Skip to main content
Current Genomics logoLink to Current Genomics
. 2012 Aug;13(5):395–415. doi: 10.2174/138920212801619232

Bladder Cancer: A Simple Model Becomes Complex

Giovanni Battista Di Pierro 1, Caterina Gulia 1, Cristiano Cristini 1, Giorgio Fraietta 2, Lorenzo Marini 1, Pietro Grande 1, Vincenzo Gentile 1, Roberto Piergentili 3,*
PMCID: PMC3401896  PMID: 23372425

Abstract

Bladder cancer is one of the most frequent malignancies in developed countries and it is also characterized by a high number of recurrences. Despite this, several authors in the past reported that only two altered molecular pathways may genetically explain all cases of bladder cancer: one involving the FGFR3 gene, and the other involving the TP53 gene. Mutations in any of these two genes are usually predictive of the malignancy final outcome. This cancer may also be further classified as low-grade tumors, which is always papillary and in most cases superficial, and high-grade tumors, not necessarily papillary and often invasive. This simple way of considering this pathology has strongly changed in the last few years, with the development of genome-wide studies on expression profiling and the discovery of small non-coding RNA affecting gene expression. An easy search in the OMIM (On-line Mendelian Inheritance in Man) database using “bladder cancer” as a query reveals that genes in some way connected to this pathology are approximately 150, and some authors report that altered gene expression (up- or down-regulation) in this disease may involve up to 500 coding sequences for low-grade tumors and up to 2300 for high-grade tumors. In many clinical cases, mutations inside the coding sequences of the above mentioned two genes were not found, but their expression changed; this indicates that also epigenetic modifications may play an important role in its development. Indeed, several reports were published about genome-wide methylation in these neoplastic tissues, and an increasing number of small non-coding RNA are either up- or down-regulated in bladder cancer, indicating that impaired gene expression may also pass through these metabolic pathways. Taken together, these data reveal that bladder cancer is far to be considered a simple model of malignancy. In the present review, we summarize recent progress in the genome-wide analysis of bladder cancer, and analyse non-genetic, genetic and epigenetic factors causing extensive gene mis-regulation in malignant cells.

Keywords: Bladder carcinoma, urinary tract, NMIBC, MICB, carcinoma in situ, CIS, FGFR3, TP53, epigenetics, small non-coding RNA, environmental causes of bladder carcinoma.

INTRODUCTION

Epidemiology of Bladder Carcinoma

Bladder carcinoma (BC) is the most common malignancy of the urinary tract [1] and one of the most frequent cancers worldwide. According to the US National Cancer Institute (NCI) website (accession: June 2012) (http://www.cancer.gov/), cases diagnosed in 2005-2009 from 18 SEER (Surveillance, Epidemiology and End Results) geographic areas are 37.0 per 100,000 men and 8.9 per 100,000 women, with a men:women ratio of approximately 4:1. Estimated cases from NCI for year 2012 show that, among cancers, BC represents the sixth cause of disease and the second urological malignancy after prostate cancer; moreover, BC is the most common urological tumor in China [2]. Recurrence with metastasis is a frequent cause of death, with some studies highlighting this phenomenon in up to 70% of patients [3]. Indeed, overall tumor-specific 5-year survival rate for years 2002-2008 is around 77%, but it is only 5.5% after cancer has metastasized (NCI, June 2012). This also means that BC is associated to the highest costs in patients surveillance compared with other tumors; according to available data (http://progressreport.cancer.gov/) approximately 3.5 billion dollars per year are spent in the US on bladder cancer treatment.

Non-Genetic Factors Affecting the Epidemiology of Bladder Carcinoma

Active and passive tobacco smoking is the most well established risk factor for BC in both sexes [4, 5]. There is a direct relationship between incidence of BC and the exposure to smoke, in both duration (years since starting) and quantity (cigarettes per day); BC incidence is also higher in people who started smoking at a younger age or are exposed to environmental tobacco smoke during childhood [6, 7]. The second most important risk factor for bladder cancer is occupational exposure [7], a problem known since at least year 1895 [8] and related to 20-25% of all bladder cancer cases.

Dietary factors are connected to BC formation and/or development. It is established that this tumor is directly related to chronic exposure to arsenic, such as that present in drinking water. This is evident in places in which Blackfoot disease (a typical illness related to high arsenic levels in blood) is endemic, as in Taiwan [9], Japan [10], Argentina [11], Chile [12] and the New Hampshire in the United States [13]. Other trace chemical elements were evaluated in literature, and it has been shown that the increase of copper and the decrease of iron and zinc may be involved in BC occurrence [14]. Also the anti-oxidant role of selenium has been studied [15]; interestingly, the role of Vitamin E has been demonstrated as well [16], indicating that anti-oxidants are powerful tools in BC prevention. Although some scientific reports seemed to confirm this, recent data exclude any relationship between BC and alcohol drinking [17, 18]. This result is particularly interesting and somehow surprising, since a relationship between alcohol and BC would be plausible, due to (i) the causal association between alcohol drinking and several types of cancer [19-22], and (ii) the presence inside the bladder and urine of acetaldehyde [23], a metabolite of alcohol and a known carcinogenic compound in humans [20, 24]. These results are applicable considering either the quantity or the different types of alcoholic beverages [18, 25]. Instead, milk consumption seems to show an inverse relationship with BC [26], another unexpected result considering that high milk intake is a risk factor for both ovarian [27] and prostate [28] cancers, two tumors affecting the urogenital system. In the light of earlier studies on milk components, the Authors suggest that the effects of milk may reside in the fats it contains. A recent meta analysis suggests that it is also possible that no relationship at all exists between milk intake and BC development [29], thus this topic is still controversial [30].

BC, and especially the invasive squamous cell carcinoma (SCC), is directly related to the presence of chronic urinary tract infection. Bladder schistosomiasis (bilharzia) is a very common parasitic infection with about 600 million people exposed to infection. This pathology has been considered a definitive cause of urinary bladder cancer with an associated five-fold risk [31]. Actually, it is acknowledged that molecules mediating immune response are also involved in BC development, such as interleukin-2 (IL-2) [32] and, possibly, IL-4, IL-4R and IL-13 [33]. Moreover, in mice it is has been recently shown a relationship among induced inflammation, miR and TP53 expression [34], all of these being involved in BC incidence (see next sections).

External beam radiation therapy (EBRT) for urogenital malignancies increases the rate of secondary bladder malignancies: standardized incidence ratios for bladder cancer developing after radical prostatectomy (RP), EBRT, brachytherapy (BT), and EBRT-BT were significantly higher than that of the general United States population. Thus, the increased risk of bladder cancer in patients undergoing ERBT, BT or ERBT-BT should be taken into account during follow-up, particularly for young patients [35]. Age is also important for survival after radical cystectomy (RC): it has been demonstrated that greater age is associated with adverse outcome [36]. As for chemotherapy, the use of cyclophosphamide, an alkylating agent used for treatment of lymphoproliferative diseases and other non-neoplastic diseases, has been correlated with later development of muscle-invasive bladder cancer (MIBC), with 6-13 years of latency. Similarly, acrolein, a metabolite of cyclophosphamide, is responsible for the increase in the incidence of BC and this effect occurs independently of the association of hemorrhagic cystitis with the same treatment [37, 38].

BC differently affects the two sexes. As stated before, males are more prone to BC than females (4:1 ratio); however, after RC of urothelial cell carcinoma (UCC), the prognosis of female patients is worse than that of male ones [39, 40-42]. Interestingly, survival of females after any cancer treatment is always better than that of males [43] except for BC. Social and cultural parameters may be taken into account to better explain survival of females to general cancers, such as a major awareness of women for their health, a minor exposure to risk factors, a lower age-related mortality interacting with cancer-specific survival. Thus, in this perspective, BC is a tumor behaving differently from the others. Moreover, this is particularly evident if the comparison is made according to cancer stage (Table 1): extrapolation of these data reveals that BC staging T1 to T3 affects both sexes equally as for survival, but a difference is evident for stage T4a [42]. Explanations of these data are still missing, although some environmental parameters (presence of specific carcinogens, anatomical and/or hormonal differences) are discussed in literature [44, 45].

Table 1.

2009 Tumor-Nodes-Metastasis (TNM) Classification of Urinary Bladder Cancer [50]

T Primary Tumor
TX Primary Tumor cannot be Assessed
T0 No Evidence of Primary Tumor
Ta Non-invasive Papillary Carcinoma
Tis Carcinoma in situ (‘Flat’ Tumor)
T1 Tumor invades Sub-epithelial Connective Tissue
T2 Tumor Invades Muscle
T2a Tumor invades Superficial Muscle (Inner Half)
T2b Tumor invades Deep Muscle (Outer Half)
T3 Tumor invades Perivesical Tissue
T3a Microscopically
T3b Macroscopically (Extravesical Mass)
T4 Tumor invades any of the following: Prostate, Uterus, Vagina, Pelvic Wall, Abdominal Wall
T4a Tumor invades Prostate, Uterus or Vagina
T4b Tumor invades Pelvic Wall or Abdominal Wall
N Lymph Nodes
NX Regional Lymph Nodes cannot be Assessed
N0 No Regional Lymph Node Metastasis
N1 Metastasis in a Single Lymph Node in the True Pelvis (Hypogastric, Obturator, External Iliac or Presacral)
N2 Metastasis in Multiple Lymph Nodes in the True Pelvis (Hypogastric, Obturator, External Iliac or Presacral)
N3 Metastasis in a Common Iliac Lymph Node(s)
M Distant Metastasis
MX Distant Metastasis cannot be Assessed
M0 No Distant Metastasis
M1 Distant Metastasis

Socio-economic explanations may be also taken into account when discussing ethnic differences in BC survival. Black people are known to have poorer survival to BC compared to whites, Hispanics and Asian/Pacific Islanders in the United States, especially because blacks show higher BC stage at presentation [46, 47]. However, differences in survival are present even if patients show similar BC stage, grade and treatment [48]. Some Authors [47] conclude that these disparities may be due, at least partly, to disparity in quality of treatment, access to and quality of care, surveillance after primary treatment, and comorbidity. Even marital status might be taken into account, when evaluating cancer survival [49].

Diagnosis, Prognosis and Treatment of Bladder Carcinoma

BC patients at time of presentation may be broadly subdivided into two groups: those having a non-muscle-invasive bladder cancer (NMIBC) staged Ta and T1, and those with a muscle-invasive disease (MIBC), staged T2 to T4 (Table 1) [50, 51]. The former group represents approximately the 75% of the patients, and the latter the remaining 25%. Among patients with NMIBC, 60% are expected to show recurrence of the disease and 1 out of 5 patients will develop a MIBC [52]. Survival rates after 5 years for NMIBC patients is around 88-98%; instead, as for the MIBC patients, about half of them are expected to die within 5 years [53], mainly because of metastases [54] (Table 2). Risk factors associated to progression to MIBC include deeper invasion of the lamina propria, tumor grade and size, concurrent presence of carcinoma in situ (CIS), tumor multiplicity, and recurrence of NMIBC [55]. Tumor types of the BC are urothelial cell carcinoma (UCC, previously called transitional cell carcinoma, TCC), squamous cell carcinoma (SCC), adenocarcinoma, and other sporadic lesions [56]. UCC begins in the cells lining the inner-most tissue layer of the bladder; SCC arises from the squamous cells of the bladder epithelium and it is frequently associated to long-term infection or irritation of this layer; finally, adenocarcinomas usually arise from bladder secretory cells, frequently of urachal origin. At diagnosis, 90% of all BC are UCC, and three fourths of them are papillary tumors localized in the urothelium or in the lamina propria. Less than 8% are classified as SCC and 2% are adenocarcinomas. Recurrence is the main problem for NMIBC patients, since up to 80% of them may relapse; instead, progression is the main problem for patients in T1 stage and CIS [52, 57, 58]. For this type of lesion a ‘risk calculator’ had been created [52], which is available online in the European Organisation for Research and Treatment of Cancer (EORTC) web site (http://www.eortc.org/). For MIBC patients, the main predictors of outcome are lymph node involvement, tumor stage and grade, lymphovascular invasion and histological subtype as for the tumor features; time from diagnosis to surgery, patient age and gender as for clinical factors [59].

Table 2.

Metastases of Invasive BC According to [54]

Place Frequency (%)
Lymph Nodes 78
Liver 38
Lung 36
Bone 27
Adrenal Gland 21
Intestine 13
Heart, Brain, Kidney, Spleen, Pancreas, Meninges, Uterus, Ovary, Prostate, Testes 1-8

Diagnosis of NMIBC (Ta/T1 grade BC plus the flat, high-grade CIS tumors confined to the mucosa) may be performed by urine cytology, ultrasonography, and cystoscopy with description of the tumor (site, size, number and appearance) and mucosal abnormalities. However, a complete and correct transurethral resection (TUR) is essential to make a correct diagnosis and remove all visible lesions [50]. Instead, the standard treatment for patients with MIBC (T2-T4a, N0-Nx, M0) is radical cystectomy (RC) with lymphadenectomy [51]. However, this ‘gold standard’ only provides 5-year survival in about 50% of patients [60]. In order to improve these unsatisfactory results, the use of peri-operative chemotherapy has been introduced. Some studies demonstrate that neoadjuvant cisplatin-containing combination chemotherapy improves overall survival by 5-7% at 5 years and should be considered in MIBC, irrespective of definitive treatment [61, 62], but its use is still a matter of debate [63]. Patients with immobile tumors (stage T4b) will receive chemotherapy or radiotherapy, occasionally followed by salvage cystectomy [53].

To date, recurrence of BC is explained by two different theories: the field-cancerization hypothesis, and the intraluminal seeding and implantation hypothesis. The first theory hypothesizes that multi-focal tumors are a consequence of carcinogen exposure of the entire urothelial layer [64, 65]. The second theory, supported by molecular data, suggests that multi-focal tumors are a consequence of clonal evolution from a single transformed cell [66, 67]. Notably, a recent paper demonstrates that the normal mucosa of human bladder contains multiple stem cells, each responsible for the turnover of the surrounding cells during bladder layers renewals, and that any clone may be replaced by surrounding clones [68].

THE GENETICS OF BLADDER CARCINOMA

Genes Controlling Cell Cycle Regulation

Several Authors report that BC is an example of human malignancy where molecular profiling can be restricted to the analysis of the function of only two genes, FGFR3 and TP53 [69-72]. However, in the last years several data show that this vision of the problem may be limited (Table 3). Several functions are necessary for the maintenance of cell homoeostasis, and alterations in genome stability, cell cycle control, apoptosis, angiogenesis, cell signalling and hormone receptor functions may all drive to neoplastic transformation and promote tissue invasion. The tumor suppressor gene TP53 is the most commonly mutated gene in cancers, including MIBC [73], since it plays essential roles in the regulation of cell proliferation, apoptosis and inhibition of angiogenesis [74]. Many studies demonstrate that altered function of this gene, as well as that of its relative TP63 [75], either because of a mutation in the coding sequence or for alterations in its regulation, may be predictive of a poor outcome in both NMIBC and MIBC; moreover, the levels of the TP53 protein increase in normal urothelium to NMIBC to CIS to MIBC to metastatic BC [76-78]. However, some scientific reports discourage the use of TP53 status alone in BC as a final outcome predictor [73, 79, 80]. TP53 controls the proper function of TP21, another key protein in cell cycle regulation. TP21 [81], together with TP27, is an inhibitor of the G1/S cell cycle transition thanks to its ability to regulate cyclin dependent kinases cdk2/4 [82, 83]. Thus, also these two proteins are good markers of the BC final outcome [78, 83-85]. Indeed, TP27 regulation is under control of Nkx.28, an NK-2 homeobox containing protein that also regulates cyclin D1, FOXO3a (a transcription factor) and the MEK/ERK pathway [86]. The contemporary malfunction of TP53 and TP21 is associated to higher relapse rate and worse survival after RC, compared to TP53 alterations only [87]. Instead, TP27 is a good predictor of outcome for MIBC, but not for NMIBC [76, 77]. As a consequence of the role of TP21/TP27, also cyclins are potentially good markers of BC progression. The primary protein in this stage, with importance in BC, is cyclin E1, whose decreased expression is frequently associated with advanced, metastatic disease [88]. Another cell cycle regulator involved in BC development is the Retinoblastoma (Rb) protein. Rb plays a role in stem cell maintenance, replication and differentiation, by controlling the G1/S transition; nonetheless, its ability to become a molecular predictor of BC outcome seems limited [89]. Finally, it has been recently demonstrated that also Polo-like kinase 1 (Plk1) has a significantly higher expression in BC and it is positively correlated with its grade, stage, recurrence, and metastasic invasion [90].

Table 3.

