CN112345755B - Biomarker for breast cancer and application thereof - Google Patents

Biomarker for breast cancer and application thereof Download PDF

Info

Publication number
CN112345755B
CN112345755B CN202011006719.8A CN202011006719A CN112345755B CN 112345755 B CN112345755 B CN 112345755B CN 202011006719 A CN202011006719 A CN 202011006719A CN 112345755 B CN112345755 B CN 112345755B
Authority
CN
China
Prior art keywords
cip2a
braf
akap4
brca2
breast cancer
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.)
Active
Application number
CN202011006719.8A
Other languages
Chinese (zh)
Other versions
CN112345755A (en
Inventor
孙苏彭
朱得坤
隗啸南
杨盼盼
周静
康美华
孙立平
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.)
Hangzhou Kaibaoluo Biological Science & Technology Co ltd
Original Assignee
Hangzhou Kaibaoluo Biological Science & 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 Hangzhou Kaibaoluo Biological Science & Technology Co ltd filed Critical Hangzhou Kaibaoluo Biological Science & Technology Co ltd
Priority to CN202011006719.8A priority Critical patent/CN112345755B/en
Publication of CN112345755A publication Critical patent/CN112345755A/en
Priority to PCT/CN2021/119819 priority patent/WO2022063156A1/en
Application granted granted Critical
Publication of CN112345755B publication Critical patent/CN112345755B/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
    • G01N33/57488Immunoassay; 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 involving compounds identifable in body fluids
    • 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
    • G01N33/57415Specifically defined cancers of breast

Abstract

The invention provides a biomarker for breast cancer, which is an autoantibody combination and comprises at least three autoantibodies respectively resisting the following tumor antigens: BRAF, CIP2A, AKAP, GIPC1, BRCA2, P53, SOX2, NY-ESO-1 and MUC-1. Early screening for breast cancer can be achieved by detecting the biomarker. The invention also provides an antigen protein combination for detecting the biomarker, a kit comprising the antigen protein combination, and a corresponding detection or diagnosis method.

