Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jul 18.
Published in final edited form as: Expert Rev Anticancer Ther. 2008 Jul;8(7):1111–1123. doi: 10.1586/14737140.8.7.1111

Bladder tumor markers: from hematuria to molecular diagnostics – where do we stand?

Samir P Shirodkar 1, Vinata B Lokeshwar 2,
PMCID: PMC5515477  NIHMSID: NIHMS823948  PMID: 18588456

Abstract

Bladder cancer is a common malignancy in the USA. Currently, the detection of initial tumors and recurrent disease is based on evaluation of voided urinary specimens, often followed by cystoscopy. With the high rate of recurrence, cystoscopies are regularly repeated with the aim of halting progression of the disease. For patients, this process is fraught with anxiety, pain and high cost. As a result, intense work is being done in the field of bladder tumor markers with the goal of identifying bladder cancer earlier, both in the initial diagnosis and in recurrences of known tumor. The possibility of identifying a marker that could noninvasively differentiate benign and malignant causes of hematuria, and identify recurrences prior to their pathologic progression is the objective of this area of research. Currently, a large number of tumor markers exist, each scrutinized in both the laboratory and in clinical trials. Here we present many of the most widely used and tested markers. Background details are provided as to the mechanism of detection of malignant cells, the results of recent trials and future directions of study. Some novel modalities for tumor detection are also presented. The next few years will no doubt bring newer markers and lead to the elimination of others. Studies continue to refine the role of these markers in clinical practice, but their ultimate efficacy will need to be borne out in large-scale clinical trials in a multitude of settings.

Keywords: bladder cancer, diagnosis, surveillance, tumor marker


The National Cancer Institute and the American Cancer Society estimate that approximately 67,000 new cases of bladder cancer were diagnosed in 2007, with approximately 14,000 deaths expected to occur – almost 40 per day [1]. The vast majority of patients are over 55 years of age. The lifetime risk for a man developing this cancer is approximately one in 30, and for a woman, one in 90. Racial differences also exist in bladder cancer epidemiology, with African–Americans having a lower incidence yet more severe forms of the disease compared with Caucasians [2], possibly explained by diagnostic delay in these groups.

For patients with bladder cancer, the extent of the disease at presentation has a significant impact on treatment options and outcomes. At the time of diagnosis, between 16 and 30% of patients will have muscle-invasive bladder cancer. This bodes a much worse prognosis and in many cases the treatment of choice is cystectomy [3,4]. A total of 70–85% of patients will present with nonmuscle-invasive disease of either high or low grade. Often these patients are initially treated endoscopically without the need for radical surgery. Unfortunately for these patients, bladder cancer has the highest recurrence rate of any solid tumor [5]. This often results in extensive follow-up surveillance and testing. Without such close observation, a percentage of patients will recur with muscle-invasive disease, leading to the morbidity of invasive surgery and, possibly, the mortality of metastatic disease [6].

Great strides have been made over the last decades in the treatment of bladder cancer, including the use of chemotherapy [7], novel surgical and bladder-sparing approaches, and intravesical treatments, which have proven very effective in the appropriate clinical scenario. Throughout this time the mainstay of the diagnosis of lower tract urothelial cancer has been cystoscopic and cytologic evaluation of the bladder, most often initiated upon the finding of microscopic or gross hematuria. Given the intermittent nature and specificity of hematuria, the American Urological Association (AUA) currently recommends cytology and cystoscopy (along with upper tract imaging) in high-risk patients, identified as those with a history of smoking, those over 40 years of age, and those with certain chemical exposures among other risk factors [8].

The cost of such testing is extremely high. In 2003, Botteman et al. calculated a cost of almost US$200,000 for treating a patient with bladder cancer for a lifetime. This included initial treatment, follow-up with frequent cystoscopic and laboratory analysis, and subsequent treatments of recurrent and metastatic disease [9,10]. Frequent testing also exposes the patient to other risks, including infection from instrumentation of the lower urinary tract, bleeding from minor trauma and contrast reaction from upper tract imaging.

As a consequence of the costly and invasive nature of cystoscopy and the large number of negative evaluations obtained with voided urine cytology, much research is ongoing into the field of bladder tumor markers. The search is underway for a marker or test that can reliably predict those patients at highest risk of bladder cancer and possibly stratify them further by likelihood of having muscle-invasive disease or high-grade tumors [11,12].

The characteristics of an ideal tumor marker have been examined in detail in the literature [12,13]. A brief overview will be presented here. Of primary concern is the sensitivity or specificity of any marker. A decrease in either of these areas puts the patient at risk: a diminished sensitivity will result in tumors present in the bladder not picked up by the testing, with the associated risk of progression; diminished specificity will result in an unnecessary cystoscopy for a patient without tumor. The marker or test should also be easily and accurately reproducible. Since the goal is to avoid unnecessary cystoscopy, the test should be noninvasive, at most requiring catheterization of the patient but preferably being able to evaluate voided urine samples. As a screening test, an inexpensive cost and ease of use is highly desirable. Yossepowitch et al. and Vriesema et al. also showed that patients desire an accurate test, being unwilling to forgo cystoscopy based on markers than have less than 90% sensitivity [14,15].

Currently, a number of biomarkers exist for evaluation of bladder cancer (Table 1). They can be broken down into a number of subgroups based on the target of their assessment. These groups include soluble antigens, cellular morphology, cell surface antigens and molecular genetic alterations. Soluble antigens, which include BTA-Stat and BTA/TRAK (Polymedco Inc., NY, USA), NMP-22, BCLA-4, survivin, hyaluronic acid–hyaluronidase (HA–HAase), hematuria and urinary bladder check (UBC), can be detected without tumor cells being present in the sample. These tests use proxy markers initially associated with urothelial cells, which can then be assayed in the urine [11]. Cellular morphology is the evaluation of a voided cytology specimen. This requires direct microscopic visualization of the cells by a pathologist. Tests evaluating cell-based antigens include Ucyt, UroVysion (Abbott Labs, IL, USA), DD23, microsatellite DNA analysis (MSA), cytokeratins, and telomere repeat amplification protocol (TRAP) and h-TERT assays (telomerase). These tests detect tumor cell-associated proteins (Ucyt and DD23) or antigens, chromosomal aberrations (UroVysion and MSA) and ribonucleoprotein enzyme assays (telomerase). Finally, a newer area of intense research aims to evaluate genetic aberrations associated with tumor formation and propagation. Recent advances in the use of tissue microarrays and molecular profiling have allowed a large number of cell-cycle- and angiogenesis-related markers to be identified and evaluated.

Table 1.

Comparison of current bladder tumor markers.

Marker Sensitivity Specificity Company Advantages Disadvantages Future directions
Hematuria 47–74% isolated;
90–95% repeat
Inexpensive Poor sensitivity Continued initial
screening test
Cytology 80–90% in high grade;
35–65% in low grade
90–100% Highly specific Requires pathologic evaluation.
Decreased utility in low grade/stage
Continued component
of hematuria work-up
BTA-Stat/BTA-TRAK 8–89% 19–70% Bard Diagnostics,
WA, USA
Inexpensive Poor utility in patients with benign
disease. Variable sensitivity in prior
studies
US FDA approved,
research ongoing
NMP-22 47–100% 60–80% Matritech, MA,
USA
Inexpensive, useful in lesions
not seen on cystoscopy
False-positive results in benign
urologic conditions
US FDA approved,
research ongoing
BladderChek 50–85% 40–90% Matritech, MA,
USA
Inexpensive, may be an
adjunct to cystoscopy
False-positive results in benign
urologic conditions and low sensitivity
US FDA approved,
research ongoing
Hyaluronic
acid-hyaluronidase
83–94% 60–90% High sensitivity, useful in low-
grade and -stage disease
Not in widespread use
Telomerase 9–100% 85–100% High sensitivity throughout
grades/stages
Unstable products of evaluation More research is ongoing
Survivin 64–100% 78–100% Bio-Rad, CA, USA High sensitivity Further research ongoing
FISH 73–87% 90–96% Abbott Labs, IL,
USA
High sensitivity in occult
disease
Criteria for positive test not yet
established, Significantly higher cost
than other markers and cytology
US FDA approved and
research ongoing
Microsatellite DNA
analysis
72–100% 80–100% High sensitivity throughout
stages/grades
Little consensus on markers used in
the test
Further research ongoing
uCyt 40–95% 60–70% Diagnocure,
Quebec, Canada
Sensitive in lower grade
disease
Requires pathologic evaluation for
testing
US FDA approved with
further research ongoing
Cytokeratins 13–79% 36–95% IDL Biotech,
Borlange, Sweden
Some studies show
acceptable sensitivity
Diminished sensitivity and specificity
in many studies
Limited utility based on
current evidence
BLCA-4 90–100% 90–100% High sensitivity and specificity,
Very little confounding by
benign disease
Little consensus on levels for test
evaluation
Further testing ongoing
Soluble Fas Initial results are promising Few studies evaluating this marker Further research is
ongoing

Soluble tumor markers

Hematuria detection

No discussion of bladder cancer can begin without an analysis of hematuria. Hematuria is the most common presenting symptom in bladder cancer, seen in approximately 85% of patients with the disease [16]. The origin of hematuria in bladder cancer is from direct hemorrhage of the tumor, however minor it may be. Hematuria may be either microscopic or gross. The AUA defines microscopic hematuria as more than three red blood cells per high-powered field in at least two of three specimens collected at different times [8]. Gross hematuria refers to the patient being able to directly visualize some degree of blood in the urine [17]. However, hematuria can be caused by a variety of benign conditions, including menses, urinary tract infection, trauma and prostatic hypertrophy, and, as a result, the specificity of hematuria to detect bladder cancer is quite low. Since hematuria is intermittent in nature, sensitivity of hematuria, in one-time voided urine specimen, is variable (47–74%) [18]. Repeat testing has shown a better sensitivity of between 90 and 95%. As a result, many authors advocate cystoscopic examination in the event of a specimen with hematuria even when a repeat examination shows none.