Potential Biomarkers of BC – Gene Functions and Metabolic Pathways According to Genecards (http://www.genecards.org/) Accessed June 2012. For Each Section, Genes are Listed in Alphabetical Order, According to their Full Name (Column 2)

Symbol(s) Full Name Molecular Function Metabolic Pathways Refs
Genes Controlling Cell Cycle Regulation
CCND1, Bcl1 Cyclin D1 Cyclin Proliferation, G1/S Transition [88, 164]
Cdk2/4 Cyclin dependent kinase 2 and 4 Ser/Thr cyclin dependent kinases Proliferation, G1/S Transition [82, 83]
CCNE1 Cyclin E1 Cyclin Proliferation, G1/S Transition [88]
TP21, p21, CIP1, WAF1, CDKN1A Cyclin-dependent kinase inhibitor 1A Protein Repressor Proliferation, G1/S Checkpoint [81]
TP27, p27, CDKN4, CDKN1B Cyclin-dependent kinase inhibitor 1B Protein Repressor Proliferation, G1/S Checkpoint [76, 77, 82, 83]
TP14, p14, NK4a, INK4, ARF, TP16, p16, CDKN2A Cyclin-dependent kinase inhibitor 2A Protein Repressor Proliferation [171, 187-189]
GATA2, NFE1B GATA binding protein 2 Transcriptional Activator Cell Proliferation [183]
E2F3 E2F transcription factor 3 Transcription Factor Cell Proliferation [162]
EZH2, ENX1, KMT6 Enhancer of zeste homolog 2 Transcriptional Repressor Cell Differentiation [203]
EOMES; TBR2 Eomesodermin Transcriptional Activator Embryo Development [207]
FGFR3 Fibroblast growth factor receptor 3 Tyrosine Kinase Cell Cycle Control; Angiogenesis [131, 132, 217]
GDF-9 Growth differentiation factor-9 Growth Factor Oncosuppressor; Cell Proliferation [202]
HOXA9 Homeobox A9 Transcription Factor Cell Differentiation; Morphogenesis [207]
ID-1, ID1 Inhibitor of DNA binding 1 HLH-Protein Cell Proliferation and Senescence; Cell Differentiation [165]
KRT2A/6B/6C/7/8/10/19/20 Keratins 2A/6B/6C/7/8/10/19/20 Intracellular Structure Cell Activation and Proliferation [172, 207, 208]
KRTAP13-1, KRTAP19-2, KRTAP20-2 Keratin-associated proteins 13-1, 19-2, 20-2 Intracellular Structure Cell Activation and Proliferation [208]
Nkx.28, NKX2-8 NK2 homeobox 8 Unknown Control of p27, cyclin D1, FOXO3a [86]
PLK1 Polo-like kinase 1 Ser/Thr protein kinase Control of Mitosis [90]
POU4F2 POU class 4 homeobox 2 Transcription Factor Cell Differentiation (Putative) [207]
Rb, RB1 Retinoblastoma Transcription Repressor Control of G0/G1 Transition [89]
RARB, HAP, NR1B2 Retinoic acid receptor b2 Hormone Receptor Cell Differentiation [173]
RUNX3, AML2, CBFA3 Runt-related transcription factor 3 Transcription Factor Activation and Repression of Transcription [193, 196]
SOX9, CMD1 Sex determining region Y-box 9 Transcription Factor Chondrogenesis [194]
TP53, p53 Tumor protein p53 Transcription Factor Proliferation, Apoptosis, Angiogenesis [73, 76-78]
TP63, p63 Tumor protein p63 Transcriptional Activator/Repressor Proliferation, Apoptosis [75]
TWIST, TWIST1, ACS3, BPES2 Twist homolog 1 Transcription Factor Cell Differentiation [214]
TBX2 T-box 2 Unknown Cell Differentiation [183]
TBX3 T-box 3 Transcriptional Repressor Cell Differentiation [183]
AKT1, AKT, RAC v-akt murine thymoma viral oncogene homolog 1 Ser-Thr protein kinase Cell Proliferation; Cell Survival; Angiogenesis [137, 138, 166]
c-myc, MYC v-myc myelocystomatosis viral oncogene homolog Regulation of Gene Transcription Cell Proliferation [161]
Genes Controlling Apoptosis
cIAP1, BIRC2, RNF48 Baculoviral IAP repeat containing 2 Protein Inhibitor Inhibition of Apoptosis [99]
BIRC5, Survivin Baculoviral IAP repeat containing 5 Protein Inhibitor Inhibition of Apoptosis; Cell invasion; Regulator of Mitosis [96-98]
Bcl-2, PPP1R50 B-cell CLL/lymphoma 2 Control of Mitochondrial Membrane Permeability Inhibition of Apoptosis [92, 215]
BNIP3 BCL2/adenovirus E1B interacting protein 3 Calcium Repartitioning Cell Survival [116]
c-FLIP, CFLAR CASP8 and FADD-like apoptosis regulator Protein Inhibitor Apoptosis Resistance [100]
CASP3, CPP32, Yama Caspase-3 Cysteine-Aspartic Protein Peptidase Activation of Apoptosis [92, 93]
DAPK Death Associated Protein kinase 1 Serine-Threonine Kinase Apoptosis; Cell Survival [177, 188, 215, 216]
CD95L, FASLG Fas ligand TNF superfamily 6 Ligand for Fas Activation of Apoptosis [94, 100]
FOXO3a, AF6q21 Forkhead box 3A Transcription Factor Activation of Apoptosis [86]
GDF15, MIC1, PLAB Growth Differentiation factor 15 Unknown Activation of Apoptosis [206]
PMF1 Polyamine-Modulated Factor 1 Polyamine Homeostasis Control of Cell Growth and Death [212]
RASSF1A Ras association domain family member 1 Protein Inhibitor Cell Proliferation Inhibitor [187, 189-191, 215]
TMEFF2, tomoregulin-2 Transmembrane protein with EGF-like and follistatin-like domains 2 Activator of Phosphorilation Cell Survival [206]
DR4, TNFRSF10A Tumor necrosis factor receptor superfamily 10A Death Receptor Activation of Apoptosis [101]
DR5, TNFRSF10B Tumor necrosis factor receptor superfamily 10B Death Receptor Activation of Apoptosis [101]
TRAIL, TNFRSF10C Tumor necrosis factor receptor superfamily 10C Protein Inhibitor Apoptosis Resistance [101]
Fas, APT1, TNFRSF6 Tumor necrosis factor receptor superfamily 6 Death Receptor Activation of Apoptosis [94, 95, 205]
DR3, TNFRSF25 Tumor necrosis factor receptor superfamily member 25 Death Receptor Activation of Apoptosis [205]
Genes Controlling Angiogenesis
bFGF, FGF2 Basic fibroblast growth factor Heparin Binding Pro-Angiogenesis; Mitogen [102]
EDNRB Endothelin receptor type B G-protein-coupled Receptor Regulation of Angiogenesis [204]
Prolidase, PEPD Peptidase D Metalloproteinase Collagen Metabolism; Pro-angiogenesis [169]
THBS1, TSP1 Thrombospondin-1 Adhesive Glicoprotein; Heparin Binding Inhibitor of Angiogenesis [102]
VEGF Vascular Endothelial Growth Factor Signaling Protein Cell Replication and Migration; Inhibition of Apoptosis [103, 104]
Genes Controlling Cell-Cell Interactions
ADAM12 ADAM metallopeptidase Domain 12 Metalloproteinase Multinucleate Cell Formation [127]
ADAM17 ADAM metallopeptidase Domain 17 Metalloproteinase Release of Cell Surface Proteins [127]
ADAM28 ADAM metallopeptidase Domain 28 Metalloproteinase Cell Adhesion [126]
APC Adenomatous Polyposis Coli Antagonist of Wnt Pathway Cell Migration and Adhesion; Apoptosis [191, 204]
AR, DHTR, SBMA, AIS Androgen Receptor Steroid Hormone Receptor Cell Growth, Differentiation and Function [129]
Annexin10, ANXA10 Annexin A10 Unknown Cell Migration [121, 122]
APOE, LPG Apolipoprotein E Catabolism of Lipoproteins Cell Function [130]
BLCA-4 Bladder Cancer A4 Transcription Factor Metastasis Formation [167, 168]
BAMBI BMP and activin membrane-bound inhibitor homolog Signal Receptor Metastasis invasion; Cell Movement [201]
LASS2 Ceramide synthase 2 Sphingolipid Synthesis (Putative) Metastasis Suppressor [123]
COL1A2 Collagen Type 1a2 Collagen Extracellular Matrix Formation [219]
CTTN, EMS1 Cortactin Unknown Cytoskeletal Organization; Regulation of Cell-Cell Junctions; Regulation of Invasiveness [124]
CDH1 E-cadherin Calcium-Dependent Membrane Protein Cell Adhesion [128, 129, 184]
ERa, ESR1 Estrogen Receptor Alpha Steroid Hormone Receptor Cell Growth, Differentiation and Function [129]
FGB Fibrinogen Beta Chain Precursor Polymerization of Monomers Cell Adhesion [130]
HYAL-1, LUCA1, NAT6 Hyaluronoglucosaminidase 1 Degradation of Hyaluronic Acid Cell Proliferation, Migration and Differentiation [218]
MAPK Mitogen-activated protein kinase family Ser-Thr kinase Cell Growth, Adhesion, Survival and Differentiation [137, 138]
NID2 Nidogen2 Membrane Glycoprotein Cell Adhesion [214]
PTEN, BZS, MMAC1, Phosphatase and tensin honolog Protein and Lipid Phosphatase Cell Migration Inhibition [178]
PIK3CA Phosphoinositide-3-kinase catalytic alpha polypeptide Lipid Kinase Cell Growth, Survival, Proliferation, Motility and Morphology [119, 137, 138]
PLCG1 Phospholipase C, g1 Phospholipase Actin Organization; Cell Migration [137, 138]
PFN1 Profilin-1 Actin Binding Cytoskeletal Organization [125]
PRKCI Protein kinase C Ser-Thr kinase Modulation of Membrane Structure [137, 138]
RIN1 Ras and Rab interactor 1 Ras Effector Protein Cytoskeletal Remodeling [115]
RAS Rat sarcoma viral oncogene family GTPase Activation of Mitosis [114]
SFRP Secreted frizzled receptor protein family Protein Receptor Metastasis invasion; Cell Movement [199, 200]
SERPINA1 Serpin peptidase inhibitor 1 Alpha-1 Antitrypsin Inhibition of Elastase [130]
STAT1, STAT91 Signal transducer and activator of transcription 1 Signal Transducer Response to Growth Factors [139]
TIMP-3 TIMP metallopeptidase inhibitor 3 Proteinase Cell Remodeling [213]
TSC1, LAM, hamartin Tuberous sclerosis 1 Protein Inhibitor Inhibition of Nutrient-Mediated Cell Growth [120]
UPK Uroplakin Integral Membrane Proteins Cytoskeleton Regulation [107]
HER2, ERBB2 v-erb-b2 erythroblastic leukemia viral oncogene homolog 2 Epidermal Growth Factor Receptor; Tyrosine Kinase Activation of Mitosis [108, 109]
Src, ASV v-src sarcoma viral oncogene homolog Tyrosine Kinase Cell Proliferation, Survival and Migration [137, 138]
Other Genes
hOGG1 8-oxoguanine DNA glycosilase Glycosylase DNA Base Excision Repair [142]
C10orf116 Chromosome 10 open reading frame 116 Unknown Unknown [172]
CYP1B1 Cytochrome P450 1B1 Oxidation Chemical Modification of Various Compounds; Detoxification [151]
ERCC1 Excision repair cross-complementing rodent repair deficiency, group 1 Endonuclease DNA Nucleotide Excision Repair [143]
ERCC2, XPD, TFIIH Excision repair cross-complementing rodent repair deficiency, group 2 Helicase Double-Stranded DNA Breaks Repair [142, 144]
GSTM1 Glutathione S-transferase mu 1 Chemical Conjugation Detoxification [149, 150]
GSTP1 Glutathione S-transferase pi 1 Chemical Conjugation Detoxification [149, 150]
GSTT1 Glutathione S-transferase theta 1 Chemical Conjugation Detoxification [149, 150]
HMGB1, SBP1 High mobility group box 1 DNA Bending DNA Transcription and Repair [140]
SIRT2 Histone Deacetylase Chromatin Remodeling Gene Expression Control [124]
HDAC6 Histone Deacetylase 6 Chromatin Remodeling Gene Expression Control [124]
LRG1 Leucine-rich alpha-2-glycoprotein 1 Unknown Unknown [130]
MTHFR Methylenetetrahydrofolate reductase NAD(P)H Reductase Nucleotide Biosynthesis [141]
NBS1, NBN Nibrin DNA Damage Signaling Double-Stranded DNA Breaks Repair [144]
PARP1, PPOL Poly(ADP-ribose) polymerase 1 DNA Polymerase Base Excision DNA Repair [146]
SYNPO2, Myopodin Synaptopodyn 2 Actin-binding Protein Unknown [211]
UGT UDP glucuronosyltransferase family Chemical Modification of xenobiotics Detoxification [148]
VIM Vimentin Type III Intermediate Filament Cell Structure [206]
XPC, Rad4, P125 Xeroderma pigmentosum, group C DNA Bending Nucleotide Excision DNA Repair [144]
XRCC3 X-ray repair complementing defective repair in Chinese hamster cells 3 Unknown Double-Stranded DNA Breaks Repair by Homologous Recombination [145]
ZIC4 Zic family member 4 DNA Binding Unknown [183]
ZNF154 Zinc finger protein 154 Transcriptional Regulation (Putative) Unknown [191]

Genes Controlling Apoptosis

Even if cell cycle progression is somehow deregulated, the cell may still remain under control thanks to the activation of the apoptotic pathways; indeed, their inactivation is frequent in all cancers and, as expected, also in BC [91]. Among the others, we can here remember the role of the protease Caspase-3, frequently associated to high grade BC [79, 92, 93]; Fas, a death receptor which is down-regulated in many BC samples [94, 95]; Bcl-2, an anti-apoptotic protein which is up-regulated in one third of the RC specimens and that is able to promote chemotherapy resistance in neoplastic cells [92]; and Survivin, another inhibitor of apoptosis acting on caspases and promoting tumor cell invasion; Survivin is up-regulated in BC, especially in high grade tumors [96-98]. Another good candidate as a molecular BC marker in this group is cellular inhibitor of apoptosis protein 1 (cIAP1), belonging to the same protein family (IAPs) of Survivin; cIAP1 is a nuclear shuttling protein able to block both the intrinsic and the extrinsic apoptotic pathways by inhibiting caspases activity; indeed, cIAP1 over-expression is highly correlated with BC recurrence, progression, resistance to chemotherapy and poor prognosis in MIBC [99]. Finally, we remember the FLICE-inhibitory protein (c-FLIP) long splicing form, which has a pivotal role in TRAIL- and CD95L-mediated apoptosis resistance [100], and the TRAIL protein itself that, together with its death receptors DR4 and DR5, is a good prognostic marker for BC [101].

Genes Controlling Angiogenesis

Solid malignancies need food and oxygen supplies to self-maintain. It is known, for example, that a high value of microvessel density is a potential predictor of poor prognosis in BC and that this parameter is also associated with lymph node metastasis formation [102]. Thus, also genes involved in angiogenesis play an important role in BC. Beyond the already cited TP53, proteins of the vascular endothelial growth factor (VEGF) family promote cell replication and migration, and are up-regulated in BC specimens, especially those of high grade [79]; VEGFs influence metastasis development and, consequently, prognosis [103, 104]. Similarly, the basic fibroblast growth factor (bFGF), with pro-angiogenic activity, is present in urine samples from BC patients, but it is absent in controls [105]; moreover, its amount increases with more advanced stages of the disease [102]. Thrombospondin-1 is an important component of the extracellular matrix and a potent inhibitor of angiogenesis, and is involved in BC progression as well [102]; however, its use as a prognostic marker heavily relies on the concurrent expression of TP53 [102] since the expression of these two proteins is strongly linked in BC development.

Genes Controlling Cell-Cell Interactions and Signalling

The neoplastic mass, established and supplied with nutrients, having lost internal cell cycle control and escaped intrinsic/extrinsic induction of apoptosis, must face interactions with surrounding, likely normal, tissues and organs. Uroplakins [106] are integral membrane proteins specific of the urotelium; they are expressed in approximately 50% of MIBC, but not in other tumors and they are absent in the more aggressive stages of MIBC [107]. Also signalling pathways are of great importance for cancer ‘survival’ inside the body; in this perspective, BC is not different from other cancers. Proteins like those of the epidermal growth factor receptor (EGFR) family are indeed involved in poor prognosis. For HER2, results published so far are puzzling, since some studies emphasize a correlation between its high expression and poor prognosis [108, 109] while other studies do not [110, 111]; a similar situation occurs for other members of this protein family [112, 113]. Instead, the implication of the RAS proteins [114] and of their effectors (such as RIN1 [115] or BNIP3 [116], both effectors of HRAS) in BC is widely accepted: approximately 13% of specimens has a mutation in one of the components of the RAS gene family (HRAS, NRAS, KRAS2) and one report shows that RAS and FGFR3 (see below) mutations are mutually exclusive in BC [117]. The role of RAS in activating mitosis (interaction with MAPK and PI3K) also suggests a role for these two proteins in BC. Indeed, PTEN and PIK3CA are both involved in BC, and are both part of the phosphatydilinositol 3-kinase (PI3K) pathway [118, 119]. Downstream of PI3K (improper) activation in BC, the phosphorilation of AKT promotes mTOR pathway activation [120], a typical feature of this cancer. In this pathway, inactivating mutations of tuberous sclerosis protein 1 (TSC1) may play similar roles in it. Annexin family is a group of twelve proteins playing a role in several processes, including tissue growth; among them, Annexin10 (ANXA10) had been validated as a good marker for the analysis of the aggressiveness of BC [121, 122]. Moreover, ANXA10 and TP53 act synergistically (inverse correlation) towards high grade/stage NMIBC to MIBC progression, likely because down-regulation on ANXA10 promotes cell migration [122]. Another gene related to cell migration is longevity assurance homologue 2 of yeast LAG1 (LASS2), which is a tumor metastasis suppressor gene; a recent report shows that this gene is down-regulated in BC and it is associated to poor clinical prognosis [123]. Other worth mentioning proteins involved in cytoskeleton homoeostasis and BC invasiveness are Cortactin [124] and the actin binding protein Profilin-1 [125]. ADAM28 [126], ADAM12 and ADAM17 [127] are metalloproteinases, members of a family of membrane-anchored glycoproteins with adhesive properties, that are frequently expressed in several types of tumors; among them, ADAM28 is considered another good molecular marker of BC [126]. To date, the role of androgen, oestrogen and progesterone receptors in BC are a debated topic [127]; however, recent scientific reports had been published indicating that the estrogen receptor beta may be a prognostic marker of recurrence-free rate in NMIBC, potentially through suppression of the cadherin switch [128, 129]. Also androgen receptor and oestrogen receptor alpha may be deregulated in BC [129]. Finally, other possible candidate proteins as BC molecular markers have been reported in a recent proteomic analysis [130], the most promising of which are apolipoprotein E (APOE), leucine-rich alpha-2-glycoprotein (LRG1) and, above all, fibrinogen beta chain precursor (FGB) and alpha-1 antitrypsin (SERPINA1).

Among the signalling proteins, one of the best characterized in BC is the fibroblast growth factor receptor 3 (FGFR3) [131, 132]. This protein is involved in several types of diseases and cancers, such as skeletal dysplasias (achondroplasia, thanatophoric dysplasia), skin disorders and cancer (epidermal nevi, seborrheic keratosis, acanthosis nigricans, myeloma, seminoma, bladder and cervical carcinoma), cartilage growth anomalies (inhibition of chondrocyte proliferation) [133]. FGFR3 is a tyrosine kinase receptor implicated in cell cycle control and angiogenesis. Improper activation of this gene (by mutations in the coding sequence or by alterations in its regulatory pathways) is present in 60% of BC samples, as underlined for example in studies of tissue microarrays [134, 135]; indeed, FGFR3 improper function is a typical feature of NMIBC [136]. FGFR3 direct and indirect interactors include MAPK, phospholipase Cγ, protein kinase C, Src, PI3K, AKT [137, 138] and STAT1 (leading to TP21 accumulation) [133, 139].

Genes Involved in Other Cellular Functions Important for BC Development

None of the aforementioned genetic alterations, taken alone, is sufficient to predict the outcome of the disease, nor simply to assess the stage and/or grade of the BC. Cancer is a multi-step disease, thus several genes must be mutated or deregulated in a cell to promote its neoplastic transformation. Thus, also genes controlling genome integrity (such as those involved in DNA replication or repair) are of crucial importance because of their “mutagenic” effect. Genes like that coding for the high-mobility group box 1 protein (HMGB1, having intranuclear and extracellular functions and involved in DNA transcription and repair) [140], methylene tethrahydrofolate reductase (MTHFR, an enzyme necessary for nucleotide biosynthesis) [141], the single strand DNA damage repair proteins hOGG1 (a glycosylase involved in base excision repair), XPD (one of the proteins affected in Xeroderma pigmentosum disease) [142] and ERCC1 (involved in nucleotide excision repair) [143], and the double-stranded DNA breaks repair proteins ERCC2, NBS1, XPC [144], XRCC3 [145] and PARP1 [146], are all involved in the formation/development of BC.

The alteration of the function of detoxifying proteins [147] such as UDP-glucuronosyltransferases (UGTs), which facilitate cellular removal of bioactivated forms of aromatic amines found in tobacco smoke and industrial chemicals [148], or glutathione S-transferases (GSTT1, GSTM1, GSTP1) [149, 150] and CYP1B1 (a member of the cytochrome P450 superfamily of enzymes) [151], all implicated in the inactivation of procarcinogens, are likely causes of BC susceptibility, by increasing the number of (eventually unrepaired) DNA lesions.

Of course, the more genes are analyzed, the more precise the diagnosis is; however, using mathematical approaches, it has been demonstrated that only a “few” genes – as low as eleven [266] or even less [153] – may be actually analysed to have a sufficiently accurate diagnosis.

