Next Article in Journal
Detection and Correlation of Single and Concomitant TP53, PTEN, and CDKN2A Alterations in Gliomas
Next Article in Special Issue
Epigenetics of Urological Cancers
Previous Article in Journal
Innovative MRI Techniques in Neuroimaging Approaches for Cerebrovascular Diseases and Vascular Cognitive Impairment
Previous Article in Special Issue
Characterization of Histone Deacetylase Expression Within In Vitro and In Vivo Bladder Cancer Model Systems
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

DNA-Methylation-Based Detection of Urological Cancer in Urine: Overview of Biomarkers and Considerations on Biomarker Design, Source of DNA, and Detection Technologies

by
Louise Katrine Larsen
1,
Guro Elisabeth Lind
2,
Per Guldberg
1 and
Christina Dahl
1,*
1
Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
2
Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, the Norwegian Radium Hospital, NO-0424 Oslo, Norway
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(11), 2657; https://doi.org/10.3390/ijms20112657
Submission received: 29 April 2019 / Revised: 23 May 2019 / Accepted: 24 May 2019 / Published: 30 May 2019
(This article belongs to the Special Issue Epigenetics of Urological Cancers)

Abstract

:
Changes in DNA methylation have been causally linked with cancer and provide promising biomarkers for detection in biological fluids such as blood, urine, and saliva. The field has been fueled by genome-wide characterization of DNA methylation across cancer types as well as new technologies for sensitive detection of aberrantly methylated DNA molecules. For urological cancers, urine is in many situations the preferred “liquid biopsy” source because it contains exfoliated tumor cells and cell-free tumor DNA and can be obtained easily, noninvasively, and repeatedly. Here, we review recent advances made in the development of DNA-methylation-based biomarkers for detection of bladder, prostate, renal, and upper urinary tract cancers, with an emphasis on the performance characteristics of biomarkers in urine. For most biomarkers evaluated in independent studies, there was great variability in sensitivity and specificity. We discuss issues that impact the outcome of DNA-methylation-based detection of urological cancer and account for the great variability in performance, including genomic location of biomarkers, source of DNA, and technical issues related to the detection of rare aberrantly methylated DNA molecules. Finally, we discuss issues that remain to be addressed to fully exploit the potential of DNA-methylation-based biomarkers in the clinic, including the need for prospective trials and careful selection of control groups.

1. Introduction

Urological cancers encompass a clinically and molecularly heterogeneous group of neoplasms affecting any region of the urological system. Cancer of the bladder, kidneys, upper urinary tract (ureter and renal pelvis), and prostate are all relatively common and pose specific requirements for diagnosis and follow-up. While kidney and upper urinary tract cancers are detected using imaging techniques, standard work-up for bladder and prostate cancer involves semi-invasive procedures (i.e., cystoscopy and digital rectal examination (DRE), respectively). Cystoscopy is extensively used in clinical practice and poses a significant burden on the healthcare system because it is used as a first-line rule-out test for cancer in patients with relevant symptoms, primarily, hematuria. Other downsides include patient discomfort and anxiety, risk of infectious complications, and high rates of false-positive and false-negative results. Subsequent histopathological assessment of biopsy tissue and surgical resection specimens is the gold standard for cancer diagnosis but also has its limitations, including the subjective evaluation by a pathologist, the need for tissue that is of a certain quality and representative of the tumor, and constraints on sampling frequency. Given these challenges, there is a major unmet need to develop noninvasive methods that could provide clinicians with rapid, objective, and accurate routines for detection of urological cancers.
An important development in cancer care is “liquid biopsy”, which involves the analysis of genetic material or tumor cells shed from primary or metastatic tumors into bodily fluids. A rapidly increasing number of studies have demonstrated the potential of liquid biopsies for a wide range of clinical applications, such as initial diagnosis, early detection, and disease monitoring after therapy [1]. Most of this work involves the analysis of cell-free DNA (cfDNA) circulating in the blood; however, for urological cancers, a more convenient liquid biopsy source is voided urine, which is easily and repeatably accessible and contains exfoliated cells and cfDNA from different sites of the urinary system [2]. The advent of high-sensitivity PCR-based technologies has enabled reliable detection of cancer-specific alterations in urine DNA. Because tumor-derived DNA in urine is often present in a large background of DNA derived from normal cells, the most useful DNA biomarkers are those that provide high specificity for malignancy or premalignancy, including mutations, translocations, gene fusions, and aberrant hypermethylation of specific CpG sites. Among these, DNA hypermethylation events represent the most versatile biomarker type because they are common in most cancers and can be easily assessed using well-established techniques. Furthermore, DNA methylation changes are considered early events in tumorigenesis and thus provide potential biomarkers for early diagnosis [3].
Urine-based DNA tests for urological cancer can be divided into two categories depending on the a priori availability of information on the patient’s tumor DNA. For detection of recurrence and evaluation of treatment response, DNA from the original tumor can be analyzed to identify specific alterations that may serve as “personalized” biomarkers. For other applications, such as initial examination of patients with symptoms of urological cancer, the genetic and epigenetic makeup of the possible tumor is unknown. In these situations, there is a need for a “universal” or “generic” test that can detect, in principle, any cancer. Because no genetic or epigenetic alteration is present in all cases of a urological cancer type, it is necessary to use a combination of biomarkers. The initial assembly of a biomarker panel is facilitated by information about the performance characteristics of individual biomarkers in terms of sensitivity (the true positive rate), specificity (the true negative rate), and predictive values [4]. If the test is used in individuals with unknown disease status to reduce the use of invasive procedures, the most important performance characteristics are sensitivity and negative predictive value (NPV) to achieve the lowest possible rate of false-negative results. As discussed below, specificity is a less well-defined characteristic that varies based on a number of factors, including control populations and the definition of a false-positive result.
Although the promises of DNA-methylation-based detection of cancer have been recognized since the early 2000s [5], including the potential for detection and management of urological cancers [6], only a few DNA methylation biomarkers have been implemented into routine clinical practice. Navigating towards clinical utility is challenging, requiring optimal study designs (representative and large patient series) as well as robustly designed biomarker assays. Here, we provide an overview of current DNA-methylation-based biomarkers for urological cancer, with an emphasis on their performance characteristics in urine. We discuss potential causes of performance variability across studies and other challenges that must be overcome before clinically useful tests can be developed and implemented. Some of these issues have been discussed in detail elsewhere [7,8,9] and are only reviewed briefly here.

2. Performance Characteristics of DNA Methylation Biomarkers

To provide an overview of current DNA-methylation-based biomarkers for urine-based detection of urological cancer, we undertook systematic literature searches in PubMed and Embase until February 2019. Details on search strategy, criteria for selection of relevant studies, and data extraction are provided in Supplementary Methods.

2.1. Bladder Cancer

A total of 57 studies met the inclusion criteria (Supplementary Table S1). Fifty-two studies analyzed urine from bladder cancer patients at first diagnosis, using a total of 114 different DNA methylation biomarkers. The sensitivities for 23 biomarkers investigated in more than three studies are shown in Figure 1. Two of these biomarkers each reached a median sensitivity of >80% (ZNF154 and POU4F2) with, however, large variability across studies. The specificities for these biomarkers are shown in Supplementary Figure S1, also demonstrating large interstudy variability. Fifty-two different biomarker combinations, comprising between 2 and 12 individual biomarkers, have been tested for initial diagnosis of bladder cancer. Twenty of these combinations achieved a sensitivity of ≥90% (listed in Table 1).
Eleven studies investigated recurrent bladder cancer, using 18 individual biomarkers and 8 biomarker panels (Table 2). ZNF154 and EOMES achieved the highest sensitivity (94%), however with a specificity of <70%. The biomarker combination with the highest sensitivity (CFTR, SALL3, and TWIST1; 90%) had a very low specificity (31%). Only one individual biomarker (TWIST1) and two panels have been evaluated in more than one study.

2.2. Prostate Cancer

Twenty-seven studies met the inclusion criteria (Supplementary Table S2). Of these, 26 studies contained data on single biomarker performance (48 different biomarkers), with 9 biomarkers tested in 2 or more studies (shown in Figure 2). GSTP1 was the most extensively studied biomarker (19 studies). The highest median sensitivity was reported for HOXD3 (76%); most other biomarkers had sensitivities of <50%. The corresponding specificities for these biomarkers are shown in Supplementary Figure S2. Fifteen studies combined two or more biomarkers, with five studies achieving a sensitivity of ≥90% (Table 3).

2.3. Renal Cancer

Five studies met the inclusion criteria (Table 4). Data on single biomarkers were available from four studies for 15 different biomarkers, with sensitivities between 5% and 79% and a generally high specificity (89–100%). TCF21 was the only biomarker tested in more than one study. Both studies achieved 100% specificity, but the sensitivity varied from 28% to 79%. Three studies evaluated biomarker combinations, with the best performing combination (VHL, p16, ARF, APC, RASSF1A, and TIMP3) achieving a sensitivity of 88% and a specificity of 100%. A similar sensitivity was reported by combining nine biomarkers (APC, ARF, CDH1, GSTP1, MGMT, p16, RARB2, RASSF1A, and TIMP3), with no indication of specificity.

2.4. Upper Urinary Tract Cancer

Only two studies met the inclusion criteria, investigating a total of 10 biomarkers (Table 5). VIM was the only biomarker investigated in both studies, achieving a sensitivity of 82% and 73% and a specificity of 100% and 61%. HSPA2 achieved a sensitivity of 83% but with a specificity of only 36%. Among seven biomarker combinations tested, a combination of VIM and GDF15 reached a sensitivity of 91% and a specificity of 100% in a study with 22 cases and 20 healthy controls, whereas in a study with 98 cases and 113 controls with benign urologic conditions, these two biomarkers in combination with CDH1, RASSF1A, and HSPA2 only reached a sensitivity of 82% and a specificity of 65%.

