WO2018190382A1 - Method for detecting pd-l1-positive cancer cells - Google Patents

Method for detecting pd-l1-positive cancer cells Download PDF

Info

Publication number
WO2018190382A1
WO2018190382A1 PCT/JP2018/015274 JP2018015274W WO2018190382A1 WO 2018190382 A1 WO2018190382 A1 WO 2018190382A1 JP 2018015274 W JP2018015274 W JP 2018015274W WO 2018190382 A1 WO2018190382 A1 WO 2018190382A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
fluorescent dye
antibody
recognizes
cell
Prior art date
Application number
PCT/JP2018/015274
Other languages
French (fr)
Japanese (ja)
Inventor
理美 八木
勝也 遠藤
雅之 樋口
清太 中村
上原 寿茂
泰浩 洪
信之 山本
Original Assignee
日立化成株式会社
公立大学法人和歌山県立医科大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 日立化成株式会社, 公立大学法人和歌山県立医科大学 filed Critical 日立化成株式会社
Priority to JP2019512558A priority Critical patent/JPWO2018190382A1/en
Publication of WO2018190382A1 publication Critical patent/WO2018190382A1/en

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer

Definitions

  • the present invention relates to a method for detecting PD-L1-positive cancer cells.
  • Immuno checkpoint inhibition therapy is known as one of cancer treatment methods.
  • immune checkpoint inhibition therapy immune cells are activated by inhibiting reaction pathways (immune checkpoints) that suppress immune responses.
  • the PD-1 / PD-L1 pathway is known as a reaction pathway that suppresses the immune response, and the development of immune checkpoint inhibitors “pembrolizumab” and “nivolumab” that inhibit this pathway is progressing.
  • pembrolizumab and “nivolumab” that inhibit this pathway is progressing.
  • the efficacy of the inhibitor acting on the PD-1 / PD-L1 pathway is low. Therefore, it is important to confirm the efficacy of the inhibitor before actually administering the inhibitor by detecting PD-L1 in the cancer cells of the patient.
  • Non-patent document 1 As a method for detecting cancer cells expressing PD-L1 (PD-L1-positive cancer cells), a method of collecting a tissue sample from a patient and staining PD-L1 in the sample is known (for example, Non-patent document 1).
  • the conventional method for detecting PD-L1-positive cancer cells requires a large burden on the patient because it requires collecting tumor tissue from the patient.
  • An object of the present invention is to detect PD-L1-positive cancer cells without imposing a heavy burden on a subject.
  • the present invention is a method for detecting PD-L1-positive cancer cells in a blood sample, comprising: (a) collecting cells from the blood sample; and (b) a primary antibody that recognizes PD-L1 in the cells. Contacting and then contacting a secondary antibody that recognizes the primary antibody and labeled with a fluorescent dye, or a cell that is an antibody that recognizes PD-L1 and labeled with a fluorescent dye And (c) irradiating a cell with excitation light of a fluorescent dye to detect fluorescence emitted from the cell.
  • the fluorescent dye may be a first fluorescent dye, and at any stage after step (a) and before step (c), (x1) a cell is contacted with a primary antibody that recognizes a leukocyte marker protein. And then contacting a secondary antibody that recognizes the primary antibody and that is labeled with a second fluorescent dye, or an antibody that recognizes leukocytes and a second fluorescent dye (X2) contacting the cell with a primary antibody that recognizes a marker protein of epithelial cells, and then a secondary antibody that recognizes the primary antibody with a third fluorescent dye.
  • the cells are irradiated with excitation light of the first, second, third and fourth fluorescent dyes, respectively.
  • the fluorescence of the first, second, third, and fourth fluorescent dyes emitted from the cells may be detected, respectively.
  • the primary antibody recognizing PD-L1 or the antibody recognizing PD-L1 may be derived from a 28-8 or SP142 clone.
  • Step (a) may be a step of capturing a cell on a filter by filtering a blood sample with a filter.
  • the leukocyte marker protein may be CD45.
  • the epithelial cell marker protein may be cytokeratin.
  • the first, second, and third fluorescent dyes may be selected from the group consisting of fluorescein, Alexa Fluor (registered trademark) 594, and Alexa Fluor (registered trademark) 647, wherein the fourth fluorescent dye is 4 ', It may be 6-diamidino-2-phenylindole.
  • the PD-L1 positive cancer cell may be derived from lung cancer.
  • the step (x1) may be performed before the step (b), and the step (x2) and the step (x3) may be performed simultaneously after the step (b).
  • PD-L1-positive cancer cells can be detected easily and in a short time without imposing a heavy burden on the subject by using a blood sample.
  • FIG. 2 is a sectional view taken along line II-II in FIG. 2 is an image of cells fluorescently labeled in Example 1.
  • FIG. 2 is an image of fluorescently labeled cells in Example 2.
  • FIG. 2 is an image of fluorescently labeled cells in Example 2.
  • FIG. 4 is an image of cells fluorescently labeled in Example 3.
  • 4 is an image of cells fluorescently labeled in Example 3.
  • 4 is an image of cells fluorescently labeled in Example 4.
  • the method for detecting PD-L1-positive cancer cells in a blood sample comprises (a) a step of collecting cells from a blood sample, and (b) contacting the cells with a primary antibody that recognizes PD-L1. Next, a step of bringing a secondary antibody that recognizes the primary antibody into contact with a secondary antibody that is labeled with a fluorescent dye, or a cell that is an antibody that recognizes PD-L1 and is labeled with a fluorescent dye A step of bringing the antibody into contact, and (c) a step of irradiating the cell with excitation light of a fluorescent dye to detect fluorescence emitted from the cell.
  • CTC circulating tumor cells
  • CTCs are those in which cancer cells in the lungs, liver, stomach, head and neck, bladder, urothelium, esophagus, biliary tract, breast, ovary, uterus, liver, prostate, or pancreas have entered blood vessels and lymph vessels.
  • PD-L1-positive cancer cells can be detected by using a blood sample of a subject without collecting cancer tissue in these organs.
  • CTC and “cancer cells in a blood sample” are treated as synonymous.
  • blood collected from a subject may be used as it is, or blood diluted with a buffer solution such as phosphate buffered saline (PBS) or other suitable medium may be used.
  • PBS phosphate buffered saline
  • the blood sample may be added with additives that are usually added to blood samples, such as anticoagulants and fixatives.
  • the cells can be collected from the blood sample by, for example, filtering the blood sample with a filter and capturing the cells in the blood sample on the filter.
  • leukocytes have the same diameter as CTC, so that some leukocytes are captured together with CTC on the filter.
  • detection of PD-L1-positive cancer cells can be performed on the filter as it is. That is, all the steps in the present invention can be performed on the cells captured on the filter.
  • Capture means that the liquid containing the cells is filtered through, leaving the cells on the filter.
  • the filter is not particularly limited as long as it can capture CTC present in the blood sample, and a conventionally known filter can be used.
  • the filter may be, for example, a metal or resin filter, and is provided with a substrate and a through-hole provided on the substrate, preferably having a pore diameter of 5 ⁇ m to 15 ⁇ m, more preferably 6 ⁇ m to 12 ⁇ m, and even more preferably 7 ⁇ m to 10 ⁇ m. You may have.
  • the hole diameter of the through hole refers to the maximum value of the diameter of a sphere that can pass through the through hole.
  • contacting a substance with a cell can be performed, for example, by immersing the cell in the substance or a solution of the substance.
  • cleaning liquid with a cell can be performed by filtering these solutions with a filter.
  • the flow rate of the solution is preferably 50 ⁇ L / min to 3000 ⁇ L / min, more preferably 100 ⁇ L / min to 1000 ⁇ L / min, and 200 ⁇ L / min to 600 ⁇ L / min. Further preferred.
  • the cells may be washed.
  • the washing step is performed, for example, by bringing a washing solution containing a known buffer solution such as PBS into contact with the cells.
  • the washing solution may contain additives such as bovine serum albumin (BSA) or ethylenediaminetetraacetic acid (EDTA). Washing is not limited to after step (a), and can be performed appropriately after each step.
  • BSA bovine serum albumin
  • EDTA ethylenediaminetetraacetic acid
  • cells may be immobilized after step (a).
  • the cells can be fixed by contacting the cells with a known fixing agent such as formaldehyde. By fixing the cells, cell spoilage or aggregation can be further reduced.
  • the immobilized cells may then be permeabilized.
  • a cell can be permeabilized by contacting the cell with a known permeabilizing agent.
  • a permeation treatment agent for example, poly (oxyethylene) octylphenyl ether can be used.
  • a secondary antibody that recognizes the primary antibody is contacted with a primary antibody that recognizes PD-L1, and is then labeled with a fluorescent dye (first fluorescent dye). Is contacted (two-step fluorescent labeling).
  • an antibody that recognizes PD-L1 and is labeled with a fluorescent dye (first fluorescent dye) is brought into contact with cells (one-step fluorescent labeling).
  • PD-L1 fluorescent labeling may be performed in two steps or one step.
  • the primary antibody that recognizes PD-L1 or the antibody that recognizes PD-L1 and is labeled with a fluorescent dye is, for example, 28-8, SP142, E1L3N (registered trademark), And a clone selected from the group consisting of EPR1161 (2), or a polyclonal antibody (for example, catalog number: 4059 from Prosci).
  • a fluorescent dye for example, 28-8, SP142, E1L3N (registered trademark)
  • a clone selected from the group consisting of EPR1161 (2), or a polyclonal antibody for example, catalog number: 4059 from Prosci.
  • the primary antibody that recognizes PD-L1 or the antibody that recognizes PD-L1 is preferably derived from 28-8 or SP142.
  • the antibodies derived from these clones are all anti-PD-L1 rabbit monoclonal antibodies.
  • the fluorescent dye is not particularly limited as long as it is a fluorescent dye usually used for fluorescent labeling of antibodies.
  • the first fluorescent dye is, for example, Alexa Fluor (registered trademark) 647 or Cy (registered trademark) 5.
  • step (c) the cells are irradiated with excitation light of a fluorescent dye to detect fluorescence emitted from the cells.
  • a cell in which fluorescence due to the fluorescent dye (first fluorescent dye) is detected (positive) is identified as a PD-L1-positive cancer cell.
  • the detected PD-L1-positive cancer cells can then be analyzed for DNA, RNA or protein.
  • sequencer, next-generation sequencer, DNA chip, microarray, comparative genomic hybridization, fluorescence in situ hybridization, digital PCR, quantitative reverse transcription PCR, ELISA, Western plotting, TOF -MS, MALDI-MS, Raman spectroscopy, chromatography, X-ray crystallography, two-dimensional electrophoresis, nuclear magnetic resonance spectroscopy, flow cytometer (FCM), etc. can be used for analysis.
  • step (b) an antibody recognizing PD-L1 binds to PD-L1-negative cells, and fluorescence indicating PD-L1 may be observed from PD-L1-negative cells (false positive). ). From the viewpoint of reducing such false positives and obtaining a more reliable detection result, the following steps (x1) to (x3) are performed at any stage after step (a) and before step (c). Further, it is preferable to carry out.
  • a cell is contacted with a primary antibody that recognizes a marker protein of leukocytes, and then a secondary antibody that recognizes the primary antibody and is labeled with a second fluorescent dye is contacted (Two-step fluorescent labeling).
  • a secondary antibody that recognizes the primary antibody and is labeled with a second fluorescent dye is contacted.
  • cells are contacted with an antibody that recognizes leukocytes and is labeled with a second fluorescent dye (one-step fluorescent labeling).
  • leukocytes are fluorescently labeled.
  • the fluorescent labeling of leukocytes may be performed in either two steps or one step as described above.
  • Leukocyte marker protein is, for example, CD45 expressed in all hematopoietic stem cells.
  • a primary antibody that recognizes a leukocyte marker protein, a secondary antibody that is labeled with a second fluorescent dye, and an antibody that recognizes a leukocyte marker protein and is labeled with a second fluorescent dye It is not limited, A polyclonal antibody or a monoclonal antibody may be sufficient.
