Journal List > Investig Clin Urol > v.61(Suppl 1) > 1141864

Kim, Kim, and Kim: Advances in urinary biomarker discovery in urological research

Abstract

A disease-specific biomarker (or biomarkers) is a characteristic reflecting a pathological condition in human body, which can be used as a diagnostic or prognostic tool for the clinical management. A urine-based biomarker(s) may provide a clinical value as attractive tools for clinicians to utilize in the clinical setting in particular to bladder diseases including bladder cancer and other bladder benign dysfunctions. Urine can be easily obtained by patients with no preparation or painful procedures required from patients' side. Currently advanced omics technologies and computational power identified potential omics-based novel biomarkers. An unbiased profiling based on transcriptomics, proteomics, epigenetics, metabolomics approaches et al. found that expression at RNA, protein, and metabolite levels are linked with specific bladder diseases and outcomes. In this review, we will discuss about the urine-based biomarkers reported by many investigators including us and how these biomarkers can be applied as a diagnostic and prognostic tool in clinical trials and patient care to promote bladder health. Furthermore, we will discuss how these promising biomarkers can be developed into a smart medical device and what we should be cautious about toward being used in real clinical setting.

INTRODUCTION

The bladder is a hollow, soft muscular organ located in the lower abdomen, which stores urine until it is ready to excrete. In urological diseases, the incidence of bladder diseases is quite high. The common bladder diseases include bladder cancer (BC), bladder dysfunction (cystitis, urinary incontinence, overactive bladder, etc.) and other bladder problems. BC is the sixth most common cancer in the United States, accounting for 4.7% of cancer cases [1]. About 45,000 men and 17,000 women in the United States are diagnosed as BC every year.
Interstitial cystitis (IC) is the most common disease in bladder dysfunction. According to the International Continence Society, the definition of IC is “the complaint of suprapubic pain related to bladder filling, accompanied by other symptoms such as increased daytime and nighttime frequency, in the absence of proven urinary infection or other obvious pathology.” [2]. The morbidity of IC in the general population is 0.26% to 12.6% [34]. The estimated morbidity of IC in women is 45/100,000, which is 4 to 5 times than that in men, with the morbidity of 8/100,000 [5]. In the United States, 3.3 million women are diagnosed as IC every year [6]. At present, one of the most important methods to diagnose bladder diseases is cystoscopy, but this technique is invasive and may lead to urinary tract infection. Compared to cystoscopy, urine testing is easier to perform in clinical practice. Urine can be obtained non-invasively and shows increased stability over serum or blood, which allows for easy multiple sampling. With the direct contact between urine and bladder diseases, the use of urinary biomarkers detection in bladder diseases becomes more and more important.

WILL IT BE USEFUL THE URINE-BASED DIAGNOSTIC BIOMARKERS TO DETECT AND MONITOR THE BLADDER DISEASES?

Urinary biomarkers are particularly attractive due to the direct contact of the urine with the urothelial tumor cells and the ease of sample collection. Urine-based diagnostic biomarkers are reviewed in our paper from the following aspects: gene mutations and gene expression-based biomarkers, proteomic biomarkers, metabolomic biomarkers, and DNA methylation biomarkers.

1. Gene mutations associated with BC

The exact cause of BC is still unclear. There are several risk factors related to BC, including environment, smoking, toxic industrial chemicals and gases, bladder inflammation, and gene mutations. As a noninvasive method, detecting mutant genes in urine plays an important role in the diagnosis of BC.
A study of Zhu et al. [7] indicated 14 important mutation genes related to BC by searching the Catalogue Of Somatic Mutations In Cancer (COSMIC) database. The mutation genes included P53, fibroblast growth factor receptor 3 (FGFR3), TSC complex subunit 1 (TSC1), stromal antigen 2 (STAG2), HRas proto-oncogene (HRAS), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), Erb-B2 receptor tyrosine kinase 3 (ERBB3), neurofibromatosis type 1 (NF1), ERBB2, FGFR1, cyclin-dependent kinase inhibitor 2A (CDKN2A), AT-rich interaction domain 1A (ARID1A), histone-lysine N-methyltransferase 2D (KTM2D), and CREB binding protein (CREBBP). Several studies showed that the development of BC is associated with the mutations of P53 gene [891011]. Sidransky et al. [12] first described the mutations of P53 gene in the urine of BC patients in 1991. They found that alterations in P53 gene were associated with poor differentiation, advanced urothelial cell carcinoma and poor prognosis [89]. Traczyk-Borszynska et al. [13] showed that the mutations of P53 gene were more common in clinically and histologically advanced carcinoma, and were the negative prognostic factor in BC. FGFR3 mutations also participated in the development of BC. A study showed that mutations of the FGFR3 gene were surrogate markers for the detection of genome stable bladder tumors [14]. Another study indicated that FGFR3 mutations were the feature of well-differentiated BC but not the prognostic marker in BC [13]. In other studies, van Rhijn et al. [15] showed that the combination of FGFR3 with MIB-1 (Ki67) had a more accurate prediction of the progression and survival in BC. Ploussard et al. [16] found that the progression and recurrence of FGFR3 mutations in disease depended on allele loss of 9p22. Also, Rebouissou et al. [17] found that the progression of FGFR3 mutations in non-muscle-invasive disease depended on the homozygous deletion of 9p21. HRAS is a proto-oncogene, which may promote tumorigenesis in several organs including the bladder. HRAS gene mutations in bladder cells were associated with BC, but the mutation rate was low. A study showed that the mutation rate of HRAS gene varies greatly in BC (0%–30%) [18]. Beukers et al. [19] indicated that HRAS gene mutations were more likely to occur in young BC patients (<20 years) compared with older patients. It suggested that mosaicism of oncogenic HRAS mutations may increase the risk of developing BC at a young age.
Several studies showed that TSC1 had inactive point mutations on 9q34 in10% to 15% of BC patients, resulting in complete loss of function of TSC1 [202122]. Also, the deletion of a single TSC1 allele may promote the growth of bladder epithelial cells and therefore promote the development of BC [23]. STAG2 mutations were recently identified in BC patients. However, the significance of STAG2 mutations remains controversial. Solomon et al. [24] showed that loss of STAG2 promoted the lymph node metastases in BC and increased the risk of recurrence and mortality. However, different subtypes of BC may exhibit different mutations [25]. In several studies, loss of STAG2 was reported to be associated with BC in low stage and low grade [262728]. Lelo et al. [29] found that STAG2 mutations were much more common in non-muscle invasive BC (32%) than in muscle invasion BC (12%). These studies suggested that STAG2 could be a potentially useful biomarker for predicting recurrence and progression in non-muscle invasive BC. BRCA1-associated protein 1 (BAP1) is a nuclear ubiquitin carboxyterminal hydrolase or deubiquitinating enzyme which can regulate several cellular functions, including cell cycle, differentiation, proliferation, and DNA damage response [30]. The recent research indicated that BAP1 mutations were related to BRCA pathway alterations in BC. Lin et al. [31] indicated that patients carrying BAP1 genetic variant alleles of rs12163565 had an increased risk of developing BC, although the increased risk was not statistically significant (odds ratio, 1.17; p=0.070). There were studies showed that PIK3CA gene alterations, including mutations, copy gains or amplifications, were associated with non-muscle invasive BC [3233]. Dueñas et al. [33] showed that PIK3CA gene alterations were frequent and associated with low recurrence and low progression in non-muscle invasive BC, which indicated that PIK3CA may be a potential biomarker for predicting recurrence and progression in non-muscle invasive BC. Collectively, many genes mutations have been found in BC patients. Further studies are required to discover more gene mutations and new biomarkers in BC before they can be used in clinical practice.

