Lung(구체적으로 non-small cell lung cancer), Skin(구체적으로 melanoma), Bile duct(구체적으로 gallbladder), Bladder(구체적으로 bladder cancer), Kidney(구체적으로 renal cell carcinoma), Liver (구체적으로 hepatocellular carcinoma), Stomach(구체적으로 advanced gastric cancer), Urothelial(구체적으로 urothelial cancer)에 암이 걸린 환자들의 혈액에서 PBMC를 분리하여 무 자극 기저상태의 pSTAT3를 분석하였다. 또한, CD27+CD45RA+CD4 T cell에서도 다양한 암종에서의 무 자극 기저상태의 pSTAT3를 분석하였다.We isolated PBMCs from the blood of patients with lung (specifically non-small cell lung cancer), skin (specifically melanoma), bile duct (specifically gallbladder), bladder (specifically bladder cancer), kidney (specifically renal cell carcinoma), liver (specifically hepatocellular carcinoma), stomach (specifically advanced gastric cancer), and urothelial (specifically urothelial cancer) cancers and analyzed pSTAT3 in the unstimulated basal state. In addition, we analyzed pSTAT3 in the unstimulated basal state in various cancers from CD27+CD45RA+CD4 T cells.
그 결과, skin cancer를 제외한 다양한 암종에서 무 자극 기저상태의 pSTAT3의 발현을 확인할 수 있었다(도 6a 내지 6d). 이를 통해, 혈액 내 존재하는 면역세포(특히 CD27+CD45RA+CD4 T cell)가 발현하는 pSTAT3를 관찰하여 암의 여부를 진단할 수 있음을 알 수 있다. As a result, we were able to confirm the expression of pSTAT3 in the unstimulated basal state in various cancers except skin cancer (Figs. 6a to 6d). This suggests that cancer can be diagnosed by observing pSTAT3 expressed by immune cells (especially CD27+CD45RA+CD4 T cells) present in the blood.
실시예 10: 사이토카인을 이용한 건강한 사람의 혈중 면역 세포 자극Example 10: Stimulation of blood immune cells in healthy individuals using cytokines
건강한 사람의 PBMC에 다양한 사이토카인(10ng/ml)을 처리하여 30분간 37℃ CO2 incubator에서 배양한 뒤 pSTAT3를 분석하였다. 그 결과, pSTAT3를 유도하는 사이토카인은 IL-6, IL-10, IL-21 및 IFN-b인 것을 확인할 수 있었다(도 7a). PBMCs from healthy people were treated with various cytokines (10 ng/ml) and cultured in a CO 2 incubator at 37°C for 30 minutes, and then pSTAT3 was analyzed. As a result, it was confirmed that the cytokines that induce pSTAT3 are IL-6, IL-10, IL-21, and IFN-b (Fig. 7a).
또한, 환자의 무 자극 기저상태의 pSTAT3 및 건강한 사람의 사이토카인(IL-6, IL-21, IFN-β, IL-10 또는 자극 없음(NS); 0.5 ng/ml)으로 유도된 pSTAT3를 다양한 면역 세포에서 확인 및 비교하였다. 이와 같이 각 사이토카인에 대한 각 면역 세포의 반응성을 확인한 결과, IL-6만이 무자극 기저상태의 pSTAT3와 동일한 발현 패턴을 나타냈다(도 7b, 7c).In addition, pSTAT3 in the unstimulated basal state of patients and pSTAT3 induced by cytokines (IL-6, IL-21, IFN-β, IL-10, or no stimulation (NS); 0.5 ng/ml) of healthy people were identified and compared in various immune cells. As a result of confirming the responsiveness of each immune cell to each cytokine, only IL-6 showed the same expression pattern as pSTAT3 in the unstimulated basal state (Fig. 7b, 7c).
