Role and efficacy of current biomarkers in bladder cancer
Review Article: Oncology: Genitourinary Cancer

Role and efficacy of current biomarkers in bladder cancer

Andrea Giordano, Francesco Soria

Division of Urology, Department of Surgical Sciences, San Giovanni Battista Hospital, University of Studies of Torino, Turin, Italy

Contributions: (I) Conception and design: Both authors; (II) Administrative support: Both authors; (III) Provision of study materials or patients: Both authors; (IV) Collection and assembly of data: Both authors; (V) Data analysis and interpretation: Both authors; (VI) Manuscript writing: Both authors; (VII) Final approval of manuscript: Both authors.

Correspondence to: Francesco Soria. Division of Urology, Department of Surgical Sciences, San Giovanni Battista Hospital, University of Studies of Torino, C.so Bramante 88, 10126 Turin, Italy. Email: francesco.soria@unito.it.

Abstract: In order to overcome the current limitations in the diagnosis, therapy and follow-up of bladder cancer (BCa), the detection of biomarkers in urine, tissue and blood has been proposed as essential components of the precision medicine. The introduction of these modern molecular tests may help to identify disease earlier, to risk-stratify patients, to improve the prediction of oncological outcomes and to optimize target therapies. On the other hand, the main limitation for their spread in clinical practice is the lack of evidence of a meaningful improvement in clinical decision-making due to their unsatisfactory performances. Recent Literature made a great effort with the aim to describe novel biomarkers and their potential role, but this jigsaw puzzle scenario makes difficult the orienteering into their correct uses and indications. We reviewed the current state of the art and the performance of available and most promising biomarkers. Despite some good results, especially regarding the improvement of sensitivity compared to urinary cytology in the diagnostic setting, the use of these molecules has not been yet introduced into daily clinical practice by international guidelines, because of both the difficulty in identifying the correct scenario of use and the lack of high-quality prospective trials, with a subsequent low level of evidence. Further prospective investigations and large international collaborations are needed to define the role of these promising biomarkers.

Keywords: Urinary biomarkers; tissue-based biomarkers; non-muscle invasive bladder cancer (NMIBC); response to treatment


Received: 03 January 2020; Accepted: 06 February 2020; Published: 25 March 2020.

doi: 10.21037/amj.2020.02.06


Introduction

Despite continuous research and improvements of available tools over the last years, above all endoscopic and imaging ones, the cornerstones for the diagnosis and surveillance of bladder cancer (BCa) have not changed and are still represented by cystoscopy and urine cytology (1,2). Cystoscopy is a routinely used exam performed almost in every urologic center and, due to its high sensitivity and specificity (2), is still considered the gold standard for BCa detection. However, this is not a side-effect free procedure: due to its invasiveness, it still represents an important cause of patients’ discomfort that negatively affects patients’ quality of life, and a costly procedure that severely impacts on the health-care system (3). Voided urinary cytology is a non-invasive test used in combination with cystoscopy in the diagnosis and follow up of high-grade tumors. Actually, urinary cytology has an overall high specificity and a good sensitivity for high-grade tumors, while its accuracy dramatically decreases in low-grade tumors (1). Recent innovations aimed to fill this lack by following the direction drawn by modern medical research towards the era of “precision medicine”, which consists in taking into account patient’s and tumor’s variability, thereby allowing the personalization of therapy. In this scenario biomarkers play a key role. Precisely, biomarkers may potentially improve patients’ risk stratification by increasing the accuracy of available diagnostic tools and by predicting oncological outcomes and response to therapies. Many biomarkers have been studied over the last years in different clinical scenarios, such as for screening, surveillance and follow-up. However, despite significant efforts dedicated to a better understanding of their principles and roles, none of them is, to date, endorsed by international guidelines, and therefore used in clinical routine (4). The main limitation for their spread in clinical practice is the lack of evidence of a meaningful improvement in clinical decision-making due to their unsatisfactory performances. This could be the consequence of multiple factors. As said before, biomarkers could be theoretically applied to different clinical scenarios such as BCa diagnosis, screening, risk stratification, prognostication of outcomes, and prediction of response to therapies. Obviously, different abilities are required for each setting: for example, to simplify, a biomarker used in a screening setting should have a high specificity in order to avoid unnecessary investigations in healthy patients, while during the follow-up of non-muscle invasive bladder cancer (NMIBC), a high sensitivity should be required, aiming to reduce the risk of missing a clinically significant tumor. Therefore, the first challenge in the detection of a reliable marker is to identify the correct scenario of application based on its accuracy. Moreover, as described by Goebell et al. (5), a complex process of validation should be followed by biomarkers to be introduced into clinical practice (Table 1): they have to be analyzed into pre-clinical exploratory studies, clinical assay development and validation studies, small clinical retrospective studies, external validation in larger cohorts, prospective clinical trials and post-approval studies. This rigorous validation process is rarely observed, and this deficiency may be identified as the second cause for the presence of numerous promising markers that have never been subsequently reproduced or validated: most of biomarkers either do not follow or fall behind this strict validation path. Despite all these limitations, and the difficulty to find orientation surfing the big wave of biomarkers in literature, their hypothetical role still remains interesting and potentially useful in the management of BCa. The aim of this review is to elucidate the current state of the art of the most promising biomarkers which may entry in clinical practice in the next future.

Table 1

Proposed structured approach to systematic validation of biomarkers

Level Sample details/bias Target Main features
Preclinical testing In vitro or animal models (an appropriate model may be lacking) A hypothesis-generating step
Phase 0 On a small group of patients (sample of population is limited) Development of clinically reproducible preliminary assays Do not take into account the potential benefit
Phase 1 On a small group of patients (defined from biomarker profile, sample of population is limited) Determination of the benefit (define the marker, establish the prediction rules, and determine the assay cut-off points)
Phase 2 On a relevant, representative target population Independent validation of the accuracy of the assay (reproducibility and robustness of the assay, determination of reference) Retrospective design, internal validation
Phase 3 On large patient populations (multi-institutional collaboration) To yield the sensitivity and specificity of the biomarker, to show an earning in clinical decision-making Randomized trial, external validation
Phase 4 Define if burden of disease is reduced by the biomarker Post-approval reporting and testing for other disease processes or disease stages

Different biomarkers for different clinical scenarios: screening, risk stratification and surveillance

For the detection of BCa and its first diagnosis, two main clinical scenarios can be identified: for screening purpose in high-risk population, such as heavy smokers and workers with occupational exposure to agents known to increase the risk of BCa, and for risk-stratification of patients with asymptomatic microhematuria (AMH). Because of the low prevalence of disease even in high-risk categories (6), in these scenarios the ideal urinary marker should have a high positive predictive value (PPV) and high specificity. However, data from recent trials indicate that even in patients with high-risk of developing BCa the incidence is so low [from 0.1% to 3.3% in heavy smokers and from 0.01% to 8% in exposed workers (7,8)] that systematic screening cannot be clearly recommended. The second clinical scenario is represented by the risk-stratification of patients with AMH. In this population previous studies have suggested a significant risk of BCa [approximately 10% (9)], thus making cystoscopic workup mandatory. The critical point in this setting is the high prevalence of AMH in the adult population, from 10% to 18%, yet only 2% of which have BCa. Therefore, the introduction of urinary biomarkers for risk-stratification purposes maintains an increasing appeal, in order to identify those patients in whom cystoscopy, an exam not side-effect free as we already discussed, can be avoided. However, only limited prospective data on the use of molecular urinary biomarkers in patients presenting with AMH are available, and to date no adequate evidence is disposable. Another potential role of biomarkers in BCa consists in their application during surveillance and follow-up of patients with previous NMIBC. This may be perceived in two major usages: as substitute to cystoscopy for low/intermediate risk NMIBC patients or as an adjunct to the endoscopic workup for high-risk NMIBC patients.


Urinary biomarkers overview

Immunocytology (ImmunoCyt and uCyt)

Urine cytology is a well-studied test approved and strongly recommended by international guidelines which has in its limited sensitivity for low grade tumors and in its variability of interpretation the main points of weakness (1). In order to overcome the low sensitivity, ImmunoCyt and uCyt assays were developed. These immunocytological tests are based upon microscopic detection of high molecular weight form of carcinoembryonic antigen (CEA) and BCa associated-mucins contain fluorescein-labelled monoclonal antibodies M344 and LDQ10 (against mucin glycoproteins) and Texas red-linked antibody 19A211 (against CEA). The strength of these assays is represented by the higher sensitivity for low grade tumors compared to conventional urine cytology. On average, the detection rate for low-grade tumors was 75%, and sensitivity for G2 and high-grade tumors was approximately 85%. Overall specificity was 75% (10-12). Conversely, costs are higher, mostly due to lab equipment, long time of processing and reading the specimens and the high experience needed for interpretation of results (13). Reported confounding factors for the uCyt TM assay seem to be benign prostatic enlargement, hematuria, urolithiasis, and inflammatory conditions but recent reports suggest that the impact of these conditions on test performance seems to be limited (14).

UroVysion FISH

The UroVysion multicolor FISH test provides the detection of chromosomal aberrations (probes for chromosomes 3, 7 and 17 and the 9p21 locus) associated to BCa. This assay is approved by the Food and Drug Administration (FDA) for diagnosis and surveillance of BCa. Many cut-off levels for the positivity of the test have been introduced (15), but in general a minimum of 25 morphologically abnormal cells are analyzed and it is considered positive when a minimum of 4 cells have a gain of two or more of the aforesaid chromosomes or 12 of more cells have a loss of the 2 copies of locus 9p21. Schlomer et al. suggested to use the UroVysion FISH test when urine cytology provides dubious findings (16). The main limitations for the use of UroVysion are related to the high costs, the need of specialized laboratories for adequate interpretation, the significative rate of false-positive and non-informative findings in approximately 10% of the cases (17). Despite the broad range of accuracy found in literature for UroVysion FISH, probably affected by different experience of investigators and cut-off values, sensitivity seem to be set from 70% to 80% and specificity approximately around 80% (18-20). Part of the relatively high rate of false-positive could be explained in the follow-up scenario as anticipatory positive results, which means that a premalignant molecular change is detected by the assay even if no macroscopic alterations can be found endoscopically: indeed, two studies showed that 85% and 88% of patients with a false-positive test finally developed a bladder recurrence within 12 months. However, the interpretation of the anticipatory positive result still remains uncertain due to the high recurrence rate of BCa (21).