3. Factors Affecting Biomarker Performance

The overall conclusion from our review of DNA-methylation-based biomarkers for detection of urological cancer is that there is great variability in sensitivity and specificity across studies. Below, we discuss clinical and technical factors, which can explain this variability and should be considered when designing studies that eventually should lead to the implementation of urine-based tests in the clinic.

3.1. Urine Collection and Processing

Urine is a complex biological fluid that contains inorganic salts, organic compounds, and multiple cell types, including leukocytes, urothelial cells, renal cells, and prostate cells. Tumor-derived DNA can be present in both the cellular and cell-free fractions of urine, and the procedures used for collection and processing of DNA will greatly impact the outcome of biomarker analysis. Several sources of DNA have been utilized, including (i) whole urine (containing cellular DNA and cfDNA), (ii) urine sediment obtained by centrifugation (containing cellular DNA), (iii) urine supernatant (containing cfDNA), and (iv) cells obtained by immune capture [44] or filtration [14,45,46]. Because cells and DNA in urine are susceptible to degradation upon storage depending on time and temperature, correct storage is important when urine samples are not processed immediately. One study found that DNA in urine stored at room temperature was stable only upon addition of preserving agents but also found that DNA remained stable without the addition of preservatives for up to 28 days when stored at −20 °C or −80 °C [47]. A confounding factor in many studies is that samples containing DNA of insufficient quantity or quality were excluded from analysis. Such sample selection bias may lead to an overestimation of performance characteristics.
The vast majority of studies included in this review utilized sedimented urine as the source of DNA. The procedure for collection of urine sediments is simple and inexpensive but has several limitations in addition to storage challenges and processing time, including the co-sedimentation of normal cells and the presence of crystals and substances that may inhibit downstream PCR analyses [48]. A recent study showed that the sensitivity for detection of bladder cancer using TERT promoter mutations as a biomarker was higher in sedimented samples compared with cfDNA [2]. However, in leukocyte-rich urine, the sensitivity was higher in cfDNA using next-generation sequencing (NGS), probably because of a higher ratio of tumor-to-wildtype DNA compared with urine sediments. An alternative approach to enriching for tumor DNA is size-based cell selection, utilizing a filter with a pore size that enables capture of tumor cells with the passage of smaller-sized normal cells, at the same time removing inhibitory substances. One study comparing sedimentation and filtration of urine samples from patients with bladder cancer showed a higher sensitivity for filtration, particularly for low-grade tumors [46].
Another factor that should be considered when designing urine-based assays for urological cancer is that the concentrations of cells and DNA in urine are not constant. Shedding of cells and release of DNA through apoptosis or necrosis are stochastic and depend on several factors, including the size and location of the tumor. In prostate cancer, higher sensitivities have been achieved after physical manipulation of the prostate, such as massage and DRE. Another approach to increase the sensitivity is repeated urine sampling. In a study of men with high-grade prostate cancer, analysis of urine cells collected by filtration on different days without prior DRE showed a great interday variation in the presence of DNA methylation biomarkers, with some samples giving a false-negative result [49]. A study of patients with small low-grade bladder tumors showed that analysis of pooled urine samples collected over 24 h resulted in a sensitivity of 100%, whereas it was only 75% when a single urine sample was analyzed [50].

3.2. Bisulfite Treatment, Detection Technologies, and Sample Scoring

With few exceptions, the studies included in this review used treatment of DNA with sodium bisulfite to selectively convert unmethylated cytosines to uracil (leaving methylated cytosines as cytosine). These methylation-dependent C-to-U changes can subsequently be analyzed using PCR-based technologies to ascertain the methylation status [50]. Although the basic protocol for bisulfite conversion is simple and well described, it has a number of limitations that can introduce biases. Treatment of DNA with bisulfite introduces DNA strand breaks and results in highly fragmented single-stranded DNA, leading to degradation of up to 90% of the input DNA [51] and severely reducing the number of molecules effectively available for PCR amplification. The loss of DNA may be further aggravated by incomplete DNA recovery after bisulfite conversion. Recovery depends on the length of input DNA, with higher recovery for high-molecular-weight DNA. In urine, cfDNA has a large component of mononucleosomal (178 bp) DNA, posing specific requirements for extraction procedures [52]. Another limitation inherent to bisulfite treatment is incomplete conversion, which may lead to false-positive results because unconverted unmethylated CpG sites are falsely interpreted as methylated. A comparison of 12 commercially available bisulfite conversion kits showed a large variability in recovery and conversion efficiency [53]. Furthermore, several factors have been shown to affect the technical variability of PCR-based analysis of bisulfite-treated DNA, including the amount of bisulfite-converted template in the PCR, the amount of DNA input in the bisulfite conversion, and storage (bisulfite-converted DNA is less stable than genomic DNA) [54].
A wide range of PCR methods have been used for downstream biomarker evaluation. The most frequently used methods are methylation-specific PCR (MSP) and quantitative MSP (qMSP). Conventional MSP [55] was the method of choice in earlier studies but has also been used in more recent studies. This method is easy to perform and requires no specialized equipment but has several limitations, including the qualitative readout. The most frequently used method in more recent studies is qMSP based on the MethyLight technology [56], which provides a semiquantitative readout. Other methods include pyrosequencing [55], methylation-sensitive single nucleotide primer extension (MS-SnuPE) [57], methylation-sensitive high-resolution melting (MS-HRM) [58], and methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA). [59] A systematic evaluation and comparison of assays for measuring DNA methylation at specific CpG sites was recently conducted by the BLUEPRINT Consortium [60]. Most methods performed well in distinguishing methylated from unmethylated DNA but all had limitations in detecting low-abundant molecules. It is likely that the field will be markedly advanced with the introduction of newer technologies such as NGS and digital PCR, which enable DNA quantification with superior sensitivity and accuracy.
A general limitation in most studies reviewed here was the lack of information about assay performance in terms of limit of blank (LoB), limit of detection (LoD), and limit of quantitation (LoQ), which are critical parameters describing the smallest concentration of a biomarker that can be reliably measured [61]. In most cases, there were no predefined thresholds for interpreting assay signals, and several studies did not indicate the number of positive biomarkers required for scoring a sample positive.

3.3. Genomic Location of Biomarker Assays

The most commonly used strategy to identify and develop new DNA methylation biomarkers is targeting functionally relevant locations, such as CpG islands where methylation affects gene expression. The three biomarkers TWIST1, OTX1, and ONECUT2 included in the commercial AssureMDx test, evaluating the risk for bladder cancer in patients with hematuria, are examples of this [11]. Whereas the biomarker for TWIST1 is located in the gene promoter and associated with loss of gene expression, the assays for OTX1 and ONECUT2 are located in regions associated with increased gene expression [8].
Detailed promoter methylation studies have demonstrated that some CpG sites may influence gene expression more than others. This was first shown in 2002, when Deng et al. reported that methylation of CpG sites in a proximal region of MHL1 was associated with lack of expression, whereas CpG sites in the distal part of the promoter tended to be methylated independently of MLH1 expression [62]. Designing biomarker assays close to the transcription start site generally increases the likelihood of hitting a location where the DNA methylation status will have a functional effect.
Independent of whether DNA methylation is functionally important or not, detailed knowledge of the methylation pattern of the individual CpG sites (e.g., through TCGA data) in a genomic region of interest is useful prior to biomarker assay design. Methylation density may vary considerably within a genomic region, potentially affecting the sensitivity and specificity of a biomarker assay. From a biomarker perspective, CpG sites that most robustly separate cases from controls and reach the highest sensitivity and specificity (independent of functional effect) would be highly attractive.

3.4. Sensitivity, Specificity, and Control Populations

Most DNA methylation biomarkers reviewed here were originally discovered by analysis of DNA from tumor biopsies, using adjacent tumor-free tissue or normal tissue as control. The sensitivity of a biomarker may here be defined as the proportion of tumors positive for this biomarker. However, a biomarker with high sensitivity in tumor tissue may not necessarily provide the same sensitivity in urine because this will depend on the shedding of tumor cells or cfDNA. Only a few studies have compared urine and tumor tissue from the same patients, suggesting that the sensitivity is generally lower in urine. As larger tumors will shed more material than smaller tumors, sensitivity is highly dependent on the cohort composition, with studies having a higher proportion of advanced cancers achieving higher sensitivity. Only a few studies have evaluated the sensitivity of biomarkers in large prospective studies enrolling patients consecutively and in an unbiased manner.
The specificity of a urinary DNA methylation biomarker is the probability of a negative test result in individuals without cancer. Based on the data compiled in this review, the specificity of DNA methylation biomarkers was relatively high in renal carcinoma (>90%) but generally lower in prostate cancer and recurrent bladder cancer. Although these figures may reflect a true difference in the ability of biomarkers to discriminate between cancer and no cancer, it is important to consider that specificity is affected by choice of control group. In the ideal situation, cases and controls should be age and sex matched. Notably, because epigenetic modifications (including DNA methylation) increase with age, the use of a non-age-matched control group could introduce significant bias. Another important factor is the clinical status of the control group. In many studies, including those on renal carcinoma, the control population consisted of healthy individuals. To evaluate the specificity of a test in a more realistic setting, the control population should consist of individuals with symptoms relevant for the specific cancer. Examples of such control groups include individuals with hematuria (in the case of bladder cancer) and increased prostate-specific antigen (PSA) levels (in the case of prostate cancer). One caveat here is that some patients with a positive urine test may have early cancers or precursor lesions that are molecularly detectable but still undetectable using current scanning or endoscopic procedures.
None of the biomarkers or biomarker panels for bladder cancer detected recurrence with a sensitivity or specificity of more than 90%, despite better performance of the same biomarkers for detection of primary bladder tumors [13]. The lower sensitivity may be explained by the fact that recurrent tumors are usually smaller than primary tumors and therefore are less prone to shed material into urine [13]. The lower specificity may at least in part be ascribed to challenges in the study design. While control groups for evaluating specificity at first diagnosis of bladder cancer are usually individuals with no prior history of bladder cancer, controls for recurrence are groups of patients showing negative follow-up cystoscopy. It is possible that these patients have residual DNA biomarkers in the urine due to incomplete tumor resection or the emergence of an as-yet undetectable recurrent tumor, thereby resulting in a lower specificity. Longitudinal studies where the patient is his/her own control may be more accurate, at least when it comes to the sensitivity.

