HUMORAL ANTIBODY BIOMARKER FOR DETECTING ARTERY LESIONS, AND DETECTING CEREBROVASCULAR AND CARDIOVASCULAR DISORDER, DIABETES, CHRONIC NEPHROPATHY, OR SOLID CANCER

Abstract
An object of the invention is to provide a comprehensive biomarker for artery lesions, and diabetes mellitus, heart disease, cerebrovascular disorder, cancer, kidney disease, etc., whose onset and progress can be suppressed by health care such as early diagnosis. The inventors have found that the object can be attained by determining the level of an antibody to protein KIAA0513 or a portion of the protein, the antibody being potentially present in a body fluid sample collected from a living body; and acquiring data showing a rise in the antibody level as data showing the presence of an artery lesion or the potential or actual presence of cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer.
Description
TECHNICAL FIELD

The present invention relates to means for acquiring data concerning a disease or disorder (hereinafter also referred to simply as “disease”) or for detecting a disease. More particularly, the invention relates to means for acquiring data concerning an artery lesion or for detecting an artery lesion, and to means for acquiring data concerning cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, solid cancer, etc. or detecting such diseases, these means being based on measuring the level of a biomarker which is a humoral antibody with respect to protein KIAA0513 or a peptide having a partial sequence of the protein.


BACKGROUND ART

A variety of disease-specific biomarkers have been found in recent years, and these biomarkers are further refined from many aspects including assessment of the disease condition, prediction of the onset and prognosis of the disease, and determination of a prophylactic/therapeutic target, through a simple blood test or the like. Conventional biomarkers include an enzyme marker and an antigen marker, and a DNA/RNA marker is reported in recent years. In contrast, a limited number of humoral antibody markers have been reported or implemented. Meanwhile, many antibody markers have been identified with respect to cerebrovascular and cardiovascular disorders. For example, Hsp60 (Non-Patent Document 1), RPA2 (Non-Patent Document 2), SOSTDC1 (Non-Patent Document 3), PDCD11 (Non-Patent Document 4), MMP1, CBX1, and CBX5 (Non-Patent Document 5) are reported as antibody markers with respect to acute ischemic stroke (AIS); ATP2B4 (Non-Patent Document 6), BMP-1 (Non-Patent Document 2, 6), DHPS (Non-Patent Document 7), SH3BP5 (Non-Patent Document 8), and prolylcarboxypeptidase (Non-Patent Document 9) are reported as markers with respect to atherosclerosis; nardilysin (NRD1) is reported as a marker with respect to acute coronary syndrome (Non-Patent Document 10); and TUBB2C (Non-Patent Document 11), insulin (Non-Patent Document 12), glutamic acid decarboxylase (Non-Patent Document 13), adiponectin (Non-Patent Document 14), and GADD34 (Non-Patent Document 15) are reported as markers with respect to diabetes mellitus (DM).


Regarding antibody biomarkers with respect to cancers, an anti-p53 antibody has been implemented as a marker which can be effectively employed for the diagnosis, monitoring, and prediction of prognosis of esophageal squamous cell carcinoma (ESCC) or head and neck cancer (Non-Patent Documents 16 and 17). The group to which the present inventors belong previously conducted screening through serological identification of antigens by recombinant cDNA expression cloning (SEREX), and found and reported Trop2/TACSTD2 (Non-Patent Document 18), SLC2A1 (Non-Patent Document 19), TRIM21 (Non-Patent Document 20), myomegalin (Non-Patent Document 21), makorin 1 (Non-Patent Document 22), ECSA (Non-Patent Document 23), and cyclin L2 (CCNL2) (Non-Patent Document 24) as antibody markers with respect to ESCC; FIR/PUF60 (Non-Patent Document 25) as an antibody marker with respect to colorectal cancer; and SH3GL1 (Non-Patent Document 26) and anti-filamin C (Non-Patent Document 27) as antibody markers with respect to glioma.


PRIOR ART DOCUMENTS
Non-Patent Documents





    • Non-Patent Document 1: Kramer J, Harcos P, Prohaszka Z, et al. Frequencies of certain complement protein alleles and serum levels of anti-heat-shock protein antibodies in cerebrovascular diseases. Stroke. 2000; 31: 2648-2652. PMID: 11062289

    • Non-Patent Document 2: Machida T, Kubota M, Kobayashi E, et al. Identification of stroke-associated-antigens via screening of recombinant proteins from the human expression cDNA library (SEREX). J Translat Med. 2015; 13:71. PMID: 25890248

    • Non-Patent Document 3: Goto K, Sugiyama T, Matsumura R, et al. Identification of cerebral infarction-specific antibody markers from autoantibodies detected in patients with systemic lupus erythematosus. J Mol Biomark Diagnos. 2015; 6:2. doi.org/10.4172/2155-9929.1000219

    • Non-Patent Document 4: Yoshida Y, Wang H, Hiwasa T, et al. Elevation of autoantibody level against PDCD11 in patients with transient ischemic attack. Oncotarget. 2018; 9, 8836-8848. PMID: 29507658

    • Non-Patent Document 5: Wang H, Zhang X M, Tomiyoshi G, et al. Association of serum levels of antibodies against MMP1, CBX1, and CBX5 with transient ischemic attack and cerebral infarction. Oncotarget. 2018; 9: 5600-5613. PMID: 29464021

    • Non-Patent Document 6: Hiwasa T, Machida T, Zhang X M, et al. Elevated levels of autoantibodies against ATP2B4 and BMP-1 in sera of patients with atherosclerosis-related diseases. Immunome Res. 2015; 11:097. doi.org/10.4172/1745-7580.1000097

    • Non-Patent Document 7: Nakamura R, Tomiyoshi G, Shinmen N, et al. An anti-deoxyhypusine synthase antibody as a marker of atherosclerosis-related cerebral infarction, myocardial infarction, diabetes mellitus, and chronic kidney disease. S M Atheroscler J. 2017; 1:1001. doi: http://smjournals.com/atherosclerosis/in-press.php#x

    • Non-Patent Document 8: Hiwasa T, Tomiyoshi G, Nakamura R, et al. Serum SH3BP5-specific antibody level is a biomarker of atherosclerosis. Immunome Res. 2017; 13:132. doi: 10.4172/17457580.1000132

    • Non-Patent Document 9: Zhang X M, Wang H, Mine S, et al. Association of serum anti-prolylcarboxypeptidase antibody marker with atherosclerotic diseases accompanied by hypertension. J Mol Biomark Diagn. 2017; 8:361. doi: 10.4172/2155-9929.1000361

    • Non-Patent Document 10: Chen P M, Ohno M, Hiwasa T, et al. Nardilysin is a promising biomarker for the early diagnosis of acute coronary syndrome. Int J Cardiol. 2017; 243: 1-8. PMID: 28747015

    • Non-Patent Document 11: Hiwasa T, Zhang X M, Kimura R, et al. Association of serum antibody levels against TUBB2C with diabetes and cerebral infarction. Integ Biomed Sci. 2015; 1: 49-63. doi: 10.18314/gjbs.vli2.27

    • Non-Patent Document 12: Palmer J P, Asplin C M, Clemons P, et al. Insulin antibodies in insulin-dependent diabetics before insulin treatment. Science. 1983; 222:1337-1339. PMID: 6362005

    • Non-Patent Document 13: Baekkeskov S, Aanstoot H, Christgau S, et al. Identification of the 64K autoantigen in insulin dependent diabetes as the GABA-synthesizing enzyme glutamic acid decarboxylase. Nature. 1990; 347: 151-156. PMID: 1697648

    • Non-Patent Document 14: Hiwasa T, Zhang X M, Kimura R, et al. Elevated adiponectin antibody levels in sera of patients with atherosclerosis-related coronary artery disease, cerebral infarction, and diabetes mellitus. J Circ Biomark. 2016; 5:8. doi: 10.5772/63218

    • Non-Patent Document 15: Sugimoto K, Tomiyoshi G, Mori M, et al. Identification of serum anti-GADD34 antibody as a common marker of diabetes mellitus and Parkinson disease. J Alzheimers Dis Parkinsonism. 2017; 7:358. doi: 10.4172/2161-0460.1000358

    • Non-Patent Document 16: Shimada H, Takeda A, Arima M, et al. Serum p53 antibody is a useful tumor marker in superficial esophageal squamous cell carcinoma. Cancer. 2000; 89: 1677-1683. PMID: 11042560

    • Non-Patent Document 17: Shimada H, Ochiai T, Nomura F, et al. Japan p53 Antibody Research Group. Titration of serum p53 antibodies in 1,085 patients with various types of malignant tumors: a multiinstitutional analysis by the Japan p53 Antibody Research Group. Cancer. 2003; 97: 682-689. PMID: 12548611

