You are on page 1of 6

10970215, 2006, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.21852 by Readcube (Labtiva Inc.), Wiley Online Library on [07/12/2022].

See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Int. J. Cancer: 119, 1513–1518 (2006)
' 2006 Wiley-Liss, Inc.

MINI REVIEW
Understanding urothelial carcinoma through cancer pathways
Wolfgang A. Schulz*
Department of Urology, Heinrich-Heine-University D€
usseldorf, Germany

Urothelial carcinoma (UC), the common histological subtype of This designation is now obsolete, as they are recognized to exhibit
bladder cancer, presents as a papillary tumor or as an invasive, of- considerable clinical and molecular differences.
ten lethal form. To study UC molecular biology, candidate gene
and genome-wide approaches have been followed. Here, it is argued Molecular biological research on bladder cancer is challenged
that a Ôcancer pathwayÕ perspective is useful to integrate findings to yield advances for its prevention, diagnostics and therapy. In
from both approaches. According to this view, papillary cancers fact, several ideas referred in this article have already influenced
typically exhibit activation of the MAPK pathway, as a conse- the latest WHO classification of urothelial cancers and thus con-
quence of oncogenic mutations in FGFR3 or HRAS, with increased tributed to practical improvements.1 In the future, most urgently
Cyclin D1 expression. In contrast, invasive UC are characterized by needed are (1) markers identifying those patients with papillary
severe disturbances in proximate cell cycle regulators, e.g. RB1 and and early stage invasive tumors at high risk for recurrence and
CDKN2A/p16INK4A, which decrease dependency on mitogenic sig- progression, (2) reliable and affordable non-invasive techniques
naling. In addition, these disturbances permit, promote and are in
turn exacerbated by chromosomal instability, which is further en- for monitoring, (3) markers distinguishing between metastatic and
hanced by loss of TP53 function. In another vicious cycle, defective localized invasive disease and (4) more efficacious therapies for
cell cycle regulation interacts with DNA methylation alterations. patients with locally progressed and metastatic cancer.
The transition toward invasive UC may require concomitant and
interacting defects in cell cycle regulation and the control of
genomic stability. Intriguingly, neither canonical WNT/b-Catenin Approaches to the molecular biology of UC
nor hedgehog signaling appear to play major roles in UC. This
may reflect its origin from more differentiated urothelial cells Two major approaches have been followed to analyze UC mo-
possessing a high regenerative potential rather than a stem cell lecular biology: candidate gene analysis and genome-wide screens.
population. In 1981, the first human oncogene, a mutated HRAS, was isolated
' 2006 Wiley-Liss, Inc. from UC cell lines.2 Since then, almost every newly emerging
oncogene or tumor suppressor was investigated for a role in UC.
Key words: cell cycle regulation; TP53; DNA methylation; Several tumor suppressors were indeed found to be frequently af-
chromosomal instability; cancer stem cells fected, including TP53, RB1 and CDKN2A. Several oncogenes were
observed to be activated in a fraction of cases, including HRAS, MYC
and CCND1.3,4 More recently, FGFR35–7 and E2F38,9 were identified
as oncogenes activated by point mutations and amplification, respec-
Bladder cancer is the fifth most frequent cancer in industrialized tively.
countries, accounting for up to 5% of all cancers. Histologically, Familial cases of bladder cancer are very rare. In the case of an
urothelial carcinoma (UC), formerly designated Ôtransitional cell exceptionally young index patient, an inherited translocation led
carcinoma,Õ is distinguished from rarer types like squamous cell to the identification of a novel oncogene, CDC91L1 at 20q.10 Its
carcinoma (SCC) and adenocarcinoma. UC are derived from the significance in sporadic cases remains to be ascertained.11 In gen-
urothelium, the epithelium lining the urinary tract from the renal eral, identification of genes important in bladder cancer must rely
pelvis to the urethra. Accordingly, some cases occur outside the on analyses of somatic alterations in tumor tissues and cell lines.
bladder in other segments of the urinary tract. Urothelial carcino- Identification of tumor suppressors is usually performed through
mas express markers of urothelial differentiation, including CK7 delineation of common regions of deletion followed by mutational
and CK13 cytokeratins, and even uroplakins, proteins of the apical and methylation analysis of candidate genes. Genome-wide
membrane of terminally differentiated umbrella cells in the top screens using LOH analyses or cytogenetical techniques revealed
urothelial layer. Clinically, the most important distinction is that several consistently altered chromosomal regions in bladder can-
between papillary and invasive UC. Papillary UC (pTa) are char- cer, but also a high background of chromosomal instability, with
acterized by hyperproliferation leading to lateral and vertical ex- essentially each chromosome affected in some cases.12,13 Some
tension of the urothelial layer. As a consequence, the epithelium recurrent chromosomal gains or losses reflect alterations in known
folds up around connective tissue papillae into a polypous struc- oncogenes or tumor suppressors. Losses at 9p21, 13q14 and 17p
ture protruding into the lumen of the urinary tract. Papillary UC reflect the established inactivation of CDKN2A, RB1 and TP53,
can be treated by local transurethral resection (TUR). Unfortu- respectively. Some targets of less common losses can also be
nately, the disease tends to recur in up to 75% of the patients, assigned, e.g., 10q losses may usually reflect PTEN inactivation.