Description

Biomarker for breast cancer and application thereof
Technical Field
The invention relates to the field of biotechnology and medical diagnosis, in particular to an autoantibody biomarker for breast cancer, an antigen combination for detecting the autoantibody biomarker, and application of the autoantibody biomarker and the antigen combination in breast cancer detection.
Background
Breast cancer is a malignant tumor that originates in the breast, and is a common malignant disease in women, with a high incidence in women between 40-55 years of age. In recent years, breast cancer has a tendency to be younger, and the incidence rate has been rising in women over 20 years old, and it is presumed that the recent years have been associated with an improvement in early screening and diagnosis means.
Early screening of breast cancer is an important secondary prevention because of the lack of effective etiology precautions. Imaging screening techniques have been traditionally used to detect breast cancer, such as mammography, ultrasound, breast MRI. However, these imaging screening techniques have drawbacks in that the detection results are related to the experience and quality of the physician, and the resolution is low, the specificity and sensitivity cannot be guaranteed, and therefore, more accurate, simple operation, low risk and non-invasive techniques need to be developed to supplement or replace.
Detection of tumor antigen markers has been proposed for screening of breast cancer, such as human epidermal growth factor receptor 2 (HER 2) antigen and carbohydrate antigen 15-3 (CA 15-3); however, it is difficult to demonstrate that these markers are breast cancer specific and susceptible to tumor burden, with limited value in early screening. In addition, detectable gene markers such as BRCA1 and BRCA2 or cfDNA or MicroRNAs (miRNAs) have been proposed, but also suffer from the disadvantage of not helping to reduce mortality from breast cancer or not being mature.
Autoantibodies refer to antibodies to self tissues, organs, cells, and cellular components. In early stages of cancer development, exposure of tumor-associated antigens can be recognized by the human immune system, producing tumor-associated autoantibodies, which can be sensitively detected by conventional means in the art, and which can maintain high levels of autoantibodies even in peripheral blood. It is well accepted in the art that autoantibodies generated by tumor antigens are good indicators for early diagnosis of tumors, and that the utilization of autoantibodies generated by tumor induction to reflect the disease progression of tumor pathogenesis in patients is becoming an important direction for finding new targets for early diagnosis and prognosis of tumors.
Studies have proposed the use of single autoantibody detection or combined detection of several autoantibodies in breast cancer screening. For example, the individual detection of p53 autoantibody, MUC1 autoantibody, HSP60 autoantibody, HSP90 autoantibody, HER2 autoantibody and the like and the simultaneous detection of several autoantibody direct targets prove to have certain sensitivity and specificity. However, due to tumor heterogeneity and differences in immune system response between individuals, the sensitivity of individual tumor autoantibodies in tumor patients is not high enough (typically 5% -20%). Thus, a single autoantibody biomarker is insufficient to provide information on the occurrence and progression of breast cancer, and the combined use of multiple different tumor autoantibodies can increase detection sensitivity.
Experiments prove that the existing autoantibody combination for detecting breast cancer, especially early breast cancer, still has the defects of insufficient sensitivity and the like, and new biomarkers, such as the autoantibody combination related to breast cancer specific antigens, still need to be discovered for early screening and auxiliary early diagnosis of the breast cancer.
Furthermore, breast cancer can be pathologically classified into three types, ER (+) positive, HER2 (+) positive and triple negative breast cancer. Triple negative breast cancer is defined as the lack of Estrogen Receptor (ER), the expression of the Progestin Receptor (PR) and the lack of overexpression or amplification of HER2, representing about 15% of breast cancer cases. This breast cancer has a poor prognosis and occurs more in young people. In addition, triple negative breast cancers also have a high diversity and can be divided into 6 subtypes: basal-like (BL 1 and BL 2), mesenchymal-like (M), mesenchymal stem cell-like (MSL), immunoregulatory (IM), luminal epithelial androgen receptor (luminal androgen receptor, LAR) and unidentified group (unspecified group, UNS). Approximately 20% of patients with triple negative breast cancer have good efficacy by standard treatment methods such as surgery, radiation and chemotherapy, while the remaining 80% develop fatal tumor metastasis (Turner N C, and Reis-Filho J S Clin Cancer Res 2013; 19:6380-6388).
Since triple negative breast cancer is easy to develop into a fatal and rapidly proliferated tumor, early detection of triple negative breast cancer has important significance. However, mammography has low sensitivity to high breast density, and triple negative breast cancer is frequently missed; while MRI is very sensitive, false positive rates are also very high. Therefore, noninvasive early detection of the blood markers of the triple negative breast cancer has important auxiliary significance in clinical application. However, the traditional antigenic markers CEA, CA27.29 and CA15-3 have higher sensitivity in advanced breast cancer, so that they are not suitable for early detection of breast cancer, and are only suitable for disease monitoring of metastatic breast cancer. Recently, there have emerged some potential blood markers as markers for early detection of triple negative breast cancer. Serum apoC-I mRNA and protein expression in triple-negative breast cancer is higher than in other types of breast cancer, and serum apoC-I can be used for early detection and prognosis of triple-negative breast cancer (Diagnostic and prognostic significance of serum apolipoprotein C-I in triple-negative breast cancer based on mass electrometry. Song D. Et al. Cancer Biol Ther.2016jun;17 (6): 635-647). In addition, a multicenter clinical trial found 3 tumor autoantibody markers for triple negative breast cancer, and the combination of the three markers found 27% of cases of triple negative breast cancer (A blinded multicenter phase II study of a panel of plasma biomarkers for the detection of triple negative breast cancer; karen SA et al.cancer Res 2015May1; 75 (9 Suppl): abstract P4-05)
Therefore, there is also a need to develop novel autoantibody combinations that also have good early screening effects on triple negative breast cancer.
Disclosure of Invention
The clinical significance of detecting breast cancer is that the earlier the tumor is found, the better the prognosis of the patient. Therefore, in order to solve the technical problems, the invention finally identifies a group of autoantibodies which can be used for screening breast cancer, especially early breast cancer, by detecting the autoantibodies aiming at different antigen targets in the blood of a breast cancer patient. The autoantibody combination is used as a biomarker, has high enough sensitivity in especially early tumors, and is especially used in experimental Chinese people; while also having a sufficiently high detection specificity.
