CN111562375A - Immunofluorescence kit for detecting expression of peripheral blood circulating tumor cells PD-L1 of gastric cancer patient and detection method - Google Patents

Immunofluorescence kit for detecting expression of peripheral blood circulating tumor cells PD-L1 of gastric cancer patient and detection method Download PDF

Info

Publication number
CN111562375A
CN111562375A CN202010311184.9A CN202010311184A CN111562375A CN 111562375 A CN111562375 A CN 111562375A CN 202010311184 A CN202010311184 A CN 202010311184A CN 111562375 A CN111562375 A CN 111562375A
Authority
CN
China
Prior art keywords
peripheral blood
gastric cancer
fluorescence
ctc
filter
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202010311184.9A
Other languages
Chinese (zh)
Other versions
CN111562375B (en
Inventor
李胜
李�浩
崔凯
夏梅
高德海
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shandong Kaige Intelligent Machine Co ltd
Original Assignee
Shandong First Medical University and Shandong Academy of Medical Sciences
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 Shandong First Medical University and Shandong Academy of Medical Sciences filed Critical Shandong First Medical University and Shandong Academy of Medical Sciences
Priority to CN202010311184.9A priority Critical patent/CN111562375B/en
Publication of CN111562375A publication Critical patent/CN111562375A/en
Priority to PCT/CN2021/087721 priority patent/WO2021213262A1/en
Application granted granted Critical
Publication of CN111562375B publication Critical patent/CN111562375B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • 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
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • C12N5/0694Cells of blood, e.g. leukemia cells, myeloma cells
    • 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
    • G01N33/57407Specifically defined cancers
    • G01N33/57446Specifically defined cancers of stomach or intestine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • C12N2509/10Mechanical dissociation
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70532B7 molecules, e.g. CD80, CD86

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • General Health & Medical Sciences (AREA)
  • Food Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Analytical Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Hospice & Palliative Care (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The invention discloses an immunofluorescence kit and a detection method for detecting the expression of peripheral blood circulating tumor cells PD-L1 of a gastric cancer patient, wherein Circulating Tumor Cells (CTC) in peripheral blood of a gastric cancer patient at a late stage are obtained by separation through a membrane filter device, and the expression condition of PD-L1 on the CTC is further detected by using an immunofluorescence technology. The detection method provided by the invention can detect the PD-L1 expression condition of a patient with advanced or recurrent gastric cancer without taking a tissue sample by puncture biopsy. The method provided by the invention has the advantages of good separation of circulating tumor cells, capability of avoiding the interference of blood cells, capability of avoiding false positive results caused by edge effect possibly generated in the dyeing process, good stability, reduction of cell loss and improvement of detection accuracy. The marker PD-L1 shows good specificity and sensitivity when used, and the application of the marker PD-L1 in gastric cancer is improved.