2. Gene expression-based BC biomarkers

Gene expression-based urinary biomarkers have good sensitivity and specificity in the detection of BC. They are less likely to be affected by inflammatory and other benign conditions. Several important genetic changes in BC have been identified in the past two decades. Based on the technology of rapid nucleic acid extraction and the proven stability of DNA and RNA in urine, gene expression-based biomarkers play an important role in the detection of BC.
A study from Beukers et al. [34] showed that FGFR3, telomerase reverse transcriptase (TERT), and orthodenticle homeobox 1 (OTX1) were significant in the diagnosis of BC. They acted as a urinary biomarker combination with a sensitivity of 57% in low grade primary BC patients and 83% in pT1 or muscle invasive BC. In a study of Holyoake et al. [35], the researchers used microarray data from BC patients and healthy controls to generate a panel of genes that were differentially expressed in various stages and grades of BC patients and normal controls. They tested the markers in voided-urine samples to generate an mRNA panel, including cyclin-dependent kinase-1 (CDK1; also known as CDC2), midkine (MDK), insulin like growth factor binding protein 5 (IGFBP5), and homeobox A3 (HOXA3), which could predict the presence of BC with a sensitivity of 48% to 100% and a specificity of 85%. Park et al. [36] examined aurora kinase A (AURKA) gene amplification in exfoliated cells in urine samples. They concluded that AURKA could be a biomarker for the detection of BC with a specificity of 96.6% and a sensitivity of 87%, and the degree of gene amplification was also associated with high grade BC. Urquidi et al. [37] used Affymetrix arrays of 92 patients (52 BC and 40 controls) and derived a 14 gene panel that could predict the presence of BC, with high sensitivity and specificity (90% and 100%, respectively) and AUC (area under the receiver operating curve) of 0.98. The 14 genes were: carbonic anhydrase 9 (CA9), transmembrane protein 45A (TMEM45A), C-C motif chemokine ligand 18 (CCL18), matrix remodeling associated 8 (MXRA8), matrix metallopeptidase 9 (MMP9), semaphorin 3D (SEMA3D), ERBB2, vascular endothelial growth factor A (VEGFA), desmocollin 2 (DSC2), Ras-related protein Rab-1A (RAB1A), angiotensinogen (AGT), synaptogyrin 1 (SYNGR1), deleted in malignant brain tumors 1 (DMBT1), angiogenin (ANG). The first seven genes were upregulated and the last seven genes were down-regulated in the urines of BC patients. Bongiovanni et al. [38] found that the expression levels of septin 4 (SEPT4) were up-regulated in the urine of BC patients, with a sensitivity of 93%, a specificity of 65%, and AUC of 0.798. All these studies have shown promise in the diagnosis of BC. However, the majority of them remain in the discovery phase.
MicroRNAs (miRNAs) are a class of small, endogenous, noncoding RNA. They regulate gene expression by affecting mRNA translation and stability or by modulating promoter activity of their target genes. In oncology, miRNAs are considered as promising biomarkers for early diagnosis, prognosis evaluation and therapeutic response prediction of the tumor. A large number of studies showed that miRNAs acted as diagnostic biomarkers in urine samples of BC patients [3639404142]. Some miRNAs were down-regulated such as miR-125b, miR-140-5p, miR-141, miR-200a, miR-200c, and others were up-regulated such as miR-18a, miR-92a, miR-96. Other studies indicated that miR-126, miR-152, miR-222, and miR-452 were up-regulated in BC [434445]. However, miR-200 family, miR-155, miR-192, miR-205, and miR-143 were found to be down-regulated in studies [4446]. Eissa et al. [47] found that the levels of miR-324-5p, miR-4738-3p, and FOSB mRNA were up-regulated in the urine of BC patients, whereas lncRNA miR-497-HG and RCAN1 mRNA were down-regulated in BC patients, compared with patients with benign lesions and healthy controls. The sensitivities and accuracies of the RNAs were significantly higher than those of cytology. In the urinary ceRNA: lncRNA-miRNA-mRNA network, 2 mRNAs (FOS B and RCAN1) displayed the highest accuracy for the diagnosis of BC. A study of Chen [48] showed that miR-101 was decreased in BC patients, and was negatively associated with aggressive clinical characteristics, with a sensitivity of 82.0% and a specificity of 80.9% in BC.
Most of the studies on miRNAs were different in methodology, with little overlap, and no results were fully validated. At present, there are no valid conclusions about urinary miRNAs in the detection of BC patients. Multicenter prospective validation studies in large clinical settings are needed in the future.

3. Proteomics profiling revealed urinary biomarkers for BC

The urinary proteome enriched in proteins reflects the development and invasion of the tumor through direct contact with BC. The study of urinary proteomic biomarkers has been mainly used to help diagnose primary and recurrent BC and to assess the aggressiveness of the disease.
Nuclear matrix protein 22 (NMP22) is one urinary biomarker approved by the U.S. Food and Drug Administration (FDA) using enzyme-linked immunosorbent assay (ELISA) test and BladderChek point-of-care test [4950]. However, in a meta-analysis of 19 studies for the detection of BC, the sensitivity of NMP22 was 52% to 59% and the specificity was 87–89%, with an AUC of 0.83 [51]. Another biomarker approved by FDA is the bladder tumor antigen (BTA), also known as human complement factor H related protein (hCFHrp). In a meta-analysis of 13 studies using BTA STAT test, the sensitivity of BTA was 64% to 69% and the specificity was 73% to 77% [52]. In a meta-analysis of 5 studies using BTA STAT test, the sensitivity of BTA was 62% to 71% and the specificity was 45% to 81% [53]. Both of the two markers above were not good in sensitivities and specificities.
An ideal protein biomarker should be the one with high sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and AUC values [54]. Several studies showed that Apo-A1, BLCA-4, and hyaluronidase in urine were independently validated in BC with high sensitivities and specificities [555657585960]. Apo-A1 is the primary protein component of high-density lipoprotein, which may improve tumor angiogenesis through kinase activation [6162]. But the association between lipoproteins and BC progression is still not very clear. Studies showed that Apo-A1 was independently validated in BC with the sensitivity of 89% to 95% and the specificity of 85% to 92% [555657]. BLCA-4 is a nuclear transcription factor found in the early stages of BC. Cai et al. [58] found that BLCA-4 was independently validated with the sensitivity of 93% and the specificity of 97% through an analysis of nine studies. Hyaluronidase could improve cellular proliferation and motility through hyaluronic acid [63]. Studies of Eissa et al. [59] and Pham et al. [60] showed the sensitivity and specificity of hyaluronidase ranged from 87% to 100% and 89% to 98% respectively. Besides the three proteins, there were several additional urine proteins that exhibited with high sensitivities and specificities, but they have not yet been independently validated, including ANG, apolipoprotein E (APOE), CA-9, interleukin-8 (IL-8), MMP, MMP10, plasminogen activator inhibitor 1 (PAI-1), VEGF [6364]. Goodison et al. [63] found that the eight-biomarker panel above achieved a sensitivity of 92% and a specificity of 97%, while the BTA TRAK ELISA test achieved a sensitivity of 78% and a specificity of 83% in the same cohort for BC detection. Another study of Urquidi et al. [65] showed that urine CCL18 achieved a sensitivity of 88% and a specificity of 86%, while BTA TRAK ELISA achieved a sensitivity of 80% and a specificity of 84% in the same cohort for BC detection. All of the biomarkers above had better sensitivities and specificities than BTA. These head-to-head studies compared the biomarkers with the FDA-approved test in the same patient cohort, increasing the validity of the studies.

