CN111273012A - Method for combined detection of serum autoantibodies - Google Patents

Method for combined detection of serum autoantibodies Download PDF

Info

Publication number
CN111273012A
CN111273012A CN202010142815.9A CN202010142815A CN111273012A CN 111273012 A CN111273012 A CN 111273012A CN 202010142815 A CN202010142815 A CN 202010142815A CN 111273012 A CN111273012 A CN 111273012A
Authority
CN
China
Prior art keywords
ctag1a
elavl4
uchl1
gage7
magea4
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
CN202010142815.9A
Other languages
Chinese (zh)
Other versions
CN111273012B (en
Inventor
王红叶
李永哲
韩晓红
姚林
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.)
Yiling Beijing Biotechnology Co Ltd
Beijing Sanpin Medical Technology Co Ltd
Original Assignee
Yiling Beijing Biotechnology Co Ltd
Beijing Sanpin Medical Technology Co Ltd
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 Yiling Beijing Biotechnology Co Ltd, Beijing Sanpin Medical Technology Co Ltd filed Critical Yiling Beijing Biotechnology Co Ltd
Priority to CN202010142815.9A priority Critical patent/CN111273012B/en
Publication of CN111273012A publication Critical patent/CN111273012A/en
Application granted granted Critical
Publication of CN111273012B publication Critical patent/CN111273012B/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
    • 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
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54306Solid-phase reaction mechanisms
    • 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
    • G01N33/57423Specifically defined cancers of lung

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention belongs to the technical field of diagnosis or detection of tumor-related markers, and particularly relates to an application of an autoantibody or a detection reagent of the autoantibody in preparation of a product for diagnosing tumors and a method for combined detection of serum autoantibody. The antigen protein used in the method is selected from the combination of more than two of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ETHE 1. The invention coats antigen protein on solid phase carrier, and can detect the level of autoantibody with affinity to protein by detecting signal molecule strength. The method can well distinguish cancer from healthy people, and provides an optimization method for cancer screening by taking the autoantibody as an index.