Description

Immunofluorescence kit for detecting expression of peripheral blood circulating tumor cells PD-L1 of gastric cancer patient and detection method
Technical Field
The invention provides an immunofluorescence kit for detecting peripheral blood circulation tumor cell PD-L1 expression of a gastric cancer patient and a detection method, and belongs to the technical field of molecular biology.
Background
Gastric Cancer (GC) is a highly fatal malignancy with the fourth world incidence (95 thousands of new cases annually) and the third in 2012 tumor-related deaths. Despite the recent improvements in gastric cancer diagnosis and treatment, the 5-year survival rate of gastric cancer patients is less than 30%, and about 50% of gastric cancer patients suffer from tumor recurrence or metastasis after curative resection. Tumor recurrence and metastasis are the leading cause of death in gastric cancer patients.
Circulating Tumor Cells (CTC) are tumor cells that have been shed from solid tumors into the peripheral blood circulation, and since their discovery in 1989, various methods have been used to detect Circulating tumor cells in the peripheral blood. Recent studies have shown that their detection is of great clinical significance for assessing the prognosis of patients with tumours, especially patients with advanced tumours, and for selecting appropriate individualized treatments. CTC detection is called liquid biopsy of tumor because of its characteristics of minimal invasion, real-time detection, etc.
Immunotherapy with PD-1/PD-L1 as an immune target has brought new eosin for the treatment of gastric cancer. The research finds that the immunosuppression is closely related to immune escape and the over-expression of tumor cells PD-L1, and the tumor cells can be combined with PD-1 on the surface of T cells of immune cells through PD-L1 on the surface of the tumor cells to transmit inhibitory signals, so that the T cells cannot recognize the tumor cells and send attack signals to the tumor cells, and the immune escape of the tumor cells is caused. Based on this theory, it was hypothesized that the appearance of apoptosis, immune clearance or survival, metastatic outcome of Circulating Tumor Cells (CTCs) shed from primary foci into the circulatory system is closely related to PD-L1 expression. The curative effect of PD-1 or PD-L1 immune preparations is related to the immunohistochemical expression level of PD-L1 in tumor tissues, and the expression level of PD-L1 is possibly a biomarker for predicting the curative effect of PD-1 immunotherapy; further studies have shown that high expression of PD-L1 in gastric cancer tissues is positively correlated with tumor invasiveness. According to the research of documents, the current detection methods of different applications of breast cancer, prostate cancer, colorectal cancer and liver cancer have few reports on the expression detection of Circulating Tumor Cells (CTC) PD-L1, but the detection of the circulating tumor cells PD-L1 of the stomach cancer is not reported at home and abroad.
The immunofluorescence analysis technology is a method for researching the distribution of specific protein antigens in cells by combining an immunological method (antigen-antibody specific binding) with a fluorescence labeling technology. The fluorescence emitted by the fluorescein can be detected under a fluorescence microscope, the fluorescein emits bright fluorescence (yellow green or orange red) under the irradiation of exciting light, the cells or tissues where the fluorescence is located can be seen, and the content is measured by using a quantitative technology, so that the cells are qualitatively and locally analyzed.
In current clinical practice, a specimen detected by PD-L1 of a gastric cancer patient is mainly tumor tissue, is from operation or aspiration biopsy, and is difficult to detect for multiple times or in real time. Therefore, the detection of the expression condition of the Circulating Tumor Cell (CTC) PD-L1 has important value for gastric cancer prognosis and immunotherapy curative effect evaluation.
At present, units such as Shandong province first medical university, Shandong province drug research institute combined with Shandong Qixin Biotechnology limited company, Shandong well-known Biotechnology limited company, Jinan Xin Biotechnology limited company, Shandong discovery biotechnology limited company and the like, carry out industrialized popularization on the key technology of detection and identification of circulating tumor cells, the project is a Shandong province major scientific and technological innovation project, the project takes the Shandong province drug research institute in Jinan school of Shandong first medical university as the core, realizes a registration system, relies on the core diagnostic technology of detection and identification of circulating tumor cells, and further registers, identifies and diagnoses a kit, and comprises PD1, PD-L1, ER, PR, Her-2, GPC-3, VEGF, P53, Vimentin, EGFR, RAS, CK, ALK-D5F3, CD20, ALK/EML4, Beta-catendin, E-Cacatenin, EP-CAM, HPV, IDH-1, PSA, PSMA, VEGF, GFAP, cytokeratin, AE1/AE3, estrogen receptor, progestin receptor, BCA-225, CA 125, CEA, EMA, ERCC1, HPV, Ki-67, P53, TOP2A and the like are used as tracers for expression of CTCs, and the identification and diagnosis kit is registered to be an ultrasensitive, ultrafast, high-coverage, low-cost, accurate and specific kit, and industrially popularized by cooperating with Shandong Qicheng Xin Biotech Co., Ltd, Shandong well-known Biotech Co., Ltd, Jinan En Biotech Co., Ltd, Shandong discovery Biotech Co., Ltd registered in Jinan.