추가적으로, 건강한 사람의 PBMC에 IL-6(0.5 ng/ml)로 자극을 준 후 각 면역 세포에서 발현하는 pSTAT3를 확인하였다. 그 결과, IL-6를 처리한 건강한 사람의 PBMC 내에서도 CD4 T cell과 CD8 T cell에서만 pSTAT3가 유도되며, 그 중에서도 CD27+CD45RA+인 그룹에서 pSTAT3의 발현이 가장 높은 것을 확인하였다 (도 7d 내지 7f).Additionally, after stimulating PBMCs from healthy people with IL-6 (0.5 ng/ml), pSTAT3 expressed in each immune cell was confirmed. As a result, pSTAT3 was induced only in CD4 T cells and CD8 T cells in PBMCs from healthy people treated with IL-6, and among them, pSTAT3 expression was highest in the CD27+CD45RA+ group (Figs. 7d to 7f).
실시예 11: 혈장을 이용한 NSCLC 환자의 무 자극 기저상태의 pSTAT3 유도 인자 규명Example 11: Identification of pSTAT3 inducers in unstimulated basal state in NSCLC patients using plasma
NSCLC 환자와 건강한 사람의 혈액에서 PBMC와 혈청을 분리한 뒤, 1)환자의 PBMC에서는 무 자극 기저상태의 pSTAT3 발현 정도를 측정하고(pSTAT3ex vivo), 2)혈청에서는 염증성 사이토카인의 농도 및 3)이를 건강한 사람의 PBMC와 공동 배양하였을 때의 혈청 유도 pSTAT3(Serum-induced pSTAT3)를 측정하였다(도 8a).After separating PBMC and serum from the blood of NSCLC patients and healthy individuals, 1) the expression level of pSTAT3 in the unstimulated basal state was measured in the patient's PBMC (pSTAT3 ex vivo ), 2) the concentration of inflammatory cytokines in the serum, and 3) serum-induced pSTAT3 when co-cultured with PBMC from healthy individuals was measured (Fig. 8a).
NSCLC 환자의 혈청 내에서 IL-6의 농도는 다른 염증성 사이토카인들에 비해 월등히 높은 것을 확인하였다(도 8b). 또한 NSCLC 환자의 혈청을 건강한 사람의 PBMC와 공동 배양하였을 때, 혈청 유도 pSTAT3는 T cell 내에서, 그 중에서도 특히 CD27+CD45RA+ 그룹 내에서 많이 발현되었다(도 8c 내지 8e).The concentration of IL-6 in the serum of NSCLC patients was confirmed to be significantly higher than that of other inflammatory cytokines (Fig. 8b). In addition, when the serum of NSCLC patients was co-cultured with PBMCs from healthy people, serum-induced pSTAT3 was highly expressed in T cells, especially in the CD27+CD45RA+ group (Figs. 8c to 8e).
이러한 혈청 유도 pSTAT3는 혈청 내 IL-6의 농도에 정비례하였고(도 8f), 혈청과 PBMC를 공동 배양할 때 anti-IL-6 또는 anti-IL-6R 항체를 넣어 IL-6 신호 전달을 방해한 경우, 앞서 관찰된 혈청 유도 pSTAT3가 전혀 관찰되지 않았다(도 8g, 8h). 추가적으로, NSCLC 환자의 PBMC에서 분석한 무 자극 기저상태의 pSTAT3 발현정도와, 혈청 유도 pSTAT3 및 혈청 내 IL-6 농도는 비례 관계를 보였다(도 8i, 8j). 이 때, 무 자극 기저상태의 pSTAT3 분석이 혈청 내 IL-6 농도 분석에 비해 월등한 민감도를 보였다. 즉, NSCLC 환자를 무 자극 기저상태의 pSTAT3 발현 정도에 따라 pSTAT3ex vivo-lo, pSTAT3ex vivo-int, 그리고 pSTAT3ex vivo-hi로 나누었을 때(도 8k), pSTAT3ex vivo-lo와 pSTAT3ex vivo-int는 혈청 내 IL-6 농도만으로는 구분이 불가능했다(도 8l).This serum-induced pSTAT3 was directly proportional to the concentration of IL-6 in the serum (Fig. 8f), and when the IL-6 signaling was disrupted by adding anti-IL-6 or anti-IL-6R antibodies during co-culture of serum and PBMCs, the serum-induced pSTAT3 observed previously was not observed at all (Figs. 8g, 8h). In addition, the expression level of pSTAT3 in the unstimulated basal state analyzed in PBMCs of NSCLC patients, serum-induced pSTAT3, and serum IL-6 concentration showed a proportional relationship (Figs. 8i, 8j). In this case, the analysis of pSTAT3 in the unstimulated basal state showed superior sensitivity compared to the analysis of serum IL-6 concentration. That is, when NSCLC patients were divided into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to the level of pSTAT3 expression in the unstimulated basal state (Fig. 8k), pSTAT3 ex vivo -lo and pSTAT3 ex vivo -int could not be distinguished based on serum IL-6 concentration alone (Fig. 8l).