4. Proteomics profiling revealed urinary biomarkers for IC

IC/bladder pain syndrome (BPS) is the most common disease in bladder dysfunction. At present, the etiology of IC/BPS is still not fully understood. There are several possible mechanisms, including infection, inflammation, toxic substances absorption, mucus layer with deficient glycosaminoglycan, hypoxia, and genetics. So far there are no gold standards in the diagnosis of IC/BPS. Some invasive testing including biopsy, urodynamic, and cystoscopy are applied to help diagnose the disease. However, there is still a lack of tools to facilitate accurate diagnosis and objective follow-up. Therefore, it is significant to investigate urinary biomarkers that can be used in clinical practice.
A study by Magalhaes et al. [66] reviewed the urinary biomarkers associated with IC/BPS. They found potential biomarkers investigated in urine specimens included macrophage inhibitory factor (MIF), nerve growth factor (NGF), methylhistamine, histamine, IL-6, antiproliferative factor (APF), epithelial growth factor (EGF), heparin-binding (HB)-EGF, glycoprotein G5P1, and a chemokine profile. Tonyali et al. [67] detected urinary NGF and nerve density in the bladder mucosa. They found that urinary NGF/Cr was significantly increased in IC/BPS patients comparing to control groups, which was similar to nerve density. Corcoran et al. [68] assessed both urine samples and bladder biopsy samples to determine the profile of 23 chemokines in 10 IC/BPS patients and 10 controls. The results indicated that univariate analysis showed no significant differences in any of the urinary proteins assessed, but multivariate analysis showed that VCAM-1 and ICAM-1 in urine were significantly different between IC/BPS and controls. A study of Vera et al. [69] studied urinary MIF concentrations in subgroups of BPS with and without Hunner lesions and control groups. They verified that urinary MIF was significantly higher in BPS patients with Hunner lesion compared with patients without Hunner and with controls, with a sensitivity of 74.4%, a specificity of 71.8%, and AUC 0.718. For the urinary MIF/Cr ratio, the sensitivity was 47%, the specificity was 91% and AUC was 0.730 in identifying patients with IC/BPS and Hunner lesions. Lamale et al. [70] investigated urinary histamine, IL-6, and methylhistamine in IC/BPS patients and controls. They found that urinary concentrations of histamine and IL-6 were increased in IC/BPS patients. However, methylhistamine levels had no significant differences between IC/BPS patients and controls. Further logistic regression analysis demonstrated that the best predictor for IC/BPS was a combined model with IL-6 and methylhistamine, with an AUC of 0.788. Furthermore, Keay et al. [71] found that APF was increased in IC/BPS patients compared to controls, but HB-EGF concentrations were decreased in IC/BPS patients. Byrne et al. [72] demonstrated that glycoprotein G5P1 concentration in urine was lower in IC/BPS patients than that in controls.
In general, urine proteomic biomarkers of bladder diseases have great promise, but the best biomarkers with the highest clinical utility remain to be discovered. There is still a need for more comprehensive screening of urine proteomic markers through extensive multi-institution validation. Table 1 shows the urinary biomarkers suggested for BC and IC diagnosis [5152535556575859606365676869707172].

5. Metabolomic biomarkers for BC

At present, urinary metabolomic biomarker studies are primarily conducted either by NMR-based or mass spectrometry (MS)-based identification. Three metabolites (2,5-furandicarboxylic acid, ribitol, and ribonic acid) were found to be lower in the urine of BC patients than in healthy controls [737475]. Taurine is the metabolite known as a free-radical scavenger that can prevent cell damage. Studies showed that taurine was elevated in the urine of BC patients than in healthy controls [7576]. Several studies showed that urinary citrate, succinate, and hippurate were reduced in BC patients compared with control groups, which suggested that citrate changes were related to an altered tricarboxylic acid (TCA) cycle in BC metabolism [7374757677]. On the study of glycolysis-related metabolites, decreased fructose levels and increased lactate levels were showed in BC patients [7375]. Urinary acetylcarnitine and adipate in BC patients were elevated, which were the results of disturbed fatty acid transportation, altered mitochondrial TCA cycle, and energy metabolism processes or an excess of acetyl-CoA production [747577].
Wittmann et al. [75] identified between 178 and 233 discriminating metabolites (depending on the respective comparison) in a retrospective MS study. They compared current BC patients with three different control groups: patients with haematuria, controls with BC in the past but without the current disease, and a mixed group of patients with haematuria, those with BC in the past and some healthy subjects. They found that 3-hydroxybutyrate and gluconate were the most highly increased in BC patients, while anserine, 3-hydroxyphenylacetate and pyridoxate showed the lowest values in BC patients. In another high-resolution liquid chromatography (LC)-MS study, glycolysis and acylcarnitines were increased in BC than a combined control group (patients with haematuria and healthy controls) [78]. Besides, amino acid metabolism and fatty acid oxidation were also important factors in BC pathology. A study showed that the acylcarnitines, decanoylcarnitine, decenoylcarnitine, hydroxynonanoylcarnitine and hydroxybutyrylcarnitine were all increased in BC patients [79]. These urinary metabolomic biomarkers may have potential significance in the diagnosis of BC.

6. Metabolomic biomarkers for IC

In the research of IC/BPS, Parker et al. [80] used LC-MS in urine samples of 40 women with IC/BPS and 40 controls to determine metabolomic profiles. They found six metabolites were closely associated with IC/BPS. One of them was etiocholan-3alpha-ol-17-one (Etio-S). The elevated Etio-S was a good predictor of IC/BPS, with a sensitivity of 91.2%, a specificity of 87.4%, and AUC of 0.92. Longitudinal analysis of women in this cohort showed that the differences in Etio-S persisted, indicating that these changes could last long.
The results from these early studies on metabolomic biomarkers suggest that urine may act as a potential tool on screening or monitoring bladder diseases in the clinical field, but it is still in the discovery phase. More large multicenter studies with independent validation cohorts are needed to advance the field. Table 2 shows the urinary biomarkers suggested for IC diagnosis [7374757677787980].