Nuclear matrix proteins (NMPs)

NMPs are important biomarkers and antigens of BCa. They contribute to the structure of the nucleus and are fundamental for supporting nuclear shape. One member of this jigsaw puzzle of NMPs is NMP22, which is much more prevalent in malignant urothelial cells compared to normal ones, because of their higher rate of apoptosis. Both qualitative point-of-care tests and quantitative ELISA tests exist, and this assay is FDA approved. According to several meta-analyses, the benefit of these protein markers compared to urine cytology seems to be a higher sensitivity both for low (median of 50% vs. 27%) and high (median of 83% vs. 78%) grade tumors (22-24). Tumor volume, in terms of numbers of lesions or size, seems to reflect the positivity of the test, as described by Poulakis et al. who found sensitivities of 79%, 90% and 97% in patients with 1, 2–3 and >3 lesions, respectively (25). The main limitation for the NMP22 application in clinical practice is its lower specificity compared to urine cytology. Indeed, urological pathologic conditions, such as inflammation, infection, stones, hematuria and also diagnostic and therapeutic procedures such as endoscopic ones, could affect the assay, thus causing false-positive results. This is due by the multiple causes of apoptosis and subsequent release of NMPs in urine. Sharma et al. tried to exclude these clinical categories, with and improvement of specificity and PPV of NMP22 (95.6%, 87.5%). As for FISH, also for NMP22 was described an anticipatory positive result (26). A recent prospective trial on 1,303 patients identified NMP22 as the strongest predictor of BCa, higher than urine cytology, and its addition to a predictive model for BCa recurrence, significantly improved the accuracy of the model (27).

BTA test

Another test approved by FDA in adjunct to cystoscopy is BTA (Bladder Tumor Antigen), both in its two newer versions, BTA stat and TRAK (which differ from the original in detecting different proteins) (28). The qualitative point of care and the quantitative Elisa test detect the human complement factor H related protein (that is thought to help cells to evade the host immune system) and complement factor H in urine samples. BTA stat is a cheap, single-step, easy to reproduce, fast to perform test and requires minimal experience from the laboratory staff (29). BTA TRAK needs instead a more sophisticated technology and more time to be performed. Manufacturer recommended the cut-off of 14 U/mL for the positivity of the test (28). BTA stat and BTA TRAK are not approved by FDA for diagnostic and screening purpose, but in surveillance setting as adjunct to cystoscopy. Glas et al. in their meta-analysis, helped our understanding about BTA assays by reporting a sensitivity of 70% (95% CI: 66–74) and a specificity of 75% (95% CI: 64–84) for BTA stat, while for BTA TRAK test sensitivity and specificity were 66% (95% CI: 62–71) and 65% (95% CI: 45–81), respectively. Again, findings may be affected by concomitant urological pathologies and procedures. BTA tests in not recommended alone in the detection of BCa (30).


Urinary biomarkers for the screening of high-risk population

The low prevalence of BCa even in categories considered at high-risk of developing urothelial tumor together with the low cancer-specific mortality (CSM) and the incidence of low-grade tumors affect the effectiveness of screening programs (7,8). The low prevalence of disease inevitably affects biomarkers’ PPV and negative predictive value (NPV) (Table 2). Lotan et al. tested NMP22 in high-risk population (1,175 men and 327 women with at least 10 years of smoking or 15 years of exposure to carcinogenic substances). Eighty-five (5.7%) subjects had a positive NMP22. Of these, 69 patients underwent further evaluation, and only 3 (3.5%) had a BCa diagnosis (all low-grade tumors). After 12 months, 2 of 1,309 (0.15%) participants developed low-grade BCa (24). Steiner et al. studied 183 heavy smokers and reported urine cytology, NMP22 and FISH levels. Forty-one percent of subjects had at least one positive biomarker, but only 5 cases of tumors and 12 cases of precancerous lesions were recorded (23). To date, no urinary biomarkers has met the criteria for screening purpose and, actually, guidelines do not advise their use, even if future large controlled trials may help to define their potential role in this setting.

Table 2

Summary of studies assessing the role of biomarkers for screening in high-risk populations

Study Number of patients Test Study population Results
Lotan et al. (24) 1,502 NMP22 Smokers (≥10 years), occupational exposure Eighty-five positive tests (8.5%), of which 69 cystoscopies were performed, 3 with abnormal findings. After a median F-U of 78.4 months no case of MIBC in subpopulation of 925 patients
Steiner et al. (23) 183 Dipstick, NMP22, cytology, FISH Smokers (≥40 pack years) Seventy-five patients with at least one positive marker (40.9%), at least one marker positive in all tumor cases
Pesch et al. (8) 1,609 Dipstick, NMP22, FISH, cytology Workers with occupational exposure Four hundred and ninety-three positive tests, 8 cases detected by cytology. 20 abnormal findings
Bonberg et al. (31) 1,609 UroVysion Workers with occupational exposure Nine of 21 BCa detected in 20 cases had UroVysion test +, including 7 HG carcinomas and 7 overlapping results with cytology
Banek et al. (32) 1,609 FISH Workers with occupational exposure Sixteen incidental BCa detected and 5 recurrent tumors in 20 cases. FISH was positive in 9 BCa cases of which 7 were HG. FISH overlapped with cytology in 7 cases. Sensitivity was 45.0% and PPV 16.4% in all and 53.85% and 13.21% in HG tumors. Specificity and NPV were 96.97% and 99.26% in all BCa
Huber et al. (33) 1,772 NMP22 Workers with occupational exposure Higher NMP22 concentrations in 224 patients, which correctly predicted BCa in 6 cases (sensitivity 97.2%, specificity 28.5%; NPV 99.04%, PPV 12.24%)

MIBC, muscle invasive bladder cancer; BCa, bladder cancer; PPV, positive predictive value; NPV, negative predictive value; NMP, nuclear matrix protein.


Urinary biomarkers for the risk-stratification of asymptomatic microscopic hematuria

While painless gross hematuria is generally considered an indication for cystoscopy, the management of painless microhematuria is controversial and differs widely. However, the need of stratifying patients by the risk of presenting BCa is univocal. As said before, the lack of prospective studies and the not distinction of gross hematuria and AMH in many studies represent a big obstacle for studies’ quality (Table 3). Cha et al. performed a retrospective analysis of 1,182 patients by testing a predictive model including cytology, imaging, cystoscopy, and immunocytology, and reported an accuracy for BCa detection of 90.8%. Within this nomogram, a positive immunocytology was the strongest predictor of presence of BCa (34). Lotan et al. tested another nomogram including NMP22, and predictive accuracy decreased to 80.2% (35). Therefore, even if prospective trials are lacking, retrospective experience seems to suggest that some biomarkers could help risk-stratifying patients with AMH.

Table 3

Summary of studies assessing the role of biomarkers in patients presenting hematuria

Study Number of patients Test Study population Results
Cha et al. (34) 1182 Immunocytology Hematuria (68% AMH) Construction of a nomogram with 90.8% of accuracy of prediction of BCa (immunocytology was the strongest predictor)
Lotan et al. (35) 381 NMP22 Hematuria Construction of a nomogram with 80.2 % of accuracy of prediction of BCa
Beukers et al. (36) 169 Methylation analysis of OSR1, SIM2, OTX1, MEIS1 and ONECUT2 Hematuria (86 AMH, 83 gross hematuria) Construction of a nomogram with sensitivity of 85% and specificity of 87% for the prediction of BCa (performance of the model was better in patients with gross hematuria compared AMH)
Todenhöfer et al. (37) 2,365 Urovysion, NMP22, immunocytology AMH The extent of AMH significantly influences the performance of noninvasive urine markers, underlining the relevance of the grade of AMH for the interpretation of tests
Lucca et al. (38) 227 HDAC9, HDAC3, tRNA methyltransferase1, DNA methyltransferase 1 AMH The addition of HDAC3 to a base risk factor model improved its accuracy by 1.4%
Davidson et al. (39) 571 CxbT Hematuria High NPV was found for the clinical pathway composed by imaging and CxbT, significantly reducing the need of cystoscopy

BCa, bladder cancer; NPV, negative predictive value; NMP, nuclear matrix protein; AMH, asymptomatic microhematuria.


Urinary biomarkers for the surveillance of NMIBC

Some guidelines currently recommended the use of biomarkers in adjunct to cystoscopy, but not its replacement. Awareness of markers’ positivity seems to improve the detection rate of BCa recurrence, as described in a prospective, single-blind, randomized multicenter trial, in which 448 patients were included. When urologists performing cystoscopy were informed about the results of urinary biomarkers, 42 recurrences were detected in 131 cystoscopies, whereas urologists that were not aware of the positivity of the marker, observed only 6 recurrences in 120 cases (40). Shariat et al. described a possible relationship between biomarkers’ positivity and tumor aggressiveness at transurethral resection of the bladder (TURBT). Positive cytology and NMP22 were associated with a 33-fold risk of a ≥ pT1 tumor and a 21-fold risk of a G3 tumor (41). A multicenter study analyzed and compared the sensitivity and specificity of main urine biomarkers for the diagnosis of BCa recurrence (NMP22, Cxbladder Detect, UroVysion FISH). Cxbladder Monitor (designed to analyze the expression of five genes, CDC2, HOXA13, MDK, IGFBP5 and CXCR2) resulted highly sensitive for the diagnosis of recurrent BCa and with a low rate of false negative results and, therefore, it outperformed the remaining markers (42). Based on these promising findings, Waitemata Disctrict Health Board in New Zealand (one of publicly funded health care providers) accepted Cxbladder Monitor as a substitute of cystoscopy in all low-risk NMIBC patients. The use of a panel of biomarkers, instead of only one of them, has been studied, and it has been shown that this approach, including voided urine cytology and other markers, does not improve the predictive value of a test that based on only one biomarker (high grade disease was diagnosed with both tests simultaneously) (43,44). Another possible approach is related to the use of biomarkers as reflex test (45). Instead of a simultaneous analysis of a panel of biomarkers, for those patients with negative result of one test, subsequent and highly sensitive markers were added, thus leading to an increased accuracy of follow up. The subsequent combination of two among the four widely adopted markers (cytology, immunocytology, FISH and NMP22) resulted in a sensitivity and NPV of 89.8% (ImmunoCyt + NMP22) and 92.1% (FISH + ImmunoCyt), respectively. If urine cytology is supplemented with any of the four tests, corresponding sensitivity and NPV are 86.7% (NMP22) and 91.3% (immunocytology), respectively. Adding FISH to conventional urine cytology is associated with a sensitivity of 80.5% (94.0% for high-risk tumors) and a NPV of 90.1% (98.8% for high-risk tumors). In conclusion, while European Association of Urology (EAU) and American Urological Associations (AUA) do not recommend urinary markers in the routine surveillance of patients with NMIBC (40,46), AUA Guidelines assess their use to define response to intravesical bacillus Calmette-Guérin (BCG) (UroVysion FISH) and adjudicate equivocal cytology (UroVysion FISH and ImmunoCyt) (47). Serial measurements of UroVysion FISH in patients treated with BCG revealed that abnormal results at baseline and during follow-up are significantly associated with both cancer recurrence and progression (cancer progressed after 2 years in half of those with positive FISH test and in only 3% of those who had normal result). The authors concluded that patients with a negative cystoscopy with abnormal FISH findings may be good candidates for clinical trials due their worse prognosis (48). FISH and ImmunoCyt were also investigated in patients with atypical cytology (49). UroVysion FISH has been shown to have 100% sensitivity and 100% NPV in those with negative cystoscopy, while ImmunoCyt was found to have 73% sensitivity in detecting recurrent bladder tumor in patients with atypical cytology with corresponding NPV of 80%. A summary of diagnostic performance of biomarkers in the detection of NMIBC can be observed in Table 4.