  • the animal from which the antibody is derived is not particularly limited as long as the animal from which the primary antibody is derived is different from the animal from which the secondary antibody is derived.
  • the second fluorescent dye is not particularly limited as long as it is a fluorescent dye usually used for fluorescent labeling of antibodies.
  • the second fluorescent dye is, for example, Alexa Fluor (registered trademark) 594 or Texas Red (registered trademark).
  • the second fluorescent dye is a fluorescent dye different from the first, third and fourth fluorescent dyes. Each fluorescent dye is distinguishable because it has a different fluorescence wavelength.
  • the first, second, and third fluorescent dyes are selected from the group consisting of fluorescein, Alexa Fluor 594, and Alexa Fluor 647, and the fourth fluorescent dye is 4 ', 6-diamidino-2-phenyl. Indole (DAPI).
  • step (x2) the cell is contacted with a primary antibody that recognizes a marker protein of epithelial cells, and then a secondary antibody that recognizes the primary antibody and is labeled with a third fluorescent dye is contacted (Two-step fluorescent labeling).
  • a secondary antibody that recognizes the primary antibody and is labeled with a third fluorescent dye is contacted.
  • cells are contacted with an antibody that recognizes a marker protein of epithelial cells and labeled with a third fluorescent dye (one-step fluorescent labeling).
  • CTC is fluorescently labeled.
  • CTC fluorescent labeling may be performed in either two steps or one step as described above.
  • epithelial cell marker proteins examples include cytokeratin, epithelial cell adhesion molecule (EpCAM), CD146, and CD176, with cytokeratin being preferred. Since CTC is derived from epithelial cells, it has a marker protein for these epithelial cells.
  • the third fluorescent dye is not particularly limited as long as it is a fluorescent dye usually used for fluorescent labeling of antibodies.
  • the third fluorescent dye is, for example, fluorescein such as fluorescein isothiocyanate (FITC) or Alexa Fluor (registered trademark) 488.
  • a primary antibody that recognizes a marker protein of epithelial cells, a secondary antibody that is labeled with a third fluorescent dye, and an antibody that recognizes a marker protein of epithelial cells and is labeled with a third fluorescent dye may be a polyclonal antibody or a monoclonal antibody.
  • the animal from which the antibody is derived is not particularly limited as long as the animal from which the primary antibody is derived is different from the animal from which the secondary antibody is derived.
  • the cell nucleus is labeled with a fourth fluorescent dye.
  • the fourth fluorescent dye for labeling the nucleus is not particularly limited as long as it is a fluorescent dye capable of binding to a nucleic acid, and a fluorescent dye usually used for fluorescently labeling a nucleus can be used.
  • Examples of the fourth fluorescent dye include DAPI and 2 ′-(4-ethoxyphenyl) -5- (4-methyl-1-piperazinyl) -2,5′-bi-1H-benzimidazole trihydrochloride (Hoechst 33342). ).
  • step (c) the cells are irradiated with excitation light of the first, second, third and fourth fluorescent dyes, respectively. Fluorescence of the first, second, third and fourth fluorescent dyes emitted from is detected respectively. PD-L1-positive cancer cells are labeled with the first, third, and fourth fluorescent dyes, but are not labeled with the second fluorescent dye. Therefore, cells in which the fluorescence of the second fluorescent dye is not detected (negative) and the fluorescence of the first, third, and fourth fluorescent dyes are detected (positive) are identified as PD-L1-positive cancer cells.
  • the cartridge shown in FIGS. 1 and 2 can be used.
  • a method for detecting PD-L1-positive cancer cells in a blood sample using a cartridge according to an embodiment of the present invention will be described. Unless otherwise stated, the details of each step and the order of the steps are as described in the above embodiment.
  • a CTC capturing cartridge (cartridge) 100 shown in FIGS. 1 and 2 has a housing having an inlet 130 to which an inflow pipe 125 into which liquid flows is connected and an outlet 140 to which an outflow pipe 135 from which liquid flows out is connected.
  • a body 120 and a filter 105 are provided.
  • the filter 105 is fixed by a casing 120 including an upper member 110 and a lower member 115.
  • the blood sample, the cleaning liquid, and other reaction liquids are introduced into the housing 120 through the inflow pipe 125, and are discharged to the outside through the filter 105 through the outflow pipe 135.
  • Such a liquid flow can be created, for example, by connecting a pump upstream of the inflow pipe 125 or downstream of the outflow pipe 135.
  • a cock may be provided upstream of the inflow pipe 125 and / or downstream of the outflow pipe 135 to control the flow of the liquid.
  • a blood sample is introduced into the cartridge 100 from the inflow tube 125, and the blood sample is filtered by the filter 105 (step (a)).
  • CTC and some white blood cells in the blood sample cannot pass through the through hole 106 of the filter 105 and remain on the surface of the filter 105.
  • Other components in the blood sample pass through the through hole 106 and are discharged out of the cartridge 100.
  • the filter 105 may be cleaned by passing a cleaning solution through the filter 105.
  • the filter 105 can be appropriately washed after the following steps.
  • a reaction solution containing a fixing agent and then a permeabilizing agent is optionally introduced into the cartridge 100, and held in the cartridge 100 for a predetermined time.
  • a fixing agent and a permeation treatment agent may be reacted with each other.
  • reaction solution containing a primary antibody that recognizes PD-L1 and then a secondary antibody that recognizes the primary antibody and is labeled with a fluorescent dye (first fluorescent dye) are applied to the cells.
  • the contained reaction solution is reacted with the cells captured on the filter 105.
  • a reaction solution containing an antibody that recognizes PD-L1 and is labeled with a fluorescent dye (first fluorescent dye) is allowed to react with the cells captured on the filter 105 (step (step ( b)).
  • the cartridge 100 is irradiated with excitation light of a fluorescent dye using a fluorescence microscope to detect fluorescence emitted from the cells captured on the filter 105 (step (c)).
  • the fluorescence is detected by, for example, observing the cartridge 100 from the upper surface in the vertical direction of the cartridge 100 and processing the fluorescence observation image.
  • the steps (x1) to (x3) can optionally be further performed.
  • Example 1 The non-small cell lung cancer cell line contained in the culture flask was cultured at 37 ° C. in a carbon dioxide incubator. Trypsin-EDTA with a concentration of 0.25% was added to the culture flask, and the cultured cells attached to the flask were detached from the flask. The detached cells were counted using a hemocytometer and a phase contrast microscope. A blood sample in which the blood of a lung cancer patient was sprinkled was prepared by adding 100 cells to the blood of a healthy person collected in a blood collection tube.
  • NCI-H820 high expression of PD-L1
  • NCI-H441 expressed in PD-L1
  • A549 low expression of PD-L1
  • NCI-H23 differed in the expression level of PD-L1, respectively.
  • Four types of blood samples were prepared using four types (PD-L1 negative).
  • a blood collection tube a blood collection tube containing EDTA-2K (ethylenediaminetetraacetic acid dipotassium salt) manufactured by Becton Dickinson & Company was used.
  • the CTC capture device includes a reservoir for introducing a blood sample and other reaction solution, and a CTC capture cartridge.
  • the CTC capture cartridge (hereinafter also referred to as cartridge) includes a thin-film metal filter (membrane area 6 mm ⁇ 6 mm, film thickness 18 ⁇ m) having a large number of through-holes having a major axis of 100 ⁇ m and a minor axis of 8 ⁇ m. This corresponds to the cartridge 100.
  • the cartridge was filled with a PBS solution containing 0.5% BSA and 2 mM EDTA (hereinafter referred to as “cleaning solution”). 7 mL of the washing solution was placed in the reservoir, and 3 mL of the blood sample was added under the washing solution so that the blood sample and the washing solution were layered.
  • the CTC capture device was activated, the blood sample and the washing solution in the reservoir were introduced into the cartridge at a flow rate of 200 ⁇ L / min, and the cells in the blood sample were captured on the filter.
  • a washing solution was introduced into the cartridge to wash away blood components remaining on the filter.
  • a reaction solution containing 1.25 mL of anti-human CD45 mouse monoclonal antibody (clone: 2D1) was introduced into the cartridge at a flow rate of 200 ⁇ L / min and reacted at room temperature for 30 minutes.
  • 1.40 mL of the washing solution was introduced into the cartridge at a flow rate of 400 ⁇ L / min, and the reaction solution in the cartridge was discharged.
  • a reaction solution containing 1.25 mL of Alexa Fluor 594-labeled anti-mouse IgG goat polyclonal antibody was introduced into the cartridge at a flow rate of 400 ⁇ L / min and reacted at room temperature for 30 minutes.
  • 1.40 mL of the washing solution was introduced into the cartridge at a flow rate of 400 ⁇ L / min, and the reaction solution in the cartridge was discharged.
  • a reaction solution containing 1.25 mL of anti-human PD-L1 rabbit monoclonal antibody (clone: 28-8) was introduced into the cartridge at a flow rate of 200 ⁇ L / min and reacted at room temperature for 60 minutes. 1.40 mL of the washing solution was introduced into the cartridge at a flow rate of 400 ⁇ L / min, and the reaction solution in the cartridge was discharged.
  • a reaction solution containing 1.25 mL of Alexa Fluor 647-labeled anti-rabbit IgG goat polyclonal antibody was introduced into the cartridge at a flow rate of 400 ⁇ L / min and reacted at room temperature for 30 minutes. 1.40 mL of the washing solution was introduced into the cartridge at a flow rate of 400 ⁇ L / min, and the reaction solution in the cartridge was discharged.
  • a reaction solution containing FITC-labeled anti-human cytokeratin mouse monoclonal antibody (clone: mixture of CK3, 6H5, AE1, and AE3) and DAPI is introduced into the cartridge at 400 ⁇ L / min and reacted at room temperature for 30 minutes. I let you. 3.00 mL of the cleaning solution was introduced into the cartridge at a flow rate of 400 ⁇ L / min, and the reaction solution in the cartridge was discharged. The cartridge was then removed from the CTC capture device.
  • the cartridge was set on a fluorescence microscope. Fluorescent mirror units (FITC, Alexa Fluor 594, Alexa Fluor 647, and DAPI) were each excited using a fluorescent mirror unit. The fluorescence emitted from each fluorescent dye was photographed, and the resulting images were synthesized.
  • Fluorescent mirror units FITC, Alexa Fluor 594, Alexa Fluor 647, and DAPI
  • H820 means NCI-H820
  • H441 means NCI-H441
  • H23 means NCI-H23 (hereinafter the same).
  • NCI-H820 and NCI-H441 which are PD-L1-positive cancer cell lines
  • the cell nucleus (DAPI), cytokeratin (FITC), and PD-L1 are positive
  • FITC cytokeratin
  • PD-L1 Alexa Fluor 647
  • a fluorescent image of cells negative for CD45 Alexa Fluor 594
  • NCI-H23 which is a PD-L1-negative cancer cell line
  • fluorescence images of cells positive for cell nuclei and cytokeratin and negative for CD45 and PD-L1 were obtained.
  • Example 2 NCI-H820 was used as a cell line. Cell fluorescence was observed in the same manner as in Example 1 except that the clone of the anti-human PD-L1 rabbit monoclonal antibody was changed to SP142.
  • FIGS. As shown in these figures, fluorescence images of cells positive for cell nuclei, cytokeratin, and PD-L1, and negative for CD45 were obtained. In the lower right of FIG. 5, leukocytes that are positive for cell nuclei and CD45, and negative for cytokeratin and PD-L1 are seen. There were no false positives due to nonspecific binding of antibodies.
  • Example 3 NCI-H820 was used as a cell line.
  • FIG. 7 shows that non-specific binding of the antibody was observed in the experiment using any antibody.
  • Example 4 Cell fluorescence was observed in the same manner as in Example 1 except that the labeling dye of the secondary antibody (anti-rabbit IgG goat polyclonal antibody) against PD-L1 was changed to Alexa Fluor 680 and Alexa Fluor (registered trademark) 700. .

