METHOD FOR DETECTING TUMOR CELL SURFACE MARKER MOLECULE PD-L1

Abstract
A method for detecting a tumor cell surface marker molecule PD-L1, comprising the following steps: providing a capture screen that has antibodies capable of specifically binding to tumor cells; making a sample to be tested flow through the capture screen, such that tumor cells in the sample to be tested bind to the capture screen; fixing captured tumor cells on the capture screen; and successively using a PD-L1 primary antibody solution, a PD-L1 secondary antibody solution labeled with a fluorophore AlexaFluor 647, a pan-CK-AlexaFluor 488 primary antibody solution, a CD45 primary antibody solution and a CD45 secondary antibody solution labeled with a fluorophore AlexaFluor 568, to incubate the cells fixed on the capture screen, and then labeling all cells on the capture screen with a nuclear fluorescent dye.
Description
TECHNICAL FIELD

The present disclosure relates to the biotechnology field, in particular to a method for detecting PD-L1 which is a tumor cell surface marker molecule.


BACKGROUND

Programmed death protein-1 (PD-1) is the main immune checkpoint receptor, and by binding to its ligand, Programmed Death Ligand-1 (PD-L1), it can down-regulate the effector function of T cells, thereby helping to maintain tolerance to tumor cells. There are currently three main inhibitors for PD-1 and PD-L1 on the market, namely pembrolizumab (trade name: Keytruda), Nibolumab (trade name: Opdivo) and Atezolizumab (trade name: Tecentriq), which can be used for the treatment of various cancers such as melanoma, non-small cells lung cancer and bladder cancer. However, not all patients can benefit from the treatment of PD-1/PD-L1 inhibitors, PD-1/PD-L1 inhibitors are currently only able to produce lasting tumor control effects in a small number of cancer patients. Therefore, detecting whether the patient has positive expression of PD-L1 can effectively help the patient choose the appropriate medicine for treatment. At present, the detection methods of PD-1/PD-L1 are mainly based on the detection of cellular protein levels, and in clinical practice, immunohistochemical methods are mainly used, and tumor tissues obtained after surgery or puncture are used for section staining. The results of immunohistochemistry are closely related to the experience of the pathologist. Therefore, a new non-invasive evaluation method and standard, and a relatively stable PD-L1 detection method are urgently needed.


Circulating tumor cells fall off from the primary tumor, enter the blood circulation, settle in remote organs or primary organs, and form metastasis foci. As a hot field of liquid biopsy, circulating tumor cell (CTC) detection has gradually emerged in the clinical manifestations such as tumor diagnosis, treatment and monitoring, and is currently the most promising non-invasive tumor diagnosis and real-time efficacy monitoring method, and has extremely significant clinical application value. Therefore, it will be of great significance if a method for detecting PD-L1 on the surface of tumor cells such as circulating tumor cells can be provided.


For example, Chinese patent CN201810312287.X discloses a method for detecting circulating tumor cell surface marker molecule PD-L1, which comprises the following steps: (1) treating whole blood with the red cell lysing solution to separate nucleated cells and fixing them with formaldehyde; (2) first positive screening by tumor immunofluorescence marker cytokeratin antibody anti-CK, incubating all cells with PD-L1 antibody, then incubating all cells with PD-L1 secondary antibody labeled with FITC fluorophore, and then labeling all cells with nuclear fluorescent dye DAPI; (3) using high-throughput multicolor imaging analysis, selecting CY5, FITC and DAPI filters, observing the fluorescence color of the channel surface, to finally achieve the detection of the circulating tumor cell surface marker molecule PD-L1. This detection method uses whole blood to be processed with red cell lysing solution to separate nucleated cells, but fails to directly separate CTC cells, where the background cells are complex and it is difficult to ensure the accuracy of the detection.


