You are on page 1of 21

135  Tumors of the Bladder

Max Kates, MD, and Trinity J. Bivalacqua, MD, PhD

EPIDEMIOLOGY incidence rates for bladder cancer are in Belgium for men and Hungary
Incidence, Prevalence, Mortality for women (Ferlay et al., 2014, 2013). These regional differences are
due to a combination of factors, including a country’s demographic
Bladder cancer is a global disease, with 540,000 incident cases and characteristics and environmental exposures.
188,000 deaths in 2015 worldwide (Fig. 135.1). In the United States
81,190 patients will be diagnosed in 2018 and 17,240 will die from Gender, Racial, and Age Differences
their disease (American Cancer Society, 2017). Although age-adjusted
incidence and mortality rates initially declined in the 1990s and early Bladder cancer typically arises from chronic, constant insults to the
part of 2000s, these rates have plateaued in recent years (Dy et al., urinary tract over time. Urothelial cancer is a disease of aging and
2017; Ferlay et al., 2014). Between 2005 and 2015 unadjusted incidence environmental exposures. As such, the incidence and prevalence
rates of bladder cancer grew 31%, which has been attributed to aging increase with age. The average age of diagnosis is 73 in the United
and population growth (Fitzmaurice et al., 2017). This increase in States with approximately 9 of 10 patients diagnosed after the age
incidence has been attributed to the increase in life expectancy over of 55. Age-specific incidence rates rise gradually around age 50 to
time and the resultant aging of the global population. Adjusted for 54 in males and females, with a sharper rise in males age 60 to 64.
demographic changes, the incidence rates for bladder cancer have The highest age-adjusted incidence rates are in the 90+ age group
remained relatively stable (Fig. 135.2). The United States has seen a for both sexes (Fig. 135.3).
decline in incidence rates from 2005 to 2014 by 0.8% per year in men Nearly three-quarters of bladder cancer cases occur in males, who
and women (American Cancer Society, 2017). The odds of developing have a higher incidence rate (9.0 per 100,000) compared with women
bladder cancer are highest in high-income countries (1 in 36 men (2.2). In the United States, 1 in 27 men will develop bladder cancer
and 1 in 165 women) and lowest in low-income countries (1 in 122 over their lifetime, and 1 in 107 US men will die of bladder cancer.
men and 1 in 310 women). In 2015 age-specific incidence rates were For women, 1 in 89 US women will develop bladder cancer, and 1
highest in North American (31.6 per 100,000), Western Europe (26.0), in 293 will die of their disease (American Cancer Society, 2017).
and Central Europe (24.1), and lowest in Oceania (4.5), Andean Several hypotheses have been proposed for increased bladder cancer
Latin America (5.8), and Central Latin America (5.9). However, since rates among men. First, globally smoking is much more common
1990, developing countries have had an increasing burden of bladder in men in comparison with women, with age-standardized prevalence
cancer incidence compared with developed countries, which has been declining in men from 41.2% in 1980 to 31.1% in 2012 in comparison
attributed to a narrowing gap in life expectancy between low- and with 10.6% to 6.2% among women (Chappidi et al., 2017). However,
high-income countries, with subsequent higher rates of cancer in an even when controlling for smoking, gender-related incidence dispari-
older population (Dy et al., 2017). ties persist (Hartge et al., 1990). It has been hypothesized that
Although incidence rates are highest in Europe and North America, although carcinogen exposure may not account for differences between
more than 60% of all bladder cancer incidence, and nearly half of genders, cellular metabolism of carcinogens may be different (Hemelt
bladder cancer deaths occur in the developing world (Antoni et al., et al., 2009). Metabolic enzymes including 5′-diphosphoglucuro-
2017). Disability-adjusted life-years (DALYs) are the primary way in nosyltransferase (UGT), which is involved in aromatic amine
which the World Health Organization (WHO) quantifies the burden of metabolism, and glutathione-S-transferase M1 (GSTM1), involved
disease from mortality and morbidity. DALYs correspond to the years in foreign substance detoxification, have been implicated in bladder
of life lost (number of deaths × remaining life expectancy) and the cancer–associated carcinogen metabolism. Both enzymes have been
years lost because of disability. Bladder cancer causes approximately shown to be differentially expressed in men and women (Karagas
3.4 million DALYs in 2015. Furthermore, although age-standardized et al., 2005; Zhang, 2013). As a result, gender differences in carcinogen
DALYs rates for bladder cancer among both sexes decreased by 9%, metabolism may explain why even among similar carcinogen
incidence rates have remained steady among high-income countries. exposures (e.g., similar smoking pack-years), gender disparities in
Because bladder cancer has fewer deaths relative to incident cases bladder cancer incidence persist.
compared with several other malignancies, bladder cancer is one of In addition, gender differences have been explained by differences
the world’s most prevalent cancers. The global 5-year prevalence of in sex steroid production and receptor expression. Among women,
bladder cancer is estimated to be 1,319,749, with 243,867 coming older age at menarche (≥15), parity (compared with nulliparous
from the United States (Ferlay et al., 2014). However, 5-year prevalence women), and use of estrogen or progestin therapy have been associ-
may not fully reflect the number of bladder cancer survivors managing ated with decreased UCB risk, suggesting that sex steroid exposure
disease sequel, as an estimated 571,518 bladder cancer survivors mitigates bladder cancer risk (Daugherty et al., 2013). From a tumor
lived in the United States in 2011 (Malats and Real, 2015). Among biology standpoint, the androgen receptor (AR) has been implicated
cancers affecting both sexes, bladder cancer ranks as the fourth most in bladder cancer development and progression. AR expression
prevalent cancer globally, and in the top 5 in prevalence in individual appears to be downregulated in immunohistochemical studies of
countries of all income levels and all regions, including the United bladder cancer, and this downregulation appears to increase with
States (96.6 patients per 100,000), Argentina (32.6), Egypt (41.2), higher stage/grade tumors (Li et al., 2017). Transgenic and N-butyl-
Belgium (158.6), Australia (59.7), and Japan (71.0) (Bray et al., N-(4-hydroxybutyl)-nitrosamine (BBN) mouse models of bladder
2013; Ferlay et al., 2014; see Fig. 135.2). Regional variations do exist, cancer have demonstrated that castration or direct AR knockout
however. Bladder cancer was the most commonly diagnosed cancer suppresses bladder cancer growth and development (Johnson et al.,
in 2015 for men in Egypt primarily because of high rates of Schistosoma 2008; Miyamoto et al., 2007).
haematobium in that country, which causes squamous cell carcinoma Although the disease is more prevalent in men, women are more
(SCC) of the bladder. In Europe, the highest world age-standardized likely to have more advanced tumors and a less-favorable prognosis

3073
3074 PART XIV  Benign and Malignant Bladder Disorders

Bladder Cancer

8.9+ 4.7–8.9 2.9–4.7 1.8–2.9 <1.8 No data

Fig. 135.1.  Global incidence rates of bladder cancer. (Data from Ferlay J, Soerjomataram I, Ervik M,
et al.: GLOBOCAN 2012 v1.1, Cancer incidence and mortality worldwide: IARC CancerBase No. 11.
Lyon, Fr. Int Agency Res Cancer, 2014 [website]: http://globocan.iarc.fr)

Males Females Persons

50

40
Incidence rate per 100,000

30

20

10

0
19

19

19

19

19

19

19

20

20

20

20

20

20

20

20

20

20

20

20

20

20

20

20
93

94

95

96

97

98

99

00

01

02

03

04

05

06

07

08

09

10

11

12

13

14

15

Year of diagnosis

Fig. 135.2.  Trends in age-standardized incidence rates for bladder cancer, 1993 to 2015. (Modified from
Cancer Center UK: Bladder cancer statistics, 2018 [website]: https://www.cancerresearchuk.org/health-
professional/cancer-statistics/statistics-by-cancer-type/bladder-cancer.)

(Scosyrev et al., 2009). Although men lose 26% of their remaining et al., 2000; Scosyrev et al., 2009). This gender disparity in the
life expectancy to bladder cancer, women lose 36% (Scosyrev et al., aggressiveness of bladder cancer at presentation has been attributed
2012). The differences in stage presentation only account for approxi- to differences in tumor biology, environmental exposures, and as
mately 30% of gender disparities in bladder cancer prognosis and well as implicit bias (also known as implicit social cognition), in
mortality; women fare worse than men stage for stage (Aydin Mungan which the conception of bladder cancer as a “male” disease leads
Chapter 135  Tumors of the Bladder 3075

Males cases Female cases Male rate Female rate


1,500 300

Average number of new cases per year


1,250 250

Incidence rate per 100,000


1,000 200

750 150

500 100

250 50

0 0 to 05 to 10 to 15 to 20 to 25 to 30 to 35 to 40 to 45 to 50 to 55 to 60 to 65 to 70 to 75 to 80 to 85 to 90+
0
04 09 14 19 24 29 34 39 44 49 54 59 64 69 74 79 84 89

Age at diagnosis

Fig. 135.3.  Age-adjusted incidence rates for bladder cancer in the United Kingdom. (Data from Cancer
Research UK: Cancer stats, 2017 [website]: https://www.cancerresearchuk.org/cancerstats.)