Hematuria screening has been widely discussed recently. For example, in their study of home-screening for hematuria, Messing et al. found that patients were detected at earlier stages of the disease and had a decreased mortality from bladder cancer [19]. The authors felt that such a screening protocol (weekly in their case) is able to detect cancers destined to become muscle invasive and intervention can occur earlier. As a result, in their series no screened patients died of bladder cancer. There was no significant difference in the percentage of patients from the screened and nonscreened group that died of nonbladder cancer causes. Clearly, no absolute conclusions can be made on the basis of this one study; however, screening shows a great deal of promise in selected populations and further testing will be needed to validate these findings.

Another almost universally used noninvasive screening tool for bladder cancer is urine cytology. This refers to the pathological evaluation of a voided urine sample by light microscopy. Many authors point out that urine cytology is not a point-of-care marker as it needs interpretation of the urothelial cells in the specimen by a trained pathologist, along with significantly time-intensive preparation. This notwithstanding, cytology is very useful in certain situations. In high-grade tumors, in particular carcinoma in situ, cytology has been shown to be very specific (close to 100%) and with very few false-negative results. Conversely, the results are less impressive with low-grade disease. Most studies report the sensitivity of cytology in these cases as between 35 and 65% [20,21]. Confounding factors in cytologic evaluation of the urine include tumor load and sampling error. Studies have shown that even in the setting of high-grade tumors, up to 23% of urine samples contain no tumor cells [22]. Most samples evaluated cytologically are voided; however, bladder washing has been shown to increase diagnostic yield of cells [23]. In addition, evidence of atypical cytology may precede cystoscopic confirmation of tumor by varying amounts of time [24]. This can expose the patient to multiple cystoscopies over time and a great deal of anxiety and trepidation.

Given the shortcomings of the widely used methods for non-invasive evaluation of bladder cancer used currently, the search for and evaluation of various bladder tumor markers is intense. Presented here is an overview of the currently tested and used markers, and the state of the latest research. While this is not a complete list, most markers under scrutiny can be found below.

BTA-Stat/BTA-TRAK

These tests detect a bladder tumor antigen now identified as human complement factor H and complement factor H-related protein in the urine [25]. BTA-Stat is a point-of-care test, performed by placing five drops of voided urine samples into the manufacturer-provided test device and qualitatively interpreting the result after 5 min [26]. BTA-TRAK is an ELISA test that measures the levels of these antigens in urine samples. These tests have been used as adjuncts, both in the initial diagnosis of bladder cancer and in surveillance for recurrent disease [27]. In the evaluation of both new and recurrent tumors, Holmang et al. found the sensitivity of detection of this test to be 75 and 54%, respectively. In his study, cytology had a greater than 92% sensitivity and specificity [28]. These results echo the findings of a large number of authors who have published data reporting sensitivities between 8 and 89%, and specificity between 50 and 70% [13,2931]. In healthy individuals, the specificity is improved, however, in patients with a history of benign urologic conditions; for example, in BPH and nephrolithiasis, it drops precipitously [32,33].

Recently, the results of the FinnBladder group’s follow-up studies have been published examining the utility of BTA-Stat in recurrent disease [34]. They note an overall 56% sensitivity and 19.2% specificity for the BTA-Stat test, but much higher sensitivity (85.7%) and specificity (98.3%) for cytology. This is comparable to the cytology values of 85.7% and 98.3%. They found a significant number of false-positive results in patients with a history of Bacille Calmette-Guérin (BCG) instillation and urinary tract infection. The authors conclude that cystoscopy is still required to evaluate recurrent disease, but BTA-Stat may have some utility in the follow-up of patients with low-grade disease.

Summary

BTA-Stat and BTA-TRAK are markers that assay for complement factor H and H-related protein in the urine. It has displayed a lower sensitivity and specificity compared with voided cytology. Its accuracy in high-grade disease is improved; however, the high rate of false-positive results in the setting of nonmalignant urologic conditions makes its clinical utility limited.

NMP-22

NMP-22 is a nuclear matrix protein involved in chromatin distribution during the replication phase of the cell cycle. Expression of this protein is increased in malignant cells, with cell death resulting in release into the urine [35]. Two different tests make use of this tumor marker: the NMP-22 ELISA and BladderChek (Matritech Inc., MA, USA). Various authors have investigated this marker and report sensitivities of 47–100% for the ELISA test. The specificity of the ELISA was reported to be between 60 and 80% [36,37]. These values depend on the concentration of the marker used as the cutoff limit, in order to infer a positive test result [38,39]. Sharma et al. identified six categories of conditions that can result in a false-positive result on the NMP22 test and these include benign inflammatory conditions (e.g., infections), renal or bladder calculi, foreign bodies (stents or nephrostomy tubes), bowel interposition, other genitourinary cancer and instrumentation [40]. The NMP test shows differences in sensitivity when applied to primary and recurrent tumors. Multiple authors have reported that sensitivity decreases markedly during detection of bladder cancer recurrence [41,42]. One hypothesis is that this is related to the reduced tumor burden in recurrent disease versus primary disease. However, Stampfer et al. reported 49% sensitivity and 92% specificity for NMP-22 to detect recurrent bladder tumors. The authors note that NMP-22 was superior to cytology without the interoperator variation seen in a subjective test. Of note, they state that 6.4 units/ml should be the most appropriate concentration used as a cutoff [43].

The BladderChek is a point-of-care test that requires four drops of urine to be placed on the device, with a result returned within 40 min [44]. Fewer studies have evaluated BladderChek given its recent arrival to the market. Witjes et al. reported a sensitivity and specificity for BladderChek of 57.1 and 89.8% compared with 42.9 and 93.2% for cytology, respectively. They conclude that, while convenient and rapid, BladderChek did not enhance urine tests already available [45]. Recently, Grossman et al. reported that the combination of BladderChek and cystoscopy was more effective in detection of bladder cancer recurrence than cystoscopy alone, although, BladderChek showed 49.5% sensitivity, suggesting limited utility of this test as a stand-alone marker [46]. In an earlier study, involving the evaluation of high-risk individuals and of patients exhibiting symptoms often seen in urothelial cancer, Grossman et al. also reported that the BladderChek could significantly increase the accuracy of cystoscopy compared with voided cytology. In that study, BladderChek was referred to as the point-of-care proteomic test that measures the nuclear matrix protein NMP22. The sensitivity of BladderChek (55.7%), although higher than cytology (15.8%), was again low for consideration as a standalone marker. The specificity of NMP-22 was 85.7% compared with 99.2% in cytology [47].

Summary

NMP-22 has shown some promise in detection of recurrent tumors. More studies are needed to reach a consensus on cutoff levels of urinary marker in testing. As with other assays, in the setting of other urologic pathology, the utility of this test is limited. While some studies do report a low sensitivity and specificity for the NMP-22 test, the overall published results are variable and work continues to be done evaluating this marker.

Hyaluronic acid–hyaluronidase test

A glycosaminoglycan that has been shown to promote tumor progression and metastasis, HA and the enzyme that cleaves it into further biologically active products, HAase, have been shown to be promising urinary markers for bladder cancer [48,49]. The HA–HAase test is a combination ELISA-like assay for which authors have reported between 83 and 94% sensitivity and 60–80% specificity [13,5052]. The lower specificity to detect recurrent disease may be related to the test’s ability to detect a bladder tumor nearly 5 months before its clinical detection [49]. One area where the HA–HAase test may show a great deal of promise is in the detection of low-grade and low-stage disease. In this scenario, the sensitivity has been reported as between 75 and 100% [5355]. Given that this group of cancers are the most difficult to detect with conventional methods and yet the most likely to recur, these findings suggest significant strides in noninvasive surveillance in this group. Sensitivity of this test in noninvasive disease (<T2) was found to be 87% and, for invasive disease (≥T2 disease), 100% [53]. Furthermore, false-positive tests have been shown to portend a four- to tenfold increase in the risk of recurrence of bladder cancer [50]. Work recently carried out by Eissa et al. has shown that HYAL1 mRNA detected by reverse transcriptase (RT)-PCR from exfoliated cells has a greater than 90% sensitivity and specificity for tumor detection [56].

Summary

HA–HAase is a promising urinary tumor marker. It is efficacious in detection of low-stage/low-grade disease, an area often underdetected by conventional methods. Further studies will need to be performed in larger cohorts; however, this test has potential for clinical utility in the outpatient setting.

Survivin

Survivin is an anti-apoptotic protein that has been found to be a promoter of bladder cancer progression [5759]. It is measured using a Bio-Dot microfiltration detection system (Bio-Rad, CA, USA). In human trials, survivin has been reported to have a sensitivity of 64–100% and specificity of 78–100% [13,6063]. Recently, Shariat et al. reported that survivin expression was associated with bladder cancer recurrence, disease-specific mortality and all-cause mortality [64]. They concluded that the addition of survivin improved the diagnostic accuracy of bladder cancer without requiring additional invasive testing. Weikert et al. performed a quantitative analysis of survivin mRNA in bladder tumor and urine specimens and found that survivin mRNA levels correlate with pathologic stage. Furthermore, high survivin mRNA expression correlates with decreased disease-free survival. Based on these results, Weikert et al. concluded that survivin mRNA may predict disease-free survival in certain patients, while being more sensitive and specific than cytology in the detection of bladder cancer [65].

Summary

Survivin has been shown to be as sensitive and specific as cytology in the detection of bladder cancer. Some authors currently advocate its use as an adjunct to cytology in the evaluation of primary hematuria or of recurrent disease [66]. More studies are necessary to clearly delineate the utility of this test.