    • Non-Patent Document 18: Nakashima K, Shimada H, Ochiai T, et al. Serological identification of TROP2 by recombinant cDNA expression cloning using sera of patients with esophageal squamous cell carcinoma. Int J Cancer. 2004; 112: 1029-1035. PMID: 15386348

    • Non-Patent Document 19: Kuboshima M, Shimada H, Liu T L, et al. Identification of a novel SEREX antigen, SLC2A1/GLUT1, in esophageal squamous cell carcinoma. Int J Oncol. 2006; 28: 463-468. PMID: 16391802

    • Non-Patent Document 20: Kuboshima M, Shimada H, Liu T L, et al. Presence of serum tripartite motif-containing 21 antibodies in patients with esophageal squamous cell carcinoma. Cancer Sci. 2006; 97: 380-386. PMID: 16630135

    • Non-Patent Document 21: Shimada H, Kuboshima M, Shiratori T, et al. Serum anti-myomegalin antibodies in patients with esophageal squamous cell carcinoma. Int J Oncol. 2007; 30: 97-103. PMID: 17143517

    • Non-Patent Document 22: Shimada H, Shiratori T, Yasuraoka M, et al. Identification of makorin 1 as a novel SEREX antigen of esophageal squamous cell carcinoma. BMC Cancer. 2009; 9:232. PMID: 19604354

    • Non-Patent Document 23: Kagaya A, Shimada H, Shiratori T, et al. Identification of a novel SEREX antigen family, ECSA, in esophageal squamous cell carcinoma. Proteome Sci. 2011; 9:31. PMID: 21696638

    • Non-Patent Document 24: Shimada H, Ito M, Kagaya A, et al. Elevated serum antibody levels against cyclin L2 in patients with esophageal squamous cell carcinoma. J Cancer Sci Ther. 2015; 7: 60-66. doi:10.4172/1948-5956.1000326

    • Non-Patent Document 25: Kobayashi S, Hoshino T, Hiwasa T, et al. Anti-FIRs (PUF60) auto-antibodies are detected in the sera of early-stage colon cancer patients. Oncotarget. 2016; 7:82493-82503. PMID: 27756887

    • Non-Patent Document 26: Matsutani T, Hiwasa T, Takiguchi M, et al. Autologous antibody to src-homology 3-domain GRB2-like 1 specifically increases in the sera of patients with low-grade gliomas. Exp Clin Cancer Res. 2012; 31:85. PMID: 23050879

    • Non-Patent Document 27: Adachi-Hayama M, Adachi A, Shinozaki N, et al. Circulating anti-filamin C antibody as a potential serum biomarker for low-grade gliomas. BMC Cancer. 2014; 14:452. PMID: 24946857

    • Non-Patent Document 28: Makrilia N, Lappa T, Xyla V, et al. The role of angiogenesis in solid tumours: an overview. Eur J Intern Med. 2009; 20: 663-671. PMID: 19818284

    • Non-Patent Document 29: Jarvandi S, Davidson N O, Schootman M. Increased risk of colorectal cancer in type 2 diabetes is independent of diet quality. PLoS One. 2013; 8:e74616. PMID: 24069323

    • Non-Patent Document 30: Fujihara S, Kato K, Morishita A, et al. Antidiabetic drug metformin inhibits esophageal adenocarcinoma cell proliferation in vitro and in vivo. Int J Oncol. 2015; 46: 2172-2180. PMID: 25709052

    • Non-Patent Document 31: Berger N A. Young Adult Cancer: Influence of the Obesity Pandemic. Obesity (Silver Spring). 2018; 26: 641-650. PMID: 29570247

    • Non-Patent Document 32: Hiwasa T, Shimada H. Autoantibody in Cancer. In: Shimada H, ed. Biomarkers in Cancer Therapy (ISBN: 978-981-13-7295-7), Singapore, Springer Nature. 2018: 25-40. https://doi.org/10.1007/978-981-13-7295-7_3





DISCLOSURE OF THE INVENTION
Problems to be Solved by the Invention

According to the Vital Statistics (2018) of the Ministry of Health, Labour and Welfare of Japan, cancers, heart diseases, cerebrovascular diseases, and renal failure are ranked as the first, second, fourth, and eighth leading causes of death of Japanese, respectively. Also, senescence, pneumonia, unexpected accidents, aspiration pneumonitis, dementia, and suicide are ranked as the third, fifth, sixth, seventh, ninth, and tenth causes of death of Japanese, respectively. According to the WHO, ischemic heart disease, cerebral stroke, trachea/bronchus/lung cancers, diabetes mellitus, and kidney diseases are ranked as the first, second, fifth, eighth, and ninth leading causes of death in the world (2019). Among these causes of death, cancers, heart diseases, cerebrovascular diseases, and renal failure, and diabetes mellitus which is a risk factor for such organic diseases, are conditions whose onset and progress can be prevented by health administration such as early diagnosis. Thus, if appropriate treatment, therapy, daily life guidance, etc. with respect to these conditions can be conducted by use of disease-specific markers, it can make a significant contribution to promotion of human health.


In the coronavirus (COVID-19) pandemic of 2020, the presence of pre-existing condition (e.g., diabetes mellitus, heart diseases, cerebrovascular diseases, cancers, and kidney diseases) is thought to have been one key factor for aggravation of COVID-19. Needless to say, such pre-existing conditions are also general factors for aggravation of infections other than COVID-19. Thus, there is keen demand for provision of a biomarker which can easily detect the presence of such a pre-existing condition, which may cause aggravation of such infections.


Under such circumstances, the related art will further be reviewed. The serum levels of the aforementioned humoral antibodies (i.e., ATP2B4, BMP-1, and DHPS), serving as markers with respect to atherosclerosis, were found to rise also in samples of esophageal cancer patients (see Non-Patent Documents 6 and 7). That is, “artery lesions” typically including arteriosclerosis (e.g., atherosclerosis) can conceivably promote carcinogenesis. Actually, “angiogenesis,” which is one example of “artery lesions,” is an essential factor for the progress of solid cancer (see Non-Patent Document 28), and diabetes mellitus and obesity, which may induce arteriosclerosis, are risk factors for colorectal cancer and esophageal cancer (see Non-Patent Documents 29, 30, and 31). Meanwhile, since internal organs interact with one another to a greater or lesser extent (see Non-Patent Document 32), detection of “artery lesions” such as arteriosclerosis and angiogenesis may capture symptoms of cerebrovascular and cardiovascular disorders, diabetes mellitus, kidney diseases, solid cancers, etc., thereby possibly leading to early detection of such diseases. If these life-threatening diseases can be detected in an early stage, or symptoms thereof can be detected, by use of a single biomarker, the value of the biomarker would be highly significant.


In addition, it would be remarkably significant to provide means for easily determining, by use of a single biomarker, the presence of a pre-existing condition, which may cause aggravation of infections.


Means for Solving the Problems

The present inventors have found that the presence of an artery lesion is detected by determining a rise in the level of an antibody to protein KIAA0513 in body fluid, whereby data showing the symptoms or presence of a disease involving the artery lesion can be acquired. The inventors have also found that data showing the potential or actual presence of cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer can also be acquired by determining a rise in the aforementioned humoral antibody level. The inventors have further found that the data showing the potential or actual presence of cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer may also be employed as data showing the presence of an artery lesion involved in the diseases. The inventors have yet further found that data showing the symptoms or presence of a disease involving an artery lesion, or data showing the potential or actual presence of cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer are acquired by detecting the presence of an artery lesion based on a rise in the level of an antibody to protein KIAA0513 in body fluid as an index, whereby the presence of a pre-existing condition which is a factor for aggravation of infections can be determined.


Accordingly, in a first aspect of the present invention, there is provided a method for acquiring data, the method comprising: determining a level of an antibody to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 which forms protein KIAA0513, or a portion of the protein, or to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 in which 10% or less of the amino acid residues (the decimal point being suppressed) are deleted, substituted, or added, or a portion of the protein, the antibody being potentially present in a body fluid sample collected from a living body; and acquiring data showing a rise in the antibody level as data showing the presence of an artery lesion (hereinafter, the method may also be referred to as the “data acquisition method of the present invention 1”).


In a second aspect of the present invention, there is provided a method for acquiring data, the method comprising: determining a level of an antibody to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 which forms protein KIAA0513, or a portion of the protein, or to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 in which 10% or less of the amino acid residues (the decimal point being suppressed) are deleted, substituted, or added, or a portion of the protein, the antibody being potentially present in a body fluid sample collected from a living body; and acquiring data showing a rise in the antibody level as data showing the potential or actual presence of cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer (hereinafter, the method may also be referred to as the “data acquisition method of the present invention 2”).


The data showing the onset of the diseases which data are obtained through the data acquisition method of the present invention 2 may also be employed as data showing the presence of an artery lesion involved in the diseases.


Unless otherwise specified, the expression “the data acquisition method of the present invention” in the present specification collectively refers to the data acquisition methods of the present invention 1 and 2.


