SlideShare uma empresa Scribd logo
1 de 14
Current perspectives in bladder cancer management
T. R. L. Griffiths, on behalf of Action on Bladder Cancer
Methods
The rationale for this review of the literature was to
discuss the latest evidence-base and to summarise
key messages for the prevention, investigation, diag-
nosis and management of bladder cancer for non-
specialists and specialists.
The literature searches were conducted using the
timeframe of February 2012 to June 2012. The data-
bases searched included PubMed and Trip, congress
abstracts [European Association of Urology (EAU)]
and current guidelines ⁄ consensus statements from
within Europe [EAU; National Institute for Health
and Clinical Excellence (NICE); The Renal Associa-
tion; British Association of Urological Surgeons
(BAUS)].
Key words used for the searches were: bladder can-
cer; risk factors; epidemiology; histopathology; diag-
nosis; haematuria; intravesical chemotherapy; BCG;
transitional cell carcinoma; cystoscopy; transurethral
resection; cystectomy; radiotherapy; systemic chemo-
therapy.
Introduction
Table 1 summarises the key points concerning the
diagnosis and management of bladder cancer for the
non-specialist healthcare professional.
Approximately 386,000 new bladder tumours were
diagnosed worldwide in 2008, making bladder cancer
the ninth most common cancer (1). In the UK, blad-
der cancer is the fourth and twelfth most common
cancer in men and women respectively (2); it is up to
three times more common in men than women, but
women tend to present with more advanced disease
and have worse outcomes (3). Around eight in 10
cases are diagnosed in individuals over the age of 65
(2), and Caucasians generally have a higher risk of
bladder cancer than people of other ethnicities (4). In
the USA, bladder cancer mortality in men decreased
from the mid-1970s to the mid-1980s and was rela-
tively stable to 2006; in women, there was a small, but
steady decrease between 1975 and 2006 (4). Within
Europe, bladder cancer mortality was stable in women
between 1980 and 2006, but consistently decreased in
SUMMARY
More than 350,000 new cases of bladder cancer are diagnosed worldwide each
year; the vast majority (> 90%) of these are transitional cell carcinomas (TCC).
The most important risk factors for the development of bladder cancer are smoking
and occupational exposure to toxic chemicals. Painless visible haematuria is the
most common presenting symptom of bladder cancer; significant haematuria
requires referral to a specialist urology service. Cystoscopy and urine cytology are
currently the recommended tools for diagnosis of bladder cancer. Excluding muscle
invasion is an important diagnostic step, as outcomes for patients with muscle
invasive TCC are less favourable. For non-muscle invasive bladder cancer, transure-
thral resection followed by intravesical chemotherapy (typically Mitomycin C or epi-
rubicin) or immunotherapy [bacillus Calmette-Gue´rin (BCG)] is the current standard
of care. For patients failing BCG therapy, cystectomy is recommended; for patients
unsuitable for surgery, the choice of treatment options is currently limited. How-
ever, novel interventions, such as chemohyperthermia and electromotive drug
administration, enhance the effects of conventional chemotherapeutic agents and
are being evaluated in Phase III trials. Radical cystectomy (with pelvic lymphaden-
ectomy and urinary diversion) or radical radiotherapy are the current established
treatments for muscle invasive TCC. Neoadjuvant chemotherapy is recommended
before definitive treatment of muscle invasive TCC; cisplatin-containing combina-
tion chemotherapy is the recommended regimen. Palliative chemotherapy is the
first-choice treatment in metastatic TCC.
Review criteria
To produce this non-systematic review, an extensive
literature search was performed with the objective
of identifying publications concerning the
management of bladder cancer. Multiple sources
were included, e.g. current European guidelines,
National Institute for Health and Clinical Excellence
guidelines, meta-analyses, randomised controlled
trials (RCT), consensus statements by the Renal
Association and British Association of Urological
Surgeons and non-randomised study evidence
where RCT evidence was not available.
Message for the clinic
Patients with a single episode of visible haematuria
(VH) and any patients with clinically relevant non-
visible haematuria (NVH) (defined in Table 1) need
prompt investigation and urological referral.
Symptoms attributable to bladder cancer can mimic
urinary tract infection (UTI). Patients with a
symptomatic UTI refractory to antibiotics or an
early recurrent UTI warrant flexible cystoscopic
evaluation before considering repeat courses of
antibiotics.
University Hospitals of Leicester
NHS Trust, Clinical Sciences
Unit, Leicester General Hospital,
Leicester, UK
Correspondence to:
T. R. Leyshon Griffiths,
University Hospitals of Leicester
NHS Trust, Clinical Sciences
Unit, Leicester General Hospital,
Gwendolen Road, Leicester LE5
4PW, UK
Tel.: +44 (0)1162 584617
Fax: +44 (0)1162 584617
Email: trlg1@le.ac.uk
Disclosures
Over the past year, I have
received an honorarium from
Alliance Pharma for advice on
creating a health-economic
model for intravesical BCG.
Over the past 5 years, I have
received honoraria from
Alliance Pharma and GE
Healthcare for involvement in
educational events including
provision of lectures. GE
Healthcare used to market
hexvixÒ
for use in
photodynamic diagnosis of
bladder cancer in the UK, but
this is no longer the case.
Charity
Registration Number:
1138532
REVIEW ARTICLE
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448. doi: 10.1111/ijcp.12075 435
Table 1 Key points for the diagnosis and management of bladder cancer
Pathology The vast majority of bladder cancers (> 90%) are TCC in origin
Risk factors Smoking and occupational exposure to toxic chemicals are the key risk factors for bladder cancer
Smoking cessation is still worthwhile following diagnosis of bladder cancer
Definitions of significant haematuria Painless VH is the most common presenting symptom of bladder cancer
NVH: ‡ 1 + blood on dipstick testing; not necessary to confirm NVH using urine microscopy; a trace
of blood only on dipstick is not regarded as significant.
Persistent a-NVH: Defined in the UK as two out of three dipsticks ‡ 1 + blood. In contrast,
the American Urological Association (AUA) defines a-NVH as ‡ 3 red blood cells per high
powered field in a properly collected urine specimen.
Clinically relevant haematuria: any episode of painless VH; any single episode of painful VH or s-NVH
in the absence of UTI; recurrent or refractory UTI with VH, or persistent a-NVH (requires urgent investigation)
Transient NVH or spurious causes of haematuria need to be excluded
Investigations within primary care All patients with s-NVH and persistent a-NVH should have their baseline blood pressure, eGFR and ACR measured
Further nephrological assessment should be considered in those aged < 40 years with persistent a-NVH if
any one of the following are present: eGFR < 60 ml ⁄ min, ACR > 30 or blood pressure > 140 ⁄ 90 mmHg
Referral to specialist urology service Routine referral: age 40–50 years with a-NVH or aged < 50 years with s-NVH
Urgent referral: age > 50 years with a-NVH or s-NVH, or age > 40 years with recurrent or persistent
UTI associated with haematuria
Tools to facilitate the diagnosis
of bladder cancer
Cystoscopy and urine cytology are currently recommended tools
Fluorescence cystoscopy (photodynamic diagnosis) is most useful for detection of CIS and guiding
biopsies in patients with positive cytology or a history of high-grade NMIBC
Narrow-band imaging cystoscopy has shown promise as an aid to facilitate detection without the need for
an intravesical photosensitiser, but needs further evaluation in randomised trials
Although currently available novel urinary biomarkers have higher sensitivity than cytology, they are not
routinely recommended because of their higher false-positive rates
Non-muscle invasive TCC Excluding muscle invasion is an important diagnostic step; one-half of patients with muscle invasive
TCC will die of bladder cancer within 5 years
EORTC risk group should be determined
Most patients with NMIBC undergo surveillance cystoscopies and intravesical treatments
Quality of first TUR can affect outcomes (tumour-free detrusor muscle ⁄ residual disease)
A single postoperative intravesical instillation of chemotherapy within 24 h of TUR is recommended for
all newly diagnosed bladder tumours
The choice between further intravesical chemotherapy or immunotherapy is guided by the EORTC risk group
Immediate radical cystectomy should be considered for patients whose risk of tumour progression is
especially high, concomitant superficial urethral TCC is present, or where they
request it in preference to intravesical BCG
Treatment with intravesical BCG is considered to have failed if: T1 disease persists at the
3-month check cystoscopy following induction BCG; high-grade Ta disease ⁄ CIS is present at
6 months; or muscle invasive TCC is detected
The incidence of concomitant UUT-TCC is low, but is increased in patients with trigonal tumours
Treatment after BCG failure Radical cystectomy remains the mainstay of treatment for patients who have failed BCG treatment
For patients who are unwilling or unfit to have a radical cystectomy after BCG failure, the
treatment options are limited
Currently available bladder-sparing treatments for those with BCG-refractory TCC are associated
with 2-year disease-free survival of approximately 50%
Treatments with the most promising clinical data, such as chemohyperthermia, are now being evaluated in Phase III trials
Muscle invasive TCC Radical cystectomy (with pelvic lymphadenectomy and urinary diversion) or radical radiotherapy are
the current established treatments for muscle invasive TCC
Neoadjuvant chemotherapy is recommended before definitive treatment of muscle invasive TCC;
cisplatin-containing combination chemotherapy is the recommended regimen
Chemotherapy concurrent with radiotherapy improves loco-regional control compared with
radiotherapy alone and confers separate benefits to neoadjuvant chemotherapy
Advanced ⁄ metastatic TCC Palliative chemotherapy is the first-choice treatment in metastatic TCC
ACR, albumin ⁄ creatinine ratio; BCG, bacillus Calmette-Gue´rin; CIS, carcinoma in situ; eGFR, estimated glomerular filtration rate; EORTC, European Organization for
Research and Treatment of Cancer; NMIBC, non-muscle invasive bladder cancer; NVH, non-visible haematuria; TCC, transitional cell carcinoma; TUR, transurethral
resection; UTI, urinary tract infection; UUT, upper urinary tract; VH, visible haematuria.
436 Bladder cancer management
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
men from around 1990 onwards (5). The bladder can-
cer mortality rate in Japan has been stable for men
since the early 1990s and has risen slightly during this
period in women (6). Putative reasons for reduced
mortality include reduced exposure to tobacco and
occupational carcinogens.
Transitional cell carcinoma (TCC; also known as
urothelial cancer) represents over 90% of bladder
cancers (7) and is consequently the focus of this
review. Less common types include squamous cell
carcinoma, adenocarcinoma and small cell carci-
noma. Around 70–85% of TCCs are superficial
[stages Ta, T1, carcinoma in situ (CIS)] at presenta-
tion and are now commonly termed non-muscle
invasive bladder cancer (NMIBC) (7–10). Unlike in
Europe or the USA, malignant non-muscle invasive
tumours that also do not invade the lamina propria
(Ta, CIS) are omitted from cancer registry statistics
in England and Wales, therefore interpretation of
epidemiological statistics varies across the world.
However, in the management of bladder cancer,
excluding muscle invasion is an important diagnostic
step, as outcomes for patients with muscle invasive
TCC (T2–T4) are less favourable; nearly one-half will
die from bladder cancer within 5 years of diagnosis
(8,10). In the Middle East and Africa, squamous cell
carcinoma was typically more common than in the
developed world, largely caused by Schistosoma hae-
matobium infection. However, improved knowledge
of schistosomiasis over the last three decades has led
to a reduction in squamous cell carcinoma such that
TCC is now also the predominant type of bladder
cancer in these countries (11,12).
Risk factors for bladder cancer
Numerous risk factors for TCC have been identified.
Smoking is implicated in approximately two-thirds
of bladder cancers in men and up to one-third in
women (13). There is a fourfold increased incidence
in current smokers relative to never-smokers (14),
and bladder cancer risk is correlated with the num-
ber of cigarettes smoked, duration of smoking and
age at smoking initiation (14–16). Those who stop
smoking reduce their bladder cancer risk by 10–40%
within 4 years (14,17), although former smokers
retain a twofold higher incidence than never-smokers
(14,16).
Data from Western Europe suggest that around 4–
7% of bladder cancers in men are attributable to a
known occupational carcinogen; the latent period
between exposure and the development of cancer is
about 20 years (18). Exposure to aniline dyes, aro-
matic amines (used in the manufacture of textiles,
paints, plastics and rubber industries) and polycyclic
aromatic amines are the primary toxins. Continuous
arsenic exposure and ingestion has been reported to
increase the risk of bladder cancer by as much as
one thousand times (19).
Carcinogen-metabolising enzymes are in part con-
trolled by genetic polymorphism. Slow acetylation of
N-acetyltransferase-2 (20,21), rapid cytochrome
P1A2 activity (22) and glutathione S-transferase M1
null genotype (21) are associated with an increased
risk of TCC. Approximately 20% of Europeans
affected by bladder cancer are homozygous for a
non-coding single nucleotide polymorphism
(8q24.21) located close to the c-Myc oncogene (23).
Older studies have shown that pelvic radiotherapy
for cervical cancer was associated with a twofold to
fourfold increased risk of secondary bladder cancer
(24,25). However, with contemporary radiotherapy,
this risk has been reduced to a minimal level (26). In
men with prostate cancer who received external
beam radiotherapy and ⁄ or brachytherapy between
1973 and 1999, at 5 years, after the initial diagnosis
of the primary cancer, 1–1.5% had developed a sec-
ondary bladder malignancy (27). However, it is esti-
mated that with modern radiotherapy techniques,
men with prostate cancer now have < 1% risk of a
secondary malignancy (26).
It has been reported that treatment with cyclo-
phosphamide for primary malignancies and autoim-
mune disease increases the risk of bladder cancer by
fourfold to ninefold, and is higher with greater
cumulative doses and longer duration of exposure
(28–31). Mesna (2-mercaptoethanesulfonic acid) is
commonly co-administered with cyclophosphamide
to decrease the development of cyclophosphamide-
induced bladder cancer. It is speculated that Mesna,
which is almost exclusively excreted by the kidneys
counteracts the toxic effects of acrolein, an inactive
metabolite of cyclophosphamide (32). Historically, it
has been reported that chronic abusers of the analge-
sic phenacetin had a four times higher risk of blad-
der malignancy relative to non-users (33,34). The
major metabolite of phenacetin is acetaminophen
(paracetamol), which is often present in modern
analgesics. However, heavy use of acetaminophen-
containing analgesics did not increase bladder cancer
risk in a number of studies (34–36).
The oral antidiabetic drug pioglitazone is currently
under post marketing pharmacovigilance, as a small
excess of bladder cancer cases was shown in two
studies evaluating its role in the management of dia-
betes. The risk was associated with drug exposure in
excess of 2 years (37,38). However, the benefits of
pioglitazone are considered to outweigh this small
risk for those who respond to treatment and in
whom there is no history or other risk of bladder
Bladder cancer management 437
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
cancer. As a general principle, people with diabetes
and non-visible haematuria (NVH) should be con-
sidered for further investigation in the same way as
any other individuals with NVH.
There are also reported associations between
increased risk of urothelial cancers and Aristolochia
fangchi (a Chinese herb found in some over-the-
counter diet pills) (39).
Presenting symptoms
The majority of patients diagnosed with bladder can-
cer present with painless visible haematuria (VH)
(13). Some patients, especially where CIS is present,
may present with persistent irritative urinary symp-
toms that may be accompanied by haematuria pres-
ent at dipstick testing [symptomatic NVH (s-NVH)].
Other tumours may be detected following investiga-
tion for asymptomatic NVH (a-NVH), or rarely with
renal failure caused by bilateral ureteric obstruction
or symptoms of metastatic disease (13). Around 19%
presenting with VH will have a urinary tract malig-
nancy detected in a one-stop haematuria clinic set-
ting, compared with around 5% presenting with
NVH; this is more likely if they have symptoms
(40,41). However, there is currently no evidence-base
to support population-based screening (7).
What is significant haematuria?
Recent publications from the UK have clarified what
constitutes clinically relevant haematuria, which
patients should be referred for clinical assessment,
and whether they should be referred to a urologist, a
nephrologist or both (summarised in Table 1)
(42,43). Briefly, these are: NVH (‡ 1 + blood on dip-
stick); persistent a-NVH (2 of 3 ‡ 1 + blood on dip-
stick; clinically relevant haematuria [any episode of
painless VH, painful VH or s-NVH in the absence of
urinary tract infection (UTI), recurrent or refractory
UTI with VH, or persistent a-NVH]. The latter
requires urgent investigation.
The American Urological Association (AUA) has
also recently issued guidelines relating to the diagno-
sis and management of a-NVH (44). A notable dif-
ference between these guidelines and the UK-based
publications is that the AUA defines a-NVH as ‡ 3
red blood cells per high powered field on a properly
collected urine specimen. The AUA Guidelines state
that a positive dipstick does not define a-NVH. The
strength of evidence for this AUA recommendation
is based on expert opinion.
Causes of transient NVH include single episodes
of UTI (45), exercise (46) and benign prostatic
hyperplasia (44). Spurious causes of NVH include
menstrual contamination, sexual intercourse, certain
foods (beetroot, blackberries, rhubarb), rhabdomyol-
ysis, drugs (doxorubicin, chloroquine, rifampicin)
and chronic lead or mercury poisoning, and these
should be excluded. However, haematuria in patients
receiving anticoagulant therapy should not be attrib-
uted solely to these agents (47,48). Recurrent or per-
sistent UTI associated with haematuria is significant
and merits urgent urological referral.
What initial investigations should be
performed in primary care?
A joint consensus statement published by the Renal
Association and BAUS, followed by a review article,
emphasised the importance of a basic nephrological
screen in all patients with NVH (42,43). Almost one-
half of patients referred to urological haematuria
clinics have NVH; however in this setting, patients
with NVH are twice as likely to have nephrological
disease (10%) than bladder cancer (5%) (49). The
overall incidence of urological malignancy in screen-
detected a-NVH is < 1% (50).
All patients with s-NVH and persistent a-NVH
should have their baseline blood pressure (BP), esti-
mated glomerular filtration rate (eGFR), and albu-
min:creatinine ratio (ACR) measured (Table 1).
Further nephrological assessment should be consid-
ered in those aged < 40 years with persistent a-NVH
if any one of eGFR < 60 ml ⁄ min ⁄ 1.73 m2
, ACR > 30
or BP > 140 ⁄ 90 mmHg are present.
Which patients with haematuria
require urological referral?
In the UK, referral guidelines for suspected cancer
were published by NICE in 2005 (51). These included
referral guidelines for haematuria (summarised in
Table 1). If a patient is aged < 40 years and has persis-
tent a-NVH, referral to a urology service is not consid-
ered necessary unless there are risk factors for bladder
cancer. If aged 40–50 years with persistent a-NVH or
aged < 50 years with s-NVH, routine referral to urol-
ogy is recommended. If aged > 50 years with persis-
tent a-NVH or s-NVH, urgent urological referral is
recommended. If aged > 40 years with recurrent or
persistent UTI associated with haematuria, an urgent
urological referral should be made.
What tools are available to facilitate
detection of bladder cancer?
Currently recommended tools
Cystoscopy remains the gold standard for the detec-
tion of both new and recurrent bladder cancer.
438 Bladder cancer management
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
Despite this, its sensitivity is limited to approxi-
mately 94% for the detection of tumours during fol-
low-up (52), and may be as low as 60% for the
detection of CIS (53).
First described in 1945, the specificity of urine cytol-
ogy for underlying urothelial malignancy is > 95%.
However, the overall sensitivity of voided urine cytol-
ogy is only 30–50%, as low-grade tumours do not shed
malignant cells. It is therefore most sensitive in
patients with high-grade tumours and CIS (54).
Photodynamic diagnosis (PDD) requires the intra-
vesical instillation of a photosensitising agent, usually
5-aminolevulinic acid (5-ALA) or its hexyl ester
[hexaminolevulinate (HAL)], at least 1 h prior to
cystoscopy. The derivative is preferentially taken up
by tumour cells and produces orange fluorescence
when blue light of 400 nm wavelength is applied.
Two meta-analyses, including data up to 2009, con-
cluded that compared with white-light cystoscopy
(WLC), PDD improved detection of bladder tumours
(especially CIS), and PDD use was associated with a
lower rate of tumour recurrence (55,56). There was
no evidence that PDD affected tumour progression
or survival. However, the true added value of PDD
in reducing tumour recurrence in routine practice is
controversial because studies vary in the choice of
photosensitiser (5-ALA or HAL) and use of immedi-
ate single-dose intravesical chemotherapy post tran-
surethral resection (TUR). Four randomised
controlled trials (RCTs) of PDD vs. WLC published
since 2009 have shown conflicting data (57–60).
Tools undergoing development
In general, novel urinary biomarkers possess greater
sensitivity than cytology for the diagnosis of bladder
cancer, at the expense of lower specificity, but none
have sufficient sensitivity to obviate the need to per-
form cystoscopy (54). Contemporary US Food and
Drug Administration-approved and available tests
are point-of-care (Nuclear Matrix Protein 22) and
laboratory-based (ImmunoCytTM
, Scimedx Corpora-
tion, Denville, NJ, USA and UroVysionTM
, Abbott
Molecular Inc, Des Plaines, IL, USA). Their most
promising application may be as part of surveillance
protocols after treatment, but large prospective trials
validating such protocols are lacking. There is con-
cern regarding their use as a diagnostic adjunct in
patients presenting with haematuria, as the increased
false-positive rate would lead to unnecessary invasive
investigations in patients with otherwise normal cys-
toscopic appearances and imaging.
Narrow-band imaging (NBI) enhances the contrast
between the bladder mucosa and vascular structures
by filtering white light into two narrow bands (415
and 540 nm) without the need for a preoperative
instillation of contrast agent. Improved detection of
primary and recurrent tumours has been shown in
small non-randomised studies (61), and recurrence
rates may be reduced when used as part of follow-up
(62). The role of NBI-cystoscopy as an aid to TUR
and in surveillance needs further evaluation in RCTs.
Pivotal considerations in the
management of NMIBC (TCC)
An algorithm outlining the primary management of
NMIBC (TCC) is presented in Figure 1.
Allocation to a risk group
Patients with NMIBC have a diverse prognosis. The
European Organisation for Research and Treatment
of Cancer (EORTC) has identified six key risk factors
for the prediction of tumour recurrence and progres-
sion to muscle invasive disease (63). These are: num-
ber of tumours; prior recurrence rate; tumour size;
stage (T-category); histological grade (1973 World
Health Organization grade); presence of concomitant
CIS. From these, risk scores are developed to enable
patients to be classified into low-, intermediate-and
high-risk groups for recurrence and progression.
Risks of recurrence and progression at 5 years fol-
lowing diagnosis vary from 31% to 78% and 0.8% to
45% respectively (63).
TUR bladder tumour (papillary)
& immediate single-dose post-operative
intravesicial chemotherapy instillation
NMIBC (TCC)
Intermediate riskLow risk
5 year follow-up only
(if no recurrence)
Very high risk &/or
long life-expectancy
Consider re-resection
and CT urogram
High risk
Surveillance
cystoscopies
Consider
further intravesical
chemotherapy
(maximum 1 year)
or
BCG + maintenance
(1–3 years)
No further
intravesical
chemotherapy
TUR : Transurethral resection
NMIBC : Non-muscle invasive bladder cancer
TCC : Transitional cell carcinoma
Intravesical BCG
+ maintenance
(1–3 years)
Consider
immediate radical
cystectomy
+ pelvic
lymphadenectomy
Figure 1 Management of non-muscle invasive bladder cancer (TCC). BCG, bacillus
Calmette-Gue´rin; CT, computed tomography; EORTC, European Organization for
Research and Treatment of Cancer; NMIBC, non-muscle invasive bladder cancer;
TCC, transitional cell carcinoma; TUR, transurethral resection
Bladder cancer management 439
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
Detrusor muscle status
The quality of the initial TUR has a substantial
impact on outcomes – removal of adequate detrusor
muscle is associated with reduced risk of recurrence
(64). Local recurrence rates for a single tumour at
first-check cystoscopy can vary from 3.5% to 20.6%
depending on the institution (65).
To exclude muscle invasion at the primary TUR,
larger tumours should be resected so that at least ex-
ophytic and deep biopsies are submitted in separate
fractions (66). Repeat TUR is recommended where
the initial resection is judged to be incomplete, either
because of the absence of detrusor muscle in a high-
grade tumour or caused by macroscopic residual dis-
ease; this is also currently recommended in all high-
grade T1 tumours even if detrusor muscle was pres-
ent in the first TUR specimen (66). Failure to adhere
to these principles can adversely affect outcomes.
Contemporary series have shown that in patients ini-
tially staged as T1, 20% were upstaged to muscle
invasive disease after repeat TUR (67) and residual
tumour rates were 25–33% (68,69). Although EAU
guidelines currently include high-grade Ta tumours
in their recommendation for repeat resection (66),
the evidence for repeat resection of high-grade Ta
tumours is less robust than for high-grade T1
tumours.
Requirement for adjuvant intravesical
treatment
The most commonly used intravesical chemothera-
peutic agents are mitomycin C (MMC) and epirubi-
cin. The intravesical immunotherapy of choice is
bacillus Calmette-Gue´rin (BCG).
A single postoperative intravesical instillation of
chemotherapy within 24 h of TUR (and ideally 6 h)
is currently recommended for all newly diagnosed
bladder tumours (66). In clinical studies, this
reduced tumour recurrence rates by 39% (70);
Another study involving patients with recurrent
NMIBC showed that the recurrence rate was
increased twofold if instillation was performed more
than 24 h after TUR (71).
Need for further instillations of chemotherapy
or BCG
The effect of a single instillation of intravesical che-
motherapy lasts for approximately 500 days (72).
The choice between further intravesical chemother-
apy or immunotherapy is guided by the patient’s risk
group. Further intravesical chemotherapy reduces the
risk of recurrence, but has not been shown to reduce
disease progression (73); BCG is more effective than
chemotherapy in reducing recurrence, provided
maintenance treatment is administered (74), but is
associated with more side effects than chemotherapy.
If maintenance therapy is given for at least 1 year,
BCG is the only intravesical agent shown to reduce
or at least delay the risk of disease progression, by
around 37% (75–78). Compared with epirubicin,
maintenance BCG was superior in preventing pro-
gression and also improving overall survival (OS)
(77). However, the relative benefit of maintenance
BCG over MMC in terms of disease progression is
more controversial (74).
For patients at low risk of recurrence and progres-
sion (EORTC recurrence and progression
scores = 0), no further treatment after TUR is rec-
ommended prior to a further recurrence. The proba-
bility of recurrence at 1 year is 15% and for
progression is 0.2% (63).
In intermediate-risk patients, the main priority is
to reduce the risk of recurrence (46–62% at 5 years),
although the risk of progression is not negligible (6–
17% at 5 years) (63). A maximum of 1 year of intra-
vesical chemotherapy or 1–3 years of intravesical
BCG is currently recommended, although the supe-
rior efficacy of BCG needs to be balanced against its
increased toxicity (66). Three-year vs. one-year BCG
maintenance at full-dose compared with one-third-
dose BCG in patients with intermediate- and selected
high-risk (solitary tumour without CIS) NMIBC has
been tested in the EORTC 30962 RCT (78). Superi-
ority could not be formally concluded for either
comparison, although patients receiving full-dose
BCG for 3 years had the highest disease-free survival
(DFS) at 5 years, whereas those receiving one-third-
dose for 1 year had the lowest. Clearly, extending
maintenance treatment for more than 1 year is a bal-
ance of efficacy, side effects and inconvenience and is
worthy of discussion with the patient. In routine
clinical practice, many urologists in the UK delay the
decision until the patient develops a further recur-
rence. Some administer a further single-dose chemo-
therapy instillation after TUR for recurrent disease,
but this has not been validated for recurrent
tumours. Others give six instillations (one instillation
weekly for 6 weeks) of intravesical chemotherapy
before the first-check cystoscopy at around 3 months
after TUR.
In high-risk patients (EORTC progression score
> 14), the 5-year probability of disease progres-
sion is 45% (63). Here, the primary goal of intra-
vesical treatment is not only to prevent
recurrence, but also to prevent progression. Main-
tenance BCG has been established as the bladder-
sparing treatment of choice in these patients. The
optimal duration of treatment has not yet been
established, but at least 1 year of maintenance is
recommended (66,78). The Southwest Oncology
440 Bladder cancer management
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
Group protocol is the most widely used and
involves giving up to 27 instillations of BCG over
a 3-year period (79).
Considerations for radical cystectomy
There are no RCTs comparing BCG with immediate
radical cystectomy. There is, however, consensus that
immediate radical cystectomy should be considered
for patients with high-risk NMIBC where the risk of
tumour progression is especially high, or concomitant
superficial urethral TCC is present, or where the
patient requests it in preference to intravesical BCG.
Added risk factors for progression in high-grade T1
disease include associated CIS (80), multifocality,
tumour size > 3 cm and persistent T1 disease at repeat
resection (81,82). If more than one of these risk factors
is present, then radical cystectomy should be strongly
considered. A growing body of urologists believe that
radical cystectomy for high-grade T1 TCC is under-
utilised at present. Their concerns are based on non-
randomised evidence that suggests worse outcomes
following cystectomy for failed intravesical treatment
compared with immediate cystectomy, although such
studies are likely to include selection bias (83,84).
Immediate radical cystectomy for NMIBC may also be
indicated when aggressive variants of TCC are present,
e.g. diffuse areas of micropapillary variant are detected
(85,86) or in those where BCG is contraindicated, e.g.
significant immunosuppression. Radical cystectomy is
also indicated for squamous cell carcinoma and radical
or partial cystectomy should be considered for adeno-
carcinoma.
Failure of BCG
The definition of BCG failure is controversial, but
the persistence of T1 disease at the 3-month check
cystoscopy after induction BCG (six instillations
administered once-weekly for 6 weeks); high-grade
Ta disease ⁄ CIS at 6 months; or detection of muscle
invasive TCC should prompt consideration of radical
cystectomy.
Increased risk of concomitant or metachronous
upper urinary tract (UUT)-TCC
The incidence of concomitant UUT-TCC is low
(1.8%), but in patients with trigonal tumours, the
incidence is 7.5% (87). During follow-up, patients
with high-grade and multifocal NMIBC are more
likely to develop UUT-TCC (88). Ultrasound is rou-
tinely performed during diagnostic work-up of blad-
der cancer, but can miss small UUT-TCC.
Computed tomography urography should therefore
be considered where the bladder tumour is high-
grade, multifocal or trigonal (66).
Prevention of bladder tumours after
nephro-ureterectomy for primary UUT-TCC
After a nephroureterectomy for UUT-TCC, up to
40% of patients will develop bladder cancer. In a
phase III RCT (ODMIT-C), a single dose of postop-
erative intravesical MMC, administered at the time
of urethral catheter removal, reduced the risk of a
bladder tumour within the first year after nephro-
ureterectomy; the absolute reduction in risk was
11%, the relative reduction was 40% and the num-
ber needed to treat to prevent one bladder tumour
was 9 (89). The trial was the largest randomised
study ever conducted in the management of patients
with UUT-TCC. However, it was not designed to
determine the most effective method of nephroure-
terectomy, and so a number of techniques were
allowed for management of the distal ureter.
Patients with a previous history of bladder cancer
were excluded.
Bladder-sparing treatments after BCG failure
Radical cystectomy remains the mainstay of treat-
ment for patients who have failed BCG treatment.
For patients who are unwilling or unfit to undergo
this procedure, the treatment options are limited.
Intravesical chemotherapeutic agents, such as gem-
citabine and docetaxel, novel immunotherapies, such
as interferon-alpha, and device-assisted treatments
have all shown promise. However, to date, much of
the evidence to support their potential benefit is
based on non-randomised or small Phase II studies.
At best, currently available bladder-sparing treat-
ments for those with BCG-refractory TCC are associ-
ated with 2-year DFS of approximately 50% (90).
Chemohyperthermia (c-HT) describes the combi-
nation of intravesical chemotherapy and hyperther-
mia, where the chemotherapy and the bladder wall
are heated to temperatures of between 44 and 45 °C.
The most common form of c-HT uses the Synergo
HT system in which local HT is administered via a
915 MHz intravesical microwave applicator. c-HT
increases cell membrane permeability, enhances uro-
thelial exposure and in particular lamina propria
exposure, alters intracellular drug trafficking and
enhances the effects of cytostatic chemotherapy (91).
Over the last 15 years, c-HT has been tested in a
variety of clinical settings, including small several
Phase II RCTs, in the BCG-naı¨ve setting (92).
Data supporting the role of c-HT (using MMC) in
BCG-refractory NMIBC has come from several
proof-of-concept studies. In patients with BCG-
refractory CIS, a complete response rate of 92% was
shown, with 50% of patients remaining disease-free
at 2-year follow-up (93). In patients with
Bladder cancer management 441
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
BCG-refractory NMIBC (77% high-risk) treated with
a maintenance c-HT schedule, the recurrence-free
rate was 56% at 2 years; 3% progressed to muscle
invasive disease and 5% withdrew from treatment
because of adverse events (94). The use of c-HT is
being further evaluated in BCG-refractory patients
with NMIBC who are unwilling or unfit for cystecto-
my in the UK-based HYMN Phase III trial (EUD-
RACT-2008-005428-99).
Electromotive drug administration (EMDA) is an
alternative way of enhancing MMC absorption and
urothelial exposure, by creating an electrical gradient
across the bladder wall using electrodes placed within
the catheter and on the lower abdominal wall. In
patients with BCG-naı¨ve high-risk NMIBC, EMDA-
MMC has shown promise (95,96). To date, no stud-
ies have specifically evaluated EMDA-MMC in the
BCG-refractory setting, although one RCT allowed
crossover of patients to EMDA-MMC alone if they
did not respond to primary BCG treatment (95).
Key considerations when treatment
intent for muscle invasive TCC is
potentially curative
An algorithm outlining the primary management of
muscle invasive bladder cancer is presented in Figure 2.
Radical cystectomy or radical radiotherapy?
Worldwide, radical cystectomy and pelvic lympha-
denectomy has been the cornerstone treatment for
muscle invasive TCC, although no RCT data exist to
support cystectomy over bladder preservation with
radical radiotherapy. Non-randomised data from sin-
gle gold-standard institutions show similar outcomes
for DFS (97,98). Typical radical radiotherapy sched-
ules are 64 Gy in 32 fractions over 6.5 weeks or
55 Gy in 20 fractions over 4 weeks. An important
confounding factor when comparing the results of
radical cystectomy and radiotherapy is the discrep-
ancy between pathological staging (cystectomy series)
and clinical staging (radiotherapy series). Clinical
staging is more likely to underestimate disease extent
and so there is an outcome bias in favour of cystec-
tomy series. For example, in one publication, only
23% of clinical stage T2 tumours (cT2) were con-
firmed to be pathological stage T2 (pT2) at cystecto-
my; 74% were pT3 ⁄ pT4 (99). In a contemporary
series assessing cystectomy and pelvic lymphadenec-
tomy alone, the 5-year OS for the entire cohort was
59% (100). In a separate study, following radical
radiotherapy alone, 5-year OS was also approxi-
mately 60% (101). When directly comparing radical
cystectomy and radical radiotherapy, no significant
difference between interventions in terms of 5-year
OS was evident (102).
Local disease control is a clinically relevant chal-
lenge in the management of muscle invasive TCC
and is consistently better after cystectomy than
radiotherapy alone, balanced against the benefits of
retaining the native bladder; many urological sur-
geons do not offer reconstructive surgery in the sal-
vage cystectomy setting. One concern is that salvage
cystectomy for local disease recurrence after radio-
therapy is technically more difficult than primary
cystectomy, with a higher risk of complications.
However, one large study demonstrated that OS
after salvage cystectomy was identical to that of pri-
mary cystectomy, but dependent on the experience
of the operator (103). The local relapse rate follow-
ing cystectomy is 10% (104), but can be as low as
8% when a pelvic lymphadenectomy is performed
(100).
Factors that would favour cystectomy over radio-
therapy include poor bladder function, widespread
CIS, large volume tumours, preexisting hydronephro-
sis, previous pelvic radiotherapy and active inflamma-
tory bowel disease. Where compliance with follow-up
cystoscopic surveillance is likely to be difficult, cystec-
tomy should be the preferred option. Overall, these
factors must be weighed against morbidities associated
with cystectomy, and patient preference.
Available technologies choices for radical
cystectomy and pelvic lymphadenectomy
Radical cystectomy is a major operation; periopera-
tive mortality is 3%, and 28% of patients develop a
complication within 3 months of surgery (105).
Ninety-day mortality rates increase with patient age:
TUR bladder tumour (solid)
Muscle invasive (T2-T2 N0-N3 M0) TCC
(potentially curative on cross sectional staging)
Advanced/metastatic (M1) TCC
on cross sectional staging
Locally advanced
down-staging systemic
chemotherapy followed by
repeat cross sectional imaging
Localised
neoadjuvant
systemic
chemotherapy
Radiotherapy
with sensitiser
Radical
radiotherapy
Radical
cystectomy
Palliative
TUR : Transurethral resection
TCC : Transitional cell carcinoma
Palliative systemic
chemotherapy
+/–
palliative radiotherapy
Figure 2 Management of muscle invasive or advanced ⁄ metastatic bladder cancer
(TCC). TCC, transitional cell carcinoma; TUR, transurethral resection
442 Bladder cancer management
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
< 70 years, 2%; 70–79 years, 5.4%; 80–89 years, 9.2%
(106). When strict reporting guidelines are imple-
mented in high volume centres, surgical morbidity
following radical cystectomy is reported to be even
higher (107,108). As well as removing the bladder
and pelvic lymph nodes, the standard procedure is
also to remove the prostate and seminal vesicles in
men, and uterus and adnexa in women. The inclu-
sion of the entire prostate in male patients and the
extent of urethrectomy and vaginal resection in
female patients have recently been questioned. There
is a substantial amount of literature about the extent
of lymphadenectomy, but consensus concerning the
optimal extent remains elusive. Available data sug-
gest that removal of least 11 lymph nodes may be
associated with more favourable outcomes (109).
Most agree that the template for standard pelvic
lymph node dissection should at least include pelvic
lymph nodes and external iliac lymph nodes up to
level of the common iliac bifurcation (110). The
extent of lymph node dissection and its impact on
survival is currently the subject of RCTs, including
the ongoing Southwest Oncology Group trial
(SWOG-S1011; NCT01224665) in the United States
and the AUO-multicentre RCT (AB 25 ⁄ 02;
NCT01215071) in Germany, which is now closed to
recruitment. Laparoscopic cystectomy and robotic-
assisted cystectomy are feasible and also currently
the subject of RCTs.
Following radical cystectomy, urinary diversion is
necessary. The most established option is the ileal
conduit. The second most common option is the
construction of a detubularised ileal orthotopic neo-
bladder that is anastomosed to the urethra. The third
option is a continence pouch that can be self-cathe-
terised via, e.g. an appendix stoma.
The presence of positive soft tissue surgical mar-
gins following radical cystoprostatectomy is a strong
predictor of recurrence-free and disease-specific sur-
vival (111). EAU guidelines recommend that urethr-
ectomy should be performed if positive surgical
margins are present at the level of the urethral dis-
section, if the tumour is located at the bladder neck
or in the urethra (in women), or if the tumour
extensively infiltrates the prostate (13). Factors that
may protect against urethral recurrence include
orthotopic reconstruction and radical radiotherapy.
Evolving radiotherapy techniques
The volume treated by radiotherapy has historically
been the whole bladder, but for unifocal disease,
techniques have evolved using a lower total dose to
the whole bladder with a boost to the tumour bed.
The potential benefits of this approach are to reduce
toxicity and allow dose-escalation to the primary dis-
ease. However, it is critical that organ motion and
verification of the intended area of treatment is
optimised.
Novel approaches to achieve this include image-
guided radiotherapy, allowing pretreatment com-
puted tomography verification to be performed on a
daily basis prior to each fraction and real-time moves
to be made. However, the cost of this technique is
significant and not available throughout the UK, but
represents a significant development in radiotherapy
implementation.
Neoadjuvant, down-staging or adjuvant
chemotherapy
Up to 30% of patients relapse with distant metastases
after radical cystectomy (100). Use of neoadjuvant
chemotherapy, prior to radical cystectomy or radio-
therapy, is postulated to reduce the risk of microm-
etastatic disease and thereby confer a survival
advantage. Cisplatin-containing combination chemo-
therapy is the recommended approach (13). Typi-
cally, three cycles are given, and the most commonly
used contemporary chemotherapy regimens are the
gemcitabine-cisplatin (GC) and methotrexate-vin-
blastine-adriamycin-cisplatin (M-VAC) combina-
tions. Neoadjuvant chemotherapy improves 5-year
OS by 5–8%, irrespective of type of subsequent local
treatment (112–114). This translates into a 14–16%
relative reduction in risk of death and a 22–26% rel-
ative improvement in DFS (112,113). Adequate renal
function and performance status (PS) is necessary
for these treatments.
Down-staging chemotherapy should be considered
for locally advanced bladder cancer (inoperable, but
non-metastatic) where the aim is to increase the pos-
sibility of either successful cystectomy or radiother-
apy. Three to six cycles are commonly given, with
cross-sectional imaging performed after three cycles
to assess response.
Currently, no RCT or meta-analysis has provided
sufficient data to support the routine use of adjuvant
chemotherapy because of underpowered studies and a
difficulty in recruitment (13). It remains unclear
whether immediate adjuvant chemotherapy or chemo-
therapy at the time of relapse is superior or equivalent
in terms of OS. However, adjuvant chemotherapy may
be discussed on an individual basis particularly in
patients with node-positive disease or locally advanced
disease with high-grade pathology who have not
already received neoadjuvant chemotherapy. Adequate
renal function and PS is again mandated.
Selective bladder preservation strategies
To optimise the success of selective bladder preserva-
tion, patients must be carefully selected to increase
Bladder cancer management 443
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
the likelihood of complete response, and meticulous
cystoscopic follow-up must be performed to identify
recurrent or persistent muscle invasive disease in
candidates suitable for early salvage cystectomy. It is
important to minimise the amount of normal tissue
in the irradiated area, especially as the small bowel is
used in an ileal conduit or in reconstruction in the
event of salvage cystectomy.
Emerging data from RCTs suggests that the stan-
dard of care for bladder preservation is likely to
become neoadjuvant chemotherapy followed by
radiotherapy with a concurrent radiosensitiser (115–
117). The Phase III BC2001 trial of 360 patients that
compared radiotherapy and concurrent 5-fluoroura-
cil and MMC chemotherapy (a non-renal toxic regi-
men) with radiotherapy alone demonstrated a
significant improvement in loco-regional recurrence-
free survival (67% compared with 54%) at 2 years
(116). A similar degree of benefit was seen across all
tumour stages (T2–T4) irrespective of age or whether
the patient had received neoadjuvant chemotherapy.
In a further Phase II study of patients receiving
chemoradiation compared with historical radiother-
apy controls, 89% of surviving patients at 3 years
had an intact bladder following chemoradiotherapy
with weekly gemcitabine (117).
Tumour hypoxia has long been considered a cause
of radiotherapy failure and one strategy to improve
radiosensitivity is therefore to increase oxygenation of
the tumour. In the BCON trial, radiotherapy adminis-
tered with carbogen breathing apparatus and nicotin-
amide tablets improved OS by 13% compared with
radiotherapy alone (115). Of long-term survivors in
the experimental arm, 83% retained intact bladders.
Quality of life after cystectomy and selective
bladder preservation
Radical cystectomy is associated with significant alter-
ation in health-related quality of life (HRQoL), with
most of the morbidity related to the use of intestinal
segments for urinary diversion. A commonly held
assumption is that patients who choose neobladders
should have improved HRQoL compared with those
who undergo ileal conduit formation. However, a sys-
tematic review has not determined a superior
approach in terms of HRQoL (118). As well as health
and body image, patients identified non-health-related
determinants, such as family, relationships, finances as
important QoL factors.
A questionnaire-based study compared the
HRQoL of 29 patients who received bladder preser-
vation therapy (combination of chemoradiotherapy
and TUR) and 30 patients who had undergone radi-
cal cystectomy (119). Parameters associated with bet-
ter HRQoL after bladder preservation than
cystectomy included physical well-being (62% vs.
53%), anxiety (28% vs. 57%) and depression (24%
vs. 47%). HRQoL after cystectomy was substantially
impacted by stoma presence and a lack of sexual
activity, but social and recreational life was only
minimally affected. Patients undergoing bladder pres-
ervation therapy reported dysuria (20%), frequency
(44%), nocturia (42%) and difficulty controlling
micturition (38%) (119).
A further study with long-term follow-up (median
6.3 years) evaluated HRQoL and bladder function
assessed by urodynamics in patients treated with
TUR, chemotherapy and radiotherapy (120). Of 32
patients, most (75%) retained normal bladder func-
tion. Urinary flow problems were reported by 6% of
patients, urgency by 15% and urinary leakage in
19%. Difficulty with bowel control occurred in seven
men and three women. Of male patients, 59%
reported they were satisfied with their sex life.
Palliative care approaches for
advanced (metastatic) bladder cancer
(TCC)
Palliative chemotherapy is the cornerstone treatment
for patients with metastatic TCC (Figure 2). It is
usually given to improve HRQoL, improve symp-
toms and also to improve prognosis, but is very unli-
kely to be a cure. Before the development of effective
chemotherapy, patients with TCC and visceral metas-
tases rarely exceeded the median survival of 3–
6 months. In ‘fit’ patients with adequate renal func-
tion, cisplatin-containing combination chemotherapy
with GC or M-VAC should be considered, which can
achieve a median survival of up to 14–15 months
(121); GC may be associated with less toxicity than
M-VAC. Carboplatin-based therapy is less effective
than cisplatin-based therapy, but may be an option
in patients ‘unfit’ for the former. Some patients who
progress after first-line chemotherapy may benefit
from agents, such as paclitaxel or vinflunine.
Palliative radiotherapy may also be administered
to help control symptoms, such as haematuria and
pain. Emerging data suggests that zoledronic acid, a
bisphosphonate, may also be helpful in patients with
bone metastases (122).
Can public and professional
recognition of bladder cancer be
improved?
A single episode of VH is a warning sign that needs
prompt investigation and urological referral. This is
a key message for both the public and doctors in Pri-
mary Care. Symptoms attributable to bladder cancer
444 Bladder cancer management
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
can mimic UTI. Failure to respond to antibiotics fol-
lowing a symptomatic UTI or an early recurrent UTI
warrants flexible cystoscopic evaluation before
considering repeat courses of antibiotics. Otherwise,
diagnosis (particularly in postmenopausal women) is
delayed and may be contributory to less favourable
outcomes in women than men. It is important to get
this message across to doctors in Primary Care and
pharmacists.
Until recently, bladder cancer has been low on the
public health agenda. However, the Department of
Health has recognised the need for earlier diagnosis
of bladder cancer, and has commenced a pilot study
in three regions of the UK to determine the best way
to promote these messages.
Action on Bladder Cancer (http://www.actionon
bladdercancer.org) is the only UK national bladder
cancer charity purely focussed on improving the lives
of patients with bladder cancer. It has three primary
aims, namely to: improve awareness; elevate the sta-
tus of bladder cancer in the public health agenda;
and improve medical knowledge. It is hoped that the
website will provide a good resource for the general
public, patients and healthcare professionals.
Acknowledgements
Sources of funding: None.
Author contributions
This project was initiated by Mr Leyshon Griffiths
(Consultant Urologist, Leicester, Secretary of Action
on Bladder Cancer) and Mr Colin Bunce (Consultant
Urologist, Middlesex, Chairman of Action on Blad-
der Cancer). Primary development of the manuscript
and co-ordination was conducted by Mr Griffiths
with support and advice from Right Angle Commu-
nications (Action on Bladder Cancer Secretariat).
The manuscript was reviewed by members of the
Action on Bladder Cancer Executive Committee: Dr
Alison Birtle (Consultant Clinical Oncologist, Pres-
ton), Dr Mark Beresford (Consultant Clinical Oncol-
ogist, Bath), Mr Roger Kockelbergh (Consultant
Urological Surgeon, Leicester), Mr Mark Feneley
(Consultant Urological Surgeon, London), Mr Jer-
emy Crew (Consultant Urological Surgeon, Oxford)
and Mr Hugh Mostafid (Consultant Urological Sur-
geon, Basingstoke).
References
1 Ferlay J, Shin HR, Bray F et al. Estimates of world-
wide burden of cancer in 2008: GLOBOCAN 2008.
Int J Cancer 2010; 127: 2893–917.
2 Cancer Research UK. Bladder cancer statistics –
key facts. http://info.cancerresearchuk.org/cancer-
stats/keyfacts/bladder-cancer/ (accessed June 2012).
3 Fajkovic H, Halpern JA, Cha EK et al. Impact of
gender on bladder cancer incidence, staging, and
prognosis. World J Urol 2011; 29: 457–63.
4 Edwards BK, Ward E, Kohler BA et al. Annual
report to the nation on the status of cancer, 1975–
2006, featuring colorectal cancer trends and impact
of interventions (risk factors, screening, and treat-
ment) to reduce future rates. Cancer 2010; 116:
544–73.
5 Bosetti C, Bertuccio P, Chatenoud L et al. Trends
in mortality from urologic cancers in Europe,
1970–2008. Eur Urol 2011; 60: 1–15.
6 Qiu D, Katanoda K, Marugame T, Sobue T. A
Joinpoint regression analysis of long-term trends
in cancer mortality in Japan (1958–2004). Int J
Cancer 2009; 124: 443–8.
7 Moyer VA, on behalf of the U.S. Preventive Ser-
vices Task Force. Screening for bladder cancer: US
Preventive Services Task Force Recommendation
Statement. Ann Intern Med 2011; 155: 246–51.
8 Keegan KA, Resnick MJ, Clark PE. Multimodal
therapies for muscle-invasive urothelial carcinoma
of the bladder. Curr Opin Oncol 2012; 24: 278–83.
9 British Association of Urological Surgeons, Section
of Oncology. BAUS Cancer Registry, analyses of
minimum data set for urological cancers, January
to December 2009. October 2010. http://www.bau-
s.org.uk/Resources/BAUS/Documents/
PDF%20Documents/Data%20and%20Audit/2009fi-
nalanalyses.pdf (accessedJune 2012).
10 Yafi FA, Aprikian AG, Chin JL et al. Contempo-
rary outcomes of 2287 patients with bladder can-
cer who were treated with radical cystectomy: a
Canadian multicentre experience. BJU Int 2011;
108: 539–45.
11 Gouda I, Mokhtar N, Bilal D, El-Bokhainy T, El-
Bokhainy NM. Bilharziasis and bladder cancer: a
time trend analysis of 9843 patients. J Egypt Natl
Canc Inst 2007; 19: 158–62.
12 Felix AS, Soliman AS, Khaled H et al. The chang-
ing patterns of bladder cancer in Egypt over the
past 26 years. Cancer Causes Control 2008; 19:
421–9.
13 Stenzl A, Witjes JA, Cowan NC, et al. European
Association of Urology Guidelines on muscle inva-
sive and metastatic bladder cancer 2011. http://
www.uroweb.org/gls/pdf/07_%20Bladder%20Can-
cer.pdf (accessed June 2012).
14 Freedman ND, Silverman DT, Hollenbeck AR,
Schatzkin A, Abnet CC. Association between
smoking and risk of bladder cancer among men
and women. JAMA 2011; 306: 737–45.
15 Zeegers MP, Tan FE, Dorant E, van Den Brandt
PA. The impact of characteristics of cigarette
smoking on urinary tract cancer risk: a meta-anal-
ysis of epidemiologic studies. Cancer 2000; 89:
630–9.
16 Baris D, Karagas MR, Verrill C et al. A case-con-
trol study of smoking and bladder cancer risk:
emergent patterns over time. J Natl Cancer Inst
2009; 101: 1553–61.
17 Hartge P, Silverman D, Hoover R et al. Changing
cigarette habits and bladder cancer risk: a case-con-
trol study. J Natl Cancer Inst 1987; 78: 1119–25.
18 Kogevinas M, Mannetje A, Cordier S et al. Occu-
pation and bladder cancer among men in Western
Europe. Cancer Causes Control 2003; 14: 907–14.
19 Chen CJ, Chuang YC, Lin TM, Wu HY. Malignant
neoplasms among residents of a blackfoot disease-
endemic area in Taiwan: high-arsenic artesian well
water and cancers. Cancer Res 1985; 45: 5895–9.
20 Marcus PM, Vineis P, Rothman N. NAT2 slow
acetylation and bladder cancer risk: a meta-analysis
of 22 case-control studies conducted in the general
population. Pharmacogenetics 2000; 10: 115–22.
21 Garcı´a-Closas M, Malats N, Silverman D. NAT2
slow acetylation, GSTM1 null genotype, and risk
of bladder cancer: results from the Spanish Blad-
der Cancer Study and meta-analyses. Lancet 2005;
366: 649–59.
22 Lee SW, Jang IJ, Shin SG et al. CYP1A2 activity as
a risk factor for bladder cancer. J Korean Med Sci
1994; 9: 482–9.
23 Kiemeney L, Thorlacius S, Sulem P et al. Sequence
variant on 8q24 confers susceptibility to urinary
bladder cancer. Nat Genet 2008; 40: 1307–12.
24 Duncan RE, Bennett DW, Evans AT, Aron BS,
Schellhas HF. Radiation-induced bladder tumors. J
Urol 1977; 118: 43–5.
25 Sella A, Dexeus FH, Chong C, Ro JY, Logothetis
CJ. Radiation therapy-associated invasive bladder
tumors. Urology 1989; 33: 185–8.
26 Turina M, Mulhall AM, Mahid SS, Yashar C, Gal-
andiuk S. Frequency and surgical management of
chronic complications related to pelvic irradiation.
Arch Surg 2008; 143: 46–52.
Bladder cancer management 445
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
27 Moon K, Stukenborg GJ, Keim J, Theodorescu D.
Cancer incidence after localized therapy for pros-
tate cancer. Cancer 2006; 107: 991–8.
28 Fairchild WV, Spence CR, Solomon HD, Gangai
MP. The incidence of bladder cancer after cyclo-
phosphamide therapy. J Urol 1979; 122: 163–4.
29 Pedersen-Bjergaard J, Ersboll J, Hansen VL et al.
Carcinoma of the urinary bladder after treatment
with cyclophosphamide for non-Hodgkin’s lym-
phoma. N Engl J Med 1988; 318: 1028–32.
30 Travis LB, Curtis RE, Glimelius B et al. Bladder
and kidney cancer following cyclophosphamide
therapy for non-Hodgkin’s lymphoma. J Natl Can-
cer Inst 1995; 87: 524–30.
31 Knight A, Askling J, Granath F, Sparen P, Ekbom
A. Urinary bladder cancer in Wegener’s granulo-
matosis: risks and relation to cyclophosphamide.
Ann Rheum Dis 2004; 63: 1307–11.
32 Links M, Lewis C. Chemoprotectants: a review of
their clinical pharmacology and therapeutic effi-
cacy. Drugs 1999; 57: 293–308.
33 Fokkens W. Phenacetin abuse related to bladder
cancer. Environ Res 1979; 20: 192–8.
34 Castelao JE, Yuan J-M, Gago-Dominguez M, Yu
MC, Ross RK. Non-steroidal anti-inflammatory
drugs and bladder cancer prevention. Br J Cancer
2000; 87: 1364–9.
35 Fortuny J, Kogevinas M, Zens MS et al. Analgesic
and anti-inflammatory use and risk of bladder
cancer: a population based case-control study.
BMC Urol 2007; 7: 13.
36 Genkinger JM, De Vivo I, Stampfer MJ, Gio-
vannucci E, Michaud DS. Nonsteroidal anti-
inflammatory drug use and risk of bladder cancer
in the health professionals follow-up study. Int J
Cancer 2007; 120: 2221–5.
37 Dormandy JA, Charbonnel B, Eckland DJ et al.
Secondary prevention of macrovascular events in
patients with type 2 diabetes in the PROactive
Study (PROspective pioglitAzone Clinical Trial In
macroVascular. Lancet 2005; 366: 1279–89.
38 Lewis JD, Ferrara A, Peng T et al. Risk of bladder
cancer among diabetic patients treated with piog-
litazone: interim report of a longitudinal cohort
study. Diabetes Care 2011; 34: 916–22.
39 Nortier JL, Martinez MC, Schmeiser HH et al.
Urothelial carcinoma associated with the use of a
Chinese herb (Aristolochia fangchi). N Engl J Med
2000; 342: 1686–92.
40 Khadra MH, Pickard RS, Charlton M, Powell PH,
Neal DE. A prospective analysis of 1,930 patients
with hematuria to evaluate current diagnostic
practice. J Urol 2000; 163: 524–7.
41 Edwards TJ, Dickinson AJ, Gosling J et al. Patient-
specific risk of undetected malignant disease after
investigation for haematuria, based on a 4-year
follow-up. BJU Int 2011; 107: 247–52.
42 Renal Association and British Association of Uro-
logical Surgeons. Joint consensus statement on the
initial assessment of haematuria. July 2008. http://
www.baus.org.uk/AboutBAUS/publications/hae-
maturia-guidelines (accessed June 2012).
43 Kelly JD, Fawcett DP, Goldberg LC. Assessment
and management of non-visible haematuria in pri-
mary care. BMJ 2009; 338: 227–32.
44 Davis R, Jones JS, Barocas DA, et al. Diagnosis,
evaluation and follow-up of asymptomatic
microhaematuria (AMH) in adults: AUA Guide-
line. http://www.auanet.org/content/media/asymp-
tomatic_microhematuria_guideline.pdf (accessed
October 2012)
45 Chung A, Arianayagam M, Rashid P. Bacterial cysti-
tis in women. Aust Fam Physician 2010; 39: 295–8.
46 Jones GR, Newhouse IJ, Jakobi JM, LaVoie NL,
Thayer R. The incidence of hematuria in middle
distance track running. Can J Appl Physiol 2001;
26: 336–49.
47 Van Savage JG, Fried FA. Anticoagulant associated
haematuria: a prospective study. J Urol 1995; 153:
1594–6.
48 Hurlen M, Eikvar L, Seljeflot I, Arnesen H. Occult
bleeding in three different antithrombotic regimes
after myocardial infarction. A WARIS-II-subgroup
analysis. Thromb Res 2006; 118: 433–8.
49 Edwards TJ, Dickinson AJ, Natale S, Gosling J,
McGrath JS. A prospective analysis of the diagnos-
tic yield resulting from the attendance of 4020
patients at a protocol-driven haematuria clinic.
BJU Int 2006; 97: 301–5.
50 Hiatt RA, Ordon˜ez JD. Dipstick urinalysis screen-
ing, asymptomatic microhematuria, and subse-
quent urological cancers in a population-based
sample. Cancer Epidemiol Biomarkers Prev 1994; 3:
439–43.
51 National Institute for Health and Clinical Excel-
lence. Referral guidelines for suspected cancer.
http://www.nice.org.uk/nicemedia/pdf/CG027pub-
licinfo.pdf (accessed June 2012).
52 Raitanen MP, Leppilahti M, Tuhkanen K et al.
Routine follow-up cystoscopy in detection of
recurrence in patients being monitored for bladder
cancer. Ann Chir Gynaecol 2001; 90: 261–5.
53 Isfoss BL. The sensitivity of fluorescent-light cys-
toscopy for the detection of carcinoma in situ
(CIS) of the bladder: a meta-analysis with com-
ments on gold standard. BJU Int 2011; 108: 1703–
7.
54 Mowatt G, Zhu S, Kilonzo M et al. Systematic
review of the clinical effectiveness and cost-effec-
tiveness of photodynamic diagnosis and urine bio-
markers (FISH, ImmunoCyt, NMP22) and
cytology for the detection and follow-up of blad-
der cancer. Health Technol Assess 2010; 14: 1–331,
iii–iv.
55 Mowatt G, N’Dow J, Vale L et al. Photodynamic
diagnosis of bladder cancer compared with white
light cystoscopy: systematic review and meta-
analysis. Int J Technol Assess Health Care 2011; 27:
3–10.
56 Kausch I, Sommerauer M, Montorsi F et al. Pho-
todynamic diagnosis in non-muscle invasive blad-
der cancer: a systematic review and cumulative
analysis of prospective studies. Eur Urol 2010; 57:
595–606.
57 Schumacher MC, Davidsson T, Friedrich B et al.
Transurethral resection of non-muscle-invasive
bladder transitional cell cancers with or without 5-
aminolevulinic acid under visible and fluorescent
light: results of a prospective randomized multi-
centre study. Eur Urol 2010; 57: 293–9.
58 Stenzl A, Burger M, Fradet Y et al. Hexaminolevu-
linate guided fluorescence cystoscopy reduces
recurrence in patients with nonmuscle invasive
bladder cancer. J Urol 2010; 184: 1907–13.
59 Hermann GG, Mogensen K, Carlsson S, Marcussen
N, Duun S. Fluorescence-guided transurethral
resection of bladder tumours reduces bladder
tumour recurrence due to less residual tumour tis-
sue in Ta ⁄ T1 patients: a randomized two-centre
study. BJU Int 2011; 108(8 Pt 2): E297–303.
60 Stenzl A, Penkoff H, Dajc-Sommerer E et al.
Detection and clinical outcome of urinary bladder
cancer with 5-aminolevulinic acid-induced fluores-
cence cystoscopy: a multicenter randomized, dou-
ble-blind, placebo-controlled trial. Cancer 2011;
117: 938–47.
61 Cauberg ECC, Kloen S, Visser M et al. Narrow
band imaging cystoscopy improves the detection
of non-muscle-invasive bladder cancer. Urology
2010; 76: 658–63.
62 Herr HW, Donat SM. Reduced bladder tumour
recurrence rate associated with narrow-band
imaging surveillance cystoscopy. BJU Int 2011;
107: 396–8.
63 Sylvester RJ, van der Meijden APM, Oosterlinck
W et al. Predicting recurrence and progression in
individual patients with Stage Ta T1 bladder can-
cer using EORTC Risk Tables: a combined analysis
of 2596 patients from seven EORTC trials. Eur
Urol 2006; 49: 466–77.
64 Mariappan P, Finney SM, Head E et al. Good
quality white-light transurethral resection of blad-
der tumours (GQ-WLTURBT) with experienced
surgeons performing complete resections and
obtaining detrusor muscle reduces early recurrence
in new non-muscle-invasive bladder cancer: valida-
tion across time and place and recommendation
for benchmarking. BJU Int 2012; 109: 1666–73.
65 Brausi M, Collette L, Kurth K et al. Variability in
the recurrence rate at first follow-up cystoscopy
after TUR in Stage Ta T1 transitional cell carci-
noma of the bladder: a combined analysis of seven
EORTC studies. Eur Urol 2002; 41: 523–31.
66 Babjuk M, Oosterlinck W, Sylvester R, et al.
European Association of Urology Guidelines on
non-muscle invasive bladder cancer (TaT1 and CIS)
2011. http://www.uroweb.org/gls/pdf/05_TaT1_
Bladder_Cancer.pdf (accessed June 2012).
67 Dalbagni G, Vora K, Kaag M et al. Clinical out-
come in a contemporary series of restaged patients
with clinical T1 bladder cancer. Eur Urol 2009; 56:
903–10.
68 Herr HW, Donat SM. Quality control in transure-
thral resection of bladder tumours. BJU Int 2008;
102: 1242–6.
69 Schips L, Augustin H, Zigeuner RE et al. Is
repeated transurethral resection justified in
patients with newly diagnosed superficial bladder
cancer? Urology 2002; 59: 220–3.
70 Sylvester RJ, Oosterlinck W, van der Meijden
APM. A single immediate postoperative instillation
of chemotherapy decreases the risk of recurrence
in patients with stage Ta T1 bladder cancer: a
meta-analysis of published results of randomized
clinical trials. J Urol 2004; 171: 2186–90.
71 Kaasinen E, Rintala E, Hellstro¨m P et al. Factors
explaining recurrence in patients undergoing
chemoimmunotherapy regimens for frequently
recurring superficial bladder carcinoma. Eur Urol
2002; 42: 167–74.
72 Hinotsu S, Akaza H, Ohashi Y, Kotake T. Intrave-
sical chemotherapy for maximum prophylaxis of
new early phase superficial bladder carcinoma
treated by transurethral resection: a combined
analysis of trials by the Japanese Urological Cancer
Research Group using smoothed hazard function.
Cancer 1999; 86: 1818–26.
446 Bladder cancer management
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
73 Pawinski A, Sylvester R, Kurth KH et al. A com-
bined analysis of European Organization for
Research and Treatment of Cancer, and Medical
Research Council randomized clinical trials for the
prophylactic treatment of stage TaT1 bladder can-
cer. European Organization for Research and
Treatment of Cancer Genitourinary Tract Cancer
Cooperative Group and the Medical Research
Council Working Party on Superficial Bladder
Cancer. J Urol 1996; 156: 1934–40.
74 Malmstrom P-U, Sylvester RJ, Crawford DE et al.
An individual patient data meta-analysis of the
long-term outcome of randomised studies com-
paring intravesical mitomycin C versus bacillus
Calmette-Gue´rin for non-muscle-invasive bladder
cancer. Eur Urol 2009; 56: 247–56.
75 Sylvester RJ, van der Meijden AP, Lamm DL.
Intravesical bacillus Calmette-Guerin reduces the
risk of progression in patients with superficial
bladder cancer: a meta-analysis of the published
results of randomized clinical trials. J Urol 2002;
168: 1964–70.
76 Bo¨hle A, Bock PR. Intravesical bacille calmette-
gue´rin versus mitomycin c in superficial bladder
cancer: formal meta-analysis of comparative stud-
ies on tumor progression. Urology 2004; 63: 682–6.
77 Sylvester RJ, Brausi MA, Kirkels WJ et al. Long-
term efficacy results of EORTC genito-urinary
group randomized phase 3 study 30911 comparing
intravesical instillations of epirubicin, bacillus Cal-
mette-Gue´rin, and bacillus Calmette-Gue´rin plus
isoniazid in patients with intermediate- and high-
risk stage Ta T1 urothelial carcinoma of the blad-
der. Eur Urol 2010; 57: 766–73.
78 Brausi MA, Oddens JR, Sylvester RJ et al. Bacillus
Calmette-Guerin: one third dose versus full dose
and one year versus three years of maintenance.
Final results of an EORTC GU Cancers Group
randomized trial in non muscle invasive bladder
cancer. 27th Annual Congress of the European
Association of Urology, February 2012. Abstract
1050.
79 Lamm DL, Blumenstein BA, Crissman JD et al.
Maintenance bacillus Calmette-Guerin immuno-
therapy for recurrent TA, T1 and carcinoma in
situ transitional cell carcinoma of the bladder: a
randomized Southwest Oncology Group Study. J
Urol 2000; 163: 1124–9.
80 Griffiths TRL, Charlton M, Neal DE, Powell PH.
Treatment of carcinoma in situ with intravesical
bacillus Calmette-Guerin without maintenance. J
Urol 2002; 167: 2408–12.
81 Herr HW, Donat SM, Dalbagni G. Can restaging
transurethral resection of T1 bladder cancer select
patients for immediate cystectomy? J Urol 2007;
177: 75–9.
82 Kulkarni GS, Hakenberg OW, Gschwend JE et al.
An updated critical analysis of the treatment strat-
egy for newly diagnosed high-grade T1 (previously
T1G3) bladder cancer. Eur Urol 2010; 57: 60–70.
83 Denzinger S, Fritsche H-M, Otto W et al. Early
versus deferred cystectomy for initial high-risk
pT1G3 urothelial carcinoma of the bladder: do
risk factors define feasibility of bladder-sparing
approach? Eur Urol 2008; 53: 146–52.
84 Herr HW, Sogani PC. Does early cystectomy
improve the survival of patients with high risk
superficial bladder tumors? J Urol 2001; 166:
1296–9.
85 Samaratunga H, Khoo K. Micropapillary variant of
urothelial carcinoma of the urinary bladder; a clin-
icopathological and immunohistochemical study.
Histopathology 2004; 45: 55–64.
86 Kamat AM, Dinney CP, Gee JR et al. Micropapil-
lary bladder cancer: a review of the University of
Texas M.D. Anderson Cancer Center experience
with 100 consecutive patients. Cancer 2007; 110:
62–7.
87 Palou J, Rodriguez-Rubio F, Huguet J et al. Multi-
variate analysis of clinical parameters of synchro-
nous primary superficial bladder cancer and upper
urinary tract tumour. J Urol 2005; 174: 859–61.
88 Millan-Rodriguez F, Chechile-Toniolo G, Salva-
dor-Bayarri J, Huguet-Perez J, Vicente-Rodriguez
J. Upper urinary tract tumours after primary
superficial bladder tumours: prognostic factors and
risk groups. J Urol 2000; 164: 1183–7.
89 O’Brien T, Ray E, Singh R, Coker B, Beard R,
British Association of Urological Surgeons Section
of Oncology. Prevention of bladder tumours after
nephroureterectomy for primary upper urinary
tract urothelial carcinoma: a prospective, multicen-
tre, randomised clinical trial of a single postopera-
tive dose of mitomycin C (the ODMIT-C trial).
Eur Urol 2011; 60: 703–10.
90 Yates DR, Roupret M. Failure of bacille Calmette-
Guerin in patients with high risk non-muscle-inva-
sive bladder cancer unsuitable for radical cystecto-
my: an update of available treatment options. BJU
Int 2010; 106: 162–7.
91 Shen Z, Shen T, Wientjes MG, O’Donnell MA, Au
JL. Intravesical treatments of bladder cancer:
review. Pharm Res 2008; 25: 1500–10.
92 Lammers RJM, Witjes JA, Inman BA et al. The
role of a combined regimen with intravesical che-
motherapy in the management of non-muscle
invasive bladder cancer: a systematic review. Eur
Urol 2011; 60: 81–93.
93 Witjes JA, Hendricksen K, Gofrit O, Risi O, Nativ
O. Intravesical hyperthermia and mitomycin-C for
carcinoma in situ of the urinary bladder: experi-
ence of the European Synergo working party.
World J Urol 2009; 27: 319–24.
94 Nativ O, Witjes JA, Hendricksen K et al. Com-
bined thermo-chemotherapy for recurrent bladder
cancer after bacillus Calmette-Guerin. J Urol 2009;
182: 1313–7.
95 Di Stasi SM, Giannantoni A, Stephen RL et al. In-
travesical electromotive mitomycin C versus pas-
sive transport mitomycin C for high risk
superficial bladder cancer: a prospective random-
ized study. J Urol 2003; 170: 777–82.
96 Di Stasi SM, Valenti M, Verri C et al. Electromo-
tive instillation of mitomycin immediately before
transurethral resection for patients with primary
urothelial non-muscle invasive bladder cancer: a
randomised controlled trial. Lancet Oncol 2011; 12:
871–9.
97 Birkmeyer NJ, Goodney PP, Stukel TA, Hillner
BE, Birkmeyer JD. Do cancer centres designated
by the National Cancer Institute have better surgi-
cal outcomes? Cancer 2005; 103: 435–41.
98 Horwich A, Dearnaley D, Huddart R et al. A ran-
domized trial of accelerated radiotherapy for local-
ized invasive bladder cancer. Radiother Oncol 2005;
75: 34–43.
99 Ficarra V, Dalpiaz O, Alrabi N, Novara G, Galfano
A, Artibani W. Correlation between clinical and
pathological staging in a series of radical cystecto-
mies for bladder carcinoma. BJU Int 2005; 95:
786–90.
100 Madersbacher S, Hochreiter W, Burkhard F et al.
Radical cystectomy for bladder cancer today – a
homogeneous series without neoadjuvant therapy.
J Clin Oncol 2003; 21: 690–6.
101 Cowan R, McBain C, Ryder WD et al. Radiother-
apy for muscle invasive carcinoma of the bladder:
results of a randomized trial comparing conven-
tional whole bladder with dose-escalated partial
bladder radiotherapy. Int J Radiat Oncol Biol Phys
2004; 59: 197–207.
102 Kotwal S, Choudhury A, Johnston C et al. Similar
treatment outcomes for radical cystectomy and
radical radiotherapy in invasive bladder cancer
treated at a United Kingdom specialist treatment
centre. Int J Radiat Oncol Biol Phys 2008; 70: 456–
63.
103 Addla SK, Naidu P, Maddineni SB, Ramani VA.
Long term survival outcome following radical cys-
tectomy for TCC of the bladder – comparison
between primary and salvage cystectomy. J Urol
2009; 4(Suppl): 633. Abstract 1754.
104 Schuster TG, Smith DC, Montie JE. Pelvic recur-
rences post cystectomy: current treatment strate-
gies. Semin Urol Oncol 2001; 19: 45–50.
105 Stein JP, Skinner DG. Results with radical cystec-
tomy for treating bladder cancer: a ‘reference stan-
dard’ for high grade invasive bladder cancer. BJU
Int 2003; 92: 12–7.
106 Liberman D, Lughezzani G, Sun M et al. Perioper-
ative mortality is significantly greater in septage-
narian and octogenarian patients treated with
radical cystectomy for urothelial cancer of the
bladder. Urology 2011; 77: 660–6.
107 Lowrance WT, Rumohr JA, Chang SS, Smith JA
Jr, Cookson MS. Contemporary open radical cys-
tectomy: analysis of perioperative outcomes. J Urol
2008; 179: 1313–8.
108 Shabsigh A, Korets R, Vora KC et al. Defining
early morbidity of radical cystectomy for patients
with bladder cancer using a standardized reporting
methodology. Eur Urol 2009; 55: 164–74.
109 Herr HW. Extent of surgery and pathology evalua-
tion has an impact on bladder cancer outcomes
after radical cystectomy. Urology 2003; 61: 105–8.
110 Burkhard FC, Roth B, Zehnder P, Studer UE.
Lymphadenectomy for bladder cancer: indications
and controversies. Urol Clin North Am 2011; 38:
397–405.
111 Novara G, Svatek RS, Karakiewicz PI et al. Soft tis-
sue surgical margin status is a powerful predictor
of outcomes after radical radiotherapy: a multicen-
tre study of more than 4400 patients. J Urol 2010;
183: 2165–70.
112 International Collaboration of Trialists, Medical
Research Council Advanced Bladder Cancer Work-
ing Party (now the National Cancer Research
Institute Bladder Cancer Clinical Studies Group),
the European Organisation for Research and
Treatment of Cancer Genito-urinary Tract Cancer
Group et al. International phase III trial assessing
neoadjuvant cisplatin, methotrexate, and vinblas-
tine chemotherapy for muscle invasive bladder
cancer; long-term results of the BA06 30894 Trial.
J Clin Oncol 2011; 29: 2171–7.
113 Advanced Bladder Cancer (ABC) Meta-analysis
collaboration. Neoadjuvant chemotherapy in
Bladder cancer management 447
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448
invasive bladder cancer update of a systematic
review and meta-analysis of individual patient data
advanced bladder cancer (ABC) meta-analysis col-
laboration. Eur Urol 2005; 48: 202–5.
114 Sherif A, Holmberg L, Rintala E et al. Nordic Uro-
thelial Cancer Group Neoadjuvant cisplatinum
based combination chemotherapy in patients with
invasive bladder cancer: a combined analysis of
two Nordic studies. Eur Urol 2004; 45: 297–303.
115 Hoskin PJ, Rojas AM, Bentzen SM, Saunders MI.
Radiotherapy with concurrent carbogen and nico-
tinamide in bladder carcinoma. J Clin Oncol 2010;
28: 4912–8.
116 James ND, Hussain SA, Hall E et al. Radiotherapy
with or without chemotherapy in muscle invasive
bladder cancer. N Engl J Med 2012; 366: 1477–88.
117 Choudhury A, Swindell R, Logue JP et al. Phase II
study of conformal hypofractionated radiotherapy
with concurrent gemcitabine in muscle-invasive
bladder cancer. J Clin Oncol 2011; 29: 733–8.
118 Somani BK, Gimlin D, Fayers P, N’Dow J. Quality
of life and body image for bladder cancer patients
undergoing radical cystectomy and urinary diver-
sion – a prospective cohort study with a systematic
review of literature. Urology 2009; 74: 1138–43.
119 Caffo O, Fellin G, Graffer U, Luciani L. Assess-
ment of quality of life after cystectomy or conser-
vative therapy for patients with infiltrating bladder
carcinoma. A survey by a self-administered ques-
tionnaire. Cancer 1996; 78: 1089–97.
120 Zietman AL, Sacco D, Skowronski U et al. Organ
conservation in invasive bladder cancer by tran-
surethral resection, chemotherapy and radiation:
results of a urodynamic and quality of life study
on long-term survivors. J Urol 2003; 170: 1772–6.
121 von der Maase H, Sengelov L, Roberts JT et al.
Long-term survival results of a randomized trial
comparing gemcitabine plus cisplatin with metho-
trexate, vinblastine, doxorubicin plus cisplatin in
patients with bladder cancer. J Clin Oncol 2005;
23: 4602–8.
122 Zaghloul MS, Boutrus R, El-Hossieny H et al. A
prospective randomized placebo-controlled trial of
zoledronic acid in bony metastatic bladder cancer.
Int J Clin Oncol 2010; 15: 382–9.
Paper received July 2012, accepted October 2012
448 Bladder cancer management
ª 2012 Blackwell Publishing Ltd
Int J Clin Pract, May 2013, 65, 5, 435–448

Mais conteúdo relacionado

Mais procurados

Management of patients with primary colorectal cancer and
Management of patients with primary colorectal cancer andManagement of patients with primary colorectal cancer and
Management of patients with primary colorectal cancer andYuvaraj Karthick
 
Colorectal liver metastases (Dr Juan Carlos Meneu Diaz). Oncocir. Clinica Ruber
Colorectal liver metastases (Dr Juan Carlos Meneu Diaz). Oncocir. Clinica Ruber Colorectal liver metastases (Dr Juan Carlos Meneu Diaz). Oncocir. Clinica Ruber
Colorectal liver metastases (Dr Juan Carlos Meneu Diaz). Oncocir. Clinica Ruber Oncocir (Unidad de Oncología Quirúrgica)
 
PREOPERATIVE DONOR WORKUP FOR LDLT
PREOPERATIVE DONOR WORKUP FOR LDLTPREOPERATIVE DONOR WORKUP FOR LDLT
PREOPERATIVE DONOR WORKUP FOR LDLTDr Amit Dangi
 
Paediatric Hepatocellular carcinoma
Paediatric Hepatocellular carcinomaPaediatric Hepatocellular carcinoma
Paediatric Hepatocellular carcinomaDrArka
 
Colorectal Cancer: Putting Prevention into Practice
Colorectal Cancer: Putting Prevention into PracticeColorectal Cancer: Putting Prevention into Practice
Colorectal Cancer: Putting Prevention into Practicelearfieldinteraction
 
Metastatic colorectal liver cancer
Metastatic colorectal liver cancerMetastatic colorectal liver cancer
Metastatic colorectal liver cancerBashir BnYunus
 
Il colangiocarcinoma: Epidemiologia, Fattori di rischio e patogenesi - Gastro...
Il colangiocarcinoma: Epidemiologia, Fattori di rischio e patogenesi - Gastro...Il colangiocarcinoma: Epidemiologia, Fattori di rischio e patogenesi - Gastro...
Il colangiocarcinoma: Epidemiologia, Fattori di rischio e patogenesi - Gastro...Gastrolearning
 
Gastric Cancer Update - 2016
Gastric Cancer Update - 2016Gastric Cancer Update - 2016
Gastric Cancer Update - 2016Mohamed Abdulla
 
Pancreatic Cancer Are We Moving Forward Yet
Pancreatic Cancer Are We Moving Forward YetPancreatic Cancer Are We Moving Forward Yet
Pancreatic Cancer Are We Moving Forward Yetfondas vakalis
 
Immunotherapy and Recurrent Ovarian Cancer: Time for New Paradigms!
Immunotherapy and Recurrent Ovarian Cancer: Time for New Paradigms!Immunotherapy and Recurrent Ovarian Cancer: Time for New Paradigms!
Immunotherapy and Recurrent Ovarian Cancer: Time for New Paradigms!National Ovarian Cancer Coalition
 
Initial chemotherapy and radiation for pancreatic cancer
Initial chemotherapy and radiation for pancreatic cancerInitial chemotherapy and radiation for pancreatic cancer
Initial chemotherapy and radiation for pancreatic cancerDr Tauqeer A Siddiqui MD FACP
 
Management of colorectal liver metastasis
Management of colorectal liver metastasis Management of colorectal liver metastasis
Management of colorectal liver metastasis Aditya Punamiya
 
Renal Cell Carcinoma Risk Stratification
Renal Cell Carcinoma Risk StratificationRenal Cell Carcinoma Risk Stratification
Renal Cell Carcinoma Risk StratificationDr.Bhavin Vadodariya
 
Pancreatic cancer Management (pancreatic adenocarcinoma)
Pancreatic cancer Management (pancreatic adenocarcinoma)Pancreatic cancer Management (pancreatic adenocarcinoma)
Pancreatic cancer Management (pancreatic adenocarcinoma)Dr mohamed Salat Gonjobe
 
Management of hcc sneha
Management of hcc snehaManagement of hcc sneha
Management of hcc snehaSneha George
 
Advances in cholangiocarcinoma
Advances in cholangiocarcinomaAdvances in cholangiocarcinoma
Advances in cholangiocarcinomaspa718
 
Staging and Surgical Management of Pancreatiic Cancer
Staging and Surgical Management of Pancreatiic CancerStaging and Surgical Management of Pancreatiic Cancer
Staging and Surgical Management of Pancreatiic Canceru.surgery
 
Barcelona clinic liver cancer (bclc) staging
Barcelona clinic liver cancer (bclc) stagingBarcelona clinic liver cancer (bclc) staging
Barcelona clinic liver cancer (bclc) stagingAbhilash Cheriyan
 

Mais procurados (20)

Cytoreductive nephrectomy
Cytoreductive nephrectomyCytoreductive nephrectomy
Cytoreductive nephrectomy
 
Breaking the Paradox: Expanding Options and New Questions in HCC Management—M...
Breaking the Paradox: Expanding Options and New Questions in HCC Management—M...Breaking the Paradox: Expanding Options and New Questions in HCC Management—M...
Breaking the Paradox: Expanding Options and New Questions in HCC Management—M...
 
Management of patients with primary colorectal cancer and
Management of patients with primary colorectal cancer andManagement of patients with primary colorectal cancer and
Management of patients with primary colorectal cancer and
 
Colorectal liver metastases (Dr Juan Carlos Meneu Diaz). Oncocir. Clinica Ruber
Colorectal liver metastases (Dr Juan Carlos Meneu Diaz). Oncocir. Clinica Ruber Colorectal liver metastases (Dr Juan Carlos Meneu Diaz). Oncocir. Clinica Ruber
Colorectal liver metastases (Dr Juan Carlos Meneu Diaz). Oncocir. Clinica Ruber
 
PREOPERATIVE DONOR WORKUP FOR LDLT
PREOPERATIVE DONOR WORKUP FOR LDLTPREOPERATIVE DONOR WORKUP FOR LDLT
PREOPERATIVE DONOR WORKUP FOR LDLT
 
Paediatric Hepatocellular carcinoma
Paediatric Hepatocellular carcinomaPaediatric Hepatocellular carcinoma
Paediatric Hepatocellular carcinoma
 
Colorectal Cancer: Putting Prevention into Practice
Colorectal Cancer: Putting Prevention into PracticeColorectal Cancer: Putting Prevention into Practice
Colorectal Cancer: Putting Prevention into Practice
 
Metastatic colorectal liver cancer
Metastatic colorectal liver cancerMetastatic colorectal liver cancer
Metastatic colorectal liver cancer
 
Il colangiocarcinoma: Epidemiologia, Fattori di rischio e patogenesi - Gastro...
Il colangiocarcinoma: Epidemiologia, Fattori di rischio e patogenesi - Gastro...Il colangiocarcinoma: Epidemiologia, Fattori di rischio e patogenesi - Gastro...
Il colangiocarcinoma: Epidemiologia, Fattori di rischio e patogenesi - Gastro...
 
Gastric Cancer Update - 2016
Gastric Cancer Update - 2016Gastric Cancer Update - 2016
Gastric Cancer Update - 2016
 
Pancreatic Cancer Are We Moving Forward Yet
Pancreatic Cancer Are We Moving Forward YetPancreatic Cancer Are We Moving Forward Yet
Pancreatic Cancer Are We Moving Forward Yet
 
Immunotherapy and Recurrent Ovarian Cancer: Time for New Paradigms!
Immunotherapy and Recurrent Ovarian Cancer: Time for New Paradigms!Immunotherapy and Recurrent Ovarian Cancer: Time for New Paradigms!
Immunotherapy and Recurrent Ovarian Cancer: Time for New Paradigms!
 
Initial chemotherapy and radiation for pancreatic cancer
Initial chemotherapy and radiation for pancreatic cancerInitial chemotherapy and radiation for pancreatic cancer
Initial chemotherapy and radiation for pancreatic cancer
 
Management of colorectal liver metastasis
Management of colorectal liver metastasis Management of colorectal liver metastasis
Management of colorectal liver metastasis
 
Renal Cell Carcinoma Risk Stratification
Renal Cell Carcinoma Risk StratificationRenal Cell Carcinoma Risk Stratification
Renal Cell Carcinoma Risk Stratification
 
Pancreatic cancer Management (pancreatic adenocarcinoma)
Pancreatic cancer Management (pancreatic adenocarcinoma)Pancreatic cancer Management (pancreatic adenocarcinoma)
Pancreatic cancer Management (pancreatic adenocarcinoma)
 
Management of hcc sneha
Management of hcc snehaManagement of hcc sneha
Management of hcc sneha
 
Advances in cholangiocarcinoma
Advances in cholangiocarcinomaAdvances in cholangiocarcinoma
Advances in cholangiocarcinoma
 
Staging and Surgical Management of Pancreatiic Cancer
Staging and Surgical Management of Pancreatiic CancerStaging and Surgical Management of Pancreatiic Cancer
Staging and Surgical Management of Pancreatiic Cancer
 
Barcelona clinic liver cancer (bclc) staging
Barcelona clinic liver cancer (bclc) stagingBarcelona clinic liver cancer (bclc) staging
Barcelona clinic liver cancer (bclc) staging
 

Destaque

Laser Therapy Applications for Osteoarthritis and Chronic Joint Pain Clinic...
Laser Therapy Applications for Osteoarthritis and Chronic Joint Pain   Clinic...Laser Therapy Applications for Osteoarthritis and Chronic Joint Pain   Clinic...
Laser Therapy Applications for Osteoarthritis and Chronic Joint Pain Clinic...Theralase Technologies Inc.
 
Efficacy of super pulsed 905 nm low level laser therapy (lllt) in the managem...
Efficacy of super pulsed 905 nm low level laser therapy (lllt) in the managem...Efficacy of super pulsed 905 nm low level laser therapy (lllt) in the managem...
Efficacy of super pulsed 905 nm low level laser therapy (lllt) in the managem...Theralase Technologies Inc.
 
Spree 2013 : Marketing Brochure
Spree 2013 : Marketing BrochureSpree 2013 : Marketing Brochure
Spree 2013 : Marketing BrochureNeil Merchant
 
презентація до уроку
презентація до урокупрезентація до уроку
презентація до урокуMarina Chmeruk
 
Earthquacke Elastic Rebound Theory Types of Waves
Earthquacke Elastic Rebound Theory  Types of WavesEarthquacke Elastic Rebound Theory  Types of Waves
Earthquacke Elastic Rebound Theory Types of WavesShakeel Ahmad
 

Destaque (12)

A Renaissance in Low Level Laser (light) lllt
A Renaissance in Low Level Laser (light) llltA Renaissance in Low Level Laser (light) lllt
A Renaissance in Low Level Laser (light) lllt
 
Theralase Investment Opportunity Aug 2013
Theralase Investment Opportunity Aug 2013Theralase Investment Opportunity Aug 2013
Theralase Investment Opportunity Aug 2013
 
Low Level Laser Therapy Class 3b vs Class 4
Low Level Laser Therapy Class 3b vs Class 4Low Level Laser Therapy Class 3b vs Class 4
Low Level Laser Therapy Class 3b vs Class 4
 
Laser Therapy Applications for Osteoarthritis and Chronic Joint Pain Clinic...
Laser Therapy Applications for Osteoarthritis and Chronic Joint Pain   Clinic...Laser Therapy Applications for Osteoarthritis and Chronic Joint Pain   Clinic...
Laser Therapy Applications for Osteoarthritis and Chronic Joint Pain Clinic...
 
Efficacy of super pulsed 905 nm low level laser therapy (lllt) in the managem...
Efficacy of super pulsed 905 nm low level laser therapy (lllt) in the managem...Efficacy of super pulsed 905 nm low level laser therapy (lllt) in the managem...
Efficacy of super pulsed 905 nm low level laser therapy (lllt) in the managem...
 
Theralase's white paper 04 10-11 pdf final
Theralase's white paper 04 10-11 pdf finalTheralase's white paper 04 10-11 pdf final
Theralase's white paper 04 10-11 pdf final
 
Theralase's White Paper Oct 2011
Theralase's White Paper Oct 2011Theralase's White Paper Oct 2011
Theralase's White Paper Oct 2011
 
Theralase Photodynamic Compounds Destroy Cancer
Theralase Photodynamic Compounds Destroy CancerTheralase Photodynamic Compounds Destroy Cancer
Theralase Photodynamic Compounds Destroy Cancer
 
narrative essay
narrative essaynarrative essay
narrative essay
 
Spree 2013 : Marketing Brochure
Spree 2013 : Marketing BrochureSpree 2013 : Marketing Brochure
Spree 2013 : Marketing Brochure
 
презентація до уроку
презентація до урокупрезентація до уроку
презентація до уроку
 
Earthquacke Elastic Rebound Theory Types of Waves
Earthquacke Elastic Rebound Theory  Types of WavesEarthquacke Elastic Rebound Theory  Types of Waves
Earthquacke Elastic Rebound Theory Types of Waves
 

Semelhante a Current perspectives on bladder cancer diagnosis and management

Endoscopy in Gastrointestinal Oncology - Slide 15 - D. Fisher - Colorectal ca...
Endoscopy in Gastrointestinal Oncology - Slide 15 - D. Fisher - Colorectal ca...Endoscopy in Gastrointestinal Oncology - Slide 15 - D. Fisher - Colorectal ca...
Endoscopy in Gastrointestinal Oncology - Slide 15 - D. Fisher - Colorectal ca...European School of Oncology
 
Management of HCC, an update
Management of HCC, an updateManagement of HCC, an update
Management of HCC, an updateMohammed A Suwaid
 
Annals of Clinical and Medical Case Reports - Acmcasereport
Annals of Clinical and Medical Case Reports - AcmcasereportAnnals of Clinical and Medical Case Reports - Acmcasereport
Annals of Clinical and Medical Case Reports - Acmcasereportsemualkaira
 
HCC: clinical update AASLD based
HCC: clinical update AASLD basedHCC: clinical update AASLD based
HCC: clinical update AASLD basedAhmed Elmoughazy
 
Liver Neoplasms
Liver   NeoplasmsLiver   Neoplasms
Liver NeoplasmsDeep Deep
 
1120_Reflections_Volume34_Winter2014_111214
1120_Reflections_Volume34_Winter2014_1112141120_Reflections_Volume34_Winter2014_111214
1120_Reflections_Volume34_Winter2014_111214H Henly
 
Primary%20and%20Secondary%20malignant%20conditions%20of%20Liver,.pptx
Primary%20and%20Secondary%20malignant%20conditions%20of%20Liver,.pptxPrimary%20and%20Secondary%20malignant%20conditions%20of%20Liver,.pptx
Primary%20and%20Secondary%20malignant%20conditions%20of%20Liver,.pptxAmandeepSingh952
 
O. Glehen - HIPEC Colorectal and Gastric
O. Glehen - HIPEC Colorectal and GastricO. Glehen - HIPEC Colorectal and Gastric
O. Glehen - HIPEC Colorectal and GastricGlehen
 
Management of malignant pleural effusion ...
 Management  of malignant pleural effusion                                   ... Management  of malignant pleural effusion                                   ...
Management of malignant pleural effusion ...Ashraf ElAdawy
 
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGS
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGSLUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGS
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGSKanhu Charan
 
Staging and investigation of ca kidney and bladder
Staging and investigation of ca kidney and bladderStaging and investigation of ca kidney and bladder
Staging and investigation of ca kidney and bladderAtulGupta369
 
carcinoma urinary bladder management
carcinoma urinary bladder management carcinoma urinary bladder management
carcinoma urinary bladder management Isha Jaiswal
 
O. Glehen - HIPEC in colorectal carcinomatosis
O. Glehen - HIPEC in colorectal carcinomatosisO. Glehen - HIPEC in colorectal carcinomatosis
O. Glehen - HIPEC in colorectal carcinomatosisGlehen
 
Post radical nephroureterectomy cystoscopic surveillance and usage of a nomog...
Post radical nephroureterectomy cystoscopic surveillance and usage of a nomog...Post radical nephroureterectomy cystoscopic surveillance and usage of a nomog...
Post radical nephroureterectomy cystoscopic surveillance and usage of a nomog...NAAR Journal
 
Liver cancer by Dr. Basil Tumaini
Liver cancer by Dr. Basil TumainiLiver cancer by Dr. Basil Tumaini
Liver cancer by Dr. Basil TumainiBasil Tumaini
 
Bladder cancer and its management
Bladder cancer and its managementBladder cancer and its management
Bladder cancer and its managementShweta Sharma
 

Semelhante a Current perspectives on bladder cancer diagnosis and management (20)

Endoscopy in Gastrointestinal Oncology - Slide 15 - D. Fisher - Colorectal ca...
Endoscopy in Gastrointestinal Oncology - Slide 15 - D. Fisher - Colorectal ca...Endoscopy in Gastrointestinal Oncology - Slide 15 - D. Fisher - Colorectal ca...
Endoscopy in Gastrointestinal Oncology - Slide 15 - D. Fisher - Colorectal ca...
 
Management of HCC, an update
Management of HCC, an updateManagement of HCC, an update
Management of HCC, an update
 
IJET-V3I2P22
IJET-V3I2P22IJET-V3I2P22
IJET-V3I2P22
 
Annals of Clinical and Medical Case Reports - Acmcasereport
Annals of Clinical and Medical Case Reports - AcmcasereportAnnals of Clinical and Medical Case Reports - Acmcasereport
Annals of Clinical and Medical Case Reports - Acmcasereport
 
HCC: clinical update AASLD based
HCC: clinical update AASLD basedHCC: clinical update AASLD based
HCC: clinical update AASLD based
 
Liver Neoplasms
Liver   NeoplasmsLiver   Neoplasms
Liver Neoplasms
 
1120_Reflections_Volume34_Winter2014_111214
1120_Reflections_Volume34_Winter2014_1112141120_Reflections_Volume34_Winter2014_111214
1120_Reflections_Volume34_Winter2014_111214
 
Ca urinary bladder management
Ca urinary bladder managementCa urinary bladder management
Ca urinary bladder management
 
Pancreatic Cancer.pptx
Pancreatic Cancer.pptxPancreatic Cancer.pptx
Pancreatic Cancer.pptx
 
Primary%20and%20Secondary%20malignant%20conditions%20of%20Liver,.pptx
Primary%20and%20Secondary%20malignant%20conditions%20of%20Liver,.pptxPrimary%20and%20Secondary%20malignant%20conditions%20of%20Liver,.pptx
Primary%20and%20Secondary%20malignant%20conditions%20of%20Liver,.pptx
 
O. Glehen - HIPEC Colorectal and Gastric
O. Glehen - HIPEC Colorectal and GastricO. Glehen - HIPEC Colorectal and Gastric
O. Glehen - HIPEC Colorectal and Gastric
 
Management of malignant pleural effusion ...
 Management  of malignant pleural effusion                                   ... Management  of malignant pleural effusion                                   ...
Management of malignant pleural effusion ...
 
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGS
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGSLUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGS
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGS
 
Staging and investigation of ca kidney and bladder
Staging and investigation of ca kidney and bladderStaging and investigation of ca kidney and bladder
Staging and investigation of ca kidney and bladder
 
carcinoma urinary bladder management
carcinoma urinary bladder management carcinoma urinary bladder management
carcinoma urinary bladder management
 
Ovarian Cancer Research and Prevention, Andrew Berchuck, MD
Ovarian Cancer Research and Prevention, Andrew Berchuck, MDOvarian Cancer Research and Prevention, Andrew Berchuck, MD
Ovarian Cancer Research and Prevention, Andrew Berchuck, MD
 
O. Glehen - HIPEC in colorectal carcinomatosis
O. Glehen - HIPEC in colorectal carcinomatosisO. Glehen - HIPEC in colorectal carcinomatosis
O. Glehen - HIPEC in colorectal carcinomatosis
 
Post radical nephroureterectomy cystoscopic surveillance and usage of a nomog...
Post radical nephroureterectomy cystoscopic surveillance and usage of a nomog...Post radical nephroureterectomy cystoscopic surveillance and usage of a nomog...
Post radical nephroureterectomy cystoscopic surveillance and usage of a nomog...
 
Liver cancer by Dr. Basil Tumaini
Liver cancer by Dr. Basil TumainiLiver cancer by Dr. Basil Tumaini
Liver cancer by Dr. Basil Tumaini
 
Bladder cancer and its management
Bladder cancer and its managementBladder cancer and its management
Bladder cancer and its management
 

Mais de Theralase Technologies Inc.

Theralase Expands Board of Directors with Strong Financing Capabilities
Theralase Expands Board of Directors with Strong Financing CapabilitiesTheralase Expands Board of Directors with Strong Financing Capabilities
Theralase Expands Board of Directors with Strong Financing CapabilitiesTheralase Technologies Inc.
 
Efficacy of super pulsed 905 nm low level laser therapy in the management of ...
Efficacy of super pulsed 905 nm low level laser therapy in the management of ...Efficacy of super pulsed 905 nm low level laser therapy in the management of ...
Efficacy of super pulsed 905 nm low level laser therapy in the management of ...Theralase Technologies Inc.
 
Theralase photo dynamic compounds effective in destruction of drug resistant ...
Theralase photo dynamic compounds effective in destruction of drug resistant ...Theralase photo dynamic compounds effective in destruction of drug resistant ...
Theralase photo dynamic compounds effective in destruction of drug resistant ...Theralase Technologies Inc.
 
Theralase photo dynamic compounds effective in destruction of drug resistant ...
Theralase photo dynamic compounds effective in destruction of drug resistant ...Theralase photo dynamic compounds effective in destruction of drug resistant ...
Theralase photo dynamic compounds effective in destruction of drug resistant ...Theralase Technologies Inc.
 
The Science of Laser Therapy and Clinical Applications
The Science of Laser Therapy and Clinical Applications The Science of Laser Therapy and Clinical Applications
The Science of Laser Therapy and Clinical Applications Theralase Technologies Inc.
 
Peer reviewed research of low level laser therapy
Peer reviewed research of low level laser therapyPeer reviewed research of low level laser therapy
Peer reviewed research of low level laser therapyTheralase Technologies Inc.
 

Mais de Theralase Technologies Inc. (20)

Theralase Inc. Executive Summary 2013
Theralase Inc. Executive Summary 2013Theralase Inc. Executive Summary 2013
Theralase Inc. Executive Summary 2013
 
2012 Year End Results
2012 Year End Results2012 Year End Results
2012 Year End Results
 
Theralase's unique value proposition
Theralase's unique value propositionTheralase's unique value proposition
Theralase's unique value proposition
 
Theralase Expands Board of Directors with Strong Financing Capabilities
Theralase Expands Board of Directors with Strong Financing CapabilitiesTheralase Expands Board of Directors with Strong Financing Capabilities
Theralase Expands Board of Directors with Strong Financing Capabilities
 
The Science of Therapeutic Laser Technology
The Science of Therapeutic Laser TechnologyThe Science of Therapeutic Laser Technology
The Science of Therapeutic Laser Technology
 
Efficacy of super pulsed 905 nm low level laser therapy in the management of ...
Efficacy of super pulsed 905 nm low level laser therapy in the management of ...Efficacy of super pulsed 905 nm low level laser therapy in the management of ...
Efficacy of super pulsed 905 nm low level laser therapy in the management of ...
 
Cold Laser Therapy - Companion book
Cold Laser Therapy - Companion bookCold Laser Therapy - Companion book
Cold Laser Therapy - Companion book
 
Equine Book _Therapeutic Laser
Equine Book _Therapeutic LaserEquine Book _Therapeutic Laser
Equine Book _Therapeutic Laser
 
Cold Laser Therapy - The Human Book
Cold Laser Therapy - The Human BookCold Laser Therapy - The Human Book
Cold Laser Therapy - The Human Book
 
Theralase Compounds Destroy Bladder Cancer
Theralase Compounds Destroy Bladder CancerTheralase Compounds Destroy Bladder Cancer
Theralase Compounds Destroy Bladder Cancer
 
Theralase photo dynamic compounds effective in destruction of drug resistant ...
Theralase photo dynamic compounds effective in destruction of drug resistant ...Theralase photo dynamic compounds effective in destruction of drug resistant ...
Theralase photo dynamic compounds effective in destruction of drug resistant ...
 
Theralase photo dynamic compounds effective in destruction of drug resistant ...
Theralase photo dynamic compounds effective in destruction of drug resistant ...Theralase photo dynamic compounds effective in destruction of drug resistant ...
Theralase photo dynamic compounds effective in destruction of drug resistant ...
 
Meta analysis of pain relief effects with laser
Meta analysis of pain relief effects with laserMeta analysis of pain relief effects with laser
Meta analysis of pain relief effects with laser
 
Theralase commences equity financing oct 2012
Theralase commences equity financing oct 2012Theralase commences equity financing oct 2012
Theralase commences equity financing oct 2012
 
Theralase expands into china rw - 10 01 2012
Theralase expands into china   rw - 10 01 2012Theralase expands into china   rw - 10 01 2012
Theralase expands into china rw - 10 01 2012
 
Osteoarthritis clinical trial
Osteoarthritis clinical trialOsteoarthritis clinical trial
Osteoarthritis clinical trial
 
Newsletter comp
Newsletter compNewsletter comp
Newsletter comp
 
The Science of Laser Therapy and Clinical Applications
The Science of Laser Therapy and Clinical Applications The Science of Laser Therapy and Clinical Applications
The Science of Laser Therapy and Clinical Applications
 
2011 year end_results_ver_4
2011 year end_results_ver_42011 year end_results_ver_4
2011 year end_results_ver_4
 
Peer reviewed research of low level laser therapy
Peer reviewed research of low level laser therapyPeer reviewed research of low level laser therapy
Peer reviewed research of low level laser therapy
 

Último

Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Premium Bangalore Call Girls Jigani Dail 6378878445 Escort Service For Hot Ma...
Premium Bangalore Call Girls Jigani Dail 6378878445 Escort Service For Hot Ma...Premium Bangalore Call Girls Jigani Dail 6378878445 Escort Service For Hot Ma...
Premium Bangalore Call Girls Jigani Dail 6378878445 Escort Service For Hot Ma...tanya dube
 
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...Genuine Call Girls
 
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...chandars293
 
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...astropune
 
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...Taniya Sharma
 
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...vidya singh
 
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋TANUJA PANDEY
 
Premium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort Service
Premium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort ServicePremium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort Service
Premium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort Servicevidya singh
 
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟ 9332606886 ⟟ Call Me For G...
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟  9332606886 ⟟ Call Me For G...Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟  9332606886 ⟟ Call Me For G...
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟ 9332606886 ⟟ Call Me For G...narwatsonia7
 
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...Taniya Sharma
 

Último (20)

Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
 
Premium Bangalore Call Girls Jigani Dail 6378878445 Escort Service For Hot Ma...
Premium Bangalore Call Girls Jigani Dail 6378878445 Escort Service For Hot Ma...Premium Bangalore Call Girls Jigani Dail 6378878445 Escort Service For Hot Ma...
Premium Bangalore Call Girls Jigani Dail 6378878445 Escort Service For Hot Ma...
 
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...
 
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
 
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
 
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
 
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
 
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
 
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
 
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
 
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
 
Premium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort Service
Premium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort ServicePremium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort Service
Premium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort Service
 
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
 
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟ 9332606886 ⟟ Call Me For G...
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟  9332606886 ⟟ Call Me For G...Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟  9332606886 ⟟ Call Me For G...
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟ 9332606886 ⟟ Call Me For G...
 
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
 

Current perspectives on bladder cancer diagnosis and management

  • 1. Current perspectives in bladder cancer management T. R. L. Griffiths, on behalf of Action on Bladder Cancer Methods The rationale for this review of the literature was to discuss the latest evidence-base and to summarise key messages for the prevention, investigation, diag- nosis and management of bladder cancer for non- specialists and specialists. The literature searches were conducted using the timeframe of February 2012 to June 2012. The data- bases searched included PubMed and Trip, congress abstracts [European Association of Urology (EAU)] and current guidelines ⁄ consensus statements from within Europe [EAU; National Institute for Health and Clinical Excellence (NICE); The Renal Associa- tion; British Association of Urological Surgeons (BAUS)]. Key words used for the searches were: bladder can- cer; risk factors; epidemiology; histopathology; diag- nosis; haematuria; intravesical chemotherapy; BCG; transitional cell carcinoma; cystoscopy; transurethral resection; cystectomy; radiotherapy; systemic chemo- therapy. Introduction Table 1 summarises the key points concerning the diagnosis and management of bladder cancer for the non-specialist healthcare professional. Approximately 386,000 new bladder tumours were diagnosed worldwide in 2008, making bladder cancer the ninth most common cancer (1). In the UK, blad- der cancer is the fourth and twelfth most common cancer in men and women respectively (2); it is up to three times more common in men than women, but women tend to present with more advanced disease and have worse outcomes (3). Around eight in 10 cases are diagnosed in individuals over the age of 65 (2), and Caucasians generally have a higher risk of bladder cancer than people of other ethnicities (4). In the USA, bladder cancer mortality in men decreased from the mid-1970s to the mid-1980s and was rela- tively stable to 2006; in women, there was a small, but steady decrease between 1975 and 2006 (4). Within Europe, bladder cancer mortality was stable in women between 1980 and 2006, but consistently decreased in SUMMARY More than 350,000 new cases of bladder cancer are diagnosed worldwide each year; the vast majority (> 90%) of these are transitional cell carcinomas (TCC). The most important risk factors for the development of bladder cancer are smoking and occupational exposure to toxic chemicals. Painless visible haematuria is the most common presenting symptom of bladder cancer; significant haematuria requires referral to a specialist urology service. Cystoscopy and urine cytology are currently the recommended tools for diagnosis of bladder cancer. Excluding muscle invasion is an important diagnostic step, as outcomes for patients with muscle invasive TCC are less favourable. For non-muscle invasive bladder cancer, transure- thral resection followed by intravesical chemotherapy (typically Mitomycin C or epi- rubicin) or immunotherapy [bacillus Calmette-Gue´rin (BCG)] is the current standard of care. For patients failing BCG therapy, cystectomy is recommended; for patients unsuitable for surgery, the choice of treatment options is currently limited. How- ever, novel interventions, such as chemohyperthermia and electromotive drug administration, enhance the effects of conventional chemotherapeutic agents and are being evaluated in Phase III trials. Radical cystectomy (with pelvic lymphaden- ectomy and urinary diversion) or radical radiotherapy are the current established treatments for muscle invasive TCC. Neoadjuvant chemotherapy is recommended before definitive treatment of muscle invasive TCC; cisplatin-containing combina- tion chemotherapy is the recommended regimen. Palliative chemotherapy is the first-choice treatment in metastatic TCC. Review criteria To produce this non-systematic review, an extensive literature search was performed with the objective of identifying publications concerning the management of bladder cancer. Multiple sources were included, e.g. current European guidelines, National Institute for Health and Clinical Excellence guidelines, meta-analyses, randomised controlled trials (RCT), consensus statements by the Renal Association and British Association of Urological Surgeons and non-randomised study evidence where RCT evidence was not available. Message for the clinic Patients with a single episode of visible haematuria (VH) and any patients with clinically relevant non- visible haematuria (NVH) (defined in Table 1) need prompt investigation and urological referral. Symptoms attributable to bladder cancer can mimic urinary tract infection (UTI). Patients with a symptomatic UTI refractory to antibiotics or an early recurrent UTI warrant flexible cystoscopic evaluation before considering repeat courses of antibiotics. University Hospitals of Leicester NHS Trust, Clinical Sciences Unit, Leicester General Hospital, Leicester, UK Correspondence to: T. R. Leyshon Griffiths, University Hospitals of Leicester NHS Trust, Clinical Sciences Unit, Leicester General Hospital, Gwendolen Road, Leicester LE5 4PW, UK Tel.: +44 (0)1162 584617 Fax: +44 (0)1162 584617 Email: trlg1@le.ac.uk Disclosures Over the past year, I have received an honorarium from Alliance Pharma for advice on creating a health-economic model for intravesical BCG. Over the past 5 years, I have received honoraria from Alliance Pharma and GE Healthcare for involvement in educational events including provision of lectures. GE Healthcare used to market hexvixÒ for use in photodynamic diagnosis of bladder cancer in the UK, but this is no longer the case. Charity Registration Number: 1138532 REVIEW ARTICLE ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448. doi: 10.1111/ijcp.12075 435
  • 2. Table 1 Key points for the diagnosis and management of bladder cancer Pathology The vast majority of bladder cancers (> 90%) are TCC in origin Risk factors Smoking and occupational exposure to toxic chemicals are the key risk factors for bladder cancer Smoking cessation is still worthwhile following diagnosis of bladder cancer Definitions of significant haematuria Painless VH is the most common presenting symptom of bladder cancer NVH: ‡ 1 + blood on dipstick testing; not necessary to confirm NVH using urine microscopy; a trace of blood only on dipstick is not regarded as significant. Persistent a-NVH: Defined in the UK as two out of three dipsticks ‡ 1 + blood. In contrast, the American Urological Association (AUA) defines a-NVH as ‡ 3 red blood cells per high powered field in a properly collected urine specimen. Clinically relevant haematuria: any episode of painless VH; any single episode of painful VH or s-NVH in the absence of UTI; recurrent or refractory UTI with VH, or persistent a-NVH (requires urgent investigation) Transient NVH or spurious causes of haematuria need to be excluded Investigations within primary care All patients with s-NVH and persistent a-NVH should have their baseline blood pressure, eGFR and ACR measured Further nephrological assessment should be considered in those aged < 40 years with persistent a-NVH if any one of the following are present: eGFR < 60 ml ⁄ min, ACR > 30 or blood pressure > 140 ⁄ 90 mmHg Referral to specialist urology service Routine referral: age 40–50 years with a-NVH or aged < 50 years with s-NVH Urgent referral: age > 50 years with a-NVH or s-NVH, or age > 40 years with recurrent or persistent UTI associated with haematuria Tools to facilitate the diagnosis of bladder cancer Cystoscopy and urine cytology are currently recommended tools Fluorescence cystoscopy (photodynamic diagnosis) is most useful for detection of CIS and guiding biopsies in patients with positive cytology or a history of high-grade NMIBC Narrow-band imaging cystoscopy has shown promise as an aid to facilitate detection without the need for an intravesical photosensitiser, but needs further evaluation in randomised trials Although currently available novel urinary biomarkers have higher sensitivity than cytology, they are not routinely recommended because of their higher false-positive rates Non-muscle invasive TCC Excluding muscle invasion is an important diagnostic step; one-half of patients with muscle invasive TCC will die of bladder cancer within 5 years EORTC risk group should be determined Most patients with NMIBC undergo surveillance cystoscopies and intravesical treatments Quality of first TUR can affect outcomes (tumour-free detrusor muscle ⁄ residual disease) A single postoperative intravesical instillation of chemotherapy within 24 h of TUR is recommended for all newly diagnosed bladder tumours The choice between further intravesical chemotherapy or immunotherapy is guided by the EORTC risk group Immediate radical cystectomy should be considered for patients whose risk of tumour progression is especially high, concomitant superficial urethral TCC is present, or where they request it in preference to intravesical BCG Treatment with intravesical BCG is considered to have failed if: T1 disease persists at the 3-month check cystoscopy following induction BCG; high-grade Ta disease ⁄ CIS is present at 6 months; or muscle invasive TCC is detected The incidence of concomitant UUT-TCC is low, but is increased in patients with trigonal tumours Treatment after BCG failure Radical cystectomy remains the mainstay of treatment for patients who have failed BCG treatment For patients who are unwilling or unfit to have a radical cystectomy after BCG failure, the treatment options are limited Currently available bladder-sparing treatments for those with BCG-refractory TCC are associated with 2-year disease-free survival of approximately 50% Treatments with the most promising clinical data, such as chemohyperthermia, are now being evaluated in Phase III trials Muscle invasive TCC Radical cystectomy (with pelvic lymphadenectomy and urinary diversion) or radical radiotherapy are the current established treatments for muscle invasive TCC Neoadjuvant chemotherapy is recommended before definitive treatment of muscle invasive TCC; cisplatin-containing combination chemotherapy is the recommended regimen Chemotherapy concurrent with radiotherapy improves loco-regional control compared with radiotherapy alone and confers separate benefits to neoadjuvant chemotherapy Advanced ⁄ metastatic TCC Palliative chemotherapy is the first-choice treatment in metastatic TCC ACR, albumin ⁄ creatinine ratio; BCG, bacillus Calmette-Gue´rin; CIS, carcinoma in situ; eGFR, estimated glomerular filtration rate; EORTC, European Organization for Research and Treatment of Cancer; NMIBC, non-muscle invasive bladder cancer; NVH, non-visible haematuria; TCC, transitional cell carcinoma; TUR, transurethral resection; UTI, urinary tract infection; UUT, upper urinary tract; VH, visible haematuria. 436 Bladder cancer management ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 3. men from around 1990 onwards (5). The bladder can- cer mortality rate in Japan has been stable for men since the early 1990s and has risen slightly during this period in women (6). Putative reasons for reduced mortality include reduced exposure to tobacco and occupational carcinogens. Transitional cell carcinoma (TCC; also known as urothelial cancer) represents over 90% of bladder cancers (7) and is consequently the focus of this review. Less common types include squamous cell carcinoma, adenocarcinoma and small cell carci- noma. Around 70–85% of TCCs are superficial [stages Ta, T1, carcinoma in situ (CIS)] at presenta- tion and are now commonly termed non-muscle invasive bladder cancer (NMIBC) (7–10). Unlike in Europe or the USA, malignant non-muscle invasive tumours that also do not invade the lamina propria (Ta, CIS) are omitted from cancer registry statistics in England and Wales, therefore interpretation of epidemiological statistics varies across the world. However, in the management of bladder cancer, excluding muscle invasion is an important diagnostic step, as outcomes for patients with muscle invasive TCC (T2–T4) are less favourable; nearly one-half will die from bladder cancer within 5 years of diagnosis (8,10). In the Middle East and Africa, squamous cell carcinoma was typically more common than in the developed world, largely caused by Schistosoma hae- matobium infection. However, improved knowledge of schistosomiasis over the last three decades has led to a reduction in squamous cell carcinoma such that TCC is now also the predominant type of bladder cancer in these countries (11,12). Risk factors for bladder cancer Numerous risk factors for TCC have been identified. Smoking is implicated in approximately two-thirds of bladder cancers in men and up to one-third in women (13). There is a fourfold increased incidence in current smokers relative to never-smokers (14), and bladder cancer risk is correlated with the num- ber of cigarettes smoked, duration of smoking and age at smoking initiation (14–16). Those who stop smoking reduce their bladder cancer risk by 10–40% within 4 years (14,17), although former smokers retain a twofold higher incidence than never-smokers (14,16). Data from Western Europe suggest that around 4– 7% of bladder cancers in men are attributable to a known occupational carcinogen; the latent period between exposure and the development of cancer is about 20 years (18). Exposure to aniline dyes, aro- matic amines (used in the manufacture of textiles, paints, plastics and rubber industries) and polycyclic aromatic amines are the primary toxins. Continuous arsenic exposure and ingestion has been reported to increase the risk of bladder cancer by as much as one thousand times (19). Carcinogen-metabolising enzymes are in part con- trolled by genetic polymorphism. Slow acetylation of N-acetyltransferase-2 (20,21), rapid cytochrome P1A2 activity (22) and glutathione S-transferase M1 null genotype (21) are associated with an increased risk of TCC. Approximately 20% of Europeans affected by bladder cancer are homozygous for a non-coding single nucleotide polymorphism (8q24.21) located close to the c-Myc oncogene (23). Older studies have shown that pelvic radiotherapy for cervical cancer was associated with a twofold to fourfold increased risk of secondary bladder cancer (24,25). However, with contemporary radiotherapy, this risk has been reduced to a minimal level (26). In men with prostate cancer who received external beam radiotherapy and ⁄ or brachytherapy between 1973 and 1999, at 5 years, after the initial diagnosis of the primary cancer, 1–1.5% had developed a sec- ondary bladder malignancy (27). However, it is esti- mated that with modern radiotherapy techniques, men with prostate cancer now have < 1% risk of a secondary malignancy (26). It has been reported that treatment with cyclo- phosphamide for primary malignancies and autoim- mune disease increases the risk of bladder cancer by fourfold to ninefold, and is higher with greater cumulative doses and longer duration of exposure (28–31). Mesna (2-mercaptoethanesulfonic acid) is commonly co-administered with cyclophosphamide to decrease the development of cyclophosphamide- induced bladder cancer. It is speculated that Mesna, which is almost exclusively excreted by the kidneys counteracts the toxic effects of acrolein, an inactive metabolite of cyclophosphamide (32). Historically, it has been reported that chronic abusers of the analge- sic phenacetin had a four times higher risk of blad- der malignancy relative to non-users (33,34). The major metabolite of phenacetin is acetaminophen (paracetamol), which is often present in modern analgesics. However, heavy use of acetaminophen- containing analgesics did not increase bladder cancer risk in a number of studies (34–36). The oral antidiabetic drug pioglitazone is currently under post marketing pharmacovigilance, as a small excess of bladder cancer cases was shown in two studies evaluating its role in the management of dia- betes. The risk was associated with drug exposure in excess of 2 years (37,38). However, the benefits of pioglitazone are considered to outweigh this small risk for those who respond to treatment and in whom there is no history or other risk of bladder Bladder cancer management 437 ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 4. cancer. As a general principle, people with diabetes and non-visible haematuria (NVH) should be con- sidered for further investigation in the same way as any other individuals with NVH. There are also reported associations between increased risk of urothelial cancers and Aristolochia fangchi (a Chinese herb found in some over-the- counter diet pills) (39). Presenting symptoms The majority of patients diagnosed with bladder can- cer present with painless visible haematuria (VH) (13). Some patients, especially where CIS is present, may present with persistent irritative urinary symp- toms that may be accompanied by haematuria pres- ent at dipstick testing [symptomatic NVH (s-NVH)]. Other tumours may be detected following investiga- tion for asymptomatic NVH (a-NVH), or rarely with renal failure caused by bilateral ureteric obstruction or symptoms of metastatic disease (13). Around 19% presenting with VH will have a urinary tract malig- nancy detected in a one-stop haematuria clinic set- ting, compared with around 5% presenting with NVH; this is more likely if they have symptoms (40,41). However, there is currently no evidence-base to support population-based screening (7). What is significant haematuria? Recent publications from the UK have clarified what constitutes clinically relevant haematuria, which patients should be referred for clinical assessment, and whether they should be referred to a urologist, a nephrologist or both (summarised in Table 1) (42,43). Briefly, these are: NVH (‡ 1 + blood on dip- stick); persistent a-NVH (2 of 3 ‡ 1 + blood on dip- stick; clinically relevant haematuria [any episode of painless VH, painful VH or s-NVH in the absence of urinary tract infection (UTI), recurrent or refractory UTI with VH, or persistent a-NVH]. The latter requires urgent investigation. The American Urological Association (AUA) has also recently issued guidelines relating to the diagno- sis and management of a-NVH (44). A notable dif- ference between these guidelines and the UK-based publications is that the AUA defines a-NVH as ‡ 3 red blood cells per high powered field on a properly collected urine specimen. The AUA Guidelines state that a positive dipstick does not define a-NVH. The strength of evidence for this AUA recommendation is based on expert opinion. Causes of transient NVH include single episodes of UTI (45), exercise (46) and benign prostatic hyperplasia (44). Spurious causes of NVH include menstrual contamination, sexual intercourse, certain foods (beetroot, blackberries, rhubarb), rhabdomyol- ysis, drugs (doxorubicin, chloroquine, rifampicin) and chronic lead or mercury poisoning, and these should be excluded. However, haematuria in patients receiving anticoagulant therapy should not be attrib- uted solely to these agents (47,48). Recurrent or per- sistent UTI associated with haematuria is significant and merits urgent urological referral. What initial investigations should be performed in primary care? A joint consensus statement published by the Renal Association and BAUS, followed by a review article, emphasised the importance of a basic nephrological screen in all patients with NVH (42,43). Almost one- half of patients referred to urological haematuria clinics have NVH; however in this setting, patients with NVH are twice as likely to have nephrological disease (10%) than bladder cancer (5%) (49). The overall incidence of urological malignancy in screen- detected a-NVH is < 1% (50). All patients with s-NVH and persistent a-NVH should have their baseline blood pressure (BP), esti- mated glomerular filtration rate (eGFR), and albu- min:creatinine ratio (ACR) measured (Table 1). Further nephrological assessment should be consid- ered in those aged < 40 years with persistent a-NVH if any one of eGFR < 60 ml ⁄ min ⁄ 1.73 m2 , ACR > 30 or BP > 140 ⁄ 90 mmHg are present. Which patients with haematuria require urological referral? In the UK, referral guidelines for suspected cancer were published by NICE in 2005 (51). These included referral guidelines for haematuria (summarised in Table 1). If a patient is aged < 40 years and has persis- tent a-NVH, referral to a urology service is not consid- ered necessary unless there are risk factors for bladder cancer. If aged 40–50 years with persistent a-NVH or aged < 50 years with s-NVH, routine referral to urol- ogy is recommended. If aged > 50 years with persis- tent a-NVH or s-NVH, urgent urological referral is recommended. If aged > 40 years with recurrent or persistent UTI associated with haematuria, an urgent urological referral should be made. What tools are available to facilitate detection of bladder cancer? Currently recommended tools Cystoscopy remains the gold standard for the detec- tion of both new and recurrent bladder cancer. 438 Bladder cancer management ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 5. Despite this, its sensitivity is limited to approxi- mately 94% for the detection of tumours during fol- low-up (52), and may be as low as 60% for the detection of CIS (53). First described in 1945, the specificity of urine cytol- ogy for underlying urothelial malignancy is > 95%. However, the overall sensitivity of voided urine cytol- ogy is only 30–50%, as low-grade tumours do not shed malignant cells. It is therefore most sensitive in patients with high-grade tumours and CIS (54). Photodynamic diagnosis (PDD) requires the intra- vesical instillation of a photosensitising agent, usually 5-aminolevulinic acid (5-ALA) or its hexyl ester [hexaminolevulinate (HAL)], at least 1 h prior to cystoscopy. The derivative is preferentially taken up by tumour cells and produces orange fluorescence when blue light of 400 nm wavelength is applied. Two meta-analyses, including data up to 2009, con- cluded that compared with white-light cystoscopy (WLC), PDD improved detection of bladder tumours (especially CIS), and PDD use was associated with a lower rate of tumour recurrence (55,56). There was no evidence that PDD affected tumour progression or survival. However, the true added value of PDD in reducing tumour recurrence in routine practice is controversial because studies vary in the choice of photosensitiser (5-ALA or HAL) and use of immedi- ate single-dose intravesical chemotherapy post tran- surethral resection (TUR). Four randomised controlled trials (RCTs) of PDD vs. WLC published since 2009 have shown conflicting data (57–60). Tools undergoing development In general, novel urinary biomarkers possess greater sensitivity than cytology for the diagnosis of bladder cancer, at the expense of lower specificity, but none have sufficient sensitivity to obviate the need to per- form cystoscopy (54). Contemporary US Food and Drug Administration-approved and available tests are point-of-care (Nuclear Matrix Protein 22) and laboratory-based (ImmunoCytTM , Scimedx Corpora- tion, Denville, NJ, USA and UroVysionTM , Abbott Molecular Inc, Des Plaines, IL, USA). Their most promising application may be as part of surveillance protocols after treatment, but large prospective trials validating such protocols are lacking. There is con- cern regarding their use as a diagnostic adjunct in patients presenting with haematuria, as the increased false-positive rate would lead to unnecessary invasive investigations in patients with otherwise normal cys- toscopic appearances and imaging. Narrow-band imaging (NBI) enhances the contrast between the bladder mucosa and vascular structures by filtering white light into two narrow bands (415 and 540 nm) without the need for a preoperative instillation of contrast agent. Improved detection of primary and recurrent tumours has been shown in small non-randomised studies (61), and recurrence rates may be reduced when used as part of follow-up (62). The role of NBI-cystoscopy as an aid to TUR and in surveillance needs further evaluation in RCTs. Pivotal considerations in the management of NMIBC (TCC) An algorithm outlining the primary management of NMIBC (TCC) is presented in Figure 1. Allocation to a risk group Patients with NMIBC have a diverse prognosis. The European Organisation for Research and Treatment of Cancer (EORTC) has identified six key risk factors for the prediction of tumour recurrence and progres- sion to muscle invasive disease (63). These are: num- ber of tumours; prior recurrence rate; tumour size; stage (T-category); histological grade (1973 World Health Organization grade); presence of concomitant CIS. From these, risk scores are developed to enable patients to be classified into low-, intermediate-and high-risk groups for recurrence and progression. Risks of recurrence and progression at 5 years fol- lowing diagnosis vary from 31% to 78% and 0.8% to 45% respectively (63). TUR bladder tumour (papillary) & immediate single-dose post-operative intravesicial chemotherapy instillation NMIBC (TCC) Intermediate riskLow risk 5 year follow-up only (if no recurrence) Very high risk &/or long life-expectancy Consider re-resection and CT urogram High risk Surveillance cystoscopies Consider further intravesical chemotherapy (maximum 1 year) or BCG + maintenance (1–3 years) No further intravesical chemotherapy TUR : Transurethral resection NMIBC : Non-muscle invasive bladder cancer TCC : Transitional cell carcinoma Intravesical BCG + maintenance (1–3 years) Consider immediate radical cystectomy + pelvic lymphadenectomy Figure 1 Management of non-muscle invasive bladder cancer (TCC). BCG, bacillus Calmette-Gue´rin; CT, computed tomography; EORTC, European Organization for Research and Treatment of Cancer; NMIBC, non-muscle invasive bladder cancer; TCC, transitional cell carcinoma; TUR, transurethral resection Bladder cancer management 439 ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 6. Detrusor muscle status The quality of the initial TUR has a substantial impact on outcomes – removal of adequate detrusor muscle is associated with reduced risk of recurrence (64). Local recurrence rates for a single tumour at first-check cystoscopy can vary from 3.5% to 20.6% depending on the institution (65). To exclude muscle invasion at the primary TUR, larger tumours should be resected so that at least ex- ophytic and deep biopsies are submitted in separate fractions (66). Repeat TUR is recommended where the initial resection is judged to be incomplete, either because of the absence of detrusor muscle in a high- grade tumour or caused by macroscopic residual dis- ease; this is also currently recommended in all high- grade T1 tumours even if detrusor muscle was pres- ent in the first TUR specimen (66). Failure to adhere to these principles can adversely affect outcomes. Contemporary series have shown that in patients ini- tially staged as T1, 20% were upstaged to muscle invasive disease after repeat TUR (67) and residual tumour rates were 25–33% (68,69). Although EAU guidelines currently include high-grade Ta tumours in their recommendation for repeat resection (66), the evidence for repeat resection of high-grade Ta tumours is less robust than for high-grade T1 tumours. Requirement for adjuvant intravesical treatment The most commonly used intravesical chemothera- peutic agents are mitomycin C (MMC) and epirubi- cin. The intravesical immunotherapy of choice is bacillus Calmette-Gue´rin (BCG). A single postoperative intravesical instillation of chemotherapy within 24 h of TUR (and ideally 6 h) is currently recommended for all newly diagnosed bladder tumours (66). In clinical studies, this reduced tumour recurrence rates by 39% (70); Another study involving patients with recurrent NMIBC showed that the recurrence rate was increased twofold if instillation was performed more than 24 h after TUR (71). Need for further instillations of chemotherapy or BCG The effect of a single instillation of intravesical che- motherapy lasts for approximately 500 days (72). The choice between further intravesical chemother- apy or immunotherapy is guided by the patient’s risk group. Further intravesical chemotherapy reduces the risk of recurrence, but has not been shown to reduce disease progression (73); BCG is more effective than chemotherapy in reducing recurrence, provided maintenance treatment is administered (74), but is associated with more side effects than chemotherapy. If maintenance therapy is given for at least 1 year, BCG is the only intravesical agent shown to reduce or at least delay the risk of disease progression, by around 37% (75–78). Compared with epirubicin, maintenance BCG was superior in preventing pro- gression and also improving overall survival (OS) (77). However, the relative benefit of maintenance BCG over MMC in terms of disease progression is more controversial (74). For patients at low risk of recurrence and progres- sion (EORTC recurrence and progression scores = 0), no further treatment after TUR is rec- ommended prior to a further recurrence. The proba- bility of recurrence at 1 year is 15% and for progression is 0.2% (63). In intermediate-risk patients, the main priority is to reduce the risk of recurrence (46–62% at 5 years), although the risk of progression is not negligible (6– 17% at 5 years) (63). A maximum of 1 year of intra- vesical chemotherapy or 1–3 years of intravesical BCG is currently recommended, although the supe- rior efficacy of BCG needs to be balanced against its increased toxicity (66). Three-year vs. one-year BCG maintenance at full-dose compared with one-third- dose BCG in patients with intermediate- and selected high-risk (solitary tumour without CIS) NMIBC has been tested in the EORTC 30962 RCT (78). Superi- ority could not be formally concluded for either comparison, although patients receiving full-dose BCG for 3 years had the highest disease-free survival (DFS) at 5 years, whereas those receiving one-third- dose for 1 year had the lowest. Clearly, extending maintenance treatment for more than 1 year is a bal- ance of efficacy, side effects and inconvenience and is worthy of discussion with the patient. In routine clinical practice, many urologists in the UK delay the decision until the patient develops a further recur- rence. Some administer a further single-dose chemo- therapy instillation after TUR for recurrent disease, but this has not been validated for recurrent tumours. Others give six instillations (one instillation weekly for 6 weeks) of intravesical chemotherapy before the first-check cystoscopy at around 3 months after TUR. In high-risk patients (EORTC progression score > 14), the 5-year probability of disease progres- sion is 45% (63). Here, the primary goal of intra- vesical treatment is not only to prevent recurrence, but also to prevent progression. Main- tenance BCG has been established as the bladder- sparing treatment of choice in these patients. The optimal duration of treatment has not yet been established, but at least 1 year of maintenance is recommended (66,78). The Southwest Oncology 440 Bladder cancer management ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 7. Group protocol is the most widely used and involves giving up to 27 instillations of BCG over a 3-year period (79). Considerations for radical cystectomy There are no RCTs comparing BCG with immediate radical cystectomy. There is, however, consensus that immediate radical cystectomy should be considered for patients with high-risk NMIBC where the risk of tumour progression is especially high, or concomitant superficial urethral TCC is present, or where the patient requests it in preference to intravesical BCG. Added risk factors for progression in high-grade T1 disease include associated CIS (80), multifocality, tumour size > 3 cm and persistent T1 disease at repeat resection (81,82). If more than one of these risk factors is present, then radical cystectomy should be strongly considered. A growing body of urologists believe that radical cystectomy for high-grade T1 TCC is under- utilised at present. Their concerns are based on non- randomised evidence that suggests worse outcomes following cystectomy for failed intravesical treatment compared with immediate cystectomy, although such studies are likely to include selection bias (83,84). Immediate radical cystectomy for NMIBC may also be indicated when aggressive variants of TCC are present, e.g. diffuse areas of micropapillary variant are detected (85,86) or in those where BCG is contraindicated, e.g. significant immunosuppression. Radical cystectomy is also indicated for squamous cell carcinoma and radical or partial cystectomy should be considered for adeno- carcinoma. Failure of BCG The definition of BCG failure is controversial, but the persistence of T1 disease at the 3-month check cystoscopy after induction BCG (six instillations administered once-weekly for 6 weeks); high-grade Ta disease ⁄ CIS at 6 months; or detection of muscle invasive TCC should prompt consideration of radical cystectomy. Increased risk of concomitant or metachronous upper urinary tract (UUT)-TCC The incidence of concomitant UUT-TCC is low (1.8%), but in patients with trigonal tumours, the incidence is 7.5% (87). During follow-up, patients with high-grade and multifocal NMIBC are more likely to develop UUT-TCC (88). Ultrasound is rou- tinely performed during diagnostic work-up of blad- der cancer, but can miss small UUT-TCC. Computed tomography urography should therefore be considered where the bladder tumour is high- grade, multifocal or trigonal (66). Prevention of bladder tumours after nephro-ureterectomy for primary UUT-TCC After a nephroureterectomy for UUT-TCC, up to 40% of patients will develop bladder cancer. In a phase III RCT (ODMIT-C), a single dose of postop- erative intravesical MMC, administered at the time of urethral catheter removal, reduced the risk of a bladder tumour within the first year after nephro- ureterectomy; the absolute reduction in risk was 11%, the relative reduction was 40% and the num- ber needed to treat to prevent one bladder tumour was 9 (89). The trial was the largest randomised study ever conducted in the management of patients with UUT-TCC. However, it was not designed to determine the most effective method of nephroure- terectomy, and so a number of techniques were allowed for management of the distal ureter. Patients with a previous history of bladder cancer were excluded. Bladder-sparing treatments after BCG failure Radical cystectomy remains the mainstay of treat- ment for patients who have failed BCG treatment. For patients who are unwilling or unfit to undergo this procedure, the treatment options are limited. Intravesical chemotherapeutic agents, such as gem- citabine and docetaxel, novel immunotherapies, such as interferon-alpha, and device-assisted treatments have all shown promise. However, to date, much of the evidence to support their potential benefit is based on non-randomised or small Phase II studies. At best, currently available bladder-sparing treat- ments for those with BCG-refractory TCC are associ- ated with 2-year DFS of approximately 50% (90). Chemohyperthermia (c-HT) describes the combi- nation of intravesical chemotherapy and hyperther- mia, where the chemotherapy and the bladder wall are heated to temperatures of between 44 and 45 °C. The most common form of c-HT uses the Synergo HT system in which local HT is administered via a 915 MHz intravesical microwave applicator. c-HT increases cell membrane permeability, enhances uro- thelial exposure and in particular lamina propria exposure, alters intracellular drug trafficking and enhances the effects of cytostatic chemotherapy (91). Over the last 15 years, c-HT has been tested in a variety of clinical settings, including small several Phase II RCTs, in the BCG-naı¨ve setting (92). Data supporting the role of c-HT (using MMC) in BCG-refractory NMIBC has come from several proof-of-concept studies. In patients with BCG- refractory CIS, a complete response rate of 92% was shown, with 50% of patients remaining disease-free at 2-year follow-up (93). In patients with Bladder cancer management 441 ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 8. BCG-refractory NMIBC (77% high-risk) treated with a maintenance c-HT schedule, the recurrence-free rate was 56% at 2 years; 3% progressed to muscle invasive disease and 5% withdrew from treatment because of adverse events (94). The use of c-HT is being further evaluated in BCG-refractory patients with NMIBC who are unwilling or unfit for cystecto- my in the UK-based HYMN Phase III trial (EUD- RACT-2008-005428-99). Electromotive drug administration (EMDA) is an alternative way of enhancing MMC absorption and urothelial exposure, by creating an electrical gradient across the bladder wall using electrodes placed within the catheter and on the lower abdominal wall. In patients with BCG-naı¨ve high-risk NMIBC, EMDA- MMC has shown promise (95,96). To date, no stud- ies have specifically evaluated EMDA-MMC in the BCG-refractory setting, although one RCT allowed crossover of patients to EMDA-MMC alone if they did not respond to primary BCG treatment (95). Key considerations when treatment intent for muscle invasive TCC is potentially curative An algorithm outlining the primary management of muscle invasive bladder cancer is presented in Figure 2. Radical cystectomy or radical radiotherapy? Worldwide, radical cystectomy and pelvic lympha- denectomy has been the cornerstone treatment for muscle invasive TCC, although no RCT data exist to support cystectomy over bladder preservation with radical radiotherapy. Non-randomised data from sin- gle gold-standard institutions show similar outcomes for DFS (97,98). Typical radical radiotherapy sched- ules are 64 Gy in 32 fractions over 6.5 weeks or 55 Gy in 20 fractions over 4 weeks. An important confounding factor when comparing the results of radical cystectomy and radiotherapy is the discrep- ancy between pathological staging (cystectomy series) and clinical staging (radiotherapy series). Clinical staging is more likely to underestimate disease extent and so there is an outcome bias in favour of cystec- tomy series. For example, in one publication, only 23% of clinical stage T2 tumours (cT2) were con- firmed to be pathological stage T2 (pT2) at cystecto- my; 74% were pT3 ⁄ pT4 (99). In a contemporary series assessing cystectomy and pelvic lymphadenec- tomy alone, the 5-year OS for the entire cohort was 59% (100). In a separate study, following radical radiotherapy alone, 5-year OS was also approxi- mately 60% (101). When directly comparing radical cystectomy and radical radiotherapy, no significant difference between interventions in terms of 5-year OS was evident (102). Local disease control is a clinically relevant chal- lenge in the management of muscle invasive TCC and is consistently better after cystectomy than radiotherapy alone, balanced against the benefits of retaining the native bladder; many urological sur- geons do not offer reconstructive surgery in the sal- vage cystectomy setting. One concern is that salvage cystectomy for local disease recurrence after radio- therapy is technically more difficult than primary cystectomy, with a higher risk of complications. However, one large study demonstrated that OS after salvage cystectomy was identical to that of pri- mary cystectomy, but dependent on the experience of the operator (103). The local relapse rate follow- ing cystectomy is 10% (104), but can be as low as 8% when a pelvic lymphadenectomy is performed (100). Factors that would favour cystectomy over radio- therapy include poor bladder function, widespread CIS, large volume tumours, preexisting hydronephro- sis, previous pelvic radiotherapy and active inflamma- tory bowel disease. Where compliance with follow-up cystoscopic surveillance is likely to be difficult, cystec- tomy should be the preferred option. Overall, these factors must be weighed against morbidities associated with cystectomy, and patient preference. Available technologies choices for radical cystectomy and pelvic lymphadenectomy Radical cystectomy is a major operation; periopera- tive mortality is 3%, and 28% of patients develop a complication within 3 months of surgery (105). Ninety-day mortality rates increase with patient age: TUR bladder tumour (solid) Muscle invasive (T2-T2 N0-N3 M0) TCC (potentially curative on cross sectional staging) Advanced/metastatic (M1) TCC on cross sectional staging Locally advanced down-staging systemic chemotherapy followed by repeat cross sectional imaging Localised neoadjuvant systemic chemotherapy Radiotherapy with sensitiser Radical radiotherapy Radical cystectomy Palliative TUR : Transurethral resection TCC : Transitional cell carcinoma Palliative systemic chemotherapy +/– palliative radiotherapy Figure 2 Management of muscle invasive or advanced ⁄ metastatic bladder cancer (TCC). TCC, transitional cell carcinoma; TUR, transurethral resection 442 Bladder cancer management ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 9. < 70 years, 2%; 70–79 years, 5.4%; 80–89 years, 9.2% (106). When strict reporting guidelines are imple- mented in high volume centres, surgical morbidity following radical cystectomy is reported to be even higher (107,108). As well as removing the bladder and pelvic lymph nodes, the standard procedure is also to remove the prostate and seminal vesicles in men, and uterus and adnexa in women. The inclu- sion of the entire prostate in male patients and the extent of urethrectomy and vaginal resection in female patients have recently been questioned. There is a substantial amount of literature about the extent of lymphadenectomy, but consensus concerning the optimal extent remains elusive. Available data sug- gest that removal of least 11 lymph nodes may be associated with more favourable outcomes (109). Most agree that the template for standard pelvic lymph node dissection should at least include pelvic lymph nodes and external iliac lymph nodes up to level of the common iliac bifurcation (110). The extent of lymph node dissection and its impact on survival is currently the subject of RCTs, including the ongoing Southwest Oncology Group trial (SWOG-S1011; NCT01224665) in the United States and the AUO-multicentre RCT (AB 25 ⁄ 02; NCT01215071) in Germany, which is now closed to recruitment. Laparoscopic cystectomy and robotic- assisted cystectomy are feasible and also currently the subject of RCTs. Following radical cystectomy, urinary diversion is necessary. The most established option is the ileal conduit. The second most common option is the construction of a detubularised ileal orthotopic neo- bladder that is anastomosed to the urethra. The third option is a continence pouch that can be self-cathe- terised via, e.g. an appendix stoma. The presence of positive soft tissue surgical mar- gins following radical cystoprostatectomy is a strong predictor of recurrence-free and disease-specific sur- vival (111). EAU guidelines recommend that urethr- ectomy should be performed if positive surgical margins are present at the level of the urethral dis- section, if the tumour is located at the bladder neck or in the urethra (in women), or if the tumour extensively infiltrates the prostate (13). Factors that may protect against urethral recurrence include orthotopic reconstruction and radical radiotherapy. Evolving radiotherapy techniques The volume treated by radiotherapy has historically been the whole bladder, but for unifocal disease, techniques have evolved using a lower total dose to the whole bladder with a boost to the tumour bed. The potential benefits of this approach are to reduce toxicity and allow dose-escalation to the primary dis- ease. However, it is critical that organ motion and verification of the intended area of treatment is optimised. Novel approaches to achieve this include image- guided radiotherapy, allowing pretreatment com- puted tomography verification to be performed on a daily basis prior to each fraction and real-time moves to be made. However, the cost of this technique is significant and not available throughout the UK, but represents a significant development in radiotherapy implementation. Neoadjuvant, down-staging or adjuvant chemotherapy Up to 30% of patients relapse with distant metastases after radical cystectomy (100). Use of neoadjuvant chemotherapy, prior to radical cystectomy or radio- therapy, is postulated to reduce the risk of microm- etastatic disease and thereby confer a survival advantage. Cisplatin-containing combination chemo- therapy is the recommended approach (13). Typi- cally, three cycles are given, and the most commonly used contemporary chemotherapy regimens are the gemcitabine-cisplatin (GC) and methotrexate-vin- blastine-adriamycin-cisplatin (M-VAC) combina- tions. Neoadjuvant chemotherapy improves 5-year OS by 5–8%, irrespective of type of subsequent local treatment (112–114). This translates into a 14–16% relative reduction in risk of death and a 22–26% rel- ative improvement in DFS (112,113). Adequate renal function and performance status (PS) is necessary for these treatments. Down-staging chemotherapy should be considered for locally advanced bladder cancer (inoperable, but non-metastatic) where the aim is to increase the pos- sibility of either successful cystectomy or radiother- apy. Three to six cycles are commonly given, with cross-sectional imaging performed after three cycles to assess response. Currently, no RCT or meta-analysis has provided sufficient data to support the routine use of adjuvant chemotherapy because of underpowered studies and a difficulty in recruitment (13). It remains unclear whether immediate adjuvant chemotherapy or chemo- therapy at the time of relapse is superior or equivalent in terms of OS. However, adjuvant chemotherapy may be discussed on an individual basis particularly in patients with node-positive disease or locally advanced disease with high-grade pathology who have not already received neoadjuvant chemotherapy. Adequate renal function and PS is again mandated. Selective bladder preservation strategies To optimise the success of selective bladder preserva- tion, patients must be carefully selected to increase Bladder cancer management 443 ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 10. the likelihood of complete response, and meticulous cystoscopic follow-up must be performed to identify recurrent or persistent muscle invasive disease in candidates suitable for early salvage cystectomy. It is important to minimise the amount of normal tissue in the irradiated area, especially as the small bowel is used in an ileal conduit or in reconstruction in the event of salvage cystectomy. Emerging data from RCTs suggests that the stan- dard of care for bladder preservation is likely to become neoadjuvant chemotherapy followed by radiotherapy with a concurrent radiosensitiser (115– 117). The Phase III BC2001 trial of 360 patients that compared radiotherapy and concurrent 5-fluoroura- cil and MMC chemotherapy (a non-renal toxic regi- men) with radiotherapy alone demonstrated a significant improvement in loco-regional recurrence- free survival (67% compared with 54%) at 2 years (116). A similar degree of benefit was seen across all tumour stages (T2–T4) irrespective of age or whether the patient had received neoadjuvant chemotherapy. In a further Phase II study of patients receiving chemoradiation compared with historical radiother- apy controls, 89% of surviving patients at 3 years had an intact bladder following chemoradiotherapy with weekly gemcitabine (117). Tumour hypoxia has long been considered a cause of radiotherapy failure and one strategy to improve radiosensitivity is therefore to increase oxygenation of the tumour. In the BCON trial, radiotherapy adminis- tered with carbogen breathing apparatus and nicotin- amide tablets improved OS by 13% compared with radiotherapy alone (115). Of long-term survivors in the experimental arm, 83% retained intact bladders. Quality of life after cystectomy and selective bladder preservation Radical cystectomy is associated with significant alter- ation in health-related quality of life (HRQoL), with most of the morbidity related to the use of intestinal segments for urinary diversion. A commonly held assumption is that patients who choose neobladders should have improved HRQoL compared with those who undergo ileal conduit formation. However, a sys- tematic review has not determined a superior approach in terms of HRQoL (118). As well as health and body image, patients identified non-health-related determinants, such as family, relationships, finances as important QoL factors. A questionnaire-based study compared the HRQoL of 29 patients who received bladder preser- vation therapy (combination of chemoradiotherapy and TUR) and 30 patients who had undergone radi- cal cystectomy (119). Parameters associated with bet- ter HRQoL after bladder preservation than cystectomy included physical well-being (62% vs. 53%), anxiety (28% vs. 57%) and depression (24% vs. 47%). HRQoL after cystectomy was substantially impacted by stoma presence and a lack of sexual activity, but social and recreational life was only minimally affected. Patients undergoing bladder pres- ervation therapy reported dysuria (20%), frequency (44%), nocturia (42%) and difficulty controlling micturition (38%) (119). A further study with long-term follow-up (median 6.3 years) evaluated HRQoL and bladder function assessed by urodynamics in patients treated with TUR, chemotherapy and radiotherapy (120). Of 32 patients, most (75%) retained normal bladder func- tion. Urinary flow problems were reported by 6% of patients, urgency by 15% and urinary leakage in 19%. Difficulty with bowel control occurred in seven men and three women. Of male patients, 59% reported they were satisfied with their sex life. Palliative care approaches for advanced (metastatic) bladder cancer (TCC) Palliative chemotherapy is the cornerstone treatment for patients with metastatic TCC (Figure 2). It is usually given to improve HRQoL, improve symp- toms and also to improve prognosis, but is very unli- kely to be a cure. Before the development of effective chemotherapy, patients with TCC and visceral metas- tases rarely exceeded the median survival of 3– 6 months. In ‘fit’ patients with adequate renal func- tion, cisplatin-containing combination chemotherapy with GC or M-VAC should be considered, which can achieve a median survival of up to 14–15 months (121); GC may be associated with less toxicity than M-VAC. Carboplatin-based therapy is less effective than cisplatin-based therapy, but may be an option in patients ‘unfit’ for the former. Some patients who progress after first-line chemotherapy may benefit from agents, such as paclitaxel or vinflunine. Palliative radiotherapy may also be administered to help control symptoms, such as haematuria and pain. Emerging data suggests that zoledronic acid, a bisphosphonate, may also be helpful in patients with bone metastases (122). Can public and professional recognition of bladder cancer be improved? A single episode of VH is a warning sign that needs prompt investigation and urological referral. This is a key message for both the public and doctors in Pri- mary Care. Symptoms attributable to bladder cancer 444 Bladder cancer management ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 11. can mimic UTI. Failure to respond to antibiotics fol- lowing a symptomatic UTI or an early recurrent UTI warrants flexible cystoscopic evaluation before considering repeat courses of antibiotics. Otherwise, diagnosis (particularly in postmenopausal women) is delayed and may be contributory to less favourable outcomes in women than men. It is important to get this message across to doctors in Primary Care and pharmacists. Until recently, bladder cancer has been low on the public health agenda. However, the Department of Health has recognised the need for earlier diagnosis of bladder cancer, and has commenced a pilot study in three regions of the UK to determine the best way to promote these messages. Action on Bladder Cancer (http://www.actionon bladdercancer.org) is the only UK national bladder cancer charity purely focussed on improving the lives of patients with bladder cancer. It has three primary aims, namely to: improve awareness; elevate the sta- tus of bladder cancer in the public health agenda; and improve medical knowledge. It is hoped that the website will provide a good resource for the general public, patients and healthcare professionals. Acknowledgements Sources of funding: None. Author contributions This project was initiated by Mr Leyshon Griffiths (Consultant Urologist, Leicester, Secretary of Action on Bladder Cancer) and Mr Colin Bunce (Consultant Urologist, Middlesex, Chairman of Action on Blad- der Cancer). Primary development of the manuscript and co-ordination was conducted by Mr Griffiths with support and advice from Right Angle Commu- nications (Action on Bladder Cancer Secretariat). The manuscript was reviewed by members of the Action on Bladder Cancer Executive Committee: Dr Alison Birtle (Consultant Clinical Oncologist, Pres- ton), Dr Mark Beresford (Consultant Clinical Oncol- ogist, Bath), Mr Roger Kockelbergh (Consultant Urological Surgeon, Leicester), Mr Mark Feneley (Consultant Urological Surgeon, London), Mr Jer- emy Crew (Consultant Urological Surgeon, Oxford) and Mr Hugh Mostafid (Consultant Urological Sur- geon, Basingstoke). References 1 Ferlay J, Shin HR, Bray F et al. Estimates of world- wide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 2010; 127: 2893–917. 2 Cancer Research UK. Bladder cancer statistics – key facts. http://info.cancerresearchuk.org/cancer- stats/keyfacts/bladder-cancer/ (accessed June 2012). 3 Fajkovic H, Halpern JA, Cha EK et al. Impact of gender on bladder cancer incidence, staging, and prognosis. World J Urol 2011; 29: 457–63. 4 Edwards BK, Ward E, Kohler BA et al. Annual report to the nation on the status of cancer, 1975– 2006, featuring colorectal cancer trends and impact of interventions (risk factors, screening, and treat- ment) to reduce future rates. Cancer 2010; 116: 544–73. 5 Bosetti C, Bertuccio P, Chatenoud L et al. Trends in mortality from urologic cancers in Europe, 1970–2008. Eur Urol 2011; 60: 1–15. 6 Qiu D, Katanoda K, Marugame T, Sobue T. A Joinpoint regression analysis of long-term trends in cancer mortality in Japan (1958–2004). Int J Cancer 2009; 124: 443–8. 7 Moyer VA, on behalf of the U.S. Preventive Ser- vices Task Force. Screening for bladder cancer: US Preventive Services Task Force Recommendation Statement. Ann Intern Med 2011; 155: 246–51. 8 Keegan KA, Resnick MJ, Clark PE. Multimodal therapies for muscle-invasive urothelial carcinoma of the bladder. Curr Opin Oncol 2012; 24: 278–83. 9 British Association of Urological Surgeons, Section of Oncology. BAUS Cancer Registry, analyses of minimum data set for urological cancers, January to December 2009. October 2010. http://www.bau- s.org.uk/Resources/BAUS/Documents/ PDF%20Documents/Data%20and%20Audit/2009fi- nalanalyses.pdf (accessedJune 2012). 10 Yafi FA, Aprikian AG, Chin JL et al. Contempo- rary outcomes of 2287 patients with bladder can- cer who were treated with radical cystectomy: a Canadian multicentre experience. BJU Int 2011; 108: 539–45. 11 Gouda I, Mokhtar N, Bilal D, El-Bokhainy T, El- Bokhainy NM. Bilharziasis and bladder cancer: a time trend analysis of 9843 patients. J Egypt Natl Canc Inst 2007; 19: 158–62. 12 Felix AS, Soliman AS, Khaled H et al. The chang- ing patterns of bladder cancer in Egypt over the past 26 years. Cancer Causes Control 2008; 19: 421–9. 13 Stenzl A, Witjes JA, Cowan NC, et al. European Association of Urology Guidelines on muscle inva- sive and metastatic bladder cancer 2011. http:// www.uroweb.org/gls/pdf/07_%20Bladder%20Can- cer.pdf (accessed June 2012). 14 Freedman ND, Silverman DT, Hollenbeck AR, Schatzkin A, Abnet CC. Association between smoking and risk of bladder cancer among men and women. JAMA 2011; 306: 737–45. 15 Zeegers MP, Tan FE, Dorant E, van Den Brandt PA. The impact of characteristics of cigarette smoking on urinary tract cancer risk: a meta-anal- ysis of epidemiologic studies. Cancer 2000; 89: 630–9. 16 Baris D, Karagas MR, Verrill C et al. A case-con- trol study of smoking and bladder cancer risk: emergent patterns over time. J Natl Cancer Inst 2009; 101: 1553–61. 17 Hartge P, Silverman D, Hoover R et al. Changing cigarette habits and bladder cancer risk: a case-con- trol study. J Natl Cancer Inst 1987; 78: 1119–25. 18 Kogevinas M, Mannetje A, Cordier S et al. Occu- pation and bladder cancer among men in Western Europe. Cancer Causes Control 2003; 14: 907–14. 19 Chen CJ, Chuang YC, Lin TM, Wu HY. Malignant neoplasms among residents of a blackfoot disease- endemic area in Taiwan: high-arsenic artesian well water and cancers. Cancer Res 1985; 45: 5895–9. 20 Marcus PM, Vineis P, Rothman N. NAT2 slow acetylation and bladder cancer risk: a meta-analysis of 22 case-control studies conducted in the general population. Pharmacogenetics 2000; 10: 115–22. 21 Garcı´a-Closas M, Malats N, Silverman D. NAT2 slow acetylation, GSTM1 null genotype, and risk of bladder cancer: results from the Spanish Blad- der Cancer Study and meta-analyses. Lancet 2005; 366: 649–59. 22 Lee SW, Jang IJ, Shin SG et al. CYP1A2 activity as a risk factor for bladder cancer. J Korean Med Sci 1994; 9: 482–9. 23 Kiemeney L, Thorlacius S, Sulem P et al. Sequence variant on 8q24 confers susceptibility to urinary bladder cancer. Nat Genet 2008; 40: 1307–12. 24 Duncan RE, Bennett DW, Evans AT, Aron BS, Schellhas HF. Radiation-induced bladder tumors. J Urol 1977; 118: 43–5. 25 Sella A, Dexeus FH, Chong C, Ro JY, Logothetis CJ. Radiation therapy-associated invasive bladder tumors. Urology 1989; 33: 185–8. 26 Turina M, Mulhall AM, Mahid SS, Yashar C, Gal- andiuk S. Frequency and surgical management of chronic complications related to pelvic irradiation. Arch Surg 2008; 143: 46–52. Bladder cancer management 445 ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 12. 27 Moon K, Stukenborg GJ, Keim J, Theodorescu D. Cancer incidence after localized therapy for pros- tate cancer. Cancer 2006; 107: 991–8. 28 Fairchild WV, Spence CR, Solomon HD, Gangai MP. The incidence of bladder cancer after cyclo- phosphamide therapy. J Urol 1979; 122: 163–4. 29 Pedersen-Bjergaard J, Ersboll J, Hansen VL et al. Carcinoma of the urinary bladder after treatment with cyclophosphamide for non-Hodgkin’s lym- phoma. N Engl J Med 1988; 318: 1028–32. 30 Travis LB, Curtis RE, Glimelius B et al. Bladder and kidney cancer following cyclophosphamide therapy for non-Hodgkin’s lymphoma. J Natl Can- cer Inst 1995; 87: 524–30. 31 Knight A, Askling J, Granath F, Sparen P, Ekbom A. Urinary bladder cancer in Wegener’s granulo- matosis: risks and relation to cyclophosphamide. Ann Rheum Dis 2004; 63: 1307–11. 32 Links M, Lewis C. Chemoprotectants: a review of their clinical pharmacology and therapeutic effi- cacy. Drugs 1999; 57: 293–308. 33 Fokkens W. Phenacetin abuse related to bladder cancer. Environ Res 1979; 20: 192–8. 34 Castelao JE, Yuan J-M, Gago-Dominguez M, Yu MC, Ross RK. Non-steroidal anti-inflammatory drugs and bladder cancer prevention. Br J Cancer 2000; 87: 1364–9. 35 Fortuny J, Kogevinas M, Zens MS et al. Analgesic and anti-inflammatory use and risk of bladder cancer: a population based case-control study. BMC Urol 2007; 7: 13. 36 Genkinger JM, De Vivo I, Stampfer MJ, Gio- vannucci E, Michaud DS. Nonsteroidal anti- inflammatory drug use and risk of bladder cancer in the health professionals follow-up study. Int J Cancer 2007; 120: 2221–5. 37 Dormandy JA, Charbonnel B, Eckland DJ et al. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular. Lancet 2005; 366: 1279–89. 38 Lewis JD, Ferrara A, Peng T et al. Risk of bladder cancer among diabetic patients treated with piog- litazone: interim report of a longitudinal cohort study. Diabetes Care 2011; 34: 916–22. 39 Nortier JL, Martinez MC, Schmeiser HH et al. Urothelial carcinoma associated with the use of a Chinese herb (Aristolochia fangchi). N Engl J Med 2000; 342: 1686–92. 40 Khadra MH, Pickard RS, Charlton M, Powell PH, Neal DE. A prospective analysis of 1,930 patients with hematuria to evaluate current diagnostic practice. J Urol 2000; 163: 524–7. 41 Edwards TJ, Dickinson AJ, Gosling J et al. Patient- specific risk of undetected malignant disease after investigation for haematuria, based on a 4-year follow-up. BJU Int 2011; 107: 247–52. 42 Renal Association and British Association of Uro- logical Surgeons. Joint consensus statement on the initial assessment of haematuria. July 2008. http:// www.baus.org.uk/AboutBAUS/publications/hae- maturia-guidelines (accessed June 2012). 43 Kelly JD, Fawcett DP, Goldberg LC. Assessment and management of non-visible haematuria in pri- mary care. BMJ 2009; 338: 227–32. 44 Davis R, Jones JS, Barocas DA, et al. Diagnosis, evaluation and follow-up of asymptomatic microhaematuria (AMH) in adults: AUA Guide- line. http://www.auanet.org/content/media/asymp- tomatic_microhematuria_guideline.pdf (accessed October 2012) 45 Chung A, Arianayagam M, Rashid P. Bacterial cysti- tis in women. Aust Fam Physician 2010; 39: 295–8. 46 Jones GR, Newhouse IJ, Jakobi JM, LaVoie NL, Thayer R. The incidence of hematuria in middle distance track running. Can J Appl Physiol 2001; 26: 336–49. 47 Van Savage JG, Fried FA. Anticoagulant associated haematuria: a prospective study. J Urol 1995; 153: 1594–6. 48 Hurlen M, Eikvar L, Seljeflot I, Arnesen H. Occult bleeding in three different antithrombotic regimes after myocardial infarction. A WARIS-II-subgroup analysis. Thromb Res 2006; 118: 433–8. 49 Edwards TJ, Dickinson AJ, Natale S, Gosling J, McGrath JS. A prospective analysis of the diagnos- tic yield resulting from the attendance of 4020 patients at a protocol-driven haematuria clinic. BJU Int 2006; 97: 301–5. 50 Hiatt RA, Ordon˜ez JD. Dipstick urinalysis screen- ing, asymptomatic microhematuria, and subse- quent urological cancers in a population-based sample. Cancer Epidemiol Biomarkers Prev 1994; 3: 439–43. 51 National Institute for Health and Clinical Excel- lence. Referral guidelines for suspected cancer. http://www.nice.org.uk/nicemedia/pdf/CG027pub- licinfo.pdf (accessed June 2012). 52 Raitanen MP, Leppilahti M, Tuhkanen K et al. Routine follow-up cystoscopy in detection of recurrence in patients being monitored for bladder cancer. Ann Chir Gynaecol 2001; 90: 261–5. 53 Isfoss BL. The sensitivity of fluorescent-light cys- toscopy for the detection of carcinoma in situ (CIS) of the bladder: a meta-analysis with com- ments on gold standard. BJU Int 2011; 108: 1703– 7. 54 Mowatt G, Zhu S, Kilonzo M et al. Systematic review of the clinical effectiveness and cost-effec- tiveness of photodynamic diagnosis and urine bio- markers (FISH, ImmunoCyt, NMP22) and cytology for the detection and follow-up of blad- der cancer. Health Technol Assess 2010; 14: 1–331, iii–iv. 55 Mowatt G, N’Dow J, Vale L et al. Photodynamic diagnosis of bladder cancer compared with white light cystoscopy: systematic review and meta- analysis. Int J Technol Assess Health Care 2011; 27: 3–10. 56 Kausch I, Sommerauer M, Montorsi F et al. Pho- todynamic diagnosis in non-muscle invasive blad- der cancer: a systematic review and cumulative analysis of prospective studies. Eur Urol 2010; 57: 595–606. 57 Schumacher MC, Davidsson T, Friedrich B et al. Transurethral resection of non-muscle-invasive bladder transitional cell cancers with or without 5- aminolevulinic acid under visible and fluorescent light: results of a prospective randomized multi- centre study. Eur Urol 2010; 57: 293–9. 58 Stenzl A, Burger M, Fradet Y et al. Hexaminolevu- linate guided fluorescence cystoscopy reduces recurrence in patients with nonmuscle invasive bladder cancer. J Urol 2010; 184: 1907–13. 59 Hermann GG, Mogensen K, Carlsson S, Marcussen N, Duun S. Fluorescence-guided transurethral resection of bladder tumours reduces bladder tumour recurrence due to less residual tumour tis- sue in Ta ⁄ T1 patients: a randomized two-centre study. BJU Int 2011; 108(8 Pt 2): E297–303. 60 Stenzl A, Penkoff H, Dajc-Sommerer E et al. Detection and clinical outcome of urinary bladder cancer with 5-aminolevulinic acid-induced fluores- cence cystoscopy: a multicenter randomized, dou- ble-blind, placebo-controlled trial. Cancer 2011; 117: 938–47. 61 Cauberg ECC, Kloen S, Visser M et al. Narrow band imaging cystoscopy improves the detection of non-muscle-invasive bladder cancer. Urology 2010; 76: 658–63. 62 Herr HW, Donat SM. Reduced bladder tumour recurrence rate associated with narrow-band imaging surveillance cystoscopy. BJU Int 2011; 107: 396–8. 63 Sylvester RJ, van der Meijden APM, Oosterlinck W et al. Predicting recurrence and progression in individual patients with Stage Ta T1 bladder can- cer using EORTC Risk Tables: a combined analysis of 2596 patients from seven EORTC trials. Eur Urol 2006; 49: 466–77. 64 Mariappan P, Finney SM, Head E et al. Good quality white-light transurethral resection of blad- der tumours (GQ-WLTURBT) with experienced surgeons performing complete resections and obtaining detrusor muscle reduces early recurrence in new non-muscle-invasive bladder cancer: valida- tion across time and place and recommendation for benchmarking. BJU Int 2012; 109: 1666–73. 65 Brausi M, Collette L, Kurth K et al. Variability in the recurrence rate at first follow-up cystoscopy after TUR in Stage Ta T1 transitional cell carci- noma of the bladder: a combined analysis of seven EORTC studies. Eur Urol 2002; 41: 523–31. 66 Babjuk M, Oosterlinck W, Sylvester R, et al. European Association of Urology Guidelines on non-muscle invasive bladder cancer (TaT1 and CIS) 2011. http://www.uroweb.org/gls/pdf/05_TaT1_ Bladder_Cancer.pdf (accessed June 2012). 67 Dalbagni G, Vora K, Kaag M et al. Clinical out- come in a contemporary series of restaged patients with clinical T1 bladder cancer. Eur Urol 2009; 56: 903–10. 68 Herr HW, Donat SM. Quality control in transure- thral resection of bladder tumours. BJU Int 2008; 102: 1242–6. 69 Schips L, Augustin H, Zigeuner RE et al. Is repeated transurethral resection justified in patients with newly diagnosed superficial bladder cancer? Urology 2002; 59: 220–3. 70 Sylvester RJ, Oosterlinck W, van der Meijden APM. A single immediate postoperative instillation of chemotherapy decreases the risk of recurrence in patients with stage Ta T1 bladder cancer: a meta-analysis of published results of randomized clinical trials. J Urol 2004; 171: 2186–90. 71 Kaasinen E, Rintala E, Hellstro¨m P et al. Factors explaining recurrence in patients undergoing chemoimmunotherapy regimens for frequently recurring superficial bladder carcinoma. Eur Urol 2002; 42: 167–74. 72 Hinotsu S, Akaza H, Ohashi Y, Kotake T. Intrave- sical chemotherapy for maximum prophylaxis of new early phase superficial bladder carcinoma treated by transurethral resection: a combined analysis of trials by the Japanese Urological Cancer Research Group using smoothed hazard function. Cancer 1999; 86: 1818–26. 446 Bladder cancer management ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 13. 73 Pawinski A, Sylvester R, Kurth KH et al. A com- bined analysis of European Organization for Research and Treatment of Cancer, and Medical Research Council randomized clinical trials for the prophylactic treatment of stage TaT1 bladder can- cer. European Organization for Research and Treatment of Cancer Genitourinary Tract Cancer Cooperative Group and the Medical Research Council Working Party on Superficial Bladder Cancer. J Urol 1996; 156: 1934–40. 74 Malmstrom P-U, Sylvester RJ, Crawford DE et al. An individual patient data meta-analysis of the long-term outcome of randomised studies com- paring intravesical mitomycin C versus bacillus Calmette-Gue´rin for non-muscle-invasive bladder cancer. Eur Urol 2009; 56: 247–56. 75 Sylvester RJ, van der Meijden AP, Lamm DL. Intravesical bacillus Calmette-Guerin reduces the risk of progression in patients with superficial bladder cancer: a meta-analysis of the published results of randomized clinical trials. J Urol 2002; 168: 1964–70. 76 Bo¨hle A, Bock PR. Intravesical bacille calmette- gue´rin versus mitomycin c in superficial bladder cancer: formal meta-analysis of comparative stud- ies on tumor progression. Urology 2004; 63: 682–6. 77 Sylvester RJ, Brausi MA, Kirkels WJ et al. Long- term efficacy results of EORTC genito-urinary group randomized phase 3 study 30911 comparing intravesical instillations of epirubicin, bacillus Cal- mette-Gue´rin, and bacillus Calmette-Gue´rin plus isoniazid in patients with intermediate- and high- risk stage Ta T1 urothelial carcinoma of the blad- der. Eur Urol 2010; 57: 766–73. 78 Brausi MA, Oddens JR, Sylvester RJ et al. Bacillus Calmette-Guerin: one third dose versus full dose and one year versus three years of maintenance. Final results of an EORTC GU Cancers Group randomized trial in non muscle invasive bladder cancer. 27th Annual Congress of the European Association of Urology, February 2012. Abstract 1050. 79 Lamm DL, Blumenstein BA, Crissman JD et al. Maintenance bacillus Calmette-Guerin immuno- therapy for recurrent TA, T1 and carcinoma in situ transitional cell carcinoma of the bladder: a randomized Southwest Oncology Group Study. J Urol 2000; 163: 1124–9. 80 Griffiths TRL, Charlton M, Neal DE, Powell PH. Treatment of carcinoma in situ with intravesical bacillus Calmette-Guerin without maintenance. J Urol 2002; 167: 2408–12. 81 Herr HW, Donat SM, Dalbagni G. Can restaging transurethral resection of T1 bladder cancer select patients for immediate cystectomy? J Urol 2007; 177: 75–9. 82 Kulkarni GS, Hakenberg OW, Gschwend JE et al. An updated critical analysis of the treatment strat- egy for newly diagnosed high-grade T1 (previously T1G3) bladder cancer. Eur Urol 2010; 57: 60–70. 83 Denzinger S, Fritsche H-M, Otto W et al. Early versus deferred cystectomy for initial high-risk pT1G3 urothelial carcinoma of the bladder: do risk factors define feasibility of bladder-sparing approach? Eur Urol 2008; 53: 146–52. 84 Herr HW, Sogani PC. Does early cystectomy improve the survival of patients with high risk superficial bladder tumors? J Urol 2001; 166: 1296–9. 85 Samaratunga H, Khoo K. Micropapillary variant of urothelial carcinoma of the urinary bladder; a clin- icopathological and immunohistochemical study. Histopathology 2004; 45: 55–64. 86 Kamat AM, Dinney CP, Gee JR et al. Micropapil- lary bladder cancer: a review of the University of Texas M.D. Anderson Cancer Center experience with 100 consecutive patients. Cancer 2007; 110: 62–7. 87 Palou J, Rodriguez-Rubio F, Huguet J et al. Multi- variate analysis of clinical parameters of synchro- nous primary superficial bladder cancer and upper urinary tract tumour. J Urol 2005; 174: 859–61. 88 Millan-Rodriguez F, Chechile-Toniolo G, Salva- dor-Bayarri J, Huguet-Perez J, Vicente-Rodriguez J. Upper urinary tract tumours after primary superficial bladder tumours: prognostic factors and risk groups. J Urol 2000; 164: 1183–7. 89 O’Brien T, Ray E, Singh R, Coker B, Beard R, British Association of Urological Surgeons Section of Oncology. Prevention of bladder tumours after nephroureterectomy for primary upper urinary tract urothelial carcinoma: a prospective, multicen- tre, randomised clinical trial of a single postopera- tive dose of mitomycin C (the ODMIT-C trial). Eur Urol 2011; 60: 703–10. 90 Yates DR, Roupret M. Failure of bacille Calmette- Guerin in patients with high risk non-muscle-inva- sive bladder cancer unsuitable for radical cystecto- my: an update of available treatment options. BJU Int 2010; 106: 162–7. 91 Shen Z, Shen T, Wientjes MG, O’Donnell MA, Au JL. Intravesical treatments of bladder cancer: review. Pharm Res 2008; 25: 1500–10. 92 Lammers RJM, Witjes JA, Inman BA et al. The role of a combined regimen with intravesical che- motherapy in the management of non-muscle invasive bladder cancer: a systematic review. Eur Urol 2011; 60: 81–93. 93 Witjes JA, Hendricksen K, Gofrit O, Risi O, Nativ O. Intravesical hyperthermia and mitomycin-C for carcinoma in situ of the urinary bladder: experi- ence of the European Synergo working party. World J Urol 2009; 27: 319–24. 94 Nativ O, Witjes JA, Hendricksen K et al. Com- bined thermo-chemotherapy for recurrent bladder cancer after bacillus Calmette-Guerin. J Urol 2009; 182: 1313–7. 95 Di Stasi SM, Giannantoni A, Stephen RL et al. In- travesical electromotive mitomycin C versus pas- sive transport mitomycin C for high risk superficial bladder cancer: a prospective random- ized study. J Urol 2003; 170: 777–82. 96 Di Stasi SM, Valenti M, Verri C et al. Electromo- tive instillation of mitomycin immediately before transurethral resection for patients with primary urothelial non-muscle invasive bladder cancer: a randomised controlled trial. Lancet Oncol 2011; 12: 871–9. 97 Birkmeyer NJ, Goodney PP, Stukel TA, Hillner BE, Birkmeyer JD. Do cancer centres designated by the National Cancer Institute have better surgi- cal outcomes? Cancer 2005; 103: 435–41. 98 Horwich A, Dearnaley D, Huddart R et al. A ran- domized trial of accelerated radiotherapy for local- ized invasive bladder cancer. Radiother Oncol 2005; 75: 34–43. 99 Ficarra V, Dalpiaz O, Alrabi N, Novara G, Galfano A, Artibani W. Correlation between clinical and pathological staging in a series of radical cystecto- mies for bladder carcinoma. BJU Int 2005; 95: 786–90. 100 Madersbacher S, Hochreiter W, Burkhard F et al. Radical cystectomy for bladder cancer today – a homogeneous series without neoadjuvant therapy. J Clin Oncol 2003; 21: 690–6. 101 Cowan R, McBain C, Ryder WD et al. Radiother- apy for muscle invasive carcinoma of the bladder: results of a randomized trial comparing conven- tional whole bladder with dose-escalated partial bladder radiotherapy. Int J Radiat Oncol Biol Phys 2004; 59: 197–207. 102 Kotwal S, Choudhury A, Johnston C et al. Similar treatment outcomes for radical cystectomy and radical radiotherapy in invasive bladder cancer treated at a United Kingdom specialist treatment centre. Int J Radiat Oncol Biol Phys 2008; 70: 456– 63. 103 Addla SK, Naidu P, Maddineni SB, Ramani VA. Long term survival outcome following radical cys- tectomy for TCC of the bladder – comparison between primary and salvage cystectomy. J Urol 2009; 4(Suppl): 633. Abstract 1754. 104 Schuster TG, Smith DC, Montie JE. Pelvic recur- rences post cystectomy: current treatment strate- gies. Semin Urol Oncol 2001; 19: 45–50. 105 Stein JP, Skinner DG. Results with radical cystec- tomy for treating bladder cancer: a ‘reference stan- dard’ for high grade invasive bladder cancer. BJU Int 2003; 92: 12–7. 106 Liberman D, Lughezzani G, Sun M et al. Perioper- ative mortality is significantly greater in septage- narian and octogenarian patients treated with radical cystectomy for urothelial cancer of the bladder. Urology 2011; 77: 660–6. 107 Lowrance WT, Rumohr JA, Chang SS, Smith JA Jr, Cookson MS. Contemporary open radical cys- tectomy: analysis of perioperative outcomes. J Urol 2008; 179: 1313–8. 108 Shabsigh A, Korets R, Vora KC et al. Defining early morbidity of radical cystectomy for patients with bladder cancer using a standardized reporting methodology. Eur Urol 2009; 55: 164–74. 109 Herr HW. Extent of surgery and pathology evalua- tion has an impact on bladder cancer outcomes after radical cystectomy. Urology 2003; 61: 105–8. 110 Burkhard FC, Roth B, Zehnder P, Studer UE. Lymphadenectomy for bladder cancer: indications and controversies. Urol Clin North Am 2011; 38: 397–405. 111 Novara G, Svatek RS, Karakiewicz PI et al. Soft tis- sue surgical margin status is a powerful predictor of outcomes after radical radiotherapy: a multicen- tre study of more than 4400 patients. J Urol 2010; 183: 2165–70. 112 International Collaboration of Trialists, Medical Research Council Advanced Bladder Cancer Work- ing Party (now the National Cancer Research Institute Bladder Cancer Clinical Studies Group), the European Organisation for Research and Treatment of Cancer Genito-urinary Tract Cancer Group et al. International phase III trial assessing neoadjuvant cisplatin, methotrexate, and vinblas- tine chemotherapy for muscle invasive bladder cancer; long-term results of the BA06 30894 Trial. J Clin Oncol 2011; 29: 2171–7. 113 Advanced Bladder Cancer (ABC) Meta-analysis collaboration. Neoadjuvant chemotherapy in Bladder cancer management 447 ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448
  • 14. invasive bladder cancer update of a systematic review and meta-analysis of individual patient data advanced bladder cancer (ABC) meta-analysis col- laboration. Eur Urol 2005; 48: 202–5. 114 Sherif A, Holmberg L, Rintala E et al. Nordic Uro- thelial Cancer Group Neoadjuvant cisplatinum based combination chemotherapy in patients with invasive bladder cancer: a combined analysis of two Nordic studies. Eur Urol 2004; 45: 297–303. 115 Hoskin PJ, Rojas AM, Bentzen SM, Saunders MI. Radiotherapy with concurrent carbogen and nico- tinamide in bladder carcinoma. J Clin Oncol 2010; 28: 4912–8. 116 James ND, Hussain SA, Hall E et al. Radiotherapy with or without chemotherapy in muscle invasive bladder cancer. N Engl J Med 2012; 366: 1477–88. 117 Choudhury A, Swindell R, Logue JP et al. Phase II study of conformal hypofractionated radiotherapy with concurrent gemcitabine in muscle-invasive bladder cancer. J Clin Oncol 2011; 29: 733–8. 118 Somani BK, Gimlin D, Fayers P, N’Dow J. Quality of life and body image for bladder cancer patients undergoing radical cystectomy and urinary diver- sion – a prospective cohort study with a systematic review of literature. Urology 2009; 74: 1138–43. 119 Caffo O, Fellin G, Graffer U, Luciani L. Assess- ment of quality of life after cystectomy or conser- vative therapy for patients with infiltrating bladder carcinoma. A survey by a self-administered ques- tionnaire. Cancer 1996; 78: 1089–97. 120 Zietman AL, Sacco D, Skowronski U et al. Organ conservation in invasive bladder cancer by tran- surethral resection, chemotherapy and radiation: results of a urodynamic and quality of life study on long-term survivors. J Urol 2003; 170: 1772–6. 121 von der Maase H, Sengelov L, Roberts JT et al. Long-term survival results of a randomized trial comparing gemcitabine plus cisplatin with metho- trexate, vinblastine, doxorubicin plus cisplatin in patients with bladder cancer. J Clin Oncol 2005; 23: 4602–8. 122 Zaghloul MS, Boutrus R, El-Hossieny H et al. A prospective randomized placebo-controlled trial of zoledronic acid in bony metastatic bladder cancer. Int J Clin Oncol 2010; 15: 382–9. Paper received July 2012, accepted October 2012 448 Bladder cancer management ª 2012 Blackwell Publishing Ltd Int J Clin Pract, May 2013, 65, 5, 435–448