Cytokeratins

Cytokeratins are cytoskeletal intermediate filament proteins found in a large number of cells. Some, such as 8, 18, 19 and 20, have been investigated as tumor markers in bladder cancer after authors had noted increased gene expression in malignant urothelial cells compared with normal cells [67]. Two assays exist based upon cytokeratins 8 and 18: the UBC-Rapid and UBC-ELISA tests. Cytokeratin 19 is detected by the CYFRA 21 – an ELISA that assays for fragments of this filament protein dissolved in the urine. Finally, cytokeratin 20 is detected using RT-PCR, which requires the presence of cells in the specimen. Pariente et al. showed that cytokeratin 19 detection has 96% sensitivity and 74% specificity to detect bladder cancer and is superior to cytology (43% sensitivity). They also noted a significant difference in cytokeratin 19 expression among patients with urothelial cancer compared with those with benign urologic conditions and normal controls [68]. One hindrance to the use of cytokeratin-19 is the unacceptably high false-positive rate seen in patients with a history of BCG instillation [69]. The UBC tests initially returned promising results, being more accurate than NMP and BTA-Stat in some early studies [70,71]. The variability of cutoff values between different studies has hindered further comparison of UBC, as has its low sensitivity in the detection of low-stage tumors. Overall, the sensitivity of UBC varies between 13 and 75%, and for Grade 2 or higher tumors, it is between 35 and 79% [13,70,7274]. Early studies of cytokeratin-20 had demonstrated sensitivity and specificity quite comparable to cytology, but this has been difficult to replicate in follow-up analyses [10,75,76]. Inclusion of HYAL1 (a tumor-derived HAase) mRNA measurement has been shown to improve the accuracy and sensitivity of cytokeratin-20 mRNA measurement to detect bladder cancer, and the performance of the combined marker is superior to the individual markers [56].

Summary

Cytokeratins, a class of proteins found in cellular skeletal components, have shown some promise in early studies of their use in detection of primary and recurrent bladder cancer. Later studies have not replicated these results and more work is needed to elucidate the role of cytokeratins in the management of bladder cancer.

BLCA-4

A nuclear matrix protein, BLCA-4, has drawn much interest based on early reports that it was found throughout bladder epithelium (both in tumorous and nontumorous regions) in patients with bladder cancer but not seen in those without bladder cancer [77,78]. A role for this protein as a marker for the ‘field’ effect of pro-oncogenic urothelium was implied by these studies. Authors have reported over 90% sensitivity and over 90% specificity in the ELISA test [78,79]. These promising results have also been confirmed in studies of patients with chronic genitourinary disease, where BLCA-4 was not correlated with a history of urinary tract infection or chronic catheterization [80]. The authors noted that the cutoff value for their assay and optical density units may need to be adjusted based on clinical scenario.

Summary

BLCA-4 shows promise in early studies with a high sensitivity and specificity. It appears to be more resistant to the confounding effects of benign genitourinary disease. More work is needed to evaluate the most appropriate cutoff levels for this test in the setting of malignant and benign disease of the bladder.

Soluble Fas

Soluble Fas (sFas), is generated by alternative splicing of Fas mRNA [81]. Fas is a cell surface receptor for Fas-ligand (Fas-L). Upon Fas–Fas-L interaction on the cell surface, the cells undergo apoptosis via the extrinsic pathway. sFas blocks Fas–Fas-L interaction and, hence, inhibits apoptosis [82,83]. Svatek et al. found higher levels of sFas associated with higher tumor stage (≥T1) and sFas outperformed NMP22 in terms of sensitivity and specificity. They conclude that urinary sFas is an independent predictor of recurrent disease among patients with a history of noninvasive disease, and that potential exists for its use in a wider surveillance regimen [81].

Summary

sFas, an anti-apoptotic protein, is overexpressed in bladder tumor cells and has been shown to be a predictor of bladder cancer recurrence with the ability to detect higher stage tumors. Current research is ongoing into this marker and it shows promise for future clinical utility.

Matrix metalloproteinases & tissue inhibitor of metalloproteinases

Matrix metalloproteinases (MMPs) are a heterogeneous group of enzymes functioning in the degradation of the extracellular matrix. This degradation is one of the most important steps in the progression of urothelial carcinoma, and a requirement for invasive disease of the tumor. Two commonly investigated MMPs known as gelatinases, MMP-2 and MMP-9, have been evaluated as diagnostic markers for bladder cancer, as has tissue inhibitor of metalloproteinases (TIMP)-2, an inhibitor of MMP activity [84]. It is the imbalance of the normally well-regulated MMP and TIMP system that results in the invasive nature of some tumor cells. Investigations are ongoing into the utility of these markers; however, preliminary data have been promising. Recently, in a study involving 154 bladder cancer (including 136 bilharzial) and 90 nonbladder cancer patients, Eissa et al. showed that MMP-2 and TIMP-2 levels inversely correlate with tumor grade, and MMP-9/TIMP-2 levels correlate with bilharziasis [84]. Conversely, Kanayama et al. reported higher expression of MMP-2 and TIMP-2 in muscle-invasive bladder tumors than in nonmuscle-invasive tumors. Furthermore, higher MMP-2 and TIMP-2 levels correlated with poor prognosis and reduced survival even after cystectomy [85]. Sier et al. reported markedly elevated MMP-2 and MMP-9 activity in the urine of patients with invasive disease compared with those with nonmuscle-invasive bladder cancer [86]. In this study, MMP activity was also found in noninvasive tumors, suggesting a role of these matrix-degrading enzymes in the earlier stages of the disease. Thus, MMP levels probably increase and correlate with invasiveness of bladder tumors, and therefore may have diagnostic and prognostic potential in predicting bladder tumor progression [87,88].

Summary

MMPs and TIMPs have been shown to have increased urinary expression in patients with bladder cancer. There appears to be a relationship between the degree of invasiveness of the tumor and the levels of MMP expressed. Further work elucidating the role of MMPs, especially in predicting invasive and advanced disease, is currently being carried out. Currently, the use of these tests remains limited.

Cell-based antigens as tumor markers

Telomerase

Telomeres are multiply repeated sequences of ribonucleotides that are found at the end of chromosomes [89]. With each successive replication in cell division, these telomeres are lost and, eventually, the lack of telomeres induces chromosomal instability and cellular loss. Telomerase is a ribonucleoprotein that is expressed by tumor cells and that regenerates the telomeres at the end of the replicating chromosomes, essentially creating a cell able to perpetually regenerate [90,91]. The TRAP assay detects telomerase expressed in exfoliated urothelial cells in urine. Also available is a RT-PCR assay that measures a subunit of telomerase, hTERT. Eissa et al. evaluated three methods for detecting telomerase efficiency, namely the TRAP assay, RT-PCR of hTR and hTERT mRNA by quantitative RT PCR. They report that the detection of hTERT in urine improved the sensitivity and specificity in diagnosing urothelial cancer compared with the other methods [92]. Recent work by a variety of authors shows a favorable specificity of between 85% and 100% for telomerase tests. Sensitivity, however, varies quite greatly, between 9 and 100%. This variation is probably related to the poor stability of telomerase in the urine sample [9295]. Bravaccini et al. recently reported that in 50% of study subjects, in whom cytology was either negative or inconclusive, telomerase assay had 88% sensitivity and 65% specificity. They also note a high accuracy from samples containing as few as 100 urothelial cells. They conclude that the major advantage of this test is in the case of negative or inconclusive cytology [96]. A confounding factor in the use of these tests is their high false-positive rates in the setting of inflammation, which is probably related to the presence of lymphocytes in the specimen [97,98].

Summary

Telomerase is a promising marker that may have utility in the setting of negative or nondiagnostic cytology. It does have significant limitations, including false-positive results due to inflammation or infection and the instability of telomerase in the harsh urine environment. Telomerase may ultimately become a useful diagnostic marker; however, the need for very precise handling of specimens must first be addressed.

Fluorescence in situ hybridization

The basis of the FISH test, known in the marketplace as UroVysion, is that certain chromosomal aberrations are seen more commonly than others in bladder cancer [99,100]. The UroVysion test is a multitarget FISH assay for detecting the gain of chromosomes 3, 7 and 17, and the loss of chromosome locus 9p21. The cells labeled with chromosome 3, 7 and 17 centromeric fluorescent probes and a 9p21 locus-specific probe are then evaluated microscopically for positive staining, and a ‘positive’ FISH test is determined by criteria based on numbers of cells and the number of chromosomes (3, 7 and/or 17) gained or lost (9p21). One point of contention in the evaluation of FISH is that there is little widespread agreement on the criteria that distinguish a positive FISH test from a negative one [10]. An area of research where FISH appears to have a significant advantage is in the evaluation and detection of lesions that have not become visually apparent. Skacel et al. found that in nine patients with a negative cystoscopy and biopsy, a positive FISH predicted the appearance of urothelial cancer in eight out of nine (89%). They conclude that in patients with an abnormal FISH, increasingly aggressive surveillance is warranted even in the presence of a negative cystoscopy and biopsy at the time of the FISH [101]. Laudadio et al. recently reported an overall sensitivity of 73% and overall specificity of 65%. They note that, in their series, FISH detected 95% of high-grade cancers, the majority of which were missed by cytology. The detection of low-grade cancers was also improved over cytology, 56% detection rate versus 32%. They conclude that FISH may be a useful tool in the diagnosis of both recurrent and initial tumors [102]. In a number of studies, the sensitivity for FISH was reported to be between 73 and 87%, and specificity between 90 and 96% [103106]. A large number of authors advocate more widespread use of FISH given the ease of use, accuracy of detection of cancer and the ability to detect lesions not yet evident on cystoscopy [102,103]. Currently, the most significant barrier to widespread use of FISH remains the high cost [107].

Summary

The FISH test is rapidly becoming utilized in clinical practice in the diagnosis and surveillance of bladder cancer. Its high sensitivity and specificity and increased accuracy compared with cytology alone may make it useful. Further work must be carried out to reach agreement as to the detection standards for abnormal cells in this test and to resolve issues related to false-positive results, which have been encountered since the use of this test in clinical settings. As one of the most expensive bladder tumor markers, decisions must also be made based on the economic impact of this test upon bladder cancer surveillance.