Innovative Approaches for the Identification of New Genes Involved in BC

Recently, new approaches were developed to “indirectly” identify genes involved in BC. One method is the identification of peptides inside urine, to discover patients with MIBC using a minimally invasive technique. The approach is promising, however the reported panel of peptides may be not highly specific [152, 153]. A second approach takes advantage of the metabonomics, i.e., the investigation of the presence of metabolites potentially useful as cancer biomarkers and not directly related to any genotype, expression profile, or even proteome [154, 155]. Using this approach, Huang and co-workers [156] were able to demonstrate that carnitine C9:1 and component I may be used together to discriminate, with high specificity and sensitivity, patients with BC, especially those with a low grade disease; however, why these molecules are particularly abundant in these patients is not yet understood. Other twelve potential markers were also identified, which need further validation or have lower specificity/sensitivity. Identifying the proteins (and, consequently, the corresponding genes) responsible for the alteration in the total amount of these metabolites may shed light on other, still unknown, metabolic malfunctions leading to neoplastic transformation in BC.

EPIGENETIC CHANGES IN BLADDER CARCINOMA: TRANSCRIPTIONAL CONTROL

Genetic Up- Or Down-Regulation Occurs in BC Cells

Genetic changes in gene expression and/or function are inheritable, frequently irreversible and due to permanent modifications of DNA, either because of small nucleotide sequence changes (point mutations) or because of gross chromosomal rearrangements (translocations, deletions, duplications and its correlated copy-number variations that may quantitatively increase gene expression). Indeed, chromosome 9 total and/or partial loss [157] is associated to BC recurrence, and at least four regions of this chromosome seem crucial in this phenomenon [158]. Epigenetic changes are similarly inheritable, but they are potentially reversible and they are not mediated by alterations in DNA sequence or chromosome structure; instead, they often involve changes in interphase chromatin architecture and, consequently, function. Well known epigenetic gene control mechanisms involve histone modifications (which influence gene expression at the transcriptional level), modification of the DNA itself, such as methylation (that acts at the transcriptional level as well), or even changes mediated by non-coding RNAs (post-transcriptional silencing, see next section). Several epigenetic alterations had been identified in BC in the last years [159, 160].

As for gene over-expression, it is long known its role in BC, although the involved mechanisms are not always clear (gene amplification by copy number variations, chromatin hypo-methylation, small chromosomal rearrangements invisible at the light microscope). Examples of this type involve the (hyper-)activation of (onco)genes such as c-myc [161], E2F3 [162], Rb [163], CCND1 [164], ID-1 [165] and AKT1 [166], or the increased level of the highly specific marker BCLA-4 (a transcription factor usually over-expressed in both BC and surrounding benign tissues, but not in normal urothelium [167, 168]). Also prolidase (a metalloproteinase) and its regulator, nitric oxide (NO), may promote angiogenesis and metastasis formation [169]. On the other hand, hypo-activation of key genes is involved in BC development as well, engaging for example TP53, Rb again [170] and p16 [171] genes. Direct analysis of mRNA from blood cells allowed the identification of two more mis-regulated genes in BC vs. control cells, namely C10orf116 (of unknown function) and the KRT19 keratin-coding gene [172]. Several studies are still running, and the list of altered gene expression is getting longer every week [173].

The Role of Histone Modification in Chromatin Remodeling and Gene Expression

Histone modifications have been extensively described in BC, at the genomic level, in recent works [174, 175]. This phenomenon may lead to either gene silencing or over-expression; both these phenomena were found in BC [175], and the in-depth study of down-regulated sequences allowed the identification of seven genome regions which are significantly associated with the presence of CIS. On the other hand, those bioptic samples identified for harbouring FGFR3 mutations, did not display the identified silencing profile. Thus, the silencing phenotype is specifically associated with CIS, high tumor grade and stage, MIBC development and very low frequency of FGFR3 mutations. Interestingly, some of these seven regions are shared with other cancers, but their number and association is tumor-specific. This means that not only any region itself is necessary yet insufficient for neoplastic transformation, but also that particular combinations of different regions may be distinctive of different types of cancer. Thus, any histone-mediated silenced region specific for BC may not be silenced in another tumor, and vice versa. As expected, alterations in the amount of proteins directly related to histone modification are responsible as well of the BC final prognosis. For example, histone deacetylase 6 (HDAC6) and histone deacetylase SIRT2 are both involved in metastasis formation in BC, mainly (but not only) because they also target the cortactin gene, encoding for a cytoskeletal protein [124].

The Role of Chromatin Remodeling by DNA Methylation

DNA methylation plays an important role in gene silencing and BC development, too [176]. Several genes were found to have an altered expression as a consequence of methylation-mediated gene silencing, especially in the CpG islands surrounding the open reading frames of key genes [177-183]. In the last years, the record of genes epigenetically silenced is growing at high speed. A low level of expression of E-cadherin (a trans-membrane protein involved in cell adhesion) is associated to neoplastic transformation and its lower levels correlate with higher invasiveness and metastatic properties of human cancers, including BC; one of the means of its down-regulation is indeed methylation of the CpG island located at the 5’ end of the gene [184]. Interestingly, in their report [184], the Authors also show that this same methylation may occur spontaneously in elderly individuals. In this perspective, it is worth remembering that hyper-methylation is an aspect of cell reaction to the environment (broad sense), too; not only age, but also gender, smoking history, and cancer development (stage, grade, localization, progression) influence chromatin remodelling [178, 185, 186]. Therefore, both ‘external’ (environment-derived, such as smoke) and ‘internal’ (age, gender) signals of gene silencing may act synergistically to promote cell transformation and BC development. Together with E-cadherin, also the methylation-derived low levels of TP16, TP14, DAPK and RASSF1A may be used to predict BC recurrence and prognosis [187-189]. In particular, RASSF1A [190], together with APC, may be a good prognostic factor for tumor reappearance; moreover, patients with methylated APC or RASSF1A coding genes were also significantly associated with shorter recurrence-free survival [191]. Nonetheless, RASSF1A role as tumor stage and grade marker is still a debated topic [191]. As for p16, its role has been demonstrated in BC as a consequence of silencing and/or haplo-insufficiency [171]; remarkably, p16 silencing may be also a consequence of uropathogenic E. coli infection [192]. Also transcription factors are implicated, as expected, in these phenomena. RUNX3 and SOX9, for example, are silenced in BC in at least 70% of the analysed samples [193, 194]; RUNX3 turned out to be a good candidate for the analysis of BC recurrence and development [195, 196], while SOX9 is part of a list of 26 hypermethylated genes in BC-derived tissues [194]. The Wnt genes are part of a well studied pathway involved in neoplastic transformation, and their role has been repeatedly demonstrated in BC, especially when the pathway is up-regulated as a consequence of hyper-methylation of its antagonists [197, 198]. For instance, epigenetic silencing of the genes encoding the secreted frizzled receptor proteins (SFRP), antagonists of the WNT pathway, leads to constitutive WNT signalling and promotes the invasive phenotype of tumors [199, 200]. The activin membrane-bound inhibitor (BAMBI) gene is epigenetically silenced in high grade BC and it is correlated to high aggressiveness and invasiveness [201]. It is likely that BAMBI plays its role by fine-tuning the TGF beta signaling pathway [201], which in turn has an essential role in many cellular processes such as cell growth and differentiation, apoptosis and cellular homeostasis. It is important to remember that also growth differentiation factor-9 (GDF-9), which belongs to the TGF beta superfamily of proteins, has been recently identified as a potential oncosuppressor in BC development [202]. Also the role of the EZH2 gene (which acts as a repressor on various target promoters) had been investigated, showing that EZH2 protein expression and APAF-1 methylation are related to TCC progression and invasiveness [203]. Another hyper-methylated signalling pathway in BC involves the endothelin receptor type B (EDNRB), a G-protein-coupled receptor that activates a phosphatidylinositol-calcium second messenger system. This protein, together with APC (adenomatous polyposis coli), TERT_a and TERT_b (telomerase subunits), are part of a panel of markers able to discriminate between tumors and controls using a minimally invasive technique [204]. Notably, in the same report the Authors also found that the tumor necrosis factor receptor superfamily member 25 (TNFRSF25) coding gene is instead hyper-methylated in controls, but not in BC cells. Conversely, another member of the same superfamily, namely TNFRSF6 (also known as Fas, involved in apoptosis) is down-regulated at both mRNA and protein level in BC cell lines and tissue samples of bladder urothelial carcinoma because of DNA methylation [205]. The same technique, that is the analysis of methylated genes from voided urine, allowed two more groups to identify other genes which are silenced in the same way, (i) GDF15 (growth differentiation factor 15, involved in apoptosis), TMEFF2 (tomoregulin-2, important for cell survival), VIM (Vimentin, a type III intermediate filament) [206], and (ii) ZNF154 (a zinc finger protein coding gene), POU4F2 (a transcription factor), HOXA9 (a homeobox motif containing protein), EOMES (eomesodermin, containing a DNA binding domain) [207]. In particular, the latter work [207] also highlights that BC-derived cells are a mosaic of genomic loci that may be either hypo- or hyper-methylated. Besides the hyper-methylated genes recorded above, there is hypo-methylation, and thus over-expression in malignant cells, of the small proline rich proteins (SRPP) coding genes located on chromosome 1 (SPRR1A/2D/3); of five keratins on chromosome 12 (KRT2A/6B/6C/7/8) (see also [208]); of three keratins on chromosome 17 (KRT10/19/20); of the keratin-associated proteins coding genes on chromosome 21 (KRTAP13-1, KRTAP19-2 and KRTAP20-2); the entire chromosome 21 seems indeed a target of differential methylation in BC cells vs. non malignant cells [207], and this could be related to the “protection” against urological cancers of patients with the Down syndrome [209]. These data fit well with the above-mentioned relationships of histone-mediated silenced/ activated regions; indeed, Down patients are protected against BC but are more prone to leukaemia [210], indicating that the pattern of genomic loci activated/silenced is tumor-specific as suggested before.

The list of methylated genes whose inactivation is related to BC formation and development is increasing every week. Similarly to what described in the previous section, also in this case many cellular processes may be affected. As for intracellular events, we may remember the Myopodin, an actin-binding protein [211]; PMF1, involved in polyamine homeostasis that, in turn, controls cell growth and death [212]; the ARF tumor suppressor [213]; the oncogene TWIST, a transcription factor [214]; several pro-apoptotic proteins such as BCL2, TERT, RASSFIA [215] and DAPK kinase [177, 215, 216]; and last but not least, as expected, FGFR3 [217]. As for extracellular events, i.e., proteins acting mainly in the pericellular matrix, controlling cell adhesion and potentially able to promote metastatic invasion, we may remember TIMP-3, a metalloproteinase-3 inhibitor [213]; Nidogen2 (NID2) [214]; HYAL-1, involved in the degradation of the hyaluronic acid [218]; and collagen type 1α2 [219].

EPIGENETIC CHANGES IN BC: THE ROLE OF SMALL NON-CODING RNAs IN POST- TRANSCRIPTIONAL SILENCING

Non-Coding RNAs and BC Development

It is well known that not all transcribed genes encode a protein – for example, tRNA, rRNA, telomerase RNA. In recent years, other non-coding RNAs (ncRNA) have been discovered and involved in numerous mechanisms of gene expression control, mostly silencing. Many of these ncRNA had been identified in BC and several reports indicate their pivotal role in its development (Table 4). Among the others, the long (>400 nucleotides) non-coding RNA (lncRNA) named UCA1 has been recently added to the class of ncRNA involved in BC formation [220, 221]. In particular, this lncRNA seems to play a central role in cell cycle regulation by indirectly acting on the PI3K-AKT pathway through the CREB protein deregulation [222].

Table 4.

Record of Non Coding RNAs (ncRNA) Cited in the Text that are Important in BC Formation and/or Development

ncRNA Target(s) Reference(s)
miR1 SRSF9/SRp30c [244]
miR100 FGFR3 [235]
miR125b E2F3 [242, 243]
miR129 GALNT1 [259]
SOX4
miR143 ERK5/MAPK7 [254, 255]
AKT
miR1826 CTNNB1 [253]
MEK1
VECFG
miR195 CDK-4 [246]
miR200 ERRF-1 [247]
ZEB1 [248-250]
miR205 TP53 [245]
PTEN
c-erbB-3
cdc42
Yes
mir21 TP53 [228, 234]
TIMP3 [236]
Bcl-2
PTEN [237, 238]
TPM1 [239]
MSH2 [240]
E2F3 [162, 241]
miR221 TRAIL [257, 258]
miR31 FGFR3 [228]
mir449a CDK6 [251]
CDC25a
TP130
miR493 FZD4 [252]
RhoC
miR99a FGFR3 [234]
UCA1 CREB [222]

Note that most of the Listed Target Genes are also Present in Table 3, Indicating that the Same Gene Might be Involved in this Pathology Either Because of Mutations in the Coding Sequence or Because of its Mis-Regulation.

The small ncRNA (micro ncRNA, also known as miRNA or miR) are a hot topic for the study of their role in cancer [223]. miRNA are usually 22nt long nucleotide sequences which are either transcribed of their own, or they are part of another RNA [224]; they may also be arranged in cluster in some genomic loci, counting more than 50 members [224, 225]. miR structure, indispensable for their function, is an hairpin able to bind target protein-coding mRNA by complementarity. The target is usually one of the ends of an mRNA molecule, but also internal sequences may be used as binding sites; the pairing might not necessarily be a perfect match, indicating that a single ncRNA is potentially able to target several mRNA [226]. Silencing may be obtained either by impeding translation, or by mediating target destruction; however, also up-regulation had been found [226, 227]. Their number in the human genome is now above 1,100 (http://www.microrna.org/microrna/home.do) and still rising. The discovery that miR are involved in neoplastic transformation attracted several scholars, and the number of scientific reports identifying new miR in carcinogenesis is increasing steadily. As expected, both miR [228], and Dicer (one of the proteins responsible for their maturation) [229] are involved in urologic cancers. Interestingly, the role of miR on gene expression depends upon their expression itself, which may be regulated either genetically or epigenetically [225, 230, 231], increasing the level of complexity of this mechanism of gene control. Thus, in some cases it has been found that some miR are down-regulated (thus, likely, this will bring to up-regulation of target mRNA); in other cases, miR are up-regulated, causing the opposite effects [232, 233].

miR and Their Targets in BC Formation and Development

If we think of up-regulated target mRNA as oncogenes, and down-regulated mRNA as oncosuppressors, the link between miR and BC becomes evident (Table 4). Indeed, it has been demonstrated that the FGFR3 mRNA is a target of miR31, miR99a and miR100 [228, 234, 235]; miR21 targets TP53 [228, 234], TIMP3 (a metalloproteinase involved in the degradation of the extracellular matrix) and Bcl-2 (a regulator of apoptosis) mRNA [236], the tumor suppressor genes PTEN [237, 238] and tropomyosin 1 (TPM1) [239] and, probably, other predicted mRNA targets involved in BC genesis such as MSH2 (coding a mismatch DNA repair protein) [240] and the transcription factor E2F3 [162, 241], which is also a target of miR125b [242]; E2F3 regulation by miR is also dependent upon the Pumilio translational repressor [243]; miR1 induces apoptosis by inhibiting the mRNA of the serine/arginine-rich 9 (SRSF9/SRp30c) protein, important for alternative pre-mRNA splicing of apoptosis-related genes [244]; miR205 is potentially able to target at the same time TP53, PTEN, c-erbB-3 (a member of the EGFR family of receptor tyrosine kinases), cdc42 (a small GTPase of the Rho-subfamily) and Yes (a tyrosine kinase coding gene belonging to the src family of genes) [245]; miR195, which in other cancer types recognizes different mRNA, in BC seems to identify the cyclin-dependent kinase 4 (CDK-4) mRNA as a preferential target [246]; miR200 family acts on the expression of both ERRF-1, a regulator of EGFR [247] and of ZEB1, a transcriptional repressor of E-cadherin [248]; miR200 family may in turn be silenced by TWIST1 (a transcription factor), thus repressing E-cadherin gene by up-regulation of ZEB1 [249, 250]; mir449a acts as a tumor suppressor by down-regulating CDK6, CDC25a (which controls Rb phosphorilation status) and promoting p130 (a Rb-related protein) expression increase [251]; miR493 silences the expression of FZD4 (a transmembrane receptor) and RhoC (a G protein, part of the Ras superfamily of signaling proteins) genes, thus acting itself as an oncosuppressor [252]; miR1826 down-regulates at the same time CTNNB1 (Wnt/beta-catenin pathway), MEK1 (Ras pathway) and VECFG (a growth factor, part of the Ras pathway) [253]; ERK5/MAPK7 and AKT kinases mRNA are both targets of miR143 [254, 255] which, in turn, if down-regulated, promotes the up-regulation of the proto-oncogene plasminogen activator inhibitor-1 (PAI-1), a common marker of solid tumors [256]; miR221 controls apoptosis through its action on TRAIL [257, 258]; miR129 has two putative targets, namely GALNT1 (involved in post-transcriptional glycosylation of proteins in the Golgi apparatus) and SOX4 (a transcription factor) [259]. To further complicate this scenario, it is noteworthy to remember that some of the abovementioned proteins, regulated by miR, are in turn able to regulate miR expression, like TP53, TP63 and TP73 [260]; furthermore, some of the components of the miR maturation machinery are themselves under miR control [260, 261].

Genome-Wide Approaches Allow the Identification of New miR Involved in BC

Likely, the list of miR targeting genes involved in BC formation will get longer in the next few years. The approach to identify new miR involved in BC is usually a large-scale genetic profiling, where the expression of RNA cellular content is analysed in parallel for several hundreds of genes, in both patients and healthy controls. In this way, in year 2007 it was possible for the first time to identify 10 up-regulated miR in BC [257]. Two years later other studies revealed the presence in BC of both up- and down-regulated miR [259, 262, 263]; then, it was reported that their presence might be revealed also in urine samples of BC patients [264, 265]. In the same years it was also suggested that miR analysis might permit distinguishing high- and low-grade BC; in particular, although with some exceptions, down-regulation may be typical of low-grade BC, while up-regulation is more common in high-grade cancers [234]. Interestingly, also miR expression profile of low-grade BC is different from the high-grade one, indicating that diverse miR are involved in BC and that they may have different effects, likely because they target distinct mRNA [235, 266]. As for regulation, recent data seems to suggest, though, that in BC there is a predominance of miR down-regulation [267, 268], while up-regulation is not common, irrespective of tumor stage and grade [267]. Using genomic approaches, also similarities may be studied: in a recent work [233] the Authors found a number of miR which are deregulated in the same way in three different genitourinary cancers, specifically transitional cell carcinoma (TCC), clear cell renal cell carcinoma (ccRCC) and testicular germ cell tumor (TGCT), indicating that some deregulations are tissue-specific while others are cancer-specific. In more detail: regulation of cell adhesion is impaired in all three tumors, but calcium signalling pathways seem to be specific for TCC and TGCT only; instead, several pathways are deregulated in both TCC and ccRCC, such as TP53 signalling, regulation of cell cycle, actin filaments behaviour and focal adhesion. On the other hand, pathways such as cytokine-cytokine receptors or PPAR signalling are impaired in both TGCT and ccRCC, but not in TCC. Finally, also TCC-specific pathways had been identified; among the others, there are drug metabolism and P450-mediated metabolism, MAPK signalling, chemokine signalling and renin-angiotensin system. This approach also allowed the Authors to subdivide analyzed miR into three clusters: the first, dominated by up-regulated oncogenes; the second, enriched in down-regulated (candidate) tumor suppressor genes; the third, maybe the most interesting, in which the same genes are up-regulated in ccRCC but down-regulated in both TCC and TGCT. This last cluster is probably a “cancer specific” set of genes, while the first two may potentially be the “tissue specific” genes responsible of neoplastic transformation of the genitourinary tract cells.