Table 4

Summary of diagnostic performance of biomarkers in the follow-up of NMIBC

Study Number of patients Sens (%) Spec (%) PPV (%) NPV (%)
VUC
   Raitanen et al. (50) 445 19.2 98.3 82.8 73.6
   Casetta et al. (51) 102 70.0 75.0 85.9 53.3
   Raitanen et al. (52) 510 19.2 85.7
   Nisman et al. (53) 154 61.9 96.2 28.6 94.3
   Babjuk et al. (54) 88 19.8 99.0 89.5 74.9
   Hosseini et al. (55) 144 44.2 83.7 60.5 72.6
   Messing et al. (56) 327 23.0 93.0
NMP22
   Casetta et al. (51) 102 64.0 64.0 78.3 45.2
   Serretta et al. (57) 137 71.5 61.0 44.7 82.8
   Horstmann et al. (58) 221 68.0 49.0 57.0 60.0
   Shariat et al. (59) 2,871 57.0 81.0 64.0 77.0
   Grossman et al. (60) 668 49.5 87.3
   Gupta et al. (61) 145 85.7 77.5 70.6 89.6
BTA stat
   Raitanen et al. (50) 445 56.0 85.7 63.1 81.7
   Pode et al. (43) 162 73.7 67.6 69.8 (accuracy)
   Raitanen et al. (52) 510 56.0 85.7 42.5 81.4
   Lokeshwar et al. (62) 70 60.7 74.1 87.9 37.7
BTA TRAK
   Casetta et al. (51) 102 60.0 60.0 75.9 39.6
   Babjuk et al. (54) 88 38.5–53.8 83.9–88.6 58.6 81.1
   Miyanaga et al. (63) 57 9.3 86.2 23.5 67.5
ImmunoCyt
   Vriesema et al. (64) 104 50.0 73.0 39.0 81.0
   Sullivan et al. (65) 100 76.0 63.0 43.0 88.0
   Horstmann et al. (58) 221 73.0 72.0 72.0 74.0
   Messing et al. (56) 327 81.0 75.0 38.0 95.0
FISH
   Sarosdy et al. (66) 176 71.0 65.8 53.0 80.7
   Horstmann et al. (58) 221 76.0 63.0 68.0 71.0
   Youssef et al. (67) 123 23.5 94.3 40.0 88.5
Cxbladder monitor
   Lotan et al. (68) 803 91.0 96.0
   Kavalieris et al. (69) 763 93.0 97.0

NMIBC, non-muscle invasive bladder cancer; VUC, voided urine cytology; PPV, positive predictive value; NPV, negative predictive value; NMP, nuclear matrix protein.


Urinary biomarkers for the prediction of response to intravesical BCG immunotherapy

As already anticipated, urinary biomarkers have been analyzed also as predictors of response to intravesical BCG (Table 5). In a systematic review, Kamat et al. concluded that, beside the standard clinicopathologic features (which still represent the most effective predictors of BCG response), single biomarkers, such as tumor p53 and urinary interleukin-2 (IL-2) expression, have limited success in predicting BCG response, while more comprehensive molecular panels (mostly urinary cytokines), have a robust correlation with response, assuming a potential future role for them in stratifying patients for outcomes after BCG treatment (48). In another analysis including 130 patients, a nomogram (CyPRIT) constructed using urinary levels of nine inducible cytokines (IL-2, IL-6, IL-8, IL-18, IL-1ra, TRAIL, IFN-γ, IL-12, and TNF-α) predicted the likelihood of recurrence with 85.5% accuracy (95% CI: 77.9–93.1%). This cytokine panel and nomogram was described as having a potential for identifying patients at risk of tumor recurrence during BCG treatment to guide modification of the dose and duration of BCG immunotherapy (76). To date current evidences are still inadequate to include biomarkers in clinical practice for the prediction of response to intravesical BCG, even if future randomized controlled trials could overcome this obstacle and so revealing their true potential.

Table 5

Summary of studies assessing the role of biomarkers for the prediction of response to intravesical BCG immunotherapy

Study Number of patients Test Results
Esuvaranathan et al. (70) 47 Rb Underexpression of Rb may be predictive of non-response to BCG
Lima et al. (71) 94 Sialyl-Tn and sialyl-6-T High expression leads to effective BCG treatment
Langle et al. (72) 11 FGFR3 Down regulation of FGFR3 predicts good response to BCG
Leibovici et al. (73) 519 IL-6 High levels related to tumor recurrence after BCG
Zhao et al. (74) 224 Glutathione peroxidase 1 Wild type had higher recurrence after BCG
Lima et al. (75) 204 Multiple genes Multi-gene panel as predictor of outcomes after BCG

BCG, bacillus Calmette-Guérin.


Prognostic and predictive tissue-based biomarkers in NMIBC

Prediction of oncological outcomes, such as recurrence-free survival (RFS) and progression-free survival (PFS), as well as the prediction of response to intravesical BCG are the most important aims of tissue-based biomarkers in NMIBC. The predictive accuracy of currently existing risk stratification systems may be improved by their application and, therefore, could be used to develop personalized treatment and follow up schemes based on different risk of recurrence and progression. To date, biomarkers associated with pathways important for tumor growth and spread have been evaluated, including cycle-cell regulators, angiogenesis, apoptosis, signaling proteins and hormonal biomarkers.

Prognostic tissue-based biomarkers of recurrence

In general, tissue biomarkers reflect tumor aggressiveness and invasiveness and, therefore, they may be indicators of progression of disease rather than tumor recurrence. Numerous markers have been analyzed with heterogeneous results, and p53 was the most extensively studied tissue-based biomarker (Table 6). p53 is the most common oncosuppressor gene mutated in almost all human cancers and its role in BCa have led to conflicting results. Some researchers found a correlation between altered p53 and poorer outcomes of BCa, due to its association with features of tumor aggressiveness such as higher stage, higher grade and lympho-vascular invasion (LVI) (85,107). Alterations of p53 were also found in normal mucosa of patients with NMBIC who experienced recurrence, maybe due to the presence of premalignant lesions surrounding the cancer area (79). Shariat et al. found a positive correlation between mutant p53 and BCa recurrence (81). However, other authors did not confirm these findings (80,92). Due to these controversial findings, researchers shifted their attention from a single gene to a series of combined alterations of multiple genes. A retrospective study including 404 patients investigated a 26-gene signature that resulted negative for tumor recurrence (108), while another retrospective multicenter study investigated the prognostic role of fibroblast growth factor receptor 3 (FGFR3) status and three molecular markers (MIB-1, P53, and P27kip1) in 286 patients with NMIBC and a mean follow-up of 5.5 years. The combination of FGFR3 and MIB-1 was found to be an independent predictor of disease recurrence rate (109). Finally, also miRNAs have been investigated as biomarkers for tumor recurrence, but to date results are incomplete (110,111).

Table 6

Summary of the roles of p53 as predictor of outcomes of BCa

Clinical setting Study Number of patients Results
NMIBC
   Predictor of recurrence Gontero et al. (77) 214 p53 has no role in prediction of recurrence of BCa
Shariat et al. (78) 43 p53 has no role in prediction of recurrence of BCa
Friedrich et al. (79) 53 p53 has only slight correlation with tumor rec, but its accumulation in healthy bladder mucosa correlated strongly with disease recurrence (maybe for premalignant alterations)
Liukkonen et al. (80) 207 p53 may be a predictor of recurrence of BCa
Shariat et al. (81) 83 p53 may help in stratifying patients with higher risk of rec
   Predictor of progression Wolf et al. (82) 30 p53 overexpression significantly decreases tumor-free survival
Hermann et al. (83) 143 p53 overexpression should be considered as a criteria for immediate RC, because of higher progression rate
Sarkis et al. (84) 43 BCas exhibiting nuclear overexpression of p53 protein have a higher probability of disease progression
Serth et al. (85) 69 85.7% of patients with more than 20% of cells positive for p53 had disease progression with muscle-invasive growth compared with only 1.8% of patients negative for p53 (P<0.01); confirm of the role of p53 in the progression of BCa
   Predictor of response to intravesical BCG Lacombe et al. (86) 98 p53 overexpression before BCG therapy did not predict response to BCG therapy
Caliskan et al. (87) 30 p53 appears to be a prognostic indicator of tumor unresponsive to BCG
Pfister et al. (88) 60 p53 mutations have a useful predictive value for BCG therapy response in BCa
Saint et al. (89) 102 p53 nuclear overexpression is associated with cancer death after BCG therapy
Hegazy et al. (90) 88 p53 positivity is a predictor of recurrence and/or progression after BCG adjuvant therapy
MIBC
   Predictor of outcomes after RC alone Shariat et al. (91) 692 p53 expression was independently associated with rec and CSM
Malats et al. (92) 10,026 A prognostic value of p53 overexpression was observed in 9/34 studies (27%)
Esrig et al. (93) 243 p53 status was an independent predictor of rec and OS
Shariat et al. (94) 80 p53 is the strongest predictor of BCa outcomes in patients undergoing RC
Shariat et al. (95) 91 Including p53 in standard predictors improves predictive accuracy of outcomes
   Predictor of response to NAC Chang et al. (96) In vitro Mutant p53 protein may enhance chemosensitivity
Sarkis et al. (97) 90 p53 overexpression has independent prognostic value for survival in patients with MIBC treated with NAC
Plimack et al. (98) 44 p53 mutation did not predict response or toxicity of NAC
   Predictor of response to systemic chemotherapy Stadler et al. (99) 499 The prognostic value of p53 was not confirmed
Siu et al. (100) 118 Expression of p53 did not predict response to systemic therapy
Qureshi et al. (101) 83 p53 did not predict response to systemic therapy, but it had prognostic significance within the chemoresistant subgroup
Kong et al. (102) 89 Prognostic differences related to P53 expression in invasive BCas may be partly due to lower chemosensitivity
   Predictor of response to systemic immunotherapy Not assessed for clinical, biological and pathophysiological reasons
   Predictor of response to radiotherapy Rödel et al. (103) 70 p53 was found to be not predictive of radiosensitivity
Lipponen et al. (104) 400 p53 may be predictive of radiosensitivity
Koga et al. (105) 35 No significant association with chemoradiation response was observed for immunohistochemical expression p53
Matsumoto et al. (106) 62 p53 was found to be not predictive of radiosensitivity