necessitating long-term monitoring by cystoscopy and repeated Gains at 7p12, 8q24 and 11q13 can result in overexpression of
treatments. Recurrences can be partly prevented by optimized re- EGFR, MYC and Cyclin D1, respectively.3,4 On a note of caution,
section and by adjuvant intravesical instillation therapy with cyto- additional or alternative genes may be targeted by losses and am-
toxic drugs or with BCG, a mycobacterium vaccine preparation. plifications in some cases. Recurrent amplifications at 6p22.3 in
Thus, while papillary UC is seldom lethal, it causes considerable advanced stage cancers are even more difficult to interpret because
suffering and high expenses. Moreover, a small but significant per-
centage of papillary UC, usually characterized by poor differentia-
tion, progress to invasive stages. Invasive bladder cancers are Grant sponsor: Deutsche Forschungsgemeinschaft; Grant number: LI
mostly derived from another precursor, carcinoma in situ (pTIS), 1038/3-1.
flat highly dysplastic lesions. They are lethal by local progression *Correspondence to: Urologische Klinik, Heinrich-Heine-Universit€at,
or metastasis, unless completely removed by cystectomy at early Moorenstr. 5, 40225 D€usseldorf, Germany. Fax: 149-211-81-15846.
invasive stages. Chemo- and radiotherapy are efficacious, but E-mail: wolfgang.schulz@uni-duesseldorf.de
Received 15 September 2005; Accepted 23 December 2005
rarely curative. In the older literature, papillary tumors of various DOI 10.1002/ijc.21852
grades, carcinoma in situ, and carcinomas with invasion of the Published online 23 March 2006 in Wiley InterScience (www.interscience.
lamina propria only (pT1), were often summarized as Ôsuperficial.Õ wiley.com).

Publication of the International Union Against Cancer


10970215, 2006, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.21852 by Readcube (Labtiva Inc.), Wiley Online Library on [07/12/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1514 SCHULZ

they are highly heterogeneous. Their most likely target is E2F3, the variability may have biological causes. Rampant chromosomal
but several other genes in the region are over-expressed to a com- instability in UC ought to lead to variable dose changes and
parable or greater extent in individual cases.8,9,14,15 This example according differences in the expression of many genes, independ-
illustrates the limitations to identifying targets of chromosomal ently of whether they are causally involved in cancer development.
aberrations in cancers with pronounced genomic instability. Con- For instance, individual bladder cancer cell lines over-express dif-
sequently, the significance of many chromosomal gains in UC is ferent genes from the 6p22.3 amplification unit up to a 100 fold.15
uncertain. Moreover, gene expression profiling tends to identify those genes
This predicament extends to chromosome losses, even highly reacting most strongly to primary genetic or epigenetic alterations
prevalent ones at 3p and 8p. The most exasperating problem con- and not the causative change itself. Obviously, in a heterogeneous
cerns chromosome 9q. Loss of (or LOH at) chromosome 9 is the cancer like UC, data interpretation needs to be anchored in a con-
most frequent chromosomal alteration in bladder cancer through- ceptual framework to gain the most from genome-wide analyses.
out all stages and grades, including well-differentiated papillary
cancers and occasionally preneoplastic hyperplasia.3,4,12,13 On 9p, TP53 as a biomarker in UC
loss and recombination are centered around CDKN2A at 9p21.
This gene encodes p16INK4A, a CDK inhibitor and thereby activa- An important goal of array investigations of UC is the develop-
tor of RB1, and p14ARF, an indirect activator of TP53, in different ment of biomarkers for prognostic purposes. This aim has so also
reading frames. In UC, this general tumor suppressor is most fre- been pursued by investigations focused on individual genes, the
quently inactivated by homozygous deletion.16,17 Point mutations, most prominent one being TP53.32–34 The TP53 tumor suppressor
typically inactivating both reading frames, and hypermethylation gene harbors missense mutations in up to 50% of bladder cancers.
of either or both alternative promoters contribute in a smaller frac- The frequency of mutations increases with tumor stage and partic-
tion of the cases. While often the entire chromosome is affected, ularly with tumor grade. Specifically, their frequency is enhanced
allelic losses at 9q occur also independently of changes at 9p. One in early stage cancers considered as high-risk from clinical experi-
or several tumor suppressors are therefore assumed to be located ence, such as carcinoma in situ and poorly differentiated papillary
on 9q. Candidates comprise PTCH1 and TSC1 already implicated and early stage invasive tumors (pTaG3 and pT1G3). Moreover,
in other cancers, and the novel, cautiously named DBCCR1 LOH at 17p is preferentially found in advanced cases. Evidently,
(deleted in bladder cancer candidate region 1). Mutations inacti- TP53 mutations are associated with an increased risk of progres-
vating the second allele, meeting standard expectations for tumor sion in bladder cancer.3,4,32–34 The moot point is whether detection
suppressors, have been detected only in TSC1, at a low fre- of TP53 mutations can predict the natural course of the disease or
quency.18,19 Inherited TSC1 mutations predispose to several tumor responses to therapy better than histopathological parameters. This
types, but not obviously to bladder cancer. Methylation of DBCCR1 issue, too, is confounded by technical as well as biological factors.