Meanwhile, protein KIAA0513 is known to include three splicing variants: isoform c (301 amino acids, NP_001284695.1: SEQ ID NO: 1), isoform b (301 amino acids, NP_001273495.1), and isoform a (411 amino acids, NP_055547.1: SEQ ID NO: 2).


The 301 full-length amino acids of KIAA0513 isoforms c or b completely coincide with the initial 301 amino acids of KIAA0513 isoform a. The nucleotide sequence of an mRNA precursor encoding KIAA0513 isoform c differs from that encoding isoform b. However, the amino acid sequence of protein KIAA0513 formed via expression of mature mRNA formed through splicing as a template is common. Thus, in the present specification, without being particularly specified, the expression “protein KIAA0513” refers to protein KIAA0513 isoforms b and/or c (SEQ ID NO: 1). A particular specification is given, in the case where “protein KIAA0513” is intended to refer to isoform a.


Additionally, the present inventors have obtained purified GST-KIAA0513 isoforms c and a, and have investigated the serum level of an antibody to protein KIAA0513 in serum samples of healthy donors (HD), acute ischemic stroke (AIS) patients, and cardiovascular disorder (CVD) patients (Example 6).


As a result, they have found that both KIAA0513 isoforms c and a exhibit a higher serum antibody level in AIS patients and CVD patients, as compared with in HDs (FIGS. 7a and b). The reactivity of KIAA0513 isoform c is slightly higher than that of KIAA0513 isoform a. However, the relative reactivity values closely correlate with each other (FIG. 7c).


The test results have revealed that the main epitope site of a humoral antibody to protein KIAA0513 is present in KIAA0513 isoform c and the initial 301 amino acids of KIAA0513 isoform b. Therefore, “a portion of protein KIAA0513” for use in detection of the humoral antibody to protein KIAA0513 based on antigen-antibody reaction preferably includes the aforementioned overlapping (common) amino acid sequence of the KIAA0513 isoforms c, b, and a.


The data acquisition method of the present invention may also be expressed as, for example, “a method for detecting an artery lesion, the method comprising: determining a level of an antibody to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 which forms protein KIAA0513, or a portion of the protein, or to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 in which 10% or less of the amino acid residues (the decimal point being suppressed) are deleted, substituted, or added, or a portion of the protein, the antibody being potentially present in a body fluid sample collected from a living body; and detecting an artery lesion based on a rise in the antibody level as an index,” and a method for diagnosing or detecting cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer, the method comprising: determining a level of an antibody to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 which forms protein KIAA0513, or a portion of the protein, or to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 in which 10% or less of the amino acid residues (the decimal point being suppressed) are deleted, substituted, or added, or a portion of the protein, the antibody being potentially present in a body fluid sample collected from a living body; and diagnosing or detecting cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer based on a rise in the antibody level as an index.”


The present invention also provides a kit for acquiring data, the kit being employed in carrying out the data acquisition method 1 or 2 of the present invention (hereinafter, the kit may also be referred to simply as the “kit of the present invention”).


The data acquiring kit of the present invention may also be expressed as, for example, “a kit for carrying out a method for detecting an artery lesion, the method comprising: determining a level of an antibody to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 which forms protein KIAA0513, or a portion of the protein, or to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 in which 10% or less of the amino acid residues (the decimal point being suppressed) are deleted, substituted, or added, or a portion of the protein, the antibody being potentially present in a body fluid sample collected from a living body; and detecting an artery lesion based on a rise in the antibody level as an index,” and “a kit for carrying out a method for diagnosing or detecting cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer, the method comprising: determining a level of an antibody to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 which forms protein KIAA0513, or a portion of the protein, or to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 in which 10% or less of the amino acid residues (the decimal point being suppressed) are deleted, substituted, or added, or a portion of the protein, the antibody being potentially present in a body fluid sample collected from a living body; and diagnosing or detecting cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer based on a rise in the antibody level as an index.”


Effects of the Invention

According to the present invention, firstly, data showing the presence of an artery lesion can be acquired. Based on the data, the symptoms and presence of a disease involving the artery lesion can be grasped. Through taking measures including daily-life guidance and medication on the basis of the data, promotion of health as well as prevention/treatment of diseases can be successfully achieved. Secondarily, there can be acquired data showing the potential or actual presence of cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer. Based on the data, the onset of the aforementioned grave diseases can be predicted, and these diseases can be detected in an early stage. Through taking measures including daily-life guidance and medication on the basis of the data, prevention/treatment of diseases can be successfully achieved.


The present invention further provides basic data for determining the presence of a pre-existing condition, which is a factor for aggravation of infections.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows the results of determination of serum KIAA0513 antibody levels in healthy donors (HD), acute ischemic stroke (AIS) patients, and transient ischemic attack (TIA) patients through the AlphaLISA method. Graph a is a distribution diagram showing the results when protein KIAA0513 is used as an antigen. The vertical axis of the diagram indicates alpha counts corresponding to the antibody level. The bars of each segment of the diagram represent an average value and a value of average and ±SD. Graphs b and c show the results of ROC analysis. In each ROC diagram, there are provided AUC value, cut-off value, sensitivity, specificity, and 95% confidence interval.



FIG. 2 shows the results of determination of serum KIAA0513 antibody levels in HDs and diabetes mellitus (DM) patients through the AlphaLISA method. Graph a is a distribution diagram showing the results when protein DIDO1 is used as an antigen. Graph b shows the results of ROC analysis. The legends for the distribution diagram and ROC diagram are the same as those for FIG. 1.



FIG. 3 shows the results of determination of serum KIAA0513 antibody levels in HDs, cardiovascular disorder (CVD) patients, and obstructive sleep apnea syndrome (OSAS) patients through the AlphaLISA method. Graph a is a distribution diagram, and Graphs b and c show the results of ROC analysis. The legends for the distribution diagram and ROC diagrams are the same as those for FIG. 1.



FIG. 4 shows the results of determination of serum KIAA0513 antibody levels in HDs and chronic kidney disease (CKD) patients through the AlphaLISA method, when protein KIAA0513 was used as an antigen. Graph a is a distribution diagram, and Graphs b, c, and d show the results of ROC analysis. The legends for the distribution diagram and ROC diagrams are the same as those for FIG. 1.



FIG. 5 shows the results of determination of serum KIAA0513 antibody levels in HDs and cancer patients through the AlphaLISA method. The serum antibody levels are determined in HDs and cancer patients (esophageal cancer (EC), gastric cancer (GC), colorectal cancer (CC), lung cancer (LC), and mammary cancer (MC)), when protein KIAA0513 is used as an antigen. Graph a is a distribution diagram, and Graphs b to f show the results of ROC analysis. The legends for the distribution diagram and ROC diagrams are the same as those for FIG. 1.



FIG. 6 shows comparison in percent survival after surgery of esophageal cancer (EC) and gastric cancer (GC), with cases of KIAA0513 antibody positive and negative. Percent survival values after surgery are compared through Kaplan-Meyer plotting.



FIG. 7 shows comparison in serum antibody level when KIAA0513 isoforms c and a are used as antigens. Graphs are distribution diagrams showing serum antibody levels in HDs, AIS patients, and CVD patients, with respect to KIAA0513 isoform c (Graph a) and KIAA0513 isoform a (Graph b). Graph c is a correlation diagram showing antibody levels (alpha counts) in samples, when KIAA0513 isoforms c and a are used an antigen, respectively.





MODES FOR CARRYING OUT THE INVENTION
(1) Data Acquisition Method of the Present Invention
(a) Data to be Acquired

The data acquired through the data acquisition method of the present invention 1 are “data showing the presence of an artery lesion.” The artery lesion corresponds to abnormality in the structure and components of an artery, and modes thereof include occlusion, expansion, and fistulation. The progress of artery lesion involves arteriosclerosis (atherosclerosis, arteriolosclerosis, or medial sclerosis), anomalous angiogenesis in chronic inflammation or solid cancer, and other conditions. In many cases, arteriosclerosis is known to be triggered by progress of diabetes mellitus and obesity. Particularly, cerebrovascular and cardiovascular disorders, chronic kidney disease, aortic aneurysm, retinopathy, feet gangrene, etc. are known to be triggered by progress of arteriosclerosis, in particular, atherosclerosis. Diabetes mellitus and obesity are also risk factors for solid cancers (colorectal cancer, esophageal cancer, etc.). Thus, the “data showing the presence of an artery lesion” obtained by the data acquisition method of the present invention 1 may be employed as data showing the possibility of onset or progress of diabetes mellitus, arteriosclerosis, solid cancer, etc. In addition, the data may also be employed as data showing the potential or actual presence of severe circulatory diseases such as cerebrovascular and cardiovascular disorders, chronic kidney disease, aortic aneurysm, retinopathy, and feet gangrene.