4. Conclusions

Studies over more than a decade have demonstrated the great potential of DNA methylation biomarkers for urine-based detection of urological cancer. However, the bewildering number of biomarkers currently under evaluation and the great variability in biomarker performance across studies hamper successful translation into clinically useful tests. We have highlighted a number of factors, which directly impact the performance of urinary DNA methylation biomarkers, including technical issues related to the design and implementation of biomarker assays. Guidelines for these procedural issues should be clearly defined to ensure reproducibility and eventually facilitate the development of clinically useful urinary tests for urological cancer.

Supplementary Materials

Supplementary materials can be found at https://www.mdpi.com/1422-0067/20/11/2657/s1. References [63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111] are cited in the supplementary materials.

Author Contributions

Conceptualization, L.K.L, G.E.L., P.G., and C.D.; Methodology, L.K.L.; Formal Analysis, L.K.L. and C.D.; Writing–Original Draft Preparation, L.K.L., G.E.L., P.G., and C.D.; Writing—Review and Editing, L.K.L., G.E.L., P.G., and C.D.; Supervision, P.G.; Funding Acquisition, P.G.

Funding

This work was supported by a grant from the Danish Cancer Society.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Heitzer, E.; Haque, I.S.; Roberts, C.E.S.; Speicher, M.R. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat. Rev. Genet. 2019, 20, 71–88. [Google Scholar] [CrossRef]
  2. Stasik, S.; Salomo, K.; Heberling, U.; Froehner, M.; Sommer, U.; Baretton, G.B.; Ehninger, G.; Wirth, M.P.; Thiede, C.; Fuessel, S. Evaluation of tert promoter mutations in urinary cell-free DNA and sediment DNA for detection of bladder cancer. Clin. Biochem. 2019, 64, 60–63. [Google Scholar] [CrossRef]
  3. Baylin, S.B.; Jones, P.A. A decade of exploring the cancer epigenome—Biological and translational implications. Nat. Rev. Cancer. 2011, 11, 726–734. [Google Scholar] [CrossRef]
  4. Trevethan, R. Sensitivity, specificity, and predictive values: Foundations, pliabilities, and pitfalls in research and practice. Front Public Health 2017, 5, 307. [Google Scholar] [CrossRef] [PubMed]
  5. Laird, P.W. The power and the promise of DNA methylation markers. Nat. Rev. Cancer 2003, 3, 253–266. [Google Scholar] [CrossRef]
  6. Cairns, P. Gene methylation and early detection of genitourinary cancer: The road ahead. Nat. Rev. Cancer 2007, 7, 531–543. [Google Scholar] [CrossRef] [PubMed]
  7. Kandimalla, R.; van Tilborg, A.A.; Zwarthoff, E.C. DNA methylation-based biomarkers in bladder cancer. Nat. Rev. Urol. 2013, 10, 327–335. [Google Scholar] [CrossRef] [PubMed]
  8. Koch, A.; Joosten, S.C.; Feng, Z.; de Ruijter, T.C.; Draht, M.X.; Melotte, V.; Smits, K.M.; Veeck, J.; Herman, J.G.; Van Neste, L.; et al. Analysis of DNA methylation in cancer: Location revisited. Nat. Rev. Clin. Oncol. 2018, 15, 459–466. [Google Scholar] [CrossRef] [PubMed]
  9. Bosschieter, J.; Lutz, C.; Segerink, L.I.; Vis, A.N.; Zwarthoff, E.C.; van Moorselaar, R.J.A.; van Rhijn, B.W.; Heymans, M.W.; Jansma, E.P.; Steenbergen, R.D.; et al. The diagnostic accuracy of methylation markers in urine for the detection of bladder cancer: A systematic review. Epigenomics 2018, 10, 673–687. [Google Scholar] [CrossRef] [PubMed]
  10. Chihara, Y.; Kanai, Y.; Fujimoto, H.; Sugano, K.; Kawashima, K.; Liang, G.; Jones, P.A.; Fujimoto, K.; Kuniyasu, H.; Hirao, Y. Diagnostic markers of urothelial cancer based on DNA methylation analysis. BMC Cancer 2013, 13, 275. [Google Scholar] [CrossRef]
  11. Wang, Y.; Yu, Y.; Ye, R.; Zhang, D.; Li, Q.; An, D.; Fang, L.; Lin, Y.; Hou, Y.; Xu, A.; et al. An epigenetic biomarker combination of pcdh17 and pou4f2 detects bladder cancer accurately by methylation analyses of urine sediment DNA in han chinese. Oncotarget 2016, 7, 2754–2764. [Google Scholar] [CrossRef]
  12. Yeh, C.M.; Chen, P.C.; Hsieh, H.Y.; Jou, Y.C.; Lin, C.T.; Tsai, M.H.; Huang, W.Y.; Wang, Y.T.; Lin, R.I.; Chen, S.S.; et al. Methylomics analysis identifies znf671 as an epigenetically repressed novel tumor suppressor and a potential non-invasive biomarker for the detection of urothelial carcinoma. Oncotarget 2015, 6, 29555–29572. [Google Scholar] [CrossRef]
  13. Yegin, Z.; Gunes, S.; Buyukalpelli, R. Hypermethylation of twist1 and nid2 in tumor tissues and voided urine in urinary bladder cancer patients. DNA Cell Biol. 2013, 32, 386–392. [Google Scholar] [CrossRef]
  14. Andersson, E.; Dahmcke, C.M.; Steven, K.; Larsen, L.K.; Guldberg, P. Filtration device for on-site collection, storage and shipment of cells from urine and its application to DNA-based detection of bladder cancer. PLoS ONE 2015, 10, e0131889. [Google Scholar] [CrossRef]
  15. Renard, I.; Joniau, S.; van Cleynenbreugel, B.; Collette, C.; Naome, C.; Vlassenbroeck, I.; Nicolas, H.; de Leval, J.; Straub, J.; Van Criekinge, W.; 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. [Google Scholar] [CrossRef]
  16. Costa, V.L.; Henrique, R.; Danielsen, S.A.; Duarte-Pereira, S.; Eknaes, M.; Skotheim, R.I.; Rodrigues, A.; Magalhaes, J.S.; Oliveira, J.; Lothe, R.A.; 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. [Google Scholar] [CrossRef]
  17. Yu, J.; Zhu, T.; Wang, Z.; Zhang, H.; Qian, Z.; Xu, H.; Gao, B.; Wang, W.; Gu, L.; Meng, J.; 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. [Google Scholar] [CrossRef]
  18. Sun, J.; Chen, Z.; Zhu, T.; Yu, J.; Ma, K.; Zhang, H.; He, Y.; Luo, X.; Zhu, J. Hypermethylated sfrp1, but none of other nine genes “informative” for western countries, is valuable for bladder cancer detection in mainland china. J. Cancer Res. Clin. Oncol. 2009, 135, 1717–1727. [Google Scholar] [CrossRef]
  19. Pietrusinski, M.; Kepczynski, L.; Jedrzejczyk, A.; Borkowska, E.; Traczyk-Borszynska, M.; Constantinou, M.; Kauzewski, B.; Borowiec, M. Detection of bladder cancer in urine sediments by a hypermethylation panel of selected tumor suppressor genes. Cancer Biomark. 2017, 18, 47–59. [Google Scholar] [CrossRef]
  20. Roperch, J.P.; Grandchamp, B.; Desgrandchamps, F.; Mongiat-Artus, P.; Ravery, V.; Ouzaid, I.; Roupret, M.; Phe, V.; Ciofu, C.; Tubach, F.; et al. Promoter hypermethylation of hs3st2, septin9 and slit2 combined with fgfr3 mutations as a sensitive/specific urinary assay for diagnosis and surveillance in patients with low or high-risk non-muscle-invasive bladder cancer. BMC Cancer 2016, 16, 704. [Google Scholar] [CrossRef]
  21. Chan, M.W.; Chan, L.W.; Tang, N.L.; Tong, J.H.; Lo, K.W.; Lee, T.L.; Cheung, H.Y.; Wong, W.S.; Chan, P.S.; Lai, F.M.; et al. Hypermethylation of multiple genes in tumor tissues and voided urine in urinary bladder cancer patients. Clin. Cancer Res. 2002, 8, 464–470. [Google Scholar] [PubMed]
  22. Wu, Y.; Jiang, G.; Zhang, N.; Liu, S.; Lin, X.; Perschon, C.; Zheng, S.L.; Ding, Q.; Wang, X.; Na, R.; et al. Hoxa9, pcdh17, pou4f2, and onecut2 as a urinary biomarker combination for the detection of bladder cancer in chinese patients with hematuria. Eur. Urol. Focus 2018. [Google Scholar] [CrossRef] [PubMed]
  23. Dahmcke, C.M.; Steven, K.E.; Larsen, L.K.; Poulsen, A.L.; Abdul-Al, A.; Dahl, C.; Guldberg, P. A prospective blinded evaluation of urine-DNA testing for detection of urothelial bladder carcinoma in patients with gross hematuria. Eur. Urol. 2016, 70, 916–919. [Google Scholar] [CrossRef] [PubMed]