increases in UC, but does not seem consistently associated with Many studies have used techniques incapable of detecting all
gene silencing.20 Nevertheless, DBCCR1 overexpression inhibits TP53 mutations, e.g. single-strand conformation polymorphism
cell proliferation.21 The mechanism involved and the effects of analysis plus sequencing starting from total tumor tissue. More-
physiological expression levels remain to be elucidated. over, accumulation of TP53 protein has often been used as a surro-
PTCH1 is a tumor suppressor in the (sonic) hedgehog pathway gate parameter for mutation. The correlation between mutations
and nuclear accumulation of TP53 is very good in bladder cancer,
and is often inactivated in basal cell carcinoma of the skin and in
but not perfect. In addition, TP53 immunohistochemistry poses its
medulloblastoma.22 These cancers are generally characterized by
own vagaries, although standardized techniques have meanwhile
activation of the hedgehog pathway, which can be alternatively
been developed.32–34
brought about by oncogenic mutations in SMO, a membrane sig-
naling protein normally inhibited by PTCH1. Activation of intra- These technical difficulties are exacerbated by the fact that
cellular hedgehog signaling by either alteration results in increased TP53 acts within a molecular network. Loss of TP53 function can
expression of several pathway components including the transcription therefore not only be caused by mutations and deletions of the
activators GLI1 and GLI2, and the inhibitory membrane proteins gene itself, but also by alterations in the mechanisms acting
HIP1 and PTCH1, through the increased activity of GLI-dependent ÔupstreamÕ or ÔdownstreamÕ of TP53 in the network. Such altera-
promoters. No evidence of such a response was found in UC cell tions may not have as severe effects as loss of TP53 itself, but
lines.23 In cancer tissues, PTCH1 expression is approximately halved may suffice to impede crucial functions of the nodal protein in the
in cases with 9q loss in accord with a pure dosage effect. PTCH1 network. ÔUpstreamÕ alterations in the network diminish TP53 acti-
mutations are extremely rare in UC and none have been reported in vation and include loss of p14ARF (encoded by CDKN2A) in a
other pathway components.24 Taken together, the evidence indicates large fraction of cases and amplification of HDM2 in a few.3,4,34
Defects in the function of the protein kinases ATM, CHK2 and
that PTCH1 hemizygosity is not pathogenetic in UC.
DNA-PK, which signal DNA damage to TP53, are implicated
Increasingly, global approaches are being used to investigate mo- too.35 Activation of these kinases in pTa and pT1 tumors may pre-
lecular changes in UC. These include array CGH and SNP arrays vent or delay their progression, but subsides in high-stage cancers
at the DNA level25,26 microarray expression profiling at the RNA in spite of increased chromosomal instability. ÔDownstreamÕ alter-
level,27–30 and proteomics approaches.31 Early studies addressed dif- ations diminish the efficacy of TP53 action through cell cycle
ferences among cell lines, cancer versus normal tissues and papillary inhibitors like p21CIP1 (see later) and pro-apoptotic proteins.
versus invasive cancers. More recently, more sophisticated issues
have been tackled, such as response to therapy or differential progno-
sis in cases with similar histopathological parameters, e.g. the clini- Arguments for a ‘‘cancer pathway’’ approach to UC
cally problematic pTaG3 tumors. Reassuringly, cluster analyses of The example of TP53, like that of PTCH1 and the hedgehog
expression profiles separate most cases of papillary from muscle- pathway discussed earlier, illustrates how productive it is to con-
invasive cancers and cancers from normal tissue, with interesting sider mutational and epigenetic gene alterations in the context of
exceptions that deserve further study. relevant networks or pathways. This approach may be particularly
Unfortunately, microarray expression studies have yielded few useful in a cancer with pronounced genomic instability such as
overlapping results with regard to the expression of individual UC. Hanahan and Weinberg36 have postulated that the distinctive
genes, which makes it difficult to draw any conclusions on molec- properties of cancers are brought about by the activation or inacti-
ular mechanisms in UC. Some of the inconsistencies may derive vation of a limited number of regulatory systems termed Ôcancer
from technical reasons and can presumably be resolved by im- pathways.Õ In addition to the TP53 network and the hedgehog
proved standardization of arrays and of sample preparation, e.g. pathway, these include the cell cycle regulatory system and the
through microdissection. Furthermore, a significant proportion of (canonical) MAPK pathway discussed later, the PI3K, STAT and
10970215, 2006, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.21852 by Readcube (Labtiva Inc.), Wiley Online Library on [07/12/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
UNDERSTANDING UROTHELIAL CARCINOMA THROUGH CANCER PATHWAYS 1515
NFjB pathways and the TGFb response not discussed here
because of space limitations, and the WNT/b-Catenin pathway,
which can serve as another example for the value of this approach.
Constitutive activation of this pathway is the ÔgatekeeperÕ
change in several cancers, notably colorectal carcinoma. Activa-
tion is caused by loss of function of the tumor suppressors APC or
AXIN1, or by oncogenic mutations in b-Catenin. Either results in
the increased activity of promoters regulated by TCF4/b-Cate-
nin.22 The few published studies on this pathway in UC report no
or very few genetic alterations in pathway components.37,38 In-
deed, promoter-reporter constructs monitoring TCF/b-Catenin de-
pendent transcription were found to be inactive in all UC lines
tested and in normal urothelial cells, indicating lack of pathway
activity.39 Furthermore, in most UC lines and in normal cells, tran-
scription could not even be induced by overexpressed oncogenic
b-Catenin, suggesting that it is actively repressed. Interestingly,
exceptional inducible cell lines lacked expression of E-Cadherin,
a modulator of the pathway.39 Thus, some bladder cancers may ac-
quire the ability to respond to strong WNT signaling through loss
of E-Cadherin, but constitutive activation is apparently very rare.
Importantly, many cancer pathways interact with each other and
the effects of further regulators, e.g. cell adhesion molecules and
extracellular proteases, are partly mediated through these pathways.
The level of regulatory pathways may thus be optimal to integrate
results from single candidate gene analyses and large-scale ge-
nomics approaches. Not least, the identification of relevant cancer
pathways yields immediate options for pharmacological therapy.

Cell cycle regulation as a ‘‘cancer pathway’’


Many cancer pathways converge towards cell cycle regulation,
a regulatory network in itself (Fig. 1). Since enhanced cell prolif-
eration is a property of human cancers by definition, altered cell
cycle regulation seems a prerequisite for cancer development.