Disclosure of Invention
Aiming at the defects that in the prior art, a patient with advanced tumor or recurrent gastric cancer cannot obtain a tissue sample by real-time or repeated puncture, and further cannot evaluate the real-time dynamic state of PD-L1 of the patient, and false positive and false negative easily occur in the existing detection method, the invention provides an immunofluorescence kit and a detection method for expressing peripheral blood circulating tumor cells PD-L1 of the patient with gastric cancer, wherein a membrane filtering device is used for separating and obtaining Circulating Tumor Cells (CTC) in peripheral blood of the patient with advanced gastric cancer, and the immunofluorescence technology is further used for detecting the expression condition of PD-L1 on the CTC.
The invention is realized by the following technical scheme:
an immunofluorescence kit for detecting the expression of peripheral blood circulation tumor cells PD-L1 of a gastric cancer patient comprises 45mL of diluent, 1mL of destaining solution, 0.5mL of staining solution A, 1mL of staining solution B, 200 μ L of methanol, 200 μ L of 2% PFA, 100 μ L of 10% goat serum, 100 μ L of primary antibody suspension consisting of mouse anti-CK, rat anti-CD 45 and rabbit anti-PD-L1, 100 μ L of secondary antibody suspension consisting of fluorescence-labeled goat anti-mouse, fluorescence-labeled goat anti-rat and fluorescence-labeled goat anti-rabbit, and a DAPI blocking tablet;
in the primary anti-suspension, the mouse anti-CK, the rat anti-CD 45 and the rabbit anti-PD-L1 are respectively diluted according to the ratio of 1:100, the ratio of 1:400 and the ratio of 1:500, and the total volume is 100 mu L;
and diluting the secondary antibody suspension with fluorescence-labeled goat-anti mouse, fluorescence-labeled goat-anti rat and fluorescence-labeled goat-anti rabbit at a ratio of 1: 500.
Preferably, the diluent consists of: 1mmol/L EDTA +0.1% BSA +0.1% trehalose +0.2% tyrosine + PBS buffer 150mmol/L, wherein the percentage is volume ratio.
Preferably, the decoloring liquid is composed of 95% alcohol and 100% xylene in a volume ratio of 1: 1.
Preferably, the staining solution A is a DAB staining solution; the staining solution B is hematoxylin staining solution.
The method for detecting the expression of the peripheral blood circulation tumor cells PD-L1 of the gastric cancer patient by the kit in a non-diagnosis purpose comprises the following steps:
(1) separating and obtaining peripheral blood of patients with advanced or recurrent gastric cancer who can not obtain tissue specimens by using a membrane filtering device: collecting 5ml of peripheral blood of the median cubital vein of a patient with late stage or recurrent gastric cancer who can not obtain a tissue specimen;
(2) peripheral blood sample pretreatment: diluting the collected peripheral blood sample by 10 times by using a diluent, and adding polyformaldehyde to fix the peripheral blood sample for 10 minutes after dilution, wherein the fixed final concentration is 0.25%;
(3) and (3) filtering the peripheral blood sample by using a membrane filtration tumor cell separation device, and separating to obtain peripheral blood CTC: adding the pretreated peripheral blood sample into a blood sample container of a membrane filtration tumor cell separation device, and naturally filtering the blood sample by means of gravity;
(4) after the filtration is finished, taking the filter out of the membrane filtration tumor cell separation device, adding 0.5ml of circulating tumor cell staining solution A into the filter, staining for 3min, and washing with PBS buffer solution; after the filtrate is completely filtered, 1ml of staining solution B is added, the dyeing is carried out for 2min, and 1ml of pure water is used for washing for 2 times;
(5) adding 200 μ l of 2% PFA into the filter, fixing at room temperature for 5min, and rinsing with 0.5ml PBS for 3 times, each for 2 min;
(6) adding 200 μ l of precooled methanol into the filter, fixing at 4 ℃ for 15min, taking down the filter membrane, placing on a glass slide, drying, and observing under a microscope to determine whether CTC exists;
(7) and detecting the expression condition of PD-L1 of the CTC in the peripheral blood by using an immunofluorescence method.
Preferably, the specific method for detecting the expression of PD-L1 in the CTC in the peripheral blood in the step (7) is as follows:
(1) and (3) decoloring: taking down the filter membrane with CTC from the glass slide, soaking in a decolorizing solution for 4-6 hours, removing the CTC staining solution, and washing with PBS for 2min × 3 times;
(2) and (3) sealing: dropping 100 μ l10% goat serum onto the filter membrane, standing at room temperature for 30min, and removing excess serum (diluting goat serum with PBS);
(3) primary antibody incubation: dripping 100 μ L primary antibody suspension composed of mouse anti-CK, rat anti-CD 45 and rabbit anti-PD-L1 onto the filter membrane, incubating at 37 deg.C for 1h or overnight at 4 deg.C, and washing with PBS for 3min × 3 times;
(4) and (3) secondary antibody incubation: dripping 100 μ l of secondary antibody suspension composed of fluorescence labeled goat-anti mouse, fluorescence labeled goat-anti rat, and fluorescence labeled goat-anti rabbit onto the filter membrane, incubating at room temperature for 30min, and washing with PBS for 2min × 3 times;
(5) sealing the piece by using a sealing agent containing DAPI, reading the piece and collecting a drawing;
(6) after the image collection was completed, the sections were removed and then stained with giemsa renbergii, which was compared with the IF results.
The device for separating and circulating tumor cells by membrane filtration comprises a filter, a blood sample container, a waste liquid tank and an iron stand, wherein the iron stand is provided with a base, a vertical frame and a support, the blood sample container is arranged at the upper part of the iron stand through the support, the filter is arranged below the blood sample container, the filter is communicated to the waste liquid tank through an infusion apparatus, and the waste liquid tank is arranged on the base.
The filter comprises a filter upper opening, a filter membrane carrying platform and a filter lower opening, and the filter membrane is arranged on the filter membrane carrying platform; the upper port of the filter is connected with a blood sample container, and the lower port of the filter is connected with a waste liquid cylinder through an infusion apparatus.
The filter membrane is made of hydrophobic material, and filter holes with the caliber of 8 microns are uniformly distributed on the filter membrane; tumor cells are typically greater than 15 microns in diameter, while blood cells (including red blood cells, white blood cells) are typically less than 8 microns in diameter, so that when peripheral blood containing CTCs is filtered, the blood cells are filtered by being smaller in diameter than the filter pores, and the CTCs are retained on the filter membrane by being larger in diameter than the filter pores.
Advantageous effects
(1) The detection method provided by the invention can detect the PD-L1 expression condition of a patient with advanced or recurrent gastric cancer without obtaining a tissue sample by puncture biopsy, and can realize real-time dynamic detection by utilizing a minimally invasive technology.
(2) The method provided by the invention has the advantages of good separation of circulating tumor cells, capability of avoiding the interference of blood cells, capability of avoiding false positive results caused by edge effect possibly generated in the dyeing process, good stability, reduction of cell loss and improvement of detection accuracy.
Drawings
FIG. 1 is a schematic structural view of a membrane filtration apparatus according to the present invention;
FIG. 2 is a schematic sectional view showing the structure of a filter of the membrane filtration apparatus of the present invention;
FIG. 3 is a schematic view showing the structure of a filter membrane of the membrane filtration apparatus of the present invention;
FIG. 4 is an immunofluorescence staining image of peripheral blood circulating tumor cells PD-L1 of a patient with advanced gastric cancer.
In the figure: 1 iron stand, 2 blood sample containers, 3 filters, 4 transfusion devices, 5 waste liquid jars, 6 filter upper ports, 7 filter membranes, 8 filter membrane platforms, 9 filter lower ports, 10 filter holes, 11 bases, 12 vertical frames and 13 supports.
Detailed Description
The invention is elucidated below with reference to the figures and embodiments.
The specific specification of the kit used in the embodiment of the invention is shown in table 1:
TABLE 1
Figure 290890DEST_PATH_IMAGE002
The primary antibody suspension consists of mouse anti-CK, rat anti-CD 45 and rabbit anti-PD-L1, wherein the mouse anti-CK, the rat anti-CD 45 and the rabbit anti-PD-L1 are respectively diluted by BD wash buffer according to the ratio of 1:100, the ratio of 1:500 and the ratio of 1:400, and 10 mu L of mouse anti-CK, 50 mu L of rat anti-CD 45 and 40 mu L of rabbit anti-PD-L1 are taken to form the primary antibody suspension after dilution;
the secondary antibody suspension consists of a fluorescence-labeled goat-Anti mouse, a fluorescence-labeled goat-Anti rat and a fluorescence-labeled goat-Anti rabbit, which are respectively Alexa Fluor 546 gat Anti-mouse, Alexa Fluor 488 gat Anti-rat and Alexa Fluor 647 gat Anti-rabbitt sold in the market, and equal amounts of the three fluorescence-labeled secondary antibodies are respectively diluted by BD washbuffer according to a ratio of 1:500 and mixed uniformly to obtain the secondary antibody suspension.