  24. Roupret, M.; Hupertan, V.; Yates, D.R.; Comperat, E.; Catto, J.W.; Meuth, M.; Lackmichi, A.; Ricci, S.; Lacave, R.; Gattegno, B.; et al. A comparison of the performance of microsatellite and methylation urine analysis for predicting the recurrence of urothelial cell carcinoma, and definition of a set of markers by bayesian network analysis. BJU Int. 2008, 101, 1448–1453. [Google Scholar] [CrossRef]
  25. Reinert, T.; Borre, M.; Christiansen, A.; Hermann, G.G.; Orntoft, T.F.; Dyrskjot, L. Diagnosis of bladder cancer recurrence based on urinary levels of eomes, hoxa9, pou4f2, twist1, vim, and znf154 hypermethylation. PLoS ONE 2012, 7, e46297. [Google Scholar] [CrossRef] [PubMed]
  26. Fernandez, C.A.; Millholland, J.M.; Zwarthoff, E.C.; Feldman, A.S.; Karnes, R.J.; Shuber, A.P. A noninvasive multi-analyte diagnostic assay: Combining protein and DNA markers to stratify bladder cancer patients. Res. Rep. Urol. 2012, 4, 17–26. [Google Scholar] [CrossRef]
  27. Zuiverloon, T.C.; Beukers, W.; van der Keur, K.A.; Munoz, J.R.; Bangma, C.H.; Lingsma, H.F.; Eijkemans, M.J.; Schouten, J.P.; Zwarthoff, E.C. A methylation assay for the detection of non-muscle-invasive bladder cancer (nmibc) recurrences in voided urine. BJU Int. 2012, 109, 941–948. [Google Scholar] [CrossRef]
  28. van der Heijden, A.G.; Mengual, L.; Ingelmo-Torres, M.; Lozano, J.J.; van Rijt-van de Westerlo, C.C.M.; Baixauli, M.; Geavlete, B.; Moldoveanud, C.; Ene, C.; Dinney, C.P.; et al. Urine cell-based DNA methylation classifier for monitoring bladder cancer. Clin. Epigenetics 2018, 10, 71. [Google Scholar] [CrossRef]
  29. Shindo, T.; Shimizu, T.; Nojima, M.; Niinuma, T.; Maruyama, R.; Kitajima, H.; Kai, M.; Itoh, N.; Suzuki, H.; Masumori, N. Evaluation of urinary DNA methylation as a marker for recurrent bladder cancer: A 2-center prospective study. Urology 2018, 113, 71–78. [Google Scholar] [CrossRef]
  30. Kandimalla, R.; Masius, R.; Beukers, W.; Bangma, C.H.; Orntoft, T.F.; Dyrskjot, L.; van Leeuwen, N.; Lingsma, H.; van Tilborg, A.A.; Zwarthoff, E.C. A 3-plex methylation assay combined with the fgfr3 mutation assay sensitively detects recurrent bladder cancer in voided urine. Clin. Cancer Res. 2013, 19, 4760–4769. [Google Scholar] [CrossRef]
  31. Zuiverloon, T.C.; Beukers, W.; van der Keur, K.A.; Nieuweboer, A.J.; Reinert, T.; Dyrskjot, L.; Orntoft, T.F.; Zwarthoff, E.C. Combinations of urinary biomarkers for surveillance of patients with incident nonmuscle invasive bladder cancer: The european fp7 uromol project. J. Urol. 2013, 189, 1945–1951. [Google Scholar] [CrossRef] [PubMed]
  32. Su, S.F.; de Castro Abreu, A.L.; Chihara, Y.; Tsai, Y.; Andreu-Vieyra, C.; Daneshmand, S.; Skinner, E.C.; Jones, P.A.; Siegmund, K.D.; Liang, G. A panel of three markers hyper- and hypomethylated in urine sediments accurately predicts bladder cancer recurrence. Clin. Cancer Res. 2014, 20, 1978–1989. [Google Scholar] [CrossRef] [PubMed]
  33. Moreira-Barbosa, C.; Barros-Silva, D.; Costa-Pinheiro, P.; Torres-Ferreira, J.; Constancio, V.; Freitas, R.; Oliveira, J.; Antunes, L.; Henrique, R.; Jeronimo, C. Comparing diagnostic and prognostic performance of two-gene promoter methylation panels in tissue biopsies and urines of prostate cancer patients. Clin. Epigenetics 2018, 10, 132. [Google Scholar] [CrossRef] [PubMed]
  34. Olkhov-Mitsel, E.; Zdravic, D.; Kron, K.; van der Kwast, T.; Fleshner, N.; Bapat, B. Novel multiplex methylight protocol for detection of DNA methylation in patient tissues and bodily fluids. Sci. Rep. 2014, 4, 4432. [Google Scholar] [CrossRef]
  35. Rogers, C.G.; Gonzalgo, M.L.; Yan, G.; Bastian, P.J.; Chan, D.Y.; Nelson, W.G.; Pavlovich, C.P. High concordance of gene methylation in post-digital rectal examination and post-biopsy urine samples for prostate cancer detection. J. Urol. 2006, 176, 2280–2284. [Google Scholar] [CrossRef] [PubMed]
  36. Brikun, I.; Nusskern, D.; Decatus, A.; Harvey, E.; Li, L.; Freije, D. A panel of DNA methylation markers for the detection of prostate cancer from fv and dre urine DNA. Clin. Epigenetics 2018, 10, 91. [Google Scholar] [CrossRef] [PubMed]
  37. Torres-Ferreira, J.; Ramalho-Carvalho, J.; Gomez, A.; Menezes, F.D.; Freitas, R.; Oliveira, J.; Antunes, L.; Bento, M.J.; Esteller, M.; Henrique, R.; et al. Mir-193b promoter methylation accurately detects prostate cancer in urine sediments and mir-34b/c or mir-129-2 promoter methylation define subsets of clinically aggressive tumors. Mol. Cancer 2017, 16, 26. [Google Scholar] [CrossRef] [PubMed]
  38. Hoque, M.O.; Begum, S.; Topaloglu, O.; Jeronimo, C.; Mambo, E.; Westra, W.H.; Califano, J.A.; Sidransky, D. Quantitative detection of promoter hypermethylation of multiple genes in the tumor, urine, and serum DNA of patients with renal cancer. Cancer Res. 2004, 64, 5511–5517. [Google Scholar] [CrossRef] [PubMed]
  39. Costa, V.L.; Henrique, R.; Danielsen, S.A.; Eknaes, M.; Patricio, P.; Morais, A.; Oliveira, J.; Lothe, R.A.; Teixeira, M.R.; Lind, G.E.; et al. Tcf21 and pcdh17 methylation: An innovative panel of biomarkers for a simultaneous detection of urological cancers. Epigenetics 2011, 6, 1120–1130. [Google Scholar] [CrossRef]
  40. Xin, J.; Xu, R.; Lin, S.; Xin, M.; Cai, W.; Zhou, J.; Fu, C.; Zhen, G.; Lai, J.; Li, Y.; et al. Clinical potential of tcf21 methylation in the diagnosis of renal cell carcinoma. Oncol. Lett. 2016, 12, 1265–1270. [Google Scholar] [CrossRef]
  41. Battagli, C.; Uzzo, R.G.; Dulaimi, E.; De Caceres, I.I.; Krassenstein, R.; Al-Saleem, T.; Greenberg, R.E.; Cairns, P. Promoter hypermethylation of tumor suppressor genes in urine from kidney cancer patients. Cancer Res. 2003, 63, 8695–8699. [Google Scholar] [PubMed]
  42. Dulaimi, E.; Uzzo, R.G.; Greenberg, R.E.; Al-Saleem, T.; Cairns, P. Detection of bladder cancer in urine by a tumor suppressor gene hypermethylation panel. Clin. Cancer Res. 2004, 10, 1887–1893. [Google Scholar] [CrossRef] [PubMed]
  43. Friedrich, M.G.; Weisenberger, D.J.; Cheng, J.C.; Chandrasoma, S.; Siegmund, K.D.; Gonzalgo, M.L.; Toma, M.I.; Huland, H.; Yoo, C.; Tsai, Y.C.; et al. Detection of methylated apoptosis-associated genes in urine sediments of bladder cancer patients. Clin. Cancer Res. 2004, 10, 7457–7465. [Google Scholar] [CrossRef] [PubMed]
  44. Macgregor-Ramiasa, M.; McNicholas, K.; Ostrikov, K.; Li, J.; Michael, M.; Gleadle, J.M.; Vasilev, K. A platform for selective immuno-capture of cancer cells from urine. Biosens. Bioelectron. 2017, 96, 373–380. [Google Scholar] [CrossRef] [PubMed]
  45. Birkhahn, M.; Mitra, A.P.; Williams, A.J.; Barr, N.J.; Skinner, E.C.; Stein, J.P.; Skinner, D.G.; Tai, Y.C.; Datar, R.H.; Cote, R.J. A novel precision-engineered microfiltration device for capture and characterisation of bladder cancer cells in urine. Eur. J. Cancer 2013, 49, 3159–3168. [Google Scholar] [CrossRef] [Green Version]
  46. Andersson, E.; Steven, K.; Guldberg, P. Size-based enrichment of exfoliated tumor cells in urine increases the sensitivity for DNA-based detection of bladder cancer. PLoS ONE 2014, 9, e94023. [Google Scholar] [CrossRef]
  47. Bosschieter, J.; Bach, S.; Bijnsdorp, I.V.; Segerink, L.I.; Rurup, W.F.; van Splunter, A.P.; Bahce, I.; Novianti, P.W.; Kazemier, G.; van Moorselaar, R.J.A.; et al. A protocol for urine collection and storage prior to DNA methylation analysis. PLoS ONE 2018, 13, e0200906. [Google Scholar] [CrossRef]
  48. Lin, S.Y.; Linehan, J.A.; Wilson, T.G.; Hoon, D.S.B. Emerging utility of urinary cell-free nucleic acid biomarkers for prostate, bladder, and renal cancers. Eur. Urol. Focus 2017, 3, 265–272. [Google Scholar] [CrossRef]