Microsatellite DNA analysis

MSA is a PCR-based assay for microsatellite DNA, the term for the 2–6 base pair short tandem repeats identified throughout the human genome. A multitude of different locations of microsatellite alterations have been identified in bladder cancer, including chromosomes 4p, 8p, 9p and q, and 11p [108,109]. Two distinct alterations take place, which form the basis of detection in MSA: loss of heterozygosity (LOH) and microsatellite instability. In testing, a large number of microsatellites are used over a number of chromosomes to improve the accuracy of detection. One drawback of this test is that a larger amount of malignant cellular DNA is required to detect LOH compared with microsatellite instability. Over a large number of studies, microsatellite DNA analysis is shown to have a sensitivity of 72–100%, and a specificity of greater than 80% in detecting both primary and recurrent tumors [110114]. As in FISH, MSA may detect tumor recurrence before it becomes clinically evident. In one study, 79% of patients with an LOH were discovered to have a recurrence between 4 and 31 months after transurethral resection. In this study, Shigyo et al. concluded that MSA is a helpful predictor of recurrent disease in which intravesical therapy may be particularly advantageous [115]. The detection of low-grade/low-stage tumors is one area where MSA shows particular promise. As noted above, these tumors are least likely to be detected by conventional methods yet most likely to recur. However, these authors pointed out that microsatellite alterations are reflections of a urothelium with changes that may make tumor development more likely, as opposed to predicting clear tumor formation.

Summary

MSA is a urinary marker with significant potential for clinical utility. It has high sensitivity and specificity, is able to detect low-grade and low-stage tumors well, and is highly predictive of tumor recurrence. A consensus must be reached, however, as to which DNA markers should be assayed for optimal detection with reasonable cost and complexity.

uCyt

uCyt was developed as a test to aid in the detection of malignant cells in cytology. It is an immunofluorescence test that detects carcinoembryonic antigen and sulfated mucin glycoproteins that are overexpressed in bladder cancer cells [116,117]. Multiple studies have reported the sensitivity of the test as between 40 and 95%, with specificity ranging from 60 to 70% [117120]. A number of authors have reported increased sensitivity compared with cytology in the surveillance of recurrent bladder cancer. Pianton et al. studied 694 patients with either gross or microscopic hematuria. They found that sensitivity of uCyt was much higher than that of cytology alone in detection of bladder tumors (80–88% vs 40–47%). However, cytology was more specific in these cases. They conclude that uCyt could be used in place of cytology in the evaluation of gross hematuria [121]. It has been reported that a combination of UCyt and cytology yields best results in the initial diagnosis of bladder cancer, especially low-grade tumors [119,122]. In reports by Lodde et al., sensitivity for G1 tumors increased from 5% with cytology alone to 85% with the addition of uCyt. Similarly, in Ta tumors sensitivity increased from 14 to 86% with uCyt as an adjunct to cytology [123].

Summary

uCyt is a useful addition to cytology in the evaluation of both initial and recurrent bladder tumors. Some of the variability seen in studies of uCyt can be ascribed to the interobserver variability in interpretation of the sample and the experience of the person evaluating specimens.

Oncogenes & DNA methylation

Multiple genetic derangements have been found in bladder cancer. These include presence of mutated oncogenes and methylation of promoters of tumor suppressor genes [124127]. Promising investigations into these markers have yet to have an impact in clinical practice as testing is expensive and difficult to replicate without specialized equipment, and little consensus has been reached on specific markers that could be used in an overall assay. Nonetheless, very recent studies have linked hypermethylation of known tumor suppressor genes, such as the retinoblastoma gene and p14/ARF, with the presence of urothelial malignancy. Control subjects with both normal and benign urologic conditions showed none of these changes [128,129].

Summary

The search for presence of mutated oncogenes and tumor suppressor genes in bladder cancer continues to be a very active area of research. A wide variety of targets are being assessed with positive results. Some of these studies have shown as much as a twofold increase in the accuracy of cytology when combined with these markers. Further investigation must be carried out into the most accurate markers for bladder cancer and how best to detect them. At the current time, multiple disadvantages, including cost and complexity, keep this group of markers from having a clinical impact, although this may change with further research.

Newer molecular markers for prognosis

An area of intense research in the last few years is that of molecular markers for bladder cancer detection. Based on the large number of events necessary for cellular transformation of a normal urothelial cell to a malignant one, researchers are actively evaluating markers from each step in this transformation and their clinical significance. Newer techniques of identification and targeting of molecular signaling pathways have allowed this young field to advance quickly. The areas of most active study include cell-cycle genetic mutations, inhibition of apoptotic pathways, proangiogenic factors and a combination thereof.

As mentioned earlier, mutations in the genes that regulate the cell cycle are frequently seen in bladder carcinoma. Many studies have been focused on p21, p27, pRb, cyclins and p53; however, the results have been mixed up to this point. p53 has been the most studied regulatory target, with authors linking genetic alterations to recurrence and progression in a number of studies [130]. Some recent data also suggest that a higher proportion of deranged p53 correlates with increasingly invasive tumors [131]. To date, however, p53 has not been positively linked to disease prognosis in early trials [132,133]. P27, pRB and cyclins continue to undergo intense scrutiny but results for their prognostic implications are mixed [134].

Apoptosis is a vitally important pathway in cell regulation and alterations in this pathway are reported in most cancers. Survivin and sFas are the two most scrutinized molecules implicated in apoptotic inhibition. Other targets, such as caspase-3, have been evaluated and show promise in early studies in predicting stage and, in some studies, survival [135].

Additionally, angiogenesis is a necessary step in tumor progression. Multiple pro-angiogenic markers have been identified, as has downregulation of angiogenic inhibitors in bladder cancer. These have been linked to disease recurrence and progression. Zu et al. was able to correlate increased VEGF expression with positive lymph node status and poor prognosis [136].

Summary

Molecular markers are a subject of very active research. Many molecular markers have been identified that play a role in cell cycle dysregulation, inhibition of apoptosis and enhancement of angiogenesis in the tumor field. These markers have been shown to correlate with disease severity (including stage and grade), prognosis, recurrence and even survival. Most authors currently advocate a combination of the above markers, given the poor sensitivity and specificity for each taken individually. Many barriers exist for these markers to have clinical significance in the practice setting; however, it seems likely that once better delineated, molecular markers may have a very important role in bladder cancer.

Expert commentary

The field of tumor markers in bladder cancer is rapidly evolving. A number of markers investigated heavily within the last decade have since been discarded, while newer assays continue to be developed and enhanced. The current mainstay of clinical practice remains cystoscopic and cytologic evaluation of a patient presenting with either hematuria or another risk factor for bladder cancer, or surveillance for recurrent disease. The addition of one or more of the markers examined above has been shown in many cases to enhance the detection of primary or recurrent disease when combined with cystoscopy and cytology. Work is being carried out to examine which patients may be followed noninvasively using a combination of the above markers. A more recent shift has been toward genetic studies evaluating the chromosomal makeup of cells in a bladder with or without obvious tumor and identifying those that have an increased risk of undergoing malignant transformation or recurrence. In many cases this may raise further questions, including appropriate surveillance protocols in low- versus high-risk patients, the role of ‘anticipatory testing’ and patient selection for appropriate testing. While studies continue to define the role of biomarkers in bladder cancer, their utility in the clinical setting continues to be debated and, as yet, no consensus has been reached.

Five-year view

Due to the opportunity of testing of any biomarker (soluble antigens or cell-based antigens) noninvasively, using voided urine specimens, bladder cancer will continue to dominate as the cancer of choice for developing new diagnostic tests. The new biomarkers most certainly will be developed based on the results of whole genome-wide array, cDNA microarray, micro-RNA and proteomic analyses. However, for such biomarkers to be adopted in standard clinical practice, more traditional tests (e.g., ELISA, point-of-care device or PCR-based assays) will have to be developed for a single marker or a panel of markers identified by the array data mining studies. Traditionally, most bladder tumor markers have better accuracy than many established tumor markers, such as the prostate-specific antigen (PSA) blood test for prostate cancer. However, bladder tumor markers are not routinely used in the diagnosis of bladder cancer because both urologists and patients are reluctant to forgo cystoscopic evaluation of the bladder due to the fear of missing a tumor via noninvasive testing. Thus, it is difficult to predict whether within next 5 years, bladder tumor markers, collectively, will overcome the issues related to their acceptance in standard clinical practice.

Key issue.

  • Bladder cancer remains a significant cause of mortality and morbidity in the USA.

  • The work-up of bladder cancer is generally precipitated by the finding of hematuria.

  • The current work-up includes cystoscopic evaluation, radiographic evaluation of the upper urinary tract and urine cytology.

  • Extensive work has been performed in the field of biomarkers in bladder cancer.

  • The goals of this work are to maximize the diagnostic yield and minimize the invasive nature of the hematuria work-up.

  • A number of studies have been conducted to evaluate the utility of these biomarkers in the detection of bladder cancer.

  • A wide variability exists in the utility and proposed effectiveness of various markers currently under investigation.

  • At the present time, no marker has become widely used clinically either alone or as an adjunct to cystoscopy in the diagnosis of bladder cancer.

  • Further work, including widespread clinical trials, will need to be performed to fully evaluate these markers.

  • Future tumor marker trends include molecular markers, including genetic mutations in pathways leading to or precluding malignant transformation.

Acknowledgments

Supported by NCI/NIH 5R01 CA-72821–10.

Footnotes

Financial & competing interests disclosure

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

Contributor Information

Samir P Shirodkar, Department of Urology (M-800), Miller School of Medicine University of Miami, P.O. Box 016960, Miami, Florida 33101, USA, Tel.: +1 305 243 3670, Fax: +1 305 243 6893, samirps@yahoo.com.

Vinata B Lokeshwar, Departments of Urology, Cell Biology & Anatomy and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, University of Miami, P.O. Box 016960, Miami, Florida 33101, USA, Tel.: +1 305 243 6321, Fax: +1 305 243 6893, vlokeshw@med.miami.edu.