이러한 결과를 통하여, 환자의 혈중세포(특히 CD4 Tn 세포)에서 무 자극 기저상태의 pSTAT3를 측정하는 방법은, 환자의 혈중 IL-6 농도를 측정하는 기존 방법에 비해 월등히 높은 민감도를 나타낸다는 것을 확인하였다.Through these results, we confirmed that the method of measuring pSTAT3 in the unstimulated basal state in patients' blood cells (especially CD4 Tn cells) showed significantly higher sensitivity than the existing method of measuring the concentration of IL-6 in patients' blood.
실시예 12: NSCLC 환자의 무 자극 기저상태의 pSTAT3와 종양 침윤 면역 세포 분화 간 연관성 확인Example 12: Correlation between unstimulated baseline pSTAT3 and tumor-infiltrating immune cell differentiation in NSCLC patients
NSCLC 환자의 무 자극 기저상태의 pSTAT3와 종양 침윤 면역 세포 분화의 연관성을 확인하기 위해, NSCLC 환자의 PBMC와 종양 침윤 림프구(Tumor-infiltrating lymphocytes; TIL)을 조사하였다.To determine the association between baseline pSTAT3 and tumor-infiltrating immune cell differentiation in NSCLC patients, PBMCs and tumor-infiltrating lymphocytes (TILs) from NSCLC patients were examined.
먼저 PBMC에서 조사한 CD27+CD45RA+ CD4 T cell의 무 자극 기저상태의 pSTAT3의 발현 정도와, TIL에서 얻은 종양관련 대식세포(Tumor-associated macrophage; TAM)의 CD206 발현 정도(항암 면역을 방해하는 M2-like TAM의 마커)의 연관성을 조사하였다. 그 결과, 혈액 내 무 자극 기저상태의 pSTAT3와 TAM의 CD206 발현 사이의 높은 연관성을 확인할 수 있었다(도 9a). 즉, pSTAT3ex vivo-lo보다 pSTAT3ex vivo-int 및 pSTAT3ex vivo-hi 환자군에서 CD206+% TAM이 많았다(도 9b, 9c).First, we investigated the correlation between the expression level of pSTAT3 in the unstimulated basal state of CD27+CD45RA+ CD4 T cells from PBMCs and the expression level of CD206 (a marker of M2-like TAM that interferes with anticancer immunity) in tumor-associated macrophages (TAMs) obtained from TILs. As a result, we were able to confirm a high correlation between the expression of pSTAT3 in the unstimulated basal state in the blood and CD206 in TAMs (Fig. 9a). That is, CD206+% TAMs were higher in the pSTAT3 ex vivo -int and pSTAT3 ex vivo -hi patient groups than in the pSTAT3 ex vivo -lo (Figs. 9b, 9c).