BCa, bladder cancer; NMIBC, non-muscle invasive bladder cancer; BCG, bacillus Calmette-Guérin; MIBC, muscle-invasive bladder cancer; RC, radical cystectomy; CSM, cancer-specific mortality; OS, overall survival; NAC, neoadjuvant chemotherapy.

Prognostic tissue-based biomarkers of progression

Many tissue-based biomarkers have been studied as predictors of tumor progression, but only few of them were founded to be clinically relevant. As already discussed, p53 is the most frequently studied biomarker and its overexpression or mutation seems to be correlated with tumor progression and aggressiveness. Wolf et al. showed how p53 may identify patients with most aggressive high-risk cancers, and therefore eligible for immediate radical cystectomy (RC) (82). Shariat et al. and Chan et al. focused their work on a combination of genes’ alterations, all related to cell cycle regulation (p53, pRb, P21, P27, MMP-2, PAI-1), finding that a series of genes rather than a single gene may affect tumor progression (81,112). In its retrospective study, van Rhijn et al. described a correlation between FGFR3 mutation together with an increased MIB-1 expression and risk of tumor progression (109). Similarly, an important prospective study of van Kessel et al., including 1,239 patients, confirmed the important role of FGFR3 mutational status and methylation of GATA2 in improving the EAU NMIBC risk stratification for the prediction of tumor progression (113). Despite these interesting results, information obtained by molecular biomarkers is not sufficient alone to be introduced in clinical practice, and its integration with established clinico-pathologic variables remains mandatory.

Predictive tissue-based biomarkers of response to intravesical therapy

Numerous biomarkers have been tested as predictor of response to intravesical therapy such as BCG treatment. Malmström et al. in a multicenter trial evaluated the role of ezrin, CK20, and Ki-67 as predictors of response to BCG and intravesical chemotherapy (114). However, none of the analyzed biomarkers showed to be able to predict response and only tumor multifocality was associated with disease progression.


Prognostic and predictive markers in muscle-invasive bladder cancer (MIBC)

MIBC still remains an aggressive disease characterized by a generally unfavorable prognosis and high rates of progression to metastatic disease. The standard treatment of MIBC is RC, eventually preceded by neoadjuvant chemotherapy (NAC) (115,116). The current predictive models of prognosis, essential for patients’ stratification and decision-making regarding perioperative systemic therapies are mainly based on clinico-pathologic variables such as clinical stage, presence of concomitant CIS, variant histology and LVI at TURBT, tumor grade and presence of pre-operative hydronephrosis. In this context, several biomarkers have been proposed in order to strengthen and improve the precision of these prognostic models.

Prognostic markers after RC alone

As for NMIBC, p53 has been the most studied biomarker for the prediction of outcomes also in patients with MIBC. Shariat et al. demonstrated how its expression was independently associated with recurrence and CSM (91). These findings were confirmed by Malats et al. in its meta-analysis (a prognostic value of p53 was found in 27% of cases) (92). Human epidermal growth factor receptor 2 (HER2) has also been tested in patients with MIBC with contrasting results. Soria et al. found a correlation between overexpression of HER2 and worse anatomopathological features at RC such as lymph node involvement, but this did not translate into a worse prognosis (117); conversely, Bolenz et al. found an association between HER2 and both disease recurrence and CSM (118). The retinoblastoma protein (RB1) and PTEN, tumor suppressor genes, and their inactivation seem to be correlated with worse cancer-specific survival in MIBC, while their mutation seem to be absent or rare in low-grade and superficial tumors (of the 38 patients with muscle-invasive tumors, 13 had altered expression of Rb, and only one of 10 patients with superficial carcinomas had this alteration) (119). An important role of survivin was described by Shariat et al.: its expression was detected only in tumor sections and in none of the normal bladder specimens (both in cystectomy and nodes specimens). At multivariable analyses that adjusted for the effect of standard prognosticators, survivin expression was associated with disease recurrence (P=0.040), disease-specific mortality and all-cause mortality in patients treated with RC. The addition of survivin improved the accuracy of the model over standard clinico-pathologic features for prediction of disease recurrence and CSS and raised the potential for survivin-targeted therapy for BCa (120). Despite some interesting results coming from these biomarkers, their potential role as single predictors of outcomes remains poor since their accuracy over standard prognosticators has proven to be unsatisfactory. Some works tried to assess the role of a panel of established BCa biomarkers. A combined analysis of p53, pRB, p21, p27, and cyclin E1 in 191 patients with pTa3N0M0 BCa treated with RC and bilateral lymphadenectomy (median follow-up: 3.1 years) showed that the number of altered biomarkers had the highest predictive accuracy for both disease recurrence (76.8%, P<0.001) and CSM (78.3%, P<0.001), and its addiction increased the predictive accuracy of a nomogram based on the TNM staging system for disease recurrence and CSM by 10.9% (83.4% vs. 72.5%, P<0.001) and 8.6% (86.9% vs. 78.3, P<0.001), respectively (121,122). The Cancer Genome Atlas (TCGA) project reported an analysis of 131 urothelial carcinomas to provide a comprehensive landscape of molecular alterations, detecting recurrent mutations in 32 genes, including multiple genes involved in cell-cycle regulation, chromatin regulation, and kinase signaling pathways (123). RNA sequencing revealed four expression subtypes: the “cluster I” tumors presented a papillary-like morphology, with overexpression of FGFR3, decreased expression of miR-99a and miR-100 and high expression of GATA3 and FOXA1; cluster II tumors presented overexpression of uroplakins, E-cadherin, members of the miR-200 family, ERBB2 and estrogen receptor-β. Cluster III tumors, defined also as basal/squamous like, were characterized by overexpression of epithelial lineage genes. Starting from that, an international collaboration led by Kamoun et al. tried to facilitate the clinical use of molecular classes by the identification of six classes of MIBC on the basis of oncogenic mechanisms: luminal papillary (24%), luminal non-specified (8%), luminal unstable (15%), stroma-rich (15%), basal/squamous (35%), and neuroendocrine-like (3%). Luminal unstable, basal/squamous and neuroendocrine-like subtypes were associated with worse oncological outcomes after RC (124). These findings may have important future implications such as those related to targeted therapies, helping the development of MIBC precision medicine by providing a robust framework to connect clinical contexts to molecular findings.

Predictive markers of response to NAC

A plethora of biomarkers involved in cell cycle as DNA repair genes, regulators of apoptosis, receptor tyrosine kinases, genes involved in cellular efflux and molecular subtypes have been analyzed for understanding the response to cisplatin-based NAC. Regarding products of DNA repair genes, a significant pathological response (pT0-1) at RC after NAC was observed in 66% (24 of 39) of patients with low/intermediate BRCA1 levels compared to 22% (4 of 18) of patients with high BRCA1 levels (P=0.01). Median overall survival (OS) was 168 months in patients with low/intermediate levels and 34 months in patients with high BRCA1 levels (P=0.002). In the multivariable analysis for survival, only BRCA1 expression levels and the presence of LVI emerged as independent prognostic factors (125). Regarding ERCC1, results are conflicting. Some authors have denied a correlation between expression of ERCC1 and response to NAC (126), while others as Ozcan et al. reported a median OS of 9.3 vs. 26.7 months (P=0.058) in patients with low and high ERCC1 expression, respectively (127). Pathological complete response after NAC (HR: 0.1, 95% CI: 0.012–0.842, P=0.034) and high ERCC1 expression (HR: 3.7, 95% CI: 1.2–11.2, P=0.019) were significantly associated with disease-free survival. Van Allen et al. performed a whole-exome sequencing on DNA from 50 patients with muscle-invasive urothelial carcinoma who received neoadjuvant cisplatin-based chemotherapy followed by RC (25 pT0/pTis “responders”, 25 pT2+ “not-responders”) and resulted that ERCC2 was the only significantly mutated gene enriched in the cisplatin responders compared with not-responders (128). In 2015 a prospective study showed how an alteration in one or more of the three DNA repair genes ATM, RB1, and FANCC predicted pathologic response (P<0.001; 87% sensitivity, 100% specificity) and better OS (P=0.007) after cisplatin-based chemotherapy for MIBC (98). The ERRB family of tyrosine kinase growth-factor receptors was also analyzed of response to NAC, and main results derived from a cohort composed of 38 patients with complete response (ypT0N0) and of 33 patients not-responders (higher than ypT2). Using a sequencing of 178 genes it was found that 9 of 38 complete responders had ERBB2 mutations, whereas none of 33 not-responders had ERBB2 mutations (P=0.003), thus concluding that ERBB2 may characterize a subgroup of MIBC patients with an excellent response to NAC (129). Regarding the regulators of apoptosis, patients with nuclear overexpression of p53 had a significantly higher proportion of cancer deaths and, at multivariable analysis, p53 overexpression was independently associated with survival (P=0.001; relative risk ratio, 3.1). Long-term survival was low (41%) in patients with p53 overexpression vs. 77% in those without p53 overexpression (P=0.007) (97). High affinity copper uptake protein 1 (CTR1) is a protein involved in cellular efflux and plays a role in the influx of platinum. An evaluation of CTR1 expression in 47 patients with MIBC treated with NAC seems to suggest that CTR1 expression may be associated with pathological response (130). Lastly, while past studies seemed to correlate response to NAC to basal, luminal and p53-like subtypes, based on different molecular expression, a recent international consensus about molecular classification of MIBC did not find a significant association between the molecular subtypes and response to NAC treatment (124).