7. DNA methylation biomarkers for BC

DNA methylation has been recognized to be important in developmental biology and cancer etiology [81]. Aberrant DNA methylation is a major characteristic of BC and it plays an important role in tumor occurrence and progression [828384]. Compared to RNA or protein, DNA is inherently stable, so it is more powerful in cancer detection. Chan et al. [85] examined the DNA methylation of seven genes (the retinoid acid receptor-β [RARβ]), death associated protein kinase 1 (DAPK1), Ecadherin, CDKN2A (p16), p15INK4b (p15), glutathione S-transferase Pi 1 (GSTP1), and O-6-methylguanine-DNA methyltransferase (MGMT) in voided urine of BC patients and ageand sexmatched controls. Four biomarkers DAPK1, RARβ, E-cadherin, and p16) achieved a sensitivity of 91% and a specificity of 76% for detecting BC. And cytology achieved a sensitivity of 46% and a specificity of 100% by comparison. Friedrich et al. [86] examined DNA methylation of apoptosisassociated genes (DAPK, TERT, and apoptosis regulator [BCL2]) in the urine of BC patients. They found that combined methylation analyses achieved both high sensitivity and specificity (78% and 100%, respectively) for detecting BC. In another study, Hoque et al. [87] examined the DNA methylation of nine genes (adenomatous polyposis coli (APC), ARF tumor suppressor (p14ARF), cadherin-1 (CDH1), GSTP1, MGMT, cyclin dependent kinase inhibitor 2A (CDKN2A), retinoic acid receptor beta (RARβ2), ras association domain family member 1 (RASSF1A), and TIMP metallopeptidase inhibitor 3 (TIMP3). They found that combined methylation analysis based on four genes (CDKN2A, p14ARF, MGMT, and GSTP1) achieved a sensitivity of 69% and a specificity of 100%. Recently there were some studies on Twist family BHLH transcription factor 1 (TWIST1) and nidogen 2 (NID2) genes. Renard et al. [88] reported that TWIST1 and NID2 genes were frequently methylated in BC patients in a total of 496 urine samples collected from three urology clinical sites. The sensitivity of this 2 gene panel was significantly better than that of cytology (90% and 48%, respectively), with the specificity of 93% and 96%, respectively. The PPV and NPV of the 2 gene panel was 86% and 95%, respectively. However, the sensitivities of these two genes were poor in the studies of Abern et al. [89] and Fantony et al. [90]. In other studies, Reinert et al. [91] found a 4marker panel (zinc finger protein 154 [ZNF154], homeobox protein Hox-A9 (HOXA9), POU class 4 homeobox 2 (POU4F2), and eomesodermin (EOMES) achieved a sensitivity of 84% and a specificity of 96% for detecting BC in urine samples from 119 BC patients and 59 controls. Another study of Reinert et al. [92] found a 6-marker panel (EOMES, HOXA9, POU4F2, TWIST1, vimentin (VIM), and ZNF154) had a sensitivity of 82% to 89% and a specificity of 94% to 100% for detecting BC in urine samples from 184 BC patients and 35 controls.
In a study of 368 urine samples collected from 90 non-muscle invasive BC patients, Su et al. [93] reported that a panel of 3 markers (SRY-box transcription factor 1 [SOX1], interleukin 1 receptor associated kinase 3 [IRAK3], and L1MET) discriminated between patients with recurrence and with no recurrence, with a sensitivity of 86% and a specificity of 89% of patients with recurrence, compared with the sensitivity of 80% and specificity of 97% of patients with no recurrence in validation sets. The results demonstrated that the combination of SOX1, IRAK3, and L1MET could detect disease recurrence with high sensitivity and specificity. Another study selected seven DNA methylation biomarkers (CDH13, cystic fibrosis transmembrane conductance regulator [CFTR], NID2, spalt like transcription factor 3 (SALL3), transmembrane protein with EGF like and two follistatin like domains 2 [TMEFF2], TWIST1, and VIM2) from four recently published BC studies [819495]. They found that the best possible combination to discriminate against BC from controls was the combination CFTR, SALL3, and TWIST1 [96]. The three-gene methylation classifier achieved an AUC of 0.874, with a sensitivity of 85% and a specificity of 68%. The discovery of highly sensitive methylation biomarkers may allow us to lower the number of follow-up cystoscopies in patients with BC, which can improve the life quality of the patients.

8. DNA methylation biomarkers for IC

In the research of IC/BPS, Magalhaes et al. [66] concluded that DNA methylation in urine samples was associated with IC/BPS. Bradley et al. [97] determined DNA methylation profiles in IC/BPS and controls. After Bonferroni correction, there was no genome-scale significantly different methylation in CpG sites. Among the methylated CpG sites, the most prominent enrichment pathway was the mitogen-activated protein kinase (MAPK) pathway. This pathway had 86% of sites with hypomethylation in IC/BPS patients compared to the controls.
There is evidence that DNA methylation biomarkers are more sensitive than cytology although there were biomarkers tested on cohorts that varied between studies. And some markers showed specificity comparable with that of cytology. A highly selective panel of methylation biomarkers may increase the sensitivity and specificity of urine analysis in the clinical studies [98]. Standardized assays and cutoff values should be used in a large and well-designed cohort in future studies. Table 3 summarizes urine epigenetics-based biomarkers for BC and IC [34353637384344454647668586878889909192939697].

CONCLUSIONS

Many studies have shown that urinary-based biomarkers have high sensitivity and specificity in the diagnosis of bladder diseases (such as BC and IC), which confirms the feasibility of using urinary exfoliated epithelium as an analyzer to diagnose bladder diseases. As shown in Table 4 [3451525399], commercially available biomarker kits for diagnosis of bladder disease such as BC have been introduced in market. If this method is accurate and reliable enough, it can be used not only for the diagnosis of bladder diseases but also for the screening of diseases in the population. However, further researches are needed to apply urinary biomarkers to clinical practice. More efforts should be made to improve and validate the biomarker panel and promote the progress of urine-based biomarker analysis, which will be applied to clinical work as soon as possible.

Figures and Tables

Table 1

Proteomics-based urinary biomarkers

icu-61-S8-i001

AUC, area under the receiver operating curve; BC, bladder cancer; NMP22, nuclear matrix protein 22; ELISA, enzyme-linked immunosorbent assay; FDA, U.S. Food and Drug Administration; BTA, bladder tumor antigen; NO, not reported; OUC, controls with other urinary conditions; qPCR, quantitative polymerase chain reaction; HC, healthy controls; PPV, positive predictive value; ANG, angiogenin; APOE, apolipoprotein E; CA9, carbonic anhydrase 9; IL, interleukin; MMP, matrix metallopeptidase; PAI-1, plasminogen activator inhibitor 1; VEGF, vascular endothelial growth factor; CCL18, C-C motif chemokine ligand 18; NPV, negative predictive value; IC/BPS, interstitial cystitis/bladder pain syndrome; NGF, nerve growth factor; MIF, macrophage inhibitory factor; APF, antiproliferative factor; HB-EGF, heparin-binding epithelial growth factor.

Table 2

Metabolomics urinary biomarkers

icu-61-S8-i002

AUC, area under the receiver operating curve; BC, bladder cancer; GC×GC-TOFMS, two-dimensional gas chromatography time-of-flight mass spectrometry; NO, not reported; UHPLC-MS/MS, ultrahigh-performance liquid chromatography/tandem mass spectrometry; GC-MS, gas chromatography-mass spectrometry; LC-MS, liquid chromatography-mass spectrometry; Etio-S, etiocholan-3alpha-ol-17-one.