다음으로, 앞서 관찰한 무 자극 기저상태의 pSTAT3와 CD8 T cell 분화와의 연관성을 조사하였다. 이 때, pSTAT3ex vivo-lo에서 pSTAT3ex vivo-int 환자 군으로 갈수록 암에 특이적이라고 알려진 CD103+CD39+ CD8+ TIL의 비율이 높아지다가, pSTAT3ex vivo-hi 환자군에서 다시 낮아지는 것을 확인할 수 있었다(도 9d 내지 9f). 이는 무 자극 기저상태의 pSTAT3의 발현 정도는 TIL의 분화와 밀접한 관계를 가지고 있으며, 그 관계가 정비례(또는 반비례) 관계가 아니라, 종형곡선(bell curve) 관계일 수도 있다는 것을 보여준다.Next, we investigated the relationship between pSTAT3 in the unstimulated basal state and CD8 T cell differentiation observed previously. At this time, it was confirmed that the proportion of CD103+CD39+ CD8+ TIL, which is known to be specific for cancer, increased from the pSTAT3 ex vivo -lo to the pSTAT3 ex vivo -int patient group, and then decreased again in the pSTAT3 ex vivo -hi patient group (Figs. 9d to 9f). This shows that the expression level of pSTAT3 in the unstimulated basal state is closely related to the differentiation of TIL, and that the relationship may not be a direct (or inverse) relationship, but a bell curve relationship.
실시예 13: NSCLC 환자의 무 자극 기저상태의 pSTAT3와 면역관문억제제에 대한 반응성 연관관계 확인 Example 13: Correlation between baseline pSTAT3 and response to immune checkpoint inhibitors in NSCLC patients
NSCLC 환자의 무 자극 기저상태의 pSTAT3와 면역항암치료에 대한 반응성과의 연관성을 확인하였다. Pembrolizumab 단독요법 또는 병용요법(Pemetrexed + cisplatin)을 항암 1차 치료로 받은 4기 NSCLC 환자들의 치료 전 혈액에서 CD27+CD45RA+ CD4 T cell의 무 자극 기저상태의 pSTAT3를 확인하고, 치료 후 3달째 반응성과의 연관성을 조사하였다.We investigated the association between baseline unstimulated pSTAT3 and response to immunotherapy in NSCLC patients. We identified baseline unstimulated pSTAT3 of CD27+CD45RA+ CD4 T cells in the blood of stage IV NSCLC patients who received pembrolizumab monotherapy or combination therapy (pemetrexed + cisplatin) as first-line chemotherapy, and investigated the association with
무 자극 기저상태의 pSTAT3에 따라 해당 환자들을 pSTAT3ex vivo-lo, pSTAT3ex vivo-int, 및 pSTAT3ex vivo-hi로 나누었을 때(도 10a), 앞선 실시예 11에서의 결과를 바탕으로 혈청 내 IL-6가 많이 검출될 것으로 예상되는 pSTAT3ex vivo-hi는, 다른 환자들(pSTAT3ex vivo-lo/int)에 비해 부분반응(partial response; PR)을 보인 환자의 비율이 적었다(도 10b).When the patients were divided into pSTAT3 ex vivo -lo, pSTAT3 ex vivo -int, and pSTAT3 ex vivo -hi according to pSTAT3 in the unstimulated basal state (Fig. 10a), pSTAT3 ex vivo -hi, in which a high level of IL-6 in the serum was expected to be detected based on the results of the previous Example 11, showed a lower proportion of patients who showed a partial response (PR) compared to other patients (pSTAT3 ex vivo -lo/int) (Fig. 10b).