Abstract

This PD-L1-positive cancer cell detection method comprises: (a) a step for collecting cells from a blood sample; (b) a step for bringing the cells into contact with a primary antibody capable of recognizing PD-L1 and further bringing the cells into contact with a secondary antibody which is capable of recognizing the primary antibody and which is labeled with a fluorescent dye, or a step for bringing the cells into contact with an antibody capable of recognizing PD-L1 and labeled with a fluorescent dye; and (c) a step for irradiating the cells with excitation light for the fluorescent dye so as to detect fluorescence emitted from the cells. This method enables detecting a PD-L1-positive cancer cell without imposing too much burden on a test subject.

Description

PD-L1陽性癌細胞の検出方法Method for detecting PD-L1-positive cancer cells
 本発明は、PD-L1陽性癌細胞の検出方法に関する。 The present invention relates to a method for detecting PD-L1-positive cancer cells.
 癌の治療方法の一つとして「免疫チェックポイント阻害療法」が知られている。免疫チェックポイント阻害療法では、免疫反応を抑制する反応経路(免疫チェックポイント)を阻害することにより、免疫細胞を活性化させる。免疫反応を抑制する反応経路としてPD-1/PD-L1経路が知られており、この経路を阻害する免疫チェックポイント阻害剤「ペムブロリズマブ」及び「ニボルマブ」の開発が進んでいる。患者の癌細胞がPD-L1を有さない場合、PD-1/PD-L1経路に作用する上記阻害剤の奏効性は低い。そのため、患者の癌細胞におけるPD-L1を検出することにより、阻害剤の奏効性を、実際に阻害剤を投与する前に確認することが重要である。 “Immune checkpoint inhibition therapy” is known as one of cancer treatment methods. In immune checkpoint inhibition therapy, immune cells are activated by inhibiting reaction pathways (immune checkpoints) that suppress immune responses. The PD-1 / PD-L1 pathway is known as a reaction pathway that suppresses the immune response, and the development of immune checkpoint inhibitors “pembrolizumab” and “nivolumab” that inhibit this pathway is progressing. When the patient's cancer cells do not have PD-L1, the efficacy of the inhibitor acting on the PD-1 / PD-L1 pathway is low. Therefore, it is important to confirm the efficacy of the inhibitor before actually administering the inhibitor by detecting PD-L1 in the cancer cells of the patient.
 PD-L1を発現している癌細胞(PD-L1陽性癌細胞)を検出する方法として、患者から組織検体を採取して、検体におけるPD-L1を染色する方法が知られている(例えば、非特許文献1)。 As a method for detecting cancer cells expressing PD-L1 (PD-L1-positive cancer cells), a method of collecting a tissue sample from a patient and staining PD-L1 in the sample is known (for example, Non-patent document 1).
 PD-L1陽性癌細胞を検出する従来の方法は、患者から腫瘍組織を採取することを要するため、患者への負担が大きかった。本発明は、被験者に大きな負担をかけることなくPD-L1陽性癌細胞を検出することを目的とする。 The conventional method for detecting PD-L1-positive cancer cells requires a large burden on the patient because it requires collecting tumor tissue from the patient. An object of the present invention is to detect PD-L1-positive cancer cells without imposing a heavy burden on a subject.
 本発明は、血液試料中のPD-L1陽性癌細胞を検出する方法であって、(a)血液試料から細胞を採取する工程と、(b)細胞に、PD-L1を認識する一次抗体を接触させ、次いで一次抗体を認識する二次抗体であって蛍光色素で標識されている二次抗体を接触させる工程、又は、細胞に、PD-L1を認識する抗体であって蛍光色素で標識されている抗体を接触させる工程と、(c)細胞に蛍光色素の励起光を照射して、細胞から発せられる蛍光を検出する工程と、を備える方法を提供する。 The present invention is a method for detecting PD-L1-positive cancer cells in a blood sample, comprising: (a) collecting cells from the blood sample; and (b) a primary antibody that recognizes PD-L1 in the cells. Contacting and then contacting a secondary antibody that recognizes the primary antibody and labeled with a fluorescent dye, or a cell that is an antibody that recognizes PD-L1 and labeled with a fluorescent dye And (c) irradiating a cell with excitation light of a fluorescent dye to detect fluorescence emitted from the cell.
 上記蛍光色素は第一の蛍光色素であってよく、工程(a)の後かつ工程(c)の前の任意の段階で、(x1)細胞に、白血球のマーカータンパク質を認識する一次抗体を接触させ、次いで一次抗体を認識する二次抗体であって第二の蛍光色素で標識されている二次抗体を接触させる工程、又は、細胞に、白血球を認識する抗体であって第二の蛍光色素で標識されている抗体を接触させる工程と、(x2)細胞に、上皮細胞のマーカータンパク質を認識する一次抗体を接触させ、次いで一次抗体を認識する二次抗体であって第三の蛍光色素で標識されている二次抗体を接触させる工程、又は、細胞に、上皮細胞のマーカータンパク質を認識する抗体であって第三の蛍光色素で標識されている抗体を接触させる工程と、(x3)細胞の核を第四の蛍光色素で標識する工程と、を任意の順でさらに備えてもよく、工程(c)において、細胞に、第一、第二、第三及び第四の蛍光色素の励起光をそれぞれ照射して、細胞から発せられる第一、第二、第三、及び第四の蛍光色素の蛍光をそれぞれ検出してもよい。 The fluorescent dye may be a first fluorescent dye, and at any stage after step (a) and before step (c), (x1) a cell is contacted with a primary antibody that recognizes a leukocyte marker protein. And then contacting a secondary antibody that recognizes the primary antibody and that is labeled with a second fluorescent dye, or an antibody that recognizes leukocytes and a second fluorescent dye (X2) contacting the cell with a primary antibody that recognizes a marker protein of epithelial cells, and then a secondary antibody that recognizes the primary antibody with a third fluorescent dye. A step of contacting a labeled secondary antibody, or a step of contacting a cell with an antibody that recognizes an epithelial cell marker protein and labeled with a third fluorescent dye, and (x3) a cell The nucleus of the fourth And a step of labeling with a fluorescent dye in any order. In step (c), the cells are irradiated with excitation light of the first, second, third and fourth fluorescent dyes, respectively. The fluorescence of the first, second, third, and fourth fluorescent dyes emitted from the cells may be detected, respectively.
 PD-L1を認識する一次抗体又はPD-L1を認識する抗体は、28-8又はSP142のクローンに由来してもよい。工程(a)は、血液試料をフィルターでろ過してフィルター上に細胞を捕捉する工程であってもよい。白血球のマーカータンパク質はCD45であってよい。上皮細胞のマーカータンパク質はサイトケラチンであってよい。第一、第二、及び第三の蛍光色素は、フルオレセイン、Alexa Fluor(登録商標) 594、及びAlexa Fluor(登録商標) 647からなる群より選ばれてよく、第4の蛍光色素は4′,6-ジアミジノ-2-フェニルインドールであってよい。PD-L1陽性癌細胞は肺癌由来であってよい。工程(x1)を工程(b)の前に行い、工程(x2)及び工程(x3)を工程(b)の後に、同時に行ってもよい。 The primary antibody recognizing PD-L1 or the antibody recognizing PD-L1 may be derived from a 28-8 or SP142 clone. Step (a) may be a step of capturing a cell on a filter by filtering a blood sample with a filter. The leukocyte marker protein may be CD45. The epithelial cell marker protein may be cytokeratin. The first, second, and third fluorescent dyes may be selected from the group consisting of fluorescein, Alexa Fluor (registered trademark) 594, and Alexa Fluor (registered trademark) 647, wherein the fourth fluorescent dye is 4 ', It may be 6-diamidino-2-phenylindole. The PD-L1 positive cancer cell may be derived from lung cancer. The step (x1) may be performed before the step (b), and the step (x2) and the step (x3) may be performed simultaneously after the step (b).
 本発明の方法によれば、血液試料を使用することにより、被験者に大きな負担をかけることなく、簡単かつ短時間でPD-L1陽性癌細胞を検出することができる。 According to the method of the present invention, PD-L1-positive cancer cells can be detected easily and in a short time without imposing a heavy burden on the subject by using a blood sample.
細胞捕捉カートリッジの一実施形態を示す斜視図である。It is a perspective view which shows one Embodiment of a cell capture | acquisition cartridge. 図1におけるII-II線断面図である。FIG. 2 is a sectional view taken along line II-II in FIG. 実施例1において蛍光標識された細胞の画像である。2 is an image of cells fluorescently labeled in Example 1. FIG. 実施例2において蛍光標識された細胞の画像である。2 is an image of fluorescently labeled cells in Example 2. FIG. 実施例2において蛍光標識された細胞の画像である。2 is an image of fluorescently labeled cells in Example 2. FIG. 実施例3において蛍光標識された細胞の画像である。4 is an image of cells fluorescently labeled in Example 3. 実施例3において蛍光標識された細胞の画像である。4 is an image of cells fluorescently labeled in Example 3. 実施例4において蛍光標識された細胞の画像である。4 is an image of cells fluorescently labeled in Example 4.
 本発明の、血液試料中のPD-L1陽性癌細胞を検出する方法は、(a)血液試料から細胞を採取する工程と、(b)細胞に、PD-L1を認識する一次抗体を接触させ、次いで一次抗体を認識する二次抗体であって蛍光色素で標識されている二次抗体を接触させる工程、又は、細胞に、PD-L1を認識する抗体であって蛍光色素で標識されている抗体を接触させる工程と、(c)細胞に蛍光色素の励起光を照射して、細胞から発せられる蛍光を検出する工程と、を備える。癌患者の血液中には、血中循環癌細胞(Circulating Tumor Cell、以下、「CTC」ともいう。)と呼ばれ、血管及びリンパ管を通じて体内を循環する癌細胞が存在する場合がある。CTCは、例えば、肺、肝臓、胃、頭頸、膀胱、尿路上皮、食道、胆道、乳腺、卵巣、子宮、肝臓、前立腺、又は膵臓における癌の細胞が血管及びリンパ管に侵入したものである。したがって、これらの器官における癌組織を採取せずとも、被験者の血液試料を使用することで、PD-L1陽性癌細胞(PD-L1陽性のCTC)を検出することができる。本明細書において、「CTC」と「血液試料中の癌細胞」は同義として扱う。 The method for detecting PD-L1-positive cancer cells in a blood sample according to the present invention comprises (a) a step of collecting cells from a blood sample, and (b) contacting the cells with a primary antibody that recognizes PD-L1. Next, a step of bringing a secondary antibody that recognizes the primary antibody into contact with a secondary antibody that is labeled with a fluorescent dye, or a cell that is an antibody that recognizes PD-L1 and is labeled with a fluorescent dye A step of bringing the antibody into contact, and (c) a step of irradiating the cell with excitation light of a fluorescent dye to detect fluorescence emitted from the cell. In the blood of cancer patients, there are cases where cancer cells circulating in the body through blood vessels and lymph vessels are called circulating tumor cells (hereinafter also referred to as “CTC”). CTCs are those in which cancer cells in the lungs, liver, stomach, head and neck, bladder, urothelium, esophagus, biliary tract, breast, ovary, uterus, liver, prostate, or pancreas have entered blood vessels and lymph vessels. . Therefore, PD-L1-positive cancer cells (PD-L1-positive CTC) can be detected by using a blood sample of a subject without collecting cancer tissue in these organs. In this specification, “CTC” and “cancer cells in a blood sample” are treated as synonymous.
 血液試料としては、被験者から採取した血液をそのまま使用してもよいし、リン酸緩衝生理食塩水(PBS)等の緩衝液又はその他適当な媒体で希釈された血液を使用してもよい。血液試料には、抗凝固剤及び固定剤等、通常血液試料に添加される添加剤が添加されていてもよい。 As a blood sample, blood collected from a subject may be used as it is, or blood diluted with a buffer solution such as phosphate buffered saline (PBS) or other suitable medium may be used. The blood sample may be added with additives that are usually added to blood samples, such as anticoagulants and fixatives.
 工程(a)において、細胞は、例えば、血液試料をフィルターでろ過して、フィルター上に血液試料中の細胞を捕捉することで、血液試料から採取することができる。血液中に含まれる細胞のうち、白血球はCTCと同程度の直径を有するため、フィルター上にはCTCとともに一部の白血球が捕捉される。フィルターにより血液試料中の細胞を採取する場合、PD-L1陽性癌細胞の検出は、そのままフィルター上で行うことができる。すなわち、本発明における全ての工程は、フィルター上に捕捉された細胞に対して行うことができる。「捕捉」とは、細胞を含有する液体をフィルターでろ過して、細胞をフィルター上に残留させることを意味する。 In step (a), the cells can be collected from the blood sample by, for example, filtering the blood sample with a filter and capturing the cells in the blood sample on the filter. Among cells contained in blood, leukocytes have the same diameter as CTC, so that some leukocytes are captured together with CTC on the filter. When cells in a blood sample are collected by a filter, detection of PD-L1-positive cancer cells can be performed on the filter as it is. That is, all the steps in the present invention can be performed on the cells captured on the filter. “Capture” means that the liquid containing the cells is filtered through, leaving the cells on the filter.