Table 3

Epigenetic urinary biomarkers

icu-61-S8-i003

AUC, area under the receiver operating curve; BC, bladder cancer; FGFR3, fibroblast growth factor receptor 3; TERT, telomerase reverse transcriptase; OTX1, orthodenticle homeobox 1; RT-PCR, reverse transcriptase-polymerase chain reaction; NO, not reported; MDK, midkine; IGFBP5, insulin like growth factor binding protein 5; HOXA3, homeobox A3; AURKA, aurora kinase A; FISH, fluorescence in situ hybridization; CA9, carbonic anhydrase 9; TMEM45A, transmembrane protein 45A; CCL18, C-C motif chemokine ligand 18; MXRA8, matrix remodeling associated 8; MMP9, matrix metallopeptidase 9; SEMA3D, semaphorin 3D; ERBB2, Erb-B2 receptor tyrosine kinase 2; VEGFA, vascular endothelial growth factor A; DSC2, desmocollin 2; RAB1A, Ras-related protein Rab-1A; AGT, angiotensinogen; SYNGR1, synaptogyrin 1; DMBT1, deleted in malignant brain tumors 1; ANG, angiogenin; SEPT4, septin 4; RT-qPCR, quantitative reverse transcriptase-polymerase chain reaction; DAPK, death associated protein kinase; RARβ, retinoid acid receptor-β; MSP, methylation-specific polymerase chain reaction; CDKN2A, cyclin dependent kinase inhibitor 2A; MGMT, O-6-methylguanine-DNA methyltransferase; GSTP1, glutathione S-transferase Pi 1; qMSP, specific high-resolution melting; TWIST1, Twist family BHLH transcription factor 1; NID2, nidogen 2; ZNF154, zinc finger protein 154; HOXA9, homeobox protein Hox-A9; POU4F2, POU class 4 homeobox 2; EOMES, eomesodermin; VIM, vimentin; MS-HRM, methylation-specific high-resolution melting; SOX1, SRY-box transcription factor 1; IRAK3, interleukin 1 receptor associated kinase 3; CFTR, cystic fibrosis transmembrane conductance regulator; SALL3, spalt like transcription factor 3; HB-EGF, heparin-binding epithelial growth factor.

Table 4

Commercially available biomarker kits

icu-61-S8-i004

FDA, U.S. Food and Drug Administration; NMP22, nuclear matrix protein 22; BTA, bladder tumor antigen; FGFR3, fibroblast growth factor receptor 3.

ACKNOWLEDGMENTS

The authors acknowledge support from National Institutes of Health grants (1U01DK103260), Department of Defense grants (W81XWH-15-1-0415), Centers for Disease Controls and Prevention (1U01DP006079). The research was supported by the International Science and Business Belt Program through the Ministry of Science, ICT and Future Planning (2017K000490) Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2017R1D1A3B03031486), and the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (2018R1A2B2005473).

Notes

CONFLICTS OF INTEREST The authors have nothing to disclose.

AUTHORS' CONTRIBUTIONS

  • Research conception and design: Wun-Jae Kim.

  • Data acquisition: Jayoung Kim.

  • Data analysis and interpretation: Jayoung Kim.

  • Drafting of the manuscript: Jayoung Kim.

  • Critical revision of the manuscript: Won Tae Kim.

  • Administrative, technical, or material support: Won Tae Kim.

  • Supervision: Wun-Jae Kim.

  • Approval of the final manuscript: all authors.