Another example is the Chinese patent CN201610705258.0 discloses a method for detecting the PDL1 gene of circulating tumor cells in the peripheral blood of patients with non-small cell lung cancer, which comprises the following steps: (1) membrane filtering the peripheral blood samples from patients with non-small cell lung cancer, to obtain circulating tumor cells in the peripheral blood; (2) fixing the filter membrane obtained in step 1; (3) detecting the filter membrane obtained in step 2 to determine the expression of PD-L1. This detection method cannot accurately obtain CTCs with PD-L1 expression only by relying on these three steps, and must be combined with HE staining and expert reading to accurately identify CTCs. On the one hand, such steps are cumbersome and complicated; on the other hand, expert reading not only has a strong subjectivity to affect the judgment result, but also has strong professionalism to make it difficult to be promoted and used.


SUMMARY

The present disclosure is aimed to provide a novel method for detecting a tumor cell surface marker molecule PD-L1, which improves detection accuracy and is easier in terms of detection operation.


To achieve the above-mentioned purposes, the technical solution employed by the present disclosure is:


a method for detecting a tumor cell surface marker molecule PD-L1, comprises the following steps:


A—providing a capture screen that comprises a mesh substrate and EpCAM antibodies formed on the mesh substrate by means of incubation;


B—making nucleated cells separated from a body fluid flow through the capture screen, such that tumor cells in the nucleated cells bind to the capture screen;


C—fixing the captured tumor cells on the capture screen by using formaldehyde;


D—successively using a PD-L1 primary antibody solution, a PD-L1 secondary antibody solution labeled with a fluorophore AlexaFluor 647, a pan-CK-AlexaFluor 488 primary antibody solution, a CD45 primary antibody solution and a CD45 secondary antibody solution labeled with a fluorophore AlexaFluor 568, to incubate the cells fixed on the capture screen, and then labeling all cells on the capture screen with a nuclear fluorescent dye.


According to some preferred implementation aspects of the present disclosure, in step A, the mesh substrate comprises a stainless-steel body and a protective layer covering the surface of the stainless-steel body, the protective layer is made of a precious metal or an alloy thereof, and the EpCAM antibodies are arranged on the protective layer.


According to some preferred implementation aspects of the present disclosure, the EpCAM antibodies are attached to the protective layer via Traut's reagent or thiolate molecules with biotin-avidin.


According to some preferred implementation aspects of the present disclosure, the mesh substrate has a size of 2-10 mm×2-10 mm, and the screen has pores of 20 μm-100 μm.


According to some preferred implementation aspects of the present disclosure, in step B, after the nucleated cells flow through the capture screen and tumor cells bind to the capture screen, the capture screen is washed with a cell washing solution to remove debris or cells that are not bound to the capture screen.


According to some preferred implementation aspects of the present disclosure, in step B, the nucleated cells are separated from blood, urine or peritoneal fluid, and the above-mentioned body fluid is blood, urine or peritoneal fluid. In a specific embodiment, the nucleated cells are separated from whole blood, and the specific implementation is as follows: processing a collected whole blood with red blood cell lysing solution or lymphocyte separation solution to separate nucleated cells. The tumor cells captured by the capture screen are circulating tumor cells (CTCs) or urinary tumor cells (UTCs). Among them, circulating tumor cells refer to tumor cells shed into the blood circulation from the primary foci or metastasis of solid tumors; urinary tumor cells refer to tumor cells that enter the urine.


According to some preferred implementation aspects of the present disclosure, in step C, the capture screen bound with the tumor cells is placed in a 4% paraformaldehyde solution, fixed at room temperature for 10˜60 min, and washed with phosphate buffer.


According to some preferred implementation aspects of the present disclosure, in step D, the cells is specifically incubated as follows:


adding a PD-L1 primary antibody solution at a ratio of 1:(50˜1000), incubating the cells fixed on the capture screen at room temperature for 20˜80 min, and then washing with phosphate buffer;


adding a PD-L1 secondary antibody solution labeled with fluorophore AlexaFluor 647 at a ratio of 1:(50˜1000), incubating at room temperature for 20˜80 min, and then washing with phosphate buffer;


adding a pan-CK-FITC primary antibody solution at a ratio of 1:(50˜1000), incubating the cells fixed on the capture screen at room temperature for 20˜80 min, and then washing with phosphate buffer;


adding a CD45 primary antibody solution at a ratio of 1:(50˜1000), incubating the cells fixed on the capture screen at room temperature for 20˜80 min, and then washing with phosphate buffer;


adding a CD45 secondary antibody solution labeled with fluorophore AlexaFluor 568 at a ratio of 1:(50˜1000), incubating at room temperature for 20˜80 min, and then washing with phosphate buffer.