Description

Method for combined detection of serum autoantibodies
Technical Field
The invention belongs to the technical field of diagnosis or detection of tumor-related markers, and particularly relates to an application of an autoantibody or a detection reagent of the autoantibody in preparation of a product for diagnosing tumors and a method for combined detection of serum autoantibody.
Background
According to the data of the world health organization, in 2018, the number of new cancer patients reaches 1810 ten thousand, and 960 ten thousand cancer patients die. 209 ten thousand new cases of lung cancer occurred globally in 2018, ranked first among all cancer types. According to the statistics of the national cancer center in 2017, the incidence and mortality of lung cancer are ranked first in malignant tumors. The incidence was first ranked in men (20.27%) and second only to breast cancer in women (17.07%) at 14.94%. Mortality rates were first in both men and women, 23.89% and 17.70%, respectively. It is predicted that lung cancer mortality in china may increase by about 40% in 2015 to 2030 years.
Lung cancer can be divided histologically into two main categories: small Cell Lung Cancer (SCLC) and non-small cell lung cancer (NSCLC). NSCLC accounts for approximately 79% of the diagnosed lung cancers, including adenocarcinoma, squamous cell carcinoma, and large cell carcinoma. Despite the great advances in recent years in targeted therapies and immunotherapy for lung cancer, surgical resection, adjuvant radiotherapy and/or chemotherapy remain the first-choice methods for early treatment of NSCLC patients. Despite the improved survival rates of all cancers in recent years, the survival rates of lung cancer are still at a low level. In 2012 and 2015, the survival rate of lung cancer in men was 16.8%, which was 62.5% lower than that of thyroid cancer with the highest survival rate, and the survival rate of women was only 25.1%. The low survival rate of lung cancer is associated with limitations in diagnostic techniques, and most lung cancer patients are found at an advanced stage at the time of diagnosis, losing the best treatment opportunity. Currently, high-resolution (or low-dose) computed tomography (LDCT) of the breast is the only screening test that is effective in reducing early mortality from lung cancer. Many regions have established a project for free screening of high risk populations by LDCT. One study in the united states has shown that LDCT screening has a better early screening potential for lung cancer, or permits a 20% reduction in mortality in high risk groups with lung cancer. Clinical transformation remains a problem. LDCT also has the problem that a high false positive rate can lead to over-diagnosis to some extent. Repeated exposure to radiation may also lead to potential health hazards. Therefore, the discovery of non-invasive serological biomarkers for early lung cancer diagnosis would greatly facilitate lung cancer intervention and prevention. The method of LDCT after initial screening based on biomarkers or combining biomarker testing with LDCT becomes a better means for diagnosing lung cancer.
The tumor-associated autoantibodies detection provides an alternative to cancer detection, which is expected to be achieved by detecting high expression (relative to healthy and benign tumors) of autoantibodies in the serum of cancer patients.currently, in clinical use, more of the tumor-associated antigens, such as Carbohydrate Antigen (CA)125, CA19-9, carcinoembryonic antigen (CEA) and α fetal protein (AFP), are used in the increased amount in serum, so that the sensitivity and specificity of tumor-associated antigens in the early stages of cancer are not very poor, and in infection, tumor or pregnancy, the presence of these markers in serum is increased with the increase in tumor size, the false positive results of these markers may be obtained in the early stages of clinical use, the false positive results of the autoantibodies may be obtained in the early stages of detection, and the detection of autoantibodies may be achieved by a number of greater than the early stages of detection.
For a single antigenic protein, its autoantibodies appear only in 10% -30% of patients and are less sensitive as a diagnostic tool. This may be associated with tumor heterogeneity and diversity of immune responses. Since immune system responses vary among patients even for the same cancer, it is necessary to bind a plurality of autoantibodies to improve detection sensitivity while maintaining high specificity. In recent years, many studies have been directed to the detection of a range of autoantibodies for cancer diagnosis. For example, Zhang et al established a combination of seven autoantibody markers (14-3-3 ζ, c-Myc, MDM2, NPM1, p16, p53 and cyclin B1) to detect the corresponding autoantibody levels in the sera of lung cancer patients and healthy controls. The results show that this combination can increase the sensitivity to 68.9% and the specificity to 79.5%. They also tested a series of sera from lung cancer patients before they were diagnosed and found that elevated autoantibody levels could be detected more than four years before lung cancer diagnosis. The sensitivity and specificity were 76.0% and 73.2%, respectively (Dai, Liping, et al, "Autoantibodidesagar tumor-associated antigens in the early detection of Long cancer99(2016): 172-.
Autoantibodies to tumor-associated antigens can increase significantly months or even years prior to clinical symptoms. Autoantibodies are more easily detected than antigens through multiple amplification of B cell proliferation. The problem of low sensitivity of a single autoantibody is solved by simultaneously detecting a plurality of autoantibodies, and a method with high specificity and high sensitivity is provided for early diagnosis and postoperative monitoring of cancer.
Disclosure of Invention
The invention aims to provide a marker for diagnosing tumor, wherein the marker is an autoantibody, and early diagnosis of a tumor patient is realized by detecting the autoantibody. Solves the problem of poor specificity and sensitivity of taking antigen protein as a diagnostic marker, can be diagnosed five years before clinical symptoms appear, and provides a method with high specificity and high sensitivity for early diagnosis and postoperative detection of cancer.
In order to achieve the above object, the present invention provides an autoantibody or an application of a detection reagent for the autoantibody in preparing a product for diagnosing a tumor, wherein the autoantibody is selected from a combination of two or more of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ete 1.