기존의 혈청 내 IL-6 농도를 측정하는 방식으로는 구분되지 않지만, 본 발명의 무 자극 기저상태의 pSTAT3 측정방식으로는 확연하게 구분되는 환자들(즉, pSTAT3ex vivo-lo 대비 pSTAT3ex vivo-int) 간 세부 비교를 수행하여, 면역관문억제제에 대한 반응성 차이를 더욱 명확히 확인하였다.A detailed comparison was performed between patients who were clearly distinguished by the unstimulated basal pSTAT3 measurement method of the present invention (i.e., pSTAT3 ex vivo -lo vs. pSTAT3 ex vivo -int), which were not distinguished by the conventional method of measuring serum IL-6 concentrations, to more clearly confirm the difference in responsiveness to immune checkpoint inhibitors.
pSTAT3ex vivo-int 그룹에서 특히 PR 환자의 비율이 높았고, pSTAT3ex vivo-lo 그룹은 pSTAT3ex vivo-hi 그룹만큼이나 반응성이 좋지 못했다(도 10c). 즉, 혈액 내 무 자극 기저상태의 pSTAT3가 약 20 내지 60%인 환자 군에서 면역관문억제제에 대한 반응성이 가장 높은 것으로 예측 가능함을 확인하였다.The proportion of PR patients was particularly high in the pSTAT3 ex vivo -int group, and the pSTAT3 ex vivo -lo group was not as responsive as the pSTAT3 ex vivo -hi group (Fig. 10c). In other words, it was confirmed that the highest responsiveness to immune checkpoint inhibitors can be predicted in the patient group in which the basal pSTAT3 level in the blood was approximately 20 to 60% of unstimulated level.
Claims (15)
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| KR10-2023-0091894 | 2023-07-14 | ||
| KR20230091894 | 2023-07-14 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025018697A1 true WO2025018697A1 (en) | 2025-01-23 |
Family
ID=94282188
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/KR2024/009914 Pending WO2025018697A1 (en) | 2023-07-14 | 2024-07-11 | Sample treatment method for cancer diagnosis, and method for providing information about cancer diagnosis by using same |
Country Status (2)
| Country | Link |
|---|---|
| KR (1) | KR20250011598A (en) |
| WO (1) | WO2025018697A1 (en) |
Citations (5)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| KR19980702119A (en) * | 1995-02-10 | 1998-07-15 | 스티븐슨 린다 에스 | Human Pancreatic Cell Line: Development and Uses |
| KR20070026303A (en) * | 2003-09-11 | 2007-03-08 | 휴비트 제노믹스 가부시키가이샤 | Methods and kits for detecting proliferative diseases leading to cure, prophylactic and / or therapeutic agents for proliferative diseases leading to cure, and methods and kits for identifying substances effective for preventing and / or treating proliferative diseases leading to cure |
| JP2011516431A (en) * | 2008-04-03 | 2011-05-26 | ベイジン バイオスター テクノロジーズ,リミテッド | Phostricin derivative and its pharmaceutical use |
| JP2017074048A (en) * | 2004-03-31 | 2017-04-20 | ザ ジェネラル ホスピタル コーポレイション | Methods for determining cancer responsiveness to treatments targeting epithelial cell growth factor receptor |
| JP2019536426A (en) * | 2016-08-26 | 2019-12-19 | イマティクス バイオテクノロジーズ ゲーエムベーハー | Novel peptides and scaffolds for use in immunotherapy for head and neck squamous cell carcinoma and other cancers |
Family Cites Families (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| KR20210103212A (en) | 2020-02-13 | 2021-08-23 | (주) 프로탄바이오 | Multiple Biomarkers for Lung Cancer Diagnosis and Uses thereof |
-
2024
- 2024-07-11 KR KR1020240091882A patent/KR20250011598A/en active Pending
- 2024-07-11 WO PCT/KR2024/009914 patent/WO2025018697A1/en active Pending
Patent Citations (5)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| KR19980702119A (en) * | 1995-02-10 | 1998-07-15 | 스티븐슨 린다 에스 | Human Pancreatic Cell Line: Development and Uses |
| KR20070026303A (en) * | 2003-09-11 | 2007-03-08 | 휴비트 제노믹스 가부시키가이샤 | Methods and kits for detecting proliferative diseases leading to cure, prophylactic and / or therapeutic agents for proliferative diseases leading to cure, and methods and kits for identifying substances effective for preventing and / or treating proliferative diseases leading to cure |