The data acquired through the data acquisition method of the present invention 2 are “data showing the potential or actual presence of diabetes mellitus, cerebrovascular or cardiovascular disorder, chronic kidney disease, or solid cancers.” Examples of the cerebrovascular or cardiovascular disorder include cerebral infarction, cerebral stroke, myocardial infarction, and angina pectoris. Examples of the solid cancer include esophageal cancer, gastric cancer, colorectal cancer, lung cancer, and mammary cancer. The rise in the level of the humoral antibody to protein KIAA0513 can be employed as reliable data showing the potential or actual presence of esophageal cancer, colorectal cancer, gastric cancer, or lung cancer, particularly esophageal cancer. The data acquisition method of the present invention 2 often overlaps with the data acquisition method of the present invention 1.


For example, the onset of any of the aforementioned diseases in a subject can be accurately predicted by periodically conducting the data acquisition method of the present invention 1 or 2 on the patient, including monitoring of the level of the humoral antibody to “protein KIAA0513.” Particularly, once a subject has developed a severe circulatory disease such as cerebral infarction or myocardial infarction or a solid cancer, the life of the subject is endangered. Even if death is avoided, the subject may suffer a severe sequela. Therefore, prediction of the onset of such diseases is the best solution, and a humoral anti-KIAA0513 antibody marker may be employed as a biomarker for acquiring data concerning prediction of the onset.


When epidemic of a specific infection is recognized, the presence of a pre-existing condition, which is a factor for aggravation of the infection, is determined on the basis of the data acquired through the data acquisition method of the present invention 1 or 2. As a result, the patient suffering the infection can be more suitably treated. In the case where the obtained data indicate the presence of, for example, an artery lesion, the patient contracting the infection is preferentially hospitalized so as to prevent aggravation. In the case where the data indicate no artery lesions, the subject is made to stay at home, whereby hospital capacity including medical staff, beds, etc. can be effectively furnished. In this case, the infected subjects can be appropriately treated in accordance with their degree of aggravation risk. Thus, the risk of medical care breakdown, which would otherwise be caused by shortage of medical staff and beds due to epidemic of infection, can be reduced.


The aforementioned infection includes various viral diseases, bacterial diseases, etc. No particular limitation is imposed on the infection, so long as the presence of a pre-existing condition is an aggravation factor of the infection. Particularly in the case of infections in the respiratory system, which occasionally cause outbreak, the aggravation risk of the infections is desirably estimated by mass examination (screening). The present invention, which can be implemented in a simple mode (e.g., immunochromatography), is particularly suited for such mass examination. Examples of the aforementioned infections include, but are not limited to, respiratory system infections such as infections with beta-coronaviruses (for example, COVID-19, SARS, and MARS); infections with influenza viruses; and infections with RS virus, adenovirus, rhinovirus, and the like.


(b) Body Fluid Sample Collected from Living Body


The term “body fluid” of “a body fluid sample collected from a living body” which is a target for data acquisition in the data acquisition method of the present invention, includes blood, lymph, or the like. The term “sample” refers to body fluid itself as collected from a body, or a processed product of the fluid. Among such body fluid samples, a blood sample is preferred. Examples of the blood sample include whole blood samples, serum samples, and plasma samples. Of these, serum samples and plasma samples are preferred, with serum samples being particularly preferred. The blood samples may undergo anti-clotting treatment (e.g., treatment with heparin) in advance.


(c) Humoral Antibody-Capturing Antigen (KIAA0513 Antigen)

In the data acquisition method of the present invention, protein KIAA0513 having an amino acid sequence represented by SEQ ID NO: 1 or 2 is the basis for capturing a humoral antibody. The entirety or a portion of protein KIAA0513 may be used as a humoral antibody-capturing antigen. Furthermore, in order to establish a more efficient detection system, a shorter chain amino acid sequence (peptide) may be selected. In this case, the number of amino acids of the amino acid sequence is about 7 to about 30, more preferably about 10 to 20.


In the entirety or a portion of any of the amino acid sequences, 10% or less of the amino acid residues (the decimal point being suppressed) may undergo modification (deletion, substitution, or addition), so long as the target humoral antibody (i.e., a humoral antibody to protein KIAA0513 or to a peptide having a partial sequence thereof) can be captured. In other words, when the amino acid-modified antigen candidate for capturing a target humoral antibody is used for capturing the humoral antibody through, for example, a technique similar to the technique described in the Examples of the present specification, data concerning the target artery lesion or disease must be acquired on the basis of the level of the captured antibody.


The term “deleting” refers to deletion of any amino acid residue in the relevant amino acid sequence. The amino acid residue at the N-terminal side of the deleted amino acid residue and that at the C-terminal side of the deleted amino acid residue are linked via a peptide bond. In the case of deletion of the N-terminal amino acid residue or the C-terminal amino acid residue, no linkage is present. The number of deleted residues is counted as “the number of amino acid deletions.” The term “substituting” refers to substitution of any amino acid residue in the relevant amino acid sequence, “with another amino acid residue.” The new amino acid residue is linked to the amino acid residue at the N-terminal side thereof and that at the C-terminal side thereof via a peptide bond. In the case of substitution of the N-terminal side or C-terminal side amino acid residue, the amino acid residue is linked via a peptide bond to another C-terminal side or N-terminal side amino acid residue. The number of substituted residues is counted as “the number of amino acid substitutions.” The term “adding” refers to addition of one or more new amino acid residues to one or more peptide bond sites in the relevant amino acid sequence, to thereby form a new peptide bond(s). Addition of an amino acid residue to the C terminal or the N terminal is also encompassed in the concept “addition.” The number of added substituted amino acid residues is counted as “the number of amino acid additions.” Hereinafter, “a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 which forms protein KIAA0513, or a portion of the protein, or a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 in which 10% or less of the amino acid residues (the decimal point being suppressed) are deleted, substituted, or added, or a portion of the protein” is also referred to as a “KIAA0513 antigen.”


The KIAA0513 antigen having a specific amino acid sequence may be obtained through a customary method. In one specific procedure, based on the nucleotide sequence of the KIAA0513 gene represented by SEQ ID NO: 1 or 2, nucleic acid amplification primers for amplifying a double-strand DNA including the entirety or a portion of nucleotide sequence are designed. Through PCR or a similar technique employing the amplification primers, a gene amplification product is yielded as the entirety or a portion of the KIAA0513 gene. The amplification product is incorporated into a prokaryotic cell expression vector (e.g., pGEX-4T), and a transformant including the vector is selected. Expression of the entirety or a portion of the KIAA0513 gene is induced by adding a chemical agent such as IPTG (isopropyl-β-D-thiogalactoside). The entirety or a portion of the thus-formed protein KIAA0513 may be purified through, for example, affinity chromatography by use of glutathione-Sepharose (product of GE Healthcare Life Sciences).


If needed, a modification structure may be appropriately added to the protein or the peptide used as such a KIAA0513 antigen. For example, as mentioned hereinbelow, there may be employed a GST (glutathione S-transferase)-fused protein or peptide, a biotin-fused protein or peptide, etc., which are used in AlphaLISA (Amplified Luminescence Proximity Homogeneous Assay-Linked ImmunoSorbent Assay).


(d) Specific Procedure of Data Acquisition

According to the data acquisition method of the present invention, the level of an antibody to the KIAA0513 antigen in a body fluid sample (e.g., a blood sample) collected from a living body is determined. When the determined value is greater than a standard value (cut-off value), the increase in KIAA0513 antigen level in the body fluid sample donor is confirmed. The data can be employed as an index for showing the presence of an artery lesion in the body fluid sample donor, or for showing the potential or actual presence of cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer in the body fluid sample donor.


The standard value of the humoral antibody level with respect to the KIAA0513 antigen in a test sample may be derived through the following procedure. First, a control sample population is established from subjects exhibiting no abnormality (e.g., diabetes, a respiratory disease, or a cancer) in a medical examination (i.e., healthy donors). The level of a humoral antibody to the KIAA0513 antigen is determined, with respect to the test samples obtained from the control sample population. The determination data are statistically processed, to thereby obtain an average, a standard deviation, etc., and derive the standard value including the cut-off value, on the basis of the data.


In one possible embodiment of the humoral antibody level determination, the KIAA0513 antigen which is immobilized on a substrate is brought into contact with a body fluid sample collected from a living body. Bonding (based on antigen-antibody reaction) of a humoral antibody to the KIAA0513 antigen in the body fluid sample collected from the living body is detected as a signal. More specifically, the determination can be performed through immuno-chromatography, AlphaLISA, ELISA, indirect immunofluorescence, Western blotting (immunoblotting), turbidimetry, nephelometry, latex-coagulation turbidimetry, CLEIA, or the like. All the exemplified determination techniques have been established for determining the level of a target substance (antibody) in a body fluid sample collected from a living body.