to delays in diagnosis from initial hematuria symptoms to pathological Cancer statistics, 2018
diagnoses. Multiple series using insurance claims data have demon-
strated that women have a delay from their initial hematuria claim
to their bladder cancer claim compared with men and that women All races
are more likely to be referred for urinary tract infection (UTI) evalu- Urinary bladder†
ation and therefore have a delay in referral to a urologist (Buteau White 100
et al., 2014; Cohn et al., 2014; Garg et al., 2014). These delays in
diagnosis may partially explain why women are more likely to present Black 7778
80
with advanced stage disease. 7070
Bladder cancer is most common among Caucasian Americans, 61 64
with an incidence rate 1.5 times that of African-Americans, twice 60
that of Hispanic Americans, and six times that of Native Americans.
However, similar to gender differences, African-American patients 40 3536
28
are more likely to have muscle-invasive disease compared with
Caucasian patients (Scosyrev et al., 2009; Fig. 135.4). In addition, 20
for reasons that are unclear, African-Americans, and African-American 5 5 6
women in particular, have increased rates of non-urothelial histology. 0
Rates of non-urothelial histology among African-American women

s
ed

nt
l

ge
na
(10.5%) are greater than African-American men (5.9%), Caucasian
ta
liz

ta
io

is
ca

ls
eg

D
American men (2.3%), or Caucasian American women (4.3%)
Lo

Al
R

(Scosyrev et al., 2009). Stage for stage, 5-year relative survival rates
are worse for African-Americans with localized disease (61% vs. 70%) Fig. 135.4.  Five-year relative survival rates for selected cancers by race and
and regionally advanced disease (28% vs. 36%) compared with all stage at diagnosis, United States, 2007 to 2013. The standard error is between
Americans (Siegel et al., 2018). 5 and 10 percentage points. †The survival rate for carcinoma in situ of the
urinary bladder is 96% in all races, 96% in whites, and 91% in blacks. (Data
from Siegel RL, Miller KD, Jemal A: Cancer statistics, 2018. CA Cancer J
Clin 68:7–30, 2018.)
ECONOMIC IMPACT
Bladder cancer is the most expensive cancer to treat per patient over
a patient’s lifetime (Curtis, 2016). The lifetime treatment cost per
patient is between $129,000 to $251,000. Annual directed medical metastatic disease resulting from large costs of late-stage therapeutics
costs are estimated to exceed $4 billion in the United States and and end-of-life care (Svatek et al., 2014).
€4.9 billion in the European Union (EU) (Curtis, 2016; Leal et al.,
2016). However, bladder cancer health care costs vary widely between
countries and disease states. For example in Latvia €2257 is spent RISK FACTORS
per bladder cancer case compared with €11 937 in France (Leal et al.,
2016). Europe as a whole spends €45 per 10 EU citizens compared Like skin and lungs, the bladder is an organ that comes in con-
with €97 per 10 US citizens. These variations between countries and stant contact with the environment and is therefore sensitive to
between patients in the same country can be attributed to regional environmental carcinogens and inflammation. Aromatic amines
variations in the management of bladder cancer, particularly non– (2-naphthylamine, 4-aminobiphenyl, and benzidine) are thought
muscle-invasive disease. In addition, treatment costs vary by institution to be the central causative agent in carcinogen-mediated bladder
and region (Svatek et al., 2014). Two areas that have been proposed carcinogenesis. Tobacco smoking and occupation account for the
for future research and interventions to improve bladder cancer costs two most frequent routes of environmental exposure. Tobacco smoke
are in the management of patients with non–muscle-invasive bladder is full of aromatic amines, as are other environmental carcinogens
cancer (NMIBC) in whom intense surveillance strategies often lead implicated in bladder cancer. These amines, when hydroxylated,
to procedures and laboratory testing annually, and in patients with lead to DNA adduction and damage.
3076 PART XIV  Benign and Malignant Bladder Disorders

Genetics meta-analyses to be a risk factor for bladder cancer development


(Choi et al., 2018; Koebnick et al., 2008; Sun et al., 2015; Zhao
The most-studied genes associated with bladder cancer are et al., 2017). One reason for this link is the strong association between
N-acetyltransferase 2 (NAT2) and a deletion of glutathione S-transferase smoking and obesity; however, BMI remains associated with bladder
µ (GSTM1) (Garcia-Closas et al., 2005; Golka et al., 1997; Karagas cancer even after controlling for smoking status. There is also a
et al., 2005). Both of these genes are associated with the ability to dose-response relationship in which bladder cancer risk appears to
metabolize aromatic amines and thus play an important role in increase with BMI. Furthermore, among patients already diagnosed
the subset of individuals with environmental carcinogen exposure. with bladder cancer, obesity appears to attenuate prognosis and is
NAT2 detoxifies nitrosamines, regulating the rate of acetylation of associated with an increased risk of tumor recurrence and a shorter
aromatic amine compounds found in cigarette smoke (Garcia-Closas time interval to recurrence (Wyszynski et al., 2014). The mechanism
et al., 2005). Recent evidence suggests that there are likely several of excess weight contributing to cancer risk includes insulin resistance,
underlying genetic risk factors, that when combined with occupational, chronic hyperinsulinemia, increased bioavailability of steroid hor-
environmental, and behavioral exposures, potentiate bladder cancer mones, and localized inflammation. Elevated insulin production
risk and development (Czerniak et al., 2016). may drive tumor growth and stimulate pro-inflammatory cytokines.
The exact mechanism by which excess weight may contribute to
Hereditary bladder cancer risk is unknown and likely includes a multitude of
inflammatory mediators.
Although first-degree relatives of patients with bladder cancer have
a twofold increased risk of developing urothelial bladder cancer, Occupational Risk
high-risk bladder cancer families are exceedingly rare. Hereditary
bladder cancer does not seem to be associated with a Mendelian Occupational exposures account for between 5% and 10% of
inheritance pattern, making classical family-tree linkage studies all bladder cancer (Cumberbatch et al., 2015; Rushton et al., 2010;
unhelpful. More likely the cause is multifactorial, in which certain Table 135.1). Occupations that work directly with chemicals and
genes magnify environmental exposures (Kiemeney, 2008). Patients dyes have the greatest lifetime risk. Particularly at-risk occupations
with Lynch syndrome (hereditary non-polyposis colon carcinoma, that work with aromatic amines include tobacco, dye, and rubber
HNPCC) are at increased risk of developing urothelial cancer. This workers, hairdressers, painters, and leather workers. Those that works
increased risk is primarily found among the mismatch repair gene with polycyclic aromatic hydrocarbons are also at risk, including
MSH2 mutations carriers (van der Post et al., 2010). chimney sweeps, nurses, waiters, alumni workers, petroleum workers,
and seamen (Cumberbatch et al., 2015). Many of these professions
Smoking are increasingly women, and it has been reported that occupational
exposures may be increasing among women in particular.
Tobacco is the main known cause of bladder cancer and accounts
for 30% to 40% of all urothelial carcinoma. Worldwide there are Medical Conditions
more than 1 billion current smokers, and smokers have a 2- to
3-fold increased risk of bladder cancer (Islami et al., 2015). Although Medical conditions have the potential to increase bladder cancer
cigarette smoking is the most common tobacco product associated risk directly or indirectly as a toxicity of treatment. Typically, direct
with bladder cancer risk, pipe smoking and cigar smoking have also carcinogenesis is mediated by chronic inflammation and the develop-
been associated with urothelial carcinoma formation (Cumberbatch ment of keratinizing squamous metaplasia. For example, patients
et al., 2016). Current cigarette smoking among US adults declined with neurogenic bladder and spinal cord injuries who have chronic
from 20.9% (45.1 million US adults) in 2005 to 15.1% (36.5 indwelling catheters are at a modestly increased risk of developing
million) in 2015. This represents a 27.7% decline in a 10-year period SCC of the bladder.
(Jamal et al., 2016). Aromatic amines are the primary carcinogens Bladder calculi, urinary outflow obstruction, recurrent UTIs, and
in tobacco smoke that lead to cancer. Persons with NAT2 slow inflammation from direct catheter trauma have been postulated to be
acetylation polymorphisms have been shown to be at a particularly additive in the development of keratinizing squamous metaplasia (Ho
high risk when they smoke cigarettes, because NAT2 allows for the et al., 2015; West et al., 1999). In addition, the malignant potential
detoxification of these amines. Although there is some evidence that of bladder exstrophy has been well documented, with the reported
dietary beta-carotene may have protective effects, supplementary
beta-carotene and vitamin supplementation (with C, E, or folate)
have not been shown to mitigate bladder cancer risk among those TABLE 135.1 Relative Risk of Bladder Cancer
with environmental exposures (Roswall et al., 2009). Smoking by Occupation
cessation has been shown to decrease the risk of urothelial cancer
formation: smokers who have stopped for 1 to 3 years have a 2.6 OCCUPATION RELATIVE RISK
relative risk compared with a 1.1 relative risk among those who have
stopped smoking for more than 15 years (Koshiaris et al., 2017). Tobacco workers 1.72
Approximately 20% of patients with bladder cancer are smokers at Dye workers 1.58
the time of diagnosis, and these patients have an increased risk for Chimney sweeps 1.53
local recurrence when actively smoking with NMIBC and increased Nurses 1.49
risk of bladder cancer death when actively smoking with muscle- Rubber workers 1.49
invasive bladder cancer (MIBC). In addition, many patients with Waiters 1.43
smoking-related cancer continue to smoke even after their bladder Aluminum workers 1.41
cancer is discovered (Koshiaris et al., 2017). The survival benefit of Hairdressers 1.32
quitting early after a bladder cancer diagnosis is controversial and Plumbers 1.2
still an area of active investigation. However, continued smoking
Metal workers 1.14
increases the risk of a second primary cancer and the risk of other
smoking-related diseases, including peripheral vascular disease and Fishermen 1.13
chronic obstructive pulmonary disorder (Health NC, 2014). Physicians 1.11
Mechanics 1.11
Body Mass Index
Data from Cumberbatch MG, Cox A, Teare D, et al.: Contemporary
Increased body mass index (BMI) has been shown to be a risk factor occupational carcinogen exposure and bladder cancer: a systematic review
for multiple cancers. Increasing BMI has been shown in multiple and meta-analysis. J Am Med Assoc Oncol 1(9):1282–1290, 2015.
Chapter 135  Tumors of the Bladder 3077