According to some preferred implementation aspects of the present disclosure, volume ratios of the antibody diluent and the antibody stock solution in the PD-L1 primary antibody solution, the PD-L1 secondary antibody solution labeled with fluorophore AlexaFluor 647, the pan-CK-AlexaFluor 488 primary antibody solution, the CD45 primary antibody solution and the CD45 secondary antibody solution labeled with fluorophore AlexaFluor 568 are (50˜1000):1.


According to some preferred implementation aspects of the present disclosure, in step D, all the cells on the capture screen are labeled with the nuclear fluorescent dye DAPI.


According to some preferred implementation aspects of the present disclosure, the detection method further comprises the following step:


E—observing fluorescence color of each channel to detect the tumor cell surface marker molecule PD-L1, through high-throughput multicolor imaging analysis using filters of CY5, FITC, PE and DAPI.


Here, the term “room temperature” refers to 20˜25° C.


Due to the use of the above solutions, the present disclosure has the following advantages over the prior art:


using a capture screen to capture tumor cells in body fluids (such as blood, urine or peritoneal fluid) and then incubating and detecting through PD-L1 and other antibodies, effectively avoids the effects of various other cells, cytokines and proteins in body fluid on the incubation and detection of PD-L1 and other antibodies, which improves detection accuracy and reliability, and has better specificity; and the capture screen is directly used as the carrier for incubating and detecting the captured and separated tumor cells, which is simple and convenient.





BRIEF DESCRIPTION OF THE DRAWINGS

For more clearly explaining the technical solutions in the embodiments of the present disclosure, the accompanying drawings used to describe the embodiments are simply introduced in the following. Apparently, the below described drawings merely show a part of the embodiments of the present disclosure, and those skilled in the art can obtain other drawings according to the accompanying drawings without creative work.



FIG. 1a and FIG. 1b are schematic diagrams of detecting positive and negative expression of CK in tumor cells by a FITC filter;



FIG. 2a and FIG. 2b are schematic diagrams of detecting positive and negative expression of PD-L1 in tumor cells by a PD-L1 (primary antibody)-Alexa Fluor 647 (secondary antibody) filter;



FIG. 3a and FIG. 3b are schematic diagrams of detecting negative expression of CK45 in tumor cells by a PE filter;



FIG. 4a and FIG. 4b are schematic diagrams of detecting whether tumor cells are nucleated cells by a DAPI filter;



FIG. 5a and FIG. 5b are schematic diagrams of detecting the positive and negative expression of PD-L1 in tumor cells by combining four kinds of filters.





DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS

In the following, the preferable embodiments of the present disclosure are explained in detail combining with the accompanying drawings so that the advantages and features of the present disclosure can be easily understood by the skilled persons in the art. It should be noted that the explanation on these implementations is to help understanding of the present disclosure, and is not intended to limit the present disclosure. Further, the technical features involved in the various embodiments of the present disclosure described below may be combined with each other as long as they do not conflict with each other.


Embodiment 1

This embodiment detected the tumor cell surface marker molecule PD-L1, which was specifically carried out as follows.


(1) Providing the capture screen


The capture screen comprises a stainless-steel body and a protective layer covering the surface of the stainless-steel body. The material of the protective layer is gold or gold alloy (such as, AuPd), and the EpCAM antibodies are arranged on the protective layer. The protective layer is an AuPd layer deposited on the stainless-steel body by magnetron sputtering or electrochemical methods. The EpCAM antibodies was attached to the protective layer via Traut's reagent, and thiolate molecules with biotin-avidin can be replace the Traut's reagent.


The specific preparation process of the above-mentioned capture screen was as follows.