In one mode of the immuno-chromatographic method, a body fluid sample and a buffer liquid containing a gold colloid-bound anti-human IgG antibody are added to a cellulose membrane onto which the KIAA0513 antigen has been applied, and the liquid mixture is slowly moved on the membrane. If an antibody to the KIAA0513 antigen is present in the body fluid sample, a line is formed at a position where the antigen is applied on the surface of the membrane. The line can be visually observed. Since the immuno-chromatographic method can be conducted in a simpler manner, as compared with other assay means, the method is particularly suited for mass examination. In other words, when the data acquisition method of the present invention is carried out through the immuno-chromatographic method, desired disease-related data can be acquired in a short period of time (about 10 minutes) even in small facilities (for example, a clinic), a public health center, and other facilities.


In the case of AlphaLISA, when a GST-fused KIAA0513 antigen is used as an antigen for capturing a humoral antibody, glutathione-conjugated donor beads are used. The antigen for capturing a humoral antibody, a blood sample, and acceptor beads to which an anti-human IgG antibody has been bound are mixed together, and the mixture is incubated at room temperature for several hours to several days, to thereby form an antigen-antibody complex. The complex is irradiated with light (680 nm), and the resultant light having a wavelength of 520 to 620 nm is detected, whereby the target humoral antibody can be quantitated. In indirect immunofluorescence assay, a body fluid sample collected from a living body is brought into contact with a protein array in which the KIAA0513 antigen has been immobilized on a substrate, to thereby form a complex of the KIAA0513 antigen with an anti-KIAA0513 humoral antibody. Then, a fluorescent-labeled secondary antibody is brought into contact with the humoral antibody complex, whereby a humoral antibody to protein KIAA0513 can be quantitated. In ELISA, the same secondary antibody as employed in indirect immunofluorescence assay is used, except that the label is changed to an enzyme, to thereby conduct quantitation. The label for the secondary antibody can be selected from various enzymes. In Western blotting, a KIAA0513 antigen is subjected to electrophoresis by use of SDS-polyacrylamide gel, and the resultant band is transferred to a carrier such as a nitrocellulose membrane. Then, a body fluid sample collected from a living body is brought into contact with the carrier, to thereby form a (KIAA0513 antigen)-(anti-KIAA0513 humoral antibody) complex. Detection of the complex is performed by use of a secondary antibody, to thereby achieve quantitation. In turbidimetry or nephelometry, a body fluid sample collected from a living body is brought into contact with the KIAA0513 antigen, to thereby form a (KIAA0513 antigen)-(anti-KIAA0513 humoral antibody) complex. Detection of the complex is performed on the basis of change in turbidity (in the case of turbidimetry) or scattered light (in the case of nephelometry), to thereby achieve quantitation. In latex agglutination turbidimetry, a body fluid sample collected from a living body is brought into contact with latex particles to which the KIAA0513 antigen has been bound, to thereby form agglutinated latex particles via interaction between the humoral anti-KIAA0513 antibodies bounded to latex particles, and the thus-formed agglutinated latex particles are quantitated. In one possible mode of CLEIA assay, a body fluid sample collected from a living body is brought into contact with magnetic particles to which the KIAA0513 antigen has been bound, to thereby form, on the surfaces of the magnetic particles, a (KIAA0513 antigen)-(anti-KIAA0513 antibody) complex. Unreacted matter is removed through collecting magnetism, and the product is subjected to an appropriate fluorescent chemical treatment or the like. Thus thus-treated complex is quantitated.


(2) Kit of the Present Invention

An embodiment of the kit of the present invention employs AlphaLISA as an assay means and has a set including a GST-fused KIAA0513 antigen, glutathione-conjugated donor beads, and anti-human IgG-conjugated acceptor beads.


Another embodiment of the kit of the present invention employs ELISA as an assay means and has a set including a plate on which the aforementioned KIAA0513 antigen has been immobilized, a labeled secondary antibody to the anti-KIAA0513 antibody, and a reagent for developing the label of the secondary antibody.


Still another embodiment of the kit of the present invention employs latex agglutination turbidimetry and includes latex particles on which the aforementioned KIAA0513 antigen has been immobilized.


Yet another embodiment of the kit of the present invention employs CLEIA and includes magnetic particles on which the aforementioned KIAA0513 antigen has been immobilized, a labeled secondary antibody with respect to the anti-KIAA0513 antibody, and a reagent for developing the label of the secondary antibody.


Notably, the aforementioned embodiments of the kit are illustrated merely as examples, and kits employing other assay means are also encompassed in the scope of the kit of the present invention. Furthermore, the number of the aforementioned kit elements may be further reduced so as to encourage self-supply or out-sourcing of medical tests. In contrast, elements such as diluent and reagent tubes are added to the kit so as to realize immediate use of the kit of the present invention. Other additional elements suited for the mode of tests may also be added.


EXAMPLES

Examples of the present invention will next be described.


Production Examples and Analytical Techniques
<Antigen Proteins Employed in the Examples>
(1) Identification of Antigen Protein

In the Examples, by means of a human protein microarray (ProtoArray (registered trademark), product of Thermo Fisher Scientific), proteins KIAA0513 were identified as antigens which are recognized by a serum antibody of atherosclerosis patients.


In a specific procedure, the N-terminal of “KIAA0513 isoform a” (SEQ ID NO: 2) was modified with GST. The full-length region of 411 amino acids, or a portion of 301 amino acids from the N-terminal was used. Notably, the latter “portion of 301 amino acids from the N-terminal” corresponds to KIAA0513 isoform c (SEQ ID NO: 1).


(2) Preparation of Antigen Proteins (Formation and Purification of Proteins KIAA0513)

Human proteins KIAA0513 were prepared through the following procedure.


Each of the cDNA fragments encoding human KIAA0513 isoforms a and c was inserted into an EcoRI/SalI site of a plasmid vector pGEX-4T-1 (GE Healthcare Life Sciences, Pittsburgh, PA), in which a glutathione S-transferase (GST) gene site is provided on the upstream side of the cloning site, to thereby produce recombinant plasmids pGEX-4T-1-KIAA0513 (isoform a) and pGEX-4T-1-KIAA0513 (isoform c) for gene expression, respectively. Each plasmid was incorporated into E. coli BL-21 for transformation, and expression of the corresponding cDNA encoding protein KIAA0513 was induced through a treatment with 0.1 mM IPTG (isopropyl-β-D-thiogalactoside) at 25° C. for 4 hours. Thereafter, pellets of E. coli were recovered and dissolved by use of BugBuster Master Mix (Merck Millipore, Darmstadt, Germany), to thereby yield a protein extract. Each of the GST-protein KIAA0513 isoforms a and c present in the protein extract was purified through column chromatography with glutathione-Sepharose (GE Healthcare Life Sciences). Through changing the buffer to PBS, the aforementioned “proteins KIAA0513” were obtained. GST as a control was purified in the same manner.


<Analysis Through AlphaLISA>

Prior to disclosure of specific working examples, the analysis through AlphaLISA employed in the Examples will next be outlined.


Specifically, the AlphaLISA (Amplified Luminescence Proximity Homogeneous Assay) was performed by use of a 384-well microtiter plate (white opaque OptiPlate™, Perkin Elmer, Waltham, MA). To each well, a serum (2.5 μL) 100-fold diluted with AlphaLISA buffer (25 mM HEPES, pH: 7.4, 0.1% casein, 0.5% Triton X-100, 1-mg/mL dextran-500, 0.05% Proclin-300) was added. GST or protein KIAA0513 (i.e., KIAA0513 isoform a or c) (10 μg/mL) (antigen), diluted with AlphaLISA buffer, was mixed with the serum in each well. The mixture was incubated at room temperature for 6 to 8 hours. Separately, anti-human IgG-conjugated acceptor beads (2.5 μL at 40 μg/mL), and glutathione- or streptavidin-conjugated donor beads (2.5 μL at 40 μg/mL) were diluted with AlphaLISA buffer, and the thus-diluted product was mixed with the incubated product. The resultant mixture was allowed to stand at room temperature for 1 to 14 days. Thereafter, generated photons were counted by means of EnSpire Alpha microplate reader (Perkin Elmer), to thereby determine “Alpha Count.” In the case of protein KIAA0513, a control GST value was subtracted, whereas in the case of the biotinylated peptide, a control buffer value was subtracted, whereby the antibody level specific to each KIAA0513 antigen was determined.


Example 1

Studies of biomarker in transient ischemic attack (TIA) patients and acute ischemic stroke (AIS) patients through AlphaLISA


Acute ischemic stroke (AIS) refers to a pathological condition in which a patient receives sudden damage in motor function or sensory function due to stagnation of blood flow in the brain. Transient ischemic attack (TIA) refers to a disease in which cerebral nerve cells are damaged through the same mechanism as that of cerebral infarction, but the damage is restored within 24 hours, generally several minutes. In both cases, blood serum samples are collected from relevant patients ≤2 weeks after the onset thereof.