incidence of 3.3% to 7.5% of the general exstrophy population—several increased rates of urothelial carcinoma. In one study, the attributable
times higher than age-matched controls in the general population risk for survivors exposed to more than 0.005 Gy was estimated to
(Rieder et al., 2006; Smeulders and Woodhouse, 2001). Adenocarci- be 16% and rose to 60% for those with more than 1 Gy of exposure.
noma accounts for more than 90% of cases and squamous cell and Urothelial cancer formation after radiation therapy does not appear
urothelial carcinoma account for the remaining 10%. As patients with age related; however, the estimated latency period is estimated to
spinal cord injuries and congenital bladder abnormalities continue to be 15 to 30 years. External beam radiation for cervical cancer is
live longer lives because of advances in the care of these individuals, associated with a two- to fourfold higher incidence of secondary
rates of bladder cancer may rise as these patients age. bladder cancer compared with the nonirradiated population (Boice
et al., 1985; Kleinerman et al., 1995). Studies regarding the use of
Schistosomiasis external beam radiation therapy (EBRT) and brachytherapy are
conflicting and likely confounded by other risk factors for bladder
Schistosomiasis is an infectious disease endemic in as many as 76 cancer. However, they do suggest that EBRT for prostate may denote
developing countries and affecting up to 218 million people (Word a 1.5-fold to twofold increased risk of bladder cancer (Chrouser
Health Organization, 2017). Despite widespread WHO-led eradication et al., 2005; Nieder et al., 2008). Brachytherapy appears to be associ-
campaigns, schistosomiasis remains a major contributor to disease ated with a smaller risk, and several investigators have suggested
and bladder cancer in many tropical countries. Schistosomes are that intensity-modulated radiation therapy denotes little to no
parasitic blood flukes that have mammalian hosts as well as intermedi- increased risk of bladder tumor formation (Hall and Wuu, 2003).
ate hosts (e.g., freshwater snails). Of the four human schistosomes,
S. haematobium is the one linked to squamous cell bladder cancer. Chemotherapy
S. haematobium live in the venules of the human urinary bladder,
where they lay eggs, producing irritation and tissue fibrosis. The Chemotherapy destroys malignant cells by inducing DNA damage and
mechanism of SCC development likely involves a TH-2 type pro- cell death; however, in organs with rapid cell turnover, chemotherapy
inflammatory immune response (Bernardo et al., 2016). In addition, can induce dysregulation for normal cells. The only chemotherapy
underlying S. haematobium infection seems to promote susceptibility that has been shown to cause bladder cancer is cyclophosphamide.
to other bladder cancer risk factors—notably smoking. Phosphoramide mustard is the primary mutagenic metabolite that
causes cyclophosphamide-associated bladder cancer. The risk of
Recurrent Infection bladder cancer is 4.5-fold higher among patients who underwent
cyclophosphamide treatment and appears to be dose dependent and
Multiple investigators have suggested that persistent or recurrent bacte- most pronounced among those who receive more than 20 g. For
rial UTIs may increase the risk of bladder carcinogenesis. Multiple patients who receive more than 50 g of cyclophosphamide, the excess
epidemiologic studies suggest that there may be a modest, increased risk increases to approximately 7 excess cases per 100 patients treated
risk of bladder cancer associated with chronic UTIs; however, these (Travis et al., 1995). In addition, the risk appears to increase after 5
studies are confounded by chronic intermittent catheterization, chronic years of cyclophosphamide treatment and peaks in years 10 to 14, with
inflammatory bladder stone formation, and other risk factors including standardized incidence ratios of up to 15% (Faurschou et al., 2015).
smoking status and occupational status (Jiang et al., 2009; Vermeulen
et al., 2015). In addition, previous gonorrhea infections have been Environmental Pollution
associated with bladder cancer formation. One prospective study found
a nearly twofold relative risk of bladder cancer formation in men with Exposure to arsenic in drinking water has been associated with the
a history of gonorrhea, and these tumors are more likely to be invasive development of urothelial bladder cancer. Arsenic is a toxic metalloid
(Michaud et al., 2007). The mechanism of action may be related to that enters drinking water from weathering rock. West Bengal,
the production of carcinogens such as nitrosamines that can produce Bangladesh, and Taiwan are the most affected regions worldwide
chronic UTIs. However, there have been no prospective studies examining with concentrations in excess of 4700 µg/L, a 470-fold increase over
the association between UTIs and bladder cancer risk, and much of the recommended amount of 10 µg/L. Studies in these regions and
the early data demonstrating a connection have not been validated parts of Chile, where arsenic exposure is particularly high, have
in more contemporary cohorts. Furthermore, recent explorations into confirmed a significantly higher rate of bladder cancer in these regions
the microbiome of the urinary tract suggest that in some instances (Burger et al., 2013). The mechanism of arsenic-mediated carcino-
certain bacterial and viral colonization of the bladder epithelium have genesis is thought to be multifactorial, including oxidative stress,
a protective effect against tumor formation. Host microbiota have been epigenetic effects, and alterations in DNA repair.
shown to be crucial to maintaining homeostasis in mucosal surfaces,
including the gut and oral cavity; current efforts are underway to define
these mechanisms in the bladder (Whiteside et al., 2015). Diet
Fluids
Indirect Medical Risk Factors
Similar to other cancers, dietary factors have been an area of much
Indirect medical causes of bladder cancer are typically unintended study with regard to bladder cancer risk. Fluid intake is one area of
side effects of treatment. controversy, as initial retrospective studies demonstrated decreased
bladder cancer risk with increased water consumption (Burger et al.,
Pioglitazone 2013). However, these findings were not demonstrated in the European
Prospective Investigation into Cancer and Nutrition (EPIC), which
Pioglitazone is an antidiabetic drug in the thiazolidinedione class showed no link between total fluid intake and bladder cancer risk
of drugs that lower glucose levels among patients with non–insulin- among 250,000 patients with a mean of 9 years follow-up (Ros
dependent diabetes. In 2005 the PROactive randomized trial reported et al., 2011). Similarly, caffeine consumption through ingestion of
an unexpected finding that the pioglitazone arm had higher cases coffee and tea have not been shown to be associated with bladder
of bladder cancer compared with placebo. These findings were cancer risk (Al-Zalabani et al., 2016). The studies that initially
controversial and were subsequently confirmed in some follow-up demonstrated a modest link between coffee consumption and bladder
studies and refuted in others (Boice et al., 1985). cancer were confounded by smoking (Villanueva et al., 2009). Alcohol
is a known risk factor for several cancers primarily involving the
Radiation gastrointestinal tract and the hepatobiliary system. However, there
is currently no evidence to support a link between alcohol consump-
The association of radiation exposure with bladder cancer has been tion and bladder cancer risk, regardless of the amount of alcohol
noted since atomic bomb survivors were found to have markedly consumed (Bagnardi et al., 2015).
3078 PART XIV  Benign and Malignant Bladder Disorders

Foods detecting hematuria is 91% to 100%, the reported specificity of


dipstick tests is 65%, which can lead to unnecessary hematuria
In addition to fluid intake, many carcinogens are ingested via food evaluations (Liu et al., 2016). In contrast, microscopic urinalysis
and excreted in the urine resulting in direct exposure to the urothelium directly examines the urine sediment for the presence of RBCs. For
and potential for increased bladder cancer risk. However, no con- this reason, urine dipstick is discouraged as the sole screening test
clusive data support consumption of a specific food or food group for microhematuria and should be confirmed with microscopic
to mitigate bladder cancer risk. In other malignancies dietary intake urinalysis (American Urological Association, 2015).
of meat has been associated with increased risk. However, among A full hematuria evaluation for bladder cancer includes a focused
more than 500,000 persons in the EPIC study, red meat consumption history and physical, cystoscopy, upper tract imaging, and a urine
was not associated with bladder cancer risk. Dietary intake of fruits culture. For patients with gross hematuria upper tract imaging
and vegetables has been investigated because they are an abundant should include a multi-phase computed tomography (CT) scan with
source of phytochemicals with purported anti-carcinogenic effects. delayed phase images for patients who can tolerate it. Although 2012
In the EPIC study, a higher consumption of fruits and vegetables AUA guidelines suggest a CT urogram (CTU) for the evaluation of
was associated with lower risk of bladder cancer among never and asymptomatic microhematuria (AMH) as well, recent studies suggest
former smokers but not in current smokers. Furthermore, in meta- CTU can be safely replaced with a renal and bladder ultrasound in
analyses adjusting for smoking status, no associations between bladder patients with AMH because upper tract malignancy is exceedingly rare
cancer and total fruits and vegetables, total vegetable, or total fruit in these patients (Tan et al., 2018). For patients with gross hematuria it
intake were identified (Ros et al., 2011; Vieira et al., 2015). is reasonable to obtain a urine cytology and/or urine markers. However,
according to the AUA guidelines, providers should avoid obtaining
Dietary Supplements urine cytology or urine markers as part of the routine evaluation of
the asymptomatic patient with microhematuria (Davis et al., 2012).
Similar to fluid and food intake, no supplemental vitamin and A focused history should inquire into patient risk factors as outlined
minerals have been shown to have carcinogenic or protective effects earlier as well as family history. Although physical exam will rarely
for bladder cancer. In a prospective series of 80,000 persons in the uncover a bladder tumor, a bimanual exam should be performed at
VITamins study, no supplements were identified to be related to the time of transurethral resection of the bladder tumor if the tumor
bladder cancer risk. Similarly, in the Selenium and Vitamin E Cancer appears invasive. A prostate-specific antigen (PSA) blood test may be
Prevention Trial (SELECT), no association between selenium and considered after a discussion between the patient and provider because
vitamin E and bladder cancer was identified. 10% of patients with recurrent gross hematuria will have prostate cancer.