References

1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018; 68:7–30.
crossref pmid
2. Abrams P, Cardozo L, Fall M, Griffiths D, Rosier P, Ulmsten U, et al. The standardisation of terminology of lower urinary tract function: report from the Standardisation Sub-committee of the International Continence Society. Am J Obstet Gynecol. 2002; 187:116–126.
crossref pmid
3. Choe JH, Son H, Song YS, Kim JC, Lee JZ, Lee KS. Prevalence of painful bladder syndrome/interstitial cystitis-like symptoms in women: a population-based study in Korea. World J Urol. 2011; 29:103–108.
crossref pmid
4. Temml C, Wehrberger C, Riedl C, Ponholzer A, Marszalek M, Madersbacher S. Prevalence and correlates for interstitial cystitis symptoms in women participating in a health screening project. Eur Urol. 2007; 51:803–808.
crossref pmid
5. Patnaik SS, Laganà AS, Vitale SG, Butticè S, Noventa M, Gizzo S, et al. Etiology, pathophysiology and biomarkers of interstitial cystitis/painful bladder syndrome. Arch Gynecol Obstet. 2017; 295:1341–1359.
crossref pmid
6. Berry SH, Elliott MN, Suttorp M, Bogart LM, Stoto MA, Eggers P, et al. Prevalence of symptoms of bladder pain syndrome/interstitial cystitis among adult females in the United States. J Urol. 2011; 186:540–544.
crossref pmid pmc
7. Zhu F, Zhang Y, Shi L, Wu CL, Chen SQ, Zheng H, et al. Gene mutation detection of urinary sediment cells for NMIBC early diagnose and prediction of NMIBC relapse after surgery. Medicine (Baltimore). 2019; 98:e16451.
crossref
8. van Rhijn BW, van der Kwast TH, Vis AN, Kirkels WJ, Boevé ER, Jöbsis AC, et al. FGFR3 and P53 characterize alternative genetic pathways in the pathogenesis of urothelial cell carcinoma. Cancer Res. 2004; 64:1911–1914.
crossref pmid
9. Ecke TH, Sachs MD, Lenk SV, Loening SA, Schlechte HH. TP53 gene mutations as an independent marker for urinary bladder cancer progression. Int J Mol Med. 2008; 21:655–661.
crossref pmid
10. Goebell PJ, Groshen SG, Schmitz-Dräger BJ. International Study-Initiative on Bladder Cancer (ISBC). p53 immunohistochemistry in bladder cancer--a new approach to an old question. Urol Oncol. 2010; 28:377–388.
crossref pmid
11. Malats N, Bustos A, Nascimento CM, Fernandez F, Rivas M, Puente D, et al. P53 as a prognostic marker for bladder cancer: a meta-analysis and review. Lancet Oncol. 2005; 6:678–686.
crossref pmid
12. Sidransky D, Von Eschenbach A, Tsai YC, Jones P, Summerhayes I, Marshall F, et al. Identification of p53 gene mutations in bladder cancers and urine samples. Science. 1991; 252:706–709.
crossref pmid
13. Traczyk-Borszynska M, Borkowska E, Jablonowski Z, Jedrzejczyk A, Pietrusinski M, Kaluzewski B, et al. Genetic diversity of urinary bladder cancer and the risk of recurrence based on mutation analysis. Neoplasma. 2016; 63:952–960.
crossref pmid
14. Junker K, van Oers JM, Zwarthoff EC, Kania I, Schubert J, Hartmann A. Fibroblast growth factor receptor 3 mutations in bladder tumors correlate with low frequency of chromosome alterations. Neoplasia. 2008; 10:1–7.
crossref pmid pmc
15. van Rhijn BW, Zuiverloon TC, Vis AN, Radvanyi F, van Leenders GJ, Ooms BC, et al. Molecular grade (FGFR3/MIB-1) and EORTC risk scores are predictive in primary non-muscle-invasive bladder cancer. Eur Urol. 2010; 58:433–441.
crossref pmid
16. Ploussard G, Soliman H, Dubosq F, Méria P, Vérine J, Desgrand-Champs F, et al. The prognostic value of FGFR3 mutational status for disease recurrence and progression depends on allelic losses at 9p22. Am J Cancer Res. 2011; 1:498–507.
17. Rebouissou S, Hérault A, Letouzé E, Neuzillet Y, Laplanche A, Ofualuka K, et al. CDKN2A homozygous deletion is associated with muscle invasion in FGFR3-mutated urothelial bladder carcinoma. J Pathol. 2012; 227:315–324.
crossref pmid
18. Fernández-Medarde A, Santos E. Ras in cancer and developmental diseases. Genes Cancer. 2011; 2:344–358.
pmid pmc
19. Beukers W, Hercegovac A, Zwarthoff EC. HRAS mutations in bladder cancer at an early age and the possible association with the Costello Syndrome. Eur J Hum Genet. 2014; 22:837–839.
crossref pmid
20. Knowles MA, Habuchi T, Kennedy W, Cuthbert-Heavens D. Mutation spectrum of the 9q34 tuberous sclerosis gene TSC1 in transitional cell carcinoma of the bladder. Cancer Res. 2003; 63:7652–7656.
pmid
21. Knowles MA, Platt FM, Ross RL, Hurst CD. Phosphatidylinositol 3-kinase (PI3K) pathway activation in bladder cancer. Cancer Metastasis Rev. 2009; 28:305–316.
crossref pmid pmc
22. Pymar LS, Platt FM, Askham JM, Morrison EE, Knowles MA. Bladder tumour-derived somatic TSC1 missense mutations cause loss of function via distinct mechanisms. Hum Mol Genet. 2008; 17:2006–2017.
crossref pmid pmc
23. Goebell PJ, Knowles MA. Bladder cancer or bladder cancers? genetically distinct malignant conditions of the urothelium. Urol Oncol. 2010; 28:409–428.
crossref pmid
24. Solomon DA, Kim JS, Bondaruk J, Shariat SF, Wang ZF, Elkahloun AG, et al. Frequent truncating mutations of STAG2 in bladder cancer. Nat Genet. 2013; 45:1428–1430.
crossref pmid pmc
25. Aquila L, Ohm J, Woloszynska-Read A. The role of STAG2 in bladder cancer. Pharmacol Res. 2018; 131:143–149.
crossref pmid
26. Guo G, Sun X, Chen C, Wu S, Huang P, Li Z, et al. Whole-genome and whole-exome sequencing of bladder cancer identifies frequent alterations in genes involved in sister chromatid cohesion and segregation. Nat Genet. 2013; 45:1459–1463.
crossref pmid pmc
27. Balbás-Martínez C, Sagrera A, Carrillo-de-Santa-Pau E, Earl J, Márquez M, Vazquez M, et al. Recurrent inactivation of STAG2 in bladder cancer is not associated with aneuploidy. Nat Genet. 2013; 45:1464–1469.
crossref pmid pmc
28. Taylor CF, Platt FM, Hurst CD, Thygesen HH, Knowles MA. Frequent inactivating mutations of STAG2 in bladder cancer are associated with low tumour grade and stage and inversely related to chromosomal copy number changes. Hum Mol Genet. 2014; 23:1964–1974.
crossref pmid
29. Lelo A, Prip F, Harris BT, Solomon D, Berry DL, Chaldekas K, et al. STAG2 is a biomarker for prediction of recurrence and progression in papillary non-muscle-invasive bladder cancer. Clin Cancer Res. 2018; 24:4145–4153.
crossref pmid pmc
30. Carbone M, Yang H, Pass HI, Krausz T, Testa JR, Gaudino G. BAP1 and cancer. Nat Rev Cancer. 2013; 13:153–159.
crossref pmid pmc
31. Lin M, Zhang L, Hildebrandt MAT, Huang M, Wu X, Ye Y. Common, germline genetic variations in the novel tumor suppressor BAP1 and risk of developing different types of cancer. Oncotarget. 2017; 8:74936–74946.
crossref
32. Segovia C, Martínez-Fernández M, Dueñas M, Rubio C, López-Calderón FF, Costa C, et al. Opposing roles of PIK3CA gene alterations to EZH2 signaling in non-muscle invasive bladder cancer. Oncotarget. 2017; 8:10531–10542.
crossref pmid pmc
33. Dueñas M, Martínez-Fernández M, García-Escudero R, Villacampa F, Marqués M, Saiz-Ladera C, et al. PIK3CA gene alterations in bladder cancer are frequent and associate with reduced recurrence in non-muscle invasive tumors. Mol Carcinog. 2015; 54:566–576.
crossref pmid
34. Beukers W, van der Keur KA, Kandimalla R, Vergouwe Y, Steyerberg EW, Boormans JL, et al. FGFR3, TERT and OTX1 as a urinary biomarker combination for surveillance of patients with bladder cancer in a large prospective multicenter study. J Urol. 2017; 197:1410–1418.