However, cancer types differ in the mechanisms that lead to
altered cell cycle regulation and it its degree. In bladder cancer,
genetic and epigenetic alterations of proximate regulators of the
cell cycle abound.3,4 Deletion and mutational inactivation of RB1
is found in a significant fraction of invasive cancers.33,40 A large
fraction of cancers of all stages harbor defects in CDKN2A, with
loss of the CDK4 inhibitor p16INK4A.16,17 In exceptional cases
CDK4 is overexpressed due to amplification of the gene at 12q1341
or mutations interfere with p16INK4A binding. In contrast, overex-
pression of Cyclin D1 is widely found. Overexpression of MYC
could also be counted as a cell-cycle related alteration. Like overex-
pression of Cyclin D1, it is caused by chromosomal gains or gene
amplification only in a small number of tumors, but is more regu-
larly a consequence of deregulation.37,42 The fact that overexpres-
sion of MYC is caused by amplification in some cases and by dereg-
ulation in others could explain why overexpression is not entirely
consistently associated with tumor stage and grade. These altera-
tions are compounded by additional changes in the CIP/KIP family
of CDK inhibitors. The p21CIP1 and p27KIP1 inhibitory proteins are
down-regulated in the majority of invasive cancers18,33,43,44 and ex-
pression of p57KIP2 is diminished or ablated by a combination of FIGURE 1 – Signaling pathways converging on cell cycle regulation
genetic and epigenetic mechanisms.45 in normal urothelial cells (a), typical papillary superficial UC (b) and
While disturbances of cell cycle regulation are doubtless crucial typical invasive UC (c). Inactive pathways: white, properly regulated
active pathways: plain grey; deregulated pathways or proteins: patchy
in all UC, closer analyses reveal interesting variations. Alterations grey; aberrantly inactivated pathways or proteins: hatched.
of RB1 are almost exclusively observed in muscle-invasive can-
cers and are associated with a significantly worse prognosis.33,39
In contrast, Cyclin D1 overexpression is characteristic of early There is thus convincing evidence that ÔprimaryÕ alterations in
stage and papillary cancers and rather associated with less malig- cell cycle regulators such as inactivation of RB1 and p16INK4A,
nant behavior.3,4 CDKN2A alterations are overall independent of usually by genetic mechanisms, together with down-regulation of
stage, grade and clinical course.16,17 Obviously, the degree of dis- CIP/KIP CDK inhibitors, predominantly by epigenetic mecha-
turbance of cell cycle regulation parallels the biological and clini- nisms, are crucial changes in invasive UC. It is not quite as clear
cal aggressiveness of bladder cancers. In line with this supposi- whether this conclusion extends to papillary cancers. A large pro-
tion, losses of p21CIP1, p27KIP1, and p57KIP2 are more frequent in portion of well-differentiated papillary cancers harbor oncogenic
muscle-invasive tumors,18,33,43–45 although their independent prog- mutations in FGFR3 which encodes a receptor for fibroblast
nostic value is debated. In contrast, many papillary tumors over- growth factors like FGF1 and FGF8. As a rule, tumors exhibiting
express p21CIP1. this change show fewer recurrences and their proportion decreases
10970215, 2006, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.21852 by Readcube (Labtiva Inc.), Wiley Online Library on [07/12/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1516 SCHULZ
5–7,46
strongly at higher stages. FGFR3 mutations are inversely
associated with TP53 mutations, but positively with Cyclin D1
overexpression.46 Overactivity of FGFR3 would be expected to
lead to activation of the MAPK pathway through RAS and RAF
and hence to induction of Cyclin D1 and cell cycle activation.
Indeed, HRAS mutations were reported to occur in a complemen-
tary pattern to those in FGFR3 suggesting they belong to the same
pathway.47 Thus, the growth of typical papillary cancers could be
driven mainly by activation of the MAPK pathway, either through
FGFR3 mutations, HRAS mutations, or through activation of
other tyrosine kinase receptors, e.g. EGFR (Fig. 1).
The proliferation of normal urothelium, e.g. in response to tissue
damage, is likewise thought to be stimulated by growth factors act-
ing on tyrosine kinase receptors, in particular by EGF family mem-
bers acting in an autocrine or paracrine fashion.48 These mecha-
nisms can be studied in primary cultures of urothelial cells which
proliferate spontaneously when placed in appropriate serum-free
culture media. Their proliferation is elicited by autocrine growth
factors such as HB-EGF and further enhanced by addition of exoge-
neous EGF. Inhibition of MAPK signaling decreases proliferation
of normal urothelial cells, but cell lines from invasive UC are much FIGURE 2 – Mutual interaction of defective cell cycle regulation
less sensitive to MAP kinase inhibitors.49 Accordingly, while phos- with mitogenic signaling, chromosomal stability and maintenance of
phorylated ERK and MEK can be detected in some UC lines, their DNA methylation patterns in UC.