 フィルターは、血液試料中に存在するCTCを捕捉できるフィルターであれば特に限定されず、従来公知のフィルターを使用できる。フィルターは、例えば、金属又は樹脂製のフィルターであってよく、基板と、基板上に設けられた、好ましくは5μm~15μm、より好ましくは6μm~12μm、さらに好ましくは7μm~10μmの孔径の貫通孔と、を有してもよい。貫通孔の孔径は、貫通孔を通過できる球の直径の最大値をいう。 The filter is not particularly limited as long as it can capture CTC present in the blood sample, and a conventionally known filter can be used. The filter may be, for example, a metal or resin filter, and is provided with a substrate and a through-hole provided on the substrate, preferably having a pore diameter of 5 μm to 15 μm, more preferably 6 μm to 12 μm, and even more preferably 7 μm to 10 μm. You may have. The hole diameter of the through hole refers to the maximum value of the diameter of a sphere that can pass through the through hole.
 本明細書において、細胞に物質を「接触させる」ことは、例えば、細胞をその物質若しくはその物質の溶液に浸すことにより行うことができる。細胞の採取にフィルターを使用する場合、細胞に反応液又は洗浄液を接触させることは、これらの溶液をフィルターでろ過することにより行うことができる。ろ過の際、細胞へのダメージを最小限に抑える観点から、溶液の流速は、50μL/分~3000μL/分が好ましく、100μL/分~1000μL/分がより好ましく、200μL/分~600μL/分がさらに好ましい。 In this specification, “contacting” a substance with a cell can be performed, for example, by immersing the cell in the substance or a solution of the substance. When using a filter for collection | recovery of a cell, contacting a reaction liquid or a washing | cleaning liquid with a cell can be performed by filtering these solutions with a filter. From the viewpoint of minimizing damage to cells during filtration, the flow rate of the solution is preferably 50 μL / min to 3000 μL / min, more preferably 100 μL / min to 1000 μL / min, and 200 μL / min to 600 μL / min. Further preferred.
 工程(a)の後、細胞を洗浄してもよい。洗浄工程は、例えば、PBS等の既知の緩衝液を含む洗浄液を、細胞に接触させることで行う。洗浄液には、牛血清アルブミン(BSA)又はエチレンジアミン四酢酸(EDTA)等の添加物が含まれていてよい。洗浄は、工程(a)の後に限らず、各工程の後に適宜行うことができる。 After the step (a), the cells may be washed. The washing step is performed, for example, by bringing a washing solution containing a known buffer solution such as PBS into contact with the cells. The washing solution may contain additives such as bovine serum albumin (BSA) or ethylenediaminetetraacetic acid (EDTA). Washing is not limited to after step (a), and can be performed appropriately after each step.
 さらに、工程(a)の後、細胞を固定化してもよい。ホルムアルデヒド等の公知の固定剤を細胞に接触させることで、細胞を固定化できる。細胞を固定化することにより、細胞の腐敗又は凝集をより軽減することができる。 Furthermore, cells may be immobilized after step (a). The cells can be fixed by contacting the cells with a known fixing agent such as formaldehyde. By fixing the cells, cell spoilage or aggregation can be further reduced.
 固定化した細胞を、次いで透過処理してもよい。公知の透過処理剤を細胞に接触させることで、細胞を透過処理することができる。透過処理剤としては、例えば、ポリ(オキシエチレン)オクチルフェニルエーテルを使用することができる。 The immobilized cells may then be permeabilized. A cell can be permeabilized by contacting the cell with a known permeabilizing agent. As the permeation treatment agent, for example, poly (oxyethylene) octylphenyl ether can be used.
 工程(b)では、細胞に、PD-L1を認識する一次抗体を接触させ、次いで一次抗体を認識する二次抗体であって蛍光色素(第一の蛍光色素)で標識されている二次抗体を接触させる(二段階の蛍光標識)。あるいは、細胞に、PD-L1を認識する抗体であって蛍光色素(第一の蛍光色素)で標識されている抗体を接触させる(一段階の蛍光標識)。この工程によりPD-L1が蛍光標識される。PD-L1の蛍光標識は、上記のように、二段階又は一段階のいずれで行ってもよい。 In the step (b), a secondary antibody that recognizes the primary antibody is contacted with a primary antibody that recognizes PD-L1, and is then labeled with a fluorescent dye (first fluorescent dye). Is contacted (two-step fluorescent labeling). Alternatively, an antibody that recognizes PD-L1 and is labeled with a fluorescent dye (first fluorescent dye) is brought into contact with cells (one-step fluorescent labeling). Through this step, PD-L1 is fluorescently labeled. As described above, PD-L1 fluorescent labeling may be performed in two steps or one step.
 PD-L1を認識する一次抗体又はPD-L1を認識する抗体であって蛍光色素(第一の蛍光色素)で標識されている抗体は、例えば、28-8、SP142、E1L3N(登録商標)、及びEPR1161(2)からなる群より選ばれるクローンに由来してもよく、ポリクローナル抗体(例えば、Prosci社のカタログ番号:4059)であってもよい。より高い感度でPD-L1陽性癌細胞を検出する観点から、PD-L1を認識する一次抗体又はPD-L1を認識する抗体は、28-8又はSP142に由来することが好ましい。また、28-8に由来する抗体又はSP142に由来する抗体を使用することで、抗体の非特異的な結合を低減することができ、したがって、偽陽性の少ない、より信頼性の高い検出結果を得ることができる。これらのクローンに由来する抗体は、いずれも抗PD-L1ウサギモノクローナル抗体である。 The primary antibody that recognizes PD-L1 or the antibody that recognizes PD-L1 and is labeled with a fluorescent dye (first fluorescent dye) is, for example, 28-8, SP142, E1L3N (registered trademark), And a clone selected from the group consisting of EPR1161 (2), or a polyclonal antibody (for example, catalog number: 4059 from Prosci). From the viewpoint of detecting PD-L1-positive cancer cells with higher sensitivity, the primary antibody that recognizes PD-L1 or the antibody that recognizes PD-L1 is preferably derived from 28-8 or SP142. In addition, by using an antibody derived from 28-8 or an antibody derived from SP142, non-specific binding of the antibody can be reduced, and thus a more reliable detection result with fewer false positives. Obtainable. The antibodies derived from these clones are all anti-PD-L1 rabbit monoclonal antibodies.
 蛍光色素(第一の蛍光色素)は、抗体の蛍光標識に通常使用される蛍光色素であれば特に限定されない。第一の蛍光色素は、例えば、Alexa Fluor(登録商標) 647又はCy(登録商標)5である。 The fluorescent dye (first fluorescent dye) is not particularly limited as long as it is a fluorescent dye usually used for fluorescent labeling of antibodies. The first fluorescent dye is, for example, Alexa Fluor (registered trademark) 647 or Cy (registered trademark) 5.
 最後に、工程(c)において、細胞に蛍光色素の励起光を照射して、細胞から発せられる蛍光を検出する。蛍光色素(第一の蛍光色素)による蛍光が検出される(陽性)細胞が、PD-L1陽性癌細胞として同定される。 Finally, in step (c), the cells are irradiated with excitation light of a fluorescent dye to detect fluorescence emitted from the cells. A cell in which fluorescence due to the fluorescent dye (first fluorescent dye) is detected (positive) is identified as a PD-L1-positive cancer cell.
 検出されたPD-L1陽性癌細胞に、その後、DNA、RNA又はタンパク質の解析を行うことができる。例えば、検出されたPD-L1陽性癌細胞に対し、シークエンサー、次世代シークエンサー、DNAチップ、マイクロアレイ、比較ゲノムハイブリダイゼーション、蛍光インサイツハイブリダイゼーション、デジタルPCR、定量逆転写PCR、ELISA、ウェスタンプロッティング、TOF-MS、MALDI-MS、ラマン分光スペクトル、クロマトグラフィー、X線結晶解析、二次元電気泳動、核磁気共鳴分光法、フローサイトメーター(FCM)等を利用した解析を行うことができる。 The detected PD-L1-positive cancer cells can then be analyzed for DNA, RNA or protein. For example, sequencer, next-generation sequencer, DNA chip, microarray, comparative genomic hybridization, fluorescence in situ hybridization, digital PCR, quantitative reverse transcription PCR, ELISA, Western plotting, TOF -MS, MALDI-MS, Raman spectroscopy, chromatography, X-ray crystallography, two-dimensional electrophoresis, nuclear magnetic resonance spectroscopy, flow cytometer (FCM), etc. can be used for analysis.
 血液試料中には、PD-L1陽性癌細胞の他に、PD-L1陰性癌細胞、白血球等のその他の細胞が多数存在する。このため、工程(b)において、PD-L1を認識する抗体がPD-L1陰性の細胞に結合し、PD-L1陰性の細胞からPD-L1を示す蛍光が観察される場合がある(偽陽性)。このような偽陽性を減らし、より信頼性の高い検出結果を得る観点から、工程(a)の後かつ工程(c)の前の任意の段階で、以下の工程(x1)~(x3)をさらに行うことが好ましい。 In addition to PD-L1-positive cancer cells, there are many other cells such as PD-L1-negative cancer cells and leukocytes in the blood sample. Therefore, in step (b), an antibody recognizing PD-L1 binds to PD-L1-negative cells, and fluorescence indicating PD-L1 may be observed from PD-L1-negative cells (false positive). ). From the viewpoint of reducing such false positives and obtaining a more reliable detection result, the following steps (x1) to (x3) are performed at any stage after step (a) and before step (c). Further, it is preferable to carry out.
 工程(x1)では、細胞に、白血球のマーカータンパク質を認識する一次抗体を接触させ、次いで一次抗体を認識する二次抗体であって第二の蛍光色素で標識されている二次抗体を接触させる(二段階の蛍光標識)。あるいは、細胞に、白血球を認識する抗体であって第二の蛍光色素で標識されている抗体を接触させる(一段階の蛍光標識)。この工程により、白血球が蛍光標識される。白血球の蛍光標識は、上記のように二段階又は一段階のいずれで行ってもよい。 In the step (x1), a cell is contacted with a primary antibody that recognizes a marker protein of leukocytes, and then a secondary antibody that recognizes the primary antibody and is labeled with a second fluorescent dye is contacted (Two-step fluorescent labeling). Alternatively, cells are contacted with an antibody that recognizes leukocytes and is labeled with a second fluorescent dye (one-step fluorescent labeling). By this step, leukocytes are fluorescently labeled. The fluorescent labeling of leukocytes may be performed in either two steps or one step as described above.
 白血球のマーカータンパク質は、例えば、全造血幹細胞に発現するCD45である。 Leukocyte marker protein is, for example, CD45 expressed in all hematopoietic stem cells.
 白血球のマーカータンパク質を認識する一次抗体、第二の蛍光色素で標識されている二次抗体、及び白血球のマーカータンパク質を認識する抗体であって第二の蛍光色素で標識されている抗体は、特に限定されず、ポリクローナル抗体又はモノクローナル抗体であってよい。抗体が由来する動物は、一次抗体が由来する動物と二次抗体が由来する動物とが異なる動物である限り、特に限定されない。 A primary antibody that recognizes a leukocyte marker protein, a secondary antibody that is labeled with a second fluorescent dye, and an antibody that recognizes a leukocyte marker protein and is labeled with a second fluorescent dye, It is not limited, A polyclonal antibody or a monoclonal antibody may be sufficient. The animal from which the antibody is derived is not particularly limited as long as the animal from which the primary antibody is derived is different from the animal from which the secondary antibody is derived.
 第二の蛍光色素は、抗体の蛍光標識に通常使用される蛍光色素であれば特に限定されない。第二の蛍光色素は、例えば、Alexa Fluor(登録商標) 594又はTexas Red(登録商標)である。第二の蛍光色素は、第一、第三及び第四の蛍光色素とは別の蛍光色素である。各蛍光色素は異なる蛍光波長を有するため、識別可能である。好ましくは、第一、第二、及び第三の蛍光色素は、フルオレセイン、Alexa Fluor 594、及びAlexa Fluor 647からなる群より選ばれ、第4の蛍光色素は4′,6-ジアミジノ-2-フェニルインドール(DAPI)である。 The second fluorescent dye is not particularly limited as long as it is a fluorescent dye usually used for fluorescent labeling of antibodies. The second fluorescent dye is, for example, Alexa Fluor (registered trademark) 594 or Texas Red (registered trademark). The second fluorescent dye is a fluorescent dye different from the first, third and fourth fluorescent dyes. Each fluorescent dye is distinguishable because it has a different fluorescence wavelength. Preferably, the first, second, and third fluorescent dyes are selected from the group consisting of fluorescein, Alexa Fluor 594, and Alexa Fluor 647, and the fourth fluorescent dye is 4 ', 6-diamidino-2-phenyl. Indole (DAPI).