Wherein, the two or more can be two, three, four, five, six, seven or eight.
Preferably, the autoantibody is IgG and/or IgA. Further preferably, the autoantibody comprises IgG1, IgG2, IgG3, IgG4, IgA1, IgA2, IgM, IgE or IgD.
Preferably, the autoantibody is selected from the group consisting of five, six, seven or eight of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ete 1.
Preferably, the detection reagent comprises an antigenic protein selected from the group consisting of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ete 1.
Wherein, the two or more can be two, three, four, five, six, seven or eight.
Further preferably, the antigenic protein is selected from the group consisting of five, six, seven or eight of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ete 1.
In one embodiment of the invention, the autoantibody is selected from any one of the following combinations:
(a)CTAG1A,ELAVL4,UCHL1,p53,ETHE1;
(b)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,ANXA1;
(c)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,MAGEA4;
(d)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,GAGE7;
(e)ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(f)CTAG1A,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(g)CTAG1A,ANXA1,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(h)CTAG1A,ANXA1,MAGEA4,UCHL1,GAGE7,p53,ETHE1;
(i)CTAG1A,ANXA1,MAGEA4,ELAVL4,GAGE7,p53,ETHE1;
(j)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,p53,ETHE1;
(k)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,ETHE1;
(l) CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p 53; or
(m)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1。
Preferably, the tumor is lung cancer. The lung cancer includes Small Cell Lung Cancer (SCLC) and non-small cell lung cancer (NSCLC). Further preferred include, but are not limited to, lung adenocarcinoma, lung squamous cell carcinoma, large cell lung cancer or small cell lung cancer.
The invention also provides a product for diagnosing tumors, which comprises a detection reagent of the autoantibody, wherein the autoantibody is selected from the combination of more than two of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ETHE 1.
Wherein, the two or more can be two, three, four, five, six, seven or eight.
Preferably, the autoantibody is IgG and/or IgA. Further preferably, the autoantibody comprises IgG1, IgG2, IgG3, IgG4, IgA1, IgA2, IgM, IgE or IgD.
Preferably, the autoantibody is selected from the group consisting of five, six, seven or eight of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ete 1.
Preferably, the detection reagent comprises an antigenic protein selected from the group consisting of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ete 1.
Wherein, the two or more can be two, three, four, five, six, seven or eight.
Further preferably, the antigenic protein is selected from the group consisting of five, six, seven or eight of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ete 1.
In one embodiment of the invention, the autoantibody is selected from any one of the following combinations:
(a)CTAG1A,ELAVL4,UCHL1,p53,ETHE1;
(b)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,ANXA1;
(c)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,MAGEA4;
(d)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,GAGE7;
(e)ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(f)CTAG1A,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(g)CTAG1A,ANXA1,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(h)CTAG1A,ANXA1,MAGEA4,UCHL1,GAGE7,p53,ETHE1;
(i)CTAG1A,ANXA1,MAGEA4,ELAVL4,GAGE7,p53,ETHE1;
(j)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,p53,ETHE1;
(k)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,ETHE1;
(l) CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p 53; or
(m)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1。
Preferably, the tumor is lung cancer. The lung cancer includes Small Cell Lung Cancer (SCLC) and non-small cell lung cancer (NSCLC). Further preferred include, but are not limited to, lung adenocarcinoma, lung squamous cell carcinoma, large cell lung cancer or small cell lung cancer.
The invention further provides a method for detecting the autoantibody, wherein the autoantibody is selected from the combination of more than two of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ETHE 1.
Wherein, the two or more can be two, three, four, five, six, seven or eight.
Preferably, the autoantibody is IgG and/or IgA. Further preferably, the autoantibody comprises IgG1, IgG2, IgG3, IgG4, IgA1, IgA2, IgM, IgE or IgD.
Further preferably, the autoantibody is selected from the group consisting of five, six, seven or eight of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ete 1.
Preferably, the autoantibody is selected from any one of the following combinations:
(a)CTAG1A,ELAVL4,UCHL1,p53,ETHE1;
(b)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,ANXA1;
(c)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,MAGEA4;
(d)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,GAGE7;
(e)ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(f)CTAG1A,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(g)CTAG1A,ANXA1,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(h)CTAG1A,ANXA1,MAGEA4,UCHL1,GAGE7,p53,ETHE1;
(i)CTAG1A,ANXA1,MAGEA4,ELAVL4,GAGE7,p53,ETHE1;
(j)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,p53,ETHE1;
(k)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,ETHE1;
(l) CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p 53; or
(m)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1。
Preferably, the detection method comprises immobilizing the antigenic protein on a solid support. Wherein the antigen protein is selected from combination of more than two of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ETHE 1. Wherein, the two or more can be two, three, four, five, six, seven or eight. Further preferably, the antigenic protein is selected from the group consisting of five, six, seven or eight of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ete 1.
Preferably, the solid phase carrier includes but is not limited to one or a combination of more than two of an enzyme-labeled micro-porous plate, a micro-particle, a micro-sphere, an affinity membrane, a liquid phase chip, a glass slide, a test strip and a plastic sphere.
Preferably, the method for immobilizing the antigen protein includes a direct coating method and an indirect coating method. Wherein, the direct coating method is to directly fix the antigen protein on a solid phase carrier. The indirect coating method is to indirectly fix the antigen on the solid phase carrier through the specific reaction between biotin and streptavidin.