The technical method is applied to the embodiment of separating, obtaining and identifying 8 gastric cancer patients (8 normal human samples are simultaneously detected as negative controls) peripheral blood circulation tumor cells.
Example 1
Firstly, separating and acquiring CTCs in peripheral blood of patients with advanced or recurrent gastric cancer, wherein the patients cannot obtain tissue specimens, and determining whether the CTCs exist:
collecting 5ml of fasting 8-12 hours fasting blood from the median cubital vein, diluting peripheral blood with 45ml of diluent, and then adding 3ml of 4% paraformaldehyde to fix the diluted blood sample for 10 minutes;
at fixed intervals, a membrane filtration device was assembled: as shown in fig. 1, 2 and 3, the filter device comprises a filter 3, a filter membrane 7, a blood sample container 2, a waste liquid tank 5 and an iron stand 1;
wetting the filter 3 with 10ml of PBS, then adding the fixed peripheral blood sample into the blood sample container 2 of the membrane filtration device, allowing it to naturally filter by gravity, and the CTC being trapped on the filter membrane 7;
the tumor cells are typically larger than 15 microns in diameter, while the blood cells (including red blood cells, white blood cells) are typically smaller than 8 microns in diameter, so that when peripheral blood containing CTCs is filtered, the blood cells can be filtered by being smaller than filter pores 10, while the CTCs are retained on filter membrane 7 by being larger than filter pores 10.
After the filtration is finished, taking the filter 3 from the filter device, opening and removing the upper opening 6 of the filter, adding 0.5ml of circulating tumor cell staining solution A into the filter, staining for 3min, and washing with PBS buffer solution; filtering the filtrate completely, adding solution B, 1ml, staining for 2min, and pure water 1ml, washing filter 3 with PBS buffer solution, taking down filter membrane 7 with ophthalmic forceps with cell surface facing upwards, and placing on glass slide;
the filters were dried and observed under a microscope to determine the presence of CTCs, with the results shown in table 2.
By observation, no CTCs were detected in 8 healthy volunteers; CTC was detected in 6 cases (Table 2) except that CTC was not detected in 1 recurrent gastric cancer patient, and the positive rate of this detection was 87.5%. It is worth noting that when the diluent contains 1mmol/LEDTA +0.1% BSA +0.1% trehalose +0.2% tyrosine + PBS buffer solution 150mmol/L, the circulating tumor cells in human peripheral blood can be effectively separated, blood cells (red blood cells, white blood cells and the like) are prevented from adhering to the tumor cells, the color development of CTC is influenced, and the final detection result is influenced.
Table 2 example 1 CTC assay results
Figure 614555DEST_PATH_IMAGE004
Secondly, detecting the expression condition of PD-L1 of CTC by using an immunofluorescence technique:
taking down the filter membrane 7 carrying the CTC on the glass slide from the glass slide, soaking in a decoloration solution which is uniformly mixed by 95% alcohol and 100% dimethylbenzene according to the volume ratio of 1:1 for 4-6 hours, removing the CTC staining solution, and washing with PBS for 2min multiplied by 3 times; dripping 100 μ l10% goat serum, standing at room temperature for 30min, sucking off excessive serum, dripping 100 μ l primary antibody suspension onto the filter membrane, incubating at 37 deg.C for 1h, and washing with PBS for 3min × 3 times; then, 100 mul of secondary antibody suspension is dripped on the filter membrane, and the mixture is incubated for 30min at room temperature, and then washed for 2min multiplied by 3 times by PBS; sealing the piece by using a sealing agent containing DAPI, reading the piece and collecting a drawing; after the image collection was completed, the sections were removed and then stained with giemsa renbergii, which was compared with the IF results.
FIG. 4 is an immunofluorescence staining image of peripheral blood circulating tumor cells of a patient with advanced hepatocellular carcinoma, which shows that the tumor cells have large cell size and abnormal karyoplasm ratio according to the immunological and morphological manifestations, and the immunological manifestations are typical CTCs. The detected circulating tumor cells are subjected to immunofluorescence to verify the expression of PD-L1 and are compared with the result of a gastric cancer general specimen PD-L1, the difference is observed, the target treatment of the gastric cancer is guided mainly for patients with negative expression of the general specimen PD-L1 and positive expression of the circulating tumor cells, and a new thought is provided for the target treatment of the gastric cancer.