KEY POINTS Cystoscopy


• Bladder cancer is related to age and exposure to The gold-standard test for the diagnosis of bladder cancer is cystoscopy
environmental carcinogens. and biopsy. All adult patients with gross hematuria and all patients
• Smoking accounts for 30% to 40% of all urothelial 35 years and older with microscopic hematuria should undergo
carcinoma. cystoscopic evaluation for malignancy. Flexible office cystoscopy is
• Although bladder cancer is more than 3 times as prevalent as reliable for the diagnosis of bladder cancer as rigid endoscopy
in men, women are more likely to be seen initially with (Grossfeld et al., 2001). Currently white light cystoscopy (WLC)
more advanced tumors and less favorable prognosis. remains the standard of care for the diagnosis of a bladder tumor
• Bladder cancer is the most expensive cancer to treat per and enables the urologist to map and resect all visible tumors.
patient over a patient’s lifetime. Although WLC has excellent sensitivity and specificity for the diagnosis
• No dietary interventions have been successful at reducing of large papillary tumors, it is less reliable for diagnosing small
risk from bladder cancer. papillary tumors and CIS. Porphyrin-induced fluorescence cystoscopy
uses photoactive porphyrins, such as hexaminolevulinate (HAL),
that accumulate preferentially in neoplastic tissue and emit red
DETECTION OF UROTHELIAL CARCINOMA fluorescence under blue-wavelength light with a wavelength of 360
to 450 nm. Although HAL and 5-aminolevulinic acid (5-ALA) have
Unlike many cancers, bladder cancer is rarely incidentally discovered been assessed in clinical studies, only HAL is FDA approved, with
at the time of autopsy. Painless gross hematuria is present in 85% indications for bladder cancer surveillance for rigid and flexible
of patients with newly diagnosed bladder cancer, and microscopic cystoscopy (Video 135.1). In a meta-analysis assessing the utility of
hematuria is present in nearly all patients (Alishahi et al., 2002; HAL-blue light cystoscopy (BLC), BLC detected 14.7% more Ta tumors
Edwards et al., 2006). Hematuria is typically intermittent and can and 40.8% more CIS lesions compared with WLC (Burger et al.,
be related to Valsalva maneuvers. Importantly, any episode of gross 2013). In 26.7% of patients, CIS was detected only by BLC, and
hematuria must be evaluated even if subsequent urinalysis is negative. detection was significantly improved for primary bladder cancer
Use of anti-platelet therapy and anticoagulation should similarly diagnosis and recurrent cancer diagnosis. In the outpatient setting,
not preclude a full hematuria evaluation (Davis et al., 2012). Irritative flexible BLC identified a tumor recurrence not seen under WLC in
voiding symptoms (e.g., frequency, urgency) also may be signs of 20.6% of patients (Daneshmand et al., 2018).
bladder cancer, particularly carcinoma in situ (CIS). Narrow band imaging (NBI) is an endoscopic optical image
Approximately 50% of patients with gross hematuria have an enhancement technique that enhances contrast between mucosal
identifiable cause, and the risk of malignancy in these patients is surfaces and microvascular surfaces without the use of dyes. The
20.4%. By comparison, the risk of malignancy among patients with depth of light penetration into the bladder wall increases with
microscopic hematuria is 2.6% to 5.2% (Davis et al., 2012; Khadra increasing wavelength. NBI illuminates the mucosal surface with
et al., 2000). The risk of malignancy among patients with recurrent light of a narrow bandwidth in the blue (415 nm) and green (540 nm)
gross or microscopic hematuria who had a complete negative light spectrum, which are strongly absorbed by hemoglobin. Con-
hematuria workup is nearly 0 within the first 6 years. As a result, it sequently the vascular structures appear dark brown or green against
is reasonable to delay a repeat hematuria evaluation in patients with a pink or white mucosal background. Studies evaluating NBI have
persistent hematuria 3 to 5 years after their first negative evaluation been performed primarily in single-institution settings or have been
(Davis et al., 2012; Khadra et al., 2000). underpowered; nevertheless, NBI appears to improve the detection
Hematuria is defined as the presence of red blood cells (RBCs) of CIS compared with WLC alone. However, in the most robust
in the urine. The urine dipstick is a common test that screens for study performed thus far, a multicenter randomized trial comparing
hematuria by using reagents that detect peroxidase activity of heme NBI-assisted transurethral resection of bladder tumor (TURBT) to
in RBCs. However, although the sensitivity is for urine dipsticks in conventional white light TURBT, NBI and WLC achieved similar
Chapter 135  Tumors of the Bladder 3079

overall 12-month recurrence rates (27% in WLC and 25.4% in NBI), Nuclear matrix protein 22 (NMP22) is part of a family of proteins
but NBI-assisted TURBT significantly reduced disease recurrence in that provides structure to cell nucleus and is 20 times more prevalent
low-risk patients (27% in WLC vs. 5.6% in NBI) (Naito et al., 2016). in malignant urothelial cells compared with normal cells. There are
currently 2 NMP22 tests FDA approved for use in BC surveillance:
Urine Cytology the original NMP22 laboratory-based quantitative test and a point-
of-care qualitative test. In a meta-analysis of 19 studies, sensitivity
Since 1945, when George Papanicolaou first hypothesized that exfoli- of quantitative NMP22 was 0.69 (95% CI 0.62–0.75), and specificity
ated cells in urine could reliably detect urinary tract malignancies, urine was 0.77 (95% CI 0.70–0.83). Diagnostic accuracy appears to be
cytology has been a standard diagnostic test to aid in the diagnosis similar for initial diagnosis as well as surveillance. Sensitivity of the
of bladder cancer. The contemporary sensitivity and specificity for qualitative point of care NMP22 tests was 0.58 (95% CI 0.39–0.75)
urine cytology in detecting bladder cancer is 31% to 62% and 94% to and specificity was 0.88 (95% CI 0.78 to 0.94), although these
100%, respectively (Planz et al., 2005; Raitanen et al., 2002; van Rhijn ranges are limited by relatively few studies.
et al., 2005). Furthermore, significant variation in cytologic reporting
exists between institutions, further complicating the interpretation of Fluorescence in Situ Hybridization
this subjective diagnostic modality (Karakiewicz et al., 2006; Raitanen
et al., 2002). In 2013 Paris Reporting System for urinary cytology was Fluorescence in situ hybridization (FISH) has been designed for
first introduced at the International Congress of Cytology to standard- bladder cancer detection since 2000 and identified fluorescently
ize cytologic interpretation of urine samples (Fig. 135.5; Rosenthal labeled DNA probes that bind to intranuclear chromosomes (Sokolova
et al., 2017). This reporting schema emphasizes specific diagnostic et al., 2000). The current commercially available FISH assay
categories and cytomorphologic criteria for the reliable diagnosis of (UroVysion) detects aneuploidy of chromosomes 3,7, and 17, and
high-grade urothelial carcinoma. Preliminary reports suggest that homozygous loss of the 9p21 locus in exfoliated urothelial cells. A
the Paris classification schema may decrease the rate of inconclusive positive test is defined as 5 or more urinary cells with gains of two
“atypia” calls and improve risk stratification for patients with high-grade or more chromosomes, 10 or more cells with gain of a single chromo-
urothelial carcinoma (Cowan et al., 2017; Wang et al., 2018). Because some, or homozygous deletion of 9p21 in more than 20% of exfoliated
urine cytology is highly specific, a cytology suspicious or positive for cells. Overall sensitivity of FISH is 0.63 (95% CI 0.50–0.75) and
high-grade urothelial carcinoma should be investigated. In a patient specificity 0.87 (95% CI 0.79–0.93) (Chou et al., 2015). As a reflex
with a negative cystoscopy and persistent positive high-grade urine test in the setting of an atypical cytology, a negative FISH correlates
cytology, it is therefore reasonable to perform random biopsies of the with benign cytologic changes. Among patients with an atypical
bladder and prostatic urethra, as well as ureteral washings for cytology cytology and a negative cystoscopy, 3-year recurrence-free survival
with or without direct ureteroscopic inspection of the upper tracts. increases from 34% to 67% in the setting of a positive FISH (Kim
Urine-based biomarkers have been developed as an adjunct to et al., 2014; Yoder et al., 2007). FISH has relative low sensitivity in
standard diagnostic modalities for bladder cancer diagnosis and detecting low-grade bladder tumors, and there continues to be a
monitoring. Noninvasive testing with improved sensitivity over urine lack of consensus regarding criteria used to evaluate abnormal cells.
cytology has been proposed as a desirable alternative to cystoscopy,
which is costly and uncomfortable. Several urine-based biomarkers Bladder Tumor Antigen
have been developed with improved sensitivity compared with urine
cytology, although most have yet to reach its specificity. We will The bladder tumor antigen (BTA) assay detects two basement mem-
discuss the most widely available and reported urinary biomarkers that brane antigens, human complement factor H-related protein and
have relevance in current and future clinical practice (Table 135.2). complement factor H using monoclonal antibodies. Two forms of

Approach to diagnosis in urinary tract

Cytologic atypia
No present? Yes

Are there
Mild Degree of atypia Severe
fibrovascular cores?
1. N:C>0.5 (required) 1. N:C>0.7 (required)
Plus at least one of: 2. Hyperchromasia (required)
No Yes 2. Hyperchromasia Plus at least one of:
3. Course chromatin 3. Course chromatin
4. Irregular chromatinic rim 4. Irregular chromatinic rim
Check endoscopy,
radiology, and
clinical impression Reason for mild atypia? Quality of atypical cells?
(treatment, etc.)