transcriptional response to MAPK activation is muted.49,50 These
findings suggest several interesting conclusions. Firstly, the more
massive alterations in cell cycle regulators found in invasive bladder bances. Chromosomal instability in invasive bladder cancers indi-
cancers apparently diminish the dependence on exogenous growth cates a dysfunction of these checkpoints. The prevalent loss of
factors and MAPK signaling, at least with regard to cell prolifera- TP53 function in these carcinomas certainly contributes to this
tion. Secondly, MAPK signaling is known to not only stimulate cell dysfunction. In cancers retaining intact TP53, down-regulation of
proliferation, but also cell differentiation and to induce inhibitors p21CIP1 or loss of RB1 should compromise its ability to arrest the
such as p21CIP1 which can induce cell cycle arrest and senescence. cell cycle upon check point activation. In fact, wild-type TP53
Therefore, activation of the pathway in papillary cancers could lead transfected into UC lines elicits apoptosis rather than cell cycle
to a self-limiting tumor growth. This would explain the generally arrest.54 Accordingly, recent evidence suggests that disturbances
lower malignancy of papillary cancers with FGFR3 mutations. Con- of checkpoint regulation are not due to TP53 loss only. The check-
tinuous tumor growth would require additional alterations that pre- point kinases ATM, CHK2 and CHK1 were observed in activated
vent terminal differentiation and replicative senescence induced states in papillary UC, but more weakly active in invasive can-
eventually by activation of G1 checkpoints through TP53 and RB1. cers.35 Apparently, dysregulation of DNA replication caused by
Alterations compromising these networks in UC may therefore also oncogenic FGFR3, RAS or MYC or by defects in the RB1 cell
prevent replicative senescence. Specifically, complete or even par- cycle regulatory network elicit activation of S-phase and G2/M
tial inactivation of CDKN2A also common in pTa cases17 may com- checkpoints. Overcoming these checkpoints through severe
plement activation of the MAPK pathway. Interestingly, mutant defects in cell cycle regulation or inactivation of checkpoint effec-
RAS proteins may more efficiently induce premature senescence tors is therefore a prerequisite for continued tumor growth and
through a different pathway dependent on p38MAPK.51 Tumors with progression. Indeed, checkpoint responses become increasingly
mutant HRAS could therefore behave differently from those with compromised as bladder cancers progress. While the underlying
FGFR mutations, in spite of similar effects on canonical MAPK mechanisms are incompletely understood, loss of RB1 in particu-
pathway activity. Thirdly and importantly, papillary and invasive lar has been linked to chromosomal instability.40 This relationship
UC may respond quite differently to therapeutic MAPK pathway in- may have several reasons. Firstly, checkpoint signaling may not
hibition, with additional differences within each group. elicit G1 arrest in cells lacking functional RB1. Secondly, defects
The notion that papillary UC are typically characterized by in replication and mitosis may occur more frequently during shorter
MAPK activation, and invasive UC by defects in the TP53 and and less coordinated cell cycles. Thirdly, RB1 is implied directly in
RB1 networks is corroborated by animal models. Transgenic mice the control of mitosis.40 In summary, therefore, the chromosomal
that overexpress a mutated Ha-Ras in urothelial cells develop uro- instability characteristic of invasive bladder cancers may partly rep-
thelial hyperplasia and papillary UC,52 whereas animals express- resent a consequence of severe disturbances of cell cycle regulation.
ing the SV40 large-T antigen, which inactivates Rb1 and Tp53, Conversely, chromosomal instability causes genetic alterations that
present with carcinoma in situ or invasive cancers.53 exacerbate cell cycle deregulation (Fig. 2).
Another vicious cycle in UC links cell cycle deregulation to
alterations of DNA methylation, which promote tumor growth and
Consequences of cell cycle disturbances in UC genomic instability (Fig. 2). As in other cancers, 2 kinds of meth-
ylation alterations are observed, hypermethylation at CpG-rich
The almost universal disturbance of the cell cycle regulatory promoter regions of selected genes and a genome-wide decrease
network in bladder cancers has consequences beyond increased in overall methylation, which is most evident at retrotransposons
cell proliferation (Fig. 2). Hyperproliferative signals elicited by and satellites sequences densely methylated in normal cells. Hyper-
activated oncogenes induce cellular senescence through CDKN2A. methylation events are more frequent in invasive than in papillary
Inactivation of this locus and down-regulation of other CDK in- UC,55 while hypomethylation is essentially universal in invasive
hibitors that can mediate cellular senescence, p21CIP1 and p57KIP2, cancers.56 The mechanisms leading to these changes are complex
would thus allow bladder cancers to tolerate persistently high pro- and far from understood, but clearly proper coordination between
liferation signals, emerging from mutated FGFR3, mutated RAS, the activity of DNA methyltransferases and DNA replication is cru-
over-expressed EGFR, or amplified MYC. cial for the maintenance of methylation patterns in normal cells.
Proper cell cycle control is also required for the function of cel- Specifically, the major enzyme in somatic cells, DNMT1, is coordi-
lular checkpoints responding to DNA damage and mitotic distur- nated with the cell cycle by RB1 via E2F transcription factors. Loss
10970215, 2006, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.21852 by Readcube (Labtiva Inc.), Wiley Online Library on [07/12/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
UNDERSTANDING UROTHELIAL CARCINOMA THROUGH CANCER PATHWAYS 1517
of proper RB1 function would be expected to disturb this coordi- tance of these pathways might relate to the urothelial origin of
nation. Indeed, DNMT1 expression was found to become un- UC. Urothelium is a quiescent tissue, but capable of rapid prolifer-
coupled from increased cell proliferation in UC.57 Thus, while ation in response to injury. The proliferating cells come from the
DNMT1 expression even increases in some UC, the enzyme can- basal and intermediate layers of the tissue. During acute injury,
not maintain normal methylation patterns. In turn, methylation stem cell recruitment does not seem to be required. Thus, the func-
alterations affect cell cycle regulators. The CDK inhibitor p57KIP2, tions of WNT and hedgehog signaling in maintaining stem cell
e.g., is down-regulated during UC progression by promoter hyper- compartments may not be as important in normal urothelial tissue
methylation as well as hypomethylation of a more distant region as in continuously proliferating tissues like the skin or the gut epi-
controlling its imprinting.45 thelium. Consequentially, UC may arise from more differentiated
cells, in which these pathways are suppressed. Indeed, UC usually
express proteins characteristic of differentiated urothelial cells.