Predictive markers of response to systemic chemotherapy

Regulators of the cell cycle have been studied also as predictors for response to systemic chemotherapy in advanced/metastatic setting. In this scenario, cyclins and cyclin-dependent kinases have a key-role (131). Chemotherapy was showed as particularly beneficial in patients with high nuclear CyclinD1 expression (100), and CCND1 amplification status and CyclinD1 expression are independent risk factors for developing metastatic BCa (132). Analyzing the role of mi-RNAs in 83 patients with advanced MIBC, higher levels of miR-21 (P=0.01, HR: 2.01) and miR-372 (P=0.05, HR: 1.70) were associated with a shorter PFS, even if this study did not confirm the role of mi-RNAs in modulating platinum sensitivity (133). P53 was studied also as predictor of response to systemic and adjuvant CT. Despite initial promising results (93), a phase III randomized controlled trial did not confirm the predictive role of p53 in invasive disease: in a cohort of 499 patients, overall 5-year probability of disease recurrence after CT was not related to the p53 status (99). Currently, the most promising biomarkers as predictors of response to CT are the proteins involved in DNA damage detection and repair (DDR), such as BRCA-1, BRCA-2, RAD51, PAR, PARP1, ERCC1, ERCC2, and RRM1. In a study including 100 patients treated with platinum-based chemotherapy, the ones with DDR alterations showed an improved PFS (9.3 vs. 6.0 months, P=0.007) and OS (23.7 vs. 13.0 months, P=0.006) (134). Finally, molecules involved in drug transport such as MDR1 gene, and in cell growth signaling such as EGFR, HER2 and FGF, were studied as predictors to therapy, with miscellaneous and not conclusive results (100,135).

Predictive markers of response to systemic immunotherapy

The recent approval of two classes of drugs (PD-L1 targeting agents and PD-1 blocking monoclonal antibodies) for the treatment of urothelial cancer by the FDA dramatically transformed the therapeutic landscape of advanced/metastatic BCa. Several biomarkers have been explored in this context and the evidence that a significant proportion of patients has a good response to treatment despite being tested negatively for a molecule (and vice-versa) underlines the need of larger prospective trials (136-140). PD-L1 expression has been widely studied also in BCa, with contrasting results. In a study of 315 patients the PD-L1 expression status on infiltrating immune cells (ICs) in the tumor microenvironment was defined by the percentage of PD-L1-positive ICs: IC0 (<1%), IC1 (≥1% but <5%), and IC2/3 (≥5%). Objective response rates were 26% (95% CI: 18–36) in the IC2/3 group, 18% (95% CI: 13–24) in the IC1/2/3 group, and 15% (95% CI: 11–19) overall in all 310 patients (136). Conversely, the CheckMate 275 trial showed clinical responses to nivolumab independently of PD-L1 expression levels (137). Many causes can be identified as responsible of these discrepancies: lack of standardized testing and evaluation of PD-L1, variations in percentage cut-offs and, lastly, that PD-L1 level is frequently assessed on a single biopsy and is therefore unable to reflect the intratumoral variability of BCa. The IMvigor210 trial found that molecular subtypes of BCa were independently associated with response to atezolizumab treatment (136). PD-L1 immune cell prevalence was higher in the basal subtype rather than in the luminal one (60% vs. 23%, P<0.0001), with expression of 15% in papillary-like luminal cluster I, 34% in cluster II, 68% in squamous-like basal cluster III, and 50% in basal cluster IV. Response to atezolizumab was significantly higher in luminal cluster II (objective response rate of 34%, P=0.0017). Conversely, CheckMate 275 showed that basal 1 subtype had the highest proportion of responders (137). As other biomarkers, TGFβ was shown as attenuator of tumor response to PD-L1 blockade (136), while higher values of IFN-γ were associated with good response to nivolumab and higher PD-L1 expression (137). Finally, a correlation between high mutational burden and better responses to immunotherapeutic agents have been identified (136). Despite these interesting and promising results, more research is needed before tissue biomarkers could enter in the clinical practice and drive decision regarding systemic immunotherapy.

Predictive markers of response to radiotherapy

Currently, patients unfit for RC can benefit of radiotherapic treatment in the context of trimodal therapy (TMT), combined with transurethral tumor resection and chemotherapy, with quality of life and oncological outcomes apparently comparable to surgical treatment (141,142). It is mandatory to stratify patients on the basis of the level of response to radiotherapy, in order to identify optimal candidates for TMT. Rödel et al. correlated high Ki-67 index (≥8%) to complete response and local control with preserved bladder at 5 years after radiotherapy (103). Tanabe et al., in their retrospective analysis, demonstrated that, among various clinicopathologic variables, high Ki-67 expression (≥20%) was independently associated with better CSS (5-year CSS rate, 78% vs. 46% for high vs. low Ki-67 levels; P=0.019) (143). Matsumoto et al., describing the role of molecules involved in apoptotic pathways as p53, Bcl-2, Bax, found no correlation between their expression and response to radiotherapy (106). As already described with systemic chemotherapy, the prognostic impact of DDR gene alterations, most commonly identified in ERCC2, showed a trend for improved response to radiotherapy, with higher bladder (HR: 0.32, P=0.070) or metastatic (HR: 0.37, P=0.070) RFS after 2 years of follow-up (144). Differently from other experiences, no impact of MRE11 protein expression on outcomes was detected (145).


Conclusions

The great interest about biomarkers and the related amount of literature published in the last decade reflects the fact that we are moving from the era of “one fits all” to that of precision medicine. Despite tremendous and continuous research, to date, biomarkers are not yet recommended for their use in daily clinical routine, because of both low level of evidence due to the poor quality of available data and to the difficulty in identifying the correct scenario of use for each biomarker, as shown in this systematic review. Despite this, there is no doubt that the potential of these biomarkers is hugely significant and that the path followed is the correct one. It derives that it is essential to pour even more energies investing in randomized controlled trials and in large international collaborations to overcome the obstacles we have encountered so far. Until then, the role of biomarkers should be limited to experimental settings.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editor (Marco Moschini) for the series “Bladder Cancer” published in AME Medical Journal. The article has undergone external peer review.