Yes No Rare, <5-10 cells Many

Negative LGUN Atypical Suspicious HGUC Positive HGUC

Fig. 135.5.  Paris system for urine cytology. HGUC, High-grade urothelial carcinoma; LGUN, low-grade
urothelial neoplasm; N:C, nucleus to cytoplasm ratio. (Modified from Barkan GA, Wojcik EM, Nayar R,
et al.: The Paris System for reporting urinary cytology: the quest to develop a standardized terminology.
Acta Cytol 60:185–197, 2016.)
3080 PART XIV  Benign and Malignant Bladder Disorders

TABLE 135.2  Test Performance of Urinary Biomarkers for Diagnosis of Bladder Cancer

VARIABLE SENSITIVITY SPECIFICITY POSITIVE LR (95% CI) NEGATIVE LR (95% CI)


Sensitivity (95% CI); Specificity (95% CI);
Studies, n_τ2 (P Value) Studies, n_τ2 (P Value)

QUANTITATIVE NMP22
Overall 0.69 (0.62–0.75); 19 0.77 (0.70–0.83); 19 3.05 (2.28–4.10) 0.40 (0.32–0.50)
0.33 (P = 0.0005) 0.62 (P < 0.0001)
Evaluation of 0.67 (0.55–0.77); 9 0.84 (0.75–0.90); 7 4.20 (2.65–6.67) 0.40 (0.29–0.55)
symptoms 0.34 (P = 0.04) 0.45 (P = 0.02)
Surveillance 0.61 (0.49–0.71); 10 0.71 (0.60–0.81); 8 2.10 (1.58–2.80) 0.55 (0.44–0.69)
0.45 (P = 0.04) 0.54 (P = 0.01)

QUALITATIVE NMP22
Overall 0.58 (0.39–0.75); 4 0.88 (0.78–0.94); 4 4.89 (3.23–7.40) 0.48 (0.33–0.71)
0.57 (P = 0.14) 0.50 (P = 0.13)
Evaluation of 0.47 (0.33–0.61); 2 0.93 (0.81–0.97); 2 6.27 (2.98–13.2) 0.58 (0.46–0.72)
symptoms 0.12 (P = 0.38) 0.52 (P = 0.31)
Surveillance 0.70 (0.40–0.89); 2 0.83 (0.75–0.89); 2 4.20 (3.22–5.47) 0.36 (0.16–0.81)
0.74 (P = 0.36) 0.74 (P = 0.31)

QUALITATIVE BTA
Overall 0.64 (0.58–0.69); 22 0.77 (0.73–0.81); 21 2.80 (2.31–3.39) 0.47 (0.30–0.55)
0.26 (P <0.0001) 0.27 (P < 0.0001)
Evaluation of 0.76 (0.67–0.83); 8 0.78 (0.66–0.87); 6 3.42 (2.04–5.74) 0.31 (0.21–0.46)
symptoms 0.21 (P = 0.05) 0.50 (P = 0.02)
Surveillance 0.60 (0.55–0.65); 11 0.76 (0.69–0.83); 8 2.53 (1.92–3.34) 0.52 (0.47–0.59)
0.02 (P = 0.27) 0.26 (P = 0.02)

QUANTITATIVE BTA
Overall 0.65 (0.54–0.75); 4 0.74 (0.64–0.82); 4 2.52 (1.86–3.41) 0.47 (0.37–0.61)
0.10 (P = 0.32) 0.14 (P = 0.27)
Evaluation of 0.76 (0.61–0.87) 1 0.53 (0.38–0.68) 1 1.61 (1.14–2.28) 0.46 (0.26–0.81)
symptoms
Surveillance 0.58 (0.46–0.69); 2 0.79 (0.72–0.85); 2 2.77 (1.66–4.61) 0.54 (0.39–0.76)
<0.0001 (P = 1.0) <0.0001 (P = 1.0)

FISH
Overall 0.63 (0.50–0.75); 11 0.87 (0.79–0.93); 11 5.02 (2.93–8.60) 0.42 (0.30–0.59)
0.74 (P = 0.01) 0.90 (P = 0.003)
Evaluation of 0.73 (0.50–0.88); 2 0.95 (0.87–0.98); 1 14.2 (5.2–39) 0.29 (0.14–0.60)
symptoms 0.36 (P = 0.40) <0.0001 (P = 1.0)
Surveillance 0.55 (0.36–0.72); 7 0.80 (0.66–0.89); 6 2.75 (1.95–3.89) 0.56 (0.42–0.76)
0.85 (P = 0.03) 0.80 (P = 0.03)

IMMUNOCYT
Overall 0.78 (0.68–0.85); 14 0.78 (0.72–0.82); 14 3.49 (2.82–4.32) 0.29 (0.20–0.41)
0.71 (P = 0.003) 0.25 (P = 0.001)
Evaluation of 0.85 (0.78–0.90); 6 0.83 (0.77–0.87); 7 4.89 (3.79–6.30) 0.18 (0.12–0.26)
symptoms 0.10 (P = 0.30) 0.11 (P = 0.07)
Surveillance 0.75 (0.64–0.83); 7 0.76 (0.70–0.81); 8 3.09 (2.56–3.72) 0.33 (0.24–0.46)
0.34 (P = 0.05) 0.14 (P = 0.04)

CXBLADDER
Evaluation of 0.82 (0.70–0.90) 1 0.85 (0.81–0.88) 1 5.53 (4.28–7.15) 0.21 (0.13–0.36)
symptomsa

BTA, Bladder tumor antigen; CI, confidence interval; FISH, fluorescence in situ hybridization; LR, likelihood ratio; NMP22, nuclear matrix protein 22.
a
Based on threshold selected for a specificity of 0.85.
Data from Chou R, Gore JL, Buckley D, et al.: Urinary biomarkers for diagnosis of bladder cancer: a systematic review and meta-analysis. Ann Int
Med 2015 (online journal): http://annals.org/aim/fullarticle/2466370/urinary-biomarkers-diagnosis-bladder-cancer-systematic-review-meta-analysis.
Chapter 135  Tumors of the Bladder 3081