Selection of screen: gold-coated stainless-steel screen: 51 μm holes were chosen and magnetron sputtering was used to coat AuPd, the size of the screen was 2×2 mm2. Pre-functionalization: various cleaning methods were used to prepare the screen before functionalization, including high-pressure steam sterilization, oxygen plasma cleaning, and ultrasonic cleaning in a variety of solutions, including piranha solutions, ammonia-hydrogen peroxide mixing liquid. For example, the screen was ultrasonic treated in a detergent for 15 minutes, rinsed, ultrasonic treated in 70% ˜99% ethanol solution for 15 minutes, and rinsed with high purity water for 5 minutes.


EpCAM antibodies: 2˜10 μl of EpCAM antibodies were taken and freezed, and then used to prepare a reaction mixture (i.e., EpCAM antibodies+PBS with EDTA).


Traut's Reagent: it was freezed quickly after purchase. The volume ratio of Traut's reagent and EpCAM antibodies was 10˜20:1. Other methods, such as thiolate molecules with biotin-avidin may replace the Traut's reagent to connect to the screen to form capture screen.


Incubation time of Traut's reagent with antibody: the best reaction time is 1 hour. Incubation of the screen in the above solution: the screen with the above incubated antibody-containing Traut's reagent was incubated at 4° C., room temperature or 37° C. for 10 minutes to within 12 hours, so that the EpCAM antibodies were connected to the screen.


(2) A patient's peripheral blood was collected, and red blood cell lysing solution or lymphocyte separation solution was used to separate nucleated cells;


the nucleated cells flowed through the capture screen once or repeatedly, during this process, the tumor cells were bound to the EpCAM antibodies on the capture screen, thereby being captured;


the capture screen was washed with a cell washing solution, and other impurities or cells that did not specifically bind to the EpCAM antibodies on the capture screen were eluted and removed, and only tumor cells that specifically binded to the EpCAM antibodies were left on the capture screen.


(3) The capture screen bound with the tumor cells was placed in 500 μL of 4% paraformaldehyde solution, fixed at room temperature for 20 min, and washed with phosphate buffer for 2 times.


(4) 200 μL of PD-L1 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:200), and the cells fixed on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(5) 200 μL of PD-L1 secondary antibody solution labeled with fluorophore AlexaFluor 647 was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells were incubated at 25° C. for 60 min, and then washed with phosphate buffer for 2 times.


(6) 200 μL of pan-CK-AlexaFluor 488 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells fixed on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(7) 200 μL of CD45 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:200), and the cells fixed on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(8) 200 μL of CD45 secondary antibody solution labeled with fluorophore AlexaFluor 568 was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells were incubated at 25° C. for 60 min, and then washed with phosphate buffer for 2 times.


(9) All the cells on the capture screen were labeled with 200 μL of nuclear fluorescent dye DAPI.


(10) Through high-throughput multicolor imaging analysis using filters of CY5, FITC, PE and DAPI, the fluorescence color of each channel was observed to detect the tumor cell surface marker molecule PD-L1.


The results of the detection of the positive and negative expression of the tumor cell surface marker molecule PD-L1 by a pan-CK filter (green), a PD-L1 (primary antibody)-Alexa Fluor 647 (secondary antibody) filter (red), a CD45 (primary antibody)-Alexa Fluor 568 (secondary antibody) filter (red), a DAPI filter (blue) and the combination of the four are shown in FIG. 1a to FIG. 5a and FIG. 1b to FIG. 5b, respectively.


Experiments on the Influence of Incubation Sequence on Detection Accuracy


1. Test sample: collecting 4 mL of healthy human peripheral blood, adding about 200 cells of the transfected NCI-H226 reference cell line to it, using lymphocyte separation solution to separate nucleated cells PBMC, and using the nucleated cells PBMC as the test sample.


2. Experimental process


Experimental group: performing detection according to the detection method of this application (the incubation sequence is: PD-L1+ secondary antibody, pan-CK, CD45+ secondary antibody, DAPI staining), the specific process was as follows.


(1) Preparing the capture screen: referring to the corresponding step (1) in Embodiment 1 for the preparation of the capture screen.