It is therefore an object of the present invention to provide a biomarker for breast cancer that is an autoantibody combination.
Based on this autoantibody combination as biomarker, it is another object of the invention to provide reagents for detecting this autoantibody combination, such as antigen protein combinations; and provides the application of the autoantibody combination or the detection reagent in preparing products for predicting the risk of breast cancer, screening, prognosis evaluation, treatment effect monitoring or recurrence monitoring and the like.
It is a further object of the present invention to provide a kit and a method for the prediction of risk of developing breast cancer, screening, prognosis evaluation, treatment effect monitoring or recurrence monitoring, etc. accordingly.
The technical scheme of the invention is as follows.
In one aspect, the invention provides a biomarker for breast cancer, the biomarker being an autoantibody combination comprising at least three of autoantibodies respectively against the following tumor antigens: BRAF, CIP2A, AKAP, GIPC1, BRCA2, P53, SOX2, NY-ESO-1 and MUC-1.
Preferably, the autoantibody combination comprises autoantibodies against the following tumor antigens, respectively: BRAF, CIP2A and AKAP4.
More preferably, the autoantibody combination further comprises at least one of autoantibodies against the following tumor antigens, respectively: GIPC1, BRCA2, MUC-1, P53, SOX2 and NY-ESO-1. Wherein the autoantibody combination preferably comprises at least one, at least two or at least three of the autoantibodies against the following tumor antigens: GIPC1, BRCA2, MUC-1 and P53. For example, the autoantibody combination comprises autoantibodies against the following tumor antigens: BRCA2 and/or MUC-1; or: GIPC1 and/or BRCA2.
According to a specific embodiment of the invention, the autoantibody combination comprises autoantibodies against the following tumor antigens, respectively:
(1)BRAF、CIP2A、AKAP4;
(2)BRAF、CIP2A、AKAP4、GIPC1;
(3)BRAF、CIP2A、AKAP4、BRCA2;
(4)BRAF、CIP2A、AKAP4、MUC1;
(5)BRAF、CIP2A、AKAP4、P53;
(6)BRCA2、MUC-1、SOX2、P53;
(7)NY-ESO-1、GIPC1、MUC1、SOX2;
(8)NY-ESO-1、P53、SOX2、MUC1;
(9)GIPC1、P53、SOX2、BRCA2;
(10)BRAF、CIP2A、AKAP4、BRCA2、MUC-1;
(11)BRAF、CIP2A、AKAP4、GIPC1、BRCA2;
(12)BRAF、CIP2A、AKAP4、BRCA2、P53、MUC-1;
(13)BRAF、CIP2A、AKAP4、GIPC1、BRCA2、P53、SOX2;
(14)BRAF、CIP2A、AKAP4、GIPC1、BRCA2、P53、SOX2、MUC-1;
(15)BRAF、CIP2A、AKAP4、GIPC1、BRCA2、P53、SOX2、NY-ESO-1、MUC-1。
according to the invention, the autoantibodies are autoantibodies in whole blood, serum, plasma, tissue or cells, interstitial fluid, cerebrospinal fluid or urine of a subject; wherein preferably the tissue or cell is a breast tissue or cell, a breast cancer tissue or cell or a paracancestral tissue or cell of breast cancer.
Preferably, the subject is a mammal, preferably a primate mammal, more preferably a human.
Preferably, the autoantibody is IgA (e.g., igA1, igA 2), igM, or IgG (e.g., igG1, igG2, igG3, igG 4).
Preferably, the breast cancer includes ductal carcinoma in situ and invasive ductal carcinoma; wherein the invasive ductal carcinoma includes invasive ductal carcinoma, invasive micro-papillary carcinoma, invasive lobular carcinoma and mucous secretion carcinoma.
Preferably, the breast cancer includes triple negative and non-triple negative breast cancers.
According to the invention, the biomarker, i.e. the autoantibody combination, can be detected in a sample (e.g. plasma or serum) of the subject. In the present invention, "presence" or "absence" of autoantibodies is used interchangeably with "positive" or "negative"; this is judged as conventional in the art. For example, detection can be by a tumor-associated antigen and antigen-antibody specific reaction therebetween that results in the presence of any autoantibody in the combination. Accordingly, in a further aspect, the invention also provides a reagent for detecting a biomarker of the invention.
Depending on the specific technical means, the reagents may be reagents for enzyme-linked immunosorbent assay (ELISA), protein/peptide fragment chip detection, immunoblotting, microbead immunoassay, microfluidic immunoassay, or the like. Preferably, the reagents are used to detect the biomarkers of the invention by antigen-antibody reaction, for example by ELISA or fluorescent or chemiluminescent immunoassay.
In this aspect, the agent may be an antigen protein combination comprising at least three tumor antigens selected from the group consisting of: BRAF, CIP2A, AKAP, GIPC1, BRCA2, P53, SOX2, NY-ESO-1 and MUC-1.
Preferably, the antigen protein combination comprises the following tumor antigens: BRAF, CIP2A and AKAP4.
More preferably, the antigen protein combination further comprises at least one of the following tumor antigens: GIPC1, BRCA2, MUC-1, P53, SOX2 and NY-ESO-1. Wherein the antigen protein combination preferably comprises at least one, at least two or at least three of the following tumor antigens: GIPC1, BRCA2, MUC-1 and P53. For example, the antigen protein combination includes the following tumor antigens: BRCA2 and/or MUC-1; or: GIPC1 and/or BRCA2.
Further preferred, the antigen protein combination comprises the following tumor antigens:
(1)BRAF、CIP2A、AKAP4;
(2)BRAF、CIP2A、AKAP4、GIPC1;
(3)BRAF、CIP2A、AKAP4、BRCA2;
(4)BRAF、CIP2A、AKAP4、MUC1;
(5)BRAF、CIP2A、AKAP4、P53;
(6)BRCA2、MUC-1、SOX2、P53;
(7)NY-ESO-1、GIPC1、MUC1、SOX2;
(8)NY-ESO-1、P53、SOX2、MUC1;
(9)GIPC1、P53、SOX2、BRCA2;
(10)BRAF、CIP2A、AKAP4、BRCA2、MUC-1;
(11)BRAF、CIP2A、AKAP4、GIPC1、BRCA2;
(12)BRAF、CIP2A、AKAP4、BRCA2、P53、MUC-1;
(13)BRAF、CIP2A、AKAP4、GIPC1、BRCA2、P53、SOX2;
(14)BRAF、CIP2A、AKAP4、GIPC1、BRCA2、P53、SOX2、MUC-1;
(15)BRAF、CIP2A、AKAP4、GIPC1、BRCA2、P53、SOX2、NY-ESO-1、MUC-1。
in yet another aspect, the invention provides the use of said biomarker or agent in the manufacture of a product for the prediction of risk of developing breast cancer, screening, prognostic assessment, treatment effect monitoring or recurrence monitoring.
Preferably, the breast cancer includes ductal carcinoma in situ and invasive ductal carcinoma; wherein the invasive ductal carcinoma includes invasive ductal carcinoma, invasive micro-papillary carcinoma, invasive lobular carcinoma and mucous secretion carcinoma.
Preferably, the breast cancer includes triple negative and non-triple negative breast cancers.
Preferably, the product is a kit; more preferably, the kit is a kit for enzyme-linked immunosorbent assay (ELISA), protein/peptide fragment chip detection, immunoblotting, microbead immunoassay or microfluidic immunoassay; preferably, the kit is for detecting the biomarker by antigen-antibody reaction, for example an ELISA kit or a fluorescent or chemiluminescent immunoassay kit.
In yet another aspect, the invention provides a kit comprising the agent of the invention.
Depending on the specific technical means, the kit may be a kit for enzyme-linked immunosorbent assay (ELISA), protein/peptide fragment chip detection, immunoblotting, microbead immunoassay, microfluidic immunoassay, or the like. Preferably, the kit is used for detection of the biomarker of the invention by antigen-antibody reaction, for example an ELISA kit or a fluorescent or chemiluminescent immunoassay kit.