  49. Larsen, L.K.; Jakobsen, J.S.; Abdul-Al, A.; Guldberg, P. Noninvasive detection of high grade prostate cancer by DNA methylation analysis of urine cells captured by microfiltration. J. Urol. 2018, 200, 749–757. [Google Scholar] [CrossRef]
  50. Dahl, C.; Guldberg, P. DNA methylation analysis techniques. Biogerontology 2003, 4, 233–250. [Google Scholar] [CrossRef]
  51. Grunau, C.; Clark, S.J.; Rosenthal, A. Bisulfite genomic sequencing: Systematic investigation of critical experimental parameters. Nucleic Acids Res. 2001, 29, E65. [Google Scholar] [CrossRef]
  52. Russo, I.J.; Ju, Y.; Gordon, N.S.; Zeegers, M.P.; Cheng, K.K.; James, N.D.; Bryan, R.T.; Ward, D.G. Toward personalised liquid biopsies for urothelial carcinoma: Characterisation of ddpcr and urinary cfdna for the detection of the tert 228 g>a/t mutation. Bladder Cancer 2018, 4, 41–48. [Google Scholar] [CrossRef]
  53. Worm Orntoft, M.B.; Jensen, S.O.; Hansen, T.B.; Bramsen, J.B.; Andersen, C.L. Comparative analysis of 12 different kits for bisulfite conversion of circulating cell-free DNA. Epigenetics 2017, 12, 626–636. [Google Scholar] [CrossRef]
  54. Pharo, H.D.; Honne, H.; Vedeld, H.M.; Dahl, C.; Andresen, K.; Liestol, K.; Jeanmougin, M.; Guldberg, P.; Lind, G.E. Experimental factors affecting the robustness of DNA methylation analysis. Sci. Rep. 2016, 6, 33936. [Google Scholar] [CrossRef]
  55. Tost, J.; Gut, I.G. DNA methylation analysis by pyrosequencing. Nat. Protoc. 2007, 2, 2265–2275. [Google Scholar] [CrossRef]
  56. Eads, C.A.; Danenberg, K.D.; Kawakami, K.; Saltz, L.B.; Blake, C.; Shibata, D.; Danenberg, P.V.; Laird, P.W. Methylight: A high-throughput assay to measure DNA methylation. Nucleic Acids Res. 2000, 28, E32. [Google Scholar] [CrossRef]
  57. Gonzalgo, M.L.; Jones, P.A. Rapid quantitation of methylation differences at specific sites using methylation-sensitive single nucleotide primer extension (ms-snupe). Nucleic Acids Res. 1997, 25, 2529–2531. [Google Scholar] [CrossRef]
  58. Wojdacz, T.K.; Dobrovic, A. Methylation-sensitive high resolution melting (ms-hrm): A new approach for sensitive and high-throughput assessment of methylation. Nucleic Acids Res. 2007, 35, e41. [Google Scholar] [CrossRef]
  59. Nygren, A.O.; Ameziane, N.; Duarte, H.M.; Vijzelaar, R.N.; Waisfisz, Q.; Hess, C.J.; Schouten, J.P.; Errami, A. Methylation-specific mlpa (ms-mlpa): Simultaneous detection of cpg methylation and copy number changes of up to 40 sequences. Nucleic Acids Res. 2005, 33, e128. [Google Scholar] [CrossRef]
  60. consortium, B. Quantitative comparison of DNA methylation assays for biomarker development and clinical applications. Nat. Biotechnol. 2016, 34, 726–737. [Google Scholar] [CrossRef] [Green Version]
  61. Armbruster, D.A.; Pry, T. Limit of blank, limit of detection and limit of quantitation. Clin. Biochem. Rev. 2008, 29 (Suppl. 1), S49–S52. [Google Scholar]
  62. Deng, G.; Peng, E.; Gum, J.; Terdiman, J.; Sleisenger, M.; Kim, Y.S. Methylation of hmlh1 promoter correlates with the gene silencing with a region-specific manner in colorectal cancer. Br. J. Cancer 2002, 86, 574–579. [Google Scholar] [CrossRef]
  63. Fantony, J.J.; Longo, T.A.; Gopalakrishna, A.; Owusu, R.; Lance, R.S.; Foo, W.C.; Inman, B.A.; Abern, M.R. Urinary nid2 and twist1 methylation to augment conventional urine cytology for the detection of bladder cancer. Cancer Biomark. 2017, 18, 381–387. [Google Scholar] [CrossRef]
  64. Sathyanarayana, U.G.; Maruyama, R.; Padar, A.; Suzuki, M.; Bondaruk, J.; Sagalowsky, A.; Minna, J.D.; Frenkel, E.P.; Grossman, H.B.; Czerniak, B.; et al. Molecular detection of noninvasive and invasive bladder tumor tissues and exfoliated cells by aberrant promoter methylation of laminin-5 encoding genes. Cancer Res. 2004, 64, 1425–1430. [Google Scholar] [CrossRef]
  65. Urakami, S.; Shiina, H.; Enokida, H.; Kawakami, T.; Kawamoto, K.; Hirata, H.; Tanaka, Y.; Kikuno, N.; Nakagawa, M.; Igawa, M.; et al. Combination analysis of hypermethylated wnt-antagonist family genes as a novel epigenetic biomarker panel for bladder cancer detection. Clin. Cancer Res. 2006, 12, 2109–2116. [Google Scholar] [CrossRef]
  66. Hoque, M.O.; Begum, S.; Topaloglu, O.; Chatterjee, A.; Rosenbaum, E.; Van Criekinge, W.; Westra, W.H.; Schoenberg, M.; Zahurak, M.; Goodman, S.N.; et al. Quantitation of promoter methylation of multiple genes in urine DNA and bladder cancer detection. J. Natl. Cancer Inst. 2006, 98, 996–1004. [Google Scholar] [CrossRef]
  67. Yates, D.R.; Rehman, I.; Meuth, M.; Cross, S.S.; Hamdy, F.C.; Catto, J.W. Methylational urinalysis: A prospective study of bladder cancer patients and age stratified benign controls. Oncogene 2006, 25, 1984–1988. [Google Scholar] [CrossRef]
  68. Bayramov, B.; Gunes, S.; Buyukalpelli, R.; Aydin, O.; Henkel, R. Promoter methylation analysis of cdh1 and p14arf genes in patients with urothelial bladder cancer. Oncol. Targets 2018, 11, 4189–4196. [Google Scholar] [CrossRef]
  69. Guo, R.Q.; Xiong, G.Y.; Yang, K.W.; Zhang, L.; He, S.M.; Gong, Y.Q.; He, Q.; Li, X.Y.; Wang, Z.C.; Bao, Z.Q.; et al. Detection of urothelial carcinoma, upper tract urothelial carcinoma, bladder carcinoma, and urothelial carcinoma with gross hematuria using selected urine-DNA methylation biomarkers: A prospective, single-center study. Urol. Oncol. 2018, 36, 342.e315–342.e323. [Google Scholar] [CrossRef]
  70. Mijnes, J.; Veeck, J.; Gaisa, N.T.; Burghardt, E.; de Ruijter, T.C.; Gostek, S.; Dahl, E.; Pfister, D.; Schmid, S.C.; Knuchel, R.; et al. Promoter methylation of DNA damage repair (ddr) genes in human tumor entities: Rbbp8/ctip is almost exclusively methylated in bladder cancer. Clin. Epigenetics 2018, 10, 15. [Google Scholar] [CrossRef]
  71. Fantony, J.J.; Abern, M.R.; Gopalakrishna, A.; Owusu, R.; Jack Tay, K.; Lance, R.S.; Inman, B.A. Multi-institutional external validation of urinary twist1 and nid2 methylation as a diagnostic test for bladder cancer. Urol. Oncol. 2015, 33, 387.e381–387.e386. [Google Scholar] [CrossRef]
  72. Maldonado, L.; Brait, M.; Michailidi, C.; Munari, E.; Driscoll, T.; Schultz, L.; Bivalacqua, T.; Schoenberg, M.; Sidransky, D.; Netto, G.J.; et al. An epigenetic marker panel for recurrence risk prediction of low grade papillary urothelial cell carcinoma (lgpucc) and its potential use for surveillance after transurethral resection using urine. Oncotarget 2014, 5, 5218–5233. [Google Scholar] [CrossRef]
  73. Hayashi, M.; Bernert, H.; Kagohara, L.T.; Maldonado, L.; Brait, M.; Schoenberg, M.; Bivalacqua, T.; Netto, G.J.; Koch, W.; Sidransky, D.; et al. Epigenetic inactivation of vgf associated with urothelial cell carcinoma and its potential as a non-invasive biomarker using urine. Oncotarget 2014, 5, 3350–3361. [Google Scholar] [CrossRef]
  74. Abern, M.R.; Owusu, R.; Inman, B.A. Clinical performance and utility of a DNA methylation urine test for bladder cancer. Urol. Oncol. 2014, 32, 51.e21–51.e26. [Google Scholar] [CrossRef]
  75. Beukers, W.; Kandimalla, R.; van Houwelingen, D.; Kovacic, H.; Chin, J.F.; Lingsma, H.F.; Dyrskjot, L.; Zwarthoff, E.C. The use of molecular analyses in voided urine for the assessment of patients with hematuria. PLoS ONE 2013, 8, e77657. [Google Scholar] [CrossRef]
  76. Garcia-Baquero, R.; Puerta, P.; Beltran, M.; Alvarez, M.; Sacristan, R.; Alvarez-Ossorio, J.L.; Sanchez-Carbayo, M. Methylation of a novel panel of tumor suppressor genes in urine moves forward noninvasive diagnosis and prognosis of bladder cancer: A 2-center prospective study. J. Urol. 2013, 190, 723–730. [Google Scholar] [CrossRef]