The situation recently became even more complicated. Recent studies reported that, at least in some cases of BC, it is not the total level of any one miR, but the ratio between two miR that might be important in carcinogenesis. For example, it has been showed that the miR126:miR152 ratio in urine permits the detection of bladder cancer with a sensitivity of 72% and a specificity of 82% [264], while another group explained that the miR21:miR205 expression ratio allows the distinction between invasive and non-invasive BC with a sensitivity of 93% and a specificity of 87% [245].

CONCLUSIONS

Bladder Carcinoma is not a “Simple” Pathology

Far from being a simple model of tumor, recent studies on BC revealed that several genes and metabolic pathways [147] are involved in its formation and development, and that these alterations are critical for the final outcome. Alterations in gene function may be found at any level – chromosome loss and/or aberrations, point mutations in coding sequences, pre- or post- transcriptional up- and down-regulations, environmental exposure of healthy tissues. As it happens for many tumors, some genes (RAS, TP53, FGFR3) are statistically more frequently altered than others, but in any case it is extremely rare to find any BC sample showing just one genetic lesion. In this perspective, BC – like all malignancies – is a multi-step disease and, as such, tens of cellular malfunctions should be present to induce neoplastic transformation and subsequent malignancy survival. Thus, the original view of BC as a model tumor whose diagnosis and prognosis may be referred to just two main metabolic pathways – controlled by TP53 and FGFR3, respectively – has radically changed in the last few years.

What Needs to be Done for Bladder Carcinoma?

For many features (for example, the involvement of some key genes such as TP53 and FGFR3 that are mutated/altered in many other cancers, or the influence of carcinogenic compounds in its formation), BC is not really different from other tumors. This means that prevention should be a priority in its management. As described in the Introduction, several non-genetic factors affect its epidemiology. Most of them (smoke, professional exposure, diet) might be controlled with a minimal economic effort and, hopefully, reduce its incidence as a consequence. This would greatly help saving lives, improving the life quality of affected patients and, last but not least, also free some additional economical resources that may be used for other medical purposes. Of course, prevention is not the only way to handle this pathology. Genetic predisposition is still cause of a high number of new cases. In this perspective, the greater the knowledge of the biological basis of BC, the higher is the possibility to tailor a therapy according to patients’ characteristics. For example, knowing that the patient with a non-invasive disease has pro-angiogenetic mutations might help in selecting a specific treatment and monitoring this aspect of the disease. But only a profound understanding of the molecular mechanisms underlying this pathology will likely allow obtaining a better and patient-oriented diagnosis and a more efficient handling of BC. The number and quality of molecular studies, and especially the genome-wide oriented ones, are the right way to proceed.

ACKNOWLEDGEMENT

Declared none.

CONFLICT OF INTEREST

The author(s) confirm that this article content has no conflicts of interest.