Thus, preferably, the kit is an enzyme-linked immunosorbent assay (ELISA) detection kit. That is, the kit is used to detect whether or not the autoantibody biomarker in the sample of the subject is positive by the enzyme-linked immunosorbent assay. Accordingly, the kit may also include other components required for ELISA detection of autoantibody biomarkers, all as is well known in the art. For detection purposes, for example, the antigen protein in the kit may be linked to a tag peptide, such as His tag, streptavidin tag, myc tag; for another example, the kit may include a solid support, such as a support having microwells to which antigen proteins can be immobilized, such as an elisa plate; or a microbead or magnetic bead solid phase carrier. It may also include an adsorption protein for immobilizing an antigen protein on a solid carrier, a dilution of blood such as serum, a washing solution, a secondary antibody with an enzyme label or a fluorescent or chemiluminescent substance, a color development solution, a stop solution, etc. The concentration of the corresponding antibody in the body fluid is detected by the principle that the antigen protein indirectly or directly coated on the surface of the solid carrier reacts with the antibody in serum/plasma/tissue fluid and the like to form an antigen-antibody complex.
In yet another aspect, the present invention provides a method for predicting risk of developing, screening, prognosis evaluation, treatment effect monitoring or recurrence monitoring of breast cancer, comprising the steps of:
(1) Quantifying each autoantibody in the autoantibody combination provided by the invention in a sample from a subject;
(2) Comparing the amount of the autoantibody to a reference threshold, and when it is above the reference threshold, determining that the subject is at risk of having breast cancer, has poor prognosis, or has poor treatment for breast cancer.
In step (1), the quantification comprises detection of each autoantibody in the autoantibody combination using the reagent (i.e. antigen protein combination) or the kit comprising the reagent provided by the invention.
According to the invention, the subject is a mammal, preferably a primate mammal, more preferably a human. And, preferably, the breast cancer is ductal carcinoma in situ, invasive ductal carcinoma, triple negative breast cancer, invasive lobular carcinoma, and breast mucinous carcinoma.
According to the invention, the sample is whole blood, serum, plasma, tissue or cells, interstitial fluid, cerebrospinal fluid or urine of a subject; wherein preferably the tissue or cell is a breast tissue or cell, a breast cancer tissue or cell or a paracancestral tissue or cell of breast cancer.
In step (2), the reference threshold may be a reference level from a healthy person or population of healthy people; for example, it may be defined as the mean value plus 2 standard deviations of the population confirmed by physical examination as not having cancer.
Compared with the prior art, the invention provides a novel biomarker for breast cancer, which is a brand-new group of tumor autoantibodies. The autoantibodies have higher positive detection rate in early breast cancer and have independent positive contribution rate; in addition, according to breast cancer stage analysis, the autoantibody combination has good detection sensitivity and high enough detection specificity, and can reach more than 50% of sensitivity even in early stage of breast cancer. In addition, experiments prove that the autoantibody combination has similar detection sensitivity to the triple negative breast cancer and the non-triple negative breast cancer, and the detection sensitivity can reach more than 50 percent.
Drawings
Embodiments of the present invention are described in detail below with reference to the attached drawing figures, wherein:
FIG. 1 is a scatter plot of the horizontal distribution of each tumor autoantibody in tumor and control groups.
FIG. 2 shows the results of sensitivity analysis of autoantibody combinations for patients with triple negative and non-triple negative breast cancer.
Detailed Description
In the present invention, the term "antigen" or the term "antigenic protein" is used interchangeably. Furthermore, the present invention relates to the following experimental operations or definitions. It should be noted that the present invention may also be practiced using other techniques conventional in the art and is not limited to the following experimental procedures.
Preparation of recombinant antigen proteins
The cDNA fragment of the tumor antigen was cloned into PET28 (a) expression vector containing the 6XHIS tag. At the N-or C-terminus of the antigen, streptavidin or the like (biotin-binding tag protein) is introduced. The obtained recombinant expression vector is transformed into escherichia coli for expression. The protein expressed by the supernatant was purified by Ni-NTA affinity column and ion column. When the protein is expressed in inclusion bodies, the protein is denatured by 6M guanidine hydrochloride, renaturated and folded in vitro according to a standard method, and then purified by a Ni-NTA affinity column through a 6XHIS tag, so that antigen protein is obtained.
(II) preparation and preservation of serum or plasma
Serum or plasma from a patient with breast cancer is collected when the patient is initially diagnosed with breast cancer and has not received any radiotherapy or chemotherapy or surgical treatment. Plasma or serum was prepared according to standard clinical procedures and stored in a-80 ℃ refrigerator for long periods of time.
(III) ELISA detection
The concentration of autoantibodies in the sample was quantified by enzyme-linked immunosorbent assay (ELISA).
The purified tumor antigen is immobilized to the microwell surface by its tag streptavidin or the like. Microwells were pre-coated with biotin-labeled Bovine Serum Albumin (BSA). Serum or plasma samples were diluted 1:110 fold with phosphate buffer and reacted by adding microwells (50 ml/well). After washing unbound serum or plasma components with wash solution, horseradish peroxidase (HRP) -conjugated anti-human IgG was added to each well for reaction. Then, TMB (3, 3', 5' -tetramethylbenzidine) as a reaction substrate was added for color development. Stop solution (1N HCl) was added and absorbance was measured at 450nm using a microplate reader (OD). Serum autoantibody concentrations were quantified using a standard curve.
Critical value (cutoff value) of autoantibody
The cutoff value of an autoantibody is defined as the mean value of the detected absorbance values plus 2 Standard Deviations (SD) or the mean value plus 3 Standard Deviations (SD) in a control normal population confirmed by physical examination to have no cancer. Determination of the cutoff value for each autoantibody was based on the following principle: 1. in the case of using two different values (average plus two standard deviations and average plus three standard deviations) as reference thresholds, the detection specificity of each autoantibody to the control normal population is 95% or more; 2. and under the condition that the two different values are adopted as reference thresholds, obtaining the specificity of each autoantibody to the control normal population and the sensitivity of each autoantibody to the breast cancer patient population, calculating the sum of the specificity and the sensitivity, and selecting the value with the sum being larger as the determined cutoff value of the autoantibody.
(V) determination of the positivity and negativity of an individual autoantibody
For each autoantibody assay, positive response is defined as quantifying the level of autoantibody in the sample, and then comparing it with the cutoff value, which is not less than the cutoff value positive; accordingly, a negative response is defined as < cutoff value negative.