(2) Capturing cells: collecting 4 mL of healthy human peripheral blood, adding about 200 cells of the transfected NCI-H226 reference cell line to it, and using lymphocyte separation solution to separate nucleated cells PBMC. The nucleated cells PBMC flowed through the capture screen once or repeatedly, during this process, the tumor cells were bound to the EpCAM antibodies on the capture screen, MCF-7 cells were captured, and finally, the capture screen was washed with a cell washing solution, to elute and remove other impurities or cells that did not specifically bind to the EpCAM antibodies on the capture screen, and only tumor cells that specifically binded to the EpCAM antibodies were left on the capture screen.


(3) Fixing the cells: the capture screen bound with the tumor cells was placed in 500 μL of 4% paraformaldehyde solution, fixed at room temperature for 20 min, and washed with phosphate buffer for 2 times.


(4) 200 μL of PD-L1 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:200), and the cells fixed on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(5) 200 μL of PD-L1 secondary antibody solution labeled with fluorophore AlexaFluor 647 was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells were incubated at 25° C. for 60 min, and then washed with phosphate buffer for 2 times.


(6) 200 μL of pan-CK-AlexaFluor 488 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells fixed on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(7) 200 μL of CD45 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:200), and the cells fixed on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(8) 200 μL of CD45 secondary antibody solution labeled with fluorophore AlexaFluor 568 was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells were incubated at 25° C. for 60 min, and then washed with phosphate buffer for 2 times.


(9) All the cells on the capture screen were labeled with 200 μL of nuclear fluorescent dye DAPI.


(10) Through high-throughput multicolor imaging analysis using filters of CY5, FITC, PE and DAPI, the fluorescence color of each channel was observed.


Control group 1: On basis of the detection method of this application, the incubation sequence was changed (the sequence was CD45+ secondary antibody, PD-L1+ secondary antibody, pan-CK, DAPI staining), the specific process was as follows.


(1) Providing the capture screen: same as the experimental group;


(2) Capturing cells: same as the experimental group;


(3) Fixing cells: same as the experimental group;


(4) 200 μL of CD45 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:200), and the cells fixed on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(5) 200 μL of CD45 secondary antibody solution labeled with fluorophore AlexaFluor 568 was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells were incubated at 25° C. for 60 min, and then washed with phosphate buffer for 2 times.


(6) 200 μL of PD-L1 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:200), and the cells fixed on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(7) 200 μL of PD-L1 secondary antibody solution labeled with fluorophore AlexaFluor 647 was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells were incubated at 25° C. for 60 min, and then washed with phosphate buffer for 2 times.


(8) 200 μL of pan-CK-AlexaFluor 488 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells fixed on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(9) All the cells on the capture screen were labeled with 200 μL of nuclear fluorescent dye DAPI.


(10) Through high-throughput multicolor imaging analysis using filters of CY5, FITC, PE and DAPI, the fluorescence color of each channel was observed to detect the tumor cell surface marker molecule PD-L1. This procedure was also the same as in the experimental group.


Control group 2: On basis of the detection method of this application, the incubation sequence was changed (the sequence was CD45+ secondary antibody, pan-CK, PD-L1+ secondary antibody, DAPI staining), the specific process was as follows:


(1) Providing the capture screen: same as the experimental group;


(2) Capturing cells: same as the experimental group;


(3) Fixing cells: same as the experimental group;


(4) 200 μL of CD45 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:200), and the cells on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(5) 200 μL of CD45 secondary antibody solution labeled with fluorophore AlexaFluor 568 was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells were incubated at 25° C. for 60 min, and then washed with phosphate buffer for 2 times.


(6) 200 μL of pan-CK-AlexaFluor 488 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(7) 200 μL of PD-L1 primary antibody solution was added (the volume ratio of antibody stock solution and PBS was 1:200), and the cells fixed on the capture screen were incubated at 25° C. for 60 min, and then washed with 200 μL of phosphate buffer for 2 times.


(8) 200 μL of PD-L1 secondary antibody solution labeled with fluorophore AlexaFluor 647 was added (the volume ratio of antibody stock solution and PBS was 1:100), and the cells were incubated at 25° C. for 60 min, and then washed with phosphate buffer for 2 times.


(9) All the cells on the capture screen were labeled with 200 μL of nuclear fluorescent dye DAPI.