 工程(x2)では、細胞に、上皮細胞のマーカータンパク質を認識する一次抗体を接触させ、次いで一次抗体を認識する二次抗体であって第三の蛍光色素で標識されている二次抗体を接触させる(二段階の蛍光標識)。あるいは、細胞に、上皮細胞のマーカータンパク質を認識する抗体であって第三の蛍光色素で標識されている抗体を接触させる(一段階の蛍光標識)。この工程により、CTCが蛍光標識される。CTCの蛍光標識は、上記のように二段階又は一段階のいずれで行ってもよい。 In step (x2), the cell is contacted with a primary antibody that recognizes a marker protein of epithelial cells, and then a secondary antibody that recognizes the primary antibody and is labeled with a third fluorescent dye is contacted (Two-step fluorescent labeling). Alternatively, cells are contacted with an antibody that recognizes a marker protein of epithelial cells and labeled with a third fluorescent dye (one-step fluorescent labeling). By this step, CTC is fluorescently labeled. CTC fluorescent labeling may be performed in either two steps or one step as described above.
 上皮細胞のマーカータンパク質としては、例えば、サイトケラチン、上皮細胞接着分子(EpCAM)、CD146、及びCD176が挙げられ、サイトケラチンが好ましい。CTCは上皮細胞に由来するため、これら上皮細胞のマーカータンパク質を有する。 Examples of epithelial cell marker proteins include cytokeratin, epithelial cell adhesion molecule (EpCAM), CD146, and CD176, with cytokeratin being preferred. Since CTC is derived from epithelial cells, it has a marker protein for these epithelial cells.
 第三の蛍光色素は、抗体の蛍光標識に通常使用される蛍光色素であれば特に限定されない。第三の蛍光色素は、例えば、フルオレセインイソチオシアネート(FITC)等のフルオレセイン又はAlexa Fluor(登録商標) 488である。 The third fluorescent dye is not particularly limited as long as it is a fluorescent dye usually used for fluorescent labeling of antibodies. The third fluorescent dye is, for example, fluorescein such as fluorescein isothiocyanate (FITC) or Alexa Fluor (registered trademark) 488.
 上皮細胞のマーカータンパク質を認識する一次抗体、第三の蛍光色素で標識されている二次抗体、及び上皮細胞のマーカータンパク質を認識する抗体であって第三の蛍光色素で標識されている抗体は、特に限定されず、ポリクローナル抗体又はモノクローナル抗体であってよい。抗体が由来する動物は、一次抗体が由来する動物と二次抗体が由来する動物とが異なる動物である限り、特に限定されない。 A primary antibody that recognizes a marker protein of epithelial cells, a secondary antibody that is labeled with a third fluorescent dye, and an antibody that recognizes a marker protein of epithelial cells and is labeled with a third fluorescent dye Without being particularly limited, it may be a polyclonal antibody or a monoclonal antibody. The animal from which the antibody is derived is not particularly limited as long as the animal from which the primary antibody is derived is different from the animal from which the secondary antibody is derived.
 工程(x3)では、細胞の核を第四の蛍光色素で標識する。核を標識する第四の蛍光色素は、核酸に結合することができる蛍光色素であれば特に限定されず、核を蛍光標識するのに通常用いられる蛍光色素を使用することができる。第四の蛍光色素としては、例えば、DAPI及び2′-(4-エトキシフェニル)-5-(4-メチル-1-ピペラジニル)-2,5′-ビ-1H-ベンゾイミダゾール三塩酸塩(Hoechst33342)が挙げられる。 In step (x3), the cell nucleus is labeled with a fourth fluorescent dye. The fourth fluorescent dye for labeling the nucleus is not particularly limited as long as it is a fluorescent dye capable of binding to a nucleic acid, and a fluorescent dye usually used for fluorescently labeling a nucleus can be used. Examples of the fourth fluorescent dye include DAPI and 2 ′-(4-ethoxyphenyl) -5- (4-methyl-1-piperazinyl) -2,5′-bi-1H-benzimidazole trihydrochloride (Hoechst 33342). ).
 任意であるこれらの工程(x1)~(x3)を行う場合、工程(c)では、細胞に、第一、第二、第三及び第四の蛍光色素の励起光をそれぞれ照射して、細胞から発せられる第一、第二、第三及び第四の蛍光色素の蛍光をそれぞれ検出する。PD-L1陽性癌細胞は、第一、第三、及び第四の蛍光色素で標識されているが、第二の蛍光色素では標識されていない。したがって、第二の蛍光色素の蛍光が検出されず(陰性)、第一、第三、及び第四の蛍光色素の蛍光が検出される(陽性)細胞が、PD-L1陽性癌細胞として同定される。 When these optional steps (x1) to (x3) are performed, in step (c), the cells are irradiated with excitation light of the first, second, third and fourth fluorescent dyes, respectively. Fluorescence of the first, second, third and fourth fluorescent dyes emitted from is detected respectively. PD-L1-positive cancer cells are labeled with the first, third, and fourth fluorescent dyes, but are not labeled with the second fluorescent dye. Therefore, cells in which the fluorescence of the second fluorescent dye is not detected (negative) and the fluorescence of the first, third, and fourth fluorescent dyes are detected (positive) are identified as PD-L1-positive cancer cells. The
 上記の方法により血液試料中のPD-L1陽性癌細胞を検出するときは、例えば、図1及び図2に示すカートリッジを用いることができる。以下、本発明の一実施形態であって、カートリッジを使用して血液試料中のPD-L1陽性癌細胞を検出する方法について述べる。別段の記載がない限り、各工程についての詳細及び工程の順番は、上記実施形態で述べたとおりである。 When detecting PD-L1-positive cancer cells in a blood sample by the above method, for example, the cartridge shown in FIGS. 1 and 2 can be used. Hereinafter, a method for detecting PD-L1-positive cancer cells in a blood sample using a cartridge according to an embodiment of the present invention will be described. Unless otherwise stated, the details of each step and the order of the steps are as described in the above embodiment.
 図1及び図2に示すCTC捕捉カートリッジ(カートリッジ)100は、液体が流入する流入管125が接続された流入口130と、液体が流出する流出管135が接続された流出口140とを有する筐体120と、フィルター105とを備える。フィルター105は、上部部材110及び下部部材115から構成される筐体120により固定されている。血液試料、洗浄液及びその他の反応液は、流入管125を通って筐体120の内部に導入され、フィルター105を通って、流出管135から外部に排出される。このような液体の流れは、例えば、流入管125の上流又は流出管135の下流にポンプを接続することにより作り出すことができる。また、流入管125の上流及び/又は流出管135の下流にコックを設け、液体の流れを制御してもよい。 A CTC capturing cartridge (cartridge) 100 shown in FIGS. 1 and 2 has a housing having an inlet 130 to which an inflow pipe 125 into which liquid flows is connected and an outlet 140 to which an outflow pipe 135 from which liquid flows out is connected. A body 120 and a filter 105 are provided. The filter 105 is fixed by a casing 120 including an upper member 110 and a lower member 115. The blood sample, the cleaning liquid, and other reaction liquids are introduced into the housing 120 through the inflow pipe 125, and are discharged to the outside through the filter 105 through the outflow pipe 135. Such a liquid flow can be created, for example, by connecting a pump upstream of the inflow pipe 125 or downstream of the outflow pipe 135. Further, a cock may be provided upstream of the inflow pipe 125 and / or downstream of the outflow pipe 135 to control the flow of the liquid.
 はじめに、血液試料を流入管125からカートリッジ100内に導入して、血液試料をフィルター105でろ過する(工程(a))。血液試料中のCTC及び一部の白血球は、フィルター105の貫通孔106を通過できず、フィルター105の表面に残留する。血液試料中のその他の成分は、貫通孔106を通過し、カートリッジ100の外へと排出される。次いで、洗浄液をフィルター105に通液してフィルター105を洗浄してもよい。フィルター105の洗浄は、以下の各工程の後にも、適宜行うことができる。 First, a blood sample is introduced into the cartridge 100 from the inflow tube 125, and the blood sample is filtered by the filter 105 (step (a)). CTC and some white blood cells in the blood sample cannot pass through the through hole 106 of the filter 105 and remain on the surface of the filter 105. Other components in the blood sample pass through the through hole 106 and are discharged out of the cartridge 100. Next, the filter 105 may be cleaned by passing a cleaning solution through the filter 105. The filter 105 can be appropriately washed after the following steps.
 さらに、フィルター105上に細胞が捕捉された後、固定剤、次いで透過処理剤を含む反応液を、それぞれカートリッジ100内に任意で導入して、カートリッジ100内に所定時間保持することで、細胞と固定剤及び透過処理剤とをそれぞれ反応させてもよい。 Further, after the cells are captured on the filter 105, a reaction solution containing a fixing agent and then a permeabilizing agent is optionally introduced into the cartridge 100, and held in the cartridge 100 for a predetermined time. A fixing agent and a permeation treatment agent may be reacted with each other.
 同様にして、細胞に、PD-L1を認識する一次抗体を含む反応液、次いで一次抗体を認識する二次抗体であって蛍光色素(第一の蛍光色素)で標識されている二次抗体を含む反応液を、それぞれフィルター105上に捕捉された細胞と反応させる。あるいは、細胞に、PD-L1を認識する抗体であって蛍光色素(第一の蛍光色素)で標識されている抗体を含む反応液を、フィルター105上に捕捉された細胞と反応させる(工程(b))。 Similarly, a reaction solution containing a primary antibody that recognizes PD-L1, and then a secondary antibody that recognizes the primary antibody and is labeled with a fluorescent dye (first fluorescent dye) are applied to the cells. The contained reaction solution is reacted with the cells captured on the filter 105. Alternatively, a reaction solution containing an antibody that recognizes PD-L1 and is labeled with a fluorescent dye (first fluorescent dye) is allowed to react with the cells captured on the filter 105 (step (step ( b)).
 最後に、蛍光顕微鏡を使用してカートリッジ100に蛍光色素の励起光を照射して、フィルター105上に捕捉された細胞から発せられる蛍光を検出する(工程(c))。蛍光の検出は、例えば、カートリッジ100の垂直方向上面からカートリッジ100を観察し、蛍光観察像を処理することにより行う。上記実施形態で述べたとおり、工程(x1)~(x3)を任意でさらに行うこともできる。 Finally, the cartridge 100 is irradiated with excitation light of a fluorescent dye using a fluorescence microscope to detect fluorescence emitted from the cells captured on the filter 105 (step (c)). The fluorescence is detected by, for example, observing the cartridge 100 from the upper surface in the vertical direction of the cartridge 100 and processing the fluorescence observation image. As described in the above embodiment, the steps (x1) to (x3) can optionally be further performed.
(実施例1)
 培養フラスコに入った非小細胞肺癌細胞株を、二酸化炭素インキュベーター内で、37℃で培養した。培養フラスコに、濃度0.25%のトリプシン-EDTAを添加し、フラスコに張り付いた培養細胞をフラスコから剥離した。剥離させた細胞を血球計算盤及び位相差顕微鏡を用いて計数した。採血管に採血した健常人の血液に、100個の細胞を添加することで、肺癌患者の血液を摸した血液試料を調製した。細胞株としては、それぞれPD-L1の発現程度の異なる、NCI-H820(PD-L1高発現)、NCI-H441(PD-L1中発現)、A549(PD-L1低発現)、及びNCI-H23(PD-L1陰性)の4種類を用いて、4種類の血液試料を準備した。採血管としては、ベクトン・ディッキンソンアンドカンパニー社製のEDTA-2K(エチレンジアミン四酢酸二カリウム塩)入り採血管を使用した。
Example 1
The non-small cell lung cancer cell line contained in the culture flask was cultured at 37 ° C. in a carbon dioxide incubator. Trypsin-EDTA with a concentration of 0.25% was added to the culture flask, and the cultured cells attached to the flask were detached from the flask. The detached cells were counted using a hemocytometer and a phase contrast microscope. A blood sample in which the blood of a lung cancer patient was sprinkled was prepared by adding 100 cells to the blood of a healthy person collected in a blood collection tube. As cell lines, NCI-H820 (high expression of PD-L1), NCI-H441 (expressed in PD-L1), A549 (low expression of PD-L1), and NCI-H23 differed in the expression level of PD-L1, respectively. Four types of blood samples were prepared using four types (PD-L1 negative). As a blood collection tube, a blood collection tube containing EDTA-2K (ethylenediaminetetraacetic acid dipotassium salt) manufactured by Becton Dickinson & Company was used.
 採血から1時間以内に、CTC捕捉装置を用いて上記4種類の血液試料中のPD-L1陽性癌細胞を以下のように検出した。CTC捕捉装置は、血液試料及びその他の反応液を導入するリザーバーと、CTC捕捉カートリッジとを備える。CTC捕捉カートリッジ(以下、カートリッジともいう)は、長径100μm、短径8μmの貫通孔を多数有する薄膜の金属フィルター(膜面積6mm×6mm、膜厚18μm)を内部に備え、上記実施形態で説明したカートリッジ100に相当する。 Within one hour after blood collection, PD-L1-positive cancer cells in the above four blood samples were detected using a CTC capture device as follows. The CTC capture device includes a reservoir for introducing a blood sample and other reaction solution, and a CTC capture cartridge. The CTC capture cartridge (hereinafter also referred to as cartridge) includes a thin-film metal filter (membrane area 6 mm × 6 mm, film thickness 18 μm) having a large number of through-holes having a major axis of 100 μm and a minor axis of 8 μm. This corresponds to the cartridge 100.