References

Papers of special note have been highlighted as:

• of interest

•• of considerable interest

  • 1.Hayat MJ, Howlader N, Reichman ME, Edwards BK. Cancer statistics, trends, and multiple primary cancer analyses from the Surveillance, Epidemiology and End Results (SEER) Program. Oncologist. 2007;12:20–37. doi: 10.1634/theoncologist.12-1-20. [DOI] [PubMed] [Google Scholar]
  • 2.Prout GR, Jr, Wesley MN, McCarron PG, et al. Survival experience of black patients and white patients with bladder carcinoma. Cancer. 2004;100(3):621–630. doi: 10.1002/cncr.11942. [DOI] [PubMed] [Google Scholar]
  • 3.Parekh DJ, Bochner BH, Dalbagni G. Superficial and muscle-invasive bladder cancer: principles of management for outcomes assessments. J. Clin. Oncol. 2006;24(35):5519–5527. doi: 10.1200/JCO.2006.08.5431. [DOI] [PubMed] [Google Scholar]
  • 4.Herr HW, Dotan Z, Donat SM, Bajorin DF. Defining optimal therapy for muscle invasive bladder cancer. J. Urol. 2007;177(2):437–443. doi: 10.1016/j.juro.2006.09.027. [DOI] [PubMed] [Google Scholar]
  • 5.Hendricksen K, Witjes J. Current strategies for first and second line intravesical therapy for nonmuscle invasive bladder cancer. Curr. Opin. Urol. 2007;17(5):352–357. doi: 10.1097/MOU.0b013e3281c55f2b. [DOI] [PubMed] [Google Scholar]
  • 6.Sylvester RJ. Natural history, recurrence, and progression in superficial bladder cancer. Sci. World J. 2006;6:2617–2625. doi: 10.1100/tsw.2006.404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.von der Maase H. Current and future perspectives in advanced bladder cancer: is there a new standard? Semin. Oncol. 2002;29:3–14. doi: 10.1053/sonc.2002.30750. [DOI] [PubMed] [Google Scholar]
  • 8.Grossfeld GD, Wolf JS, Jr, Litwan MS, et al. Asymptomatic microscopic hematuria in adults: summary of the AUA best practice policy recommendations. Am. Fam. Physician. 2001;63(6):1145–1154. [PubMed] [Google Scholar]
  • 9.Botteman MF, Pashos CL, Redaelli A, Laskin B, Hauser R. The health economics of bladder cancer: a comprehensive review of the published literature. Pharmacoeconomics. 2003;21(18):1315–1330. doi: 10.1007/BF03262330. [DOI] [PubMed] [Google Scholar]
  • 10.Schrag D, Hsieh LJ, Rabbani F, Bach PB, Herr H, Begg CB. Adherence to surveillance among patients with superficial bladder cancer. J. Natl Cancer Inst. 2003;95(8):588–597. doi: 10.1093/jnci/95.8.588. [DOI] [PubMed] [Google Scholar]
  • 11.Liou LS. Urothelial cancer biomarkers for detection and surveillance. Urology. 2006;67:25–33. doi: 10.1016/j.urology.2006.01.034. [DOI] [PubMed] [Google Scholar]
  • 12. Lokeshwar VB, Habuchi T, Grossman HB, et al. Bladder tumor markers beyond cytology: International Consensus Panel on bladder tumor markers. Urology. 2005;66:35–63. doi: 10.1016/j.urology.2005.08.064. •• Detailed review of the marker process and evaluation.
  • 13.Lokeshwar VB, Selzer MG. Urinary bladder tumor markers. Urol. Oncol. 2006;24:528–537. doi: 10.1016/j.urolonc.2006.07.003. [DOI] [PubMed] [Google Scholar]
  • 14.Yossepowitch O, Herr HW, Donat SM. Use of urinary biomarkers for bladder cancer surveillance: patient perspectives. J. Urol. 2007;177:1277–1282. doi: 10.1016/j.juro.2006.11.066. [DOI] [PubMed] [Google Scholar]
  • 15.Vriesema JL, Povcki MH, Kiemeney LA, Witjes JA. Patient opinion of urinary tests versus flexible urethrocystoscopy in follow-up examination for superficial bladder cancer: utility analysis. Urology. 2000;56:793–797. doi: 10.1016/s0090-4295(00)00777-9. [DOI] [PubMed] [Google Scholar]
  • 16.Pashos CL, Botteman MF, Laskin BL, Redaelli A. Bladder cancer: epidemiology, diagnosis, and management. Cancer Pract. 2002;10:311–322. doi: 10.1046/j.1523-5394.2002.106011.x. [DOI] [PubMed] [Google Scholar]
  • 17.Yun EJ, Meng MV, Carroll PR. Evaluation of the patient with hematuria. Med. Clin. North Am. 2004;88:329–343. doi: 10.1016/S0025-7125(03)00172-X. [DOI] [PubMed] [Google Scholar]
  • 18.Grossfeld GD, Carroll PR. Evaluation of asymptomatic microscopic hematuria. Urol. Clin. North Am. 1998;25(4):661–676. doi: 10.1016/s0094-0143(05)70055-0. [DOI] [PubMed] [Google Scholar]
  • 19.Messing EM, Madeb R, Young T, et al. Long-term outcome of hematuria home screening for bladder cancer in men. Cancer. 2006;107:2173–2179. doi: 10.1002/cncr.22224. [DOI] [PubMed] [Google Scholar]
  • 20.Zein TA, Milad MF. Urine cytology in bladder tumors. Int. Surg. 1991;76:52–54. [PubMed] [Google Scholar]
  • 21.Gil-Salom M, Sánchez MC, Chuan P, Clar F, Santamaría J, García-Sisamón F. Multiple mucosal biopsies and postoperative urinary cytology in patients with bladder cancer. Eur. Urol. 1990;17:281–285. doi: 10.1159/000480134. [DOI] [PubMed] [Google Scholar]
  • 22.Badalament RA. The sensitivity of bladder wash flow cytometry, bladder wash cytology, and voided cytology in the detection of bladder carcinoma. Cancer. 1987;60:1423–1427. doi: 10.1002/1097-0142(19871001)60:7<1423::aid-cncr2820600702>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
  • 23.Konchuba AM, Schellhammer PF, Alexander JP, Wright GL., Jr Flow cytometric study comparing paired bladder washing and voided urine for bladder cancer detection. Urology. 1989;33:89–96. doi: 10.1016/0090-4295(89)90002-2. [DOI] [PubMed] [Google Scholar]
  • 24.Brown FM. Urine cytology. It is still the gold standard for screening? Urol. Clin. North Am. 2000;27:25–37. doi: 10.1016/s0094-0143(05)70231-7. [DOI] [PubMed] [Google Scholar]
  • 25.Kinders R, Jones T, Root R, et al. Complement factor H or a related protein is a marker for transitional cell cancer of the bladder. Clin. Cancer Res. 1998;4:2511–2520. [PubMed] [Google Scholar]
  • 26.Package insert. BTA-Stat Test. [Google Scholar]
  • 27.Pode D, Shapiro A, Wald M, Nativ O, Laufer M, Kaver I. Noninvasive detection of bladder cancer with the BTA stat test. J. Urol. 1999;161:443–446. [PubMed] [Google Scholar]
  • 28.Boman H, Hedelin H, Holmäng S. Four bladder tumor markers have a disappointingly low sensitivity for small size and low grade recurrence. J. Urol. 2002;167:80–83. [PubMed] [Google Scholar]
  • 29.Leyh H, Marberger M, Conort P, et al. Comparison of the BTA stat test with voided urine cytology and bladder wash cytology in the diagnosis and monitoring of bladder cancer. Eur. Urol. 1999;35:52–56. doi: 10.1159/000019819. [DOI] [PubMed] [Google Scholar]
  • 30.Giannopoulos A, Manousakas T, Gounari A, Constantinides C, Choremi-Papadopoulou H, Dimopoulos C. Comparative evaluation of the diagnostic performance of the BTA stat test, NMP22 and urinary bladder cancer antigen for primary and recurrent bladder tumors. J. Urol. 2001;166:470–475. [PubMed] [Google Scholar]
  • 31.Gutiérrez Baños JL, Rebollo Rodrigo MH, Antolín Juárez FM, Martín García B. NMP 22, BTA stat test and cytology in the diagnosis of bladder cancer: a comparative study. Urol. Int. 2001;66:185–190. doi: 10.1159/000056612. [DOI] [PubMed] [Google Scholar]
  • 32.Wald M, Halachmi S, Amiel G, et al. Bladder tumor antigen stat test in nonurothelial malignant urologic conditions. Isr. Med. Assoc. J. 2002;4:174–175. [PubMed] [Google Scholar]
  • 33.Raitanen MP, Kaasinen E, Lukkarinen O, et al. Finnbladder Group. Analysis of false-positive BTA STAT test results in patients followed up for bladder cancer. Urology. 2001;57:680–684. doi: 10.1016/s0090-4295(00)01055-4. [DOI] [PubMed] [Google Scholar]
  • 34.Raitanen MP The FinnBladder Group. The role of BTA Stat Test in follow-up of patients with bladder cancer: results from FinnBladder studies. World J. Urol. 2008;26:45–50. doi: 10.1007/s00345-007-0230-3. [DOI] [PubMed] [Google Scholar]
  • 35.Carpinito GA, Stadler WM, Briggman JV, et al. Urinary nuclear matrix protein as a marker for transitional cell carcinoma of the urinary tract. J. Urol. 1996;156:1280–1285. [PubMed] [Google Scholar]
  • 36.Tsui KH, Chen SM, Wang TM, et al. Comparisons of voided urine cytology, nuclear matrix protein-22 and bladder tumor associated antigen tests for bladder cancer of geriatric male patients in Taiwan, China. Asian J. Androl. 2007;9:711–715. doi: 10.1111/j.1745-7262.2007.00218.x. [DOI] [PubMed] [Google Scholar]