(10) Through high-throughput multicolor imaging analysis using filters of CY5, FITC, PE and DAPI, the fluorescence color of each channel was observed to detect the tumor cell surface marker molecule PD-L1. This procedure was also the same as in the experimental group.


3. Results and Analysis


CTC criterion: For the cells in the same position, keep the capture screen sample still, switch the filter of the microscope, observe the fluorescence effect of the cells after staining with different dyes, the sequence to be switches is green fluorescence (CK), blue fluorescence (DAPI), red fluorescence (CD45), and magenta fluorescence (PD-L1). If the cell fluorescence captured on the chip is CK-positive, DAPI-positive, and CD45-negative (which can effectively eliminate false positives), it can be identified as a CTC.


The criterion for PD-L1 expression: after confirming that the cell is a CTC, identify the PD-L1 stained fluorescence at the position with the CTC, and if the CTC has PD-L1 expression (i.e., magenta fluorescence), then the CTC has PD-L1 expression; if there is no magenta fluorescence, the CTC does not have PD-L1 expression.


According to the above criteria, according to the experimental conditions of each group, the detection results are summarized as shown in Table 1.









TABLE 1







Fluorescence detection results of experimental group and control groups













Total
The number of
The ratio of CTCs




number
CTCs with PD-L1
with PD-L1 expression


Items
Staining sequence
of CTCs
expression
to the total CTCs














Experimental
PD-L1 + secondary
60
60
 100%


group
antibody, pan-CK,



CD45 + secondary



antibody, DAPI


Control
CD45 + secondary
73
62
84.9%


group 1
antibody, PD-L1 +



secondary antibody,



pan-CK, DAPI


Control
CD45 + secondary
51
41
80.4%


group 2
antibody, pan-CK,



PD-L1 + secondary



antibody, DAPI





Note:


The numbers of CTC cells in the above table corresponds to the numbers of CTC cells in a certain area on the capture screen, not all CTC cells in the test sample; the selected areas before and after the staining in the same group of experiments are the same.






From the results in the above table, it can be seen that the proportion of CTCs with PD-L1 expression in the control group 1 and the control group 2 is significantly lower than the result in the experimental group, indicating that some CTCs with PD-L1 expression in control group 1 and control group 2 were not detected. It shows that using the staining sequence in the control group 1 and the control group 2, the fluorescence of PD-L1 of some CTCs is weak, and some do not even have expression of PD-L1, that is, cells with positive PD-L1 expression are not stained. The reason for the analysis to cause such results may be: using the staining sequence in the control group 1 and the control group 2, the AlexaFluor 647-labeled PD-L1 secondary antibody binds to the CD45 primary antibody, which reduces the binding efficiency of the PD-L1 primary antibody and the AlexaFluor 647-labeled secondary antibody, which leads to inaccurate detection of CTCs with PD-L1 expression, and reduces the expression efficiency of PD-L1.


In the embodiment of this application, a capture screen connected with EpCAM antibodies is used to specifically capture tumor cells in nucleated cells by controlling the flow rate of the system through the microfluidics; then to the capture screen that captures tumor cells, a PD-L1 antibody solution is used and combined with the use of three fluorescent antibodies, namely DAPI, CK and CD45, to identify the CTCs with PD-L1 expression. That is to say, the embodiment only needs to use the capture screen to specifically capture the cells, and only perform two steps of incubation and combined immunofluorescence analysis to accurately identify the CTCs with PD-L1 expression.


Through the technical solutions of the embodiments of this application, in addition to the excellent effects brought about by the above-mentioned staining sequence, it also has the following advantages:


1. this application adopts the method of physical+antigen-antibody binding, which can specifically bind tumor cells chemically to the mesh substrate, and control the flow rate of the system through microfluidics to specifically capture tumor cells in nucleated cells and capture thoroughly, it belongs to the microfluidic product, the capture accuracy is high, there will be no missed capturing or capturing too much due to the size of the cells in the filter retention scheme, and there will be no complicated background cells in the final detection process, which ensures the accuracy of detection and identification.