 まず、カートリッジを、0.5%BSA及び2mM EDTAを含有したPBS溶液(以下、「洗浄液」という。)で満たした。リザーバーに、洗浄液を7mL入れ、洗浄液の下に、上記血液試料を3mL、血液試料と洗浄液が層をなすように加えた。CTC捕捉装置を作動させ、流速200μL/分でリザーバー中の血液試料及び洗浄液をカートリッジに導入し、血液試料中の細胞をフィルター上に捕捉した。カートリッジに洗浄液を導入し、フィルターに残留した血液成分を洗い流した。 First, the cartridge was filled with a PBS solution containing 0.5% BSA and 2 mM EDTA (hereinafter referred to as “cleaning solution”). 7 mL of the washing solution was placed in the reservoir, and 3 mL of the blood sample was added under the washing solution so that the blood sample and the washing solution were layered. The CTC capture device was activated, the blood sample and the washing solution in the reservoir were introduced into the cartridge at a flow rate of 200 μL / min, and the cells in the blood sample were captured on the filter. A washing solution was introduced into the cartridge to wash away blood components remaining on the filter.
 1.25mLの抗ヒトCD45マウスモノクローナル抗体(クローン:2D1)を含む反応液を流速200μL/分でカートリッジに導入し、室温にて30分反応させた。1.40mLの洗浄液を流速400μL/分でカートリッジに導入し、カートリッジ内の上記反応液を排出した。1.25mLのAlexa Fluor 594標識抗マウスIgGヤギポリクロナール抗体を含む反応液を流速400μL/分でカートリッジに導入し、室温にて30分反応させた。1.40mLの洗浄液を流速400μL/分でカートリッジに導入し、カートリッジ内の上記反応液を排出した。 A reaction solution containing 1.25 mL of anti-human CD45 mouse monoclonal antibody (clone: 2D1) was introduced into the cartridge at a flow rate of 200 μL / min and reacted at room temperature for 30 minutes. 1.40 mL of the washing solution was introduced into the cartridge at a flow rate of 400 μL / min, and the reaction solution in the cartridge was discharged. A reaction solution containing 1.25 mL of Alexa Fluor 594-labeled anti-mouse IgG goat polyclonal antibody was introduced into the cartridge at a flow rate of 400 μL / min and reacted at room temperature for 30 minutes. 1.40 mL of the washing solution was introduced into the cartridge at a flow rate of 400 μL / min, and the reaction solution in the cartridge was discharged.
 ホルムアルデヒドを0.5質量%~4質量%含有するPBS溶液1.25mLを、流速400μL/分でカートリッジに導入し、室温にて10分反応させることにより、細胞を固定化した。1.40mLの洗浄液を流速400μL/分でカートリッジに導入し、カートリッジ内の上記反応液を排出した。 1.25 mL of PBS solution containing formaldehyde in an amount of 0.5% to 4% by mass was introduced into the cartridge at a flow rate of 400 μL / min and reacted at room temperature for 10 minutes to immobilize the cells. 1.40 mL of the washing solution was introduced into the cartridge at a flow rate of 400 μL / min, and the reaction solution in the cartridge was discharged.
 Triton X-100(シグマアルドリッチ社製)を0.05質量%~0.1質量%含有するPBS溶液1.25mLを、流速400μL/分でカートリッジに導入し、室温にて10分反応させることにより、細胞を透過処理した。1.40mLの洗浄液を流速400μL/分でカートリッジに導入し、カートリッジ内の上記反応液を排出した。 By introducing 1.25 mL of a PBS solution containing 0.05 mass% to 0.1 mass% of Triton X-100 (manufactured by Sigma Aldrich) into the cartridge at a flow rate of 400 μL / min and reacting at room temperature for 10 minutes. The cells were permeabilized. 1.40 mL of the washing solution was introduced into the cartridge at a flow rate of 400 μL / min, and the reaction solution in the cartridge was discharged.
 1.25mLの抗ヒトPD-L1ウサギモノクローナル抗体(クローン:28-8)を含む反応液を流速200μL/分でカートリッジに導入し、室温にて60分反応させた。1.40mLの洗浄液を流速400μL/分でカートリッジに導入し、カートリッジ内の上記反応液を排出した。1.25mLのAlexa Fluor 647標識抗ウサギIgGヤギポリクロナール抗体を含む反応液を流速400μL/分でカートリッジに導入し、室温にて30分反応させた。1.40mLの洗浄液を流速400μL/分でカートリッジに導入し、カートリッジ内の上記反応液を排出した。 A reaction solution containing 1.25 mL of anti-human PD-L1 rabbit monoclonal antibody (clone: 28-8) was introduced into the cartridge at a flow rate of 200 μL / min and reacted at room temperature for 60 minutes. 1.40 mL of the washing solution was introduced into the cartridge at a flow rate of 400 μL / min, and the reaction solution in the cartridge was discharged. A reaction solution containing 1.25 mL of Alexa Fluor 647-labeled anti-rabbit IgG goat polyclonal antibody was introduced into the cartridge at a flow rate of 400 μL / min and reacted at room temperature for 30 minutes. 1.40 mL of the washing solution was introduced into the cartridge at a flow rate of 400 μL / min, and the reaction solution in the cartridge was discharged.
 FITC標識抗ヒトサイトケラチンマウスモノクローナル抗体(クローン:CK3、6H5、AE1、及びAE3の混合物)、及びDAPIを含む反応液1.25mLを、400μL/分でカートリッジに導入し、室温にて30分反応させた。3.00mLの洗浄液を流速400μL/分でカートリッジに導入し、カートリッジ内の上記反応液を排出した。次いで、カートリッジをCTC捕捉装置から外した。 1.25 mL of a reaction solution containing FITC-labeled anti-human cytokeratin mouse monoclonal antibody (clone: mixture of CK3, 6H5, AE1, and AE3) and DAPI is introduced into the cartridge at 400 μL / min and reacted at room temperature for 30 minutes. I let you. 3.00 mL of the cleaning solution was introduced into the cartridge at a flow rate of 400 μL / min, and the reaction solution in the cartridge was discharged. The cartridge was then removed from the CTC capture device.
 カートリッジを蛍光顕微鏡に設置した。蛍光ミラーユニットを使用して、細胞上の蛍光色素(FITC、Alexa Fluor 594、Alexa Fluor 647、及びDAPI)をそれぞれ励起させた。それぞれの蛍光色素から発せられた蛍光を撮影し、得られた画像を合成した。 The cartridge was set on a fluorescence microscope. Fluorescent mirror units (FITC, Alexa Fluor 594, Alexa Fluor 647, and DAPI) were each excited using a fluorescent mirror unit. The fluorescence emitted from each fluorescent dye was photographed, and the resulting images were synthesized.
 結果を図3に示す。図中、H820はNCI-H820、H441はNCI-H441、H23はNCI-H23を、それぞれ意味する(以下、同じ)。PD-L1陽性の癌細胞株であるNCI-H820及びNCI-H441を用いた実験においては、細胞核(DAPI)、サイトケラチン(FITC)、及びPD-L1(Alexa Fluor 647)が陽性であり、かつ、CD45(Alexa Fluor 594)が陰性である細胞の蛍光画像が得られた。PD-L1陰性の癌細胞株であるNCI-H23を用いた実験においては、細胞核及びサイトケラチンが陽性であり、かつ、CD45及びPD-L1が陰性である細胞の蛍光画像が得られた。一方、いずれの実験においても、細胞核及びCD45が陽性であり、かつ、サイトケラチン及びPD-L1が陰性である、白血球の蛍光画像も得られた(図には示していない)。抗体の非特異的な結合による偽陽性はみられなかった。なお、「陽性」とは蛍光が検出されたことを意味し、「陰性」とは蛍光が検出されなかったことを意味する。 The results are shown in FIG. In the figure, H820 means NCI-H820, H441 means NCI-H441, and H23 means NCI-H23 (hereinafter the same). In experiments using NCI-H820 and NCI-H441, which are PD-L1-positive cancer cell lines, the cell nucleus (DAPI), cytokeratin (FITC), and PD-L1 (Alexa Fluor 647) are positive, and , A fluorescent image of cells negative for CD45 (Alexa Fluor 594) was obtained. In an experiment using NCI-H23, which is a PD-L1-negative cancer cell line, fluorescence images of cells positive for cell nuclei and cytokeratin and negative for CD45 and PD-L1 were obtained. On the other hand, in each experiment, a leukocyte fluorescence image in which cell nuclei and CD45 were positive and cytokeratin and PD-L1 were negative was also obtained (not shown). There were no false positives due to nonspecific binding of antibodies. Note that “positive” means that fluorescence was detected, and “negative” means that fluorescence was not detected.
(実施例2)
 細胞株としてNCI-H820を用いた。抗ヒトPD-L1ウサギモノクローナル抗体のクローンをSP142に変更した以外は実施例1と同様の方法により、細胞の蛍光を観察した。
(Example 2)
NCI-H820 was used as a cell line. Cell fluorescence was observed in the same manner as in Example 1 except that the clone of the anti-human PD-L1 rabbit monoclonal antibody was changed to SP142.
 結果を図4及び図5に示す。これらの図に示すように、細胞核、サイトケラチン、及びPD-L1が陽性であり、かつ、CD45が陰性である細胞の蛍光画像が得られた。また、図5の右下には、細胞核及びCD45が陽性であり、かつ、サイトケラチン及びPD-L1が陰性である、白血球がみられる。抗体の非特異的な結合による偽陽性はみられなかった。 The results are shown in FIGS. As shown in these figures, fluorescence images of cells positive for cell nuclei, cytokeratin, and PD-L1, and negative for CD45 were obtained. In the lower right of FIG. 5, leukocytes that are positive for cell nuclei and CD45, and negative for cytokeratin and PD-L1 are seen. There were no false positives due to nonspecific binding of antibodies.
(実施例3)
 細胞株としてNCI-H820を用いた。抗ヒトPD-L1ウサギモノクローナル抗体を、E1L3N(登録商標)由来の抗体、EPR1161(2)由来の抗体、及びポリクローナル抗体(Prosci社製、カタログ番号:4059)に変更した以外は実施例1と同様の方法により、細胞の蛍光を観察した。結果を図6及び図7に示す。
(Example 3)
NCI-H820 was used as a cell line. The same as Example 1 except that the anti-human PD-L1 rabbit monoclonal antibody was changed to an antibody derived from E1L3N (registered trademark), an antibody derived from EPR1161 (2), and a polyclonal antibody (manufactured by Prosci, catalog number: 4059). The fluorescence of the cells was observed by this method. The results are shown in FIGS.
 図6及び図7に示すように、いずれの抗体を用いた実験においても、細胞核、サイトケラチン、及びPD-L1が陽性であり、かつ、CD45が陰性である細胞の蛍光画像が得られた。PD-L1の蛍光強度は、E1L3N由来の抗体を用いた実験において高く、ポリクローナル抗体を用いた実験において低かった。EPR1161(2)由来の抗体を用いた実験においては、PD-L1の蛍光は僅かに確認できる程度であった。 As shown in FIG. 6 and FIG. 7, in the experiments using any of the antibodies, fluorescence images of cells that were positive for cell nucleus, cytokeratin, and PD-L1, and negative for CD45 were obtained. The fluorescence intensity of PD-L1 was high in the experiment using the antibody derived from E1L3N and low in the experiment using the polyclonal antibody. In an experiment using an antibody derived from EPR1161 (2), the fluorescence of PD-L1 was only slightly confirmed.
 図7に示すように、いずれの抗体を用いた実験においても、細胞核及びCD45が陽性であり、かつ、サイトケラチン及びPD-L1が陰性である、白血球がみられる。一方、図7は、いずれの抗体を用いた実験においても、抗体の非特異的な結合が観察されたことをも示す。 As shown in FIG. 7, in the experiment using any of the antibodies, cell nuclei and white blood cells that are positive for CD45 and negative for cytokeratin and PD-L1 are observed. On the other hand, FIG. 7 also shows that non-specific binding of the antibody was observed in the experiment using any antibody.
(実施例4)
 PD-L1に対する二次抗体(抗ウサギIgGヤギポリクロナール抗体)の標識色素をAlexa Fluor 680及びAlexa Fluor(登録商標) 700に変更した以外は実施例1と同様の方法により、細胞の蛍光を観察した。
Example 4
Cell fluorescence was observed in the same manner as in Example 1 except that the labeling dye of the secondary antibody (anti-rabbit IgG goat polyclonal antibody) against PD-L1 was changed to Alexa Fluor 680 and Alexa Fluor (registered trademark) 700. .
 結果を図8に示す。Alexa Fluor 680及びAlexa Fluor 700を用いた場合、PD-L1の蛍光強度が低かった。 The results are shown in FIG. When Alexa Fluor 680 and Alexa Fluor 700 were used, the fluorescence intensity of PD-L1 was low.
 100…CTC捕捉カートリッジ、105…フィルター、106…貫通孔、110…上部部材、115…下部部材、120…筐体、125…流入管、130…流入口、135…流出管、140…流出口。 DESCRIPTION OF SYMBOLS 100 ... CTC capture cartridge, 105 ... Filter, 106 ... Through hole, 110 ... Upper member, 115 ... Lower member, 120 ... Housing, 125 ... Inflow pipe, 130 ... Inlet, 135 ... Outlet, 140 ... Outlet