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://amj.amegroups.com/article/view/10.21037/amj.2020.02.06/coif). The series “Bladder Cancer” was commissioned by the editorial office without any funding or sponsorship. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Yafi FA, Brimo F, Steinberg J, et al. Prospective analysis of sensitivity and specificity of urinary cytology and other urinary biomarkers for bladder cancer. Urol Oncol 2015;33:66.e25-31. [Crossref] [PubMed]
  2. Kurth KH, Schellhammer PF, Okajima E, et al. Current methods of assessing and treating carcinoma in situ of the bladder with or without involvement of the prostatic urethra. Int J Urol 1995;2:8-22. [Crossref] [PubMed]
  3. Krajewski W, Kościelska-Kasprzak K, Rymaszewska J, et al. How different cystoscopy methods influence patient sexual satisfaction, anxiety, and depression levels: a randomized prospective trial. Qual Life Res 2017;26:625-34. [Crossref] [PubMed]
  4. Soria F, Droller MJ, Lotan Y, et al. An up-to-date catalog of available urinary biomarkers for the surveillance of non-muscle invasive bladder cancer. World J Urol 2018;36:1981-95. [Crossref] [PubMed]
  5. Goebell PJ, Groshen SL, Schmitz-Drager BJ, et al. Guidelines for development of diagnostic markers in bladder cancer. World J Urol 2008;26:5-11. [Crossref] [PubMed]
  6. Ferlay J, Soerjomataram I, Ervik M, et al. GLOBOCAN 2012: estimated cancer incidence, mortality and prevalence worldwide in 2012. Lyon: International Agency for Research on Cancer, 2013.
  7. Rouprêt M. Smoking status is not sufficient to accurately target patients who would benefit from screening for bladder and kidney cancer. Eur Urol Focus 2015;1:52-3. [Crossref] [PubMed]
  8. Pesch B, Taeger D, Johnen G, et al. Screening for bladder cancer with urinary tumor markers in chemical workers with exposure to aromatic amines. Int Arch Occup Environ Health 2014;87:715-24. [Crossref] [PubMed]
  9. Mariani AJ, Mariani MC, Macchioni C, et al. The significance of adult hematuria: 1,000 hematuria evaluations including a risk-benefit and cost-effectiveness analysis. J Urol 1989;141:350-5. [Crossref] [PubMed]
  10. Mian C, Pycha A, Wiener H, et al. ImmunoCyt: a new tool for detecting transitional cell cancer of the urinary tract. J Urol 1999;161:1486-9. [Crossref] [PubMed]
  11. Olsson H, Zackrisson B. ImmunoCyt a useful method in the follow-up protocol for patients with urinary bladder carcinoma. Scand J Urol Nephrol 2001;35:280-2. [Crossref] [PubMed]
  12. Feil G, Zumbragel A, Paulgen-Nelde HJ, et al. Accuracy of the ImmunoCyt assay in the diagnosis of transitional cell carcinoma of the urinary bladder. Anticancer Res 2003;23:963-7. [PubMed]
  13. Beiche B, Ebert T, Schmitz-Dräger BJ. Immunzytologie in der Diagnostik des Urothelkarzinoms—ein reproduzierbares Testverfahren. Urologe A 2002;41:45.
  14. Schmitz-Dräger B, Tirsar LA, Schmitz-Dräger C, et al. Immunocytology in the assessment of patients with painless gross haematuria. BJU Int 2008;101:455-8. [PubMed]
  15. Hajdinjak T. UroVysion FISH test for detecting urothelial cancers: meta-analysis of diagnostic accuracy and comparison with urinary cytology testing. Urol Oncol 2008;26:646-51. [Crossref] [PubMed]
  16. Schlomer BJ, Ho R, Sagalowsky A, et al. Prospective validation of the clinical usefulness of reflex fluorescence in situ hybridization assay in patients with atypical cytology for the detection of urothelial carcinoma of the bladder. J Urol 2010;183:62-7. [Crossref] [PubMed]
  17. Köhler CU, Martin L, Bonberg N, et al. Automated quantification of FISH signals in urinary cells enables the assessment of chromosomal aberration patterns characteristic for bladder cancer. Biochem Biophys Res Commun 2014;448:467-72. [Crossref] [PubMed]
  18. Bergman J, Reznichek RC, Rajfer J. Surveillance of patients with bladder carcinoma using fluorescent in-situ hybridization on bladder washings. BJU Int 2008;101:26-9. [PubMed]
  19. Junker K, Fritsch T, Hartmann A, et al. Multicolor fluorescence in situ hybridization (M-FISH) on cells from urine for the detection of bladder cancer. Cytogenet Genome Res 2006;114:279-83. [Crossref] [PubMed]
  20. Krause FS, Rauch A, Schrott KM, et al. Clinical decisions for treatment of different staged bladder cancer based on multitarget fluorescence in situ hybridization assays? World J Urol 2006;24:418-22. [Crossref] [PubMed]
  21. 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. [Crossref] [PubMed]
  22. Shariat SF, Canto EI, Kattan MW, et al. Beyond prostate-specific antigen: new serologic biomarkers for improved diagnosis and management of prostate cancer. Rev Urol 2004;6:58-72. [PubMed]
  23. Steiner H, Bergmeister M, Verdorfer I, et al. Early results of bladder-cancer screening in a high-risk population of heavy smokers. BJU Int 2008;102:291-6. [Crossref] [PubMed]
  24. Lotan Y, Elias K, Svatek RS, et al. Bladder cancer screening in a high risk asymptomatic population using a point of care urine based protein tumor marker. J Urol 2009;182:52-7. [Crossref] [PubMed]
  25. Poulakis V, Witzsch U, De Vries R, et al. A comparison of urinary nuclear matrix protein-22 and bladder tumour antigen tests with voided urinary cytology in detecting and following bladder cancer: the prognostic value of false-positive results. BJU Int 2001;88:692-701. [Crossref] [PubMed]
  26. Sharma S, Zippe CD, Pandrangi L, et al. Exclusion criteria enhance the specificity and positive predictive value of NMP22 and BTA stat. J Urol 1999;162:53-7. [Crossref] [PubMed]
  27. Barbieri CE, Cha EK, Chromecki TF, et al. Decision curve analysis assessing the clinical benefit of NMP22 in the detection of bladder cancer: secondary analysis of a prospective trial. BJU Int 2012;109:685-90. [Crossref] [PubMed]
  28. Lokeshwar VB, Soloway MS. Current bladder tumor tests: does their projected utility fulfill clinical necessity? J Urol 2001;165:1067-77. [Crossref] [PubMed]
  29. 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;4:2511-20. [PubMed]
  30. Glas AS, Roos D, Deutekom M, et al. Tumor markers in the diagnosis of primary bladder cancer. A systematic review. J Urol 2003;169:1975-82. [Crossref] [PubMed]
  31. Bonberg N, Taeger D, Gawrych K, et al. Chromosomal instability and bladder cancer: the UroVysion(TM) test in the UroScreen study. BJU Int 2013;112:E372-82. [Crossref] [PubMed]
  32. Banek S, Schwentner C, Tager D, et al. Prospective evaluation of fluorescence-in situ-hybridization to detect bladder cancer: results from the UroScreen-Study. Urol Oncol 2013;31:1656-62. [Crossref] [PubMed]
  33. Huber S, Schwentner C, Taeger D, et al. Nuclear matrix protein-22: a prospective evaluation in a population at risk for bladder cancer. Results from the UroScreen study. BJU Int 2012;110:699-708. [Crossref] [PubMed]
  34. Cha EK, Tirsar LA, Schwentner C, et al. Immunocytology is a strong predictor of bladder cancer presence in patients with painless hematuria: a multicentre study. Eur Urol 2012;61:185-92. [Crossref] [PubMed]
  35. Lotan Y, Svatek RS, Krabbe LM, et al. Prospective external validation of a bladder cancer detection model. J Urol 2014;192:1343-8. [Crossref] [PubMed]
  36. Beukers W, Kandimalla R, van Houwelingen D, et al. The use of molecular analyses in voided urine for the assessment of patients with hematuria. PLoS One 2013;8:e77657. [Crossref] [PubMed]
  37. Todenhöfer T, Hennenlotter J, Tews V, et al. Impact of different grades of microscopic hematuria on the performance of urine-based markers for the detection of urothelial carcinoma. Urol Oncol 2013;31:1148-54. [Crossref] [PubMed]
  38. Lucca I, Hofbauer SL, Haitel A, et al. Urinary expression of genes involved in DNA methylation and histone modification for diagnosis of bladder cancer in patients with asymptomatic microscopic haematuria. Oncol Lett 2019;18:57-62. [PubMed]
  39. Davidson PJ, McGeoch G, Shand B. Inclusion of a molecular marker of bladder cancer in a clinical pathway for investigation of haematuria may reduce the need for cystoscopy. N Z Med J 2019;132:55-64. [PubMed]
  40. van der Aa MN, Steyerberg EW, Bangma C, et al. Cystoscopy revisited as the gold standard for detecting bladder cancer recurrence: diagnostic review bias in the randomized, prospective CEFUB trial. J Urol 2010;183:76-80. [Crossref] [PubMed]
  41. Shariat SF, Casella R, Wians FH, et al. Risk stratification for bladder tumor recurrence, stage and grade by urinary nuclear matrix protein 22 and cytology. Eur Urol 2004;45:304-13. [Crossref] [PubMed]
  42. Breen V, Kasabov N, Kamat AM, et al. A holistic comparative analysis of diagnostic tests for urothelial carcinoma: a study of Cxbladder Detect, UroVysion® FISH, NMP22® and cytology based on imputation of multiple datasets. BMC Med Res Methodol 2015;15:45. [Crossref] [PubMed]
  43. Pode D, Shapiro A, Wald M, et al. Noninvasive detection of bladder cancer with the BTA stat test. J Urol 1999;161:443-6. [Crossref] [PubMed]
  44. Doğan C, Pelit ES, Yıldırım A, et al. The value of the NMP22 test for superficial bladder cancer diagnosis and follow-up. Turk J Urol 2013;39:137-42. [Crossref] [PubMed]
  45. Todenhöfer T, Hennenlotter J, Esser M, et al. Combined application of cytology and molecular urine markers to improve the detection of urothelial carcinoma. Cancer Cytopathol 2013;121:252-60. [Crossref] [PubMed]
  46. Babjuk M, Böhle A, Burger M, et al. EAU guidelines on non-muscle-invasive urothelial carcinoma of the bladder: update 2016. Eur Urol 2017;71:447-61. [Crossref] [PubMed]
  47. Chang SS, Boorjian SA, Chou R, et al. Diagnosis and treatment of non-muscle invasive bladder cancer: AUA/SUO guideline. J Urol 2016;196:1021-9. [Crossref] [PubMed]
  48. Kamat AM, Willis DL, Dickstein RJ, et al. Novel fluorescence in situ hybridization-based definition of bacille Calmette-Guérin (BCG) failure for use in enhancing recruitment into clinical trials of intravesical therapies. BJU Int 2016;117:754-60. [Crossref] [PubMed]