pmid
35. Holyoake A, O'Sullivan P, Pollock R, Best T, Watanabe J, Kajita Y, et al. Development of a multiplex RNA urine test for the detection and stratification of transitional cell carcinoma of the bladder. Clin Cancer Res. 2008; 14:742–749.
crossref pmid
36. Park HS, Park WS, Bondaruk J, Tanaka N, Katayama H, Lee S, et al. Quantitation of Aurora kinase A gene copy number in urine sediments and bladder cancer detection. J Natl Cancer Inst. 2008; 100:1401–1411.
crossref pmid pmc
37. Urquidi V, Goodison S, Cai Y, Sun Y, Rosser CJ. A candidate molecular biomarker panel for the detection of bladder cancer. Cancer Epidemiol Biomarkers Prev. 2012; 21:2149–2158.
crossref
38. Bongiovanni L, Pirozzi F, Guidi F, Orsini M, Chiurazzi P, Bassi PF, et al. Bradeion (SEPT4) as a urinary marker of transitional cell bladder cancer: a real-time polymerase chain reaction study of gene expression. J Urol. 2012; 187:2223–2227.
pmid
39. Friedrich MG, Toma MI, Hellstern A, Pantel K, Weisenberger DJ, Noldus J, et al. Comparison of multitarget fluorescence in situ hybridization in urine with other noninvasive tests for detecting bladder cancer. BJU Int. 2003; 92:911–914.
crossref pmid
40. Sarosdy MF, Schellhammer P, Bokinsky G, Kahn P, Chao R, Yore L, et al. Clinical evaluation of a multi-target fluorescent in situ hybridization assay for detection of bladder cancer. J Urol. 2002; 168:1950–1954.
crossref pmid
41. Bollmann M, Heller H, Bánkfalvi A, Griefingholt H, Bollmann R. Quantitative molecular urinary cytology by fluorescence in situ hybridization: a tool for tailoring surveillance of patients with superficial bladder cancer? BJU Int. 2005; 95:1219–1225.
crossref
42. Kipp BR, Karnes RJ, Brankley SM, Harwood AR, Pankratz VS, Sebo TJ, et al. Monitoring intravesical therapy for superficial bladder cancer using fluorescence in situ hybridization. J Urol. 2005; 173:401–404.
crossref pmid
43. Hanke M, Hoefig K, Merz H, Feller AC, Kausch I, Jocham D, et al. A robust methodology to study urine microRNA as tumor marker: microRNA-126 and microRNA-182 are related to urinary bladder cancer. Urol Oncol. 2010; 28:655–661.
crossref pmid
44. Puerta-Gil P, García-Baquero R, Jia AY, Ocaña S, Alvarez-Múgica M, Alvarez-Ossorio JL, et al. miR-143, miR-222, and miR-452 are useful as tumor stratification and noninvasive diagnostic biomarkers for bladder cancer. Am J Pathol. 2012; 180:1808–1815.
crossref pmid
45. Yamada Y, Enokida H, Kojima S, Kawakami K, Chiyomaru T, Tatarano S, et al. MiR-96 and miR-183 detection in urine serve as potential tumor markers of urothelial carcinoma: correlation with stage and grade, and comparison with urinary cytology. Cancer Sci. 2011; 102:522–529.
crossref pmid
46. Wang G, Chan ES, Kwan BC, Li PK, Yip SK, Szeto CC, et al. Expression of microRNAs in the urine of patients with bladder cancer. Clin Genitourin Cancer. 2012; 10:106–113.
crossref pmid
47. Eissa S, Safwat M, Matboli M, Zaghloul A, El-Sawalhi M, Shaheen A. Measurement of urinary level of a specific competing endogenous RNA network (FOS and RCAN mRNA/miR-324-5p, miR-4738-3p, /lncRNA miR-497-HG) enables diagnosis of bladder cancer. Urol Oncol. 2019; 37:292.e19–292.e27.
48. Chen X. MiR-101 acts as a novel bio-marker in the diagnosis of bladder carcinoma. Medicine (Baltimore). 2019; 98:e16051.
crossref
49. Soloway MS, Briggman V, Carpinito GA, Chodak GW, Church PA, Lamm DL, et al. Use of a new tumor marker, urinary NMP22, in the detection of occult or rapidly recurring transitional cell carcinoma of the urinary tract following surgical treatment. J Urol. 1996; 156(2 Pt 1):363–367.
crossref pmid
50. Miyake M, Goodison S, Giacoia EG, Rizwani W, Ross S, Rosser CJ. Influencing factors on the NMP-22 urine assay: an experimental model. BMC Urol. 2012; 12:23.
crossref
51. Wang Z, Que H, Suo C, Han Z, Tao J, Huang Z, et al. Evaluation of the NMP22 BladderChek test for detecting bladder cancer: a systematic review and meta-analysis. Oncotarget. 2017; 8:100648–100656.
crossref
52. Guo A, Wang X, Gao L, Shi J, Sun C, Wan Z. Bladder tumour antigen (BTA stat) test compared to the urine cytology in the diagnosis of bladder cancer: a meta-analysis. Can Urol Assoc J. 2014; 8:E347–E352.
crossref
53. Glas AS, Roos D, Deutekom M, Zwinderman AH, Bossuyt PM, Kurth KH. Tumor markers in the diagnosis of primary bladder cancer. a systematic review. J Urol. 2003; 169:1975–1982.
crossref pmid
54. Chakraborty A, Dasari S, Long W, Mohan C. Urine protein biomarkers for the detection, surveillance, and treatment response prediction of bladder cancer. Am J Cancer Res. 2019; 9:1104–1117.
pmid pmc
55. Li H, Li C, Wu H, Zhang T, Wang J, Wang S, et al. Identification of Apo-A1 as a biomarker for early diagnosis of bladder transitional cell carcinoma. Proteome Sci. 2011; 9:21.
crossref
56. Li C, Li H, Zhang T, Li J, Liu L, Chang J. Discovery of Apo-A1 as a potential bladder cancer biomarker by urine proteomics and analysis. Biochem Biophys Res Commun. 2014; 446:1047–1052.
crossref pmid
57. Chen YT, Chen CL, Chen HW, Chung T, Wu CC, Chen CD, et al. Discovery of novel bladder cancer biomarkers by comparative urine proteomics using iTRAQ technology. J Proteome Res. 2010; 9:5803–5815.
crossref pmid
58. Cai Q, Wu Y, Guo Z, Gong R, Tang Y, Yang K, et al. Urine BLCA-4 exerts potential role in detecting patients with bladder cancers: a pooled analysis of individual studies. Oncotarget. 2015; 6:37500–37510.
crossref
59. Eissa S, Matboli M, Essawy NO, Kotb YM. Integrative functional genetic-epigenetic approach for selecting genes as urine biomarkers for bladder cancer diagnosis. Tumour Biol. 2015; 36:9545–9552.
crossref pmid
60. Pham HT, Block NL, Lokeshwar VB. Tumor-derived hyaluronidase: a diagnostic urine marker for high-grade bladder cancer. Cancer Res. 1997; 57:778–783.
crossref pmid
61. Gourin CG, Zhi W, Adam BL. Proteomic identification of serum biomarkers for head and neck cancer surveillance. Laryngoscope. 2009; 119:1291–1302.
crossref pmid
62. Yang HH, Chen XF, Hu W, Lv DQ, Ding WJ, Tang LJ, et al. Lipoprotein(a) level and its association with tumor stage in male patients with primary lung cancer. Clin Chem Lab Med. 2009; 47:452–457.
crossref pmid
63. Goodison S, Chang M, Dai Y, Urquidi V, Rosser CJ. A multianalyte assay for the non-invasive detection of bladder cancer. PLoS One. 2012; 7:e47469.
crossref
64. Frantzi M, Vlahou A. Ten years of proteomics in bladder cancer: progress and future directions. Bladder Cancer. 2017; 3:1–18.
crossref pmid pmc
65. Urquidi V, Kim J, Chang M, Dai Y, Rosser CJ, Goodison S. CCL18 in a multiplex urine-based assay for the detection of bladder cancer. PLoS One. 2012; 7:e37797.
crossref
66. Magalhaes TF, Baracat EC, Doumouchtsis SK, Haddad JM. Biomarkers in the diagnosis and symptom assessment of patients with bladder pain syndrome: a systematic review. Int Urogynecol J. 2019; 08. 13. DOI: 10.1007/s00192-019-04075-9. [Epub].
crossref
67. Tonyali S, Ates D, Akbiyik F, Kankaya D, Baydar D, Ergen A. Urine nerve growth factor (NGF) level, bladder nerve staining and symptom/problem scores in patients with interstitial cystitis. Adv Clin Exp Med. 2018; 27:159–163.