  77. Scher, M.B.; Elbaum, M.B.; Mogilevkin, Y.; Hilbert, D.W.; Mydlo, J.H.; Sidi, A.A.; Adelson, M.E.; Mordechai, E.; Trama, J.P. Detecting DNA methylation of the bcl2, cdkn2a and nid2 genes in urine using a nested methylation specific polymerase chain reaction assay to predict bladder cancer. J. Urol. 2012, 188, 2101–2107. [Google Scholar] [CrossRef]
  78. Karnes, R.J.; Fernandez, C.A.; Shuber, A.P. A noninvasive multianalyte urine-based diagnostic assay for urothelial cancer of the bladder in the evaluation of hematuria. Mayo Clin. Proc. 2012, 87, 835–842. [Google Scholar] [CrossRef]
  79. Zhao, Y.; Guo, S.; Sun, J.; Huang, Z.; Zhu, T.; Zhang, H.; Gu, J.; He, Y.; Wang, W.; Ma, K.; et al. Methylcap-seq reveals novel DNA methylation markers for the diagnosis and recurrence prediction of bladder cancer in a chinese population. PLoS ONE 2012, 7, e35175. [Google Scholar] [CrossRef]
  80. Eissa, S.; Zohny, S.F.; Shehata, H.H.; Hegazy, M.G.; Salem, A.M.; Esmat, M. Urinary retinoic acid receptor-beta2 gene promoter methylation and hyaluronidase activity as noninvasive tests for diagnosis of bladder cancer. Clin. Biochem. 2012, 45, 402–407. [Google Scholar] [CrossRef]
  81. Reinert, T.; Modin, C.; Castano, F.M.; Lamy, P.; Wojdacz, T.K.; Hansen, L.L.; Wiuf, C.; Borre, M.; Dyrskjot, L.; Orntoft, T.F. 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. [Google Scholar] [CrossRef]
  82. Eissa, S.; Swellam, M.; El-Khouly, I.M.; Kassim, S.K.; Shehata, H.; Mansour, A.; Esmat, M.; Nossier, A.I.; Hamdy, M.A.; Awad, N.M.; et al. Aberrant methylation of rarbeta2 and apc genes in voided urine as molecular markers for early detection of bilharzial and nonbilharzial bladder cancer. Cancer Epidemiol. Biomark. Prev. 2011, 20, 1657–1664. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, P.C.; Tsai, M.H.; Yip, S.K.; Jou, Y.C.; Ng, C.F.; Chen, Y.; Wang, X.; Huang, W.; Tung, C.L.; Chen, G.C.; et al. Distinct DNA methylation epigenotypes in bladder cancer from different chinese sub-populations and its implication in cancer detection using voided urine. BMC Med. Genom. 2011, 4, 45. [Google Scholar] [CrossRef] [PubMed]
  84. Chung, W.; Bondaruk, J.; Jelinek, J.; Lotan, Y.; Liang, S.; Czerniak, B.; Issa, J.P. Detection of bladder cancer using novel DNA methylation biomarkers in urine sediments. Cancer Epidemiol. Biomark. Prev. 2011, 20, 1483–1491. [Google Scholar] [CrossRef]
  85. Serizawa, R.R.; Ralfkiaer, U.; Steven, K.; Lam, G.W.; Schmiedel, S.; Schuz, J.; Hansen, A.B.; Horn, T.; Guldberg, P. Integrated genetic and epigenetic analysis of bladder cancer reveals an additive diagnostic value of fgfr3 mutations and hypermethylation events. Int. J. Cancer 2011, 129, 78–87. [Google Scholar] [CrossRef] [PubMed]
  86. Vinci, S.; Giannarini, G.; Selli, C.; Kuncova, J.; Villari, D.; Valent, F.; Orlando, C. Quantitative methylation analysis of bcl2, htert, and dapk promoters in urine sediment for the detection of non-muscle-invasive urothelial carcinoma of the bladder: A prospective, two-center validation study. Urol. Oncol. 2011, 29, 150–156. [Google Scholar] [CrossRef]
  87. Lin, H.H.; Ke, H.L.; Huang, S.P.; Wu, W.J.; Chen, Y.K.; Chang, L.L. Increase sensitivity in detecting superficial, low grade bladder cancer by combination analysis of hypermethylation of e-cadherin, p16, p14, rassf1a genes in urine. Urol. Oncol. 2010, 28, 597–602. [Google Scholar] [CrossRef]
  88. Pu, R.T.; Laitala, L.E.; Clark, D.P. Methylation profiling of urothelial carcinoma in bladder biopsy and urine. Acta Cytol. 2006, 50, 499–506. [Google Scholar] [CrossRef]
  89. Padrao, N.A.; Monteiro-Reis, S.; Torres-Ferreira, J.; Antunes, L.; Leca, L.; Montezuma, D.; Ramalho-Carvalho, J.; Dias, P.C.; Monteiro, P.; Oliveira, J.; et al. Microrna promoter methylation: A new tool for accurate detection of urothelial carcinoma. Br. J. Cancer 2017, 116, 634–639. [Google Scholar] [CrossRef]
  90. Shimizu, T.; Suzuki, H.; Nojima, M.; Kitamura, H.; Yamamoto, E.; Maruyama, R.; Ashida, M.; Hatahira, T.; Kai, M.; Masumori, N.; et al. Methylation of a panel of microrna genes is a novel biomarker for detection of bladder cancer. Eur. Urol. 2013, 63, 1091–1100. [Google Scholar] [CrossRef]
  91. van Kessel, K.E.; Van Neste, L.; Lurkin, I.; Zwarthoff, E.C.; Van Criekinge, W. Evaluation of an epigenetic profile for the detection of bladder cancer in patients with hematuria. J. Urol. 2016, 195, 601–607. [Google Scholar] [CrossRef]
  92. Chan, M.W.; Chan, L.W.; Tang, N.L.; Lo, K.W.; Tong, J.H.; Chan, A.W.; Cheung, H.Y.; Wong, W.S.; Chan, P.S.; Lai, F.M.; et al. Frequent hypermethylation of promoter region of rassf1a in tumor tissues and voided urine of urinary bladder cancer patients. Int. J. Cancer 2003, 104, 611–616. [Google Scholar] [CrossRef]
  93. Zhao, F.; Olkhov-Mitsel, E.; Kamdar, S.; Jeyapala, R.; Garcia, J.; Hurst, R.; Hanna, M.Y.; Mills, R.; Tuzova, A.V.; O’Reilly, E.; et al. A urine-based DNA methylation assay, procure, to identify clinically significant prostate cancer. Clin. Epigenetics 2018, 10, 147. [Google Scholar] [CrossRef]
  94. Yao, L.; Ren, S.; Zhang, M.; Du, F.; Zhu, Y.; Yu, H.; Zhang, C.; Li, X.; Yang, C.; Liu, H.; et al. Identification of specific DNA methylation sites on the y-chromosome as biomarker in prostate cancer. Oncotarget 2015, 6, 40611–40621. [Google Scholar] [CrossRef]
  95. Haluskova, J.; Lachvac, L.; Nagy, V. The investigation of gstp1, apc and rassf1 gene promoter hypermethylation in urine DNA of prostate-diseased patients. Bratislava Lek Listy 2015, 116, 79–82. [Google Scholar] [CrossRef]
  96. Zhang, L.; Zhang, Q.; Li, L.; Wang, Z.; Ying, J.; Fan, Y.; He, Q.; Lv, T.; Han, W.; Li, J.; et al. Dlec1, a 3p tumor suppressor, represses nf-kappab signaling and is methylated in prostate cancer. J. Mol. Med. (Berl.) 2015, 93, 691–701. [Google Scholar] [CrossRef]
  97. Daniunaite, K.; Jarmalaite, S.; Kalinauskaite, N.; Petroska, D.; Laurinavicius, A.; Lazutka, J.R.; Jankevicius, F. Prognostic value of rassf1 promoter methylation in prostate cancer. J. Urol. 2014, 192, 1849–1855. [Google Scholar] [CrossRef]
  98. Dimitriadis, E.; Kalogeropoulos, T.; Velaeti, S.; Sotiriou, S.; Vassiliou, E.; Fasoulis, L.; Klapsas, V.; Synesiou, M.; Apostolaki, A.; Trangas, T.; et al. Study of genetic and epigenetic alterations in urine samples as diagnostic markers for prostate cancer. Anticancer Res. 2013, 33, 191–197. [Google Scholar]
  99. Daniunaite, K.; Berezniakovas, A.; Jankevicius, F.; Laurinavicius, A.; Lazutka, J.R.; Jarmalaite, S. Frequent methylation of rassf1 and rarb in urine sediments from patients with early stage prostate cancer. Medicina (Kaunas) 2011, 47, 147–153. [Google Scholar]
  100. Prior, C.; Guillen-Grima, F.; Robles, J.E.; Rosell, D.; Fernandez-Montero, J.M.; Agirre, X.; Catena, R.; Calvo, A. Use of a combination of biomarkers in serum and urine to improve detection of prostate cancer. World J. Urol. 2010, 28, 681–686. [Google Scholar] [CrossRef]
  101. Payne, S.R.; Serth, J.; Schostak, M.; Kamradt, J.; Strauss, A.; Thelen, P.; Model, F.; Day, J.K.; Liebenberg, V.; Morotti, A.; et al. DNA methylation biomarkers of prostate cancer: Confirmation of candidates and evidence urine is the most sensitive body fluid for non-invasive detection. Prostate 2009, 69, 1257–1269. [Google Scholar] [CrossRef] [PubMed]