The serum antibody (s-KIAA0513-Abs: serum KIAA0513 antibody) level with respect to KIAA0513 isoform c (SEQ ID NO: 1) was determined in AIS patients and TIA patients. Serum samples of healthy donors (HDs), AIS patients, and TIA patients were collected from Chiba Prefectural Sawara Hospital. In AlphaLISA, the s-KIAA0513-Ab level was found to be significantly higher in TIA patients and AIS patients than in HDs (see FIG. 1).


When the cut-off value was set to “the average+2SD for HD samples,” the s-KIAA0513-Ab positive ratios of HDs, AIS patients, and TIA patients were 0.0%, 7.6%, and 15.6%, respectively (see Table 1).


Through ROC analysis, area under the curve (AUC) values of s-KIAA0513-Abs were found to be 0.6439 [95% confidence interval (CI)=0.5873-0.7004] with respect to AIS (see FIG. 1b), and 0.6604 (95% CI=0.5633-0.7575) with respect to TIA (see FIG. 1c).


Thus, TIA, which is a prodromal stage of AIS, was also found to positively correlate with the serum KIAA0513 antibody level. Therefore, the s-KIAA0513-Ab level served as a marker effectively predicting the onset of AIS.












TABLE 1







Sample information
HD
AIS
TIA





Total sample number
139
228
44


Male/Female
87/52
129/99
24/20


Age (Average ± SD)
51.6 ± 12.8
77.0 ± 11.1
68.5 ± 12.1














Subject group
Type of value
KIAA0513-Ab







HD
Average
92,849




SD
36,115




Cutoff value
165,080




Positive number
0




Positive (%)
0.0%



AIS
Average
110,913




SD
40,351




Positive number
11




Positive (%)
7.6%




P (AIS vs HD)
<0.001



TIA
Average
122,303




SD
42,845




Positive number
5




Positive (%)
15.6%




P (TIA vs HD)
<0.01










Example 2
Studies of Biomarker in Diabetes Mellitus (DM) Patients Through AlphaLISA

Next, the s-KIAA0513-Abs level of DM patients was investigated.


Serum samples of HDs and DM patients were obtained from Chiba University Hospital.


The s-KIAA0513-Abs level was found to be significantly higher in DM patients than in HDs (see FIG. 2a).


When the cut-off value was set to “the average+2SD for HD samples,” the s-KIAA0513-Ab positive ratios of HDs and DM patients were 2.5% and 26.5%, respectively (see Table 2).


The ability of the antibody marker to indicate the presence of DM was evaluated through ROC analysis. The area under the curve (AUC) value of s-KIAA0513-Abs was 0.7361, and the values of sensitivity and specificity were 50.55% and 87.65%, respectively (see FIG. 2b).













TABLE 2









Sample information
HD
DM







Total sample number
81
275



Male/Female
46/35
158/117



Type 1 DM/Type 2 DM

 26/216*



Age (Average ± SD)
45.2 ± 11.0
63.1 ± 12.0















Subject group
Type of value
KIAA0513-Ab







HD
Average
33,698




SD
18,121




Cutoff value
69,941




Positive number
2




Positive (%)
2.5%



DM
Average
55,282




SD
29,897




Positive number
73




Positive (%)
26.5%




P (vs HD)
<0.001










Example 3

Studies of biomarker in cardiovascular disorder (CVD) patients and obstructive sleep apnea syndrome (OSAS) patients through AlphaLISA


Next, the s-KIAA0513-Abs levels of CVD patients and OSAS patients were investigated.


The CVD patients in Chiba University Hospital included acute myocardial infarction patients and unstable angina patients. Generally, OSAS is known to relate to atherosclerosis and is a high risk factor for acute ischemic stroke (AIS) and CVD. Thus, serum samples of OSAS patients obtained from Chiba University Hospital were also analyzed.


As a result, the s-KIAA0513-Abs level was found to be higher in CVD patients and OSAS patients than in HDs (FIG. 3a). The s-KIAA0513-Abs positive ratios of HDs, CVD patients, and OSAS patients were 5.3%, 10.3%, and 11.6%, respectively (see Table 3).


Through ROC analysis, area under the curve (AUC) values of s-KIAA0513-Abs were found to be 0.6492 (95% CI: 0.5821-0.7163) with respect to CVD (see FIG. 3b), and 0.6463 (95% CI: 0.5616-0.7309) with respect to OSAS (see FIG. 3e).


The P value of s-KIAA0513-Ab with respect to CVD was small (<0.0001), but the P value with respect to OSAS was 0.0013. The data suggest that the correlation between the s-KIAA0513-Ab marker and OSAS is less significant than the correlation between the s-KIAA0513-Ab marker and CVD (see Table 3). Thus, conceivably, OSAS does not correlate directly with the s-KIAA0513-Ab marker, but does indirectly correlate with the marker by the mediation of CVD or the like, which is induced by OSAS.












TABLE 3







Sample information
HD
CVD
OSAS





Total sample number
76
97
86


Male/Female
43/33
81/16
59/27


Age (Average + SD)
45.1 ± 11.5
66.3 ± 11.4
57.8 ± 12.5














Subject group
Type of value
KIAA0513-Ab







HD
Average
43,398




SD
21,374




Cutoff value
86,146




Positive number
4




Positive (%)
5.3%



CVD
Average
57,531




SD
22,460




Positive number
10




Positive (%)
10.3%




P (CVD vs HD)
<0.001



OSAS
Average
55,650




SD
27,493




Positive number
10




Positive (%)
11.6%




P (OSA vs HD)
<0.01










Example 4
Studies of Biomarker in Chronic Kidney Disease (CKD) Patients Through AlphaLISA

Next, the s-KIAA0513-Abs level of CKD patients was investigated. CKD is closely related to atherosclerosis.


The studied CKD patients were divided into three groups.


Specifically, the groups were the diabetic nephropathy group (Type 1), the nephrosclerosis group (Type 2), and the glomerulonephropathy group (Type 3).


Serum samples of CKD patients were obtained from a cohort of Kumamoto Prefecture, and those of HDs were obtained from Chiba University.


The serum s-KIAA0513-Abs level was found to be significantly higher in all the three CKD groups than in HD samples (FIG. 4a). More specifically, the s-KIAA0513-Ab positive ratios of HDs, Type 1 CKD patients, Type 2 CKD patients, and Type 3 CKD patients were 6.1%, 29.0%, 37.5%, and 20.3%, respectively (see Table 4), and the highest positive ratio was identified in serum samples of Type 2 CKD patients.


Through ROC analysis, area under the curve (AUC) values of s-KIAA0513-Abs were found to be 0.7434 (95% CI: 0.6783-0.8085) with respect to Type 1 CKD (see FIG. 4b), 0.8080 (95% CI: 0.7264-0.8897) with respect to Type 2 CKD (see FIG. 4c), and 0.6906 (95% CI: 0.6177-0.7635) with respect to Type 3 CKD (see FIG. 4d).













TABLE 4







Sample information
HD
Type-1 CKD
Type-2 CKD
Type-3 CKD





Total sample number
82
145
32
123


Male/Female
44/38
106/39
21/11
70/53


Age (Average ± SD)
44.1 ± 11.2
66.0 ± 10.4
76.0 ± 9.8
62.0 ± 11.7













Alpha analysis (antibody level)
KIAA0513-Ab















HD
Average
36,007




SD
28,697




Cutoff value
93,400




Positive No.
5




Positive rate (%)
6.1%



Typ-1 CKD
Average
71,773




SD
44,434




Positive No.
42




Positive rate (%)
29.0%




P (vs HD)
<0.001



Type-2 CKD
Average
84,954




SD
47,414




Positive No.
12




Positive rate (%)
37.5%




P (vs HD)
<0.001



Type-3 CKD
Average
61,443




SD
41,110




Positive No.
25




Positive rate (%)
20.3%




P (vs HD)
<0.001










Example 5
Studies of Biomarker in Cancer Patients Through AlphaLISA

Atherosclerosis is known to be often related to cancers. Thus, the s-KIAA0513-Abs level in serum samples of esophageal cancer (EC), gastric cancer (GC), colon cancer (CC), lung cancer (LC), and mammary cancer (MC) patients was investigated. The serum samples were obtained from Toho University Medical Center.


As a result, the s-KIAA0513-Abs level was significantly higher in serum samples of all cancer patients, as compared with those of HDs. The highest average value of s-KIAA0513-Ab level was identified in the case of esophageal cancer (see FIG. 5a and Table 5). In ROC analysis, the AUC value was as high as 0.8300 in the case of esophageal cancer (see FIGS. 5b to 5f).