crossref pmid
68. Corcoran AT, Yoshimura N, Tyagi V, Jacobs B, Leng W, Tyagi P. Mapping the cytokine profile of painful bladder syndrome/interstitial cystitis in human bladder and urine specimens. World J Urol. 2013; 31:241–246.
crossref pmid
69. Vera PL, Preston DM, Moldwin RM, Erickson DR, Mowlazadeh B, Ma F, et al. Elevated urine levels of macrophage migration inhibitory factor in inflammatory bladder conditions: a potential biomarker for a subgroup of interstitial cystitis/bladder pain syndrome patients. Urology. 2018; 116:55–62.
crossref pmid pmc
70. Lamale LM, Lutgendorf SK, Zimmerman MB, Kreder KJ. Interleukin-6, histamine, and methylhistamine as diagnostic markers for interstitial cystitis. Urology. 2006; 68:702–706.
crossref pmid
71. Keay S, Zhang CO, Chai T, Warren J, Koch K, Grkovic D, et al. Antiproliferative factor, heparin-binding epidermal growth factor-like growth factor, and epidermal growth factor in men with interstitial cystitis versus chronic pelvic pain syndrome. Urology. 2004; 63:22–26.
crossref pmid
72. Byrne DS, Sedor JF, Estojak J, Fitzpatrick KJ, Chiura AN, Mulholland SG. The urinary glycoprotein GP51 as a clinical marker for interstitial cystitis. J Urol. 1999; 161:1786–1790.
pmid
73. Pasikanti KK, Esuvaranathan K, Hong Y, Ho PC, Mahendran R, Raman Nee Mani L, et al. Urinary metabotyping of bladder cancer using two-dimensional gas chromatography time-of-flight mass spectrometry. J Proteome Res. 2013; 12:3865–3873.
crossref pmid
74. Pasikanti KK, Esuvaranathan K, Ho PC, Mahendran R, Kamaraj R, Wu QH, et al. Noninvasive urinary metabonomic diagnosis of human bladder cancer. J Proteome Res. 2010; 9:2988–2995.
crossref pmid
75. Wittmann BM, Stirdivant SM, Mitchell MW, Wulff JE, Mc-Dunn JE, Li Z, et al. Bladder cancer biomarker discovery using global metabolomic profiling of urine. PLoS One. 2014; 9:e115870.
crossref
76. Srivastava S, Roy R, Singh S, Kumar P, Dalela D, Sankhwar SN, et al. Taurine-a possible fingerprint biomarker in non-muscle invasive bladder cancer: a pilot study by 1H NMR spectroscopy. Cancer Biomark. 2010; 6:11–20.
pmid
77. Huang Z, Lin L, Gao Y, Chen Y, Yan X, Xing J, et al. Bladder cancer determination via two urinary metabolites: a biomarker pattern approach. Mol Cell Proteomics. 2011; 10:M111.007922.
crossref
78. Jin X, Yun SJ, Jeong P, Kim IY, Kim WJ, Park S. Diagnosis of bladder cancer and prediction of survival by urinary metabolomics. Oncotarget. 2014; 5:1635–1645.
crossref pmid pmc
79. Liu X, Cheng X, Liu X, He L, Zhang W, Wang Y, et al. Investigation of the urinary metabolic variations and the application in bladder cancer biomarker discovery. Int J Cancer. 2018; 143:408–418.
crossref pmid
80. Parker KS, Crowley JR, Stephens-Shields AJ, van Bokhoven A, Lucia MS, Lai HH, et al. Urinary metabolomics identifies a molecular correlate of interstitial cystitis/bladder pain syndrome in a Multidisciplinary Approach to the Study of Chronic Pelvic Pain (MAPP) research network cohort. EBioMedicine. 2016; 7:167–174.
crossref pmid pmc
81. Esteller M. Epigenetics in cancer. N Engl J Med. 2008; 358:1148–1159.
crossref pmid
82. Kim WJ, Kim YJ. Epigenetic biomarkers in urothelial bladder cancer. Expert Rev Mol Diagn. 2009; 9:259–269.
crossref pmid
83. Besaratinia A, Cockburn M, Tommasi S. Alterations of DNA methylome in human bladder cancer. Epigenetics. 2013; 8:1013–1022.
crossref pmid pmc
84. Kandimalla R, van Tilborg AA, Zwarthoff EC. DNA methylation-based biomarkers in bladder cancer. Nat Rev Urol. 2013; 10:327–335.
crossref pmid
85. Chan MW, Chan LW, Tang NL, Tong JH, Lo KW, Lee TL, et al. Hypermethylation of multiple genes in tumor tissues and voided urine in urinary bladder cancer patients. Clin Cancer Res. 2002; 8:464–470.
pmid
86. Friedrich MG, Weisenberger DJ, Cheng JC, Chandrasoma S, Siegmund KD, Gonzalgo ML, et al. Detection of methylated apoptosis-associated genes in urine sediments of bladder cancer patients. Clin Cancer Res. 2004; 10:7457–7465.
crossref pmid
87. Hoque MO, Begum S, Topaloglu O, Chatterjee A, Rosenbaum E, Van Criekinge W, et al. Quantitation of promoter methylation of multiple genes in urine DNA and bladder cancer detection. J Natl Cancer Inst. 2006; 98:996–1004.
crossref pmid
88. Renard I, Joniau S, van Cleynenbreugel B, Collette C, Naômé C, Vlassenbroeck I, et al. Identification and validation of the methylated TWIST1 and NID2 genes through real-time methylation-specific polymerase chain reaction assays for the noninvasive detection of primary bladder cancer in urine samples. Eur Urol. 2010; 58:96–104.
crossref pmid
89. Abern MR, Owusu R, Inman BA. Clinical performance and utility of a DNA methylation urine test for bladder cancer. Urol Oncol. 2014; 32:51.e21–51.e26.
crossref
90. Fantony JJ, Abern MR, Gopalakrishna A, Owusu R, Jack Tay K, Lance RS, et al. Multi-institutional external validation of urinary TWIST1 and NID2 methylation as a diagnostic test for bladder cancer. Urol Oncol. 2015; 33:387.e1–387.e6.
crossref
91. Reinert T, Modin C, Castano FM, Lamy P, Wojdacz TK, Hansen LL, et al. Comprehensive genome methylation analysis in bladder cancer: identification and validation of novel methylated genes and application of these as urinary tumor markers. Clin Cancer Res. 2011; 17:5582–5592.
crossref pmid
92. Reinert T, Borre M, Christiansen A, Hermann GG, Ørntoft TF, Dyrskjøt L. Diagnosis of bladder cancer recurrence based on urinary levels of EOMES, HOXA9, POU4F2, TWIST1, VIM, and ZNF154 hypermethylation. PLoS One. 2012; 7:e46297.
crossref
93. Su SF, de Castro Abreu AL, Chihara Y, Tsai Y, Andreu-Vieyra C, Daneshmand S, et al. A panel of three markers hyper- and hypomethylated in urine sediments accurately predicts bladder cancer recurrence. Clin Cancer Res. 2014; 20:1978–1989.
crossref pmid
94. Yu J, Zhu T, Wang Z, Zhang H, Qian Z, Xu H, et al. A novel set of DNA methylation markers in urine sediments for sensitive/specific detection of bladder cancer. Clin Cancer Res. 2007; 13:7296–7304.
crossref pmid
95. Costa VL, Henrique R, Danielsen SA, Duarte-Pereira S, Eknaes M, Skotheim RI, et al. Three epigenetic biomarkers, GDF15, TMEFF2, and VIM, accurately predict bladder cancer from DNA-based analyses of urine samples. Clin Cancer Res. 2010; 16:5842–5851.
crossref pmid
96. van der Heijden AG, Mengual L, Ingelmo-Torres M, Lozano JJ, van Rijt-van de Westerlo CCM, Baixauli M, et al. Urine cell-based DNA methylation classifier for monitoring bladder cancer. Clin Epigenetics. 2018; 10:71.
crossref
97. Bradley MS, Burke EE, Grenier C, Amundsen CL, Murphy SK, Siddiqui NY. A genome-scale DNA methylation study in women with interstitial cystitis/bladder pain syndrome. Neurourol Urodyn. 2018; 37:1485–1493.
crossref pmid pmc
98. Kim YJ, Kim WJ. Can we use methylation markers as diagnostic and prognostic indicators for bladder cancer? Investig Clin Urol. 2016; 57:Suppl 1. S77–S88.
crossref
99. Liou LS. Urothelial cancer biomarkers for detection and surveillance. Urology. 2006; 67:3 Suppl 1. 25–33. discussion 33-4.
crossref
TOOLS
ORCID iDs

Jayoung Kim
https://orcid.org/0000-0002-3683-4627

Won Tae Kim
https://orcid.org/0000-0002-7482-4550

Wun-Jae Kim
https://orcid.org/0000-0002-8060-8926

Similar articles