  49. Bubendorf L, Piaton E. UroVysion® multiprobe FISH in the triage of equivocal urinary cytology cases. Ann Pathol 2012;32:e52-6, 438-43.
  50. Raitanen MP, Marttila T, Nurmi M, et al. Human complement factor h related protein test for monitoring bladder cancer. J Urol 2001;165:374-7. [Crossref] [PubMed]
  51. Casetta G, Gontero P, Zitella A, et al. BTA quantitative assay and NMP22 testing compared with urine cytology in the detection of transitional cell carcinoma of the bladder. Urol Int 2000;65:100-5. [Crossref] [PubMed]
  52. Raitanen MP. 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. [Crossref] [PubMed]
  53. Nisman B, Yutkin V, Peretz T, et al. The follow-up of patients with non-muscle-invasive bladder cancer by urine cytology, abdominal ultrasound and urine CYFRA 21-1: a pilot study. Anticancer Res 2009;29:4281-5. [PubMed]
  54. Babjuk M, Soukup V, Pešl M, et al. Urinary cytology and quantitative BTA and UBC tests in surveillance of patients with pTapT1 bladder urothelial carcinoma. Urology 2008;71:718-22. [Crossref] [PubMed]
  55. Hosseini J, Golshan AR, Mazloomfard MM, et al. Detection of recurrent bladder cancer: NMP22 test or urine cytology? Urol J 2012;9:367-72. [PubMed]
  56. Messing EM, Teot L, Korman H, et al. Performance of urine test in patients monitored for recurrence of bladder cancer: a multicenter study in the United States. J Urol 2005;174:1238-41. [Crossref] [PubMed]
  57. Serretta V, Lo Presti D, Vasile P, et al. Urinary NMP22 for the detection of recurrence after transurethral resection of transitional cell carcinoma of the bladder: experience on 137 patients. Urology 1998;52:793-6. [Crossref] [PubMed]
  58. Horstmann M, Patschan O, Hennenlotter J, et al. Combinations of urine-based tumour markers in bladder cancer surveillance. Scand J Urol Nephrol 2009;43:461-6. [Crossref] [PubMed]
  59. 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;176:919-26. [Crossref] [PubMed]
  60. Grossman HB, Soloway M, Messing E, et al. Surveillance for recurrent bladder cancer using a point-of-care proteomic assay. JAMA 2006;295:299-305. [Crossref] [PubMed]
  61. Gupta NP, Sharma N, Kumar R. Nuclear matrix protein 22 as adjunct to urine cytology and cystoscopy in follow-up of superficial TCC of urinary bladder. Urology 2009;73:592-6. [Crossref] [PubMed]
  62. 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. [Crossref] [PubMed]
  63. 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;8:369-73. [Crossref] [PubMed]
  64. Vriesema JL, Atsma F, Kiemeney LA, et al. Diagnostic efficacy of the ImmunoCyt test to detect superficial bladder cancer recurrence. Urology 2001;58:367-71. [Crossref] [PubMed]
  65. Sullivan PS, Nooraie F, Sanchez H, et al. Comparison of ImmunoCyt, UroVysion, and urine cytology in detection of recurrent urothelial carcinoma: a "split-sample" study. Cancer 2009;117:167-73. [PubMed]
  66. 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;168:1950-4. [Crossref] [PubMed]
  67. Youssef RF, Schlomer BJ, Ho R, et al. Role of fluorescence in situ hybridization in bladder cancer surveillance of patients with negative cytology. Urol Oncol 2012;30:273-7. [Crossref] [PubMed]
  68. Lotan Y. Clinical comparison of noninvasive urine tests for ruling out recurrent urothelial carcinoma. Urol Oncol 2017;35:531.e15-22. [Crossref] [PubMed]
  69. Kavalieris L, O’Sullivan P, Frampton C, et al. Performance Characteristics of a Multigene Urine Biomarker Test for Monitoring for Recurrent Urothelial Carcinoma in a Multicenter Study. J Urol 2017;197:1419-26. [Crossref] [PubMed]
  70. Esuvaranathan K, Chiong E, Thamboo TP, et al. Predictive value of p53 and pRb expression in superficial bladder cancer patients treated with BCG and interferon-alpha. Cancer 2007;109:1097-105. [Crossref] [PubMed]
  71. Lima L, Severino PF, Silva M, et al. Response of high-risk of recurrence/progression bladder tumours expressing sialyl-Tn and sialyl-6-T to BCG immunotherapy. Br J Cancer 2013;109:2106-14. [Crossref] [PubMed]
  72. Langle YV, Belgorosky D, Prack McCormick B, et al. FGFR3 down-regulation is involved in bacillus Calmette-Guérin induced bladder tumor growth inhibition. J Urol 2016;195:188-97. [Crossref] [PubMed]
  73. Leibovici D, Grossman HB, Dinney CP, et al. Polymorphisms in inflammation genes and bladder cancer: from initiation to recurrence, progression, and survival. J Clin Oncol 2005;23:5746-56. [Crossref] [PubMed]
  74. Zhao H, Liang D, Grossman HB, et al. Glutathione peroxidase 1 gene polymorphism and risk of recurrence in patients with superficial bladder cancer. Urology 2005;66:769-74. [Crossref] [PubMed]
  75. Lima L, Oliveira D, Ferreira JA, et al. The role of functional polymorphisms in immune response genes as biomarkers of bacille Calmette-Guérin (BCG) immunotherapy outcome in bladder cancer: establishment of a predictive profile in a Southern Europe population. BJU Int 2015;116:753-63. [Crossref] [PubMed]
  76. Kamat AM, Briggman J, Urbauer DL, et al. Cytokine Panel for Response to Intravesical Therapy (CyPRIT): Nomogram of Changes in Urinary Cytokine Levels Predicts Patient Response to Bacillus Calmette-Guérin. Eur Urol 2016;69:197-200. [Crossref] [PubMed]
  77. Gontero P, Casetta G, Zitella A, et al. Evaluation of P53 protein overexpression, Ki67 proliferative activity and mitotic index as markers of tumour recurrence in superficial transitional cell carcinoma of the bladder. Eur Urol 2000;38:287-96. [Crossref] [PubMed]
  78. Shariat SF, Weizer AZ, Green A, et al. Prognostic value of P53 nuclear accumulation and histopathologic features in T1 transitional cell carcinoma of the urinary bladder. Urology 2000;56:735-40. [Crossref] [PubMed]
  79. Friedrich MG, Riethdorf S, Erbersdobler A, et al. Relevance of p53 gene alterations for tumor recurrence in patients with superficial transitional cell carcinoma of the bladder. Eur Urol 2001;39:159-66. [Crossref] [PubMed]
  80. Liukkonen T, Rajala P, Raitanen M, et al. Prognostic value of MIB-1 score, p53, EGFr, mitotic index and papillary status in primary superficial (Stage pTa/T1) bladder cancer: a prospective comparative study. The Finnbladder Group. Eur Urol 1999;36:393-400. [Crossref] [PubMed]
  81. Shariat SF, Ashfaq R, Sagalowsky AI, et al. Predictive value of cell cycle biomarkers in nonmuscle invasive bladder transitional cell carcinoma. J Urol 2007;177:481-7. [Crossref] [PubMed]
  82. Wolf HK, Stöber C, Hohenfellner R, et al. Prognostic value of p53, p21/WAF1, Bcl-2, Bax, Bak and Ki-67 immunoreactivity in pT1 G3 urothelial bladder carcinomas. Tumour Biol ;22:328-36. nd.. [Crossref] [PubMed]
  83. Hermann GG, Horn T, Steven K. The influence of the level of lamina propria invasion and the prevalence of p53 nuclear accumulation on survival in stage T1 transitional cell bladder cancer. J Urol 1998;159:91-4. [Crossref] [PubMed]
  84. Sarkis AS, Dalbagni G, Cordon-Cardo C, et al. Nuclear overexpression of p53 protein in transitional cell bladder carcinoma: a marker for disease progression. J Natl Cancer Inst 1993;85:53-9. [Crossref] [PubMed]
  85. Serth J, Kuczyk MA, Bokemeyer C, et al. p53 immunohistochemistry as an independent prognostic factor for superficial transitional cell carcinoma of the bladder. Br J Cancer 1995;71:201-5. [Crossref] [PubMed]
  86. Lacombe L, Dalbagni G, Zhang ZF, et al. Overexpression of p53 protein in a high-risk population of patients with superficial bladder cancer before and after bacillus Calmette-Guérin therapy: correlation to clinical outcome. J Clin Oncol 1996;14:2646-52. [Crossref] [PubMed]
  87. Caliskan M, Türkeri LN, Mansuroglu B, et al. Nuclear accumulation of mutant p53 protein: a possible predictor of failure of intravesical therapy in bladder cancer. Br J Urol 1997;79:373-7. [Crossref] [PubMed]
  88. Pfister C, Flaman JM, Dunet F, et al. p53 mutations in bladder tumors inactivate the transactivation of the p21 and Bax genes, and have a predictive value for the clinical outcome after bacillus Calmette-Guerin therapy. J Urol 1999;162:69-73. [Crossref] [PubMed]
  89. Saint F, Le Frere Belda MA, Quintela R, et al. Pretreatment p53 nuclear overexpression as a prognostic marker in superficial bladder cancer treated with Bacillus Calmette-Guérin (BCG). Eur Urol 2004;45:475-82. [Crossref] [PubMed]
  90. Hegazy R, Kamel M, Salem EA, et al. The prognostic significance of p53, p63 and her2 expression in non-muscle-invasive bladder cancer in relation to treatment with bacille Calmette-Guerin. Arab J Urol 2015;13:225-30. [Crossref] [PubMed]
  91. Shariat SF, Bolenz C, Karakiewicz PI, et al. p53 expression in patients with advanced urothelial cancer of the urinary bladder. BJU Int 2010;105:489-95. [Crossref] [PubMed]
  92. Malats N, Bustos A, Nascimento CM, et al. P53 as a prognostic marker for bladder cancer: A meta-analysis and review. Lancet Oncol 2005;6:678-86. [Crossref] [PubMed]
  93. Esrig D, Elmajian D, Groshen S, et al. Accumulation of nuclear p53 and tumor progression in bladder cancer. N Engl J Med 1994;331:1259-64. [Crossref] [PubMed]
  94. Shariat SF, Bolenz C, Godoy G, et al. Predictive value of combined immunohistochemical markers in patients with pT1 urothelial carcinoma at radical cystectomy. J Urol 2009;182:78-84. [Crossref] [PubMed]
  95. Shariat SF, Lotan Y, Karakiewicz PI, et al. p53 predictive value for pT1-2 N0 disease at radical cystectomy. J Urol 2009;182:907-13. [Crossref] [PubMed]
  96. Chang FL, Lai MD. Various forms of mutant p53 confer sensitivity to cisplatin and doxorubicin in bladder cancer cells. J Urol 2001;166:304-10. [Crossref] [PubMed]
  97. Sarkis AS, Bajorin DF, Reuter VE, et al. Prognostic value of p53 nuclear overexpression in patients with invasive bladder cancer treated with neoadjuvant MVAC. J Clin Oncol 1995;13:1384-90. [Crossref] [PubMed]
  98. Plimack ER, Dunbrack RL, Brennan TA, et al. Defects in DNA repair genes predict response to neoadjuvant cisplatin-based chemotherapy in muscle-invasive bladder cancer. Eur Urol 2015;68:959-67. [Crossref] [PubMed]