the BTA test are available and FDA approved for the diagnosis and
follow-up of bladder cancer. The BTA stat test is a qualitative dipstick-
KEY POINTS
based point-of-care test. Sensitivity of qualitative BTA was 0.64 (95% • Painless gross hematuria is the key presenting symptom of
CI 0.58–0.69) across 22 studies, and specificity was 0.77 (95% CI bladder cancer, occurring in 85% of newly diagnosed
0.73–0.81). In addition, a quantitative, ELISA-based BTA assay (BTA patients.
TRAK) has also been developed, with reported sensitivity of 0.65 (95% • A full hematuria evaluation for bladder cancer includes a
CI 0.54–0.75) and specificity of 0.74 (95% CI 0.64 to 0.82). The high focused history and physical, cystoscopy, upper tract
false positivity of both BTA assays could be related to the presence of imaging, and a urine culture.
cross-reactivity with red blood cells because complement factor H is • The gold-standard test for the diagnosis of bladder cancer
present in high concentrations in serum and thus is associated with is cystoscopy and biopsy. Increasingly, HAL BLC and
high false positives in hematuria (Longo et al., 2018). narrow band imaging are being used as adjuncts to
cystoscopy in identifying occult malignancy.
Immunocyt • There are various urine markers that can be used to
evaluate secreted proteins or shed cells in the hope of
Immunocyt is a cell-based adjunct to urine cytology, developed in noninvasively detecting bladder cancer. None of these
the late 1990s. Fluorescent-labeled antibodies are directed against markers has a high enough sensitivity or specificity to
three antigens that are commonly expressed by exfoliated urothelial replace office cystoscopy.
cells: a glycosylated from of carcinoembryonic antigen (CEA) and two
mucins. Across all studies, the sensitivity of ImmunoCyt was 0.78 (95%
CI 0.68–0.85), and specificity was 0.78 (95% CI 0.72–0.82) (Chou inflammatory areas that can be confused for cancer. Squamous
et al., 2015). In one study, ImmunoCyt in combination with urine metaplasia is very common, affecting up to 40% of women and 5%
cytology improved sensitivity from 50% to 90%; however, specificity of men, and can be attributed to a history of urinary tract infection,
was lower (80% to 72%) when compared with cytology alone (Toma trauma, and prior surgery (Ozbey et al., 1999). A biopsy to diagnose
et al., 2004). Like urine cytology, the test is operator dependent, with glandular or squamous metaplasia is unnecessary and no treatment
high interobserver variability and poor agreement (Pode et al., 1999); is required or efficacious.
one study has suggested that the variability in test performance can
be overcome with adequate training (Messing et al., 2005). Papilloma and Inverted Papilloma
CxBladder An inverted papilloma (Fig. 135.6) is a benign proliferative lesion
that is associated with chronic inflammation or bladder outlet
CxBladder is a cell-based urine assay that comprises five mRNA obstruction and can be located throughout the bladder but most
fragments (HOXA13, CDC2, MDK, CXCR2, and IGFBP5) (Holyoake commonly on the trigone, accounting for less than 1% of all bladder
et al., 2008; O’Sullivan et al., 2012). With a limited number of studies tumors. Painless gross hematuria is the most common representing
performed thus far, test sensitivity is approximately 85% and specificity symptom, with microscopic hematuria and irritative voiding symptoms
85% (Kavalieris et al., 2015; O’Sullivan et al., 2012). Although still also common. Inverted papillomas grow in an endophytic pattern
in need of validation in independent, multi-institutional datasets, along the lamina propria to form nests and anastomosing trabeculae.
initial comparative studies have suggested the CxBladder may have When diagnosed according to strictly defined criteria (e.g., lack of
improved sensitivity and specificity over urine cytology and other cytologic atypia), inverted papillomas behave in a benign fashion
protein-based assays (Breen et al., 2015; Lough et al., 2018). with a 1% chance of recurrence.
The currently available urinary biomarkers to detect bladder cancer Urothelial papilloma (Fig. 135.7) is characterized by a benign
miss many patients with bladder cancer and yield false positives in proliferative growth of delicate stalks lined by normal-appearing
others. Head-to-head comparisons of urinary biomarkers have been urothelium. Papillomas were historically characterized as low-grade
too limited thus far to reach firm conclusions about comparative Ta malignant bladder tumors but are now thought to be benign
effectiveness. It is thus our opinion and current practice that in the with no risk of local or systemic invasion. Although papillomas have
setting of microhematuria, the addition of any current urine biomark- fibroblast growth factor receptor-3 (FGFR3) mutations, which are
ers does not preclude a cystoscopy and thus does not change manage- commonly found in bladder cancer, they lack markers of aggressive
ment. Similarly, in the case of gross hematuria, a negative test result behavior, including TP53 and RB mutations (Montironi and Lopez-
is associated with an approximately 10% probability of having bladder Beltran, 2005).
cancer (Longo et al., 2018). As a result, a negative test would also
not preclude a cystoscopy. Furthermore, the field is rapidly evolving
with the discovery of novel genomics and bladder cancer subtypes.
Multiplex gene assays have been developed by multiple teams and
are in need of further validation. Because none of the currently
available biomarkers have reached the necessary threshold for the
diagnosis of bladder cancer, they are currently used in combination
with cystoscopy as a surveillance strategy for patients with a known
history of non–muscle-invasive bladder cancer.

PATHOLOGY
There are numerous benign tumors of the bladder, each with its
own clinical presentation and disease burden.

Epithelial Metaplasia
Epithelial metaplasia is defined by focal area of transformed uro-
thelium, with otherwise normal nuclear and cellular morphology.
Typically, epithelial metaplasia is located on the trigone and may
be composed of either squamous metaplasia or glandular metaplasia.
Glandular metaplasia in particular consists of clumps of raised, red Fig. 135.6.  Inverted papilloma.
3082 PART XIV  Benign and Malignant Bladder Disorders

Fig. 135.7.  Papilloma.

Fig. 135.8.  Von Brunn nest with cystitis cystica.


Nephrogenic Adenoma
Nephrogenic adenoma is a rare tumor caused by chronic irritation
of the urothelium and can arise from trauma, prior surgery, renal
transplantation, intravesical chemotherapy, stones, catheters, and
infection. Nephrogenic adenoma is composed of glandular appear-
ing tubules similar to renal tubules that involve mucosa and sub
mucosa of the bladder. Cystoscopically, nephrogenic adenoma may
appear like a papillary neoplasm and may be hard to distinguish
from more aggressive bladder neoplasms. Lesions may cause dysuria
hematuria, prompting cystoscopic investigation. Recurrence does occur
but is uncommon and can be safely managed with repeat resection
(Yi et al., 2018).

Leukoplakia
Leukoplakia of the bladder has similar features to squamous meta-
plasia with the additional characteristic of white, flaky plaques floating
in the bladder. If not diagnosed incidentally, presenting symptoms
are typically frequent, recurrent infections with urinary urgency and
frequency. Histologic analyses demonstrate that the bladder mucosa
is chronically inflamed, displaying diffuse squamous metaplasia of Fig. 135.9.  Cystitis glandularis.
keratinizing type leukoplakia. Although on other squamous epithelial
surfaces leukoplakia is premalignant, there exists some controversy
as to whether bladder leukoplakia predisposes patients to malignancy. and pseudostratified columnar epithelium. Cells mature from the
Historic reports described leukoplakia as premalignant, with 37% basal basement membrane cells, which are small and cuboidal, to
risk of carcinoma (Benson et al., 1984). However, more recently a intermediate cells to superficial umbrella cells in an orderly fashion.
connection between leukoplakia as an independent risk factor for The surface has large umbrella cells that may have nuclear atypia
bladder cancer has been disproven. Staack et al. (2006) performed and form asymmetrical units. These umbrella cells form a urine
cytogenic studies of bladder leukoplakia in 77 cases using DNA bladder barrier preventing toxins from transforming urothelial cells.
analysis for tumor suppressor gene TP52 and found no evidence to Precursor lesions are a continuum from hyperplasia to atypia to
suggest that this condition was premalignant; therefore cystoscopy, dysplasia and finally cancer. The 2004 World Health Organization
biopsy, and further treatment were unnecessary. classification system for urothelial neoplasia classifies pre-malignant
lesions as urothelial hyperplasia (flat and papillary), reactive atypia,
Cystitis Cystica and Glandularis urothelial atypia of unknown significance (AUS), urothelial dysplasia,
and low-grade intraurothelial neoplasia (Hodges et al., 2010).
Cystitis cystica and/or glandularis is a common finding in normal Urothelial hyperplasia may be flat or papillary (Fig. 135.10) and
bladders, typically associated with inflammation or chronic obstruc- is characterized by thickening of the mucosa to 10 or more cell
tion. These benign lesions represent cystic nests that are lines by layers. Although the nuclei may be slightly enlarged, no cytologic
columnar or cuboidal cells and are generally associated with prolifera- atypia is present and mitoses are absent. Although in isolation
tion of Von Brunn nests (Fig. 135.8) (Semins and Schoenberg, 2007). urothelial hyperplasia is not thought to have malignant potential,
Although cystitis glandularis (Fig. 135.9) can transform into adeno- hyperplasia is frequently seen in conjunction with pre-malignant
carcinoma, the risk is low; however, regular endoscopic evaluation and malignant lesions. Hyperplastic flat lesions may be precursors
of these entities is recommended. Treatment is transurethral resection to low-grade papillary tumors, evidenced by mutation in FGFR3,
and relief of the obstruction or inflammatory condition. and loss of heterozygosity at 3q22-q24, 5q22-q31, 10q26, 13q14,
and 17p13 in benign hyperplastic lesions from bladders with uro-
Precursor Malignant Lesions thelial carcinoma (Majewski et al., 2008; van Oers et al., 2006). On
cystoscopy, papillary urothelial hyperplasia can be indistinguishable
Normal urothelium comprises multilayered mucosa, 4 to 7 cells from low-grade papillary lesions and is often present adjacent to
thick, which features the transition between nonkeratinizing squamous low-grade papillary tumors.
Chapter 135  Tumors of the Bladder 3083

Fig. 135.10.  Papillary urothelial hyperplasia. Fig. 135.11.  Carcinoma in situ.