Claims (9)

  1.  血液試料中のPD-L1陽性癌細胞を検出する方法であって、
     (a)血液試料から細胞を採取する工程と、
     (b)細胞に、PD-L1を認識する一次抗体を接触させ、次いで一次抗体を認識する二次抗体であって蛍光色素で標識されている二次抗体を接触させる工程、又は、細胞に、PD-L1を認識する抗体であって蛍光色素で標識されている抗体を接触させる工程と、
     (c)細胞に蛍光色素の励起光を照射して、細胞から発せられる蛍光を検出する工程と、を備える
    方法。
    A method for detecting PD-L1-positive cancer cells in a blood sample, comprising:
    (A) collecting cells from a blood sample;
    (B) contacting a cell with a primary antibody that recognizes PD-L1, and then contacting a secondary antibody that recognizes the primary antibody and labeled with a fluorescent dye, or the cell, Contacting an antibody that recognizes PD-L1 and labeled with a fluorescent dye;
    (C) irradiating a cell with excitation light of a fluorescent dye, and detecting fluorescence emitted from the cell.
  2.  前記蛍光色素が第一の蛍光色素であり、
     工程(a)の後かつ工程(c)の前の任意の段階で、
      (x1)細胞に、白血球のマーカータンパク質を認識する一次抗体を接触させ、次いで一次抗体を認識する二次抗体であって第二の蛍光色素で標識されている二次抗体を接触させる工程、又は、細胞に、白血球を認識する抗体であって第二の蛍光色素で標識されている抗体を接触させる工程と、
      (x2)細胞に、上皮細胞のマーカータンパク質を認識する一次抗体を接触させ、次いで一次抗体を認識する二次抗体であって第三の蛍光色素で標識されている二次抗体を接触させる工程、又は、細胞に、上皮細胞のマーカータンパク質を認識する抗体であって第三の蛍光色素で標識されている抗体を接触させる工程と、
      (x3)細胞の核を第四の蛍光色素で標識する工程と、
    を任意の順でさらに備え、
     工程(c)において、細胞に、第一、第二、第三及び第四の蛍光色素の励起光をそれぞれ照射して、細胞から発せられる第一、第二、第三、及び第四の蛍光色素の蛍光をそれぞれ検出する、請求項1に記載の方法。
    The fluorescent dye is a first fluorescent dye;
    At any stage after step (a) and before step (c),
    (X1) contacting a cell with a primary antibody that recognizes a leukocyte marker protein, and then contacting a secondary antibody that recognizes the primary antibody and labeled with a second fluorescent dye, or Contacting the cell with an antibody that recognizes leukocytes and is labeled with a second fluorescent dye;
    (X2) contacting a cell with a primary antibody that recognizes a marker protein of epithelial cells, and then a secondary antibody that recognizes the primary antibody and labeled with a third fluorescent dye, Or contacting a cell with an antibody that recognizes a marker protein of epithelial cells and labeled with a third fluorescent dye;
    (X3) labeling the cell nucleus with a fourth fluorescent dye;
    Further in any order,
    In the step (c), the cells are irradiated with excitation light of the first, second, third and fourth fluorescent dyes, respectively, and the first, second, third and fourth fluorescence emitted from the cells. The method according to claim 1, wherein fluorescence of each dye is detected.
  3.  PD-L1を認識する一次抗体又はPD-L1を認識する抗体が、28-8又はSP142のクローンに由来する、請求項2に記載の方法。 The method according to claim 2, wherein the primary antibody that recognizes PD-L1 or the antibody that recognizes PD-L1 is derived from a clone of 28-8 or SP142.
  4.  工程(a)が、血液試料をフィルターでろ過してフィルター上に細胞を捕捉する工程である、請求項2又は3に記載の方法。 The method according to claim 2 or 3, wherein step (a) is a step of filtering a blood sample with a filter to capture cells on the filter.
  5.  白血球のマーカータンパク質がCD45である、請求項2~4のいずれか一項に記載の方法。 The method according to any one of claims 2 to 4, wherein the leukocyte marker protein is CD45.
  6.  上皮細胞のマーカータンパク質がサイトケラチンである、請求項2~5のいずれか一項に記載の方法。 The method according to any one of claims 2 to 5, wherein the marker protein of epithelial cells is cytokeratin.
  7.  第一、第二、及び第三の蛍光色素が、フルオレセイン、Alexa Fluor(登録商標) 594、及びAlexa Fluor(登録商標) 647からなる群より選ばれ、第4の蛍光色素が4′,6-ジアミジノ-2-フェニルインドールである、請求項2~6のいずれか一項に記載の方法。 The first, second, and third fluorescent dyes are selected from the group consisting of fluorescein, Alexa Fluor (registered trademark) 594, and Alexa Fluor (registered trademark) 647, and the fourth fluorescent dye is 4 ', 6- The method according to any one of claims 2 to 6, which is diamidino-2-phenylindole.
  8.  PD-L1陽性癌細胞が肺癌由来である、請求項2~7のいずれか一項に記載の方法。 The method according to any one of claims 2 to 7, wherein the PD-L1-positive cancer cells are derived from lung cancer.
  9.  工程(x1)を工程(b)の前に行い、
     工程(x2)及び工程(x3)を工程(b)の後に、同時に行う、請求項2~8のいずれか一項に記載の方法。
    Performing step (x1) before step (b),
    The method according to any one of claims 2 to 8, wherein the step (x2) and the step (x3) are simultaneously performed after the step (b).
PCT/JP2018/015274 2017-04-13 2018-04-11 Method for detecting pd-l1-positive cancer cells WO2018190382A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2019512558A JPWO2018190382A1 (en) 2017-04-13 2018-04-11 Method for detecting PD-L1-positive cancer cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2017-080045 2017-04-13
JP2017080045 2017-04-13