2. The staining method used in this application is an in-situ staining method, in which the capture screen with the tumor cells is used as the carrier, the captured cells do not need to be transferred, it is only necessary to use the PD-L1 antibody solution in situ on the surface of the carrier and use three fluorescent antibodies, namely DAPI, CK, and CD45, in combination, and can accurately identify CTCs with PD-L1 expression, effectively eliminate false positives, such as blood-derived cell interference or non-specific adsorption interference.


a. The PD-L1 primary antibody solution and the PD-L1 secondary antibody solution labeled with the fluorophore AlexaFluor 647 are first used to label the site of PD-L1 expression in situ;


b. then the pan-CK-AlexaFluor 488 primary antibody solution is used to label CTCs (circulating tumor cells);


c. then the CD45 primary antibody solution and the CD45 secondary antibody solution labeled with the fluorophore AlexaFluor 568 are used to label the white blood cells to remove the interference of white blood cells and eliminate false positives;


d. finally, the nuclear fluorescent dye (DAPI) is used to label the nucleated cells; the combination of the above several fluorescent antibodies can accurately identify CTCs with PD-L1 expression, the results obtained without any of these fluorescent antibodies are inaccurate, and because the total CTCs and the CTCs with PD-L1 expression are labeled at the same time, therefore, the ratio of CTCs with PD-L1 expression to the total CTCs can be obtained.


3. In the embodiment of this application, the capture screen that specifically captures tumor cells is used as a carrier to detect and incubate in situ, the captured tumor cells are fixed in place on the capture screen, and the location of the capture screen can also be kept unchanged, and the microfluidic system is used to make the antibody solutions flow to the position of the capture screen for incubation, even if the antibody solutions flow up and down in the capture screen, it will not cause the captured tumor cells to fall; when performing PD-L1 identification, it only needs to directly place the capture device in the microscope for observation; it simplifies the operation process and reduces the complexity of the operation while ensuring the accuracy of the detection.


4. In the embodiment of the present application, there is no need to remove the capture screen and fix it on other carriers with an adhesive during incubation and detection, which will not affect the captured cells and simplify the operation process.


5. The pan-CK includes many kinds of CK antibodies, and in this application, the pan-CK-AlexaFluor 488 antibody is used as a tumor marker for positive screening, which has high enrichment efficiency and enrichment accuracy, avoids omission of tumor cells in different stages of differentiation, and improves the accuracy of detection.


6. Under normal circumstances, the proportion of false positives in detection samples will be greater than 10%. The incubation and identification method in this application can accurately determine the false positive samples, so that the false positive samples are eliminated from the total number of samples to obtain more accurate identification results.


7. In the embodiment of this application, the tumor cells are complete cells throughout from the initial capture to the subsequent incubation and detection, there is no step of permeabilizing the cells, and the integrity of the cell morphology and structure will not be destroyed.


8. The samples detected in the embodiment of this application are nucleated cells obtained by separating peripheral blood using red blood cell lysing solution or lymphocyte separation solution, which can further avoid the influence of other cells, cytokines and proteins in the body fluid on the PD-L1 antibody incubation detection, and improve the accuracy and reliability of the detection.


9. The technical solution based on the capture screen and the microfluidic system in this application is easier to realize automation, and the capture screen can be fixed in a specific device, and the antibody solutions can be sucked into the device for incubation, detection and other steps.


10. The detection results obtained by adopting the technical solution in this application can effectively avoid the interference of human subjective factors in the traditional HE staining combined with expert reading methods, with higher accuracy and more popularization.


The detection method of the present application is mainly used for non-diagnostic purposes of PD-L1 detection, but can also be used for diagnostic purposes.


The embodiments described above are only for illustrating the technical concepts and features of the present disclosure, are preferred embodiments, and are intended to make those skilled in the art being able to understand the present disclosure and thereby implement it, and should not be concluded to limit the protective scope of this disclosure. Any equivalent variations or modifications according to the present disclosure should be covered by the protective scope of the present disclosure.