Positive determination of autoantibody combinations
Since the single autoantibody has a low positive rate, the result is analyzed by combining the results of a plurality of autoantibodies to determine the predictive effect in order to increase the positive rate of autoantibody detection. The rules are: detecting a plurality of autoantibodies in a sample, and judging that the combined result of the antibodies is positive as long as one or more autoantibodies show positive; if all the autoantibodies are negative, the antibody combination result is judged to be negative.
(seventh) statistical analysis method
Both groups were statistically analyzed using GraphPad Prism v.6 (GraphPad Prism software, san diego, california) and IBM SPSS Statistics for Windows (IBM, new york) using the Mann-Whitney U test. In analyzing the relationship between each parameter, a Spearman correlation analysis was performed.
Eighth sensitivity and specificity determination
Sensitivity: in all breast cancer cases diagnosed by gold standard, the cases with positive detection results of autoantibodies or autoantibody combination account for the proportion of all the cases with diseases.
Specificity: among all subjects diagnosed with no disease by gold standard, the subjects whose autoantibodies or autoantibody combination detection results were negative are present in proportion to all subjects with no disease.
(nine) clinical assessment index
The Estrogen Receptor (ER), progestogen Receptor (PR) and human epidermal growth factor receptor 2 (HER 2) expression levels in breast cancer pathology reports were obtained according to conventional immunohistochemical and medical standardized pathology report analysis. ER and PR positivity are defined as > 1% of positively stained tumor cells.
In this study, ER expression levels were artificially grouped into:
ER (-), and: ER negative;
ER (medium): 1% -50% of positively stained tumor cells;
ER (high): >50% of positively stained tumor cells.
PR expression levels are artificially grouped into:
PR (-). PR negative;
PR (medium): 1% -10% of positively stained tumor cells;
PR (high): >10% of positively stained tumor cells.
The expression level of HER2 on the surface of breast cancer tissue cells was detected by immunohistochemistry, giving the results HER2 (0) to HER2 (3+). More than 10% of the cells developed a strong staining of the whole envelope of 3+. The expression level was 0 to 1+, and HER2 expression level was considered negative. If the result is that the expression level of 2+ is critical, in this case, the in situ hybridization method is further used to detect the HER2 gene amplification, and when the single copy HER2 gene is more than 6 or the HER2/CEP17 ratio is more than 2.0, the HER2 expression level is positive, otherwise, the HER2 expression level is negative.
The invention is described below with reference to specific examples. It will be appreciated by those skilled in the art that these examples are for illustration of the invention only and are not intended to limit the scope of the invention in any way. Sample collection has informed consent of the subject or patient and is approved by regulatory authorities.
The experimental methods in the following examples are conventional methods unless otherwise specified. The raw materials, reagent materials and the like used in the examples described below are commercially available products unless otherwise specified.
Example 1
Screening for tumor autoantibody markers was performed using a discovery cohort, comprising 84 healthy physical examination populations and 88 breast cancer patients. Serum specimens from breast cancer patients were from Shanghai tumor hospitals and healthy physical examination populations were from 4 different hospitals and physical examination centers. All breast cancer patient serum is collected when the patient is initially diagnosed as breast cancer and has not received any radiotherapy and chemotherapy and operation treatment, and is stored in a refrigerator at-80 ℃.
Patient information used for the discovery queue is shown in table 1.
Table 1: discovery queue patient feature table
Figure BDA0002696207410000101
Figure BDA0002696207410000111
25 breast cancer antigens are selected, expressed and purified, coated on the surface of a 96-well plate, sealed, reacted with breast cancer serum diluted by 1:110 times or serum of normal control population, then reacted with an anti-human IgG antibody-HRP horseradish catalase, then subjected to color development reaction, and detected by using an enzyme-labeled instrument OD450nm wavelength. Table 2 shows the detection sensitivity and specificity of each antigen.
Table 2: sensitivity and specificity of single tumor autoantibody markers of the discovery cohort
Figure BDA0002696207410000112
Figure BDA0002696207410000121
The 25 antigens were subjected to three-layer sorting as follows:
1. since the aim of this study was to find markers for early breast cancer detection, a portion of antigens with low positive detection rate in early (stage i+ii) breast cancer were first rejected, even though such antigens were more positive in late (stage iii+iv) breast cancer.
2. Antigens that have overlapping positive detection with other antigens and no individual positive contribution rate are eliminated.
3. The inventors found in previous studies that the control population had a greater bias in the sample of the physical examination population from different areas or hospitals. Therefore, the control crowd in a plurality of different places or hospitals can more accurately represent the actual scene of clinical application, so that the detection has more clinical significance. Thus, 100 new healthy control populations were used in the study to perform the serum test for the tumor antigen autoantibodies, and individual antigens with a specificity of 94% or less were removed from the total of 184 control serum.
A preferred set of autoantibody combinations was finally determined, including autoantibodies against the following 9 antigens: BRAF, CIP2A, AKAP4, GIPC1, BRCA2, P53, SOX2, MUC1 and NY-ESO-1, sensitivity and specificity in the queues were found to be 59.1% and 91.3%, respectively, as shown in Table 3. The detection sensitivity of these 9 autoantibody combinations in the discovery cohort was 50%, 56%, 60%, 62% and 60% in stages 0, I, II, III and IV, respectively, as analyzed by breast cancer stage, as shown in table 4.
Table 3: detection results of discovery of queued tumor autoantibody combinations
Number of people Number of positives Sensitivity to Specificity (specificity)
88 (patient) 52 59.1%
184 (health) 16 91.3%
Table 4: detection sensitivity of different stages of combination of tumor autoantibodies in queue was found
Stage by stage Number of patients Number of positives Sensitivity (%)
0 6 3 50
I 9 5 56
II 20 12 60
III 26 16 62
IV 15 9 60
Unknown 12 7 58
Example 2
The validation cohort included 100 breast cancer patients freshly collected (9 ductal carcinoma in situ, 87 invasive ductal carcinoma, 1 invasive papillary carcinoma, 1 invasive lobular carcinoma, 2 mucinous carcinomas), and 73 randomized physical examination populations collected from 4 different hospitals for validation of antibody markers. Patient information is shown in table 5.
Table 5: validation of patient characteristic table (for advanced patients, no IV phase validation queue)
Figure BDA0002696207410000131
Figure BDA0002696207410000141