Next, correlation of the s-KIAA0513-Ab level with survival rates of esophageal cancer (EC) patients and gastric cancer (GC) patients after surgery was investigated. On the basis of the cut-off value obtained through ROC analysis, analytical samples were divided into an s-KIAA0513-Ab positive group and an s-KIAA0513-Ab negative group (see FIGS. 5b and 5c). In the case of esophageal cancer, no significant difference was found in overall survival rate between the s-KIAA0513-Ab positive group and the s-KIAA0513-Ab negative group (P=0.3509), but a tendency of poor prognosis was observed in the antibody positive group between 40 to 60 weeks after surgery (FIG. 6a). In contrast, in the case of gastric cancer, no significant difference was found in survival rate between the s-KIAA0513-Ab positive group and the s-KIAA0513-Ab negative group (FIG. 6b).















TABLE 5







Sample information
HD
EC
GC
CC
LC
MC





Total sample
96
192
96
192
96
96


number


Male/Female
51/45
155/37
68/28
119/74
0/96
58/38


Age (Average ± SD)
57.9 ± 6.0
67.4 ± 9.8
68.7 ± 10.6
66.7 ± 11.7
60.9 ± 13.3
68.1 ± 9.6














Subject group
Type of value
KIAA0513-Ab







HD
Average
36,199




SD
23,001




Cutoff value
82,201




Positive number
8




Positive (%)
8.3%



EC
Average
87,535




SD
57,134




Positive number
86




Positive (%)
44.8%




P (EC vs HD)
<0.001



GC
Average
62,714




SD
54,540




Positive number
21




Positive (%)
21.9%




P (GC vs HD)
<0.001



CC
Average
69,308




SD
58,449




Positive number
53




Positive (%)
27.6%




P (CC vs HD)
<0.001



LC
Average
62,148




SD
57,101




Positive number
24




Positive (%)
25.0%




P (LC vs HD)
<0.001



MC
Average
50,270




SD
36,802




Positive number
18




Positive (%)
18.8%




P (MC vs HD)
0.0018










Example 6
Comparison of Serum Antibody Level to Antigens; i.e., Between KIAA0513 Isoforms c and a

The serum antibody levels with respect to KIAA0513 isoform c (SEQ ID NO: 1) and KIAA0513 isoform a (SEQ ID NO: 2) were determined in healthy donors (HDs), acute ischemic stroke (AIS) patients, and cardiovascular disorder (CVD) patients through AlphaLISA.


As a result, with respect to both KIAA0513 isoforms c and a, the serum antibody levels were found to be higher in AIS patients and CVD patients than in HDs (see FIGS. 7a and b). The reactivity of KIAA0513 isoform c to the serum antibody is higher than that of KIAA0513 isoform a. However, the relative reactivity values closely correlated with each other (FIG. 7c).


The test results have revealed that the main epitope site of a serum antibody to protein KIAA0513 is present in 301 amino acids of KIAA0513 isoform c.


Example 7
Correlation Analysis Between Serum Antibody Level and Test Subject Data
(1) Mann-Whitney U Analysis

Correlation analysis between s-KIAA0513-Ab levels and test subject data was carried out by use of 665 serum samples collected from subjects in Chiba Prefectural Sawara Hospital. The samples included 139 HD samples, 225 samples of AIS patients, 44 samples of TIA patients, 17 samples of asymptomatic ischemic cerebral stroke patients, 122 samples of deep and subcortical white matter hyperintensity (DSWMH) patients, 59 samples of chronic ischemic stroke (CIS) patients, and 41 control samples.


In the correlation analysis, Mann-Whitney U analysis was employed to compare s-KIAA0513-Ab levels with the parameters: sex (male/female), DM, hypertension (HT), CVD, lipidemia, and obesity [“normal or thin” when body mass index (BMI) is <25; and “obese” when BMI is ≥25], smoking habit, and drinking habit. As a result, a significant difference was found only in the case of presence/absence of HT (Table 6).












TABLE 6







Sex

Male
Female





Sample number

396
268


KIAA0513-Ab level
Average
10,136
104,138



SD
39,889
40,969


P value (vs Male)


0.1466





Other disease

DM−
DM+





Sample number

525
135


KIAA0513-Ab level
Average
101,818
101,727



SD
40,022
41,930


P value (vs DM−)


0.5031





Other disease

HT−
HT+





Sample number

239
421


KIAA0513-Ab level
Average
96,476
104,821



SD
38,988
40,905


P value (vs HT−)


0.0091





Other disease

CVD−
CVD+





Sample number

623
37


KIAA0513-Ab level
Average
101,376
108,939



SD
39,757
49,697


P value (vs CVD−)


0.1177





Other disease

Lipidemia−
Lipidemia+





Sample number

475
185


KIAA0513-Ab level
Average
103,028
98,647



SD
40,591
39,808


P value (vs Lipidemia−)


0.2716





Obesity

BMI < 25
BMI ≥ 25





Sample number

498
158


KIAA0513-Ab level
Average
102,071
101,390



SD
40,475
40,488


P value (vs Alcohol−)


0.7490





Life style

Non-smoker
Smoker





Sample number

344
319


KIAA0513-Ab level
Average
98,991
104,832



SD
38,563
42,055


P value (vs non-smoker)


0.0673





Life style

Alcohol−
Alcohol+





Sample number

238
419


KIAA0513-Ab level
Average
102,663
101,364



SD
39,883
40,759


P value (vs Alcohol−)


0.4595









(2) Spearman's Correlation Analysis

Through Spearman's correlation analysis, correlations of s-KIAA0513-Ab level with continuous variables, specifically, general information parameters of subjects including age, height, weight, and BMI; degree of arteriostenosis including maximum intima-media thickening (max-IMT); life-style factors such as period of smoking (years) and alcohol frequency (/week); and blood test data, were investigated. Significant correlations were identified between s-KIAA0513-Abs level and age, max-IMT, ALP, potassium, C-reactive protein (CRP), body sugar (BS), and smoking period (see Table 7). Inverse correlations were observed between the antibody level and height and weight. Correlation between the antibody level and max-IMT indicates that the s-KIAA0513-Ab level is associated with stenosis and atherosclerosis.


Similar results were obtained using other cohorts. In Spearman's correlation analysis of a CKD cohort (300 cases), significant correlations of the antibody level were observed with plaque score, max-IMT, and CAVI (right), which are indices for atherosclerosis (see Table 8). In the CKD cohort, correlation was observed between CRP and the s-KIAA0513-Abs level, indicating certain association of inflammation. Meanwhile, in the CKD cohort, no significant correlations of the s-KIAA0513-Abs level were observed with age, height, weight, BMI, and potassium. AIS is closely related with age, and positive correlation was observed between the s-KIAA0513-Abs level and AIS. Therefore, the s-KIAA0513-Abs may be indirectly associated with aging.













TABLE 7









Number of
KIAA0513-Ab













Parameter
XY pairs
r value
P value
















Age
663
0.1959

<0.0001




Height
657
−0.1317

0.0007




Weight
661
−0.1093

0.0049




BMI
656
−0.0414
0.2899



max-IMT
457
0.1763

0.0002




A/G
628
−0.0187
0.6402



AST (GOT)
655
0.0395
0.3134



ALT (GPT)
655
−0.0035
0.9278



ALP
600
0.0889

0.0295




LDH
631
0.0431
0.2798



tBil
637
0.0317
0.4250



CHE
506
−0.0387
0.3851



γ-GTP
609
−0.0099
0.8072



TP
633
−0.0184
0.6435



ALB
638
−0.0207
0.6014



BUN
654
−0.0454
0.2469



CRE
649
−0.0310
0.4303



eGFR
552
0.0432
0.3113



UA
492
−0.0138
0.7596



AMY
415
−0.0901
0.0667



T-CHO
563
0.0446
0.2913



HDL-C
435
−0.0280
0.5600



TG
460
−0.0198
0.6719



Na
641
−0.0347
0.3808



K
641
−0.0899

0.0228




Cl
641
−0.0392
0.3215



Ca
380
−0.0607
0.2376



IP
302
−0.0085
0.8824



Fe
311
−0.0083
0.8838



CRP
477
0.0917

0.0453




LDL-C
344
−0.0559
0.3011



WBC
650
0.0712
0.0697



RBC
650
−0.0220
0.5748



HGB
650
0.0117
0.7651



HCT
650
−0.0041
0.9174



MCV
650
0.0525
0.1814



MCH
650
0.0611
0.1199



MCHC
650
0.0401
0.3080



RDW
650
0.0437
0.2661



PLT
650
−0.0269
0.4942



MPV
650
−0.0354
0.3683



PCT
650
−0.0271
0.4900



PDW
650
−0.0342
0.3838



BS
596
0.0932

0.0229




HbA1c
505
−0.0212
0.6351



Smoking period
656
0.1297

0.0009




Alcohol frequency
662
−0.0328
0.4000







*The data of the test subjects employed include the following: age, height, weight, body mass index (BMI), maximum intima-media thickness (max IMT), albumin/globulin ratio (A/G), aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), lactate dehydrogenase (LDH), total bilirubin (tBil), cholinesterase (CHE), γ-glutamyl transpeptidase (γ-GTP), total protein (TP), albumin (ALB), blood urea nitrogen (BUN), creatinine (CRE), estimated glomerular filtrating ratio (eGFR), uric acid (UA), amylase (AMY), total cholesterol (T-CHO), high-density lipoprotein cholesterol (HDL-C), triglyceride (TG), sodium (Na), potassium (K), chlorine (CI), calcium (Ca), inorganic phosphate (IP), iron (Fe), C-reactive protein (CRP), low-density lipoprotein cholesterol (LDL-C), white blood cell (WBC), red blood cell (RBC), hemoglobin (HGB), hamatocrit (HCT), mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), MCH concentration (MCHC), red cell distribution width (RDW), platelet (PLT), mean platelet volume (MPV), procalcitonin (PCT), platelet distribution width (PDW), blood sugar (BS), and glycated hemoglobin (HbA1c), smoking period, alcohol frequency.