| JP2017074048A (en) * | 2004-03-31 | 2017-04-20 | ザ ジェネラル ホスピタル コーポレイション | Methods for determining cancer responsiveness to treatments targeting epithelial cell growth factor receptor |
| JP2011516431A (en) * | 2008-04-03 | 2011-05-26 | ベイジン バイオスター テクノロジーズ,リミテッド | Phostricin derivative and its pharmaceutical use |
| JP2019536426A (en) * | 2016-08-26 | 2019-12-19 | イマティクス バイオテクノロジーズ ゲーエムベーハー | Novel peptides and scaffolds for use in immunotherapy for head and neck squamous cell carcinoma and other cancers |
Also Published As
| Publication number | Publication date |
|---|---|
| KR20250011598A (en) | 2025-01-21 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Noël et al. | Functional Th1-oriented T follicular helper cells that infiltrate human breast cancer promote effective adaptive immunity | |
| Levitsky et al. | Immunosuppression withdrawal in liver transplant recipients on sirolimus | |
| Loewendorf et al. | Normal human pregnancy results in maternal immune activation in the periphery and at the uteroplacental interface | |
| Maes et al. | Decreased serum dipeptidyl peptidase IV activity in major depression | |
| Gane et al. | Flow cytometric monitoring of allergen induced basophil activation | |
| WO2013168876A1 (en) | Kit for monitoring immune status after transplant and monitoring method using same | |
| WO2020027446A1 (en) | Breast cancer early diagnosis and post-therapy monitoring method using liquid biopsy multiple cancer gene biomarkers | |
| Kared et al. | SLAMF7 defines subsets of human effector CD8 T cells | |
| WO2025018697A1 (en) | Sample treatment method for cancer diagnosis, and method for providing information about cancer diagnosis by using same | |
| CA2128424A1 (en) | Method for determining cytolytic t cell precursors | |
| CA2709823A1 (en) | Improved assay | |
| Prince et al. | CD69 expression reliably predicts the anti-CD3-induced proliferative response of lymphocytes from human immunodeficiency virus type 1-infected patients | |
| WO2017204446A2 (en) | Method for testing nk cell activity using synergistic activity of receptor, and method for diagnosing disease associated with nk cell activity using same | |
| WO2024101823A1 (en) | Biomarker for predicting immunotherapeutic responsiveness based on spatial transcriptome analysis and uses thereof | |
| WO2020091367A1 (en) | Biomarker for breast cancer diagnosis and use thereof | |
| US20190302116A1 (en) | Method for testing nk cell activity using synergistic activity of receptor, and method for diagnosing disease associated with nk cell activity using same | |
| WO2022265326A1 (en) | Method for predicting prognosis of pancreatic cancer, and kit therefor | |
| WO2020226366A1 (en) | Marker for diagnosing colorectal cancer and method for providing information required for diagnosis of colorectal cancer | |
| WO2020184911A1 (en) | Marker for predicting tumor reactivity of lymphocytes, and use thereof | |
| WO2020256530A1 (en) | Urine exosome biomarker for diagnosing t cell mediated rejection after kidney transplantation or predicting prognosis of patient after kidney transplantation | |
| WO2011132872A2 (en) | Information providing method for diagnosis of tuberculosis, and tuberculosis diagnostic kit for same | |
| Fleisher | Evaluation of suspected immunodeficiency | |
| Cissé et al. | Prevalence of COVID-19 at the Wahgnion-Gold mining site in Burkina Faso and use of RT-PCR initial cycle threshold to monitor the dynamics of SARS-CoV-2 load | |
| EP3611504B1 (en) | Method for detecting effector-type t-cells, t-cells analyzing device and analyzing system | |
| WO2025048570A1 (en) | Marker for predicting therapeutic response to immune checkpoint inhibitor in renal cancer, and method for predicting therapeutic response by using same |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24843415 Country of ref document: EP Kind code of ref document: A1 |