For the antigens BRAF, CIP2A, AKAP4, GIPC1, BRCA2, P53, SOX2, MUC1 and NY-ESO-1, each tumor autoantibody was represented in a scatter plot of the level distribution of tumor groups and control groups (see FIG. 1). Due to the difference of the immune system of tumor patients and the diversity of tumor generation mechanisms, the distribution sensitivity of single tumor autoantibodies in tumor patients is low, and is usually only between 5 and 20 percent. Thus, the distribution of the antibody levels of tumor autoantibodies in the tumor and control groups was statistically analyzed using Mann Whitney test, and it was found that the distribution of the levels of autoantibodies against the tumor antigens BRAF, CIP2A and BRCA2 in the tumor and control groups were significantly different (p < 0.05); whereas the level distribution of autoantibodies against tumor antigens AKAP4, GIPC1, MUC1, SOX2, P53 and NY-ESO-1 was not significantly different in the tumor group and the control group. The main reason is presumed to be that most tumor patients were negative for their tumor autoantibody levels, and that there was no significant difference between their antibody levels and those of the normal control group. However, if the autoantibody levels greater than the cutoff value in the tumor group and the control group were analyzed to be distributed in both groups of people, the autoantibodies of AKAP4, GIPC1, MUC1, SOX2, P53 and NY-ESO-1 were significantly different in both groups (P < 0.05). This is consistent with the results of the sensitivity and specificity of each autoantibody marker in table 6 below. Because of this, these markers are of great significance for highly specific detection of breast cancer tumor patients.
The sensitivity and specificity of detection of 9 tumor autoantibody markers in 100 breast cancer sera and 73 healthy control sera of the validation cohort are shown in table 6. The specificity of the single marker is 97.3% or above, and the sensitivity is 6-17%.
Table 6: validation of detection sensitivity and specificity of individual autoantibodies of the cohort
Figure BDA0002696207410000142
Figure BDA0002696207410000151
Different antigens were selected from the 9 antigens to form different antigen combinations, from which detection of the corresponding tumor autoantibody combinations was performed, and the results are shown in table 7. It can be seen that as the number of antigens increases, the detection sensitivity increases and the specificity decreases gradually. The three autoantibody combinations (anti-BRAF, CIP2A and AKAP 4) detected sensitivity and specificity were 31% and 95.9%, respectively. On the basis of the combination of the three autoantibodies, one autoantibody such as anti-GIPC 1, BRCA2, MUC1 or P53 is added, the detection sensitivity is increased to 35-36%, and the specificity is 94.5-93.1%. The combination of other 4 different antibodies, excluding the three autoantibodies against BRAF, CIP2A and AKAP4, was 28-32% sensitive and 91.8-93.1% specific. The sensitivity and specificity of the autoantibody marker combinations comprising against 9 tumor antigens BRAF, CIP2A, AKAP, GIPC1, BRCA2, P53, SOX2, MUC1 and NY-ESO-1 in the validation cohort were 53% and 84.9%, respectively.
Table 7: verifying the sensitivity and specificity of combinations of different tumor antigens in a cohort to detect autoantibody markers
Figure BDA0002696207410000152
Example 3
Autoantibody marker combinations against 9 tumor antigens BRAF, CIP2A, AKAP4, GIPC1, BRCA2, P53, SOX2, MUC1 and NY-ESO-1 were selected for susceptibility analysis of different stages and types of breast cancer.
Referring to example 2, the validation cohort included 100 breast cancer patients, 9 of whom had Ductal Carcinoma In Situ (DCIS), 87 of which had Invasive Ductal Carcinoma (IDC), 1 of which had Invasive Lobular Carcinoma (ILC), 3 of which had invasive special breast cancer (1 of invasive micro papillary carcinoma, 2 of which had mucinous carcinoma), and the sensitivity of the tumor autoantibody combinations to detection was analyzed for different stages and subtypes.
Most breast cancer patients in the verification queue are in phase 0, phase I and phase II, the proportion is 84%, 9 cases of phase 0 patients have detection sensitivity of 56%,29 cases of phase I patients have detection sensitivity of 52%;46 patients with phase II, sensitivity was 56%; the sensitivity in 5 of the phase III patients was 80%. The results are shown in Table 8.
Table 8: verifying sensitivity of different stages of the queue autoantibody combination
Staging of tumors Number of patients Sensitivity to
0 9 56%
I 29 52%
II 46 56%
III 5 80%
Unknown 11 36%
In situ ductal carcinoma Ductal Carcinoma In Situ (DCIS) develops in the ducts of the breast, without invasiveness. The percentage of newly discovered breast cancer cases is about 20%. Invasive ductal carcinoma Invasive Ductal Carcinoma (IDC) is the most common type of breast cancer, accounting for about 80% of newly discovered breast cancer cases, which is produced in the ducts of the breast but has invaded other tissues of the breast and can be divided into five different types, tubular, medullary, mucinous, papillary and screen. Invasive lobular carcinoma Invasive Lobular Carcinoma (ILC) is the second most common type of breast cancer behind IDC, where tumors occur in the lobules that produce milk and invade other tissues of the breast, accounting for about 10% of the newly discovered cases of invasive breast cancer. According to different subtypes, the sensitivity of 9 catheter in situ cancers is 56%; invasive ductal carcinoma, invasive lobular carcinoma, and mucous-secreting carcinoma in the validation cohort were summed up to 90 cases (collectively referred to as IDC in table 9) with a sensitivity of 54% for detection. Only 1 case of invasive lobular carcinoma ILC breast cancer was collected, and the pathological diagnosis was phase I, without positive detection of tumor autoantibodies. The results are shown in Table 9.
Table 9: verifying sensitivity of different subtypes of a combination of queue autoantibodies
Tumor subtype Number of patients Sensitivity to
DCIS 9 56%
IDC 90 54
ILC
1 0
Example 4
The expression levels of the three hormone receptors ER, PR, HER of breast cancer tumor cells have important effects on both the growth of the tumor cells and the selection of therapeutic regimens. In the study, the expression levels of three hormone receptors ER, PR, HER2 in pathological sections of breast cancer tumors are analyzed and classified, and whether the expression level of ER, PR, HER2 influences detection of tumor autoantibodies is studied.
The results showed that the expression levels of the three hormone receptors ER, PR, HER2 did not greatly affect the autoantibody combination positive detection rate of the above 9 tumor antibodies. Of the 100 breast cancer patients, 12 were triple negative breast cancer patients, with a tumor autoantibody combination sensitivity of 58.3%, while the remaining 88 non-triple negative breast cancer patients had a tumor autoantibody combination sensitivity of 52.2%. Tumor autoantibody combinations are similarly sensitive in detection of early breast cancer patients, in both trinocular and non-trinocular patients. The results are shown in FIG. 2.
The above description of the embodiments of the present invention is not intended to limit the present invention, and those skilled in the art can make various changes or modifications according to the present invention without departing from the spirit of the present invention, and shall fall within the scope of the appended claims.