Preferably, the detection method includes signal detection by visible light color development, chemiluminescence, fluorescence, or the like.
Preferably, the detection reagent further comprises a secondary antibody containing a marker, a blocking buffer and/or a washing buffer, and the like.
Preferably, the use thereof is in the diagnosis of tumors; further preferably, the detection method is used for auxiliary diagnosis of lung cancer.
Preferably, the tumor is lung cancer. The lung cancer includes Small Cell Lung Cancer (SCLC) and non-small cell lung cancer (NSCLC). Further preferred include, but are not limited to, lung adenocarcinoma, lung squamous cell carcinoma, large cell lung cancer or small cell lung cancer.
The antigen protein of the invention can be natural or artificial. In a specific embodiment of the present invention, the antigen protein is artificially synthesized, and specifically comprises the steps of using a cloning plasmid containing a target gene as a template, amplifying a desired gene fragment by PCR, introducing the amplified gene fragment into escherichia coli, yeast or mammalian cells, expressing and purifying the amplified gene fragment to obtain the antigen protein. The autoantibodies of the invention may be in the serum, plasma, interstitial fluid, cerebrospinal fluid or urine of a human or non-human animal. The non-human animal is a non-human mammal, including but not limited to mouse, rat, monkey, zebrafish, pig, chicken, rabbit, etc.
The "product" of the present invention includes, but is not limited to, a kit containing reagents required for the reaction of an antigenic protein with an autoantibody, reagents required for the binding of a solid support and an antigenic protein to a solid support, and reagents conventionally selected such as a reagent for eluting unbound substances.
"tumors" as referred to herein include, but are not limited to, lymphoma, non-small cell lung cancer, leukemia, ovarian cancer, breast cancer, endometrial cancer, colon cancer, rectal cancer, gastric cancer, bladder cancer, lung cancer, bronchial cancer, bone cancer, prostate cancer, pancreatic cancer, liver and biliary tract cancer, esophageal cancer, kidney cancer, thyroid cancer, head and neck cancer, testicular cancer, glioblastoma, astrocytoma, melanoma, myelodysplastic syndrome, and sarcoma. Wherein the leukemia is selected from acute lymphocytic (lymphoblastic) leukemia, acute myelogenous leukemia, chronic lymphocytic leukemia, multiple myeloma, plasma cell leukemia, and chronic myelogenous leukemia; said lymphoma is selected from Hodgkin's lymphoma and non-Hodgkin's lymphoma, including B-cell lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, marginal zone B-cell lymphoma, T-cell lymphoma, and Waldenstrom's macroglobulinemia; the sarcoma is selected from osteosarcoma, Ewing's sarcoma, leiomyosarcoma, synovial sarcoma, soft tissue sarcoma, angiosarcoma, liposarcoma, fibrosarcoma, rhabdomyosarcoma, and chondrosarcoma. Preferably, the tumor is lung cancer. In one embodiment of the invention, the tumor is lung adenocarcinoma, lung squamous cell carcinoma, large cell lung cancer or small cell lung cancer.
"diagnosis" as used herein refers to the determination of whether a patient has suffered from a disease or condition in the past, at the time of diagnosis, or in the future, or the determination of the progression or likely progression of a disease in the future, or the assessment of a patient's response to a therapy.
The invention provides a novel cancer detection method, which improves the sensitivity and specificity of detection by detecting a plurality of autoantibodies, and provides possibility for clinical cancer screening and auxiliary diagnosis; the operation is convenient and simple, the cost is low, and the serum can be used as a detection sample, so that the harm to patients is low. The invention has good application prospect.
Obviously, many modifications, substitutions, and variations are possible in light of the above teachings of the invention, without departing from the basic technical spirit of the invention, as defined by the following claims.
The foregoing aspects of the present invention are explained in further detail below with reference to specific embodiments. This should not be understood as limiting the scope of the above-described subject matter of the present invention to the following examples. All the technologies realized based on the above contents of the present invention belong to the scope of the present invention.
Drawings
Embodiments of the invention are described in detail below with reference to the attached drawing figures, wherein:
FIG. 1: comparing the levels of a plurality of autoantibodies in lung cancer patients to healthy controls, wherein figure 1A is CTAG 1A; FIG. 1B is ANXA 1; FIG. 1C is MAGEA 4; FIG. 1D is ELAVL 4; fig. 1E is UCHL 1; FIG. 1F is GAGE 7; FIG. 1G is p 53; fig. 1H is ete 1.
FIG. 2: ROC analysis of autoantibodies in plasma of lung cancer patients and healthy control sera, wherein FIG. 2A is CTAG 1A; FIG. 2B is ANXA 1; FIG. 2C is MAGEA 4; FIG. 2D is ELAVL 4; fig. 2E is UCHL 1; FIG. 2F is GAGE 7; FIG. 2G is p 53; fig. 2H is ete 1.
FIG. 3: ROC profiles of different antigen combinations in lung cancer and healthy patients, where fig. 3A is CTAG1A, ELAVL4, UCHL1, p53, ete 1; FIG. 3B shows CTAG1A, ELAVL4, UCHL1, p53, ETHE1, ANXA 1; FIG. 3C shows CTAG1A, ELAVL4, UCHL1, p53, ETHE1, MAGEA 4; FIG. 3D shows CTAG1A, ELAVL4, UCHL1, p53, ETHE1, GAGE 7; FIG. 3E shows ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53, ETHE 1; FIG. 3F shows CTAG1A, MAGEA4, ELAVL4, UCHL1, GAGE7, p53, ETHE 1; FIG. 3G is CTAG1A, ANXA1, ELAVL4, UCHL1, GAGE7, p53, ETHE 1; FIG. 3H is CTAG1A, ANXA1, MAGEA4, UCHL1, GAGE7, p53, ETHE 1; FIG. 3I is CTAG1A, ANXA1, MAGEA4, ELAVL4, GAGE7, p53, ETHE 1; FIG. 3J shows CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, p53, ETHE 1; FIG. 3K is CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, ETHE 1; FIG. 3L shows CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p 53; FIG. 3M shows CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53, ETHE 1.
Detailed Description
The technical solutions in the embodiments of the present invention will be clearly and completely described below with reference to the drawings in the embodiments of the present invention, and it is obvious that the described embodiments are only some embodiments of the present invention, and not all embodiments. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
Example 1 recombinant antigenic protein expression and purification