Claims (6)

1. An immunofluorescence kit for detecting the expression of peripheral blood circulation tumor cells PD-L1 of a gastric cancer patient is characterized by comprising 45mL of diluent, 1mL of destaining solution, 0.5mL of staining solution A, 1mL of staining solution B, 200 μ L of methanol, 200 μ L of 2% PFA, 100 μ L of 10% goat serum, 100 μ L of primary antibody suspension consisting of mouse anti-CK, rat anti-CD 45 and rabbit anti-PD-L1, 100 μ L of secondary antibody suspension consisting of a fluorescence-labeled goat anti-mouse, a fluorescence-labeled goat anti-rat and a fluorescence-labeled goat anti-rabbit, and a DAPI encapsulated tablet;
in the primary anti-suspension, the mouse anti-CK, the rat anti-CD 45 and the rabbit anti-PD-L1 are respectively diluted according to the ratio of 1:100, the ratio of 1:400 and the ratio of 1:500, and the total volume is 100 mu L;
and diluting the secondary antibody suspension with fluorescence-labeled goat-anti mouse, fluorescence-labeled goat-anti rat and fluorescence-labeled goat-anti rabbit at a ratio of 1: 500.
2. The kit according to claim 1, wherein the diluent consists of: 1mmol/L EDTA +0.1% BSA +0.1% trehalose +0.2% tyrosine + PBS buffer 150mmol/L, wherein the percentage is volume ratio.
3. The kit of claim 1, wherein the destaining solution is comprised of 95% alcohol to 100% xylene in a volume ratio of 1: 1.
4. The kit according to claim 1, wherein the staining solution A is DAB staining solution; the staining solution B is hematoxylin staining solution.
5. A method for detecting the expression of PD-L1 in peripheral blood circulation tumor cells of a gastric cancer patient for non-diagnostic purposes by using the kit of any one of claims 1 to 4, comprising the steps of:
(1) separating and obtaining peripheral blood of patients with advanced or recurrent gastric cancer who can not obtain tissue specimens by using a membrane filtering device: collecting 5ml of peripheral blood of the median cubital vein of a patient with late stage or recurrent gastric cancer who can not obtain a tissue specimen;
(2) peripheral blood sample pretreatment: diluting the collected peripheral blood sample by 10 times by using a diluent, and adding polyformaldehyde to fix the peripheral blood sample for 10 minutes after dilution, wherein the fixed final concentration is 0.25%;
(3) and (3) filtering the peripheral blood sample by using a membrane filtration tumor cell separation device, and separating to obtain peripheral blood CTC: adding the pretreated peripheral blood sample into a blood sample container of a membrane filtration tumor cell separation device, and naturally filtering the blood sample by means of gravity;
(4) after the filtration is finished, taking the filter out of the membrane filtration tumor cell separation device, adding 0.5ml of circulating tumor cell staining solution A into the filter, staining for 3min, and washing with PBS buffer solution; after the filtrate is completely filtered, 1ml of staining solution B is added, the dyeing is carried out for 2min, and 1ml of pure water is used for washing for 2 times;
(5) adding 200 μ l of 2% PFA into the filter, fixing at room temperature for 5min, and rinsing with 0.5ml PBS for 3 times, each for 2 min;
(6) adding 200 μ l of precooled methanol into the filter, fixing at 4 ℃ for 15min, taking down the filter membrane, placing on a glass slide, drying, and observing under a microscope to determine whether CTC exists;
(7) and detecting the expression condition of PD-L1 of the CTC in the peripheral blood by using an immunofluorescence method.
6. The method of claim 5, wherein the specific method for detecting PD-L1 expression in peripheral blood CTC in step (7) is as follows:
(1) and (3) decoloring: taking down the filter membrane with CTC from the glass slide, soaking in a decolorizing solution for 4-6 hours, removing the CTC staining solution, and washing with PBS for 2min × 3 times;
(2) and (3) sealing: dropping 100 μ l10% goat serum onto the filter membrane, standing at room temperature for 30min, and removing excess serum (diluting goat serum with PBS);
(3) primary antibody incubation: dripping 100 μ L primary antibody suspension composed of mouse anti-CK, rat anti-CD 45 and rabbit anti-PD-L1 onto the filter membrane, incubating at 37 deg.C for 1h or overnight at 4 deg.C, and washing with PBS for 3min × 3 times;
(4) and (3) secondary antibody incubation: dripping 100 μ l of secondary antibody suspension composed of fluorescence labeled goat-anti mouse, fluorescence labeled goat-anti rat, and fluorescence labeled goat-anti rabbit onto the filter membrane, incubating at room temperature for 30min, and washing with PBS for 2min × 3 times;
(5) sealing the piece by using a sealing agent containing DAPI, reading the piece and collecting a drawing;
(6) after the image collection was completed, the sections were removed and then stained with giemsa renbergii, which was compared with the IF results.
CN202010311184.9A 2020-04-20 2020-04-20 Immunofluorescence kit for detecting expression of peripheral blood circulating tumor cells PD-L1 of gastric cancer patient and detection method Active CN111562375B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202010311184.9A CN111562375B (en) 2020-04-20 2020-04-20 Immunofluorescence kit for detecting expression of peripheral blood circulating tumor cells PD-L1 of gastric cancer patient and detection method
PCT/CN2021/087721 WO2021213262A1 (en) 2020-04-20 2021-04-16 Immunofluorescence test kit for measuring pd-l1 expression in circulating tumor cells in peripheral blood in stomach cancer patient, and measurement method