Claims (28)

1. A reagent for detecting a biomarker of breast cancer, the biomarker being an autoantibody combination comprising autoantibodies against the following tumour antigens respectively: BRAF, CIP2A and AKAP4.
2. The reagent of claim 1, wherein the autoantibody combination further comprises at least one of autoantibodies against the following tumor antigens, respectively: GIPC1, BRCA2, MUC-1, P53, SOX2, NY-ESO-1.
3. The agent of claim 1, wherein the autoantibody combination further comprises at least one, at least two or at least three of the autoantibodies against the following tumor antigens: GIPC1, BRCA2, MUC-1 and P53.
4. The agent of claim 1, wherein the combination of autoantibodies comprises autoantibodies against the following tumor antigens, respectively:
BRAF、CIP2A、AKAP4、GIPC1;
BRAF、CIP2A、AKAP4、BRCA2;
BRAF、CIP2A、AKAP4、MUC1;
BRAF、CIP2A、AKAP4、P53;
BRAF、CIP2A、AKAP4、BRCA2、MUC-1;
BRAF、CIP2A、AKAP4、GIPC1、BRCA2;
BRAF、CIP2A、AKAP4、BRCA2、P53、MUC-1;
BRAF、CIP2A、AKAP4、GIPC1、BRCA2、P53、SOX2;
BRAF, CIP2A, AKAP, GIPC1, BRCA2, P53, SOX2, MUC-1; or (b)
BRAF、CIP2A、AKAP4、GIPC1、BRCA2、P53、SOX2、NY-ESO-1、MUC-1。
5. The reagent of any one of claims 1 to 4, wherein the autoantibody is an autoantibody in whole blood, serum, plasma, tissue or cells, interstitial fluid, cerebrospinal fluid or urine of a subject.
6. The agent of claim 5, wherein the tissue or cell is breast tissue or cell, breast cancer tissue or cell, or a paracancerous tissue or cell of breast cancer.
7. The agent of claim 5, wherein the subject is a mammal.
8. The agent of claim 5, wherein the subject is a primate mammal.
9. The agent of claim 5, wherein the subject is a human.
10. The reagent according to any one of claims 1 to 4, wherein the autoantibody is IgA, igM or IgG.
11. The agent of any one of claims 1 to 4, wherein the breast cancer comprises ductal carcinoma in situ and invasive ductal carcinoma; wherein the invasive ductal carcinoma includes invasive ductal carcinoma, invasive micro-papillary carcinoma, invasive lobular carcinoma and mucous secretion carcinoma; alternatively, the breast cancer includes triple-negative and non-triple-negative breast cancers.
12. The reagent according to any one of claims 1 to 4, wherein the reagent is a reagent for enzyme-linked immunosorbent assay, protein/peptide fragment chip detection, immunoblotting, microbead immunoassay or microfluidic immunoassay.
13. The reagent according to any one of claims 1 to 4, wherein the reagent is used for detection of the biomarker by an antigen-antibody reaction.
14. The reagent according to any one of claims 1 to 4, wherein the reagent is used for detection of the biomarker by enzyme-linked immunosorbent assay or fluorescence or chemiluminescence immunoassay.
15. The agent according to any one of claims 1 to 4, characterized in that the agent is an antigen protein combination comprising the following tumor antigens: BRAF, CIP2A and AKAP4.
16. The reagent of claim 15, wherein the antigen protein combination further comprises at least one tumor antigen selected from the group consisting of: GIPC1, BRCA2, MUC-1, P53, SOX2, NY-ESO-1.
17. The reagent of claim 15, wherein the antigen protein combination comprises at least one, at least two, or at least three of the following tumor antigens: GIPC1, BRCA2, MUC-1 and P53.
18. The reagent of claim 15, wherein the antigen protein combination comprises the following tumor antigens:
BRAF、CIP2A、AKAP4、GIPC1;
BRAF、CIP2A、AKAP4、BRCA2;
BRAF、CIP2A、AKAP4、MUC1;
BRAF、CIP2A、AKAP4、P53;
BRAF、CIP2A、AKAP4、BRCA2、MUC-1;
BRAF、CIP2A、AKAP4、GIPC1、BRCA2;
BRAF、CIP2A、AKAP4、BRCA2、P53、MUC-1;
BRAF、CIP2A、AKAP4、GIPC1、BRCA2、P53、SOX2;
BRAF, CIP2A, AKAP, GIPC1, BRCA2, P53, SOX2, MUC-1; or (b)
BRAF、CIP2A、AKAP4、GIPC1、BRCA2、P53、SOX2、NY-ESO-1、MUC-1。
19. Use of an agent according to any one of claims 1 to 18 for the preparation of a product for disease risk prediction, screening, prognosis evaluation, treatment effect monitoring or recurrence detection of breast cancer.
20. The use of claim 19, wherein the breast cancer comprises ductal carcinoma in situ and invasive ductal carcinoma; wherein the invasive ductal carcinoma includes invasive ductal carcinoma, invasive micro-papillary carcinoma, invasive lobular carcinoma and mucous secretion carcinoma; alternatively, the breast cancer includes triple-negative and non-triple-negative breast cancers.
21. Use according to claim 19 or 20, wherein the product is a kit.
22. The use according to claim 21, wherein the kit is a kit for enzyme-linked immunosorbent assay, protein/peptide fragment chip detection, immunoblotting, microbead immunoassay or microfluidic immunoassay.
23. The use according to claim 21, wherein the kit is for detecting the biomarker by an antigen-antibody reaction.
24. The use according to claim 21, wherein the kit is an enzyme-linked immunosorbent assay kit or a fluorescent or chemiluminescent immunoassay kit.
25. A kit for detecting breast cancer comprising the reagent of any one of claims 1 to 18.
26. The kit of claim 25, wherein the kit is a kit for enzyme-linked immunosorbent assay, protein/peptide fragment chip detection, immunoblotting, microbead immunoassay, or microfluidic immunoassay.
27. The kit of claim 25, wherein the kit is for detecting the biomarker by an antigen-antibody reaction.
28. The kit of claim 25, wherein the kit is an enzyme-linked immunosorbent assay kit or a fluorescent or chemiluminescent immunoassay kit.
CN202011006719.8A 2020-09-23 2020-09-23 Biomarker for breast cancer and application thereof Active CN112345755B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202011006719.8A CN112345755B (en) 2020-09-23 2020-09-23 Biomarker for breast cancer and application thereof
PCT/CN2021/119819 WO2022063156A1 (en) 2020-09-23 2021-09-23 Biomarker in breast cancer and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011006719.8A CN112345755B (en) 2020-09-23 2020-09-23 Biomarker for breast cancer and application thereof