  99. Stadler WM, Lerner SP, Groshen S, et al. Phase III study of molecularly targeted adjuvant therapy in locally advanced urothelial cancer of the bladder based on p53 status. J Clin Oncol 2011;29:3443-9. [Crossref] [PubMed]
  100. Siu LL, Banerjee D, Khurana RJ, et al. The prognostic role of p53, metallothionein, P-glycoprotein, and MIB-1 in muscle-invasive urothelial transitional cell carcinoma. Clin Cancer Res 1998;4:559-65. [PubMed]
  101. Qureshi KN, Griffiths TR, Robinson MC, et al. TP53 accumulation predicts improved survival in patients resistant to systemic cisplatin-based chemotherapy for muscle-invasive bladder cancer. Clin Cancer Res 1999;5:3500-7. [PubMed]
  102. Kong G, Shin KY, Oh YH, et al. Bcl-2 and p53 expressions in invasive bladder cancers. Acta Oncol 1998;37:715-20. [Crossref] [PubMed]
  103. Rödel C, Grabenbauer GG, Rödel F, et al. Apoptosis, p53, bcl-2, and Ki-67 in invasive bladder carcinoma: possible predictors for response to radiochemotherapy and successful bladder preservation. Int J Radiat Oncol Biol Phys 2000;46:1213-21. [Crossref] [PubMed]
  104. Lipponen PK, Aaltomaa S. Apoptosis in bladder cancer as related to standard prognostic factors and prognosis. J Pathol 1994;173:333-9. [Crossref] [PubMed]
  105. Koga F, Yoshida S, Tatokoro M, et al. ErbB2 and NFκB overexpression as predictors of chemoradiation resistance and putative targets to overcome resistance in muscle-invasive bladder cancer. PLoS One 2011;6:e27616. [Crossref] [PubMed]
  106. Matsumoto H, Wada T, Fukunaga K, et al. Bax to Bcl-2 ratio and Ki-67 index are useful predictors of neoadjuvant chemoradiation therapy in bladder cancer. Jpn J Clin Oncol 2004;34:124-30. [Crossref] [PubMed]
  107. Vorreuther R, Hake R, Borchmann P, et al. Expression of immunohistochemical markers (pcna, ki-67,486p and p53) on paraffin sections and their relation to the recurrence rate of superficial bladder tumors. Urol Int 1997;59:88-94. [Crossref] [PubMed]
  108. Dyrskjøt L, Zieger K, Real FX, et al. Gene expression signatures predict outcome in non-muscle-invasive bladder carcinoma: a multicenter validation study. Clin Cancer Res 2007;13:3545-51. [Crossref] [PubMed]
  109. van Rhijn BWG, Vis AN, van der Kwast TH, et al. Molecular grading of urothelial cell carcinoma with fibroblast growth factor receptor 3 and MIB-1 is superior to pathologic grade for the prediction of clinical outcome. J Clin Oncol 2003;21:1912-21. [Crossref] [PubMed]
  110. Andrew AS, Karagas MR, Schroeck FR, et al. MicroRNA dysregulation and non-muscle-invasive bladder cancer prognosis. Cancer Epidemiol Biomarkers Prev 2019;28:782-8. [Crossref] [PubMed]
  111. Blanca A, Sanchez-Gonzalez A, Requena MJ, et al. Expression of miR-100 and miR-138 as prognostic biomarkers in non-muscle-invasive bladder cancer. APMIS 2019;127:545-53. [Crossref] [PubMed]
  112. Chan OTM, Furuya H, Pagano I, et al. Association of MMP-2, RB and PAI-1 with decreased recurrence-free survival and overall survival in bladder cancer patients. Oncotarget 2017;8:99707-21. [Crossref] [PubMed]
  113. van Kessel KEM, van der Keur KA, Dyrskjøt L, et al. Molecular markers increase precision of the European association of urology non-muscle-invasive bladder cancer progression risk groups. Clin Cancer Res 2018;24:1586-93. [Crossref] [PubMed]
  114. Malmström PU, Hemdan T, Segersten U. Validation of the ezrin, CK20, and Ki-67 as potential predictive markers for BCG instillation therapy of non-muscle-invasive bladder cancer. Urol Oncol 2017;35:532.e1-532.e6. [Crossref] [PubMed]
  115. Stein JP, Skinner DG. Radical cystectomy for invasive bladder cancer: long-term results of a standard procedure. World J Urol 2006;24:296-304. [Crossref] [PubMed]
  116. Bassi P, Ferrante GD, Piazza N, et al. Prognostic factors of outcome after radical cystectomy for bladder cancer: a retrospective study of a homogeneous patient cohort. J Urol 1999;161:1494-7. [Crossref] [PubMed]
  117. Soria F, Moschini M, Haitel A, et al. The effect of HER2 status on oncological outcomes of patients with invasive bladder cancer. Urol Oncol 2016;34:533.e1-10. [Crossref] [PubMed]
  118. Bolenz C, Shariat SF, Karakiewicz PI, et al. Human epidermal growth factor receptor 2 expression status provides independent prognostic information in patients with urothelial carcinoma of the urinary bladder. BJU Int 2010;106:1216-22. [Crossref] [PubMed]
  119. Cordon-Cardo C, Wartinger D, Petrylak D, et al. Altered expression of the retinoblastoma gene product: prognostic indicator in bladder cancer. J Natl Cancer Inst 1992;84:1251-6. [Crossref] [PubMed]
  120. Shariat SF, Ashfaq R, Karakiewicz PI, et al. Survivin expression is associated with bladder cancer presence, stage, progression, and mortality. Cancer 2007;109:1106-13. [Crossref] [PubMed]
  121. Shariat SF, Ashfaq R, Sagalowsky AI, et al. Correlation of cyclin D1 and E1 expression with bladder cancer presence, invasion, progression, and metastasis. Hum Pathol 2006;37:1568-76. [Crossref] [PubMed]
  122. Shariat SF, Karakiewicz PI, Ashfaq R, et al. Multiple biomarkers improve prediction of bladder cancer recurrence and mortality in patients undergoing cystectomy. Cancer 2008;112:315-25. [Crossref] [PubMed]
  123. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 2014;507:315-22. [Crossref] [PubMed]
  124. Kamoun A, de Reyniès A, Allory Y, et al. A consensus molecular classification of muscle-invasive bladder cancer. Eur Urol 2019; [Epub ahead of print]. [Crossref] [PubMed]
  125. Font A, Taron M, Gago JL, et al. BRCA1 mRNA expression and outcome to neoadjuvant cisplatin-based chemotherapy in bladder cancer. Ann Oncol 2011;22:139-44. [Crossref] [PubMed]
  126. Choueiri TK, Jacobus S, Bellmunt J, et al. Neoadjuvant dose-dense methotrexate, vinblastine, doxorubicin, and cisplatin with pegfilgrastim support in muscle-invasive urothelial cancer: Pathologic, radiologic, and biomarker correlates. J Clin Oncol 2014;32:1889-94. [Crossref] [PubMed]
  127. Ozcan MF, Dizdar O, Dincer N, et al. Low ERCC1 expression is associated with prolonged survival in patients with bladder cancer receiving platinum-based neoadjuvant chemotherapy. Urol Oncol 2013;31:1709-15. [Crossref] [PubMed]
  128. Van Allen EM, Mouw KW, Kim P, et al. Somatic ERCC2 mutations correlate with cisplatin sensitivity in muscle-invasive urothelial carcinoma. Cancer Discov 2014;4:1140-53. [Crossref] [PubMed]
  129. Groenendijk FH, de Jong J, Fransen van de Putte EE, et al. ERBB2 Mutations Characterize a Subgroup of Muscle-invasive Bladder Cancers with Excellent Response to Neoadjuvant Chemotherapy. Eur Urol 2016;69:384-8. [Crossref] [PubMed]
  130. Kilari D, Iczkowski KA, Pandya C, et al. Copper transporter-CTR1 expression and pathological outcomes in platinum-treated muscle-invasive bladder cancer patients. Anticancer Res 2016;36:495-501. [PubMed]
  131. Mitra AP, Datar RH, Cote RJ. Molecular pathways in invasive bladder cancer: new insights into mechanisms, progression, and target identification. J Clin Oncol 2006;24:5552-64. [Crossref] [PubMed]
  132. Birkhahn M, Mitra AP, Cote RJ. Molecular markers for bladder cancer: the road to a multimarker approach. Expert Rev Anticancer Ther 2007;7:1717-27. [Crossref] [PubMed]
  133. Bellmunt J, Zhou CW, Mullane SA, et al. Association of tumour microRNA profiling with outcomes in patients with advanced urothelial carcinoma receiving first-line platinum-based chemotherapy. Br J Cancer 2016;115:12-9. [Crossref] [PubMed]
  134. Teo MY, Bambury RM, Zabor EC, et al. DNA Damage Response and Repair Gene Alterations Are Associated with Improved Survival in Patients with Platinum-Treated Advanced Urothelial Carcinoma. Clin Cancer Res 2017;23:3610-8. [Crossref] [PubMed]
  135. Hoffmann AC, Wild P, Leicht C, et al. MDR1 and ERCC1 expression predict outcome of patients with locally advanced bladder cancer receiving adjuvant chemotherapy. Neoplasia 2010;12:628-36. [Crossref] [PubMed]
  136. Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 2016;387:1909-20. [Crossref] [PubMed]
  137. Sharma P, Retz M, Siefker-Radtke A, et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol 2017;18:312-22. [Crossref] [PubMed]
  138. Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 2017;376:1015-26. [Crossref] [PubMed]
  139. Powles T, O'Donnell PH, Massard C, et al. Efficacy and safety of durvalumab in locally advanced or metastatic urothelial carcinoma: updated results from a phase 1/2 open-label study. JAMA Oncol 2017;3:e172411. [Crossref] [PubMed]
  140. Apolo AB, Infante JR, Balmanoukian A, et al. Avelumab, an anti-programmed death-ligand 1 antibody, in patients with refractory metastatic urothelial carcinoma: results from a multicenter, phase Ib study. J Clin Oncol 2017;35:2117-24. [Crossref] [PubMed]
  141. Efstathiou JA, Spiegel DY, Shipley WU, et al. Long-term outcomes of selective bladder preservation by combined-modality therapy for invasive bladder cancer: the MGH experience. Eur Urol 2012;61:705-11. [Crossref] [PubMed]
  142. Giacalone NJ, Shipley WU, Clayman RH, et al. Long-term outcomes after bladder-preserving tri-modality therapy for patients with muscle-invasive bladder cancer: an updated analysis of the Massachusetts General Hospital experience. Eur Urol 2017;71:952-60. [Crossref] [PubMed]
  143. Tanabe K, Yoshida S, Koga F, et al. High Ki-67 expression predicts favorable survival in muscle-invasive bladder cancer patients treated with chemoradiation-based bladder-sparing protocol. Clin Genitourin Cancer 2015;13:e243-51. [Crossref] [PubMed]
  144. Desai NB, Scott SN, Zabor EC, et al. Genomic characterization of response to chemoradiation in urothelial bladder cancer. Cancer 2016;122:3715-23. [Crossref] [PubMed]
  145. Choudhury A, Nelson LD, Teo MT, et al. MRE11 expression is predictive of cause-specific survival following radical radiotherapy for muscle-invasive bladder cancer. Cancer Res 2010;70:7017-26. [Crossref] [PubMed]
doi: 10.21037/amj.2020.02.06
Cite this article as: Giordano A, Soria F. Role and efficacy of current biomarkers in bladder cancer. AME Med J 2020;5:6.

Download Citation