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010311184.9A CN111562375B (en) 2020-04-20 2020-04-20 Immunofluorescence kit for detecting expression of peripheral blood circulating tumor cells PD-L1 of gastric cancer patient and detection method

Publications (2)

Publication Number Publication Date
CN111562375A true CN111562375A (en) 2020-08-21
CN111562375B CN111562375B (en) 2022-05-06

Family

ID=72073210

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010311184.9A Active CN111562375B (en) 2020-04-20 2020-04-20 Immunofluorescence kit for detecting expression of peripheral blood circulating tumor cells PD-L1 of gastric cancer patient and detection method

Country Status (2)

Country Link
CN (1) CN111562375B (en)
WO (1) WO2021213262A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021213262A1 (en) * 2020-04-20 2021-10-28 山东第一医科大学(山东省医学科学院) Immunofluorescence test kit for measuring pd-l1 expression in circulating tumor cells in peripheral blood in stomach cancer patient, and measurement method
CN115792245A (en) * 2023-02-02 2023-03-14 北京市肿瘤防治研究所 Method for simultaneously detecting activity and immune state of peripheral blood gastric cancer circulating tumor cells

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115184620B (en) * 2022-09-14 2023-01-24 山东子峰生物技术有限公司 Quantum dot fluorescence detection test strip and kit for PLA2R antibody and application of quantum dot fluorescence detection test strip and kit

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105588943A (en) * 2016-01-28 2016-05-18 山东省肿瘤防治研究院 Detection method for peripheral blood CTC (Circulating Tumor Cell) Her-2 gene of stomach cancer patient
CN106198984A (en) * 2016-08-22 2016-12-07 上海立闻生物科技有限公司 The detection method of Peripheral Blood of Patients with Non-small Cell Lung circulating tumor cell PDL1 gene
WO2019089740A1 (en) * 2017-11-03 2019-05-09 Dana-Farber Cancer Institute, Inc. Biomarkers of clinical response and benefit to immune checkpoint inhibitor therapy
CN110632292A (en) * 2019-06-12 2019-12-31 江苏莱尔生物医药科技有限公司 Immunofluorescence kit for detecting PD-L1 and CD8 antigens and application method

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180267046A1 (en) * 2017-03-14 2018-09-20 Micareo Taiwan Co., Ltd. Methods for diagnosing and treating cancer
CN111562375B (en) * 2020-04-20 2022-05-06 山东第一医科大学(山东省医学科学院) Immunofluorescence kit for detecting expression of peripheral blood circulating tumor cells PD-L1 of gastric cancer patient and detection method
CN111521799A (en) * 2020-04-22 2020-08-11 山东第一医科大学(山东省医学科学院) Immunofluorescence kit for detecting esophageal squamous carcinoma patient PD-L1 gene expression through peripheral blood circulation tumor cells
CN111638359A (en) * 2020-07-01 2020-09-08 山东凯歌智能机器有限公司 Immunofluorescence kit and detection method for detecting PD-L1 gene mutation of peripheral blood circulating tumor cells of small cell lung cancer patients

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105588943A (en) * 2016-01-28 2016-05-18 山东省肿瘤防治研究院 Detection method for peripheral blood CTC (Circulating Tumor Cell) Her-2 gene of stomach cancer patient
CN106198984A (en) * 2016-08-22 2016-12-07 上海立闻生物科技有限公司 The detection method of Peripheral Blood of Patients with Non-small Cell Lung circulating tumor cell PDL1 gene
WO2019089740A1 (en) * 2017-11-03 2019-05-09 Dana-Farber Cancer Institute, Inc. Biomarkers of clinical response and benefit to immune checkpoint inhibitor therapy
CN110632292A (en) * 2019-06-12 2019-12-31 江苏莱尔生物医药科技有限公司 Immunofluorescence kit for detecting PD-L1 and CD8 antigens and application method

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MENGYUAN LIU ET AL.: "Prognostic significance of PD-L1 expression on cell-surface vimentin-positive circulating tumor cells in gastric cancer patients", 《MOLECULAR ONCOLOGY》 *
王岳君 等: "胃癌中PD-L1蛋白表达及分布特征的病理学意义", 《临床与实验病理学杂志》 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021213262A1 (en) * 2020-04-20 2021-10-28 山东第一医科大学(山东省医学科学院) Immunofluorescence test kit for measuring pd-l1 expression in circulating tumor cells in peripheral blood in stomach cancer patient, and measurement method
CN115792245A (en) * 2023-02-02 2023-03-14 北京市肿瘤防治研究所 Method for simultaneously detecting activity and immune state of peripheral blood gastric cancer circulating tumor cells