Publications (2)

Publication Number Publication Date
CN112345755A CN112345755A (en) 2021-02-09
CN112345755B true CN112345755B (en) 2023-06-16

Family

ID=74357719

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011006719.8A Active CN112345755B (en) 2020-09-23 2020-09-23 Biomarker for breast cancer and application thereof

Country Status (2)

Country Link
CN (1) CN112345755B (en)
WO (1) WO2022063156A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112345755B (en) * 2020-09-23 2023-06-16 杭州凯保罗生物科技有限公司 Biomarker for breast cancer and application thereof
CN113702636B (en) * 2021-08-02 2024-03-08 中国医学科学院北京协和医院 Application of plasma autoantibody marker in early diagnosis of breast cancer and molecular subtype characterization thereof
CN115372616B (en) * 2022-07-15 2023-09-15 杭州凯保罗生物科技有限公司 Gastric cancer related biomarker and application thereof
CN115877006B (en) * 2022-12-20 2023-12-15 杭州凯保罗生物科技有限公司 Ovarian cancer-related biomarker and application thereof
CN116298289B (en) * 2023-01-30 2023-11-03 上海秤信生物科技有限公司 Biomarker for predicting lung cancer immune new adjuvant therapy effect and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108601525A (en) * 2015-10-07 2018-09-28 明策生物医学科技私人有限公司 Utilize the integrated vision form and expression of cellular proteins of resonant light scattering
CN110716043A (en) * 2019-10-23 2020-01-21 郑州大学 Serum protein marker, kit and detection method for early screening and diagnosis of breast cancer

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9810040D0 (en) * 1998-05-11 1998-07-08 Univ Nottingham Blood borne tumour markers
GB2426581A (en) * 2005-05-27 2006-11-29 Univ Nottingham Immunoassay methods
SG10201604654RA (en) * 2012-01-25 2016-07-28 Dnatrix Inc Biomarkers and combination therapies using oncolytic virus and immunomodulation
KR101431062B1 (en) * 2012-03-08 2014-08-21 (주)바이오메디앙 Multiple biomarker set for breast cancer diagnosis, method of detecting the same, and diagnosis kit for breast cancer using antibody against the same
EP2857839A1 (en) * 2013-10-01 2015-04-08 AIT Austrian Institute of Technology GmbH Breast cancer diagnostic method and means
WO2016037136A1 (en) * 2014-09-04 2016-03-10 Provista Diagnostics, Inc. Biomarkers for detection of breast cancer
US20180126003A1 (en) * 2016-05-04 2018-05-10 Curevac Ag New targets for rna therapeutics
CN111337678B (en) * 2020-02-21 2023-06-06 杭州凯保罗生物科技有限公司 Biomarker related to tumor immunotherapy effect and application thereof
CN112345755B (en) * 2020-09-23 2023-06-16 杭州凯保罗生物科技有限公司 Biomarker for breast cancer and application thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108601525A (en) * 2015-10-07 2018-09-28 明策生物医学科技私人有限公司 Utilize the integrated vision form and expression of cellular proteins of resonant light scattering
CN110716043A (en) * 2019-10-23 2020-01-21 郑州大学 Serum protein marker, kit and detection method for early screening and diagnosis of breast cancer

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
A panel of autoantibodies as potential early diagnostic serum biomarkers in patients with breast cancer;Ying Liu 等;《International Journal of Clinical Oncology》;第22卷;第291-296页 *
Assessment of CIP2A and ROCK-I expression and their prognostic value in breast cancer;Nisreen Abdel Tawab Osman1 等;《Pol J Pathol》;第71卷(第2期);第87-98页 *
Breast cancer vaccines: Heeding the lessons of the past to guide a path forward;Cinzia Solinasa 等;《Cancer Treatment Reviews》;第84卷;第1-43页 *
Elaboration of protein microarrays for rapid screening and quantification of breast cancer biomarkers;Liu Shi 等;《Other. Ecole Centrale de Lyon》;第1-235页 *
Jérôme Lacombe 等.Use of Autoantibodies to Detect the Onset of Breast Cancer.《Journal of Immunology Research》.2014,第2014卷第1-8页. *
Proteomic Approaches in Biomarker Discovery: New Perspectives in Cancer Diagnostics;Petra Hudler 等;《The Scientific World Journal》;第2014卷;第1-18页 *

Also Published As

Publication number Publication date
WO2022063156A1 (en) 2022-03-31
CN112345755A (en) 2021-02-09

Similar Documents

Publication Publication Date Title
CN112345755B (en) Biomarker for breast cancer and application thereof
CN105755112B (en) The diagnosis of colorectal cancer
CN102749447B (en) Immunoassay Methods
CN112362871B (en) Biomarker for esophageal cancer and application thereof
US20110053156A1 (en) Small cell lung carcinoma biomarker panel
KR101032607B1 (en) Proteinic markers for diagnosing hepatocellular carcinoma
EP3545309B1 (en) Antibody assay
EP3283886B1 (en) Methods for treating lung cancer
US20140271621A1 (en) Methods of prognosis and diagnosis of pancreatic cancer
TWI408370B (en) A serological maker for detecting pancreatic cancer and a method for using the serological maker
US7846653B2 (en) ASC as a marker for colorectal cancer
WO2012109233A2 (en) Methods for predicting recurrence risk in breast cancer patients
WO2016195051A1 (en) Plasma biomarker panel for diagnosing pancreatic cancer
CN109116023B (en) Lung cancer marker anti-MMP 12 autoantibody and application thereof
CN116121392A (en) Methods and reagents for diagnosis of pancreatic cystic tumours
CN115372616A (en) Gastric cancer related biomarker and application thereof
EP3102600B1 (en) Composition and method for detecting malignant neoplastic disease
US20230375550A1 (en) Method for diagnosing breast cancer by using biomarker
KR102018209B1 (en) Diagnosis composition of gastric cancer and diagnosis method of gastric cancer using the same
Sekacheva et al. Clinical validation of the novel CLIA-CA-62 assay efficacy for early-stage breast cancer detection
CN115877006B (en) Ovarian cancer-related biomarker and application thereof
US20220390453A1 (en) Ovarian cancer biomarker and methods of using same
US20130095483A1 (en) Predictive biomarkers for breast cancer
CA3163199A1 (en) Ovarian cancer biomarker and methods of using same
CN116413430A (en) Autoantibody/antigen combination and detection kit for early prediction of liver cancer

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