  102. Woodson, K.; O’Reilly, K.J.; Hanson, J.C.; Nelson, D.; Walk, E.L.; Tangrea, J.A. The usefulness of the detection of gstp1 methylation in urine as a biomarker in the diagnosis of prostate cancer. J. Urol. 2008, 179, 508–511. [Google Scholar] [CrossRef]
  103. Roupret, M.; Hupertan, V.; Yates, D.R.; Catto, J.W.; Rehman, I.; Meuth, M.; Ricci, S.; Lacave, R.; Cancel-Tassin, G.; de la Taille, A.; et al. Molecular detection of localized prostate cancer using quantitative methylation-specific pcr on urinary cells obtained following prostate massage. Clin. Cancer Res. 2007, 13, 1720–1725. [Google Scholar] [CrossRef] [PubMed]
  104. Hoque, M.O.; Topaloglu, O.; Begum, S.; Henrique, R.; Rosenbaum, E.; Van Criekinge, W.; Westra, W.H.; Sidransky, D. Quantitative methylation-specific polymerase chain reaction gene patterns in urine sediment distinguish prostate cancer patients from control subjects. J. Clin. Oncol. 2005, 23, 6569–6575. [Google Scholar] [CrossRef] [PubMed]
  105. Goessl, C.; Muller, M.; Heicappell, R.; Krause, H.; Straub, B.; Schrader, M.; Miller, K. DNA-based detection of prostate cancer in urine after prostatic massage. Urology 2001, 58, 335–338. [Google Scholar] [CrossRef]
  106. Baden, J.; Green, G.; Painter, J.; Curtin, K.; Markiewicz, J.; Jones, J.; Astacio, T.; Canning, S.; Quijano, J.; Guinto, W.; et al. Multicenter evaluation of an investigational prostate cancer methylation assay. J. Urol. 2009, 182, 1186–1193. [Google Scholar] [CrossRef] [PubMed]
  107. Vener, T.; Derecho, C.; Baden, J.; Wang, H.; Rajpurohit, Y.; Skelton, J.; Mehrotra, J.; Varde, S.; Chowdary, D.; Stallings, W.; et al. Development of a multiplexed urine assay for prostate cancer diagnosis. Clin. Chem. 2008, 54, 874–882. [Google Scholar] [CrossRef]
  108. Gonzalgo, M.L.; Pavlovich, C.P.; Lee, S.M.; Nelson, W.G. Prostate cancer detection by gstp1 methylation analysis of postbiopsy urine specimens. Clin. Cancer Res. 2003, 9, 2673–2677. [Google Scholar]
  109. Jeronimo, C.; Usadel, H.; Henrique, R.; Silva, C.; Oliveira, J.; Lopes, C.; Sidransky, D. Quantitative gstp1 hypermethylation in bodily fluids of patients with prostate cancer. Urology 2002, 60, 1131–1135. [Google Scholar] [CrossRef]
  110. Goessl, C.; Muller, M.; Heicappell, R.; Krause, H.; Miller, K. DNA-based detection of prostate cancer in blood, urine, and ejaculates. Ann. N. Y. Acad. Sci. 2001, 945, 51–58. [Google Scholar] [CrossRef]
  111. Dumache, R.; Popescu, S.; Minciu, R.; Negru, S.; Puiu, M. Molecular detection of prostate cancer by methylation-specific polymerase chain reaction from urine specimens. J. Med. Biochem. 2013, 32, 233–237. [Google Scholar] [CrossRef]
Figure 1. Reported sensitivities of DNA methylation biomarkers for detection of primary bladder cancer. *, Inconsistent nomenclature among studies.
Figure 1. Reported sensitivities of DNA methylation biomarkers for detection of primary bladder cancer. *, Inconsistent nomenclature among studies.
Ijms 20 02657 g001
Figure 2. Reported sensitivities of DNA methylation biomarkers for detection of prostate cancer. *, Inconsistent nomenclature among studies.
Figure 2. Reported sensitivities of DNA methylation biomarkers for detection of prostate cancer. *, Inconsistent nomenclature among studies.
Ijms 20 02657 g002
Table 1. DNA-methylation biomarker panels for detection of primary bladder cancer.
Table 1. DNA-methylation biomarker panels for detection of primary bladder cancer.
BiomarkerSample ProcessingCases (n)Controls (n)PathologyControl PopulationMethodSens. (%)Spec. (%)Ref.Year
SOX1, TJP2, MYOD, HOXA9_1, HOXA9_2, VAMP8, CASP8, SPP1, IFNG, CAPG, HLADPA1, RIPK3 (Positive when six or more markers are present)Sedimentation7318Ta-T4, Grade 1–3HealthyPyrosequencing100100[10]2013
POU4F2, PCDH17, GDF15Sedimentation7292Ta-T4, LG, HGMixed urologic diseases and healthyqMSP9775[11]2016
ZNF671, SFRP1, IRF8Sedimentation2619Ta-T4, LG, HGNoncancer, not specifiedqMSP9684[12]2015
POU4F2, EOMESSedimentation7292Ta-T4, LG, HGMixed urologic diseases and healthyqMSP9688[11]2016
TWIST1, NID2Sedimentation2415Ta-T3, LG, HGMixed urologic diseases, and healthyMSP9693[13]2013
BCL2, EOMES, VIM, SALL3, CCNA1, HOXA9, POU4F2Filtration (8 µm)3326Ta-T2, LH, HG, PUNLMPMixed urologic diseases, negative findingsqMSP94 [14]2015
TWIST1, NID2Sedimentation3557Ta-T2, LG, HGMixed urologic diseasesqMSP9491[15]2010
VIM, TMEFF2, GDF15Sedimentation5120Not SpecifiedHealthyqMSP94100[16]2010
VIM, TMEFF2, GDF15, HSPA2Sedimentation5120Not SpecifiedHealthyqMSP94100[16]2010
SALL3, CFTR, ABCC6, HPR1, RASSF1A, MT1A, ALX4, CDH13, RPRM, MINT1, BRCA1Sedimentation13223 Stage 0a-IVMixed urologic diseasesMSP9287[17]2007
SALL3, CFTR, MT1A, HPP1, ABCC6, RASSF1A, CDH13, RPRM, MINT1, BRCA1, SFRP1Sedimentation8215Stage pTa-IVMixed urologic diseasesMSP9273[18]2009
POU4F2, PCDH17Sedimentation5890Ta-T4, LG, HGMixed urologic diseases and healthyqMSP9193[11]2016
POU4F2, PCDH17, GDF15Sedimentation5890Ta-T4, LG, HGMixed urologic diseases and healthyqMSP9188[11]2016
p14ARF, p16INK4A, RASSF1A, DAPK, APCSedimentation113 ≥T1, PUNLMP, Grade 1–3HealthyMSP91 [19]2017
SEPTIN9, SLIT2Filtration (11 µm)167105Ta-T1 (NMIBC), LG, HGPatients with negative cystoscopy (hematuria)qMSP9171[20]2016
RARβ, DAPK, CDH1, p16Sedimentation2217NMIBC-MIBC, Grade 1–3HealthyMSP9176[21]2002
HOXA9, PCDH17, POU4F2, ONECUT2SedimentationNot specifiedNot specifiedTa-T4, LG, HG, PUNLMPMixed urologic diseases, hematuriaqMSP9173[22]2018
HS3ST2, SEPTIN9, SLIT2Filtration (11 µm)167105Ta-T1 (NMIBC), LG, HGPatients with negative cystoscopy (hematuria)qMSP9075[20]2016
HS3ST2, SLIT2Filtration (11 µm)167105Ta-T1 (NMIBC), LG, HGPatients with negative cystoscopy (hematuria)qMSP9034[20]2016
HS3ST2, SEPTIN9Filtration (11 µm)167105Ta-T1 (NMIBC), LG, HGPatients with negative cystoscopy (hematuria)qMSP9072[20]2016
SALL3, CFTR, MT1A, HPP1, ABCC6, RASSF1A, CDH13, RPRM, MINT1, BRCA1Sedimentation8215Stage Ta-IVMixed urologic diseasesMSP9080[18]2009
ONECUT2, VIM, SALL3, CCNA1, BCL2, EOMESFiltration (8 µm)99376Ta-T4, LG, HG, PUNLMPMacroscopic hematuria, no malignancyqMSP 9089[23]2016
MSP = methylation-specific PCR, qMSP = quantitative MSP, HG = high grade, LG = low grade, PUNLMP = papillary urothelial neoplasm of low malignant potential, NMIBC = nonmuscle invasive bladder cancer, MIBC = muscle invasive bladder cancer.
Table 2. DNA-methylation biomarkers and biomarker panels for detection of recurrent bladder cancer.
Table 2. DNA-methylation biomarkers and biomarker panels for detection of recurrent bladder cancer.
BiomarkerSample ProcessingCases (n)Controls (n)PathologyControl PopulationMethodSens. (%)Spec. (%)Ref.Year
APCSedimentation1525Ta-T1No recurrenceqMSP2780[24]2008
BCL2Sedimentation1525Ta-T1No recurrenceqMSP13962008
DAPKSedimentation1525Ta-T1No recurrenceqMSP0962008
CDH1Sedimentation1525Ta-T1No recurrenceqMSP7842008
EDNRBSedimentation1525Ta-T1No recurrenceqMSP20802008
EOMESSedimentation13967Ta-T1, Grade 1–3Mixed urologic diseasesqMSP9455[25]2012
HOXA9Sedimentation13967Ta-T1, Grade 1–3Mixed urologic diseasesqMSP93552012
IGFBPSedimentation1525Ta-T1No recurrenceqMSP2084[24]2008
MGMTSedimentation1525Ta-T1No recurrenceqMSP20922008
NID2 48275Ta-T3, Grade 1–3No recurrenceMSP4690[26]2012
POU4F2Sedimentation13967Ta-T1, Grade 1–3Mixed urologic diseasesqMSP8864[25]2012
RASSF1ASedimentation1525Ta-T1No recurrenceqMSP5032[24]2008
TERTSedimentation1525Ta-T1No recurrenceqMSP131002008
TNFRSF25Sedimentation1525Ta-T1No recurrenceqMSP40562008
TWIST1Sedimentation13967Ta-T1, Grade 1–3Mixed urologic diseasesqMSP9043[25]2012
TWIST1 48275Ta-T3, Grade 1–3No recurrenceMSP7569[26]2012
VIMSedimentation13967Ta-T1, Grade 1–3Mixed urologic diseasesqMSP9059[25]2012