Reactive atypia is characterized by nuclear abnormalities occurring


in the background of an inflamed urothelium (Hodges et al., 2010).
Typically patients have a history of chronic inflammation, regenera-
tion, or reaction from noxious stimuli and have irritative voiding
symptoms. Endoscopically, reactive atypia is edematous and inflamed
appearing. Microscopically, although nuclei may be prominent and
enlarged, the mucosa proceeds in an orderly fashion from basal to
intermediate to superficial cell types, and nuclear polarity is main-
tained. Similar to hyperplasia, reactive atypia does not appear to
independently evolve into carcinoma; however, atypia often coexists
with dysplasia and carcinoma in situ and thus mandates rigorous
investigation.
Atypia of unknown significance (AUS) is used when it is unclear
whether observed urothelial changes are premalignant or reactive.
Microscopically, AUS is characterized by inflammation of the lamina
propria and nuclear atypia disproportionate to the degree of atypia.
CK20 and CK44 immunohistochemical staining, which are positively
expressed in AUS, can aid in distinguishing AUS from dysplasia
(Kunju et al., 2005). AUS independently does not appear to lead to
urothelial carcinoma and is a diagnosis of limited clinical utility. Fig. 135.12.  Papillary urothelial neoplasm of low malignant potential.
Urothelial dysplasia is characterized by a normal urothelial
thickness and an altered cytologic appearance of urothelial cells.
Normal cytoplasmic clearing is lost particularly in basal and intermedi-
ate cells, with loss of nuclear polarity, nuclear enlargement, hyper- a non-cohesive cellular structure. The cells are large, pleomorphic,
chromosia, and nuclear membrane irregularity. The lamina propria chromatin clumping, and often with abnormal mitotic figures. A
is typically normal, and comparison with normal urothelium can characteristic that separates dysplasia from CIS is the loss of umbrella
help differentiate reactive atypia from dysplasia. Ck20 and P53 cells. The genetic abnormalities of CIS are more similar to high-grade,
staining typically highlight the dysplastic cells. Umbrella cells are invasive tumors of the urinary tract, furthering the concept that CIS
typically present with dysplasia but not with CIS. Dysplasia, although is a precursor to high-grade invasive carcinoma. These include altera-
difficult to distinguish from CIS in some cases, is a good indication tions to RB, TP53, and PTEN (Sanchez-Carbayo et al., 2002). Interest-
of urothelial instability and a marker of recurrence or progression ingly, no FGFR3 mutations have been reported in CIS, suggesting a
in those with a history of urothelial cancer. Approximately 14% to very different tumor lineage from low-grade papillary tumors, which
19% of patients with urothelial dysplasia will progress to urothelial have frequent FGFR3 mutations (Zieger et al., 2009). Clinically, CIS
cancer; however, in the setting of a prior history of bladder cancer, in association with invasive tumors carries a worse prognosis, with
urothelial dysplasia will lead to CIS in 60% of cases (Baithun et al., a 45% to 65% 5-year mortality rate (Montironi et al., 2009).
1988; Cheng et al., 1999, 2000). Papillary urothelial neoplasm of low malignant potential
(PUNLMP) (Fig. 135.12) constitutes a diagnosis with particularly
Malignant Lesions good prognosis, and there is controversy as to whether it should
carry the label of cancer (Montironi and Lopez-Beltran, 2005; Sauter,
Urothelial carcinoma is the most common malignancy of the urinary 2004). PUNLMP is a papillary growth with minimal cytologic atypia
tract and is the second most common cause of death among all that is more than seven cells thick, generally solitary, and typically
genitourinary tumors. Non–muscle-invasive bladder cancer, compris- located on the trigone. Cell polarity is maintained, and nuclei are
ing low- and high-grade noninvasive papillary neoplasms, CIS, and mildly enlarged. Five-year recurrence rates after PUNLMP are estimated
urothelial carcinoma with invasion into the lamina propria (T1), to be 20.1%, and these recurrences are nearly always PUNLMP or
make up approximately 70% of all bladder urothelial carcinoma. low-grade papillary lesions. Progression to MIBC among patients
CIS (Fig. 135.11) is characterized by noninvasive, flat high-grade with PUNLMP is less than 1%, and it is unclear if these rare high-grade
tumors. There is severe nuclear atypia, loss of cellular polarity, and recurrences represent true stage progression or the presence of a
3084 PART XIV  Benign and Malignant Bladder Disorders

new, separate bladder cancer (Maxwell et al., 2015). TERT promoter Staging
mutations and FGFR3 mutations have been identified in more than
60% of PUNLMP cases, which creates a similar profile to low-grade The American Joint Committee on Cancer (AJCC) in combination
papillary Ta lesions. The low rate of HRAS mutations (3%) compared with the International Union Cancer Consortium meets regularly to
with the 33% incidence in other noninvasive urothelial tumors, determine tumor, nodes, and metastases (TNM) staging classification.
however, may speak to the relative indolent, non-aggressive course The 2017 staging system is shown in Table 135.3 (Amin et al., 2017).
of PUNLMP lesions (Rodriguez Pena et al., 2017). Broadly speaking, non–muscle-invasive bladder cancer comprises
Low-grade urothelial carcinoma is typically papillary (Fig. 135.13) stage 0 (noninvasive) and stage I (invasion into subepithelial con-
with a fibrovascular stalk and frequent papillary branching with nective tissue), muscle-invasive organ-confined bladder cancer is
increased cellular size, some nuclear atypia, and occasional mitotic of stage II, muscle-invasive locally advanced bladder cancer is stage
figures. These tumors almost universally display alterations of genes III, and metastatic disease is stage IV. Whereas prior AJCC editions
in chromosome 9 and frequent mutations in FGFR3, PI3K or, viewed positive lymph nodes as stage IV regardless of the quantity
alternatively, Ras genes (Dinney et al., 2004; Lopez-Knowles et al., or location, the 2017 AJCC staging update now views nodal disease
2006; van Rhijn et al 2010). Low-grade Ta tumors frequently recur in the absence of systemic metastases as stage III (Paner et al., 2018).
48% to 71% of the time; however, stage or grade progression is rela- NMIBC is further subdivided into low-, intermediate-, and high-risk
tively infrequent, occurring 2% to 12% of the time, depending on disease. Low-risk NMIBC comprises a primary (i.e., not recurrent),
the series (Montironi and Lopez-Beltran, 2005). low-grade papillary (Ta), solitary tumors less than 3 cm. Intermediate-
High-grade papillary urothelial cancer (Fig. 135.14) is composed risk NMIBC is histologically confirmed by multiple and/or recurrent
of fused papillary stalk with high-grade cancer in the urothelial layer. and/or large (>3 cm) low-grade Ta tumors. High-risk NMIBC involves
There is a disordered growth pattern, numerous mitotic figures present, tumors with any high-grade histologic features (i.e., CIS or T1) (Kamat
and there are pleomorphic cells with exaggerated nuclei. If left et al., 2014). In addition, the 2017 AJCC recommends subcategoriza-
untreated, more than 80% of high-grade papillary tumors invade tion of T1 urothelial carcinoma into T1a (superficial) and T1b (deep)
the underlying urothelial stroma. High-grade papillary tumors are lamina propria invasion (Fig. 135.15) to help stratify the heteroge-
considered biologically, and clinically, to be a precursor to invasive neous group of T1 tumors, which are at a 50% risk of upstaging to
high-grade urothelial cancer. T2 or higher and a 33% risk of being upstaged to non–organ confined
(Badalato et al., 2011; Svatek et al., 2010). These stratifications suggest
that the deeper the tumor invades into the lamina propria, the
worse the survival.
Muscle-invasive disease is subdivided into T2a and T2b. T2a
includes invasion into the inner half of the muscularis propria,
although T2b is deeper into the outer half. It is controversial whether
depth of muscle invasion makes significant impact on disease, and
this is complicated by sampling variability of muscle on TURBT. For
this reason, the AJCC staging guidelines view T2a and T2b as similar
in the current staging schema. T3 disease constitutes invasion outside
of the bladder proper into the periadipose tissue. T3a disease involves
microscopic extension, whereas T3b is macroscopic extension. T4a
disease is invasion of the prostatic stroma, uterus, or vagina, and
T4b disease is invasion of the pelvic or abdominal wall.
Prostatic urethral cancer is associated with bladder cancer in 90%
of cases, although among patients with primary bladder urothelial
cancer, urethral involvement is relatively rare, occurring in 3% of
patients. Most prostatic urethral urothelial carcinoma is CIS, and
extension of the tumor into the prostatic urethra without stroma
invasion does not carry an adverse prognosis for patients with known
bladder cancer. These tumors can be difficult to treat with intravesical
therapy, however, and as a result among patients undergoing frequent
intravesical therapy, the rate of prostatic urethral CIS is elevated
Fig. 135.13.  Low-grade papillary urothelial neoplasm. (Table 135.4; Mungan et al., 2005). Prostate stromal invasion,
however, particularly if it is direct extension from the bladder through
the muscle into the prostate, does carry a poor prognosis, with 5-year
overall survival of less than 25% in historic series (Donat et al.,
2001; Esrig et al., 1996). For this reason only patients with prostate
stromal invasion are considered to be T4.

Molecular Biology
Major progress has been made in recent years toward improving our
understanding of the molecular makeup of urothelial carcinoma.
Several molecular alterations are common in bladder cancer, including
FGFR3 alterations, PI3K pathway alterations, activation of mitogen-
activated protein kinase (MAPK) signaling, defects in cell cycle regula-
tion, and Hedgehog and WNT signaling (Knowles and Hurst, 2015).
Genes encoding proteins that control the cell cycle are frequently
mutated in bladder cancer, particularly MIBC. Inactivation of RB1,
and CDKN2A are very common, and p53 inactivation occurs in as
many as 76% of MIBCs (Cancer Genome Atlas Research Network,
2014). Similarly loss of the tumor suppressor gene RB1 is common
in MIBC and leads to amplification and overexpression of the
oncogene E2F3 (Knowles and Hurst, 2015). The genetic alterations
Fig. 135.14.  High-grade urothelial cancer invading the lamina propria. that are the hallmark of NMIBC are alterations in the fibroblast
Chapter 135  Tumors of the Bladder 3085

TABLE 135.3  American Joint Committee on Cancer/International Union Cancer Consortium 2017 Tumor,
Nodes, and Metastases (TNM) Staging Classification