  • 37.Lee MY, Tsou MH, Cheng MH, Chang DS, Yang AL, Ko JS. Clinical application of NMP22 and urinary cytology in patients with hematuria or a history of urothelial carcinoma. World J. Urol. 2000;18:401–405. doi: 10.1007/s003450000124. [DOI] [PubMed] [Google Scholar]
  • 38.Gutiérrez Baños JL, Rebollo Rodrigo MH, Antolín Juárez FM, Martín García B. NMP 22, BTA stat test and cytology in the diagnosis of bladder cancer: a comparative study. Urol. Int. 2001;66:185–190. doi: 10.1159/000056612. [DOI] [PubMed] [Google Scholar]
  • 39.Shariat SF, Marberger MJ, Lotan Y, et al. Variability in the performance of nuclear matrix protein 22 for the detection of bladder cancer. J. Urol. 2006;176:919–926. doi: 10.1016/j.juro.2006.04.017. [DOI] [PubMed] [Google Scholar]
  • 40.Sharma S, Zippe CD, Pandrangi L, Nelson D, Agarwal A. Exclusion criteria enhance the specificity and positive predictive value of NMP22 and BTA stat. J. Urol. 1999;162:53–57. doi: 10.1097/00005392-199907000-00014. [DOI] [PubMed] [Google Scholar]
  • 41.Miyanaga N, Akaza H, Tsukamoto S, et al. Usefulness of urinary NMP22 to detect tumor recurrence of superficial bladder cancer after transurethral resection. Int. J. Clin. Oncol. 2003;8:369–373. doi: 10.1007/s10147-003-0357-1. [DOI] [PubMed] [Google Scholar]
  • 42.Serretta V, Lo Presti D, Vasile P, Gange E, Esposito E, Menozzi I. Urinary NMP22 for the detection of recurrence after transurethral resection of transitional cell carcinoma of the bladder: experience on 137 patients. Urology. 1998;52:793–796. doi: 10.1016/s0090-4295(98)00332-x. [DOI] [PubMed] [Google Scholar]
  • 43.Stampfer DS, Carpinito GA, Rodriguez-Villanueva J, et al. Evaluation of NMP22 in the detection of transitional cell carcinoma of the bladder. J. Urol. 1998;159:394–398. doi: 10.1016/s0022-5347(01)63930-2. [DOI] [PubMed] [Google Scholar]
  • 44.Package insert. NMP22® Test Kit. [Google Scholar]
  • 45.Moonen PM, Kiemeney LA, Witjes JA. Urinary NMP22 BladderChek test in the diagnosis of superficial bladder cancer. Eur. Urol. 2005;48:951–956. doi: 10.1016/j.eururo.2005.09.002. [DOI] [PubMed] [Google Scholar]
  • 46.Grossman HB, Soloway M, Messing E, Katz G, Stein B, Kassabian V, Shen Y. Surveillance for recurrent bladder cancer using a point-of-care proteomic assay. JAMA. 2006;295:299–305. doi: 10.1001/jama.295.3.299. [DOI] [PubMed] [Google Scholar]
  • 47.Grossman HB, Messing E, Soloway M, et al. Detection of bladder cancer using a point-of-care proteomic assay. JAMA. 2005;293(7):810–816. doi: 10.1001/jama.293.7.810. [DOI] [PubMed] [Google Scholar]
  • 48.Lokeshwar VB, Cerwinka WH, Lokeshwar BL. HYAL1 hyaluronidase: a molecular determinant of bladder tumor growth and invasion. Cancer Res. 2005;65:2243–2250. doi: 10.1158/0008-5472.CAN-04-2805. [DOI] [PubMed] [Google Scholar]
  • 49.Lokeshwar VB, Young MJ, Goudarzi G, et al. Identification of bladder tumor-derived hyaluronidase: its similarity to HYAL1. Cancer Res. 1999;59:4464–4470. [PubMed] [Google Scholar]
  • 50.Lokeshwar VB, Schroeder GL, Selzer MG, et al. Bladder tumor markers for monitoring recurrence and screening comparison of hyaluronic acid-hyaluronidase and BTA-Stat tests. Cancer. 2002;95:61–72. doi: 10.1002/cncr.10652. [DOI] [PubMed] [Google Scholar]
  • 51.Hautmann S, Toma M, Lorenzo Gomez MF, et al. Immunocyt and the HA-HAase urine tests for the detection of bladder cancer: a side-by-side comparison. Eur. Urol. 2004;46:466–471. doi: 10.1016/j.eururo.2004.06.006. [DOI] [PubMed] [Google Scholar]
  • 52.Lokeshwar VB, Obek C, Pham HT, et al. Urinary hyaluronic acid and hyaluronidase: markers for bladder cancer detection and evaluation of grade. J. Urol. 2000;163:348–356. doi: 10.1016/s0022-5347(05)68050-0. [DOI] [PubMed] [Google Scholar]
  • 53.Schroeder GL, Lorenzo-Gomez MF, Hautmann SH, Friedrich MG, Ekici S, Huland H, Lokeshwar V. A side by side comparison of cytology and biomarkers for bladder cancer detection. J. Urol. 2004;172:1123–1126. doi: 10.1097/01.ju.0000134347.14643.ab. [DOI] [PubMed] [Google Scholar]
  • 54.Sun Y, He DL, Ma Q, et al. Comparison of seven screening methods in the diagnosis of bladder cancer. Chin. Med. J. (Engl.) 2006;119:1763–1771. [PubMed] [Google Scholar]
  • 55.Lokeshwar VB, Block NL. A sensitive and specific method for detecting bladder cancer and evaluating its grade. Urol. Clin. North Am. 2000;27:53–61. doi: 10.1016/s0094-0143(05)70234-2. [DOI] [PubMed] [Google Scholar]
  • 56.Eissa S, Kassim SK, Labib RA, et al. Detection of bladder carcinoma by combined testing of urine for hyaluronidase and cytokeratin 20 RNAs. Cancer. 2005;103(7):1356–1362. doi: 10.1002/cncr.20902. [DOI] [PubMed] [Google Scholar]
  • 57.Chiou SK, Jones MK, Tarnawski AS. Survivin – an anti-apoptosis protein: its biological roles and implications for cancer and beyond. Med. Sci. Monit. 2003;9:25–29. [PubMed] [Google Scholar]
  • 58.Duffy MJ, O’Donovan N, Brennan DJ, Gallagher WM, Ryan BM. Survivin: a promising tumor biomarker. Cancer Lett. 2007;249:49–60. doi: 10.1016/j.canlet.2006.12.020. [DOI] [PubMed] [Google Scholar]
  • 59. Margulis V, Lotan Y, Shariat SF. Survivin: a promising biomarker for detection and prognosis of bladder cancer. World J. Urol. 2008;26:59–65. doi: 10.1007/s00345-007-0219-y. • Recent study of the biomarker, survivin.
  • 60.Wu Y, Wang G, Wei J, Wen X. Survivin protein expression positively correlated with proliferative activity of cancer cells in bladder cancer. Ind. J. Med. Sci. 2005;59:235–242. [PubMed] [Google Scholar]
  • 61.Smith SD, Wheeler MA, Plescia J, Colberg JW, Weiss RM, Altieri DC. Urine detection of survivin and diagnosis of bladder cancer. JAMA. 2001;285:324–328. doi: 10.1001/jama.285.3.324. [DOI] [PubMed] [Google Scholar]
  • 62.Pu XY, Wang ZP, Chen YR, Wang XH, Wu YL, Wang HP. The value of combined use of survivin, cytokeratin 20 and mucin 7 mRNA for bladder cancer detection in voided urine. J. Cancer Res. Clin. Oncol. 2008;134(6):659–665. doi: 10.1007/s00432-007-0331-9. [DOI] [PubMed] [Google Scholar]
  • 63.Kenney DM, Geschwindt RD, Kary MR, Linic JM, Sardesai NY, Li ZQ. Detection of newly diagnosed bladder cancer, bladder cancer recurrence and bladder cancer in patients with hematuria using quantitative rt-PCR of urinary survivin. Tumour Biol. 2007;28:57–62. doi: 10.1159/000099033. [DOI] [PubMed] [Google Scholar]
  • 64.Shariat SF, Ashfaq R, Karakiewicz PI, Saeedi O, Sagalowsky AI, Lotan Y. Survivin expression is associated with bladder cancer presence, stage, progression and mortality. Cancer. 2007;109:1106–1113. doi: 10.1002/cncr.22521. [DOI] [PubMed] [Google Scholar]
  • 65.Weikert S, Christoph F, Schrader M, Krause H, Miller K, Müller M. Quantitative analysis of survivin mRNA expression in urine and tumor tissue of bladder cancer patients and its potential relevance for disease detection and prognosis. Int. J. Cancer. 2005;116(1):100–104. doi: 10.1002/ijc.21000. [DOI] [PubMed] [Google Scholar]
  • 66.Karam JA, Lotan Y, Ashfaq R, Sagalowsky AI, Shariat SF. Survivin expression in patients with non-muscle-invasive urothelial cell carcinoma of the bladder. Urology. 2007;70:482–486. doi: 10.1016/j.urology.2007.05.009. [DOI] [PubMed] [Google Scholar]
  • 67.Helmy H, Seddek MN, Basta MT, et al. Cytokeratin shedding in urine as a biological marker for bladder cancer: monoclonal antibody-based evaluation. Br. J. Urol. 1991;68:248–253. doi: 10.1111/j.1464-410x.1991.tb15316.x. [DOI] [PubMed] [Google Scholar]
  • 68.Pariente JL, Bordenave L, Michel P, Latapie MJ, Ducassou D, Le Guillou M. Initial evaluation of CYFRA 21-1 diagnostic performances as a urinary marker in bladder transitional cell carcinoma. J. Urol. 1997;158:338–341. [PubMed] [Google Scholar]
  • 69.Nisman B, Barak V, Shapiro A, Golijanin D, Peretz T, Pode D. Evaluation of urine CYFRA 21-1 for the detection of primary and recurrent bladder carcinoma. Cancer. 2002;94:2914–2922. doi: 10.1002/cncr.10565. [DOI] [PubMed] [Google Scholar]