Conclusion and perspective According to this view, the property of unlimited growth, which
In conclusion, 2 major ‘‘cancer pathways,’’ cell cycle regulation cancer cells share with stem cells, would be acquired secondarily
and the TP53 network, appear to be affected in UC and the extent in UC, most likely by inactivation of TP53 and RB1 function.
of their respective deregulation in each tumor significantly deter- Obviously, to even better understand bladder cancer, the cancer
mines its biological and clinical characteristics (Fig. 1). Activation pathway perspective sketched here should be widened to encom-
of the canonical MAPK pathway may characterize an important pass the context of urothelial tissue organization.
subclass of papillary tumors.1,4,58 If so, this subclass may be more
responsive to appropriate drug treatment than UC in general. Intri-
guingly, neither hedgehog nor WNT/b-Catenin signaling appear Acknowledgements
to play major parts in UC, in spite of their fundamental importance
in other carcinomas. Apparently, stimulation of cell proliferation The author thanks Andrea Linnemann-Florl, Hans-Helge Seifert,
in UC is accomplished by other alterations, notably severe defects Sascha Raschke and Jiri Hatina for critical reading of the
in proximate cell cycle regulation. In addition, the lack of impor- manuscript.

References
1. Eple JN, Sauter G, Epstein JI, Sesterhenn IA, eds. Pathology and 16. Florl AR, Franke KH, Niederacher D, Gerharz CD, Seifert HH,
genetics: tumours of the urinary system and male genital organs. Schulz WA. DNA methylation and the mechanisms of CDKN2A inac-
Lyon: IARC Press, 2004. 359 p. tivation in transitional cell carcinoma of the urinary bladder. Lab
2. Parada LF, Tabin CJ, Shih C, Weinberg RA. Human EJ bladder carci- Invest 2000;80:1513–22.
noma oncogene is homologue of Harvey sarcoma virus ras gene. 17. Chapman EJ, Harnden P, Chambers P, Johnston C, Knowles MA.
Nature 1982;297:474–8. Comprehensive analysis of CDKN2A status in microdissected urothelial
3. Knowles MA. What we could do now: molecular pathology of blad- cell carcinoma reveals potential haploinsufficiency, a high frequency of
der cancer. Mol Pathol 2001;54:215–21. homozygous co-deletion and associations with clinical phenotype. Clin
4. Dinney CP, McConkey DJ, Millikan RE, Wu X, Bar-Eli M, Adam L, Cancer Res 2005;11:5740–7.
Kamat AM, Siefker-Radtke AO, Tuziak T, Sabichi AL, Grossman HB, 18. Adachi H, Igawa M, Shiina H, Urakami S, Shigeno K, Hino O.
Benedict WF. Focus on bladder cancer. Cancer Cell 2004;6:111–6. Human bladder tumors with 2-hit mutations of tumor suppressor gene
5. Cappellen D, De Oliveira C, Ricol D, de Medina S, Bourdin J, Sastre- TSC1 and decreased expression of p27. J Urol 2003;170:601–4.
Garau X, Chopin D, Thiery JP, Radvanyi F. Frequent activating muta- 19. Knowles MA, Habuchi T, Kennedy W, Cuthbert-Heavens D. Muta-
tions of FGFR3 in human bladder and cervix carcinomas. Nat Genet tion spectrum of the 9q34 tuberous sclerosis gene TSC1 in transitional
1999;23:18–20. cell carcinoma of the bladder. Cancer Res 2003;63:7652–6.
6. Sibley K, Cuthbert-Heavens D, Knowles MA. Loss of heterozygosity 20. Habuchi T, Takahashi T, Kakinuma H, Wang L, Tsuchiya N, Satoh S,
at 4p16.3 and mutation of FGFR3 in transitional cell carcinoma. Akao T, Sato K, Ogawa O, Knowles MA, Kato T. Hypermethylation
Oncogene 2001;20:686–91. at 9q32-33 tumour suppressor region is age-related in normal urothe-
7. van Rhijn BW, Lurkin I, Radvanyi F, Kirkels WJ, van der Kwast TH, lium and an early and frequent alteration in bladder cancer. Oncogene
Zwarthoff EC. The fibroblast growth factor receptor 3 (FGFR3) muta- 2001;20:531–7.
tion is a strong indicator of superficial bladder cancer with low recur- 21. Wright KO, Messing EM, Reeder JE. DBCCR1 mediates death in cul-
rence rate. Cancer Res 2001;61:1265–8. tured bladder tumor cells. Oncogene 2004;23:82–90.
8. Feber A, Clark J, Goodwin G, Dodson AR, Smith PH, Fletcher A, 22. Beachy PA, Karhadkar SS, Berman DM. Tissue repair and stem cell
Edwards S, Flohr P, Falconer A, Roe T, Kovacs G, Dennis N. renewal in carcinogenesis. Nature 2004;432:324–31.
Amplification and overexpression of E2F3 in human bladder cancer. 23. Thievessen I, Wolter M, Prior A, Seifert HH, Schulz WA. Hedgehog
Oncogene 2004;23:1627–30. signaling in normal urothelial cells and in urothelial carcinoma cell
9. Oeggerli M, Tomovska S, Schraml P, Calvano-Forte D, Schafroth S, lines. J Cell Physiol 2005;203:372–7.
Simon R, Gasser T, Mihatsch MJ, Sauter G. E2F3 amplification and 24. Aboulkassim TO, LaRue H, Lemieux P, Rousseau F, Fradet Y. Alter-
overexpression is associated with invasive tumor growth and rapid ation of the PATCHED locus in superficial bladder cancer. Oncogene
tumor cell proliferation in urinary bladder cancer. Oncogene 2004;23: 2003;22:2967–71.