The cloning plasmid containing the target gene is used as a template, and the required gene segment is obtained by PCR amplification. The amino acid sequence of the CTAG1A can be shown as SEQ ID NO. 1; the amino acid sequence of the ANXA1 can be shown as SEQ ID NO. 2; the amino acid sequence of the MAGEA4 can be shown as SEQ ID NO. 3; the amino acid sequence of ELAVL4 can be shown as SEQ ID NO. 4; the amino acid sequence of UCHL1 can be shown as SEQ ID NO. 5; the amino acid sequence of the GAGE7 can be shown as SEQ ID NO. 6; the amino acid sequence of the p53 can be shown as SEQ ID NO. 7; the amino acid sequence of the ETHE1 can be shown as SEQ ID NO. 8.
The plasmid vector pET-21a (+) is cut by BamHI and EcoRI for double enzyme cutting, after agarose electrophoresis of the cut product, the gel recovery Kit is used for recovering the large fragment of the vector, the PCR product is also cut by BamHI and EcoRI for double enzyme cutting and then is recovered, and is connected into the vector under the action of recombinase, the connection product is transformed into Escherichia coli DH5 α, screening is carried out according to the mark (anti-ampicillin) of the recombinant vector, single spots are picked, and the obtained recombinant plasmid is confirmed to contain correct exogenous fragments by colony PCR and sequencing identification.
The recombinant plasmid containing the antigen fragment obtained in the above is transformed into a competent cell of escherichia coli BL21(DE3), the induction expression is carried out through isopropyl thio- β -D-galactoside (IPTG), the obtained recombinant antigen protein is purified through a Ni-NTA column and a molecular sieve in two steps and then quantified through a Bradford method, the expression, purification and quantification results of the antigen protein are identified and confirmed through SDS-PAGE, and the obtained antigen protein is stored for later use.
Example 2 Combined detection of multiple autoantibodies
First, clinical sample
160 lung cancer patients and 107 healthy controls were selected, and the basic information is shown in table 1:
TABLE 1
Figure BDA0002399682620000101
Second, detection principle
The antigen proteins of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ETHE1 prepared in the example 1 are fixed on an enzyme-labeled pore plate, after serum is added for incubation, each antigen protein in the serum is combined with autoantibodies (mainly comprising IgG and IgA antibodies and also other types of antibodies) corresponding to the autoantibodies, the unbound antibodies and other proteins are washed away, an anti-human IgG horseradish peroxidase labeled secondary antibody is added for incubation, the unbound secondary antibody is washed away, a horseradish peroxidase substrate is added, after the substrate is developed, the substrate is detected by an enzyme reader, and the strength of the signal is in positive correlation with the number of the autoantibodies.
Third, method
The reagents used in this section are shown in table 2:
TABLE 2
Figure BDA0002399682620000102
Figure BDA0002399682620000111
1) Coating antigen protein: diluting the protein by using a coating buffer solution, respectively adding 50 ng/hole of the diluted protein into reaction holes of a polystyrene plate, and standing overnight at 4 ℃;
2) rewarming: and taking the enzyme label plate out of a refrigerator at 4 ℃, putting the enzyme label plate at room temperature for rewarming for half an hour, and washing for four times.
3) And (3) sealing: adding a confining liquid, incubating for one hour at 37 ℃, and washing twice;
4) adding a serum sample: adding a 1:200 diluted serum sample, placing the serum sample in a side-swinging shaking table at 12rpm, incubating for 2.5h at room temperature, washing for four times (the serum needs to be placed in a4 ℃ freeze-thaw in advance, and the diluted serum is vibrated and mixed uniformly and then added with the sample);
5) adding an enzyme-labeled antibody: to each reaction well, horseradish peroxidase-labeled anti-human IgG (4. mu.g/ml) was added. Placing the mixture on a side-swinging shaking table at 12rpm, incubating the mixture for 1 hour at room temperature in a dark place, and washing the mixture twice;
6) adding a chromogenic substrate: adding 50 μ l of temporarily prepared TMB substrate solution into each reaction well;
7) and (4) terminating: after the liquid in the wells turned blue, 25. mu.l of 2M H was added to each well2SO4
8) And (4) computer reading: readings of 0D 450 were measured in a TECAN F50 microplate reader and the data were saved.
Different antigen combinations were chosen as follows:
1.CTAG1A,ELAVL4,UCHL1,p53,ETHE1。
2.CTAG1A,ELAVL4,UCHL1,p53,ETHE1,ANXA1。
3.CTAG1A,ELAVL4,UCHL1,p53,ETHE1,MAGEA4。
4.CTAG1A,ELAVL4,UCHL1,p53,ETHE1,GAGE7。
5.ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1。
6.CTAG1A,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1。
7.CTAG1A,ANXA1,ELAVL4,UCHL1,GAGE7,p53,ETHE1。
8.CTAG1A,ANXA1,MAGEA4,UCHL1,GAGE7,p53,ETHE1。
9.CTAG1A,ANXA1,MAGEA4,ELAVL4,GAGE7,p53,ETHE1。
10.CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,p53,ETHE1。
11.CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,ETHE1。
12.CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53。
13.CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1。
the specificity and sensitivity of the different antigen combinations are shown in table 3:
TABLE 3
Figure BDA0002399682620000121
Fourth, result analysis
1. Based on the current sample analysis, the expression levels of CTAG1A (fig. 1A), ELAVL4 (fig. 1D), UCHL1 (fig. 1E), p53 (fig. 1G), and ete 1 (fig. 1H) autoantibodies in the plasma of lung cancer patients were shown to be statistically significantly different from healthy controls.
2. In the single-index autoantibody ROC analysis (fig. 2) of the lung cancer group and the healthy group, when the specificity was 90%, the sensitivities of CTAG1A (fig. 2A), ELAVL4 (fig. 2D), and p53 (fig. 2G) were better, 35.4%, 23.3%, and 32.3%, respectively.
3. In ROC analysis (fig. 3) of different antigen combinations to identify lung cancer and healthy people, sensitivity was seen to be significantly improved by combining different antigens when the specificity was 90% compared to a single index. Wherein the combination 7, 8, 9, 12 and 13 has a sensitivity of 51.7%, 52.4%, 51.7%, 52.0% and 51.7%, respectively, of 50% or more at a specificity of 90%.
From the above results, it can be seen that lung cancer and healthy persons can be identified by measuring the autoantibody level in a serum sample by the present method. Different autoantibody indexes are combined, which is helpful for improving the detection rate and accuracy of cancer diagnosis. The method has good application prospect in clinical auxiliary diagnosis.