Publications (1)

Publication Number Publication Date
WO2018190382A1 true WO2018190382A1 (en) 2018-10-18

Family

ID=63792606

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2018/015274 WO2018190382A1 (en) 2017-04-13 2018-04-11 Method for detecting pd-l1-positive cancer cells

Country Status (2)

Country Link
JP (1) JPWO2018190382A1 (en)
WO (1) WO2018190382A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011163830A (en) * 2010-02-05 2011-08-25 Tokyo Univ Of Agriculture & Technology Detection of circulation tumor cells using size-selective micro cavity array
WO2015174539A1 (en) * 2014-05-13 2015-11-19 学校法人順天堂 Method for detecting cells
WO2016035772A1 (en) * 2014-09-03 2016-03-10 日立化成株式会社 Filter for capturing biological substance, and system for capturing biological substance
JP2016086736A (en) * 2014-11-05 2016-05-23 日立化成株式会社 Production method of liquid containing rare cells in blood
WO2018029858A1 (en) * 2016-08-12 2018-02-15 日立化成株式会社 Detection method of circulating tumor cells and pretreatment method for detecting circulating tumor cells
KR20180048215A (en) * 2016-11-02 2018-05-10 주식회사 싸이토젠 A method for identifying a subject with cancer for pd-l1 targeted immune therapy with circulating tumor cells

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011163830A (en) * 2010-02-05 2011-08-25 Tokyo Univ Of Agriculture & Technology Detection of circulation tumor cells using size-selective micro cavity array
WO2015174539A1 (en) * 2014-05-13 2015-11-19 学校法人順天堂 Method for detecting cells
WO2016035772A1 (en) * 2014-09-03 2016-03-10 日立化成株式会社 Filter for capturing biological substance, and system for capturing biological substance
JP2016086736A (en) * 2014-11-05 2016-05-23 日立化成株式会社 Production method of liquid containing rare cells in blood
WO2018029858A1 (en) * 2016-08-12 2018-02-15 日立化成株式会社 Detection method of circulating tumor cells and pretreatment method for detecting circulating tumor cells
KR20180048215A (en) * 2016-11-02 2018-05-10 주식회사 싸이토젠 A method for identifying a subject with cancer for pd-l1 targeted immune therapy with circulating tumor cells

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
KANBAYASHI, SATOSHI: "Review on PD-L1 expression in circulating-tumour cell with respect to lung cancer", PROCEEDINGS OF THE JAPANESE LUNG CANCER SOCIETY, vol. 56, no. 6, 5 November 2016 (2016-11-05), pages 802 *
KIM, WOONG: "Differential expression of PD-L1 on circulating tumor cells among patients with advanced lung cancer", CANCER RESEARCH, vol. 76, no. suppl. 14, July 2016 (2016-07-01), pages 2257 *
KO, YASUHIRO: "Circulating tumor cells and their PD-L1 expression", JOURNAL OF MOLECULAR TARGETED THERAPY FOR CANCER, vol. 14, no. 2, 8 July 2016 (2016-07-08), pages 228 - 232 , 168, 231 *
KO, YASUHIRO: "Diachronic analysis on PD-L1 positive circulating-tumor cells in blood with respect to non-small cell lung cancer patient who received nivolumab therapy", THE JOURNAL OF THE WAKAYAMA MEDICAL SOCIETY, vol. 69, no. 1, 31 March 2018 (2018-03-31), pages 74 - 75 *
KO, YASUHIRO: "Measurement of PD-L1 expression in circulating- tumour cell with respect to ongoing lung cancer", ABSTRACTS OF ACADEMIC CONFERENCE PROGRAM OF JAPANESE SOCIETY OF MEDICAL ONCOLOGY, vol. 14 th, 2016, pages 2 - 145 *
PANIEL, KLAUS: "Circulating tumor cells: Detection, Biology and Clinical Implications", PROGRAM ABSTRACTS OF MEETING OF THE JAPANESE ASSOCIATION FOR METASTASIS RESEARCH, vol. 26 th, 2017, pages 62 *
TANAKA, AYAKA: "Evaluation of a novel automated device for size-based enrichment and isolation of CTCs in patients with advanced lung cancer", ANNALS OF THE JAPANESE RESPIRATORY SOCIETY, vol. 5, 2016, pages 373 *
TERAOKA, SHUNSUKE: "Diachronic analysis on PD-L1 positive circulating-tumor cells in blood with respect to non-small cell lung cancer patient who received nivolumab therapy", PROCEEDINGS OF THE JAPANESE LUNG CANCER SOCIETY, vol. 57, no. 5, 5 September 2017 (2017-09-05), pages 607 *

Also Published As

Publication number Publication date
JPWO2018190382A1 (en) 2020-02-27

Similar Documents

Publication Publication Date Title
JP6982327B2 (en) Methods, compositions and systems for microfluidic assays
US20220357330A1 (en) Method for detecting tumor cell surface marker molecule pd-l1
JP6639906B2 (en) Biological sample detection method
US20190078153A1 (en) Method of analyzing genetically abnormal cells
US20140106388A1 (en) Automated ctc detection
JP6936984B2 (en) How to Predict the Prognosis of Cancer Patients Using Rare Cells
JP7040816B2 (en) Androgen receptor mutant-based screening method for prostate cancer patients
JP6617516B2 (en) Method for detecting target cells contained in blood sample
WO2018029858A1 (en) Detection method of circulating tumor cells and pretreatment method for detecting circulating tumor cells
WO2018190382A1 (en) Method for detecting pd-l1-positive cancer cells
CN109752308A (en) Cell detection method and cell detecting system
CN212560306U (en) Enrichment device of circulating tumor cells
JP6913977B2 (en) Prostate Specific Membrane Antigen-Based Prostate Cancer Patient Screening Method
WO2018030547A1 (en) Detection method of circulating tumor cells and pretreatment method for detecting circulating tumor cells
WO2018116465A1 (en) Method for detecting her2-positive cancer cells
WO2018190379A1 (en) Method for predicting efficacy of immune checkpoint inhibitor on test subject
Lee et al. Enrichment of circulating tumor cells using a centrifugal affinity plate system
WO2018047311A1 (en) Pretreatment agent for detecting circulating tumor cells
JP2019056678A (en) Target cell detection method
US20180095086A1 (en) Standard cell suspension
Choi et al. A portable microfluidic chip system for cancer diagnosis with simultaneous detection methods
US20170080142A1 (en) Filtration of circulating tumor cells for theraputic purposes

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: 18784446

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019512558

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18784446

Country of ref document: EP

Kind code of ref document: A1