  • 70.Mian C, Lodde M, Haitel A, Vigl EE, Marberger M, Pycha A. Comparison of the monoclonal UBC-ELISA test and the NMP22 ELISA test for the detection of urothelial cell carcinoma of the bladder. Urology. 2000;55:223–226. doi: 10.1016/s0090-4295(99)00383-0. [DOI] [PubMed] [Google Scholar]
  • 71.Mian C, Lodde M, Haitel A, Egarter Vigl E, Marberger M, Pycha A. Comparison of two qualitative assays, the UBC rapid test and the BTA stat test, in the diagnosis of urothelial cell carcinoma of the bladder. Urology. 2000;56:228–231. doi: 10.1016/s0090-4295(00)00664-6. [DOI] [PubMed] [Google Scholar]
  • 72.May M, Hakenberg OW, Gunia S, et al. Comparative diagnostic value of urine cytology, UBC-ELISA, and fluorescence in situ hybridization for detection of transitional cell carcinoma of urinary bladder in routine clinical practice. Urology. 2007;70:449–453. doi: 10.1016/j.urology.2007.04.023. [DOI] [PubMed] [Google Scholar]
  • 73.Kibar Y, Goktas S, Kilic S, Yaman H, Onguru O, Peker AF. Prognostic value of cytology, nuclear matrix protein 22 (NMP22) test, and urinary bladder cancer II (UBC II) test in early recurrent transitional cell carcinoma of the bladder. Ann. Clin. Lab. Sci. 2006;36:31–38. [PubMed] [Google Scholar]
  • 74.Hakenberg OW, Fuessel S, Richter K, et al. Qualitative and quantitative assessment of urinary cytokeratin 8 and 18 fragments compared with voided urine cytology in diagnosis of bladder carcinoma. Urology. 2004;64:1121–1126. doi: 10.1016/j.urology.2004.08.019. [DOI] [PubMed] [Google Scholar]
  • 75.Buchumensky V, Klein A, Zemer R, Kessler OJ, Zimlichman S, Nissenkorn I. Cytokeratin 20, a new marker for early detection of bladder cell carcinoma? J. Urol. 1998;160:1971–1974. [PubMed] [Google Scholar]
  • 76.Retz M, Lehmann J, Amann E, Wullich B, Röder C, Stöckle M. Mucin 7 and cytokeratin 20 as new diagnostic urinary markers for bladder tumor. J. Urol. 2003;169:86–89. doi: 10.1016/S0022-5347(05)64042-6. [DOI] [PubMed] [Google Scholar]
  • 77.Van Le TS, Myers J, Konety BR, Barder T, Getzenberg RH. Functional characterization of the bladder cancer marker, BLCA-4. Clin. Cancer Res. 2004;10:1384–1391. doi: 10.1158/1078-0432.ccr-0455-03. [DOI] [PubMed] [Google Scholar]
  • 78.Konety BR, Nguyen TS, Brenes G, et al. Clinical usefulness of the novel marker BLCA-4 for the detection of bladder cancer. J. Urol. 2000;164:634–639. doi: 10.1097/00005392-200009010-00004. [DOI] [PubMed] [Google Scholar]
  • 79.Konety BR, Nguyen TS, Dhir R, et al. Detection of bladder cancer using a novel nuclear matrix protein, BLCA-4. Clin. Cancer Res. 2000;6:2618–2625. [PubMed] [Google Scholar]
  • 80.Van Le TS, Miller R, Barder T, Babjuk M, Potter DM, Getzenberg RH. Highly specific urine-based marker of bladder cancer. Urology. 2005;66:1256–1260. doi: 10.1016/j.urology.2005.07.010. [DOI] [PubMed] [Google Scholar]
  • 81.Svatek RS, Herman MP, Lotan Y, et al. Soluble Fas - a promising novel urinary marker for the detection of recurrent superficial bladder cancer. Cancer. 2006;106:1701–1707. doi: 10.1002/cncr.21795. [DOI] [PubMed] [Google Scholar]
  • 82.Maas S, Warskulat U, Steinhoff C, et al. Decreased Fas expression in advanced-stage bladder cancer is not related to p53 status. Urology. 2004;63:392–397. doi: 10.1016/j.urology.2003.08.023. [DOI] [PubMed] [Google Scholar]
  • 83.Perabo FG, Mattes RH, Wirger A, et al. Soluble Fas and Fas-ligand in bladder cancer in vitro and in vivo. Urol. Oncol. 2001;6:163–169. doi: 10.1016/s1078-1439(00)00130-7. [DOI] [PubMed] [Google Scholar]
  • 84.Eissa S, Ali-Labib R, Swellam M, Bassiony M, Tash F, El-Zayat TM. Noninvasive diagnosis of bladder cancer by detection of matrix metalloproteinases (MMP-2 and MMP-9) and their inhibitor (TIMP-2) in urine. Eur. Urol. 2007;52(5):1388–1396. doi: 10.1016/j.eururo.2007.04.006. [DOI] [PubMed] [Google Scholar]
  • 85.Kanayama H, Yokota K, Kurokawa Y, Murakami Y, Nishitani M, Kagawa S. Prognostic values of matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-2 expression in bladder cancer. Cancer. 1998;82:1359–1366. [PubMed] [Google Scholar]
  • 86.Sier CF, Casetta G, Verheijen JH, et al. Enhanced urinary gelatinase activities (matrix metalloproteinases 2 and 9) are associated with early-stage bladder carcinoma: a comparison with clinically used tumor markers. Clin. Cancer Res. 2000;6:2333–2340. [PubMed] [Google Scholar]
  • 87.Vasala K, Pääkkö P, Turpeenniemi-Hujanen T. Matrix metalloproteinase-2 immunoreactive protein as a prognostic marker in bladder cancer. Urology. 2003;62(5):952–957. doi: 10.1016/s0090-4295(03)00660-5. [DOI] [PubMed] [Google Scholar]
  • 88.Di Carlo A, Terracciano D, Mariano A, Macchia V. Urinary gelatinase activities (matrix metalloproteinases 2 and 9) in human bladder tumors. Oncol. Rep. 2006;15(5):1321–1326. [PubMed] [Google Scholar]
  • 89.Henderson ER, Larson DD. Telomeres – what’s new at the end? Curr. Opin. Genet. Dev. 1991;1:538–543. doi: 10.1016/s0959-437x(05)80205-9. [DOI] [PubMed] [Google Scholar]
  • 90.Rhyu MS. Telomers, telomerase and immortality. J. Natl Cancer Inst. 1995;87:884–894. doi: 10.1093/jnci/87.12.884. [DOI] [PubMed] [Google Scholar]
  • 91.Shay JW, Wright WE, Werbin H. Loss of telomeric DNA during aging may predispose cells to cancer. Int. J. Oncol. 1993;3:559–563. doi: 10.3892/ijo.3.4.559. [DOI] [PubMed] [Google Scholar]
  • 92.Eissa S, Swellam M, Ali-Labib R, Mansour A, El-Malt O, Tash FM. Detection of telomerase in urine by 3 methods: evaluation of diagnostic accuracy for bladder cancer. J. Urol. 2007;178:1068–1072. doi: 10.1016/j.juro.2007.05.006. [DOI] [PubMed] [Google Scholar]
  • 93.Sanchini MA, Gunelli R, Nanni O, et al. Relevance of urine telomerase in the diagnosis of bladder cancer. JAMA. 2005;294:2052–2056. doi: 10.1001/jama.294.16.2052. [DOI] [PubMed] [Google Scholar]
  • 94.Kinoshita H, Ogawa O, Kakehi Y, et al. Detection of telomerase activity in exfoliated cells in urine from patients with bladder cancer. J. Natl Cancer Inst. 1997;89:724–730. doi: 10.1093/jnci/89.10.724. [DOI] [PubMed] [Google Scholar]
  • 95.Bian W, Xu Z. Combined assay of CYFRA21-1, telomerase and vascular endothelial growth factor in the detection of bladder transitional cell carcinoma. Int. J. Urol. 2007;14:108–111. doi: 10.1111/j.1442-2042.2007.01670.x. [DOI] [PubMed] [Google Scholar]
  • 96.Bravaccini S, Sanchini MA, Granato AM, et al. Urine telomerase activity for the detection of bladder cancer in females. J. Urol. 2007;178:57–61. doi: 10.1016/j.juro.2007.03.025. [DOI] [PubMed] [Google Scholar]
  • 97.Weikert S, Krause H, Wolff I, et al. Quantitative evaluation of telomerase subunits in urine as biomarkers for noninvasive detection of bladder cancer. Int. J. Cancer. 2005;117:274–280. doi: 10.1002/ijc.21168. [DOI] [PubMed] [Google Scholar]
  • 98.Counter CM, Gupta J, Harley CB, Bacchetti S. Telomerase activity in normal leukocytes and in hematologic malignancies. Blood. 1995;85:2315–2320. [PubMed] [Google Scholar]
  • 99.Yokogi H, Wada Y, Moriyama-Gonda N, Igawa M, Ishibe T. Genomic heterogeneity in bladder cancer as detected by fluorescence in situ hybridization. Br. J. Urol. 1996;78:699–703. doi: 10.1046/j.1464-410x.1996.01786.x. [DOI] [PubMed] [Google Scholar]
  • 100.Skacel M, Liou LS, Pettay JD, Tubbs RR. Interphase fluorescence in-situ hybridization in the diagnosis of bladder cancer. Front. Biosci. 2002;7:e27–e32. doi: 10.2741/A902. [DOI] [PubMed] [Google Scholar]
  • 101.Yoder BJ, Skacel M, Hedgepeth R, et al. Reflex UroVysion testing of bladder cancer surveillance patients with equivocal or negative urine cytology: a prospective study with focus on the natural history of anticipatory positive findings. Am. J. Clin. Pathol. 2007;127:295–301. doi: 10.1309/ADJL7E810U1H42BJ. [DOI] [PubMed] [Google Scholar]
  • 102.Laudadio J, Keane TE, Reeves HM, et al. Fluorescence in situ hybridization for detecting transitional cell carcinoma: implications for clinical practice. BJU Int. 2005;96:1280–1285. doi: 10.1111/j.1464-410X.2005.05826.x. [DOI] [PubMed] [Google Scholar]
  • 103.Riesz P, Lotz G, Páska C, et al. Detection of bladder cancer from the urine using fluorescence in situ hybridization technique. Pathol. Oncol. Res. 2007;13:187–194. doi: 10.1007/BF02893498. [DOI] [PubMed] [Google Scholar]