Claims (10)

1. The application of the autoantibody or the detection reagent of the autoantibody in preparing a product for diagnosing tumors is characterized in that the autoantibody is selected from the combination of more than two of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ETHE 1.
2. The use of claim 1, wherein the detection reagent comprises an antigenic protein selected from the group consisting of a combination of two or more of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ete 1.
3. The use according to claim 1 or 2, wherein the autoantibody is selected from the group consisting of:
(a)CTAG1A,ELAVL4,UCHL1,p53,ETHE1;
(b)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,ANXA1;
(c)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,MAGEA4;
(d)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,GAGE7;
(e)ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(f)CTAG1A,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(g)CTAG1A,ANXA1,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(h)CTAG1A,ANXA1,MAGEA4,UCHL1,GAGE7,p53,ETHE1;
(i)CTAG1A,ANXA1,MAGEA4,ELAVL4,GAGE7,p53,ETHE1;
(j)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,p53,ETHE1;
(k)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,ETHE1;
(l) CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p 53; or
(m)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1。
4. The use of any one of claims 1-3, wherein the neoplasm is lung cancer.
5. A product for diagnosing a tumor, comprising a detection reagent for an autoantibody selected from the group consisting of two or more of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ETHE 1.
6. A method for detecting an autoantibody, wherein the autoantibody is selected from the group consisting of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ETHE 1.
7. The detection method according to claim 6, wherein the autoantibody is IgG and/or IgA; preferably, the autoantibodies comprise IgG1, IgG2, IgG3, IgG4, IgA1, IgA2, IgM, IgE or IgD.
8. The method of claim 6 or 7, wherein the autoantibody is selected from the group consisting of:
(a)CTAG1A,ELAVL4,UCHL1,p53,ETHE1;
(b)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,ANXA1;
(c)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,MAGEA4;
(d)CTAG1A,ELAVL4,UCHL1,p53,ETHE1,GAGE7;
(e)ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(f)CTAG1A,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(g)CTAG1A,ANXA1,ELAVL4,UCHL1,GAGE7,p53,ETHE1;
(h)CTAG1A,ANXA1,MAGEA4,UCHL1,GAGE7,p53,ETHE1;
(i)CTAG1A,ANXA1,MAGEA4,ELAVL4,GAGE7,p53,ETHE1;
(j)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,p53,ETHE1;
(k)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,ETHE1;
(l) CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p 53; or
(m)CTAG1A,ANXA1,MAGEA4,ELAVL4,UCHL1,GAGE7,p53,ETHE1。
9. The assay of any one of claims 6 to 8 which comprises immobilizing an antigenic protein selected from the group consisting of a combination of two or more of CTAG1A, ANXA1, MAGEA4, ELAVL4, UCHL1, GAGE7, p53 and ETHE1 on a solid support.
10. The detection method according to any one of claims 6 to 9, which is used for cancer diagnosis; preferably, the detection method is used in the auxiliary diagnosis of lung cancer.
CN202010142815.9A 2020-03-04 2020-03-04 Method for combined detection of serum autoantibodies Active CN111273012B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010142815.9A CN111273012B (en) 2020-03-04 2020-03-04 Method for combined detection of serum autoantibodies