Also Published As

Publication number Publication date
CN111562375B (en) 2022-05-06
WO2021213262A1 (en) 2021-10-28

Similar Documents

Publication Publication Date Title
CN111562375B (en) Immunofluorescence kit for detecting expression of peripheral blood circulating tumor cells PD-L1 of gastric cancer patient and detection method
CN111521796A (en) Immunofluorescence kit and detection method for detecting expression of peripheral blood circulating tumor cells PD-L1 of renal cancer patient
CN111521798A (en) Kit and method for detecting PD-L1 gene mutation of peripheral blood circulating tumor cells of renal cancer patients
CN111638359A (en) Immunofluorescence kit and detection method for detecting PD-L1 gene mutation of peripheral blood circulating tumor cells of small cell lung cancer patients
CN111638341A (en) Kit and method for detecting PD-L1 gene mutation of peripheral blood circulating tumor cells of small cell lung cancer patients
CN111521794A (en) Immunofluorescence kit and detection method for detecting NSE gene mutation of peripheral blood circulating tumor cells of small cell lung cancer patients
CN111638357A (en) Immunofluorescence kit and method for E-Cadherin mutation of peripheral blood circulating tumor cells of patient with non-small cell lung cancer
CN111521790A (en) Non-diagnosis purpose method for detecting PD-L1 gene mutation of colorectal cancer patient through peripheral blood circulating tumor cells
CN111551718A (en) Immunofluorescence kit and detection method for detecting prostate cancer patient peripheral blood circulating tumor cell PD-L1 expression
CN111521799A (en) Immunofluorescence kit for detecting esophageal squamous carcinoma patient PD-L1 gene expression through peripheral blood circulation tumor cells
CN111521792A (en) Immunofluorescence kit and method for detecting non-small cell lung cancer patient peripheral blood circulating tumor cell PD-L1 gene mutation
CN111521801A (en) Kit for detecting BRAF gene V600E mutation expression of colorectal cancer patient through peripheral blood circulation tumor cells
CN111521793A (en) Immunofluorescence kit and detection method for detecting CEA gene mutation of peripheral blood circulating tumor cells of non-small cell lung cancer patients
CN111679077A (en) Immunofluorescence kit for expressing E-Cadherin (circulating tumor cell) in peripheral blood of renal cell carcinoma patient and detection method
CN111521795A (en) Kit and method for detecting non-small cell lung cancer patient peripheral blood circulating tumor cell PD-L1 gene mutation
CN111638358A (en) Immunofluorescence kit and method for E-Cadherin mutation of peripheral blood circulating tumor cells of small cell lung cancer patients
CN111521791A (en) Kit and method for detecting NSE gene mutation of peripheral blood circulating tumor cells of small cell lung cancer patients
CN111521800A (en) Immunofluorescence kit for detecting gene expression of PD-L1 of colorectal cancer patient through peripheral blood circulating tumor cells
CN111638349A (en) Immunofluorescence kit for detecting expression of peripheral blood circulating tumor cells CA125 of gastric cancer patient and detection method
CN111534586B (en) Kit and method for detecting CEA gene mutation of peripheral blood circulating tumor cells of non-small cell lung cancer patients
CN111562376A (en) Kit and method for detecting peripheral blood circulation tumor cell PD-L1 gene mutation of gastric cancer patient
CN111596056A (en) Kit and method for detecting prostate cancer patient peripheral blood circulating tumor cell PD-L1 gene mutation
CN111551728A (en) Immunofluorescence kit and detection method for detecting expression of peripheral blood circulating tumor cells PD-L1 of pancreatic cancer patient
CN111551727A (en) Immunofluorescence kit and detection method for detecting ALK gene mutation of peripheral blood circulating tumor cells of non-small cell lung cancer patients
CN111521789A (en) Immunofluorescence kit and detection method for detecting expression of peripheral blood circulating tumor cells CA199 of pancreatic cancer patient

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
TR01 Transfer of patent right

Effective date of registration: 20240115

Address after: C416, Building 1, Overseas Students Pioneer Park, No. 69 Huayang Road, Lixia District, Jinan, Shandong 250000

Patentee after: Shandong Kaige Intelligent Machine Co.,Ltd.

Address before: No.18877, Jingshi Road, Lixia District, Jinan City, Shandong Province

Patentee before: Shandong First Medical University (Shandong Academy of Medical Sciences)

TR01 Transfer of patent right