REFERENCES

  • 1.Babjuk M, Oosterlinck W, Sylvester R, Kaasinen E, Böhle A, Palou-Redorta J. European Association of Urology (EAU). EAU guidelines on non-muscle-invasive urothelial carcinoma of the bladder. Eur. Urol. 2008;54:303–314. doi: 10.1016/j.eururo.2008.04.051. [DOI] [PubMed] [Google Scholar]
  • 2.Zhang SW, Ma JH, Li M, Na Y-Q, Chen W-Q. Incidence trends of bladder cancer in cities and counties in China. Chin. J. Urol. 2009;30:673–676. [Google Scholar]
  • 3.Hisataki T, Miyao N, Masumori N, Takahashi A, Sasai M, Yanase M, Itoh N, Tsukamoto T. Risk factors for the development of bladder cancer after upper tract urothelial cancer. Urology. 2000;55:663–667. doi: 10.1016/s0090-4295(99)00563-4. [DOI] [PubMed] [Google Scholar]
  • 4.Pelucchi C, Bosetti C, Negri E, Malvezzi M, La Vecchia C. Mechanisms of disease: the epidemiology of bladder cancer. Nat. Clin. Pract. Urol. 2006;3:327–340. doi: 10.1038/ncpuro0510. [DOI] [PubMed] [Google Scholar]
  • 5.Murta-Nascimento C, Schmitz-Drager BJ, Zeegers MP, Steineck G, Kogevinas M, Real FX, Malats N. Epidemiology of urinary bladder cancer: from tumor development to patient’s death. World J. Urol. 2007;25:285–295. doi: 10.1007/s00345-007-0168-5. [DOI] [PubMed] [Google Scholar]
  • 6.Freedman ND, Silverman DT, Hollenbeck AR, Schatzkin A, Abnet CC. Association between smoking and risk of bladder cancer among men and women. JAMA. 2011;306:737–745. doi: 10.1001/jama.2011.1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yang M. A current global view of environmental and occupational cancers. J. Environ. Sci. Health C Environ. Carcinog. Ecotoxicol. Rev. 2011;29:223–249. doi: 10.1080/10590501.2011.601848. [DOI] [PubMed] [Google Scholar]
  • 8.Dietrich HG, Golka K. Bladder tumors and aromatic amines – Historical milestones from Ludwig Rehn to Wilhelm Hueper. Front. Biosci. (Elite Ed) 2012;4:279–288. doi: 10.2741/375. [DOI] [PubMed] [Google Scholar]
  • 9.Yang CY, Chiu HF, Chang CC, Ho SC, Wu TN. Bladder cancer mortality reduction after installation of a tap-water supply system in an arseniousendemic area in southwestern Taiwan. Environ. Res. 2005;98:127–132. doi: 10.1016/j.envres.2004.07.013. [DOI] [PubMed] [Google Scholar]
  • 10.Tsuda T, Babazono A, Yamamoto E, Kurumatani N, Mino Y, Ogawa T, Kishi Y, Aoyama K. Ingested arsenic and internal cancer: a historical cohort study followed for 33 years. Am. J. Epidemiol. 1995;141:198–209. doi: 10.1093/oxfordjournals.aje.a117421. [DOI] [PubMed] [Google Scholar]
  • 11.Hopenhayn-Rich C, Biggs ML, Fuchs A. Bladder cancer mortality associated with arsenic in drinking water in Argentina. Epidemiology. 1996;7:117–124. doi: 10.1097/00001648-199603000-00003. [DOI] [PubMed] [Google Scholar]
  • 12.Fernández MI, López JF, Vivaldi B, Coz F. Long-term impact of arsenic in drinking water on bladder cancer health care and mortality rates 20 years after end of exposure. J. Urol. 2012;187:856–861. doi: 10.1016/j.juro.2011.10.157. [DOI] [PubMed] [Google Scholar]
  • 13.Lesseura C, Gilbert-Diamonda D, Andrewa AS, Ekstroma RM, Li Z, Kelseyd KT, Marsita CJ, Karagasa MR. A case-control study of polymorphisms in xenobiotic and arsenic metabolism genes and arsenic-related bladder cancer in New Hampshire. Toxicol. Lett. 2012;210:100–106. doi: 10.1016/j.toxlet.2012.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Mazdak H, Yazdekhasti F, Movahedian A, Mirkheshti N, Shafieian M. The comparative study of serum iron, copper, and zinc levels between bladder cancer patients and a control group. Int. Urol. Nephrol. 2010;42:89–93. doi: 10.1007/s11255-009-9583-4. [DOI] [PubMed] [Google Scholar]
  • 15.Reszka E. Selenoproteins in bladder cancer. Clin. Chim. Acta. 2012;413:847–854. doi: 10.1016/j.cca.2012.01.041. [DOI] [PubMed] [Google Scholar]
  • 16.Mazdak H, Zia H. Vitamin E reduces superficial bladder cancer recurrence: a randomized controlled trial. Int. J. Prev. Med. 2012;3:110–115. [PMC free article] [PubMed] [Google Scholar]
  • 17.Pelucchi C, Galeone C, Tramacere I, Bagnardi V, Negri E, Islami F, Scotti L, Bellocco R, Corrao G, Boffetta P, La Vecchia C. Alcohol drinking and bladder cancer risk: a meta-analysis. Ann. Oncol. 2011. [DOI] [PubMed]
  • 18.World Cancer Research Fund (WCRF)/American Institute for Cancer Research (AICR) Food, Nutrition, Physical Activity and the Prevention of Cancer: A Global Perspective. Washington, DC: AICR; 2007. Cancers; pp. 244–321. [Google Scholar]
  • 19.Boffetta P, Hashibe M. Alcohol and cancer. Lancet Oncol. 2006;7:149–156. doi: 10.1016/S1470-2045(06)70577-0. [DOI] [PubMed] [Google Scholar]
  • 20.Secretan B, Straif K, Baan R, Grosse Y, El Ghissassi F, Bouvard V, Benbrahim-Tallaa L, Guha N, Freeman C, Galichet L, Cogliano V. WHO International Agency for Research on Cancer Monograph Working Group. A review of human carcinogens— Part E: tobacco, areca nut, alcohol, coal smoke, and salted fish. Lancet Oncol. 2009;10:1033–1034. doi: 10.1016/s1470-2045(09)70326-2. [DOI] [PubMed] [Google Scholar]
  • 21.International Agency for Research on Cancer (IARC) Alcohol Consumption and Ethyl Carbamate. Lyon, France: IARC; 2010. [Google Scholar]
  • 22.Tramacere I, Scotti L, Jenab M, Bagnardi V, Bellocco R, Rota M, Corrao G, Bravi F, Boffetta P, La Vecchia C. Alcohol drinking and pancreatic cancer risk: a meta-analysis of the dose-risk relation. Int. J. Cancer. 2010;126L:1474–1486. doi: 10.1002/ijc.24936. [DOI] [PubMed] [Google Scholar]
  • 23.Tsukamoto S, Kanegae T, Uchigasaki S, Kitazawa M, Fujioka T, Fujioka S, Imamura Y, Nagoya T, Shimamura M, Mieda Y. Changes in free and bound alcohol metabolites in the urine during ethanol oxidation. Arukoru Kenkyuto Yakubutsu Ison. 1993;28:441–452. [PubMed] [Google Scholar]
  • 24.Boffetta P, Hashibe M, La Vecchia C, Zatonski W, Rehm J. The burden of cancer attributable to alcohol drinking. Int. J. Cancer. 2006;119:884–887. doi: 10.1002/ijc.21903. [DOI] [PubMed] [Google Scholar]
  • 25.Mao Q, Lin Y, Zheng X, Qin J, Yang K, Xie L. A meta-analysis of alcohol intake and risk of bladder cancer. Cancer Causes Control. 2010;21:1843–1850. doi: 10.1007/s10552-010-9611-9. [DOI] [PubMed] [Google Scholar]
  • 26.Mao Q-Q, Dai Y, Lin Y-W, Qin J, Xie L-P, Zheng X-Y. Milk consumption and bladder cancer risk: a meta-analysis of published epidemiological studies. Nutr. Cancer. 2011;63:1263–1271. doi: 10.1080/01635581.2011.614716. [DOI] [PubMed] [Google Scholar]
  • 27.Larsson SC, Orsini N, Wolk A. Milk, milk products and lactose intake and ovarian cancer risk: a meta-analysis of epidemiological studies. Int. J. Cancer. 2006;118:431–441. doi: 10.1002/ijc.21305. [DOI] [PubMed] [Google Scholar]
  • 28.Qin LQ, Xu JY, Wang PY, Tong J, Hoshi K. Milk consumption is a risk factor for prostate cancer in Western countries: evidence from cohort studies. Asia Pac. J. Clin. Nutr. 2007;16:467–476. [PubMed] [Google Scholar]
  • 29.Li F, An SL, Zhou Y, Liang ZK, Jiao ZJ, Jing YM, Wan P, Shi XJ, Tan WL. Milk and dairy consumption and risk of bladder cancer: a meta-analysis. Urology. 2011;78:1298–1305. doi: 10.1016/j.urology.2011.09.002. [DOI] [PubMed] [Google Scholar]
  • 30.Lampe JW. Dairy products and cancer. J. Am. Coll. Nutr. 2011;30(5 ) Suppl 1:464S–470S. doi: 10.1080/07315724.2011.10719991. [DOI] [PubMed] [Google Scholar]
  • 31.Mostafa MH, Sheweita SA, O'Connor PJ. Relationship between Schistosomiasis and bladder cancer. Clin. Microbiol. Rev. 1999;12:97–111. doi: 10.1128/cmr.12.1.97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Shen Y, Liu Y, Liu S, Zhang A. The association between -330T/G polymorphism of interleukin 2 gene and bladder cancer. DNA Cell Biol. 2012. [DOI] [PubMed]
  • 33.Chu H, Ma L, Wang M, Shi D, Qin C, Yuan L, Yin C, Zhang Z. The polymorphisms of IL-4, IL-4R and IL-13 genes and bladder cancer risk in a Chinese population: a case-control study. Mol. Biol. Rep. 2012;39:5349–5357. doi: 10.1007/s11033-011-1334-9. [DOI] [PubMed] [Google Scholar]
  • 34.Mathé E, Nguyen GH, Funamizu N, He P, Moake M, Croce CM, Hussain SP. Inflammation regulates microRNA expression in cooperation with p53 and nitric oxide. Int. J. Cancer. 2011. [DOI] [PMC free article] [PubMed]
  • 35.Nieder AM, Porter MP, Soloway MS. Radiation therapy for prostate cancer increases subsequent risk of bladder and rectal cancer: a population based cohort study. J. Urol. 2008;180:2005–2009. doi: 10.1016/j.juro.2008.07.038. [DOI] [PubMed] [Google Scholar]
  • 36.Nielsen ME, Shariat SF, Karakiewicz PI, Lotan Y, Rogers CG, Amiel GE, Bastian PJ, Vazina A, Gupta A, Lerner SP, Sagalowsky AI, Schoenberg MP, Palapattu GS. Cancer Research Consortium (BCRC). Advanced age is associated with poorer bladder cancer-specific survival in patients treated with radical cystectomy. Eur. Urol. 2007;51:699–706. doi: 10.1016/j.eururo.2006.11.004. [DOI] [PubMed] [Google Scholar]
  • 37.Kaldor JM, Day NE, Kittelmann B, Pettersson F, Langmark F, Pedersen D, Prior P, Neal F, Karjalainen S, Bell J, Choi W, Koch M, Band P, Pompe-Kirn V, Garton C, Staneczek W, Zarén B, Stovall M, Boffetta P. Bladder tumors following chemotherapy and radiotherapy for ovarian cancer: a case control study. Int. J. Cancer. 1995;63:1–6. doi: 10.1002/ijc.2910630102. [DOI] [PubMed] [Google Scholar]
  • 38.Travis LB, Curtis RE, Glimelius B, Holowaty EJ, Van Leeuwen FE, Lynch CF, Hagenbeek A, Stovall M, Banks PM, Adami J, Gospodarowicz MK, Wacholder S, Inskip PD, Tucker MA, Boice JD ., Jr Bladder and kidney cancer following cyclophosphamide therapy for non-Hodgkin’s lymphoma. J. Natl. Cancer Inst. 1995;87:524–531. doi: 10.1093/jnci/87.7.524. [DOI] [PubMed] [Google Scholar]
  • 39.Fajkovic H, Halpern JA, Cha EK, Bahadori A, Chromecki TF, Karakiewicz PI, Breinl E, Merseburger AS, Shariat SF. Impact of gender on bladder cancer incidence, staging, and prognosis. World J. Urol. 2011;29:457–463. doi: 10.1007/s00345-011-0709-9. [DOI] [PubMed] [Google Scholar]
  • 40.Tilki D, Svatek RS, Karakiewicz PI, Isbarn H, Reich O, Kassouf W, Fradet Y, Novara G, Fritsche HM, Bastian PJ, Izawa JI, Stief CG, Ficarra V, Lerner SP, Schoenberg M, Dinney CP, Skinner E, Lotan Y, Sagalowsky AI, Shariat SF. Characteristics and outcomes of patients with pT4 urothelial carcinoma at radical cystectomy: a retrospective international study of 583 patients. J. Urol. 2010;183:87–93. doi: 10.1016/j.juro.2009.08.145. [DOI] [PubMed] [Google Scholar]
  • 41.Liberman D, Alasker A, Sun M, Ismail S, Lughezzani G, Jeldres C, Budaus L, Thuret R, Shariat SF, Widmer H, Perrotte P, Graefen M, Montorsi F, Karakiewicz PI. Radical cystectomy for patients with pT4 urothelial carcinoma in a large population-based study. BJU Int. 2011;107:905–911. doi: 10.1111/j.1464-410X.2010.09590.x. [DOI] [PubMed] [Google Scholar]
  • 42.May M, Bastian PJ, Brookman-May S, Fritsche HM, Tilki D, Otto W, Bolenz C, Gilfrich C, Trojan L, Herrmann E, Moritz R, Tiemann A, Müller SC, Ellinger J, Buchner A, Stief CG, Wieland WF, Höfner T, Hohenfellner M, Haferkamp A, Roigas J, Zacharias M, Nuhn P, Burger M. Gender-specific differences in cancer-specific survival after radical cystectomy for patients with urothelial carcinoma of the urinary bladder in pathologic tumor stage T4a. Urol. Oncol. 2011. [DOI] [PubMed]
  • 43.Micheli A, Mariotto A, Giorgi Rossi A, Gatta G, Muti P. The prognostic role of gender in survival of adult cancer patients. Eur. J. Cancer. 1998;34:2271–2278. doi: 10.1016/s0959-8049(98)00324-4. [DOI] [PubMed] [Google Scholar]
  • 44.Shariat SF, Sfakianos JP, Droller MJ, Karakiewicz PI, Meryn S, Bochner BH. The effect of age and gender on bladder cancer: a critical review of the literature. BJU Int. 2010;105:300–308. doi: 10.1111/j.1464-410X.2009.09076.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Scosyrev E, Trivedi D, Messing E. Female bladder cancer: incidence, treatment, and outcome. Curr. Opin. Urol. 2010;20:404–408. doi: 10.1097/MOU.0b013e32833c7a9b. [DOI] [PubMed] [Google Scholar]
  • 46.Lee CT, Dunn RL, Williams C, Underwood 3rd W. Racial disparity in bladder cancer: trends in tumor presentation at diagnosis. J. Urol. 2006;176:927–934. doi: 10.1016/j.juro.2006.04.074. [DOI] [PubMed] [Google Scholar]
  • 47.Yee DS, Ishill NM, Lowrance WT, Herr HW, Elkin EB. Ethnic differences in bladder cancer survival. Urology. 2011;78:544–549. doi: 10.1016/j.urology.2011.02.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Underwood 3rd W, Dunn RL, Williams C, Lee CT. Gender and geographic influence on the racial disparity in bladder cancer mortality in the US. J. Am. Coll. Surg. 2006;202:284–290. doi: 10.1016/j.jamcollsurg.2005.09.009. [DOI] [PubMed] [Google Scholar]
  • 49.Gore JL, Kwan L, Saigal CS, Litwin MS. Marriage and mortality in bladder carcinoma. Cancer. 2005;104:1188–1194. doi: 10.1002/cncr.21295. [DOI] [PubMed] [Google Scholar]
  • 50.Babjuk M, Oosterlinck W, Sylvester R, Kaasinen E, Böhle A, Palou-Redorta J, Rouprêt M. EAU guidelines on non-muscle-invasive urothelial carcinoma of the bladder, the 2011 update. Eur. Urol. 2012;59:997–1008. doi: 10.1016/j.eururo.2011.03.017. [DOI] [PubMed] [Google Scholar]
  • 51.Stenzl A, Cowan NC, De Santis M, Kuczyk MA, Merseburger AS, Ribal MJ, Sherif A, Witjes JA. Treatment of muscle-invasive and metastatic bladder cancer: update of the EAU guidelines. Eur. Urol. 2012;59:1009–1018. doi: 10.1016/j.eururo.2011.03.023. [DOI] [PubMed] [Google Scholar]
  • 52.Sylvester RJ, van der Meijden AP, Oosterlinck W, Witjes JA, Bouffioux C, Denis L, Newling DW, Kurth K. Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: a combined analysis of 2596 patients from seven EORTC trials. Eur. Urol. 2006;49:466–475. doi: 10.1016/j.eururo.2005.12.031. [DOI] [PubMed] [Google Scholar]
  • 53.Kaufman DS, Shipley WU, Feldman AS. Bladder cancer. Lancet. 2009;374:239–249. doi: 10.1016/S0140-6736(09)60491-8. [DOI] [PubMed] [Google Scholar]
  • 54.Babaian RJ, Johnson DE, Llamas L, Ayala AG. Metastases from transitional cell carcinoma of urinary bladder. Urology. 1980;16:142–144. doi: 10.1016/0090-4295(80)90067-9. [DOI] [PubMed] [Google Scholar]
  • 55.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;159:91–94. doi: 10.1016/s0022-5347(01)64021-7. [DOI] [PubMed] [Google Scholar]
  • 56.Bane BL, Rao JY. Pathology and staging of bladder cancer. Semin. Oncol. 1996;23:549–570. [PubMed] [Google Scholar]
  • 57.Fernandez-Gomez J, Solsona E, Unda M, Martinez-Piñeiro L, Gonzalez M, Hernandez R, Madero R, Ojea A, Pertusa C, Rodriguez-Molina J, Camacho JE, Isorna S, Rabadan M, Astobieta A, Montesinos M, Muntañola P, Gimeno A, Blas M, Martinez-Piñeiro JA. Club Urológico Español de Tratamiento Oncológico (CUETO). Prognostic factors in patients with non-muscle-invasive bladder cancer treated with bacillus Calmette-Guerin: multivariate analysis of data from four randomized CUETO trials. Eur. Urol. 2008;53:992–1001. doi: 10.1016/j.eururo.2007.10.006. [DOI] [PubMed] [Google Scholar]
  • 58.van Rhijn BW, Burger M, Lotan Y, Solsona E, Stief CG, Sylvester RJ, Witjes JA, Zlotta AR. Recurrence and progression of disease in non-muscle-invasive bladder cancer: from epidemiology to treatment strategy. Eur. Urol. 2009;56:430–442. doi: 10.1016/j.eururo.2009.06.028. [DOI] [PubMed] [Google Scholar]
  • 59.Youssef RF, Lotan Y. Predictors of outcome of non-muscle-invasive and muscle-invasive bladder cancer. TheScientificWorld-JOURNAL. 2011;11:369–381. doi: 10.1100/tsw.2011.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Stein JP, Skinner DG. Radical cystectomy for invasive bladder cancer: long-term results of a standard procedure. World J. Urol. 2006;24:296–304. doi: 10.1007/s00345-006-0061-7. [DOI] [PubMed] [Google Scholar]
  • 61.Sternberg CN, Donat SM, Bellmunt J, Millikan RE, Stadler W, De Mulder P, Sherif A, von der Maase H, Tsukamoto T, Soloway MS. Chemotherapy for bladder cancer: treatment guidelines for neoadjuvant chemotherapy, bladder preservation, adjuvant chemotherapy, and metastatic cancer. Urology. 2007;69(1 Suppl ):62–79. doi: 10.1016/j.urology.2006.10.041. [DOI] [PubMed] [Google Scholar]
  • 62.Calabro F, Sternberg CN. Neoadjuvant and adjuvant chemotherapy in muscle-invasive bladder cancer. Eur. Urol. 2009;55:348–358. doi: 10.1016/j.eururo.2008.10.016. [DOI] [PubMed] [Google Scholar]
  • 63.Clark PE. Neoadjuvant versus adjuvant chemotherapy for muscle-invasive bladder cancer. Expert Rev. Anticancer Ther. 2009;9:821–830. doi: 10.1586/era.09.36. [DOI] [PubMed] [Google Scholar]
  • 64.Miyake H, Hara I, Kamidono S, Eto H. Multifocal transitional cell carcinoma of the bladder and upper urinary tract: molecular screening of clonal origin by characterizing CD44 alternative splicing patterns. J. Urol. 2004;172:1127–1129. doi: 10.1097/01.ju.0000129541.23460.48. [DOI] [PubMed] [Google Scholar]
  • 65.Jones TD, Eble JN, Wang M, MacLennan GT, Delahunt B, Brunelli M, Martignoni G, Lopez-Beltran A, Bonsib SM, Ulbright TM, Zhang S, Nigro K, Cheng L. Molecular genetic evidence for the independent origin of multifocal papillary tumors in patients with papillary renal cell carcinomas. Clin. Cancer Res. 2005;11:7226–7223. doi: 10.1158/1078-0432.CCR-04-2597. [DOI] [PubMed] [Google Scholar]
  • 66.Catto JW, Hartmann A, Stoehr R, Bolderson E, Rehman I, Rosario DJ, Hamdy FC, Meuth M. Multifocal urothelial cancers with the mutator phenotype are of monoclonal origin and require panurothelial treatment for tumor clearance. J. Urol. 2006;175:2323–2330. doi: 10.1016/S0022-5347(06)00256-4. [DOI] [PubMed] [Google Scholar]
  • 67.Catto JW, Yates DR, Rehman I, Azzouzi AR, Patterson J, Sibony M, Cussenot O, Hamdy FC. Behavior of urothelial carcinoma with respect to anatomical location. J. Urol. 2007;177:1715–1720. doi: 10.1016/j.juro.2007.01.030. [DOI] [PubMed] [Google Scholar]
  • 68.Gaisa NT, Graham TA, McDonald SAC, Cañadillas-Lopez S, Poulsom R, Heidenreich A, Jakse G, Tadrous PJ, Knuechel R, Wright NA. The human urothelium consists of multiple clonal units, each maintained by a stem cell. J. Pathol. 2011;225:163–171. doi: 10.1002/path.2945. [DOI] [PubMed] [Google Scholar]
  • 69.Bakkar AA, Wallerand H, Radvanyi F, Lahaye JB, Pissard S, Lecerf L, Kouyoumdjian JC, Abbou CC, Pairon JC, Jaurand MC, Thiery JP, Chopin DK, de Medina SG. FGFR3 and TP53 gene mutations define two distinct pathways in urothelial cell carcinoma of the bladder. Cancer Res. 2003;63:8108–8112. [PubMed] [Google Scholar]
  • 70.van Rhijn BW, van der Kwast TH, Vis AN, Kirkels WJ, Boevé ER, Jöbsis AC, Zwarthoff EC. FGFR3 and P53 characterize alternative genetic pathways in the pathogenesis of urothelial cell carcinoma. Cancer Res. 2004;64:1911–1914. doi: 10.1158/0008-5472.can-03-2421. [DOI] [PubMed] [Google Scholar]
  • 71.Hernandez S, Lopez-Knowles E, Lloreta J, Kogevinas M, Jaramillo R, Amorós A, Tardón A, García-Closas R, Serra C, Carrato A, Malats N, Real FX. FGFR3 and Tp53 mutations in T1G3 transitional bladder carcinomas: independent distribution and lack of association with prognosis. Clin. Cancer Res. 2005;11:5444–5450. doi: 10.1158/1078-0432.CCR-05-0122. [DOI] [PubMed] [Google Scholar]