  • 104.Bergman J, Reznichek RC, Rajfer J. Surveillance of patients with bladder carcinoma using fluorescent in-situ hybridization on bladder washings. BJU Int. 2008;101:26–29. doi: 10.1111/j.1464-410X.2007.07183.x. [DOI] [PubMed] [Google Scholar]
  • 105.Marín-Aguilera M, Mengual L, Ribal MJ, et al. Utility of a multiprobe fluorescence in situ hybridization assay in the detection of superficial urothelial bladder cancer. Cancer Genet. Cytogenet. 2007;173:131–135. doi: 10.1016/j.cancergencyto.2006.10.011. [DOI] [PubMed] [Google Scholar]
  • 106.Sarosdy MF, Schellhammer P, Bokinsky G, et al. Clinical evaluation of a multi-target fluorescent in situ hybridization assay for detection of bladder cancer. J. Urol. 2002;168:1950–1954. doi: 10.1016/S0022-5347(05)64270-X. [DOI] [PubMed] [Google Scholar]
  • 107.Krause FS, Rauch A, Schrott KM, Engehausen DG. Clinical decisions for treatment of different staged bladder cancer based on multitarget fluorescence in situ hybridization assays? World J. Urol. 2006;24:418–422. doi: 10.1007/s00345-006-0086-y. [DOI] [PubMed] [Google Scholar]
  • 108.Karran P. Microsatellite instability and DNA mismatch repair in human cancer. Semin. Cancer Biol. 1996;7:15–24. doi: 10.1006/scbi.1996.0003. [DOI] [PubMed] [Google Scholar]
  • 109.von Knobloch R, Hegele A, Brandt H, Olbert P, Heidenreich A, Hofmann R. Serum DNA and urine DNA alterations of urinary transitional cell bladder carcinoma detected by fluorescent microsatellite analysis. Int. J. Cancer. 2001;94:67–72. doi: 10.1002/ijc.1434. [DOI] [PubMed] [Google Scholar]
  • 110.Bartoletti R, Cai T, Dal Canto M, Boddi V, Nesi G, Piazzini M. Multiplex polymerase chain reaction for microsatellite analysis of urine sediment cells: a rapid and inexpensive method for diagnosing and monitoring superficial transitional bladder cell carcinoma. J. Urol. 2006;175:2032–2037. doi: 10.1016/S0022-5347(06)00283-7. [DOI] [PubMed] [Google Scholar]
  • 111.Szarvas T, Kovalszky I, Bedi K, et al. Deletion analysis of tumor and urinary DNA to detect bladder cancer: urine supernatant versus urine sediment. Oncol. Rep. 2007;18:405–409. [PubMed] [Google Scholar]
  • 112.Frigerio S, Padberg BC, Strebel RT, et al. Improved detection of bladder carcinoma cells in voided urine by standardized microsatellite analysis. Int. J. Cancer. 2007;121:329–338. doi: 10.1002/ijc.22690. [DOI] [PubMed] [Google Scholar]
  • 113.Steiner G, Schoenberg MP, Linn JF, Mao L, Sidransky D. Detection of bladder cancer recurrence by microsatellite analysis of urine. Nat. Med. 1997;3:621–624. doi: 10.1038/nm0697-621. [DOI] [PubMed] [Google Scholar]
  • 114.Amira N, Mourah S, Rozet F, et al. Noninvasive molecular detection of bladder cancer recurrence. Int. J. Cancer. 2002;101:293–297. doi: 10.1002/ijc.10561. [DOI] [PubMed] [Google Scholar]
  • 115.Shigyo M, Sugano K, Tobisu K, Tsukamoto T, Sekiya T, Kakizoe T. Molecular followup of newly diagnosed bladder cancer using urine samples. J. Urol. 2001;166:1280–1285. [PubMed] [Google Scholar]
  • 116.Fradet Y, Lockhart C. Performance characteristics of a new monoclonal antibody test for bladder cancer: ImmunoCyt. Can. J. Urol. 1997;4:400–405. [PubMed] [Google Scholar]
  • 117.Mian C, Pycha A, Wiener H, Haitel A, Lodde M, Marberger M. ImmunoCyt: a new tool for detecting transitional cell cancer of the urinary tract. J. Urol. 1999;161:1486–1489. doi: 10.1016/s0022-5347(05)68934-3. [DOI] [PubMed] [Google Scholar]
  • 118.Vriesema JL, Atsma F, Kiemeney LA, Peelen WP, Witjes JA, Schalken JA. Diagnostic efficacy of the ImmunoCyt test to detect superficial bladder cancer recurrence. Urology. 2001;58:367–371. doi: 10.1016/s0090-4295(01)01217-1. [DOI] [PubMed] [Google Scholar]
  • 119.Toma MI, Friedrich MG, Hautmann SH, et al. Comparison of the ImmunoCyt test and urinary cytology with other urine tests in the detection and surveillance of bladder cancer. World J. Urol. 2004;22:145–1479. doi: 10.1007/s00345-003-0390-8. [DOI] [PubMed] [Google Scholar]
  • 120.Têtu B, Tiguert R, Harel F, Fradet Y. ImmunoCyt/uCyt+ improves the sensitivity of urine cytology in patients followed for urothelial carcinoma. Mod. Pathol. 2005;18:83–89. doi: 10.1038/modpathol.3800262. [DOI] [PubMed] [Google Scholar]
  • 121.Schmitz-Dräger BJ, Tirsar LA, Schmitz-Dräger C, et al. Immunocytology in the assessment of patients with asymptomatic hematuria. World J. Urol. 2008;26:31–37. doi: 10.1007/s00345-007-0228-x. [DOI] [PubMed] [Google Scholar]
  • 122.Pianton E, Daniel L, Verriele V, et al. Improved detection of urothelial carcinomas with fluorescence immunocytochemistry (uCyt+ assay) and urinary cytology: results of a French Prospective Multicenter Study. Lab. Invest. 2003;83(6):845–852. doi: 10.1097/01.lab.0000074893.70675.2e. [DOI] [PubMed] [Google Scholar]
  • 123.Lodde M, Mian C, Negri G, et al. Role of uCyt+ in the detection and surveillance of urothelial carcinoma. Urology. 2003;61:243–247. doi: 10.1016/s0090-4295(02)02073-3. [DOI] [PubMed] [Google Scholar]
  • 124.Dalbagni G, Presti J, Reuter V. Genetic alterations in bladder cancer. Lancet. 1993;342:469–471. doi: 10.1016/0140-6736(93)91595-d. [DOI] [PubMed] [Google Scholar]
  • 125.Cordon-Cardo C, Reuter VE. Alterations of tumor suppressor genes in bladder cancer. Semin. Diagn. Pathol. 1997;14:123–132. [PubMed] [Google Scholar]
  • 126.Sánchez-Carbayo M, Cordon-Cardó C. Molecular alterations associated with bladder cancer progression. Semin. Oncol. 2007;34:75–84. doi: 10.1053/j.seminoncol.2006.12.013. [DOI] [PubMed] [Google Scholar]
  • 127.Lu M, Wikman F, Orntoft TF. Impact of alterations affecting the p53 pathway in bladder cancer on clinical outcome, assessed by conventional and array-based methods. Clin. Cancer Res. 2002;8:171–179. [PubMed] [Google Scholar]
  • 128.Mitra AP, Birkhahn M, Cote RJ. p53 and retinoblastoma pathways in bladder cancer. World J. Urol. 2007;25:563–571. doi: 10.1007/s00345-007-0197-0. [DOI] [PubMed] [Google Scholar]
  • 129.Le Frère-Belda MA, Gil Diez de Medina S, Daher A, et al. Profiles of the 2 INK4a gene products, p16 and p14ARF, in human reference urothelium and bladder carcinomas, according to pRb and p53 protein status. Hum. Pathol. 2004;35(7):817–824. doi: 10.1016/j.humpath.2004.01.019. [DOI] [PubMed] [Google Scholar]
  • 130.George B, Datar RH, Wu L, et al. p53 gene and protein status: the role of p53 alterations in predicting outcome in patients with bladder cancer. J. Clin. Oncol. 2007;25(34):5352–5358. doi: 10.1200/JCO.2006.10.4125. [DOI] [PubMed] [Google Scholar]
  • 131.Malats N, Bustos A, Nascimento CM, et al. P53 as a prognostic marker for bladder cancer: a meta-analysis and review. Lancet Oncol. 2005;6:678–686. doi: 10.1016/S1470-2045(05)70315-6. [DOI] [PubMed] [Google Scholar]
  • 132.Moonen PM, van Balken-Ory B, Kiemeney LA, Schalken JA, Witjes JA. Prognostic value of p53 for high risk superficial bladder cancer with long-term followup. J. Urol. 2007;177:80–83. doi: 10.1016/j.juro.2006.08.110. [DOI] [PubMed] [Google Scholar]
  • 133.Dalbagni G, Parekh DJ, Ben-Porat L, Potenzoni M, Herr HW, Reuter VE. Prospective evaluation of p53 as a prognostic marker in T1 transitional cell carcinoma of the bladder. BJU Int. 2007;99:281–285. doi: 10.1111/j.1464-410X.2006.06624.x. [DOI] [PubMed] [Google Scholar]
  • 134. Shariat SF, Ashfaq R, Sagalowsky AI, Lotan Y. Predictive value of cell cycle biomarkers in nonmuscle invasive bladder transitional cell carcinoma. J. Urol. 2007;177:481–487. doi: 10.1016/j.juro.2006.09.038. • Good review of biomarkers, with emphasis on tumor activity.
  • 135. Shariat SF, Karam JA, Lerner SP. Molecular markers in bladder cancer. Curr. Opin. Urol. 2008;18:1–8. doi: 10.1097/MOU.0b013e3282f1c5c1. •• Includes evaluation of newer diagnostic modalities.
  • 136.Zu X, Tang Z, Li Y, Gao N, Ding J, Qi L. Vascular endothelial growth factor-C expression in bladder transitional cell cancer and its relationship to lymph node metastasis. BJU Int. 2006;98:1090–1093. doi: 10.1111/j.1464-410X.2006.06446.x. [DOI] [PubMed] [Google Scholar]

RESOURCES