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010142815.9A CN111273012B (en) 2020-03-04 2020-03-04 Method for combined detection of serum autoantibodies

Publications (2)

Publication Number Publication Date
CN111273012A true CN111273012A (en) 2020-06-12
CN111273012B CN111273012B (en) 2020-12-01

Family

ID=70997553

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010142815.9A Active CN111273012B (en) 2020-03-04 2020-03-04 Method for combined detection of serum autoantibodies

Country Status (1)

Country Link
CN (1) CN111273012B (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112394176A (en) * 2020-07-14 2021-02-23 迪瑞医疗科技股份有限公司 Lung cancer autoantibody P53 protein colloidal gold detection kit and preparation method thereof
CN114113611A (en) * 2021-12-13 2022-03-01 郑州大学 Biomarker for liver cancer diagnosis and detection kit
CN114167059A (en) * 2021-11-03 2022-03-11 郑州大学 Biomarker for diagnosing esophageal squamous carcinoma and detection kit
CN114924075A (en) * 2022-05-26 2022-08-19 郑州大学第一附属医院 Cardiac adenocarcinoma diagnosis biomarker screened based on focusing array protein chip and application thereof
CN116735879A (en) * 2023-06-06 2023-09-12 北京三品医疗科技有限公司 Application of polypeptide in diagnosis of lung cancer