  • 72.Lamy A, Gobet F, Laurent M, Blanchard F, Varin C, Moulin C, Andreou A, Frebourg T, Pfister C. Molecular profiling of bladder tumors based on the detection of FGFR3 and TP53 mutations. J. Urol. 2006;176:2686–2689. doi: 10.1016/j.juro.2006.07.132. [DOI] [PubMed] [Google Scholar]
  • 73.Malats N, Bustos A, Nascimento CM, Fernandez F, Rivas M, Puente D, Kogevinas M, Real FX. 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]
  • 74.Vogelstein B, Lane D, Levine AJ. Surfing the p53 network. Nature. 2000;408:307–310. doi: 10.1038/35042675. [DOI] [PubMed] [Google Scholar]
  • 75.Choi W, Shah JB, Tran M, Svatek R, Marquis L, Lee I-L, Yu D, Adam L, Wen S, Shen Y, Dinney C, McConkey DJ, Siefker-Radtke A. p63 expression defines a lethal subset of muscle-invasive bladder cancers. PLoS One. 2012;7(1 ) doi: 10.1371/journal.pone.0030206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Shariat SF, Zlotta AR, Ashfaq R, Sagalowsky AI, Lotan Y. Cooperative effect of cell-cycle regulators expression on bladder cancer development and biologic aggressiveness. Mod. Pathol. 2007;20:445–459. doi: 10.1038/modpathol.3800757. [DOI] [PubMed] [Google Scholar]
  • 77.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. [DOI] [PubMed] [Google Scholar]
  • 78.Shariat SF, Lotan Y, Karakiewicz PI, Ashfaq R, Isbarn H, Fradet Y, Bastian PJ, Nielsen ME, Capitanio U, Jeldres C, Montorsi F, Müller SC, Karam JA, Heukamp LC, Netto G, Lerner SP, Sagalowsky AI, Cote RJ. p53 predictive value for pT1-2 N0 disease at radical cystectomy. J. Urol. 2009;182:907–913. doi: 10.1016/j.juro.2009.05.024. [DOI] [PubMed] [Google Scholar]
  • 79.Shariat SF, Karam JA, Lerner SP. Molecular markers in bladder cancer. Curr. Opin. Urol. 2008;18:1–8. doi: 10.1097/MOU.0b013e3282f1c5c1. [DOI] [PubMed] [Google Scholar]
  • 80.Bolenz C, Lotan Y. Molecular biomarkers for urothelial carcinoma of the bladder: challenges in clinical use. Nat. Clin. Pract. Urol. 2008;5:676–685. doi: 10.1038/ncpuro1259. [DOI] [PubMed] [Google Scholar]
  • 81.Yang K, Zheng X-Y, Qin J, Wang Y-B, Bai Y, Mao Q-Q, Wan Q, Wu Z-M, Xie L-P. Up-regulation of p21WAF1/Cip1 by saRNA induces G1-phase arrest and apoptosis in T24 human bladder cancer cells. Cancer Lett. 2008;265:206–214. doi: 10.1016/j.canlet.2008.02.014. [DOI] [PubMed] [Google Scholar]
  • 82.Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 1999;13:1501–1512. doi: 10.1101/gad.13.12.1501. [DOI] [PubMed] [Google Scholar]
  • 83.Shariat SF, Karakiewicz PI, Ashfaq R, Lerner SP, Palapattu GS, Cote RJ, Sagalowsky AI, Lotan Y. Multiple biomarkers improve prediction of bladder cancer recurrence and mortality in patients undergoing cystectomy. Cancer. 2008;112:315–325. doi: 10.1002/cncr.23162. [DOI] [PubMed] [Google Scholar]
  • 84.Shariat SF, Chade DC, Karakiewicz PI, Ashfaq R, Isbarn H, Fradet Y, Bastian PJ, Nielsen ME, Capitanio U, Jeldres C, Montorsi F, Lerner SP, Sagalowsky AI, Cote RJ, Lotan Y. Combination of multiple molecular markers can improve prognostication in patients with locally advanced and lymph node positive bladder cancer. J. Urol. 2010;183:68–75. doi: 10.1016/j.juro.2009.08.115. [DOI] [PubMed] [Google Scholar]
  • 85.Shariat SF, Bolenz C, Karakiewicz PI, Fradet Y, Ashfaq R, Bastian PJ, Nielsen ME, Capitanio U, Jeldres C, Rigaud J, Müller SC, Lerner SP, Montorsi F, Sagalowsky AI, Cote RJ, Lotan Y. p53 expression in patients with advanced urothelial cancer of the urinary bladder. BJU Int. 2010;105:489–495. doi: 10.1111/j.1464-410X.2009.08742.x. [DOI] [PubMed] [Google Scholar]
  • 86.Yu C, Zhang Z, Liao W, Zhao X, Liu L, Wu Y, Liu Z, Li Y, Zhong Y, Chen K, Li J, Zhou F, Song L. The tumor-suppressor gene Nkx2.8 suppresses bladder cancer proliferation through upregulation of FOXO3a and inhibition of the MEK/ERK signaling pathway. Carcinogenesis. 2012;33:678–686. doi: 10.1093/carcin/bgr321. [DOI] [PubMed] [Google Scholar]
  • 87.Stein JP, Ginsberg DA, Grossfeld GD, Chatterjee SJ, Esrig D, Dickinson MG, Groshen S, Taylor CR, Jones PA, Skinner DG, Cote RJ. Effect of p21WAF1/CIP1 expression on tumor progression in bladder cancer. J. Natl. Cancer Inst. 1998;90:1072–1079. doi: 10.1093/jnci/90.14.1072. [DOI] [PubMed] [Google Scholar]
  • 88.Shariat SF, Ashfaq R, Sagalowsky AI, Lotan Y. Correlation of cyclin D1 and E1 expression with bladder cancer presence, invasion, progression, and metastasis. Hum. Pathol. 2006;37:1568–1576. doi: 10.1016/j.humpath.2006.05.017. [DOI] [PubMed] [Google Scholar]
  • 89.Yurakh AO, Ramos D, Calabuig-Fariñas S, López-Guerrero JA, Rubio J, Solsona E, Romanenko AM, Vozianov AF, Pellin A, Llombart-Bosch A. Molecular and immunohistochemical analysis of the prognostic value of cell-cycle regulators in urothelial neoplasms of the bladder. Eur. Urol. 2006;50:506–515. doi: 10.1016/j.eururo.2006.03.027. [DOI] [PubMed] [Google Scholar]
  • 90.Zhang Z, Zhang G, Kong C. High expression of polo-like kinase 1 is associated with the metastasis and recurrence in urothelial carcinoma of bladder. Urol. Oncol. 2011. [DOI] [PubMed]
  • 91.McKnight JJ, Gray SB, O’Kane HF, Johnston SR, Williamson KE. Apoptosis and chemotherapy for bladder cancer. J. Urol. 2005;173:683–690. doi: 10.1097/01.ju.0000143194.79287.a9. [DOI] [PubMed] [Google Scholar]
  • 92.Karam JA, Lotan Y, Karakiewicz PI, Ashfaq R, Sagalowsky AI, Roehrborn CG, Shariat SF. Use of combined apoptosis biomarkers for prediction of bladder cancer recurrence and mortality after radical cystectomy. Lancet Oncol. 2007;8:128–136. doi: 10.1016/S1470-2045(07)70002-5. [DOI] [PubMed] [Google Scholar]
  • 93.Margulis V, Lotan Y, Montorsi F, Shariat SF. Predicting survival after radical cystectomy for bladder cancer. BJU Int. 2008;102:15–22. doi: 10.1111/j.1464-410X.2008.07594.x. [DOI] [PubMed] [Google Scholar]
  • 94.Yamana K, Bilim V, Hara N, Kasahara T, Itoi T, Maruyama R, Nishiyama T, Takahashi K, Tomita Y. Prognostic impact of FAS/CD95/APO-1 in urothelial cancers: decreased expression of Fas is associated with disease progression. Br. J. Cancer. 2005;93:544–551. doi: 10.1038/sj.bjc.6602732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Svatek RS, Herman MP, Lotan Y, Casella R, Hsieh JT, Sagalowsky AI, Shariat SF. 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]
  • 96.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]
  • 97.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]
  • 98.Shariat SF, Karakiewicz PI, Godoy G, Karam JA, Ashfaq R, Fradet Y, Isbarn H, Montorsi F, Jeldres C, Bastian PJ, Nielsen ME, Müller SC, Sagalowsky AI, Lotan Y. Survivin as a prognostic marker for urothelial carcinoma of the bladder: a multicenter external validation study. Clin. Cancer Res. 2009;15:7012–7019. doi: 10.1158/1078-0432.CCR-08-2554. [DOI] [PubMed] [Google Scholar]
  • 99.Che X, Yang D, Zong H, Wang J, Li X, Chen F, Chen X, Song X. Nuclear cIAP1 overexpression is a tumor stage- and grade-independent predictor of poor prognosis in human bladder cancer patients. Urol. Oncol. 2011. [DOI] [PubMed]
  • 100.Ewald F, Ueffing N, Brockmann L, Hader C, Telieps T, Schuster M, Schulz WA, Schmitz I. The role of c-FLIP splice variants in urothelial tumors. Cell Death Dis. 2011. [DOI] [PMC free article] [PubMed]
  • 101.Li Y, Jin X, Li J, Jin X, Yu J, Sun X, Chu Y, Xu C, Li X, Wang X, Kakehi Y, Wu X. Expression of TRAIL, DR4, and DR5 in bladder cancer: correlation with response to adjuvant therapy and implications of prognosis. Urology. 2012. [DOI] [PubMed]
  • 102.Shariat SF, Youssef RF, Gupta A, Chade DC, Karakiewicz PI, Isbarn H, Jeldres C, Sagalowsky AI, Ashfaq R, Lotan Y. Association of angiogenesis related markers with bladder cancer outcomes and other molecular markers. J. Urol. 2010;183:1744–1750. doi: 10.1016/j.juro.2010.01.018. [DOI] [PubMed] [Google Scholar]
  • 103.Miyata Y, Kanda S, Ohba K, Nomata K, Hayashida Y, Eguchi J, Hayashi T, Kanetake H. Lymphangiogenesis and angiogenesis in bladder cancer: prognostic implications and regulation by vascular endothelial growth factors-A, -C, and -D. Clin. Cancer Res. 2006;12:800–806. doi: 10.1158/1078-0432.CCR-05-1284. [DOI] [PubMed] [Google Scholar]
  • 104.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]
  • 105.Chodak GW, Hospelhorn V, Judge SM, Mayforth R, Koeppen H, Sasse J. Increased levels of fibroblast growth factor-like activity in urine from patients with bladder or kidney cancer. Cancer Res. 1988;48:2083–2088. [PubMed] [Google Scholar]
  • 106.Wu XR, Kong XP, Pellicer A, Kreibich G, Sun TT. Uroplakins in urothelial biology, function, and disease. Kidney Int. 2009;75:1153–1165. doi: 10.1038/ki.2009.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Huang HY, Shariat SF, Sun TT, Lepor H, Shapiro E, Hsieh JT, Ashfaq R, Lotan Y, Wu XR. Persistent uroplakin expression in advanced urothelial carcinomas: implications in urothelial tumor progression and clinical outcome. Hum. Pathol. 2007;38:1703–1713. doi: 10.1016/j.humpath.2007.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Krüger S, Weitsch G, Büttner H, Matthiensen A, Böhmer T, Marquardt T, Sayk F, Feller AC, Böhle A. HER2 overexpression in muscle-invasive urothelial carcinoma of the bladder: prognostic implications. Int. J. Cancer. 2002;102:514–518. doi: 10.1002/ijc.10731. [DOI] [PubMed] [Google Scholar]
  • 109.Kolla SB, Seth A, Singh MK, Gupta NP, Hemal AK, Dogra PN, Kumar R. Prognostic significance of Her2/neu overexpression in patients with muscle invasive urinary bladder cancer treated with radical cystectomy. Int. Urol. Nephrol. 2008;40:321–327. doi: 10.1007/s11255-007-9283-x. [DOI] [PubMed] [Google Scholar]
  • 110.Kassouf W, Black PC, Tuziak T, Bondaruk J, Lee S, Brown GA, Adam L, Wei C, Baggerly K, Bar-Eli M, McConkey D, Czerniak B, Dinney CP. Distinctive expression pattern of ErbB family receptors signifies an aggressive variant of bladder cancer. J. Urol. 2008;179:353–358. doi: 10.1016/j.juro.2007.08.087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Liedberg F, Anderson H, Chebil G, Gudjonsson S, Höglund M, Lindgren D, Lundberg LM, Lövgren K, Fernö M, Månsson W. Tissue microarray based analysis of prognostic markers in invasive bladder cancer: much effort to no avail? Urol. Oncol. 2008;26:17–24. doi: 10.1016/j.urolonc.2006.08.021. [DOI] [PubMed] [Google Scholar]
  • 112.Blehm KN, Spiess PE, Bondaruk JE, Dujka ME, Villares GJ, Zhao YJ, Bogler O, Aldape KD, Grossman HB, Adam L, McConkey DJ, Czerniak BA, Dinney CP, Bar-Eli M. Mutations within the kinase domain and truncations of the epidermal growth factor receptor are rare events in bladder cancer: implications for therapy. Clin. Cancer Res. 2006;12:4671–4677. doi: 10.1158/1078-0432.CCR-06-0407. [DOI] [PubMed] [Google Scholar]
  • 113.Villares GJ, Zigler M, Blehm K, Bogdan C, McConkey D, Colin D, Bar-Eli M. Targeting EGFR in bladder cancer. World J. Urol. 2007;25:573–579. doi: 10.1007/s00345-007-0202-7. [DOI] [PubMed] [Google Scholar]
  • 114.Zhang ZT, Pak J, Huang HY, Shapiro E, Sun TT, Pellicer A, Wu XR. Role of Ha-ras activation in superficial papillary pathway of urothelial tumor formation. Oncogene. 2001;20:1973–1980. doi: 10.1038/sj.onc.1204315. [DOI] [PubMed] [Google Scholar]
  • 115.Shan GY, Zhang Z, Chen QG, Yu XY, Liu GB, Kong CZ. Overexpression of RIN1 associates with tumor grade and progression in patients of bladder urothelial carcinoma. Tumor Biol. 2012. [DOI] [PubMed]
  • 116.Wu SY, Lan SH, Cheng DE, Chen WK, Shen CH, Lee YR, Zuchini R, Liu HS. Ras-related tumorigenesis is suppressed by BNIP3-mediated autophagy through inhibition of cell proliferation. Neoplasia. 2011;13:1171–1182. doi: 10.1593/neo.11888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Jebar AH, Hurst CD, Tomlinson DC, Johnston C, Taylor CF, Knowles MA. FGFR3 and Ras gene mutations are mutually exclusive genetic events in urothelial cell carcinoma. Oncogene. 2005;24:5218–5225. doi: 10.1038/sj.onc.1208705. [DOI] [PubMed] [Google Scholar]
  • 118.Aveyard JS, Skilleter A, Habuchi T, Knowles MA. Somatic mutation of PTEN in bladder carcinoma. Br. J. Cancer. 1999;80:904–908. doi: 10.1038/sj.bjc.6690439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Lopez-Knowles E, Hernandez S, Malats N, Kogevinas M, Lloreta J, Carrato A, Tardón A, Serra C, Real FX. PIK3CA mutations are an early genetic alteration associated with FGFR3 mutations in superficial papillary bladder tumors. Cancer Res. 2006;66:7401–7404. doi: 10.1158/0008-5472.CAN-06-1182. [DOI] [PubMed] [Google Scholar]
  • 120.Engelman JA, Luo J, Cantley LC. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat. Rev. Genet. 2006;7:606–619. doi: 10.1038/nrg1879. [DOI] [PubMed] [Google Scholar]
  • 121.Dyrskjot L, Zieger K, Real FX, Malats N, Carrato A, Hurst C, Kotwal S, Knowles M, Malmstrom PU, de la Torre M, Wester K, Allory Y, Vordos D, Caillault A, Radvanyi F, Hein AM, Jensen JL, Jensen KM, Marcussen N, Orntoft TF. Gene expression signatures predict outcome in non-muscle-invasive bladder carcinoma: a multicenter validation study. Clin. Cancer Res. 2007;13:3545–3551. doi: 10.1158/1078-0432.CCR-06-2940. [DOI] [PubMed] [Google Scholar]
  • 122.Munksgaard PP, Mansilla F, Brems Eskildsen A-S, Fristrup N, Birkenkamp-Demtröder K, Ulhøi BP, Borre M, Agerbæk M, Hermann GG, Ørntoft TF, Dyrskjøt L. Low ANXA10 expression is associated with disease aggressiveness in bladder cancer. Br. J. Cancer. 2011;105:1379–1387. doi: 10.1038/bjc.2011.404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Wang H, Wang J, Zuo Y, Ding M, Yan R, Yang D, Ke C. Expression and prognostic significance of a new tumor metastasis suppressor gene LASS2 in human bladder carcinoma. Med. Oncol. 2011. [DOI] [PubMed]
  • 124.Zuo Q, Wu W, Li X, Zhao L, Chen W. HDAC6 and SIRT2 promote bladder cancer cell migration and invasion by targeting cortactin. Oncol. Rep. 2012;27:819–824. doi: 10.3892/or.2011.1553. [DOI] [PubMed] [Google Scholar]
  • 125.Zoidakis J, Makridakis M, Zerefos PG, Bitsika V, Esteban S, Frantzi M, Stravodimos K, Anagnou NP, Roubelakis MG, Sanchez-Carbayo M, Vlahou A. Profilin 1 is a potential biomarker for bladder cancer aggressiveness. Mol. Cell. Proteomics. 2011. [DOI] [PMC free article] [PubMed]
  • 126.Yang M-H, Chu P-Y, Chen SC-J, Chung T-W, Chen W-C, Tan L-B, Kan W-C, Wang H-Y, Su S-B, Tyan Y-C. Characterization of ADAM28 as a biomarker of bladder transitional cell carcinomas by urinary proteome analysis. Biochem. Biophys. Res. Commun. 2011;411:714–720. doi: 10.1016/j.bbrc.2011.07.010. [DOI] [PubMed] [Google Scholar]
  • 127.Matsushita K, Cha EK, Matsumoto K, Baba S, Chromecki TF, Fajkovic H, Sun M, Karakiewicz PI, Scherr DS, Shariat SF. Immunohistochemical biomarkers for bladder cancer prognosis. Int. J. Urol. 2011;18:616–629. doi: 10.1111/j.1442-2042.2011.02809.x. [DOI] [PubMed] [Google Scholar]
  • 128.Han B, Cui D, Jing Y, Hong Y, Xia S. Estrogen receptor β (ERβ) is a novel prognostic marker of recurrence survival in non-muscle-invasive bladder cancer potentially by inhibiting cadherin switch. World J. Urol. 2012. [DOI] [PubMed]
  • 129.Miyamoto H, Yao JL, Chaux A, Zheng Y, Hsu I, Izumi K, Chang C, Messing EM, Netto GJ, Yeh S. Expression of androgen and oestrogen receptors and its prognostic significance in urothelial neoplasm of the urinary bladder. BJU Int. 2012. [DOI] [PubMed]
  • 130.Lindén M, Bergström Lind M, Mayrhofer C, Segersten U, Wester K, Lyutvinskiy Y, Zubarev R, Malmström P-U, Pettersson U. Proteomic analysis of urinary biomarker candidates for non-muscle invasive bladder cancer. Proteomics. 2012. [DOI] [PubMed]
  • 131.Pandith AA, Shah ZA, Siddiqi MA. Oncogenic role of fibroblast growth factor receptor 3 in tumorigenesis of urinary bladder cancer. Urol. Oncol. 2010. [DOI] [PubMed]
  • 132.Iyer G, Milowsky MI. Fibroblast growth factor receptor-3 in urothelial tumorigenesis. Urol. Oncol. 2011. [DOI] [PubMed]
  • 133.Foldynova-Trantirkova S, Wilcox WR, Krejci P. Sixteen years and counting: the current understanding of fibroblast growth factor receptor 3 (FGFR3) signaling in skeletal dysplasias. Hum. Mutat. 2012;33:29–41. doi: 10.1002/humu.21636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Gomez-Roman J, Saenz P, Conzalez J, Escuredo K, Santa Cruz S, Junquera C, Simón L, Martínez A, Gutiérrez Baños JL, López-Brea M, Esparza C, Val-Bernal JF. Fibroblast growth factor receptor 3 is overexpressed in urinary tract carcinomas and modulates the neoplastic cell growth. Clin. Cancer Res. 2005;11:459–465. [PubMed] [Google Scholar]
  • 135.Mhawech-Fauceglia P, Fischer G, Alvarez V, Ahmed A, Herrmann F. Predicting outcome in minimally invasive (T1a and T1b) urothelial bladder carcinoma using a panel of biomarkers: a high throughput tissue microarray analysis. BJU Int. 2007;100:1182–1187. doi: 10.1111/j.1464-410X.2007.07090.x. [DOI] [PubMed] [Google Scholar]
  • 136.Knowles MA. Role of FGFR3 in urothelial cell carcinoma: biomarker and potential therapeutic target. World J. Urol. 2007;25:581–593. doi: 10.1007/s00345-007-0213-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Brazil DP, Hemmings BA. Ten years of protein kinase B signalling: a hard Akt to follow. Trends Biochem. Sci. 2001;26:657–664. doi: 10.1016/s0968-0004(01)01958-2. [DOI] [PubMed] [Google Scholar]
  • 138.Qayyum T, Fyffe G, Duncan M, McArdle PA, Hilmy M, Orange C, Halbert G, Seywright M, Horgan PG, Underwood MA, Edwards J. The interrelationships between Src, Cav-1 and RhoGD12 in transitional cell carcinoma of the bladder. Br. J. Cancer. 2012. [DOI] [PMC free article] [PubMed]
  • 139.Su WC, Kitagawa M, Xue N, Xie B, Garofalo S, Cho J, Deng C, Horton WA, Fu XY. Activation of Stat1 by mutant fibroblast growth-factor receptor in thanatophoric dysplasia type II dwarfism. Nature. 1997;386:288–292. doi: 10.1038/386288a0. [DOI] [PubMed] [Google Scholar]
  • 140.Yang GL, Zhang LH, Bo JJ, Huo XJ, Chen HG, Cao M, Liu DM, Huang YR. Increased expression of HMGB1 is associated with poor prognosis in human bladder cancer. J. Surg. Oncol. 2012. [DOI] [PubMed]
  • 141.Izmirli M, Inandiklioglu N, Abat D, Alptekin D, Demirhan O, Tansug Z, Bayazit Y. MTHFR gene polymorphisms in bladder cancer in the Turkish population. Asian Pac. J. Cancer Prev. 2011;12:1833–1835. [PubMed] [Google Scholar]
  • 142.Wang YH, Yeh SD, Shen KH, Shen CH, Tung MC, Liu CT, Chiou HY. Association of hOGG1 and XPD polymorphisms with urothelial carcinoma in Taiwan. Anticancer Res. 2011;31:3939–3944. [PubMed] [Google Scholar]