Claims
  • 1. A method for detecting a tumor cell surface marker molecule PD-L1, comprising the following steps: providing a capture screen that has antibodies capable of specifically binding to tumor cells;making a sample to be tested flow through the capture screen, such that tumor cells in the sample to be tested bind to the capture screen;fixing captured tumor cells on the capture screen; andsuccessively using a PD-L1 primary antibody solution, a PD-L1 secondary antibody solution labeled with a fluorophore AlexaFluor 647, a pan-CK-AlexaFluor 488 primary antibody solution, a CD45 primary antibody solution and a CD45 secondary antibody solution labeled with a fluorophore AlexaFluor 568, to incubate the cells fixed on the capture screen, and then labeling all cells on the capture screen with a nuclear fluorescent dye.
  • 2. The detection method according to claim 1, wherein during incubating, the entire capture screen with fixed tumor cells is used as a carrier for incubation.
  • 3. The detection method according to claim 1, wherein the cells fixed on the capture screen is incubated as follows: adding a PD-L1 primary antibody solution, incubating the cells fixed on the capture screen at room temperature for 20˜80 min, and then washing with phosphate buffer;adding a PD-L1 secondary antibody solution labeled with fluorophore AlexaFluor 647, incubating at room temperature for 20˜80 min, and washing with phosphate buffer;adding a pan-CK-AlexaFluor 488 primary antibody solution, incubating the cells fixed on the capture screen at room temperature for 20˜80 min, and washing with phosphate buffer;adding a CD45 primary antibody solution, incubating the cells fixed on the capture screen at room temperature for 20˜80 min, and washing with phosphate buffer; andadding a CD45 secondary antibody solution labeled with fluorophore AlexaFluor 568, incubating at room temperature for 20˜80 min, and washing with phosphate buffer.
  • 4. The detection method according to claim 1, wherein after incubated, all nucleated cells on the capture screen are labeled with nuclear fluorescent dye DAPI.
  • 5. The detection method according to claim 1, wherein the detection method further comprises the following step: observing fluorescence color of each channel to detect the tumor cell surface marker molecule PD-L1, through high-throughput multicolor imaging analysis using filters of CY5, FITC, PE and DAPI.
  • 6. The detection method according to claim 1, wherein the capture screen comprises a mesh substrate and EpCAM antibodies arranged on the mesh substrate by incubation.
  • 7. The detection method according to claim 6, wherein the mesh substrate has a size of 2-10 mm×2-10 mm, and the screen has pores of 20 μm-100 μm.
  • 8. The detection method according to claim 6, wherein the mesh substrate comprises a stainless-steel body and a protective layer covering a surface of the stainless-steel body, the protective layer is made of a precious metal or an alloy thereof, and the EpCAM antibodies are arranged on the protective layer.
  • 9. The detection method according to claim 8, wherein the protective layer is an AuPd layer deposited on the stainless-steel body by magnetron sputtering or electrochemical methods.
  • 10. The detection method according to claim 8, wherein the EpCAM antibodies are attached to the protective layer via Traut's reagent or thiolate molecules with biotin-avidin.
  • 11. The detection method according to claim 1, wherein the sample to be detected is nucleated cells separated from body fluid.
  • 12. The detection method according to claim 11, wherein the nucleated cells are obtained by separating peripheral blood using red blood cell lysing solution or lymphocyte separation solution.
  • 13. The detection method according to claim 11, wherein after the nucleated cells flow through the capture screen and tumor cells bind to the capture screen, washing the capture screen with a cell washing solution to remove debris or cells that are not bound to the capture screen.
  • 14. The detection method according to claim 11, wherein the body fluid is blood, urine or peritoneal fluid.
  • 15. The detection method according to claim 1, wherein the capture screen bound with the tumor cells is placed in a 4% paraformaldehyde solution, maintained at room temperature for 10˜60 min, and washed with phosphate buffer, to fix the captured tumor cells on the capture screen.
Priority Claims (1)
Number Date Country Kind
201910601477.8 Jul 2019 CN national
RELATED APPLICATIONS

The present application is a U.S. National Phase of International Application Number PCT/CN2020/100189, filed Jul. 3, 2020, and claims priority to Chinese Application Number 2019106014778, filed Jul. 4, 2019.

PCT Information
Filing Document Filing Date Country Kind
PCT/CN2020/100189 7/3/2020 WO