**Table 7 shows the number of data, correlation coefficient (r), and P value in Spearman's correlation analysis. Values with significant correlation (P<0.05) are printed in boldface.
















TABLE 8







r value
P value




















Age
0.0640
0.2695



Height
0.0690
0.2342



Weight
−0.0180
0.7567



BMI
−0.0736
0.2045



Plaque score
0.2306

<0.0001




max-IMT
0.1157

0.0468




CAVI (right)
0.1244

0.0371




CAVI (left)
0.1133
0.0568



ABI (right)
−0.0164
0.7804



ABI (left)
−0.0559
0.3389



HbA1c
−0.0890
0.2821



W-PTH
0.0502
0.3864



Dia-period
−0.0176
0.7614



ARB
−0.0294
0.6116



ACE
0.0977
0.0910



PTA
0.1276

0.0273




Fe
−0.0456
0.4319



Ferritin
0.1655

0.0040




TSAT Ratio
0.0399
0.4910



Kt/V
−0.0746
0.1975



RBC
−0.0801
0.1664



HGB
−0.0518
0.3713



HCT
−0.0322
0.5787



PLT
−0.1867

0.0012




TP
−0.0368
0.5259



ALB
−0.1337

0.0205




UN
−0.1303

0.0240




CRE
−0.0645
0.2655



UA
−0.0570
0.3256



Na
0.0830
0.1515



K
−0.0043
0.9405



Cl
0.0494
0.3936



Ca
0.0039
0.9459



IP
−0.0242
0.6768



Mg
0.0675
0.2436



AST
0.0816
0.1586



ALT
0.0111
0.8486



LDH
0.0591
0.3076



γ-GTP
0.0811
0.1610



ALP
0.0031
0.9574



tBil
0.0344
0.5530



AMY
−0.0151
0.7940



CK
−0.0239
0.6799



T-CHO
−0.0655
0.2583



HDL-C
−0.1534

0.0078




LDL-C
−0.0099
0.8640



TG
0.0292
0.6147



CRP
0.1813

0.0016








Mode of representation of values printed in boldface, and means of abbreviation are the same as those employed in Table 7.






SUMMARY OF THE EXAMPLES

Through primary screening by use of a human protein microarray, protein KIAA0513 was identified as an antigen recognized by a serum antibody of atherosclerosis patients. Then, a recombinant GST-tagged protein KIAA0513 was purified. By use of the thus-purified protein as an antigen, the serum antibody level was determined through AlphaLISA. As a result, the s-KIAA0513-Abs level was found to be significantly higher in patients of AIS, TIA, DM, CVD, OSA, CKD, EC, GC, CC, LC, and MC than in HDs (see FIGS. 1 to 5, Tables 1 to 5). Among these diseases, a particularly high AUC value was observed in DM, Type 2 CKD, and EC (see FIGS. 2, 4, and 5). Furthermore, through correlation analysis, the serum antibody level to protein KIAA0513 was found to positively correlate with max-IMT, plaque score, and CAVI, which are indices for atherosclerosis-related lesions (see Tables 7 and 8). In contrast, the serum antibody level to protein KIAA0513 was found to weakly correlate with BS (P=0.0229) (see Table 7), but no correlation was found between the serum antibody level and HbA1c, which is a typical DM marker (see Table 7). The serum antibody level to protein KIAA0513 was relatively higher in DM patients than in HDs. However, conceivably, the antibody marker does not mainly reflect an elevated blood sugar level in DM, but shows an arteriosclerosis lesion induced by DM. Also, angiogenesis is an essential element for the progress of solid cancers such as EC. Thus, similar to arteriosclerosis, dysplasia of blood vessels in a solid cancer is thought to promote formation of protein KIAA0513. Actually, DM and arteriosclerosis are known to be risk factors for cancer.


Currently, no precise function of protein KIAA0513 has been elucidated. However, protein KIAA0513 has been reported to bind to a protein involved in synaptic transmission and apoptosis induction. Thus, conceivably, protein KIAA0513 may be involved in at least such signal transmission.


Generally, humoral antibody markers exhibit a higher sensitivity of detecting data concerning diseases, as compared with humoral antigen markers. Among humoral antigens, a KIAA0513 antigen has particularly high antigenicity. Thus, the anti-KIAA0513 humoral antibody marker exhibits remarkably high sensitivity. The above experiments have revealed that the present invention can acquire data concerning the potential or actual presence of diseases (e.g., cancers, cerebral infarction, myocardial infarction, diabetes mellitus, and chronic kidney disease) which are fatal to humans, as well as data showing the presence of an artery lesion (e.g., arteriosclerosis, or anomalous angiogenesis in solid cancer).


INDUSTRIAL APPLICABILITY

Hitherto, there have been known only a small number of easily employable markers for life-threatening grave diseases including cancers, cerebral infarction, myocardial infarction, diabetes mellitus, chronic kidney disease, etc., and individual disease-specific markers must be used. However, the data acquisition method of the present invention enables to acquire data concerning diseases at remarkably high sensitivity and to acquire comprehensive data concerning grave diseases. In addition, by acquiring data showing the presence of an artery lesion (e.g., arteriosclerosis, or anomalous angiogenesis in solid cancer), it is possible to use the acquired data as an index for disease prevention. Thus, the method of the present invention is suited for mass examination or the like.


As described above, according to the present invention, the presence of pre-existing conditions, which are factors for aggravation of an infection, can be comprehensively detected by use of a single biomarker. If the data acquisition method of the present invention is carried out in mass examination in response to epidemic of respiratory tract infection caused by novel coronavirus (COVID-19) or an influenza virus, hospital capacity including medical staff, beds, etc. can be effectively furnished, and better treatment for the infection can be attained.

Claims
  • 1. A method for acquiring data, the method comprising: determining a level of an antibody to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 which forms protein KIAA0513, or a portion of the protein, or to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 in which 10% or less of the amino acid residues (the decimal point being suppressed) are deleted, substituted, or added, or a portion of the protein, the antibody being potentially present in a body fluid sample collected from a living body; and acquiring data showing a rise in the antibody level as data showing the presence of an artery lesion.
  • 2. A method for acquiring data, the method comprising: determining a level of an antibody to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 which forms protein KIAA0513, or a portion of the protein, or to a protein having an amino acid sequence represented by SEQ ID NO: 1 or 2 in which 10% or less of the amino acid residues (the decimal point being suppressed) are deleted, substituted, or added, or a portion of the protein, the antibody being potentially present in a body fluid sample collected from a living body; and acquiring data showing a rise in the antibody level as data showing the potential or actual presence of cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer.
  • 3. The data acquisition method according to claim 2, wherein the data showing the potential or actual presence of cerebrovascular or cardiovascular disorder, diabetes mellitus, chronic kidney disease, or a solid cancer are acquired as data concerning an artery lesion involved in the diseases.
  • 4. The data acquisition method according to any one of claims 1 to 3, wherein a portion of the protein having an amino acid sequence represented by SEQ ID NO: 1 has 10 to 301 amino acid residues.
  • 5. The data acquisition method according to any one of claims 1 to 4, wherein the body fluid sample collected from a living body is a blood sample.
  • 6. The data acquisition method according to claim 5, wherein the blood sample is a serum or plasma sample.
  • 7. The data acquisition method according to any one of claims 1 to 6, wherein the antibody level is determined through immuno-chromatography, ELISA, AlphaLISA, indirect immunofluorescence, Western blotting (immunoblotting), turbidimetry, nephelometry, latex-coagulation turbidimetry, or CLEIA.
  • 8. A kit for acquiring data, the kit being employed in carrying out the data acquisition method as recited in any one of claims 1 to 7.
Priority Claims (1)
Number Date Country Kind
2020-170489 Oct 2020 JP national
PCT Information
Filing Document Filing Date Country Kind
PCT/JP2021/035474 9/27/2021 WO