  • 143.Kawashima A, Takayama H, Tsujimura A. A review of ERCC1 gene in bladder cancer: implications for carcinogenesis and resistance to chemoradiotherapy. Adv. Urol. 2012. 2012;812398 doi: 10.1155/2012/812398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Stern MC, Lin J, Figueroa JD, Kelsey KT, Kiltie AE, Yuan JM, Matullo G, Fletcher T, Benhamou S, Taylor JA, Placidi D, Zhang ZF, Steineck G, Rothman N, Kogevinas M, Silverman D, Malats N, Chanock S, Wu X, Karagas MR, Andrew AS, Nelson HH, Bishop DT, Sak SC, Choudhury A, Barrett JH, Elliot F, Corral R, Joshi AD, Gago-Dominguez M, Cortessis VK, Xiang YB, Gao YT, Vineis P, Sacerdote C, Guarrera S, Polidoro S, Allione A, Gurzau E, Koppova K, Kumar R, Rudnai P, Porru S, Carta A, Campagna M, Arici C, Park SS, Garcia-Closas M. International Consortium of Bladder Cancer. Polymorphisms in DNA repair genes, smoking, and bladder cancer risk: findings from the international consortium of bladder cancer. Cancer Res. 2009;69:6857–6864. doi: 10.1158/0008-5472.CAN-09-1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Zhu X, Zhong Z, Zhang X, Zhao X, Xu R, Ren W, Li S. DNA repair gene XRCC3 T241M polymorphism and bladder cancer risk in a chinese population. Genet. Test Mol. Biomarkers. 2012. [DOI] [PubMed]
  • 146.Teo MT, Landi D, Taylor CF, Elliott F, Vaslin L, Cox DG, Hall J, Landi S, Bishop DT, Kiltie AE. The role of microRNA-binding site polymorphisms in DNA repair genes as risk factors for bladder cancer and breast cancer and their impact on radiotherapy outcomes. Carcinogenesis. 2012;33:581–586. doi: 10.1093/carcin/bgr300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Menashe I, Figueroa JD, Garcia-Closas M, Chatterjee N, Malats N, Picornell A, Maeder D, Yang Q, Prokunina-Olsson L, Wang Z, Real FX, Jacobs KB, Baris D, Thun M, Albanes D, Purdue MP, Kogevinas M, Hutchinson A, Fu Y-P, Tang W, Burdette L, Tardón A, Serra C, Carrato A, García-Closas R, Lloreta J, Johnson A, Schwenn M, Schned A, Andriole G, Jr, Black A, Jacobs EJ, Diver RW, Gapstur SM, Weinstein SJ, Virtamo J, Caporaso NE, Landi MT, Fraumeni JF, Jr, Chanock SJ, Silverman DT, Rothman N. Large-scale pathway-based analysis of bladder cancer genome-wide association data from five studies of european background. PLoS One. 2012;7(1 ):e29396. doi: 10.1371/journal.pone.0029396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Tang W, Fu YP, Figueroa JD, Malats N, Garcia-Closas M, Chatterjee N, Kogevinas M, Baris D, Thun M, Hall JL, De Vivo I, Albanes D, Porter-Gill P, Purdue MP, Burdett L, Liu L, Hutchinson A, Myers T, Tardón A, Serra C, Carrato A, Garcia-Closas R, Lloreta J, Johnson A, Schwenn M, Karagas MR, Schned A, Black A, Jacobs EJ, Diver WR, Gapstur SM, Virtamo J, Hunter DJ, Fraumeni JF, Jr, Chanock SJ, Silverman DT, Rothman N, Prokunina-Olsson L. Mapping of the UGT1A locus identifies an uncommon coding variant that affects mRNA expression and protects from bladder cancer. Hum. Mol. Genet. 2012;21:1918–1930. doi: 10.1093/hmg/ddr619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Safarinejad MR, Safarinejad S, Shafiei N, Safarinejad S. Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility. Urol. Oncol. 2011. [DOI] [PubMed]
  • 150.Gong M, Dong W, An R. Glutathione S-transferase T1 polymorphism contributes to bladder cancer risk: a meta-analysis involving 50 studies. DNA Cell Biol. 2012. [DOI] [PubMed]
  • 151.Salinas-Sánchez AS, Donate-Moreno MJ, López-Garrido MP, Giménez-Bachs JM, Escribano J. Role of CYP1B1 gene polymorphisms in bladder cancer susceptibility. J. Urol. 2012;187:700–706. doi: 10.1016/j.juro.2011.10.063. [DOI] [PubMed] [Google Scholar]
  • 152.Schiffer E, Vlahou A, Petrolekas A, Stravodimos K, Tauber R, Geschwend JE, Neuhaus J, Stolzenburg JU, Conaway MR, Mischak H, Theodorescu D. Prediction of muscle-invasive bladder cancer using urinary proteomics. Clin. Cancer Res. 2009;15:4935–4943. doi: 10.1158/1078-0432.CCR-09-0226. [DOI] [PubMed] [Google Scholar]
  • 153.Chen Y-T, Chena H-W, Domanskic D, Smith DS, Liang K-H, Wu C-C, Chen C-L, Chung T, Chen M-C, Chang Y-S, Parker CE, Borchers CH, Yu J-S. Multiplexed quantification of 63 proteins in human urine by multiple reaction monitoring-based mass spectrometry for discovery of potential bladder cancer biomarkers. J. Proteomics. 2012;DOI doi: 10.1016/j.jprot.2011.12.031. [DOI] [PubMed] [Google Scholar]
  • 154.Holmes E, Wilson I D, Nicholson J K. Metabolic phenotyping in health and disease. Cell. 2008;134:714–717. doi: 10.1016/j.cell.2008.08.026. [DOI] [PubMed] [Google Scholar]
  • 155.Spratlin JL, Serkova NJ, Eckhardt SG. Clinical applications of metabolomics in oncology: a review. Clin. Cancer Res. 2009;15:431–440. doi: 10.1158/1078-0432.CCR-08-1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Huang Z, Lin L, Gao Y, Chen Y, Yan X, Xing J, Hang W. Bladder cancer determination via two urinary metabolites: a biomarker pattern approach. Mol. Cell. Proteomics. 2011. [DOI] [PMC free article] [PubMed]
  • 157.Lindgren D, Liedberg F, Andersson A, Chebil G, Gudjonsson S, Borg A, Månsson W, Fioretos T, Höglund M. Molecular characterization of early-stage bladder carcinomas by expression profiles, FGFR3 mutation status, and loss of 9q. Oncogene. 2006;25:2685–2696. doi: 10.1038/sj.onc.1209249. [DOI] [PubMed] [Google Scholar]
  • 158.Simoneau M, LaRue H, Aboulkassim TO, Meyer F, Moore L, Fradet Y. Chromosome 9 deletions and recurrence of superficial bladder cancer: identification of four regions of prognostic interest. Oncogene. 2000;19:6317–6323. doi: 10.1038/sj.onc.1204022. [DOI] [PubMed] [Google Scholar]
  • 159.Hoffman AM, Cairns P. Epigenetics of kidney cancer and bladder cancer. Epigenomics. 2011;3:19–34. doi: 10.2217/epi.10.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Dudziec E, Goepel JR, Catto JW. Global epigenetic profiling in bladder cancer. Epigenomics. 2011;3:35–45. doi: 10.2217/epi.10.71. [DOI] [PubMed] [Google Scholar]
  • 161.Christoph F, Schmidt B, Schmitz-Drager BJ, Schulz WA. Overexpression and amplification of the c-myc gene in human urothelial carcinoma. Int. J. Cancer. 1999;84:169–173. doi: 10.1002/(sici)1097-0215(19990420)84:2<169::aid-ijc13>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
  • 162.Feber A, Clark J, Goodwin G, Dodson AR, Smith PH, Fletcher A, Edwards S, Flohr P, Falconer A, Roe T, Kovacs G, Dennis N, Fisher C, Wooster R, Huddart R, Foster CS, Cooper CS. Amplification and overexpression of E2F3 in human bladder cancer. Oncogene. 2004;23:1627–1630. doi: 10.1038/sj.onc.1207274. [DOI] [PubMed] [Google Scholar]
  • 163.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]
  • 164.Dinney CP, McConkey DJ, Millikan RE, Wu X, Bar-Eli M, Adam L, Kamat AM, Siefker-Radtke AO, Tuziak T, Sabichi AL, Grossman HB, Benedict WF, Czerniak B. Focus on bladder cancer. Cancer Cell. 2004;6:111–116. doi: 10.1016/j.ccr.2004.08.002. [DOI] [PubMed] [Google Scholar]
  • 165.Hu H, Wang YL, Wang GW, Wong YC, Wang XF, Wang Y, Xu KX. A novel role of Id-1 in regulation of epithelial-to-mesenchymal transition in bladder cancer. Urol. Oncol. 2012. [DOI] [PubMed]
  • 166.Sun XF, Sun ZY, Pan B, Li L, Shen W. Alteration in methylation pattern of oncogene Akt1 promoter region in bladder cancer. Mol. Biol. Rep. 2011;39:5631–5636. doi: 10.1007/s11033-011-1369-y. [DOI] [PubMed] [Google Scholar]
  • 167.Shirodkar SP, Lokeshwar VB. Potential new urinary markers in the early detection of bladder cancer. Curr. Opin. Urol. 2009;19:488–493. doi: 10.1097/MOU.0b013e32832eb3a0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Feng CC, Wang PH, Guan M, Jiang HW, Wen H, Ding Q, Wu Z. Urinary BLCA-4 is highly specific for detection of bladder cancer in Chinese Han population and is related to tumor invasiveness. Folia Biol. (Praha) 2011;57:242–247. [PubMed] [Google Scholar]
  • 169.Gecit I, Aslan M, Gunes M, Pirincci N, Esen R, Demir H, Ceylan K. Serum prolidase activity, oxidative stress, and nitric oxide levels in patients with bladder cancer. J. Cancer Res. Clin. Oncol. 2012. [DOI] [PMC free article] [PubMed]
  • 170.Chatterjee SJ, Datar R, Youssefzadeh D, George B, Goebell PJ, Stein JP, Young L, Shi SR, Gee C, Groshen S, Skinner DG, Cote RJ. Combined effects of p53, p21, and pRb expression in the progression of bladder transitional cell carcinoma. J. Clin. Oncol. 2004;22:1007–1013. doi: 10.1200/JCO.2004.05.174. [DOI] [PubMed] [Google Scholar]
  • 171.Chapman EJ, Harnden P, Chambers P, Johnston C, Knowles MA. Comprehensive analysis of CDKN2A status in microdissected urothelial cell carcinoma reveals potential haploinsufficiency, a high frequency of homozygous co-deletion and associations with clinical phenotype. Clin. Cancer Res. 2005;11:5740–5747. doi: 10.1158/1078-0432.CCR-05-0411. [DOI] [PubMed] [Google Scholar]
  • 172.Marín-Aguilera M, Mengual L, Ribal MJ, Ars E, Ríos J, Gázquez C, Villavicencio H, Alcaraz A. Utility of urothelial mRNA markers in blood for staging and monitoring bladder cancer. Urology. 2012;79:240.e9–240. doi: 10.1016/j.urology.2011.09.006. [DOI] [PubMed] [Google Scholar]
  • 173.Riester M, Taylor J, Feifer A, Koppie TM, Rosenberg J, Downey RJ, Bochner BH, Michor F. Combination of a novel gene expression signature with a clinical nomogram improves the prediction of survival in high-risk bladder cancer. Clin. Cancer Res. 2012;18:1323–1333. doi: 10.1158/1078-0432.CCR-11-2271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Stransky N, Vallot C, Reyal F, Bernard-Pierrot I, de Medina SG, Segraves R, de Rycke Y, Elvin P, Cassidy A, Spraggon C, Graham A, Southgate J, Asselain B, Allory Y, Abbou CC, Albertson DG, Thiery JP, Chopin DK, Pinkel D, Radvanyi F. Regional copy number-independent deregulation of transcription in cancer. Nat. Genet. 2006;38:1386–1396. doi: 10.1038/ng1923. [DOI] [PubMed] [Google Scholar]
  • 175.Vallot C, Stransky N, Bernard-Pierrot I, Hérault A, Zucman-Rossi J, Chapeaublanc E, Vordos D, Laplanche A, Benhamou S, Lebret T, Southgate J, Allory Y, Radvanyi F. A novel epigenetic phenotype associated with the most aggressive pathway of bladder tumor progression. J. Natl. Cancer Inst. 2011;103:47–60. doi: 10.1093/jnci/djq470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Sánchez-Carbayo M. Hypermethylation in bladder cancer: biological pathways and translational applications. Tumor Biol. 2012;33:347–361. doi: 10.1007/s13277-011-0310-2. [DOI] [PubMed] [Google Scholar]
  • 177.Tada Y, Wada M, Taguchi K, Mochida Y, Kinugawa N, Tsuneyoshi M, Naito S, Kuwano M. The association of death-associated protein kinase hypermethylation with early recurrence in superficial bladder cancers. Cancer Res. 2002;62:4048–4053. [PubMed] [Google Scholar]
  • 178.Catto JW, Azzouzi AR, Rehman I, Feeley KM, Cross SS, Amira N, Fromont G, Sibony M, Cussenot O, Meuth M, Hamdy FC. Promoter hypermethylation is associated with tumor location, stage, and subsequent progression in transitional cell carcinoma. J. Clin. Oncol. 2005;23:2903–2910. doi: 10.1200/JCO.2005.03.163. [DOI] [PubMed] [Google Scholar]
  • 179.Hellwinkel OJC, Kedia M, Isbarn H, Budäus L, Friedrich MG. Methylation of the TPEF- and PAX6-promoters is increased in early bladder cancer and in normal mucosa adjacent to pTa tumors. BJU Int. 2007;101:753–757. doi: 10.1111/j.1464-410X.2007.07322.x. [DOI] [PubMed] [Google Scholar]
  • 180.Yates DR, Rehman I, Abbod MF, Meuth M, Cross SS, Linkens DA, Hamdy FC, Catto JW. Promoter hypermethylation identifies progression risk in bladder cancer. Clin. Cancer Res. 2007;13:2046–2053. doi: 10.1158/1078-0432.CCR-06-2476. [DOI] [PubMed] [Google Scholar]
  • 181.Jarmalaite S, Jankevicius F, Kurgonaite K, Suziedelis K, Mutanen P, Husgafvel-Pursiainen K. Promoter hypermethylation in tumor suppressor genes shows association with stage, grade and invasiveness of bladder cancer. Oncology. 2008;75:145–151. doi: 10.1159/000158665. [DOI] [PubMed] [Google Scholar]
  • 182.Eissa S, Zohny SF, Shehata HH, Hegazy MGA, Salem AM, Esmat M. Urinary retinoic acid receptor-β2 gene promoter methylation and hyaluronidase activity as noninvasive tests for diagnosis of bladder cancer. Clin. Biochem. 2012;45:402–407. doi: 10.1016/j.clinbiochem.2012.01.010. [DOI] [PubMed] [Google Scholar]
  • 183.Kandimalla R, van Tilborg AAG, Kompier LC, Stumpel DJPM, Stam RW, Bangma CH, Zwarthoff EC. Genome-wide analysis of CpG island methylation in bladder cancer identified TBX2, TBX3, GATA2, and ZIC4 as pTa-specific prognostic markers. Eur. Urol. 2012. [DOI] [PubMed]
  • 184.Bornman DM, Mathew S, Alsruhe J, Herman JG, Gabrielson E. Methylation of the E-cadherin gene in bladder neoplasia and in normal urothelial epithelium from elderly individuals. Am. J. Pathol. 2001;159:831–835. doi: 10.1016/S0002-9440(10)61758-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Marsit CJ, Houseman EA, Schned AR, Karagas MR, Kelsey KT. Promoter hypermethylation is associated with current smoking, age, gender and survival in bladder cancer. Carcinogenesis. 2007;28:1745–1751. doi: 10.1093/carcin/bgm116. [DOI] [PubMed] [Google Scholar]
  • 186.Yang W, Cui S, Ma J, Lu Q, Kong C, Liu T, Sun Z. Cigarette smoking extract causes hypermethylation and inactivation of WWOX gene in T-24 human bladder cancer cells. Neoplasma. 2012;59:216–223. doi: 10.4149/neo_2012_028. [DOI] [PubMed] [Google Scholar]
  • 187.Lin HH, Ke HL, Huang SP, Wu WJ, Chen YK, Chang LL. Increase sensitivity in detecting superficial, low grade bladder cancer by combination analysis of hypermethylation of E-cadherin, p16, p14, RASSF1A genes in urine. Urol. Oncol. 2010;28:597–602. doi: 10.1016/j.urolonc.2008.12.008. [DOI] [PubMed] [Google Scholar]
  • 188.Jablonowski Z, Reszka E, Gromadzinska J, Wasowicz W, Sosnowski M. Hypermethylation of p16 and DAPK promoter gene regions in patients with non-invasive urinary bladder cancer. Arch. Med. Sci. 2011;7:512–516. doi: 10.5114/aoms.2011.23421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Lin HH, Ke HL, Wu WJ, Lee YH, Chang LL. Hyper-methylation of E-cadherin, p16, p14, and RASSF1A genes in pathologically normal urothelium predict bladder recurrence of bladder cancer after transurethral resection. Urol. Oncol. 2012;30:177–181. doi: 10.1016/j.urolonc.2010.01.002. [DOI] [PubMed] [Google Scholar]
  • 190.Lee MG, Kim HY, Byun DS. Frequent epigenetic inactivation of RASSF1A in human bladder carcinoma. Cancer Res. 2001;61:6688–6692. [PubMed] [Google Scholar]
  • 191.Chen PC, Tsai MH, Yip SK, Jou YC, Ng CF, Chen Y, Wang X, Huang W, Tung CL, Chen GC, Huang MM, Tong JH, Song EJ, Chang DC, Hsu CD, To KF, Shen CH, Chan MW. Distinct DNA methylation epigenotypes in bladder cancer from different Chinese sub-populations and its implication in cancer detection using voided urine. BMC Med. Genomics. 2011;4:45. doi: 10.1186/1755-8794-4-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Tolg C, Sabha N, Cortese R, Panchal T, Ahsan A, Soliman A, Aitken KJ, Petronis A, Bägli DJ. Uropathogenic E. coli infection provokes epigenetic downregulation of CDKN2A (p16INK4A) in uroepithelial cells. Lab. Invest. 2011;91:825–836. doi: 10.1038/labinvest.2010.197. [DOI] [PubMed] [Google Scholar]
  • 193.Kim EJ, Kim YJ, Jeong P, Ha YS, Bae SC, Kim WJ. Methylation of the RUNX3 promoter as a potential prognostic marker for bladder tumor. J. Urol. 2008;180:1141–1145. doi: 10.1016/j.juro.2008.05.002. [DOI] [PubMed] [Google Scholar]
  • 194.Aleman A, Adrien L, Lopez-Serra L, Cordon-Cardo C, Esteller M, Belbin TJ, Sanchez-Carbayo M. Identification of DNA hypermethylation of SOX9 in association with bladder cancer progression using CpG microarrays. Br. J. Cancer. 2008;98:466–473. doi: 10.1038/sj.bjc.6604143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Ha YS, Kim JS, Yoon HY, Jeong P, Kim TH, Yun SJ, Lee SC, Kim GY, Choi YH, Moon SK, Yi Kim I, Kim WJ. Novel combination markers for predicting progression of nonmuscle invasive bladder cancer. Int. J. Cancer. 2011. [DOI] [PubMed]
  • 196.Yan C, Kim YW, Ha YS, Kim IY, Kim YJ, Yun SJ, Moon SK, Bae SC, Kim WJ. RUNX3 methylation as a predictor for disease progression in patients with non-muscle-invasive bladder cancer. J. Surg. Oncol. 2012;105:425–430. doi: 10.1002/jso.22087. [DOI] [PubMed] [Google Scholar]
  • 197.Urakami S, Shiina H, Enokida H, Kawakami T, Tokizane T, Ogishima T, Tanaka Y, Li LC, Ribeiro-Filho LA, Terashima M, Kikuno N, Adachi H, Yoneda T, Kishi H, Shigeno K, Konety BR, Igawa M, Dahiya R. Epigenetic inactivation of Wnt inhibitory factor-1 plays an important role in bladder cancer through aberrant canonical Wnt/ß-Catenin signaling pathway. Clin. Cancer Res. 2006;12:383–391. doi: 10.1158/1078-0432.CCR-05-1344. [DOI] [PubMed] [Google Scholar]
  • 198.Urakami S, Shiina H, Enokida H, Kawakami T, Kawamoto K, Hirata H, Tanaka Y, Kikuno N, Nakagawa M, Igawa M, Dahiya R. Combination analysis of hypermethylated Wnt-antagonist family genes as a novel epigenetic biomarker panel for bladder cancer detection. Clin. Cancer Res. 2006;12:2109–2116. doi: 10.1158/1078-0432.CCR-05-2468. [DOI] [PubMed] [Google Scholar]
  • 199.Stoehr R, Wissmann C, Suzuki H, Knuechel R, Krieg RC, Klopocki E, Dahl E, Wild P, Blaszyk H, Sauter G, Simon R, Schmitt R, Zaak D, Hofstaedter F, Rosenthal A, Baylin SB, Pilarsky C, Hartmann A. Deletions of chromosome 8p and loss of sFRP1 expression are progression markers of papillary bladder cancer. Lab. Invest. 2004;84:465–478. doi: 10.1038/labinvest.3700068. [DOI] [PubMed] [Google Scholar]
  • 200.Marsit CJ, Karagas MR, Andrew A, Liu M, Danaee H, Schned AR, Nelson HH, Kelsey KT. Epigenetic inactivation of SFRP genes and TP53 alteration act jointly as markers of invasive bladder cancer. Cancer Res. 2005;65:7081–7085. doi: 10.1158/0008-5472.CAN-05-0267. [DOI] [PubMed] [Google Scholar]
  • 201.Khin SS, Kitazawa R, Win N, Aye TT, Mori K, Kondo T, Kitazawa S. BAMBI gene is epigenetically silenced in subset of high-grade bladder cancer. Int. J. Cancer. 2009;125:328–338. doi: 10.1002/ijc.24318. [DOI] [PubMed] [Google Scholar]
  • 202.Du P, Ye L, Li H, Ruge F, Yang Y, Jiang WG. Growth differentiation factor-9 expression is inversely correlated with an aggressive behaviour in human bladder cancer cells. Int. J. Mol. Med. 2012;29:428–434. doi: 10.3892/ijmm.2011.858. [DOI] [PubMed] [Google Scholar]
  • 203.Hinz S, Kempkensteffen C, Weikert S, Schostak M, Schrader M, Miller K, Christoph F. EZH2 polycomb transcriptional repressor expression correlates with methylation of the APAF-1 gene in superficial transitional cell carcinoma of the bladder. Tumor Biol. 2007;28:151–157. doi: 10.1159/000103380. [DOI] [PubMed] [Google Scholar]
  • 204.Zuiverloon TCM, Beukers W, van der Keur KA, Munoz JR, Bangma CH, Lingsma HF, Eijkemans MJC, Schouten JP, Zwarthoff EC. A methylation assay for the detection of non-muscle-invasive bladder cancer (NMIBC) recurrences in voided urine. BJU Int. 2012;109:941–948. doi: 10.1111/j.1464-410X.2011.10428.x. [DOI] [PubMed] [Google Scholar]
  • 205.Li W, Xia D, Wang Y, Li Y, Xue Y, Wu X, Ye Z. Relationship between aberrant methylation of FAS promoter and biological behavior of bladder urothelial carcinoma. J. Huazhong Univ. Sci. Technolog. Med. Sci. 2011;31:794–798. doi: 10.1007/s11596-011-0679-6. [DOI] [PubMed] [Google Scholar]
  • 206.Costa VL, Henrique R, Danielsen SA, Duarte-Pereira S, Eknaes M, Skotheim RI, Rodrigues A, Magalhães JS, Oliveira J, Lothe RA, Teixeira MR, Jerónimo C, Lind GE. Three epigenetic biomarkers, GDF15, TMEFF2, and VIM, accurately predict bladder cancer from DNA-based analyses of urine samples. Clin. Cancer Res. 2010;16:5842–5851. doi: 10.1158/1078-0432.CCR-10-1312. [DOI] [PubMed] [Google Scholar]
  • 207.Reinert T, Modin C, Castano FM, Lamy P, Wojdacz TK, Hansen LL, Wiuf C, Borre M, Dyrskjøt L, Ørntoft TF. Comprehensive genome methylation analysis in bladder cancer; identification and validation of novel methylated genes and application of these as urinary tumor markers. Clin. Cancer Res. 2011;17:5582–5592. doi: 10.1158/1078-0432.CCR-10-2659. [DOI] [PubMed] [Google Scholar]
  • 208.Dyrskjøt L, Thykjaer T, Kruhøffer M, Jensen JL, Marcussen N, Hamilton-Dutoit S, Wolf H, Orntoft TF. Identifying distinct classes of bladder carcinoma using microarrays. Nat. Genet. 2003;33:90–96. doi: 10.1038/ng1061. [DOI] [PubMed] [Google Scholar]
  • 209.Satge D, Sasco AJ, Day S, Culine S. A lower risk of dying from urological cancer in Down syndrome: clue for cancer protecting genes on chromosome 21. Urol. Int. 2009;82:296–300. doi: 10.1159/000209361. [DOI] [PubMed] [Google Scholar]
  • 210.Malinge S, Izraeli S, Crispino JD. Insights into the manifestations, outcomes, and mechanisms of leukemogenesis in Down syndrome. Blood. 2009;113:2619–2628. doi: 10.1182/blood-2008-11-163501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Cebrian V, Alvarez M, Aleman A, Palou J, Bellmunt J, Gonzalez-Peramato P, Cordón-Cardo C, García J, Piulats JM, Sánchez-Carbayo M. Discovery of myopodin methylation in bladder cancer. J. Pathol. 2008;216:111–119. doi: 10.1002/path.2390. [DOI] [PubMed] [Google Scholar]
  • 212.Aleman A, Cebrian V, Alvarez M, Lopez V, Orenes E, Lopez-Serra L, Algaba F, Bellmunt J, López-Beltrán A, Gonzalez-Peramato P, Cordón-Cardo C, García J, del Muro JG, Esteller M, Sánchez-Carbayo M. Identification of PMF1 methylation in association with bladder cancer progression. Clin. Cancer Res. 2008;14:8236–8243. doi: 10.1158/1078-0432.CCR-08-0778. [DOI] [PubMed] [Google Scholar]
  • 213.Hoque MO, Begum S, Brait M, Jeronimo C, Zahurak M, Ostrow KL, Rosenbaum E, Trock B, Westra WH, Schoenberg M, Goodman SN, Sidransky D. Tissue inhibitor of metalloproteinases-3 promoter methylation is an independent prognostic factor for bladder cancer. J. Urol. 2008;179:743–747. doi: 10.1016/j.juro.2007.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Renard I, Joniau S, van Cleynenbreugel B, Collette C, Naome C, Vlassenbroeck I, Nicolas H, de Leval J, Straub J, Van Criekinge W, Hamida W, Hellel M, Thomas A, de Leval L, Bierau K, Waltregny D. Identification and validation of the methylated TWIST1 and NID2 genes through real-time methylation-specific polymerase chain reaction assays for the noninvasive detection of primary bladder cancer in urine samples. Eur Urol. 2010;58:96–104. doi: 10.1016/j.eururo.2009.07.041. [DOI] [PubMed] [Google Scholar]
  • 215.Friedrich MG, Weisenberger DJ, Cheng JC, Chandrasoma S, Siegmund KD, Gonzalgo ML, Toma MI, Huland H, Yoo C, Tsai YC, Nichols PW, Bochner BH, Jones PA, Liang G. Detection of methylated apoptosis-associated genes in urine sediments of bladder cancer patients. Clin. Cancer Res. 2004;10:7457–7465. doi: 10.1158/1078-0432.CCR-04-0930. [DOI] [PubMed] [Google Scholar]
  • 216.Sobti RC, MalekZadeh K, Nikbakht M, Sadeghi IA, Shekari M, Singh SK. Hypermethylation-mediated partial transcriptional silencing of DAP-kinase gene in bladder cancer. Biomarkers. 2010;15:167–174. doi: 10.3109/13547500903395124. [DOI] [PubMed] [Google Scholar]
  • 217.Serizawa RR, Ralfkiaer U, Steven K, Lam GW, Schmiedel S, Schüz J, Hansen AB, Horn T, Guldberg P. Integrated genetic and epigenetic analysis of bladder cancer reveals an additive diagnostic value of FGFR3 mutations and hypermethylation events. Int. J. Cancer. 2011;129:78–87. doi: 10.1002/ijc.25651. [DOI] [PubMed] [Google Scholar]
  • 218.Lokeshwar VB, Gomez P, Kramer M, Knapp J, McCornack MA, Lopez LE, Fregien N, Dhir N, Scherer S, Klumpp DJ, Manoharan M, Soloway MS, Lokeshwar BL. Epigenetic regulation of HYAL-1 hyaluronidase expression. identification of HYAL-1 promoter. J. Biol. Chem. 2008;283:29215–29227. doi: 10.1074/jbc.M801101200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Mori K, Enokida H, Kagara I, Kawakami K, Chiyomaru T, Tatarano S, Kawahara K, Nishiyama K, Seki N, Nakagawa M. CpG hypermethylation of collagen type I alpha 2 contributes to proliferation and migration activity of human bladder cancer. Int. J. Oncol. 2009;34:1593–1602. doi: 10.3892/ijo_00000289. [DOI] [PubMed] [Google Scholar]
  • 220.Wang XS, Zhang Z, Wang HC, Cai JL, Xu QW, Li MQ, Chen YC, Qian XP, Lu TJ, Yu LZ, Zhang Y, Xin DQ, Na YQ, Chen WF. Rapid identification of UCA1 as a very sensitive and specific unique marker for human bladder carcinoma. Clin. Cancer Res. 2006;12:4851–4858. doi: 10.1158/1078-0432.CCR-06-0134. [DOI] [PubMed] [Google Scholar]
  • 221.Wang F, Lia X, Xie XJ, Zhao L, Chen W. UCA1, a non-protein-coding RNA up-regulated in bladder carcinoma and embryo, influencing cell growth and promoting invasion. FEBS Lett. 2008;582:1919–1927. doi: 10.1016/j.febslet.2008.05.012. [DOI] [PubMed] [Google Scholar]
  • 222.Yang C, Li X, Wang Y, Zhao L, Chen W. Long non-coding RNA UCA1 regulated cell cycle distribution via CREB through PI3-K dependent pathway in bladder carcinoma cells. Gene. 2012;496:8–16. doi: 10.1016/j.gene.2012.01.012. [DOI] [PubMed] [Google Scholar]
  • 223.Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat. Rev. Genet. 2009;10:704–714. doi: 10.1038/nrg2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Choudhry H, Catto JW. Epigenetic regulation of microRNA expression in cancer. Methods Mol. Biol. 2010;676:165–184. doi: 10.1007/978-1-60761-863-8_12. [DOI] [PubMed] [Google Scholar]
  • 225.Dudziec E, Miah S, Choudhury HMZ, Owen HC, Blizard S, Glover M, Hamdy FC, Catto JW. Hypermethylation of CpG islands and shores around specific microRNAs and mirtrons is associated with the phenotype and presence of bladder cancer. Clin. Cancer Res. 2011;17:1287–1296. doi: 10.1158/1078-0432.CCR-10-2017. [DOI] [PubMed] [Google Scholar]
  • 226.Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233. doi: 10.1016/j.cell.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Guo H, Ingolia NT, Weissman JS, Bartel DP. Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 2010;466:835–840. doi: 10.1038/nature09267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Catto JWF, Alcaraz A, Bjartell AS, De Vere White R, Evans CP, Fussel S, Hamdy FC, Kallioniemi O, Mengual L, Schlomm T, Visakorpi T. MicroRNA in prostate, bladder, and kidney cancer: a systematic review. Eur. Urol. 2011;59:671–681. doi: 10.1016/j.eururo.2011.01.044. [DOI] [PubMed] [Google Scholar]
  • 229.Wu D, Tao J, Xu B, Li P, Lu Q, Zhang W. Downregulation of Dicer, a component of the microRNA machinery, in bladder cancer. Mol. Med. Report. 2012;5:695–699. doi: 10.3892/mmr.2011.711. [DOI] [PubMed] [Google Scholar]
  • 230.Wiklund ED, Bramsen JB, Hulf T, Dyrskjøt L, Ramanathan R, Hansen TB, Villadsen SB, Gao S, Ostenfeld MS, Borre M, Peter ME, Ørntoft TF, Kjems J, Clark SJ. Coordinated epigenetic repression of the miR-200 family and miR-205 in invasive bladder cancer. Int. J. Cancer. 2011;128:1327–1334. doi: 10.1002/ijc.25461. [DOI] [PubMed] [Google Scholar]
  • 231.Zhu J, Jiang Z, Gao F, Hu X, Zhou L, Chen J, Luo H, Sun J, Wu S, Han Y, Yin G, Chen M, Han Z, Li X, Huang Y, Zhang W, Zhou F, Chen T, Fa P, Wang Y, Sun L, Leng H, Sun F, Liu Y, Ye M, Yang H, Cai Z, Gui Y, Zhang X. A systematic analysis on DNA methylation and the expression of both mRNA and microRNA in bladder cancer. PLoS One. 2011;6(11 ):e28223. doi: 10.1371/journal.pone.0028223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Han Y, Chen J, Zhao X, Liang C, Wang Y, Sun L, Jiang Z, Zhang Z, Yang R, Chen J, Li Z, Tang A, Li X, Ye J, Guan Z, Gui Y, Cai Z. MicroRNA expression signatures of bladder cancer revealed by deep sequencing. PLoS One. 2011;6(3 ):e18286. doi: 10.1371/journal.pone.0018286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Li X, Chen J, Hu X, Huang Y, Li Z, Zhou L, Tian Z, Ma H, Wu Z, Chen M, Han Z, Peng Z, Zhao X, Liang C, Wang Y, Sun L, Chen J, Zhao J, Jiang B, Yang H, Gui Y, Cai Z, Zhang X. Comparative mRNA and microRNA expression profiling of three genitourinary cancers reveals common hallmarks and cancer-specific molecular events. PLoS One. 2011;6(7 ):e22570. doi: 10.1371/journal.pone.0022570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Catto JWF, Miah S, Owen HC, Bryant H, Myers K, Dudziec E, Larré S, Milo M, Rehman I, Rosario DJ, Di Martino E, Knowles MA, Meuth M, Harris AL, Hamdy FC. Distinct microRNA alterations characterize high- and low-grade bladder cancer. Cancer Res. 2009;69:8472–8481. doi: 10.1158/0008-5472.CAN-09-0744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Veerla S, Lindgren D, Kvist A, Frigyesi A, Staaf J, Persson H, Liedberg F, Chebil G, Gudjonsson S, Borg A, Månsson W, Rovira C, Höglund M. MiRNA expression in urothelial carcinomas: important roles of miR-10a, miR-222, miR-125b, miR-7 and miR-452 for tumor stage and metastasis, and frequent homozygous losses of miR-31. Int. J. Cancer. 2009;124:2236–2242. doi: 10.1002/ijc.24183. [DOI] [PubMed] [Google Scholar]
  • 236.Dalmay T, Edwards DR. MicroRNAs and the hallmarks of cancer. Oncogene. 2006;25:6170–6175. doi: 10.1038/sj.onc.1209911. [DOI] [PubMed] [Google Scholar]
  • 237.Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 2007;133:647–658. doi: 10.1053/j.gastro.2007.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Tao J, Lu Q, Wu D, Li P, Xu B, Qing W, Wang M, Zhang Z, Zhang W. microRNA-21 modulates cell proliferation and sensitivity to doxorubicin in bladder cancer cells. Oncol. Rep. 2011;25:1721–1729. doi: 10.3892/or.2011.1245. [DOI] [PubMed] [Google Scholar]
  • 239.Zhu S, Si ML, Wu H, Mo YY. MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1) J. Biol. Chem. 2007;282:14328–14336. doi: 10.1074/jbc.M611393200. [DOI] [PubMed] [Google Scholar]
  • 240.Catto JW, Xinarianos G, Burton JL, Meuth M, Hamdy FC. Differential expression of hMLH1 and hMSH2 is related to bladder cancer grade, stage and prognosis but not microsatellite instability. Int. J. Cancer. 2003;105:484–490. doi: 10.1002/ijc.11109. [DOI] [PubMed] [Google Scholar]
  • 241.Oeggerli M, Tomovska S, Schraml P, Calvano-Forte D, Scha-froth S, Simon R, Gasser T, Mihatsch MJ, Sauter G. E2F3 amplification and overexpression is associated with invasive tumor growth and rapid tumor cell proliferation in urinary bladder cancer. Oncogene. 2004;23:5616–5623. doi: 10.1038/sj.onc.1207749. [DOI] [PubMed] [Google Scholar]
  • 242.Huang L, Luo J, Cai Q, Pan Q, Zeng H, Guo Z, Dong W, Huang J, Lin T. MicroRNA-125b suppresses the development of bladder cancer by targeting E2F3. Int. J. Cancer. 2011;128:1758–1769. doi: 10.1002/ijc.25509. [DOI] [PubMed] [Google Scholar]
  • 243.Miles WO, Tschöp K, Herr A, Ji J-Y, Dyson NJ. Pumilio facilitates miRNA regulation of the E2F3 oncogene. Genes Dev. 2012;26:356–368. doi: 10.1101/gad.182568.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Yoshino H, Enokida H, Chiyomaru T, Tatarano S, Hidaka H, Yamasaki T, Gotannda T, Tachiwada T, Nohata N, Yamane T, Seki N, Nakagawa M. Tumor suppressive microRNA-1 mediated novel apoptosis pathways through direct inhibition of splicing factor serine/arginine-rich 9 (SRSF9/SRp30c) in bladder cancer. Biochem. Biophys. Res. Commun. 2012;417:588–593. doi: 10.1016/j.bbrc.2011.12.011. [DOI] [PubMed] [Google Scholar]
  • 245.Neely LA, Rieger-Christ KM, Silva Neto B, Eroshkin A, Garver J, Patel S, Phung NA, McLaughlin S, Libertino JA, Whitney D, Summerhayes IC. A microRNA expression ratio defining the invasive phenotype in bladder tumors. Urol. Oncol. 2010;28:39–48. doi: 10.1016/j.urolonc.2008.06.006. [DOI] [PubMed] [Google Scholar]
  • 246.Lin Y, Wu J, Chen H, Mao Y, Liu Y, Mao Q, Yang K, Zheng X, Xie L. Cyclin-dependent kinase 4 is a novel target in microRNA-195-mediated cell cycle arrest in bladder cancer cells. FEBS Lett. 2012;586:442–447. doi: 10.1016/j.febslet.2012.01.027. [DOI] [PubMed] [Google Scholar]
  • 247.Adam L, Zhong M, Choi W, Qi W, Nicoloso M, Arora A, Calin G, Wang H, Siefker-Radtke A, McConkey D, Bar-Eli M, Dinney C. miR-200 expression regulates epithelial-to-mesenchymal transition in bladder cancer cells and reverses resistance to epidermal growth factor receptor therapy. Clin. Cancer Res. 2009;15:5060–5072. doi: 10.1158/1078-0432.CCR-08-2245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Kenney PA, Wszolek MF, Rieger-Christ KM, Neto BS, Gould JJ, Harty NJ, Mosquera JM, Zeheb R, Loda M, Darling DS, Libertino JA, Summerhayes IC. Novel ZEB1 expression in bladder tumorigenesis. BJU Int. 2011;107:656–563. doi: 10.1111/j.1464-410X.2010.09489.x. [DOI] [PubMed] [Google Scholar]
  • 249.Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, Brabletz T. A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008;9:582–589. doi: 10.1038/embor.2008.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Shen A, Zhang Y, Yang H, Xu R, Huang G. Overexpression of ZEB1 relates to metastasis and invasion in osteosarcoma. J. Surg. Oncol. 2011. [DOI] [PubMed]
  • 251.Chen H, Lin Y-W, Mao Y-Q, Wu J, Liu Y-F, Zheng X-Y, Xie L-P. MicroRNA-449a acts as a tumor suppressor in human bladder cancer through the regulation of pocket proteins. Cancer Lett. 2012;320:40–47. doi: 10.1016/j.canlet.2012.01.027. [DOI] [PubMed] [Google Scholar]
  • 252.Ueno K, Hirata H, Majid S, Yamamura S, Shahryari V, Tabatabai ZL, Hinoda Y, Dahiya R. Tumor suppressor microRNA-493 decreases cell motility and migration ability in human bladder cancer cells by down-regulating RhoC and FZD4. Mol. Cancer Ther. 2012;11:244–253. doi: 10.1158/1535-7163.MCT-11-0592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Hirata H, Hinoda Y, Ueno K, Shahryari V, Tabatabai ZL, Dahiya R. MicroRNA-1826 tar gets VEGFC, beta-catenin (CTNNB1) and MEK1 (MAP2K1) in human bladder cancer. Carcinogenesis. 2012;33:41–48. doi: 10.1093/carcin/bgr239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Lin TX, Dong W, Huang J, Pan Q, Fan X, Zhang C, Huang L. MicroRNA-143 as a tumor suppressor for bladder cancer. J. Urol. 2009;181:1372–1380. doi: 10.1016/j.juro.2008.10.149. [DOI] [PubMed] [Google Scholar]
  • 255.Noguchi S, Mori T, Hoshino Y, Maruo K, Yamada N, Kitade Y, Naoe T, Akao Y. MicroRNA-143 functions as a tumor suppressor in human bladder cancer T24 cells. Cancer Lett. 2011;307:211–220. doi: 10.1016/j.canlet.2011.04.005. [DOI] [PubMed] [Google Scholar]
  • 256.Villadsen SB, Bramsen JB, Ostenfeld MS, Wiklund ED, Fristrup N, Gao S, Hansen TB, Jensen TI, Borre M, Ørntoft TF, Dyrskjøt L, Kjems J. The miR-143/-145 cluster regulates plasminogen activator inhibitor-1 in bladder cancer. Br. J. Cancer. 2012;106:366–374. doi: 10.1038/bjc.2011.520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Gottardo F, Liu CG, Ferracin M, Calin GA, Fassan M, Bassi P, Sevignani C, Byrne D, Negrini M, Pagano F, Gomella LG, Croce CM, Baffa R. Micro-RNA profiling in kidney and bladder cancers. Urol. Oncol. 2007;25:387–392. doi: 10.1016/j.urolonc.2007.01.019. [DOI] [PubMed] [Google Scholar]
  • 258.Garofalo M, Di Leva G, Romano G, Nuovo G, Suh SS, Ngankeu A, Taccioli C, Pichiorri F, Alder H, Secchiero P, Gasparini P, Gonelli A, Costinean S, Acunzo M, Condorelli G, Croce CM. miR-221 and 222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell. 2009;16:498–509. doi: 10.1016/j.ccr.2009.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 259.Dyrskjøt L, Ostenfeld MS, Bramsen JB, Silahtaroglu AN, Lamy P, Ramanathan R, Fristrup N, Jensen JL, Andersen CL, Zieger K, Kauppinen S, Ulhøi BP, Kjems J, Borre M, Orntoft TF. Genomic profiling of microRNAs in bladder cancer: miR-129 is associated with poor outcome and promotes cell death in vitro. Cancer Res. 2009;69:4851–4860. doi: 10.1158/0008-5472.CAN-08-4043. [DOI] [PubMed] [Google Scholar]
  • 260.Boominathan L. The tumor suppressors p53, p63, and p73 are regulators of microRNA processing complex. PLoS One. 2010;5(5 ):e10615. doi: 10.1371/journal.pone.0010615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Tao J, Wu D, Li P, Xu B, Lu Q, Zhang W. microRNA-18a, a member of the oncogenic miR-17-92 cluster, targets Dicer and suppresses cell proliferation in bladder cancer T24 cells. Mol. Med. Report. 2012;5:167–172. doi: 10.3892/mmr.2011.591. [DOI] [PubMed] [Google Scholar]
  • 262.Ichimi T, Enokida H, Okuno Y, Kunimoto R, Chiyomaru T, Kawamoto K, Kawahara K, Toki K, Kawakami K, Nishiyama K, Tsujimoto G, Nakagawa M, Seki N. Identification of novel microRNA targets based on microRNA signatures in bladder cancer. Int. J. Cancer. 2009;125:345–352. doi: 10.1002/ijc.24390. [DOI] [PubMed] [Google Scholar]
  • 263.Baffa R, Fassan M, Volinia S, O'Hara B, Liu CG, Palazzo JP, Gardiman M, Rugge M, Gomella LG, Croce CM, Rosenberg A. MicroRNA expression profiling of human metastatic cancers identifies cancer gene targets. J. Pathol. 2009;219:214–221. doi: 10.1002/path.2586. [DOI] [PubMed] [Google Scholar]
  • 264.Hanke M, Hoefig K, Merz H, Feller AC, Kausch I, Jocham D, Warnecke JM, Sczakiel G. A robust methodology to study urine microRNA as tumor marker: microRNA-126 and microRNA-182 are related to urinary bladder cancer. Urol. Oncol. 2010;28:655–661. doi: 10.1016/j.urolonc.2009.01.027. [DOI] [PubMed] [Google Scholar]
  • 265.Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, Galas DJ, Wang K. The microRNA spectrum in 12 body fluids. Clin. Chem. 2010;56:1733–1741. doi: 10.1373/clinchem.2010.147405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Catto JWF, Abbod MF, Wild PJ, Linkens DA, Pilarsky C, Rehman I, Rosario DJ, Denzinger S, Burger M, Stoehr R, Knuechel R, Hartmann A, Hamdy FC. The application of artificial intelligence to microarray data: identification of a novel gene signature to identify bladder cancer progression. Eur. Urol. 2010;57:398–406. doi: 10.1016/j.eururo.2009.10.029. [DOI] [PubMed] [Google Scholar]
  • 267.Wang G, Zhang H, He H, Tong W, Wang B, Liao G, Chen Z, Du C. Up-regulation of microRNA in bladder tumor tissue is not common. Int. Urol. Nephrol. 2010;42:95–102. doi: 10.1007/s11255-009-9584-3. [DOI] [PubMed] [Google Scholar]
  • 268.Chen Y-H, Wang S-Q, Wu X-L, Shen M, Chen Z-G, Chen X-G, Liu Y-X, Zhu X-L, Guo F, Duan X-Z, Han X-C, Tao Z-H. Characterization of microRNAs expression profiling in one group of Chinese urothelial cell carcinoma identified by Solexa sequencing. Urol. Oncol. 2011. [DOI] [PubMed]

Articles from Current Genomics are provided here courtesy of Bentham Science Publishers

RESOURCES