WIF1Sedimentation1525Ta-T1No recurrenceqMSP2076[24]2008
ZNF154Sedimentation13967Ta-T1, Grade 1–3Mixed urologic diseasesqMSP9467[25]2012
APC_a, TERT_a, TERT_b, EDNRBSedimentation6529Ta-T4, Grade 0–3No recurrenceMS-MLPA7255[27]2012
APC_a, TERT_a, TERT_b, EDNRBSedimentation4960Ta-T1, Grade 1–3No recurrenceMS-MLPA63582012
APC_a, TERT_a, TERT_b, EDNRBSedimentation6891Ta-T1, Grade 1–3No BCMS-MLPA 2012
CFTR, SALL3, TWIST1Sedimentation173285Ta-T1Ta-T1Pyrosequencing9031[28]2018
HS3ST2, SLIT2, SEPTIN9Filtration (11 µm)7286Ta-T4, LG, HGTa-T4, LG, HGqMSP [20]2016
miR-9-3, miR124-2, miR-124-3, miR-137Sedimentation25107Ta-T1No recurrencePyrosequencing6274[29]2018
OTX1, ONECUT2, OSR1Sedimentation9540NMIBC, Grade 1–3, (recurrence)No recurrenceSNaPshot74Fixed = 90%[30]2013
Panel consisting of 41 sequencesSedimentation136 ≥Ta, LG, HGMixed urologic diseases, and healthyMS-MLPA [31]2013
RASSF1A, ECAD, APC, DAPK, MGMT, BCL2, TERT, EDNRB, WIF1, TNFRSF25. IGFBPSedimentation1525Ta-T1No recurrenceqMSP868[24]2008
SOX1, IRAK3, L1-MET (L1-MET hypomethylated)Sedimentation2954Ta-T1, LG, HGTa-T1, LG, HGPyrosequencing8689[32]2014
SOX1, IRAK3, L1-MET (L1-MET hypomethylated)Sedimentation13425Ta-T1, LG, HGTa-T1, LG, HGPyrosequencing80972014
MSP = methylation specific PCR, qMSP = quantitative MSP, HG = high grade, LG = low grade, NMIBC = nonmuscle invasive bladder cancer, BC = bladder cancer, MS-MLPA = methylation-specific multiplex ligation-dependent probe amplification.
Table 3. DNA-methylation biomarker panels for detection of prostate cancer.
Table 3. DNA-methylation biomarker panels for detection of prostate cancer.
BiomarkersUrine CollectionSample ProcessingCancer (n)Controls (n)PathologyControl PopulationMethodSens. (%)Spec. (%)Ref.Year
GSTP1, RARβ2, APC, miR-34b/c + miR-193bMorningSedimentation8732T2-T3bAsymptomatic donorsqMSP10075[33]2018
TGFB2, HOXD3, APCPost DRESedimentation105Organ confinedCancer free (not further specified)qMSP10060[34]2014
EDNRB, APC, GSTP1Post DRE/biopsySedimentation125GS 6–7Biopsy NegativeMSP10040[35]2006
miR-34b/c + miR-193bMorningSedimentation8732T2-T3bAsymptomatic donorsqMSP9584[33]2018
GSTP1, RARβ2, APCMorningSedimentation8732T2-T3bAsymptomatic donorsqMSP94842018
≥6 positive of 19 markersFirst VoidSedimentation3235GS 6–10Negative biopsy resultsNested qMSP9471[36]2018
miR-34b/c + miR-193bNo DRESedimentation9546GS ≥ 6No urological malignancy, healthyqMSP9198[37]2017
MSP = methylation specific PCR, qMSP = quantitative MSP, DRE = digital rectal examination, GS = Gleason score.
Table 4. DNA-methylation biomarkers and biomarker panels for detection of renal cancer.
Table 4. DNA-methylation biomarkers and biomarker panels for detection of renal cancer.
BiomarkerSample ProcessingCancer (n)Controls (n)PathologyControl PopulationMethodSens. (%)Spec. (%)Ref.Year
APCSedimentation2691Not specifiedVarious conditions, malignant and nonmalignantqMSP3896[38]2004
ARFSedimentation2691Not specifiedVarious conditions, malignant and nonmalignantqMSP311002004
CDH1Sedimentation2691Not specifiedVarious conditions, malignant and nonmalignantqMSP38952004
GDF15Sedimentation1920Not specifiedHealthyqMSP5100[16]2010
GSTP1Sedimentation2691Not specifiedVarious conditions, malignant and nonmalignantqMSP15100[38]2004
HSPA2Sedimentation1920Not specifiedHealthyqMSP11100[16]2010
MGMTSedimentation2691Not specifiedVarious conditions, malignant and nonmalignantqMSP8100[38]2004
p16Sedimentation2691Not specifiedVarious conditions, malignant and nonmalignantqMSP351002004
PCDH17Sedimentation5048Not specifiedHealthyqMSP20100[39]2011
RARB2Sedimentation2691Not specifiedVarious conditions, malignant and nonmalignantqMSP3191[38]2004
RASSF1ASedimentation2691Not specifiedVarious conditions, malignant and nonmalignantqMSP65892004
TCF21Sedimentation3315Grades I–IVHealthyPyrosequencing79100[40]2016
TCF21Sedimentation5048Not specifiedHealthyqMSP28100[39]2011
TIMP3Sedimentation2691Not specifiedVarious conditions, malignant and nonmalignantqMSP4691[38]2004
TMEFF2Sedimentation1920Not specifiedHealthyqMSP11100[16]2010
VIMSedimentation1920Not specifiedHealthyqMSP51002010
PCDH17, TCF21Sedimentation5048Not specifiedHealthyqMSP32100[39]2011
APC, ARF, CDH1, GSTP1, MGMT, P16, RAR-β2, RASSF1A, TIMP3Sedimentation2691Not specifiedVarious conditions, malignant and nonmalignantqMSP88 [38]2004
VHL, p16/cdkn2a, p14ARF, APC, RASSF1A, Timp-3Sedimentation5024T1–T3Healthy, renal stones, benign renal cystsMSP88100[41]2004
MSP = methylation specific PCR, qMSP = quantitative MSP.
Table 5. DNA methylation biomarkers and biomarker panels for detection of upper urinary tract tumors.
Table 5. DNA methylation biomarkers and biomarker panels for detection of upper urinary tract tumors.
BiomarkersSample ProcessingCancer (n)Controls (n)PathologyControl PopulationMethodSens. (%)Spec. (%)RefYear
ABCC6Sedimentation98113Not specifiedBenign urologic conditionsMSP4454[42]2018
BRCA1Sedimentation98113Not specifiedBenign urologic conditionsMSP26582018
CDH1Sedimentation98113Not specifiedBenign urologic conditionsMSP28982018
GDF15Sedimentation98113Not specifiedBenign urologic conditionsMSP30902018
HSPA2Sedimentation98113Not specifiedBenign urologic conditionsMSP83362018
RASSF1ASedimentation98113Not specifiedBenign urologic conditionsMSP48732018
SALL3Sedimentation98113Not specifiedBenign urologic conditionsMSP23802018
THBS1Sedimentation98113Not specifiedBenign urologic conditionsMSP74252018
TMEFF2Sedimentation98113Not specifiedBenign urologic conditionsMSP67402018
VIMSedimentation98113Not specifiedBenign urologic conditionsMSP73612018
VIMSedimentation2220Not specifiedHealthyqMSP82100[43]2014
CDH1, VIMSedimentation98113Not specifiedBenign urologic conditionsMSP8260[42]2018
CDH1, VIM, RASSF1ASedimentation98113Not specifiedBenign urologic conditionsMSP82602018
CDH1, VIM, RASSF1A, HSPA2Sedimentation98113Not specifiedBenign urologic conditionsMSP85592018
CDH1, VIM, RASSF1A, HSPA2, GDF15Sedimentation98113Not specifiedBenign urologic conditionsMSP82652018
CDH1, VIM, RASSF1A, HSPA2, GDF15, TMEFF2Sedimentation98113Not specifiedBenign urologic conditionsMSP82682018
VIM, GDF15Sedimentation2220Papillary, invasive, LG, HGHealthy, renal cell carcinoma, prostate carcinomaqMSP91100[43]2014
VIM, GDF15, TMEFF2Sedimentation2220Papillary, invasive, LG, HGHealthy, renal cell carcinoma, prostate carcinomaqMSP911002014
MSP = methylation specific PCR, qMSP = quantitative MSP, HG = high grade, LG = low grade.

Share and Cite

MDPI and ACS Style

Larsen, L.K.; Lind, G.E.; Guldberg, P.; Dahl, C. DNA-Methylation-Based Detection of Urological Cancer in Urine: Overview of Biomarkers and Considerations on Biomarker Design, Source of DNA, and Detection Technologies. Int. J. Mol. Sci. 2019, 20, 2657. https://doi.org/10.3390/ijms20112657

AMA Style

Larsen LK, Lind GE, Guldberg P, Dahl C. DNA-Methylation-Based Detection of Urological Cancer in Urine: Overview of Biomarkers and Considerations on Biomarker Design, Source of DNA, and Detection Technologies. International Journal of Molecular Sciences. 2019; 20(11):2657. https://doi.org/10.3390/ijms20112657

Chicago/Turabian Style

Larsen, Louise Katrine, Guro Elisabeth Lind, Per Guldberg, and Christina Dahl. 2019. "DNA-Methylation-Based Detection of Urological Cancer in Urine: Overview of Biomarkers and Considerations on Biomarker Design, Source of DNA, and Detection Technologies" International Journal of Molecular Sciences 20, no. 11: 2657. https://doi.org/10.3390/ijms20112657

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop