Skip to main content
Asian Journal of Urology logoLink to Asian Journal of Urology
. 2016 Sep 14;3(4):211–222. doi: 10.1016/j.ajur.2016.09.005

Understanding the biology of urothelial cancer metastasis

Takashi Kobayashi 1
PMCID: PMC5730871  PMID: 29264189

Abstract

Management of unresectable urothelial cancer (UC) has been a clinical challenge for decades. While drug resistance is a key issue, precise understanding of biology of UC metastasis is another challenge for the improvement of treatment outcome of UC patients. Introduction of the cell biology concepts including epithelial-mesenchymal transition (EMT) and cancer stemness seems to explain UC metastasis. Molecular genetics based on gene expression profiling, next generation sequencing, and explosion of non-coding RNA world has opened the door to intrinsic molecular subtyping of UC. Next steps include, based on the recently accumulated understanding, the establishment of novel disease models representing UC metastasis in various experimental platforms, particularly in vivo animal systems. Indeed, novel knowledge molecular genetics has not been fully linked to the modeling of UC metastasis. Further understanding of bladder carcinogenesis is needed particularly with regard to cell of origin related to tumor characteristics including driver gene alterations, pathological differentiations, and metastatic ability. Then we will be able to establish better disease models, which will consequently lead us to further understanding of biology and eventually the development of novel therapeutic strategies for UC metastasis.

Keywords: Urothelial carcinoma, Metastasis, Epithelial-mesenchymal transition, Animal models

1. Introduction

Bladder cancer is the sixth most common malignancy excluding non-melanoma skin cancers with an estimation of 330,380 new cases and 123,051 deaths from bladder cancer worldwide in 2012 [1]. Approximately 95% of bladder cancers are histologically classified as urothelial carcinoma (hereafter referred as UC, formerly called transitional cell carcinoma) with the exception of a particular area where squamous cell carcinoma due to chronic infection of Schistosoma hematobiumis more prevalent. Urothelium, where UC is originated, lines all through the urinary tract except for distal urethra. Therefore, UC can arise from renal pelvis, ureter, bladder, and proximal urethra.

As many of other malignancies, bladder UC is a disease of older individuals. Patients are typically in their fifth or greater decades of life. The majority of bladder UC occur in males where there is an approximately 2- to 3-fold greater incidence compared with females. In the United States, Caucasians are at highest risk for bladder UC among all races including African, Asian, and Latin Americans. It is known that smoking and occupational or endemic exposures to certain chemicals predispose us to UC. The underlying mechanisms that link urothelial carcinogenesis to the sexual and racial disparities and risk factors including aging and carcinogens are currently not fully understood.

Treatment and prognosis of UC depend on several key factors including anatomical site, extent (stage), and histological grade of the disease. Non-muscle-invasive bladder UC (NMIBC),with the exception of carcinoma in situ (CIS), can be treated by transurethral resection with excellent survival outcomes, whereas muscle-invasive bladder UC (MIBC) and upper urinary tract UC (UTUC) often need radical cystectomy (RC) or nephroureterectomy (RNU). While these treatments are usually indicated with a curative intent, there are currently few curative treatment options for a metastatic or recurrent UC that has progressed outside of the urinary tract. MIBC is associated with higher incidence of distant metastasis compared with NMIBC. Bladder UC often metastasize to lymph nodes, bone, lung, liver, and peritoneum [2]. Systemic chemotherapy, the standard treatment for metastatic UC, can hardly achieve durable disease control. Therefore, treatment outcome of the patients with metastatic UC has been very poor with approximately 15% of overall survival rate [3].

Thus, it is apparently important to understand the underlying biology for metastatic progression of UC. Recently published series of molecular genetics of bladder cancer provided novel information for intrinsic molecular subtyping of UC [4], [5], [6], which will potentially lead us to the effective prevention and cure of this currently lethal form of the disease. This article reviews recent key findings that have been accumulated in the research field of UC metastasis, barriers that hamper our research progression, and future perspectives that may potentially overcome them.

2. Cell biology of UC metastasis

2.1. Epithelial-mesenchymal transition (EMT)

Several cellular processes are implicated in metastatic progression of UC. EMT is referred as a complex process that reprograms and transmogrifies epithelial cells to mesenchymal phenotype characterized by loss of cell adhesion and polarity. Although EMT is a phenomenon that physiologically observed during development and wound healing, it has long been implicated in cancer metastasis and treatment resistance. As essential roles of EMT in urothelial cancer metastasis was extensively discussed in an excellent review by McConkey et al. [7] in 2009, this article focuses on relatively recent findings.

One of the most particular molecular characteristics of cells undergoing EMT is downregulation of surface CDH-1 (cadherin 1, also known as E-cadherin) and EMT is best characterized by decreased expression of CDH-1 and increased expression of CDH-2 (N-cadherin). Indeed, aberrantly attenuated expression of CDH-1 was reported to be associated with high progression rate of bladder UC [8], [9]. Recently Al-Ahmadie et al. [10] observed truncating somatic mutations in the CDH-1 gene in 84% of plasmacytoid bladder cancers, a highly invasive histological variant of UC. Knock-out of CDH-1 in bladder UC cells enhanced cell migration, suggesting that loss of CDH-1 expression is not just a marker for EMT, but has some central, causal, and functional significance in tumor invasion and progression.

It is well known that numerous signaling pathways involving TGFβ, integrins, Notch, Wnt, and sonic hedgehog (SHH) induce EMT [7], [11]. Recent studies addressed molecular mechanisms involved in TGFβ-induced EMT in UC cells. Those studies revealed malat-1 [12], [13] and EIF5A2 [14] downstream mediators of TGFβ signaling pathway that induce EMT. Another study showed that PPM1A functions as a negative regulator of EMT by dephosphorylating TGFβ-activated Smad2/3 [15]. Although those reports suggest that TGFβ signaling promotes EMT in UC, another study showed that GDF15, a member of TGFβ superfamily, inhibits EMT through upregulating mammary serine protease inhibitor (MASPIN) and N-myc downstream-regulated family genes (NDRG1, NDRG2, and NDRG3) [16]. Additionally, it is yet to be fully understood what cellular interaction including auto- and para-crine mechanisms induce TGFβ signaling in the microenvironment of UC tumors.

Several recent reports suggested that integrins and associated signaling pathways are implicated in EMT of UC. Integrins mediate cellular adhesion to extracellular matrix (ECM). In the process of EMT, coordinated regulation of the integrin-mediated cell–ECM adhesion and E-cadherin-mediated cell–cell adhesion is required [17]. A report showed that knockdown of αv integrinsled UC cells to a shift towards more epithelial track characterized by increased CDH-1/CDH-2 ratio and downregulation of EMT-associated genes including SNAI2, NANOG, BMI1, ALDH1 [18]. Importantly, these phenotypic changes were associated with decreased metastatic growth ability of the cells. Integrin-linked kinase (ILK) is highly evolutionally conserved serine/threonine kinase binding to β1 integrin [19]. Like other focal adhesion molecules such as c-Src and FAK, it mediates outside-inside signal transduction from ECM–integrin interaction. It was reported that ILK expression is higher in mesenchymal UC cells compared with epithelial ones [20]. Exogenous expression of ILK led epithelial UC cells to mesenchymal shift through activation of GSK3β–Zeb1 pathway. Importantly, ILK expression is positively correlated with invasive phenotype of human and murine bladder UC. These findings indicate that ECM–integrin adhesion plays a key role for cancer cell plasticity as well as E-cadherin-mediated cell–cell adhesion.

When we consider that EMT occurs physiologically during embryonic development and tissue repair, it is not surprising that EMT in cancer cells are also regulated by the developmental signaling pathways including SHH, Wnt, and Notch pathways. Specifically, recent studies have shed light on the role of SHH signaling in urothelium and UC. Beachy's group found that basal urothelial cells expressing SHH gave a rise of whole urothelial layer [21]. They also demonstrated that SHH-expressing basal cells were essential for the development of CIS, an early form of mesenchymal UC [22]. However, they observed that SHH expression in CIS was lost as it progresses to invasive bladder UC. Additionally, they reported that inhibition of SHH dramatically accelerated tumor progression and decreased survival time [23]. These findings suggest a tumor-suppressive function of SHH signaling despite another essential role in the maintenance of epithelial stem cell or tumor-initiating cells. It is consistent with a previous report showing more abundant SHH expression in NMIBC compared with MIBC [24]. However, there have been a few reports showing positive correlations between SHH expression and clinicopathological aggressiveness of bladder UC [25], [26]. Accordingly, the role of SHH in UC is warranted to be elucidated in the future studies as it can be a potent therapeutic target.

It is well known that Wnt/β-catenin signaling pathway also regulates EMT in various cancers [27]. β-catenin interacts with E-cadherin or triggers the activation of EMT-inducing transcription factors including SNAIL (Snail 1/2/3), TWIST (Twist 1/2), and ZEB (Zeb 1/2). It was reported that homeodomain-interacting protein kinase-2 (Hipk2) negatively regulated EMT and subsequent invasion by inhibiting Wnt/β-catenin signaling pathway in UC cells [28]. Several other reports also demonstrated that Wnt/β-catenin pathway activation induced EMT in UC cells [13], [29].

Recent studies have shed light on several different aspects of the biology of Notch in bladder cancer including EMT. A report revealed that Notch acted as a tumor suppressor by inhibiting ERK pathway [30]. Another report showed that disruption of Rbpj and Psen, important mediators of Notch signaling, promoted bladder squamous cell carcinoma (SCC) with mesenchymal features in N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced mouse bladder carcinogenesis model [31]. Another group, inspired by genomic gain of Notch2 in The Cancer Genome Atlas (-TCGA) dataset, has shown that overexpression of Notch2-intracellular domain (N2ICD) promoted growth and EMT of UC cells [32]. These findings indicate that Notch1/2/3 have distinct and context-dependent functions in various aspects of biological processes in UC. Further understanding is needed for specific targeting of Notch pathways in the management of UC patients.

Finally, some carcinogenic chemical substances have been reported to induce EMT in UC cells. Benzidine is known as a strong urothelial carcinogen. A previous report showed that benzidine induced EMT of normal bladder urothelial cells through activation of ERK1/2 pathway [33]. Chronic exposure to arsenic acid causes UC as well as skin and lung cancers [34]. It was reported that arsenic acid induced EMT in lung and prostate epithelial cells [35], [36], [37]. However, precise mechanisms of arsenic acid-induced UC carcinogenesis or progression have not been elucidated. Aristolochic acid, a compound found in Chinese herbs, is known to induce progressive tubulointerstitial fibrosis, chronic renal insufficiency, and upper urinary tract UC [38]. Aristolochic acid was reported to induce EMT in human proximal tubule epithelial cells [39], while affected urothelial cells frequently undergo T→A transversion in codon 139 of exon 5 of p53 gene [40]. Mechanistic association between the two observations is not fully understood and subject to the future studies. These agents have been implicated in carcinogenesis based on their potential to induce EMT on normal epithelial cells. As described above and elsewhere [7], however, EMT is strongly associated with local invasion and metastatic progression of cancer cells as well. Indeed, it was reported that bladder cancers associated with arsenic acid exposure was more aggressive than those in unexposed patients [41].

2.2. Cancer stem cell or tumor-initiating cell

Several investigators reported that cancer stem cells or tumor initiating cells of UC were successfully isolated based on the expression of cell surface markers or some functional molecules [42], [43], [44], [45]. In addition to markers commonly proposed for tumor-initiating cells of solid cancer including CD24, CD44, CD133, and ALDH1A1, several unique markers such as CK14 have been identified as UC stem cell markers [45]. Intriguingly, tumor-initiating cells displayed distinct marker profiles according to stage and grade of original tumor, suggesting that phenotype of tumor-initiating cells defines biological and clinical aggressiveness of bladder UC [46].

Cancer stem cell hypothesis is an important concept for the understanding of cancer biology including the multistep process of tumor metastasis. Provided that cancer stem cell theory is closely associated with the process of EMT [45], [47], [48], it is not surprising that tumor-initiating cells seem to play an important role in UC metastasis, while it is also reported to be responsible for chemo-resistance of UC [45], [49]. Indeed, cancer stemness and EMT were mutually linked by common markers including OCT4, NANOG, SNAI1/2, ZEB1/2 and TWIST [42], [45]. Indeed, expressions of various stem cell markers including OCT4 [50], [51], ALDH1A1 [52], ZEB1/2 [53], and TWIST1 [54] were reported to be associated with bladder UC metastasis (extensively reviewed by van der Horst et al. [45]). However, another study on stem cell markers and EMT signature questioned the strict correlation between cancer stemness and EMT in bladder UC cells [55].

Apart from EMT, Overdevest et al. [56] reported that bladder UC cells expressing stem cell marker CD24 had higher potential of lung metastasis. CD24-expressing cells showed higher early retention to the lungs after tail vein injection, suggesting that CD24 expression promoted lung colonization. However, another group reported that CD24 expression did not alter tumor-initiating ability in the subcutaneous xenograft model [57].

Importantly, matched pair immunohistochemical analysis using primary bladder UC and lung metastasis tumors demonstrated that lung metastatic lesion expressed higher CD24 compared with corresponding primary tumors. Additionally, another study showed that CD24 expression was associated with inferior post-RC survival in bladder UC patients [57].

CD24 is considered as a luminal marker as it is expressed in superficial umbrella cells of normal urothelium [57] and luminal subtypes of UC [5]. It is notable that a luminal marker is associated with higher metastasis ability and poor prognosis of cancer. In this regard, these traits of CD24 as a molecular marker for poor prognosis and luminal subtypes have been also reported in breast cancer [58], suggesting that CD24 has a common function correlated with metastatic ability and luminal characteristics. Further studies are warranted for the elucidation of CD24 function in cancer in the future.

2.3. Role of tumor microenvironment

It is obvious that above-mentioned cellular signaling pathways (TGFβ, integrins, Notch, Wnt, and SHH) regulating EMT and tumor stemness are closely associate with tumor microenvironment, which has been increasingly studied in recent years as exemplified by the Beachy's reports showing epithelial-mesenchymal interaction mediated by SHH and Wnt in urothelial regeneration [21] and UC development and progression [22], [23]. Another report showed that, in the tumor microenvironment, UC cells and recruited mast cells cooperatively enhanced EMT and metastasis by modulating ERβ/CCL2/CCR2 signaling pathway [59]. Another group investigated infiltration of tumor-associated macrophage (TAM) by assessing CD163 expression [60]. CD163 expression was positively correlated with local expression of IL-6 and IL-10. Interestingly, CD163 expression was observed not only on TAMs but also on tumor cells in some cases. Moreover, CD163 expression on tumor cells was significantly associated with more advanced disease stage, higher histological grade, and inferior clinical outcomes. More recently, some investigators focused on exosome that promotes EMT in an autocrine manner [61]. Indeed, exosomes extracted from some mesenchymal bladder UC cells contained EMT-related proteins and reduced expression of CDH-1 and β-catenin in urothelial cells [62], [63].

Like other solid tumors [47], [64], [65], it is believed that UC requires tumor-associated stroma to maintain tumor-initiating cells [43], [45], [47]. Thus, epithelial-mesenchymal interaction in tumor microenvironment is increasingly recognized as important biological processes that are essential to induce EMT or maintain tumor-initiating cells of UC. This should be subject to further investigation in the future studies.

3. Metastasis promoters and suppressors of UC

Recent efforts in UC research identified a number of metastasis-related genes in UC. Our group identified RalGAP complex as the GTPase-activating protein (GAP) of small G-protein Ral [66], which has been implicated in bladder cancer metastasis and progression [67], [68], [69]. Indeed, activity of Ral in bladder UC cells was inversely correlated with of RalGAP expression. The expression RalGAP is downregulated in high grade bladder UC and exogenous expression of RalGAP attenuated cellular metastatic potentials. Additionally, RalGAPa2 knock-out mice developed more invasive bladder cancers compared with wild-type animals after administration of bladder carcinogen BBN [70]. Our results suggested that RalGAP functions as a tumor metastasis suppressor through regulating the Ral pathway in bladder UC.

p63, a member of p53 tumor suppressor family, is a basal cell marker and implicated in maintenance of normal urothelial stem cells. Similar to breast cancer, p63 is categorized as a basal epithelial marker in UC [5], [71]. There are several isoforms for p63 harboring (Tp63) or lacking (ΔNp63) N-terminal transactivation domain [72]. It was reported that Tp63 was abundantly expressed in non-invasive bladder UC [73]. Additionally, the same group and others reported that loss of p63 expression was associated with muscle-invasive progression or aggressive pathological phenotypes of UC [72], [74], [75], [76]. Indeed, a number of reports showed metastasis suppressor function of p63 in various cancers [77], [78]. These works collectively suggest a tumor metastasis suppressor function of Tp63 in UC. According to previous reports studying ΔNp63 variants specifically, however, a subset of muscle-invasive bladder cancer expressed ΔNp63 and maintenance of ΔNp63 expression is significantly associated with UC-specific mortality [72], [73]. Collectively, p63 isoforms seem to have distinct functional significance that is also context-dependent. Future studies should better dissociate expression profiles and molecular function of p63 isoforms at various biological steps, which would enable us to understand the role of this interesting molecule for the UC pathogenesis.

Several receptor tyrosine kinases including epithelial growth factor receptor [79], [80], FGFR (fibroblast growth factor receptor) [81] and c-Met [80], [82] have been implicated in UC metastasis (also extensively reviewed by McConkey et al. [7]). However, it is well known that dependency on FGFR signals is considered as one of the characteristics of epithelial UC. Consistently, it was reported that mesenchymal UC cells are more likely to show resistance to an FGFR receptor tyrosine kinase inhibitor [83]. As for transcription factors, FOXQ1 was reported to promote EMT of UC cells upon stimulation of TGFβ as in other malignancies [84]. KLF4 was reported to be downregulated in UC and act as a metastasis suppressor inhibiting EMT [85]. Androgen receptor (AR) was reported to promote UC metastasis by affecting CD24 [86], [87], [88]. Several investigators reported that infiltrating neutrophils [89] and B lymphocytes [90] modulated AR transactivation leading to elevated MMPs expression and cell invasion. Interestingly, these reports and others [91] showed that AR in UC cells activated in ligand-independent manners.

c-Src is one of the oldest proto-oncogene that encodes non-receptor tyrosin kinase. c-Src has been recognized to promote metastasis of cancers by modifying focal adhesion and cell–cell junction mediated by integrins and cadherins [92], [93]. However, Theodorescu's group [94] recently reported that c-Src acted as metastasis suppressor in bladder UC thorough phosphorylating RhoGDI2. Their reports showed that expression of c-Src was inversely correlated with tumor stage of bladder UC [94], [95]. Considering that a number of Src inhibitors such as dasatinib have been developed for cancer treatment, the bilateral roles of c-Src for cancer metastasis should be further elucidated in the future.

Recently microRNAs (miRNA) have been reported to regulate various biological processes including tumor invasion and metastasis. To date, miR-200 family (miR-200a, miR-200b, miR-200c, miR-141 and miR-429) have been most extensively studied [96], [97], [98], [99], [100], while many others such as miR-205 [101], miR-429 [102], miR-218 [103], miR-23b [80], [104], miR-34a [105], miR-451 [106], miR-145 [107], and miR-433 [108] have been reported to act as a metastasis promoter or suppressor. More recent studies have shed light on long non-coding RNAs (lncRNA) including lncRNA-UCA1 [109], lncRNA-HOTAIR [110], and MALAT-1 [13] as metastasis promoters. It is intriguing that these lncRNAs were reported to induce EMT through upregulating EMT-related genes such as ZEB1/2. Accordingly, lncRNAs seem to be a promising research subject for potential therapeutic targets and clinically useful urine markers.

4. Therapeutic opportunities

Recent efforts showed some promising findings that potentially lead us to effective prevention or treatment of UC metastasis. Several Cox-2 inhibitors was shown to inhibit UC cell growth as well as reversal of EMT characterized by induction of CDH-1 and reduction of SNAIL expression [111]. A micro-tubule-targeting agent vinflunine was also reported to reverse EMT [112]. Vinflunine increased CDH-1 by stabilizing it through inhibiting Hakai, E3-ubiquitin ligase of CDH-1. Silibinin, a natural flavonoid, was reported to inhibit GSK3β/β-catenin signaling, decrease ZEB1 expression, and subsequently suppress metastasis through blocking MET and stemness of UC cells [113]. A report showed the possibility that reactivation of p53 through RNA activation using small activating RNA may inhibit cell growth and EMT of UC cells [114]. A plant alkaloid tetrandrine was reported to act as GLI-1 inhibitor blocking Hedgehog signaling and subsequently suppress EMT and metastasis of UC cells [115]. Finally, Theodorescu's group [116] discovered novel small molecules RBC8 and BQU57 that inhibited Ral activity. They showed promising effects of these drugs on UC cell growth, however it is also anticipated that these drugs may also suppress metastasis of UC since Ral pathways have been implicated in metastasis of various cancers including UC [68], [69], [70] and others [117], [118], [119], [120].

5. Experimental models of UC metastasis

It is essential to have useful models that recapitulate pathogenesis, biology, and treatment response for the development of novel treatments of human disease. Currently a variety of experimental systems are available for the research on UC metastasis.

5.1. Cell culture experimental systems

A panel of UC cell lines is currently available with the information for their origin and genetic profile [121]. Cell culture-based experiments related to metastasis research include wound healing (“scratch”) assay, Boyden chamber transwell assays. Wound healing assay is an easy, well-developed and inexpensive method that has been employed for years to study cell motility [122]. Boyden chamber transwell assay utilizes chemotaxis and leads cells to go across a polycarbonate membrane with micropores (3–15 μm in diameter). If the membrane is coated with ECM such as matrigel or collagens, one can see the cellular potential to invade across basement membrane. Transwell assays with different ECM substances may provide information for key molecules that facilitate invasion such as matrix metalloproteinases (MMPs) [123]. They are useful assays to dissociate intrinsic cellular motility or migratory potential. Indeed, abilities of migration and invasion are not necessarily parallel [124]. Additionally, higher motility or invasiveness in these assays does not always mean high metastatic potential of the cells in vivo.

5.2. Cell line-based in vivo experimental systems (Table 1)

Table 1.

Urothelial carcinoma cell lines for metastasis researches.

Cell line Source Application
253Ja MIBC Orthotopic intramural injection model (LN met.) [137] (Liver met.) [130]
Orthotopic intravesical injection model (LN and lung mets.) [135], [136]
Caudal vein injection model (lung met.) [130]
J82 MIBC Intraperitoneal injection model (intraperitoneal spread) [153]
Subcutaneous injection model (liver met.) [128]
Caudal vein injection model (liver and lung mets.) [128]
UM-UC-3a Met. Subcutaneous injection model (lung met.) [129]
Caudal vein injection model (lung met.) [56], [95], [132]
Orthotopic intramural injection model (liver and lung mets.) [82]
UM-UC-14 MIBC Orthotopic intramural injection model (LN mets.) [138]
T24 MIBC Caudal vein injection model (lung met.) [131]
TSU-Pr1a,b MIBC Orthotopic intramural injection model (lung met.) [134]
Intracardiac (left ventricle) injection (lung, liver, brain, LN, bone mets.) [133], [134]
Intratibial injection (mimicking tumor growth at bone metastatic lesion) [134]
EJ MIBC Caudal vein injection model (lung met.) [114]

LN, lymph node; MIBC, muscle-invasive bladder carcinoma; Met., metastatic bladder carcinoma.

a

Including derivative sublines.

b

Derivative of T24 [154].

Various approaches currently available to model bladder cancer in vivo are mostly utilizing mouse [125]. Cell-based xenograft models are widely used with their potential advantages. Preparation of the cells is easy including genetic manipulation for functional assays or reporter expression for in vivo imaging. However, it inevitably requires immunocompromised animals. Experimental systems based on immunocompromised host animals limit the investigation of role of immune systems in UC metastasis. Additionally, the results from these systems should be carefully interpreted provided the known importance of the immune system for cancer metastasis [126]. Use of syngeneic system with mouse bladder cancer cells established from carcinogen-induced tumors seems to be a good solution in the future.

Subcutaneous injection model is one of the most widely used cell-based xenograft experimental systems. It is technically simple and allows easy access to the tumor. Renal subcapsular engraftment was also reported [127]. It seems to yield higher take rate due to more abundant blood flow but requires more sophisticated experimental skills. Although there are numerous previous reports studying tumor growth at engraftment site, there are the limited number of studies that addressed metastatic ability of the UC cells using these ectopic engraftment models [128], [129].

Direct injection of tumor cells into circulation is more widely used for metastasis researches. In the intravenous injection, caudal vein injection in most cases, tumor cells enter the right ventricular (pulmonary) circulatory system and subsequently form lung metastasis [56], [70], [95], [114], [128], [130], [131], [132]. On the other hand, in the left ventricular intracardiac injection, tumor cells enter the left ventricular (systemic) circulatory system and subsequently metastasize to bone, liver and brain [133], [134]. These experimental systems allow us to focus biological behavior of the tumor cells after intravasation into the circulation.

Orthotopic engraft models can be stratified into intravesical injection (instillation into bladder lumen) [135], [136]and intramural injection (inoculation into bladder wall) [82], [130], [137], [138]. The former primarily mimics the seeding implantation of superficial bladder cancer and the latter submucosal progression of infiltrating bladder cancer. Both methods can be used to investigate metastasis once after successful tumor engraftment and progression. These models have an advantage of representing tumor microenvironment at the primary site and reflecting multistep metastatic process more faithfully. However, the rate of metastasis formation are usually much lower compared with intravascular injection models. The primary tumor at the engraftment site often grows rapidly and the host animal has to be sacrificed before the development of metastatic lesions [139]. Additionally, the intramural inoculation used to require bladder exposure via lower abdominal incision and very sophisticated skills for precise injection. A recent report has shown a modification with less invasive and more precise procedure using ultrasonography scan [140].

5.3. Patient-derived tumor graft

Patient-derived xenograft (PDX) is a classical experimental system that has been revisited and growingly become important [141], [142]. Almost all PDX lines are subcutaneously engrafted and passaged. This seems to be the main reason for no prior report on the application of PDX lines for metastasis researches although only one previous report showed highly invasive characteristics of the xenograft [143]. In this regard, orthotopic engraftment of PDX tumor or dissociated tumor cells seems to be a resolution although it will be technically challenging.

5.4. Chemical-induced bladder cancer

Although a number of chemical carcinogens are known to induce bladder cancer in rodents, BBN is currently most widely used for bladder cancer research. Mice administered with BBN in the drinking water develop CIS and then muscle invasive bladder cancer eventually. A problem is the model is high prevalence of squamous cell carcinoma [144], [145], which accounts for less than 10% of human invasive bladder cancer [146]. Only a few studies reported the metastatic progression in this model [86], [144]. BBN-induced bladder carcinogenesis model has a potential to be combined with genetically engineered mice and can be used for functional association of metastasis-related genes. Additionally, syngeneic bladder tumors developed in immunocompetent animals are suitable for immune-oncological investigations that are becoming increasingly crucial for the era [147].

5.5. Genetically-engineered mouse (GEM)

As well as the BBN-induced model, GEM models can represent de novo carcinogenesis in immunocompetent hosts. Therefore, given relevant genetic alterations in the right population of the urothelial cells, GEM may recapitulate the pathogenesis and tumor microenvironment of human disease more faithfully. However, bladder cancer is still underrepresented by GEM models compared with other types of malignancies although there are an increasing number of GEM models of bladder cancer being reported [125]. Most models develop non-muscle-invasive disease and metastatic progression is rare even in invasive bladder cancer models with a few exceptions.

One of these exceptions is SV40T-based transgenic mouse models. Wu's group [148] first established a GEM model harboring Simian Virus 40 T antigen (SV40T) transgene driven by uroplakin II promoter. Although mice having low copy number of SV40T transgenes developed only CIS, those having high copy number developed invasive UC and some of them showed metastatic progression to lymph nodes and liver. Another group reported a similar GEM model based on SV40T transgene driven by cytokeratin 19 (Krt19) promoter [149]. This model was characterized by lung metastasis that was observed in 25% of the mice. Unfortunately, non-specific activation of Krt19 led the mice to develop tumor lesions in other organs including adrenal glands, prostate and mesothelium. In these GEM models, however, metastasis is observed in a subset of affected mice and other investigators reported that they found no metastasis in similar models [150], [151]. These findings suggest that these gene alterations are not sufficient and that the accumulation of other mutations or tumor suppressor inactivation are required metastatic progression.

Another problem in using SV40T-based model is the low relevancy of the gene alteration that drives mouse bladder carcinogenesis and progression. The role of SV40T in human UC is very limited, although inactivation of p53 and Rb, major targets of SV40T, is among the most frequently observed genetic events in human MIBC [4] and molecular profiles of SV40T-based mouse bladder cancers were reported to be conserved with those of human disease [151].

Another GEM model for MIBC metastasis is based on inactivation of Trp53 and Pten in urothelium [127]. This model is characterized by stochastic gene recombination in urothelium induced by Adeno virus expressing Cre recombinase that is directly injected into bladder lumen. It was reported that about 60% of affected mice developed macroscopically visible metastatic spread. Metastatic loci were observed most frequently in regional lymph nodes, while distant metastatic lesions were detected in spleen, liver, and diaphragm.

However, UC metastasis has not been studied extensively using these autochthonous mouse models. There seem to be several aspects of difficulties that hamper UC metastasis research using autochthonous mouse models. A primary tumor often becomes lethal causing obstructive renal insufficiency before the tumor cells metastasize to distant organs. Bladder tumor of 1 cm in diameter can kill the host mouse, while a tumor of that size is hardly identified with concomitant distant metastasis. Higher induction rate (whatever chemical exposure or gene recombination) or rapid growth can be an advantage for studies on primary tumor, but those characteristics may sometimes make metastasis research difficult. In this regard, use of lentivirus that usually yields lower infection efficiency compared with that of Adeno virus may benefit as succeeded in a lung cancer research [152]. Another issue is that anatomies of human and mouse urinary tracts are not exactly same, with particular regard to blood supply and lymphatic drainage. Mouse bladder is not enveloped with fat tissue and not insulated from peritoneal cavity by dense peritoneum like human bladder. While recent efforts have provided relatively detailed information about bladder cancer genetics [4], [6], [42], [71], molecular correlation between subtypes of human UC and GEM models are still under investigation. Since the cell of origin for MIBC is still inconclusive [22], [145], there are still limited options to induce gene alterations specifically to the urothelium. We need to overcome these barriers in order to establish more relevant and efficient GEM models for UC metastasis research in the future.

6. Future directions

The biology of UC metastasis is not fully understood. The lack of experimental models that accurately recapitulate the human disease has been one of the main barriers that has precluded our precise understanding of UC metastasis. Although several concepts in cell biology such as EMT or cancer stem cell appear to be attractive to explain biology of UC metastasis, it does not seem to be fully linked to in vivo findings in animal models. There is an urgent need for the establishment of novel animal models that recapitulate molecular, genetic, and clinical characteristics of human disease. It is anticipated that precise modeling of the disease will lead us to more profound understanding and consequently the development of novel therapeutic strategy to overcome metastasis of UC.

Conflicts of interest

The authors declare no conflict of interest.

Acknowledgments

The author apologizes to my colleagues for my inability to refer many important studies due to the limitation of the length. I thank Dr. Osamu Ogawa for helpful discussions. This work was supported by Grants-in-aid for Scientific Research to the author from Japan Society for the Promotion of Science (25713055).

Footnotes

Peer review under responsibility of Second Military Medical University.

References

  • 1.GLOBOCAN 2012: estimated cancer incidence, mortality and prevalence worldwide in 2012. 2016/05/29. http://globocan.iarc.fr/Default.aspx [Google Scholar]
  • 2.Shinagare A.B., Ramaiya N.H., Jagannathan J.P., Fennessy F.M., Taplin M.E., Van den Abbeele A.D. Metastatic pattern of bladder cancer: correlation with the characteristics of the primary tumor. AJR Am J Roentgenol. 2011;196:117–122. doi: 10.2214/AJR.10.5036. [DOI] [PubMed] [Google Scholar]
  • 3.von der Maase H., Sengelov L., Roberts J.T., Ricci S., Dogliotti L., Oliver T. Long-term survival results of a randomized trial comparing gemcitabine plus cisplatin, with methotrexate, vinblastine, doxorubicin, plus cisplatin in patients with bladder cancer. J Clin Oncol. 2005;23:4602–4608. doi: 10.1200/JCO.2005.07.757. [DOI] [PubMed] [Google Scholar]
  • 4.Cancer Genome Atlas Research Network Comprehensive molecular characterization of urothelial bladder carcinoma. Nature. 2014;507:315–322. doi: 10.1038/nature12965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Choi W., Czerniak B., Ochoa A., Su X., Siefker-Radtke A., Dinney C. Intrinsic basal and luminal subtypes of muscle-invasive bladder cancer. Nat Rev Urol. 2014;11:400–410. doi: 10.1038/nrurol.2014.129. [DOI] [PubMed] [Google Scholar]
  • 6.Damrauer J.S., Hoadley K.A., Chism D.D., Fan C., Tiganelli C.J., Wobker S.E. Intrinsic subtypes of high-grade bladder cancer reflect the hallmarks of breast cancer biology. Proc Natl Acad Sci USA. 2014;111:3110–3115. doi: 10.1073/pnas.1318376111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.McConkey D.J., Choi W., Marquis L., Martin F., Williams M.B., Shah J. Role of epithelial-to-mesenchymal transition (EMT) in drug sensitivity and metastasis in bladder cancer. Cancer Metastasis Rev. 2009;28:335–344. doi: 10.1007/s10555-009-9194-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Muramaki M., Miyake H., Terakawa T., Kumano M., Sakai I., Fujisawa M. Expression profile of E-cadherin and N-cadherin in non-muscle-invasive bladder cancer as a novel predictor of intravesical recurrence following transurethral resection. Urol Oncol. 2012;30:161–166. doi: 10.1016/j.urolonc.2010.01.005. [DOI] [PubMed] [Google Scholar]
  • 9.Breyer J., Gierth M., Shalekenov S., Aziz A., Schafer J., Burger M. Epithelial-mesenchymal transformation markers E-cadherin and survivin predict progression of stage pTa urothelial bladder carcinoma. World J Urol. 2016;34:709–716. doi: 10.1007/s00345-015-1690-5. [DOI] [PubMed] [Google Scholar]
  • 10.Al-Ahmadie H.A., Iyer G., Lee B.H., Scott S.N., Mehra R., Bagrodia A. Frequent somatic CDH1 loss-of-function mutations in plasmacytoid variant bladder cancer. Nat Genet. 2016;48:356–358. doi: 10.1038/ng.3503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Peinado H., Olmeda D., Cano A. Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer. 2007;7:415–428. doi: 10.1038/nrc2131. [DOI] [PubMed] [Google Scholar]
  • 12.Fan Y., Shen B., Tan M., Mu X., Qin Y., Zhang F. TGF-beta-induced upregulation of malat1 promotes bladder cancer metastasis by associating with suz12. Clin Cancer Res. 2014;20:1531–1541. doi: 10.1158/1078-0432.CCR-13-1455. [DOI] [PubMed] [Google Scholar]
  • 13.Ying L., Chen Q., Wang Y., Zhou Z., Huang Y., Qiu F. Upregulated MALAT-1 contributes to bladder cancer cell migration by inducing epithelial-to-mesenchymal transition. Mol Biosyst. 2012;8:2289–2294. doi: 10.1039/c2mb25070e. [DOI] [PubMed] [Google Scholar]
  • 14.Wei J.H., Cao J.Z., Zhang D., Liao B., Zhong W.M., Lu J. EIF5A2 predicts outcome in localised invasive bladder cancer and promotes bladder cancer cell aggressiveness in vitro and in vivo. Br J Cancer. 2014;110:1767–1777. doi: 10.1038/bjc.2014.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Geng J., Fan J., Ouyang Q., Zhang X., Zhang X., Yu J. Loss of PPM1A expression enhances invasion and the epithelial-to-mesenchymal transition in bladder cancer by activating the TGF-beta/Smad signaling pathway. Oncotarget. 2014;5:5700–5711. doi: 10.18632/oncotarget.2144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tsui K.H., Hsu S.Y., Chung L.C., Lin Y.H., Feng T.H., Lee T.Y. Growth differentiation factor-15: a p53- and demethylation-upregulating gene represses cell proliferation, invasion, and tumorigenesis in bladder carcinoma cells. Sci Rep. 2015;5:12870. doi: 10.1038/srep12870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Canel M., Serrels A., Frame M.C., Brunton V.G. E-cadherin-integrin crosstalk in cancer invasion and metastasis. J Cell Sci. 2013;126:393–401. doi: 10.1242/jcs.100115. [DOI] [PubMed] [Google Scholar]
  • 18.van der Horst G., Bos L., van der Mark M., Cheung H., Heckmann B., Clement-Lacroix P. Targeting of alpha-v integrins reduces malignancy of bladder carcinoma. PLoS One. 2014;9:e108464. doi: 10.1371/journal.pone.0108464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Hannigan G.E., McDonald P.C., Walsh M.P., Dedhar S. Integrin-linked kinase: not so ‘pseudo’ after all. Oncogene. 2011;30:4375–4385. doi: 10.1038/onc.2011.177. [DOI] [PubMed] [Google Scholar]
  • 20.Matsui Y., Assi K., Ogawa O., Raven P.A., Dedhar S., Gleave M.E. The importance of integrin-linked kinase in the regulation of bladder cancer invasion. Int J Cancer. 2012;130:521–531. doi: 10.1002/ijc.26008. [DOI] [PubMed] [Google Scholar]
  • 21.Shin K., Lee J., Guo N., Kim J., Lim A., Qu L. Hedgehog/Wnt feedback supports regenerative proliferation of epithelial stem cells in bladder. Nature. 2011;472:110–114. doi: 10.1038/nature09851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Shin K., Lim A., Odegaard J.I., Honeycutt J.D., Kawano S., Hsieh M.H. Cellular origin of bladder neoplasia and tissue dynamics of its progression to invasive carcinoma. Nat Cell Biol. 2014;16:469–478. doi: 10.1038/ncb2956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Shin K., Lim A., Zhao C., Sahoo D., Pan Y., Spiekerkoetter E. Hedgehog signaling restrains bladder cancer progression by eliciting stromal production of urothelial differentiation factors. Cancer Cell. 2014;26:521–533. doi: 10.1016/j.ccell.2014.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pignot G., Vieillefond A., Vacher S., Zerbib M., Debre B., Lidereau R. Hedgehog pathway activation in human transitional cell carcinoma of the bladder. Br J Cancer. 2012;106:1177–1186. doi: 10.1038/bjc.2012.55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.He H.C., Chen J.H., Chen X.B., Qin G.Q., Cai C., Liang Y.X. Expression of hedgehog pathway components is associated with bladder cancer progression and clinical outcome. Pathol Oncol Res. 2012;18:349–355. doi: 10.1007/s12253-011-9451-2. [DOI] [PubMed] [Google Scholar]
  • 26.Islam S.S., Mokhtari R.B., Noman A.S., Uddin M., Rahman M.Z., Azadi M.A. Sonic hedgehog (Shh) signaling promotes tumorigenicity and stemness via activation of epithelial-to-mesenchymal transition (EMT) in bladder cancer. Mol Carcinog. 2016;55:537–551. doi: 10.1002/mc.22300. [DOI] [PubMed] [Google Scholar]
  • 27.Ghahhari N.M., Babashah S. Interplay between microRNAs and WNT/beta-catenin signalling pathway regulates epithelial-mesenchymal transition in cancer. Eur J Cancer. 2015;51:1638–1649. doi: 10.1016/j.ejca.2015.04.021. [DOI] [PubMed] [Google Scholar]
  • 28.Tan M., Gong H., Zeng Y., Tao L., Wang J., Jiang J. Downregulation of homeodomain-interacting protein kinase-2 contributes to bladder cancer metastasis by regulating Wnt signaling. J Cell Biochem. 2014;115:1762–1767. doi: 10.1002/jcb.24842. [DOI] [PubMed] [Google Scholar]
  • 29.Jing Y., Cui D., Guo W., Jiang J., Jiang B., Lu Y. Activated androgen receptor promotes bladder cancer metastasis via Slug mediated epithelial-mesenchymal transition. Cancer Lett. 2014;348:135–145. doi: 10.1016/j.canlet.2014.03.018. [DOI] [PubMed] [Google Scholar]
  • 30.Rampias T., Vgenopoulou P., Avgeris M., Polyzos A., Stravodimos K., Valavanis C. A new tumor suppressor role for the Notch pathway in bladder cancer. Nat Med. 2014;20:1199–1205. doi: 10.1038/nm.3678. [DOI] [PubMed] [Google Scholar]
  • 31.Maraver A., Fernandez-Marcos P.J., Cash T.P., Mendez-Pertuz M., Duenas M., Maietta P. NOTCH pathway inactivation promotes bladder cancer progression. J Clin Invest. 2015;125:824–830. doi: 10.1172/JCI78185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hayashi T., Gust K.M., Wyatt A.W., Goriki A., Jager W., Awrey S. Not all NOTCH is created equal: the oncogenic role of NOTCH2 in bladder Cancer and its implications for targeted therapy. Clin Cancer Res. 2016;22:2981–2992. doi: 10.1158/1078-0432.CCR-15-2360. [DOI] [PubMed] [Google Scholar]
  • 33.Zhao L., Geng H., Liang Z.F., Zhang Z.Q., Zhang T., Yu D.X. Benzidine induces epithelial-mesenchymal transition in human uroepithelial cells through ERK1/2 pathway. Biochem Biophys Res Commun. 2015;459:643–649. doi: 10.1016/j.bbrc.2015.02.163. [DOI] [PubMed] [Google Scholar]
  • 34.Steinmaus C., Yuan Y., Bates M.N., Smith A.H. Case-control study of bladder cancer and drinking water arsenic in the western United States. Am J Epidemiol. 2003;158:1193–1201. doi: 10.1093/aje/kwg281. [DOI] [PubMed] [Google Scholar]
  • 35.Riedmann C., Ma Y., Melikishvili M., Godfrey S.G., Zhang Z., Chen K.C. Inorganic Arsenic-induced cellular transformation is coupled with genome wide changes in chromatin structure, transcriptome and splicing patterns. BMC Genomics. 2015;16:212. doi: 10.1186/s12864-015-1295-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Stueckle T.A., Lu Y., Davis M.E., Wang L., Jiang B.H., Holaskova I. Chronic occupational exposure to arsenic induces carcinogenic gene signaling networks and neoplastic transformation in human lung epithelial cells. Toxicol Appl Pharmacol. 2012;261:204–216. doi: 10.1016/j.taap.2012.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Tokar E.J., Diwan B.A., Waalkes M.P. Arsenic exposure transforms human epithelial stem/progenitor cells into a cancer stem-like phenotype. Environ Health Perspect. 2010;118:108–115. doi: 10.1289/ehp.0901059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Patel N., Arya M., Muneer A., Powles T., Sullivan M., Hines J. Molecular aspects of upper tract urothelial carcinoma. Urol Oncol. 2014;32 doi: 10.1016/j.urolonc.2012.10.002. 28 e11–20. [DOI] [PubMed] [Google Scholar]
  • 39.Li Y., Wang Z., Wang S., Zhao J., Zhang J., Huang Y. Gremlin-mediated decrease in bone morphogenetic protein signaling promotes aristolochic acid-induced epithelial-to-mesenchymal transition (EMT) in HK-2 cells. Toxicology. 2012;297:68–75. doi: 10.1016/j.tox.2012.04.004. [DOI] [PubMed] [Google Scholar]
  • 40.Stiborova M., Frei E., Schmeiser H.H. Biotransformation enzymes in development of renal injury and urothelial cancer caused by aristolochic acid. Kidney Int. 2008;73:1209–1211. doi: 10.1038/ki.2008.125. [DOI] [PubMed] [Google Scholar]
  • 41.Moore L.E., Smith A.H., Eng C., Kalman D., DeVries S., Bhargava V. Arsenic-related chromosomal alterations in bladder cancer. J Natl Cancer Inst. 2002;94:1688–1696. doi: 10.1093/jnci/94.22.1688. [DOI] [PubMed] [Google Scholar]
  • 42.Chan K.S., Espinosa I., Chao M., Wong D., Ailles L., Diehn M. Identification, molecular characterization, clinical prognosis, and therapeutic targeting of human bladder tumor-initiating cells. Proc Natl Acad Sci USA. 2009;106:14016–14021. doi: 10.1073/pnas.0906549106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Brandt W.D., Matsui W., Rosenberg J.E., He X., Ling S., Schaeffer E.M. Urothelial carcinoma: stem cells on the edge. Cancer Metastasis Rev. 2009;28:291–304. doi: 10.1007/s10555-009-9187-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ho P.L., Kurtova A., Chan K.S. Normal and neoplastic urothelial stem cells: getting to the root of the problem. Nat Rev Urol. 2012;9:583–594. doi: 10.1038/nrurol.2012.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.van der Horst G., Bos L., van der Pluijm G. Epithelial plasticity, cancer stem cells, and the tumor-supportive stroma in bladder carcinoma. Mol Cancer Res. 2012;10:995–1009. doi: 10.1158/1541-7786.MCR-12-0274. [DOI] [PubMed] [Google Scholar]
  • 46.Volkmer J.P., Sahoo D., Chin R.K., Ho P.L., Tang C., Kurtova A.V. Three differentiation states risk-stratify bladder cancer into distinct subtypes. Proc Natl Acad Sci USA. 2012;109:2078–2083. doi: 10.1073/pnas.1120605109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Borovski T., De Sousa E.M.F., Vermeulen L., Medema J.P. Cancer stem cell niche: the place to be. Cancer Res. 2011;71:634–639. doi: 10.1158/0008-5472.CAN-10-3220. [DOI] [PubMed] [Google Scholar]
  • 48.Garg M. Urothelial cancer stem cells and epithelial plasticity: current concepts and therapeutic implications in bladder cancer. Cancer Metastasis Rev. 2015;34:691–701. doi: 10.1007/s10555-015-9589-6. [DOI] [PubMed] [Google Scholar]
  • 49.Kurtova A.V., Xiao J., Mo Q., Pazhanisamy S., Krasnow R., Lerner S.P. Blocking PGE2-induced tumour repopulation abrogates bladder cancer chemoresistance. Nature. 2015;517:209–213. doi: 10.1038/nature14034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Atlasi Y., Mowla S.J., Ziaee S.A., Bahrami A.R. OCT-4, an embryonic stem cell marker, is highly expressed in bladder cancer. Int J Cancer. 2007;120:1598–1602. doi: 10.1002/ijc.22508. [DOI] [PubMed] [Google Scholar]
  • 51.Chang C.C., Shieh G.S., Wu P., Lin C.C., Shiau A.L., Wu C.L. Oct-3/4 expression reflects tumor progression and regulates motility of bladder cancer cells. Cancer Res. 2008;68:6281–6291. doi: 10.1158/0008-5472.CAN-08-0094. [DOI] [PubMed] [Google Scholar]
  • 52.Su Y., Qiu Q., Zhang X., Jiang Z., Leng Q., Liu Z. Aldehyde dehydrogenase 1 A1-positive cell population is enriched in tumor-initiating cells and associated with progression of bladder cancer. Cancer Epidemiol Biomarkers Prev. 2010;19:327–337. doi: 10.1158/1055-9965.EPI-09-0865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sayan A.E., Griffiths T.R., Pal R., Browne G.J., Ruddick A., Yagci T. SIP1 protein protects cells from DNA damage-induced apoptosis and has independent prognostic value in bladder cancer. Proc Natl Acad Sci USA. 2009;106:14884–14889. doi: 10.1073/pnas.0902042106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhao D., Besser A.H., Wander S.A., Sun J., Zhou W., Wang B. Cytoplasmic p27 promotes epithelial-mesenchymal transition and tumor metastasis via STAT3-mediated Twist1 upregulation. Oncogene. 2015;34:5447–5459. doi: 10.1038/onc.2014.473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.McConkey D.J., Lee S., Choi W., Tran M., Majewski T., Lee S. Molecular genetics of bladder cancer: emerging mechanisms of tumor initiation and progression. Urol Oncol. 2010;28:429–440. doi: 10.1016/j.urolonc.2010.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Overdevest J.B., Thomas S., Kristiansen G., Hansel D.E., Smith S.C., Theodorescu D. CD24 offers a therapeutic target for control of bladder cancer metastasis based on a requirement for lung colonization. Cancer Res. 2011;71:3802–3811. doi: 10.1158/0008-5472.CAN-11-0519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hofner T., Macher-Goeppinger S., Klein C., Schillert A., Eisen C., Wagner S. Expression and prognostic significance of cancer stem cell markers CD24 and CD44 in urothelial bladder cancer xenografts and patients undergoing radical cystectomy. Urol Oncol. 2014;32:678–686. doi: 10.1016/j.urolonc.2014.01.001. [DOI] [PubMed] [Google Scholar]
  • 58.Kwon M.J., Han J., Seo J.H., Song K., Jeong H.M., Choi J.S. CD24 overexpression is associated with poor prognosis in luminal a and triple-negative breast Cancer. PLoS One. 2015;10:e0139112. doi: 10.1371/journal.pone.0139112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Rao Q., Chen Y., Yeh C.R., Ding J., Li L., Chang C. Recruited mast cells in the tumor microenvironment enhance bladder cancer metastasis via modulation of ERbeta/CCL2/CCR2 EMT/MMP9 signals. Oncotarget. 2016;7:7842–7855. doi: 10.18632/oncotarget.5467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Maniecki M.B., Etzerodt A., Ulhoi B.P., Steiniche T., Borre M., Dyrskjot L. Tumor-promoting macrophages induce the expression of the macrophage-specific receptor CD163 in malignant cells. Int J Cancer. 2012;131:2320–2331. doi: 10.1002/ijc.27506. [DOI] [PubMed] [Google Scholar]
  • 61.Greening D.W., Gopal S.K., Mathias R.A., Liu L., Sheng J., Zhu H.J. Emerging roles of exosomes during epithelial-mesenchymal transition and cancer progression. Semin Cell Dev Biol. 2015;40:60–71. doi: 10.1016/j.semcdb.2015.02.008. [DOI] [PubMed] [Google Scholar]
  • 62.Franzen C.A., Blackwell R.H., Todorovic V., Greco K.A., Foreman K.E., Flanigan R.C. Urothelial cells undergo epithelial-to-mesenchymal transition after exposure to muscle invasive bladder cancer exosomes. Oncogenesis. 2015;4:e163. doi: 10.1038/oncsis.2015.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Jeppesen D.K., Nawrocki A., Jensen S.G., Thorsen K., Whitehead B., Howard K.A. Quantitative proteomics of fractionated membrane and lumen exosome proteins from isogenic metastatic and nonmetastatic bladder cancer cells reveal differential expression of EMT factors. Proteomics. 2014;14:699–712. doi: 10.1002/pmic.201300452. [DOI] [PubMed] [Google Scholar]
  • 64.Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 65.Visvader J.E., Lindeman G.J. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 2008;8:755–768. doi: 10.1038/nrc2499. [DOI] [PubMed] [Google Scholar]
  • 66.Shirakawa R., Fukai S., Kawato M., Higashi T., Kondo H., Ikeda T. Tuberous sclerosis tumor suppressor complex-like complexes act as GTPase-activating proteins for Ral GTPases. J Biol Chem. 2009;284:21580–21588. doi: 10.1074/jbc.M109.012112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Smith S.C., Oxford G., Baras A.S., Owens C., Havaleshko D., Brautigan D.L. Expression of ral GTPases, their effectors, and activators in human bladder cancer. Clin Cancer Res. 2007;13:3803–3813. doi: 10.1158/1078-0432.CCR-06-2419. [DOI] [PubMed] [Google Scholar]
  • 68.Smith S.C., Theodorescu D. The Ral GTPase pathway in metastatic bladder cancer: key mediator and therapeutic target. Urol Oncol. 2009;27:42–47. doi: 10.1016/j.urolonc.2008.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Smith S.C., Baras A.S., Owens C.R., Dancik G., Theodorescu D. Transcriptional signatures of Ral GTPase are associated with aggressive clinicopathologic characteristics in human cancer. Cancer Res. 2012;72:3480–3491. doi: 10.1158/0008-5472.CAN-11-3966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Saito R., Shirakawa R., Nishiyama H., Kobayashi T., Kawato M., Kanno T. Downregulation of Ral GTPase-activating protein promotes tumor invasion and metastasis of bladder cancer. Oncogene. 2013;32:894–902. doi: 10.1038/onc.2012.101. [DOI] [PubMed] [Google Scholar]
  • 71.Choi W., Porten S., Kim S., Willis D., Plimack E.R., Hoffman-Censits J. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell. 2014;25:152–165. doi: 10.1016/j.ccr.2014.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Karni-Schmidt O., Castillo-Martin M., Shen T.H., Gladoun N., Domingo-Domenech J., Sanchez-Carbayo M. Distinct expression profiles of p63 variants during urothelial development and bladder cancer progression. Am J Pathol. 2011;178:1350–1360. doi: 10.1016/j.ajpath.2010.11.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Choi W., Shah J.B., Tran M., Svatek R., Marquis L., Lee I.L. p63 expression defines a lethal subset of muscle-invasive bladder cancers. PLoS One. 2012;7:e30206. doi: 10.1371/journal.pone.0030206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Guo C.C., Dadhania V., Zhang L., Majewski T., Bondaruk J., Sykulski M. Gene expression profile of the clinically aggressive micropapillary variant of bladder Cancer. Eur Urol. 2016 doi: 10.1016/j.eururo.2016.02.056. pii:S0302-2838(16)00246-3. http://dx.doi.org/10.1016/j.eururo.2016.02.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Koga F., Kawakami S., Kumagai J., Takizawa T., Ando N., Arai G. Impaired Delta Np63 expression associates with reduced beta-catenin and aggressive phenotypes of urothelial neoplasms. Br J Cancer. 2003;88:740–747. doi: 10.1038/sj.bjc.6600764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Urist M.J., Di Como C.J., Lu M.L., Charytonowicz E., Verbel D., Crum C.P. Loss of p63 expression is associated with tumor progression in bladder cancer. Am J Pathol. 2002;161:1199–1206. doi: 10.1016/S0002-9440(10)64396-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Giacobbe A., Compagnone M., Bongiorno-Borbone L., Antonov A., Markert E.K., Zhou J.H. p63 controls cell migration and invasion by transcriptional regulation of MTSS1. Oncogene. 2016;35:1602–1608. doi: 10.1038/onc.2015.230. [DOI] [PubMed] [Google Scholar]
  • 78.Melino G. p63 is a suppressor of tumorigenesis and metastasis interacting with mutant p53. Cell Death Differ. 2011;18:1487–1499. doi: 10.1038/cdd.2011.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wallerand H., Cai Y., Wainberg Z.A., Garraway I., Lascombe I., Nicolle G. Phospho-Akt pathway activation and inhibition depends on N-cadherin or phospho-EGFR expression in invasive human bladder cancer cell lines. Urol Oncol. 2010;28:180–188. doi: 10.1016/j.urolonc.2008.09.041. [DOI] [PubMed] [Google Scholar]
  • 80.Chiyomaru T., Seki N., Inoguchi S., Ishihara T., Mataki H., Matsushita R. Dual regulation of receptor tyrosine kinase genes EGFR and c-Met by the tumor-suppressive microRNA-23b/27b cluster in bladder cancer. Int J Oncol. 2015;46:487–496. doi: 10.3892/ijo.2014.2752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Cheng T., Roth B., Choi W., Black P.C., Dinney C., McConkey D.J. Fibroblast growth factor receptors-1 and -3 play distinct roles in the regulation of bladder cancer growth and metastasis: implications for therapeutic targeting. PLoS One. 2013;8:e57284. doi: 10.1371/journal.pone.0057284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Matsumoto R., Tsuda M., Wang L., Maishi N., Abe T., Kimura T. Adaptor protein CRK induces epithelial-mesenchymal transition and metastasis of bladder cancer cells through HGF/c-Met feedback loop. Cancer Sci. 2015;106:709–717. doi: 10.1111/cas.12662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hanze J., Henrici M., Hegele A., Hofmann R., Olbert P.J. Epithelial mesenchymal transition status is associated with anti-cancer responses towards receptor tyrosine-kinase inhibition by dovitinib in human bladder cancer cells. BMC Cancer. 2013;13:589. doi: 10.1186/1471-2407-13-589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Zhu Z., Zhu Z., Pang Z., Xing Y., Wan F., Lan D. Short hairpin RNA targeting FOXQ1 inhibits invasion and metastasis via the reversal of epithelial-mesenchymal transition in bladder cancer. Int J Oncol. 2013;42:1271–1278. doi: 10.3892/ijo.2013.1807. [DOI] [PubMed] [Google Scholar]
  • 85.Li H., Wang J., Xiao W., Xia D., Lang B., Wang T. Epigenetic inactivation of KLF4 is associated with urothelial cancer progression and early recurrence. J Urol. 2014;191:493–501. doi: 10.1016/j.juro.2013.08.087. [DOI] [PubMed] [Google Scholar]
  • 86.Overdevest J.B., Knubel K.H., Duex J.E., Thomas S., Nitz M.D., Harding M.A. CD24 expression is important in male urothelial tumorigenesis and metastasis in mice and is androgen regulated. Proc Natl Acad Sci USA. 2012;109:E3588–E3596. doi: 10.1073/pnas.1113960109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Agarwal N., Dancik G.M., Goodspeed A., Costello J.C., Owens C., Duex J.E. GON4L drives cancer growth through a YY1- androgen receptor-CD24 axis. Cancer Res. 2016;76:5175–5185. doi: 10.1158/0008-5472.CAN-16-1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ding G., Yu S., Cheng S., Li G., Yu Y. Androgen receptor (AR) promotes male bladder cancer cell proliferation and migration via regulating CD24 and VEGF. Am J Transl Res. 2016;8:578–587. [PMC free article] [PubMed] [Google Scholar]
  • 89.Lin C., Lin W., Yeh S., Li L., Chang C. Infiltrating neutrophils increase bladder cancer cell invasion via modulation of androgen receptor (AR)/MMP13 signals. Oncotarget. 2015;6:43081–43089. doi: 10.18632/oncotarget.5638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ou Z., Wang Y., Liu L., Li L., Yeh S., Qi L. Tumor microenvironment B cells increase bladder cancer metastasis via modulation of the IL-8/androgen receptor (AR)/MMPs signals. Oncotarget. 2015;6:26065–26078. doi: 10.18632/oncotarget.4569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Hsieh T.F., Chen C.C., Ma W.L., Chuang W.M., Hung X.F., Tsai Y.R. Epidermal growth factor enhances androgen receptor mediated bladder cancer progression and invasion via potentiation of AR transactivation. Oncol Rep. 2013;30:2917–2922. doi: 10.3892/or.2013.2792. [DOI] [PubMed] [Google Scholar]
  • 92.Yeatman T.J. A renaissance for SRC. Nat Rev Cancer. 2004;4:470–480. doi: 10.1038/nrc1366. [DOI] [PubMed] [Google Scholar]
  • 93.Liu W., Kovacevic Z., Peng Z., Jin R., Wang P., Yue F. The molecular effect of metastasis suppressors on Src signaling and tumorigenesis: new therapeutic targets. Oncotarget. 2015;6:35522–35541. doi: 10.18632/oncotarget.5849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Wu Y., Moissoglu K., Wang H., Wang X., Frierson H.F., Schwartz M.A. Src phosphorylation of RhoGDI2 regulates its metastasis suppressor function. Proc Natl Acad Sci USA. 2009;106:5807–5812. doi: 10.1073/pnas.0810094106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Thomas S., Overdevest J.B., Nitz M.D., Williams P.D., Owens C.R., Sanchez-Carbayo M. Src and caveolin-1 reciprocally regulate metastasis via a common downstream signaling pathway in bladder cancer. Cancer Res. 2011;71:832–841. doi: 10.1158/0008-5472.CAN-10-0730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Gregory P.A., Bert A.G., Paterson E.L., Barry S.C., Tsykin A., Farshid G. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008;10:593–601. doi: 10.1038/ncb1722. [DOI] [PubMed] [Google Scholar]
  • 97.Park S.M., Gaur A.B., Lengyel E., Peter M.E. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008;22:894–907. doi: 10.1101/gad.1640608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Adam L., Zhong M., Choi W., Qi W., Nicoloso M., Arora A. 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]
  • 99.Wiklund E.D., Bramsen J.B., Hulf T., Dyrskjot L., Ramanathan R., Hansen T.B. 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]
  • 100.Chen M.F., Zeng F., Qi L., Zu X.B., Wang J., Liu L.F. Transforming growth factorbeta1 induces epithelial mesenchymal transition and increased expression of matrix metalloproteinase16 via miR200b downregulation in bladder cancer cells. Mol Med Rep. 2014;10:1549–1554. doi: 10.3892/mmr.2014.2366. [DOI] [PubMed] [Google Scholar]
  • 101.Tran M.N., Choi W., Wszolek M.F., Navai N., Lee I.L., Nitti G. The p63 protein isoform DeltaNp63alpha inhibits epithelial-mesenchymal transition in human bladder cancer cells: role of MIR-205. J Biol Chem. 2013;288:3275–3288. doi: 10.1074/jbc.M112.408104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Wu C.L., Ho J.Y., Chou S.C., Yu D.S. MiR-429 reverses epithelial-mesenchymal transition by restoring E-cadherin expression in bladder cancer. Oncotarget. 2016;7:26593–26603. doi: 10.18632/oncotarget.8557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Yamasaki T., Seki N., Yoshino H., Itesako T., Hidaka H., Yamada Y. MicroRNA-218 inhibits cell migration and invasion in renal cell carcinoma through targeting caveolin-2 involved in focal adhesion pathway. J Urol. 2013;190:1059–1068. doi: 10.1016/j.juro.2013.02.089. [DOI] [PubMed] [Google Scholar]
  • 104.Majid S., Dar A.A., Saini S., Deng G., Chang I., Greene K. MicroRNA-23b functions as a tumor suppressor by regulating Zeb1 in bladder cancer. PLoS One. 2013;8:e67686. doi: 10.1371/journal.pone.0067686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Yu G., Yao W., Xiao W., Li H., Xu H., Lang B. MicroRNA-34a functions as an anti-metastatic microRNA and suppresses angiogenesis in bladder cancer by directly targeting CD44. J Exp Clin Cancer Res. 2014;33:779. doi: 10.1186/s13046-014-0115-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Zeng T., Peng L., Chao C., Fu B., Wang G., Wang Y. miR-451 inhibits invasion and proliferation of bladder cancer by regulating EMT. Int J Clin Exp Pathol. 2014;7:7653–7662. [PMC free article] [PubMed] [Google Scholar]
  • 107.Tan J., Qiu K., Li M., Liang Y. Double-negative feedback loop between long non-coding RNA TUG1 and miR-145 promotes epithelial to mesenchymal transition and radioresistance in human bladder cancer cells. FEBS Lett. 2015;589:3175–3181. doi: 10.1016/j.febslet.2015.08.020. [DOI] [PubMed] [Google Scholar]
  • 108.Xu X., Zhu Y., Liang Z., Li S., Xu X., Wang X. c-Met and CREB1 are involved in miR-433-mediated inhibition of the epithelial-mesenchymal transition in bladder cancer by regulating Akt/GSK-3beta/Snail signaling. Cell Death Dis. 2016;7:e2088. doi: 10.1038/cddis.2015.274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Xue M., Pang H., Li X., Li H., Pan J., Chen W. Long non-coding RNA urothelial cancer-associated 1 promotes bladder cancer cell migration and invasion by way of the hsa-miR-145-ZEB1/2-FSCN1 pathway. Cancer Sci. 2016;107:18–27. doi: 10.1111/cas.12844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Berrondo C., Flax J., Kucherov V., Siebert A., Osinski T., Rosenberg A. Expression of the long non-coding RNA HOTAIR correlates with disease progression in bladder Cancer and is contained in bladder cancer patient urinary exosomes. PLoS One. 2016;11:e0147236. doi: 10.1371/journal.pone.0147236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Adhim Z., Matsuoka T., Bito T., Shigemura K., Lee K.M., Kawabata M. In vitro and in vivo inhibitory effect of three Cox-2 inhibitors and epithelial-to-mesenchymal transition in human bladder cancer cell lines. Br J Cancer. 2011;105:393–402. doi: 10.1038/bjc.2011.262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Aparicio L.A., Castosa R., Haz-Conde M., Rodriguez M., Blanco M., Valladares M. Role of the microtubule-targeting drug vinflunine on cell-cell adhesions in bladder epithelial tumour cells. BMC Cancer. 2014;14:507. doi: 10.1186/1471-2407-14-507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Wu K., Ning Z., Zeng J., Fan J., Zhou J., Zhang T. Silibinin inhibits beta-catenin/ZEB1 signaling and suppresses bladder cancer metastasis via dual-blocking epithelial-mesenchymal transition and stemness. Cell Signal. 2013;25:2625–2633. doi: 10.1016/j.cellsig.2013.08.028. [DOI] [PubMed] [Google Scholar]
  • 114.Wang C., Ge Q., Zhang Q., Chen Z., Hu J., Li F. Targeted p53 activation by saRNA suppresses human bladder cancer cells growth and metastasis. J Exp Clin Cancer Res. 2016;35:53. doi: 10.1186/s13046-016-0329-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Zhang Y., Liu W., He W., Zhang Y., Deng X., Ma Y. Tetrandrine reverses epithelial-mesenchymal transition in bladder cancer by downregulating Gli-1. Int J Oncol. 2016;48:2035–2042. doi: 10.3892/ijo.2016.3415. [DOI] [PubMed] [Google Scholar]
  • 116.Yan C., Liu D., Li L., Wempe M.F., Guin S., Khanna M. Discovery and characterization of small molecules that target the GTPase Ral. Nature. 2014;515:443–447. doi: 10.1038/nature13713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Lim K.H., O'Hayer K., Adam S.J., Kendall S.D., Campbell P.M., Der C.J. Divergent roles for RalA and RalB in malignant growth of human pancreatic carcinoma cells. Curr Biol. 2006;16:2385–2394. doi: 10.1016/j.cub.2006.10.023. [DOI] [PubMed] [Google Scholar]
  • 118.Zipfel P.A., Brady D.C., Kashatus D.F., Ancrile B.D., Tyler D.S., Counter C.M. Ral activation promotes melanomagenesis. Oncogene. 2010;29:4859–4864. doi: 10.1038/onc.2010.224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Peschard P., McCarthy A., Leblanc-Dominguez V., Yeo M., Guichard S., Stamp G. Genetic deletion of RALA and RALB small GTPases reveals redundant functions in development and tumorigenesis. Curr Biol. 2012;22:2063–2068. doi: 10.1016/j.cub.2012.09.013. [DOI] [PubMed] [Google Scholar]
  • 120.Yin J., Pollock C., Tracy K., Chock M., Martin P., Oberst M. Activation of the RalGEF/Ral pathway promotes prostate cancer metastasis to bone. Mol Cell Biol. 2007;27:7538–7550. doi: 10.1128/MCB.00955-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Earl J., Rico D., Carrillo-de-Santa-Pau E., Rodriguez-Santiago B., Mendez-Pertuz M., Auer H. The UBC-40 urothelial bladder cancer cell line index: a genomic resource for functional studies. BMC Genomics. 2015;16:403. doi: 10.1186/s12864-015-1450-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Liang C.C., Park A.Y., Guan J.L. In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc. 2007;2:329–333. doi: 10.1038/nprot.2007.30. [DOI] [PubMed] [Google Scholar]
  • 123.Albini A., Benelli R. The chemo invasion assay: a method to assess tumor and endothelial cell invasion and its modulation. Nat Protoc. 2007;2:504–511. doi: 10.1038/nprot.2006.466. [DOI] [PubMed] [Google Scholar]
  • 124.Schaeffer D., Somarelli J.A., Hanna G., Palmer G.M., Garcia-Blanco M.A. Cellular migration and invasion uncoupled: increased migration is not an inexorable consequence of epithelial-to-mesenchymal transition. Mol Cell Biol. 2014;34:3486–3499. doi: 10.1128/MCB.00694-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Kobayashi T., Owczarek T.B., McKiernan J.M., Abate-Shen C. Modelling bladder cancer in mice: opportunities and challenges. Nat Rev Cancer. 2015;15:42–54. doi: 10.1038/nrc3858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.de Visser K.E., Eichten A., Coussens L.M. Paradoxical roles of the immune system during cancer development. Nat Rev Cancer. 2006;6:24–37. doi: 10.1038/nrc1782. [DOI] [PubMed] [Google Scholar]
  • 127.Puzio-Kuter A.M., Castillo-Martin M., Kinkade C.W., Wang X., Shen T.H., Matos T. Inactivation of p53 and Pten promotes invasive bladder cancer. Genes Dev. 2009;23:675–680. doi: 10.1101/gad.1772909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Lu Y.C., Chen C.N., Wang B., Hsu W.M., Chen S.T., Chang K.J. Changes in tumor growth and metastatic capacities of J82 human bladder cancer cells suppressed by down-regulation of calreticulin expression. Am J Pathol. 2011;179:1425–1433. doi: 10.1016/j.ajpath.2011.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Wu Z., Owens C., Chandra N., Popovic K., Conaway M., Theodorescu D. RalBP1 is necessary for metastasis of human cancer cell lines. Neoplasia. 2010;12:1003–1012. doi: 10.1593/neo.101080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Kim E.Y., Seo J.M., Kim C., Lee J.E., Lee K.M., Kim J.H. BLT2 promotes the invasion and metastasis of aggressive bladder cancer cells through a reactive oxygen species-linked pathway. Free Radic Biol Med. 2010;49:1072–1081. doi: 10.1016/j.freeradbiomed.2010.06.023. [DOI] [PubMed] [Google Scholar]
  • 131.Kuwada M., Chihara Y., Luo Y., Li X., Nishiguchi Y., Fujiwara R. Pro-chemotherapeutic effects of antibody against extracellular domain of claudin-4 in bladder cancer. Cancer Lett. 2015;369:212–221. doi: 10.1016/j.canlet.2015.08.019. [DOI] [PubMed] [Google Scholar]
  • 132.Wang H., Owens C., Chandra N., Conaway M.R., Brautigan D.L., Theodorescu D. Phosphorylation of RalB is important for bladder cancer cell growth and metastasis. Cancer Res. 2010;70:8760–8769. doi: 10.1158/0008-5472.CAN-10-0952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chaffer C.L., Dopheide B., McCulloch D.R., Lee A.B., Moseley J.M., Thompson E.W. Upregulated MT1-MMP/TIMP-2 axis in the TSU-Pr1-B1/B2 model of metastatic progression in transitional cell carcinoma of the bladder. Clin Exp Metastasis. 2005;22:115–125. doi: 10.1007/s10585-005-5141-3. [DOI] [PubMed] [Google Scholar]
  • 134.Chaffer C.L., Brennan J.P., Slavin J.L., Blick T., Thompson E.W., Williams E.D. Mesenchymal-to-epithelial transition facilitates bladder cancer metastasis: role of fibroblast growth factor receptor-2. Cancer Res. 2006;66:11271–11278. doi: 10.1158/0008-5472.CAN-06-2044. [DOI] [PubMed] [Google Scholar]
  • 135.Izawa J.I., Sweeney P., Perrotte P., Kedar D., Dong Z., Slaton J.W. Inhibition of tumorigenicity and metastasis of human bladder cancer growing in athymic mice by interferon-beta gene therapy results partially from various antiangiogenic effects including endothelial cell apoptosis. Clin Cancer Res. 2002;8:1258–1270. [PubMed] [Google Scholar]
  • 136.Singh A.V., Franke A.A., Blackburn G.L., Zhou J.R. Soy phytochemicals prevent orthotopic growth and metastasis of bladder cancer in mice by alterations of cancer cell proliferation and apoptosis and tumor angiogenesis. Cancer Res. 2006;66:1851–1858. doi: 10.1158/0008-5472.CAN-05-1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Chikazawa M., Inoue K., Fukata S., Karashima T., Shuin T. Expression of angiogenesis-related genes regulates different steps in the process of tumor growth and metastasis in human urothelial cell carcinoma of the urinary bladder. Pathobiology. 2008;75:335–345. doi: 10.1159/000164218. [DOI] [PubMed] [Google Scholar]
  • 138.Karashima T., Sweeney P., Kamat A., Huang S., Kim S.J., Bar-Eli M. Nuclear factor-kappaB mediates angiogenesis and metastasis of human bladder cancer through the regulation of interleukin-8. Clin Cancer Res. 2003;9:2786–2797. [PubMed] [Google Scholar]
  • 139.Jager W., Xue H., Hayashi T., Janssen C., Awrey S., Wyatt A.W. Patient-derived bladder cancer xenografts in the preclinical development of novel targeted therapies. Oncotarget. 2015;6:21522–21532. doi: 10.18632/oncotarget.3974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Jager W., Moskalev I., Janssen C., Hayashi T., Awrey S., Gust K.M. Ultrasound-guided intramural inoculation of orthotopic bladder cancer xenografts: a novel high-precision approach. PLoS One. 2013;8:e59536. doi: 10.1371/journal.pone.0059536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Inoue T., Terada N., Kobayashi T., Ogawa O. Patient-derived xenografts as in vivo models for basic and clinical research on urological malignancies. Nat Rev Urol. 2016 doi: 10.1038/nrurol.2017.19. [in press] [DOI] [PubMed] [Google Scholar]
  • 142.Bernardo C., Costa C., Sousa N., Amado F., Santos L. Patient-derived bladder cancer xenografts: a systematic review. Transl Res. 2015;166:324–331. doi: 10.1016/j.trsl.2015.02.001. [DOI] [PubMed] [Google Scholar]
  • 143.Hay J.H., Busuttil A., Steel C.M., Duncan W. The growth and histological characteristics of a series of human bladder cancer xenografts. Radiother Oncol. 1986;7:331–340. doi: 10.1016/s0167-8140(86)80063-9. [DOI] [PubMed] [Google Scholar]
  • 144.Yamamoto S., Masui T., Murai T., Mori S., Oohara T., Makino S. Frequent mutations of the p53 gene and infrequent H- and K-ras mutations in urinary bladder carcinomas of NON/Shi mice treated with N-butyl-N-(4-hydroxybutyl) nitro-samine. Carcinogenesis. 1995;16:2363–2368. doi: 10.1093/carcin/16.10.2363. [DOI] [PubMed] [Google Scholar]
  • 145.Van Batavia J., Yamany T., Molotkov A., Dan H., Mansukhani M., Batourina E. Bladder cancers arise from distinct urothelial sub-populations. Nat Cell Biol. 2014;16:982–991. doi: 10.1038/ncb3038. 1-5. [DOI] [PubMed] [Google Scholar]
  • 146.Amin M.B. Histological variants of urothelial carcinoma: diagnostic, therapeutic and prognostic implications. Mod Pathol. 2009;22(Suppl 2):S96–S118. doi: 10.1038/modpathol.2009.26. [DOI] [PubMed] [Google Scholar]
  • 147.Mataraza J.M., Gotwals P. Recent advances in immuno-oncology and the application to urological cancers. BJU Int. 2016;118:506–514. doi: 10.1111/bju.13518. [DOI] [PubMed] [Google Scholar]
  • 148.Zhang Z.T., Pak J., Shapiro E., Sun T.T., Wu X.R. Urothelium-specific expression of an oncogene in transgenic mice induced the formation of carcinoma in situ and invasive transitional cell carcinoma. Cancer Res. 1999;59:3512–3517. [PubMed] [Google Scholar]
  • 149.Grippo P.J., Sandgren E.P. Highly invasive transitional cell carcinoma of the bladder in a simian virus 40 T-antigen transgenic mouse model. Am J Pathol. 2000;157:805–813. doi: 10.1016/S0002-9440(10)64594-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Ayala de la Pena F., Kanasaki K., Kanasaki M., Tangirala N., Maeda G., Kalluri R. Loss of p53 and acquisition of angiogenic microRNA profile are insufficient to facilitate progression of bladder urothelial carcinoma in situ to invasive carcinoma. J Biol Chem. 2011;286:20778–20787. doi: 10.1074/jbc.M110.198069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Stone R., 2nd, Sabichi A.L., Gill J., Lee I.L., Adegboyega P., Dai M.S. Identification of genes correlated with early-stage bladder cancer progression. Cancer Prev Res (Phila) 2010;3:776–786. doi: 10.1158/1940-6207.CAPR-09-0189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Winslow M.M., Dayton T.L., Verhaak R.G., Kim-Kiselak C., Snyder E.L., Feldser D.M. Suppression of lung adenocarcinoma progression by Nkx2-1. Nature. 2011;473:101–104. doi: 10.1038/nature09881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Araki D., Takayama K., Inoue M., Watanabe T., Kumon H., Futaki S. Cell-penetrating D-isomer peptides of p53 C-terminus: long-term inhibitory effect on the growth of bladder cancer. Urology. 2010;75:813–819. doi: 10.1016/j.urology.2009.10.002. [DOI] [PubMed] [Google Scholar]
  • 154.van Bokhoven A., Varella-Garcia M., Korch C., Miller G.J. TSU-Pr1 and JCA-1 cells are derivatives of T24 bladder carcinoma cells and are not of prostatic origin. Cancer Res. 2001;61:6340–6344. [PubMed] [Google Scholar]

Articles from Asian Journal of Urology are provided here courtesy of Second Military Medical University

RESOURCES