5616–23. 25. Veltman JA, Fridlyand J, Pejavar S, Olshen AB, Korkola JE, DeVries S,
10. Guo Z, Linn JF, Wu G, Anzick SL, Eisenberger CF, Halachmi S, Carroll P, Kuo WL, Pinkel D, Albertson D, Cordon-Cardo C, Jian AN.
Cohen Y, Fomenkov A, Hoque MO, Okami K, Steiner G, Engles JM, Array-based comparative genomic hybridization for genome-wide
et al. CDC91L1 is a newly discovered oncogene in human bladder screening of DNA copy number in bladder tumors. Cancer Res 2003;63:
cancer. Nat Med 2004;10:374–81. 2872–80.
11. Schultz IJ, Kiemeney LA, Witjes JA, Schalken JA, Willems JL, Swin- 26. Primdahl H, Wikman FP, von der Maase H, Zhou XG, Wolf H,
kels DW, de Kok JB. CDC91L1 (PIG-U) mRNA expression in urothe- Orntoft TF. Allelic imbalances in human bladder cancer: genome-wide
lial cell carcinomas. Int J Cancer 2005;116:282–4. detection with high-density single-nucleotide polymorphism arrays.
12. Knowles MA, Elder PA, Williamson M, Cairns JP, Shaw ME, J Natl Cancer Inst 2002;94:216–23.
Law MG. Allelotype of human bladder cancer. Cancer Res 1994;54: 27. Dyrskjot L, Thykjaer T, Kruhoffer M, Jensen JL, Marcussen N, Ham-
531–538. ilton-Dutoit S, Wolf H, Orntoft TF. Identifying distinct classes of
13. Hovey RM, Chu L, Balazs M, DeVries S, Moore D, Sauter G, Carroll PR, bladder carcinoma using microarrays. Nat Genet 2003;33:90–6.
Waldman FM. Genetic alterations in primary bladder cancers and 28. Modlich O, Prisack HB, Pitschke G, Ramp U, Ackermann R, Bojar H,
their metastases. Cancer Res 1998;58:3555–60. V€ogeli TA, Grimm MO. Identifying superficial, muscle-invasive, and
14. Bruch J, Schulz WA, Melzner I, Kemmerling R, Br€uderlein S, M€oller P, metastasizing transitional cell carcinoma of the bladder: use of cDNA
Vogel W, Hameister H. Concurrent gain of chromosomes 5p, 6p, and array analysis of gene expression profiles. Clin Cancer Res 2004;10:
20q in bladder carcinoma cell lines: delineation of the 6p22 amplifica- 3410–21.
tion unit. Cancer Res 2000;60:4526–30. 29. Blaveri E, Simko J, Korkola JE, Brewer JL, Baehner F, Mehta K,
15. Wu Q, Hoffmann MJ, Hartmann FH, Schulz WA. Amplification and deVries S, Koppie T, Pejavar S, Carroll P, Waldman FM. Bladder
overexpression of the ID4 gene at 6p22.3 in bladder cancer. BMC cancer outcome and subtype classification by gene expression. Clin
Mol Cancer 2005;4:16. Cancer Res 2005;11:4044–55.
10970215, 2006, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.21852 by Readcube (Labtiva Inc.), Wiley Online Library on [07/12/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1518 SCHULZ

30. Wild PJ, Herr A, Wissmann C, Stoehr R, Rosenthal A, Zaak D, Simon R, 45. Hoffmann MJ, Florl AR, Seifert HH, Schulz WA. Multiple mecha-
Knuechel R, Pilarsky C, Hartmann A. Gene expression profiling of nisms inactivating CDKN1C in bladder cancer. Int J Cancer 2005;
progressive papillary noninvasive carcinomas of the urinary bladder. 114:406–13.
Clin Cancer Res 2005;11:4415–29. 46. Bakkar AA, Wallerand H, Radvanyi F, Lahaye JB, Pissard S, Lecerf L,
31. Celis JE, Gromova I, Moreira JM, Cabezon T, Gromov P. Impact of pro- Kouyoumdjian JC, Abbou CC, Pairon JC, Jaurand MC, Thiery JP,
teomics on bladder cancer research. Pharmacogenomics 2004;5:381–94. Chopin OK. FGFR3 and TP53 gene mutations define two distinct path-
32. Helpap B, Schmitz-Drager BJ, Hamilton PW, Muzzonigro G, Galosi AB, ways in urothelial cell carcinoma of the bladder. Cancer Res 2003;63:
Kurth KH, Lubaroff D, Waters DJ, Droller MJ. Molecular pathology of 8108–12.
non-invasive urothelial carcinomas. Virchows Arch 2003;442:309–16. 47. Jebar AH, Hurst CD, Tomlinson DC, Johnston C, Taylor CF, Knowles MA.
33. Chatterjee SJ, Datar R, Youssefzadeh D, George B, Goebell PJ, Stein JP, FGFR3 and Ras gene mutations are mutually exclusive genetic events
Young L, Shi SR, Gee C, Groshen S, Skinner DG, Cote RJ. Combined in urothelial cell carcinoma. Oncogene 2005;24:5218–25.
effects of p53, p21, and pRb expression in the progression of bladder 48. Varley C, Hill G, Pellegrin S, Shaw NJ, Selby PJ, Trejdosiewicz LK,
transitional cell carcinoma. J Clin Oncol 2004;22:1007–13. Southgate J. Autocrine regulation of human urothelial proliferation
34. Malats N, Bustos A, Nascimento CM, Fernandez F, Rivas M, Puente D, and migration during regenerative responses in vitro. Exp Cell Res
Kogevinas M, Real FX. P53 as a prognostic marker for bladder cancer: 2005;306:216–29.
a metaanalysis and review. Lancet Oncol 2005;6:678–686. 49. Swiatkowski S, Seifert HH, Steinhoff C, Prior A, Thievessen I, Schli-
35. Bartkova J, Horejsi Z, Koed K, Kr€amer A, Tort F, Zieger K, Guldberg P, ess F, Schulz WA. Activities of MAP-kinase pathways in normal
Sehested M, Nesland JM, Lukas C, Orntoft T, Lukas J. DNA damage uroepithelial cells and urothelial carcinoma cell lines. Exp Cell Res
response as a candidate anti-cancer barrier in early human tumorigenesis. 2003;282:48–57.