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103869086A (en) * 2014-04-14 2014-06-18 杭州凯保罗生物科技有限公司 Serum autoantibody detection kit
WO2018187228A1 (en) * 2017-04-03 2018-10-11 Cdi Laboratories, Inc. Serological biomarkers for early diagnosis of lung cancer

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103869086A (en) * 2014-04-14 2014-06-18 杭州凯保罗生物科技有限公司 Serum autoantibody detection kit
WO2018187228A1 (en) * 2017-04-03 2018-10-11 Cdi Laboratories, Inc. Serological biomarkers for early diagnosis of lung cancer

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112394176A (en) * 2020-07-14 2021-02-23 迪瑞医疗科技股份有限公司 Lung cancer autoantibody P53 protein colloidal gold detection kit and preparation method thereof
CN114167059A (en) * 2021-11-03 2022-03-11 郑州大学 Biomarker for diagnosing esophageal squamous carcinoma and detection kit
CN114167059B (en) * 2021-11-03 2023-07-14 郑州大学 Biomarker and detection kit for diagnosis of esophageal squamous carcinoma
CN114113611A (en) * 2021-12-13 2022-03-01 郑州大学 Biomarker for liver cancer diagnosis and detection kit
CN114924075A (en) * 2022-05-26 2022-08-19 郑州大学第一附属医院 Cardiac adenocarcinoma diagnosis biomarker screened based on focusing array protein chip and application thereof
CN116735879A (en) * 2023-06-06 2023-09-12 北京三品医疗科技有限公司 Application of polypeptide in diagnosis of lung cancer
CN116735879B (en) * 2023-06-06 2024-04-09 北京三品医疗科技有限公司 Application of polypeptide in diagnosis of lung cancer

Also Published As

Publication number Publication date
CN111273012B (en) 2020-12-01

Similar Documents

Publication Publication Date Title
CN111273012B (en) Method for combined detection of serum autoantibodies
CN111337678B (en) Biomarker related to tumor immunotherapy effect and application thereof
Caron et al. Cancer immunomics using autoantibody signatures for biomarker discovery
Li et al. Serum carbonic anhydrase 9 level is associated with postoperative recurrence of conventional renal cell cancer
Lacombe et al. Use of autoantibodies to detect the onset of breast cancer
Shan et al. A cancer/testis antigen microarray to screen autoantibody biomarkers of non-small cell lung cancer
WO2022083673A1 (en) Biomarker for esophageal cancer, and use thereof
Yi et al. Autoantibody to tumor antigen, alpha 2-HS glycoprotein: a novel biomarker of breast cancer screening and diagnosis
JP2015500478A (en) Nucleosome adduct detection method
US20140274794A1 (en) Methods and Compositions for Diagnosis of Ovarian Cancer
CN112345755A (en) Biomarker of breast cancer and application thereof
Kobayashi et al. Identification of specific and common diagnostic antibody markers for gastrointestinal cancers by SEREX screening using testis cDNA phage library
CN109142729B (en) Lung cancer marker anti-HMGB 3 autoantibody and application thereof
KR20050114604A (en) Use of nicotinamide n-methyltransferase as a marker for colorectal cancer
Draghici et al. Epitomics: serum screening for the early detection of cancer on microarrays using complex panels of tumor antigens
EP3317671B1 (en) Use of cell-free nucleosomes as biomarkers in sputum samples
Lam et al. Interference-free HER2 ECD as a serum biomarker in breast cancer
Byström et al. Affinity proteomics exploration of melanoma identifies proteins in serum with associations to T-stage and recurrence
KR102208140B1 (en) Methods and arrays for use in biomarker detection for prostate cancer
CN110687283A (en) Use of autoantibodies for diagnosis and/or treatment of tumors
Chen et al. Development of a panel of serum IgG and IgA autoantibodies for early diagnosis of colon cancer
EP3129785B1 (en) Diagnosis of cancer by detecting dimeric il-18
CN109116023B (en) Lung cancer marker anti-MMP 12 autoantibody and application thereof
WO2008028066A2 (en) Boris isoforms and methods of detecting and treating disease
CN115372616A (en) Gastric cancer related biomarker and application thereof

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