Nature 2005;434:864–70. 50. Hoshino R, Chatani Y, Yamori T, Tsuruo T, Oka H, Yoshida O,
36. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57–70. Shimada Y, Ari-i S, Wada H, Fujimoto J, Kohno M. Constitutive acti-
37. Shiina H, Igawa M, Shigeno K, Terashima M, Deguchi M, Yamanaka vation of the 41-/43-kDa mitogen-activated protein kinase signaling
M, Ribeiro-Filho L, Kane CJ, Dahiya R. Beta-catenin mutations cor- pathway in human tumors. Oncogene 1999;18:813–22.
relate with over expression of C-myc and cyclin D1 genes in bladder 51. Deng Q, Liao R, Wu BL, Sun P. High intensity RAS signaling induces
cancer. J Urol 2002;168:2220–6. premature senescence by activating p38 pathway in primary human
38. Stoehr R, Krieg RC, Knuechel R, Hofstaedter R, Pilarsky C, Zaak D, fibroblasts. J Biol Chem 2004;279:1050–9.
Schmitt R, Hartmann A. No evidence for involvement of beta-catenin 52. Zhang ZT, Pak J, Huang HY, Shapiro E, Sun TT, Pellicer A, Wu XR.
and APC in urothelial carcinomas. Int J Oncol 2002;20:905–11. Role of Ha-ras activation in superficial papillary pathway of urothelial
39. Thievessen I, Seifert HH, Swiatkowski S, Florl AR, Schulz WA. tumor formation. Oncogene 2001;20:1973–80.
E-cadherin involved in inactivation of WNT/b-catenin signalling in 53. Zhang ZT, Pak J, Shapiro E, Sun TT, Wu XR. Urothelium-specific
urothelial carcinoma and normal urothelial cells. Br J Cancer 2003; expression of an oncogene in transgenic mice induced the formation
88:1932–8. of carcinoma in situ and invasive transitional cell carcinoma. Cancer
40. Hernando E, Nahle Z, Juan G, Diaz-Rodriguez E, Alaminos M, Res 1999;59:3512–7.
Hemann M, Michel L, Mittal V, Gerald W, Benezra R, Lowe SW, 54. Makri D, Schulz WA, Grimm MO, Clasen S, Bojar H, Schmitz-
Cordon-Cardo C. Rb inactivation promotes genomic instability by Dr€ager BJ. WAF1/p21 regulates proliferation but does not mediate
uncoupling cell cycle progression from mitotic control. Nature 2004; p53-dependent apoptosis in urothelial carcinoma cell lines. Int J Oncol
430:797–802. 1998;12:621–8.
41. Simon R, Struckmann K, Schraml P, Wagner U, Forster T, Moch H, 55. Catto JW, Azzouzi AR, Rehman I, Feeley KM, Cross SS, Amira N,
Fijan A, Bruderer J, Wilber K, Mihatsch MJ, Gasser T, Sauter G. Fromont G, Sibony M, Cussenot O, Meuth M, Hamdy FC. Promoter
Amplification pattern of 12q13-q15 genes (MDM2, CDK4, GLI) in hypermethylation is associated with tumor location, stage, and subse-
urinary bladder cancer. Oncogene 2002;21:2476–83. quent progression in transitional cell carcinoma. J Clin Oncol 2005;23:
42. Christoph F, Schmidt B, Schmitz-Dr€ager BJ, Schulz WA. Overexpression 2903–10.
and amplification of the c-myc gene in human urothelial carcinoma. Int 56. Florl AR, Loewer R, Schmitz-Dr€ager BJ, Schulz WA. DNA meth-
J Cancer 1999;84:169–73. ylation and expression of L1 LINE and HERV-K provirus sequen-
43. Clasen S, Schulz WA, Gerharz CD, Grimm MO, Christoph F, ces in urothelial and renal cell carcinoma. Br J Cancer 1999;80:
Schmitz-Dr€ager BJ. Frequent and heterogenous expression of cyclin- 1312–21.
dependent kinase inhibitor WAF1/p21 protein and mRNA in urothe- 57. Kimura F, Seifert HH, Florl AR, Santourlidis S, Steinhoff C, Swiat-
lial carcinoma. Br J Cancer 1998;77:515–21. kowski S, Mahotka C, Gerharz CD, Schulz WA. Decreased DNA
44. Franke KH, Miklosi M, Goebell P, Clasen S, Steinhoff C, Anastasiadis AG, methyltransferase 1 expression relative to cell proliferation in transi-
Gerharz CD, Schulz WA. Cyclin-dependent kinase inhibitor p27KIP1 is tional cell carcinoma. Int J Cancer 2003;104:568–78.
expressed preferentially in early stages of urothelial carcinoma. Urology 58. Wu XR. Urothelial tumorigenesis: a tale of divergent pathways. Nat
2000;56:689–95. Rev Cancer 2005;5:713–25.

You might also like