STAGE STAGE GROUPING STAGE DESCRIPTION

0a Ta The cancer is a noninvasive papillary carcinoma (Ta). It has grown toward the hollow center of the
bladder but has not grown into the connective tissue or muscle of the bladder wall.
N0 It has not spread to nearby lymph nodes (N0) or distant sites (M0).
M0
0is Tis The cancer is a flat, noninvasive carcinoma (Tis), also known as flat carcinoma in situ (CIS). The
cancer is growing in the inner lining layer of the bladder only. It has not grown inward toward the
hollow part of the bladder, nor has it invaded the connective tissue or muscle of the bladder wall.
N0 It has not spread to nearby lymph nodes (N0) or distant sites (M0).
M0
I T1 The cancer has grown into the layer of connective tissue under the lining layer of the bladder but
has not reached the layer of muscle in the bladder wall (T1).
N0 The cancer has not spread to nearby lymph nodes (N0) or to distant sites (M0).
M0
II T2a or T2b N0 The cancer has grown into the inner (T2a) or outer (T2b) muscle layer of the bladder wall, but it has
not passed completely through the muscle to reach the layer of fatty tissue that surrounds the
bladder.
M0 The cancer has not spread to nearby lymph nodes (N0) or to distant sites (M0).
IIIA T3a, T3b or T4a The cancer has grown through the muscle layer of the bladder and into the layer of fatty tissue that
surrounds the bladder (T3a or T3b).
N0 It may have spread into the prostate, uterus, or vagina, but it is not growing into the pelvic or
abdominal wall (T4a).
M0 The cancer has not spread to nearby lymph nodes (N0) or to distant sites (M0).
OR The cancer has:
T1-4a grown into the layer of connective tissue under the lining of the bladder wall (T1), OR
N1 into the muscle layer of the bladder wall (T2), OR
M0 into the layer of fatty tissue that surrounds the bladder, (T3a or T3b) OR
it may have spread into the prostate, uterus, or vagina, but it is not growing into the pelvic or
abdominal wall (T4a).
AND the cancer has spread to a nearby lymph node in the true pelvis (N1).
It has not spread to distant sites (M0).
IIIB The cancer has:
T1-T4a grown into the layer of connective tissue under the lining of the bladder wall (T1), OR
N2 or N3 M0 into the muscle layer of the bladder wall (T2), OR
into the layer of fatty tissue that surrounds the bladder (T3a or T3b), OR
it may have spread into the prostate, uterus, or vagina, but it is not growing into the pelvic or
abdominal wall (T4a).
AND the cancer has spread to 2 or more lymph nodes in the true pelvis (N2) or to lymph nodes
along the common iliac arteries (N3).
It has not spread to distant sites (M0).
IVA The cancer has grown through the bladder wall into the pelvic or abdominal wall (T4b).
T4b The cancer has not spread to nearby lymph nodes (N0) or to distant sites (M0).
N0
M0
OR
Any T The cancer may or may not have grown through the wall of the bladder into nearby organs (Any T).
Any N It may or may not have spread to nearby lymph nodes (Any N).
M1a It has spread to a distant set of lymph nodes (M1a).
IVB Any T The cancer may or may not have grown through the wall of the bladder into nearby organs (Any T).
Any N It may or may not have spread to nearby lymph nodes (Any N).
M1b It has spread to 1 or more distant organs (such as the bones, liver or lungs) (M1b).

growth factor receptor-3 (FGFR3) gene and deletions of chromosome bladder epithelium, FGFR3 is expressed in the IIIb isoform and
regions 9p and 9q. Up to 80% of low-grade papillary (Ta) bladder binds to FGF1. In bladder cancer cell lines, a switch from the IIIb
tumors have activating point mutations in FGFR3, whereas approxi- to the IIIc FGFR3 isoform may induce oncogenesis via paracrine
mately 10% to 20% of muscle-invasive bladder tumors have FGFR3 and autocrine signaling (Tomlinson et al., 2005; Zhang et al., 2006).
mutations (Cappellen et al., 1999; Zieger et al., 2005). In normal In addition, FGFR3 fusion proteins, which are present in many bladder
3086 PART XIV  Benign and Malignant Bladder Disorders

Muscularis mucosae

Muscularis mucosae

Fig. 135.15.  High grade T1a (left) and Tlb (right) urothelial cancer.

TABLE 135.4  Clinical Significance of Different Non–Muscle-Invasive Urothelial Cancer Categories in the World Health
Organization 2004 Grading System

PAPILLARY NEOPLASM OF LOW-GRADE PAPILLARY HIGH-GRADE CARCINOMA


PAPILLOMA LOW MALIGNANT POTENTIAL CARCINOMA (PAPILLARY AND CIS)

Recurrence (%) 0–8 27–47 48–71 55–58


Grade progression (%) 2 11 7 N/A
Stage progression (%) 0 0–4 2–12 27–61
Survival (%) 100 93–100 82–96 74–90

CIS, Carcinoma in situ; N/A, not applicable.


Data from Montironi R, Lopez-Beltran A: The 2004 WHO classification of bladder tumors: a summary and commentary. Int J Surg Pathol 13(2):143–53, 2005.

tumors, comprise highly activated oncoproteins of FGFR3 fused to of activated b-catenin with PTEN deletion promoted bladder cancer
TACC3 or BAI1 (Williams et al., 2013). development (Ahmad et al 2011).
The MAPK signaling pathway comprises another set of key genetic A new framework is emerging for understanding the heterogeneity
alterations in bladder cancer pathogenesis. Ras (HRAS or KRAS) of bladder cancer and how that molecular diversity plays a role in
pathway alterations are mutually exclusive from FGFR3 mutations treatment response. Historically, bladder tumors were thought to
because they are in the same RAS-MEK-ERK pathway. Ras genes arise from two distinct pathways. In the first pathway, tumors arise
are from a family of transforming oncogenes that were originally from urothelial proliferation and differentiate into low-grade non-
identified in T24 bladder cancer cell lines (McBride et al., 1982). invasive papillary tumors. These tumors are enriched in FGFR3
However, unlike FGFR3, RAS mutations occur with equal incidence mutations and have a relatively low total mutational burden. In the
in NMIBC and MIBC. second pathway, tumors start as dysplastic cells and transform into
Point mutations in PIK3CA are found in 25% of NMIBC and CIS and into high-grade invasive tumors.
less commonly in MIBC. FGFR3 and PIK3CA work together in a However, recent genome-wide expression and sequencing studies
similar pathway, and thus both mutations are often seen in the same have identified genes and pathways that are clear drivers in urothe-
tumors. PI3K pathway alterations are critical in the PTEN pathway lial cancer development. A more complex picture with molecular
as PTEN negatively regulates PI3K. Downregulation of PTEN in MIBC subclasses that are agnostic to conventional tumor stage and grade
is associated with TP53 alterations and poor prognosis (Platt et al., has emerged.
2009; Puzio-Kuter et al., 2009). In a pre-clinical transgenic model In 2014, the comprehensive molecular characterization of MIBC
of bladder cancer, dual deletion of the genes encoding p53 and was performed by the Cancer Genome Atlas (TCGA) (Network TCGAR
PTEN leads to the development of metastatic bladder cancer (Knowles 2014). In that analysis of mRNA, microRNA, and protein expression
and Hurst 2015). It appears that the protein phosphatase activity of from 129 muscle invasive tumors, four distinct clusters were identified
PTEN, and not its lipid activity, drives bladder cancer invasion and resembling intrinsic subtypes of breast cancer. Subdividing urothelial
metastasis (Gildea et al 2004). Additional studies demonstrate that bladder cancer into intrinsic subtypes based on breast cancer was
the WNT/b-catenin pathway often coordinates with PI3K/PTEN to independently confirmed by groups from Lund, MDAnderson, and
promote bladder cancer development and growth. Three dimensional UNC (Sjodahl et al 2012; Choi et al 2014; Damrauer et al 2014).
organoid cultures have revealed the importance of the WNT/b-catenin These systems partially overlap, and broadly speaking characterizes
pathway in bladder cancer cell proliferation (Yoshida et al 2018). basal and luminal tumor subtypes (Fig. 135.16). The luminal group,
These studies confirmed earlier experiments showing that expression characterized by high levels of FGFR3, GATA3, KRT20, and PPARG
Chapter 135  Tumors of the Bladder 3087

UNC Luminal Basal

MDA Luminal TP53-like Basal MIBC

TCGA I II IV III

NMIBC
Lund Uro A Genomically unstable Infiltrated SCC-like Uro B
+ MIBC

Transcription PPARG, RXRA, GATA3, PPARG, RXRA, GATA3, SNAI1 STAT3 None
factors and FOXA1 FOXA1, and FOXM1 and ZEB2 and FOXM1 identified

Immune FGFR3
Actionable FGFR3, ERBB2, Immune checkpoints and
targets and EGFR ERBB2 checkpoints and EGFR EGFR

Fig. 135.16.  Molecular subtypes of bladder cancer. MDA, MD Anderson Cancer Center nomenclature;
MIBC, muscle-invasive bladder cancer; NMIBC, non–muscle-invasive bladder cancer; SCC, small cell
carcinoma; TCGA, The Cancer Genome Atlas nomenclature; UNC, University of North Carolina nomenclature.
(Modified from Compérat E, Varinot J, Moroch J, et al.: A practical guide to bladder cancer pathology.
Nat Rev Urol 15:143–154, 2018.)

Luminal Basal/Squamous
KRT20+, GATA3, FOXA1+ KRT5,6,14+, GATA3-, FOXA1-

Luminal-papillary Luminal-infiltrated Luminal Basal/Squamous Neuronal

FGFR3 mut, fusion, amp Low purity UPKs Female SOX2


Papillary histology EMT markers (TWIST1, ZEB1) KRT20 Squamous differentiation DLX6
SHH+ miR-200 family SNX31 Basal keratin markers MSI1
Low CIS Medium CD274 (PD-L1), CTLA-4 High CD274 (PD-L1), CTLA4 PLEKHG4B
Myofibroblast markers Immune infiltrates E2F3/SOX4 amp
Wild type p53 High cell cycle

Low risk Anti-PD-L1, PD-1, CTLA-4 Targeted therapy? Anti-PD-L1, PD-1, CTLA-4 Etoposide/Cisplatin NAC
NAC* Cisplatin-based NAC** Cisplatin-based NAC
FGFR3 inhibitors
** Low response rate
* Low predicted
likelihood of response,
based on preliminary
data

Fig. 135.17.  Proposed schema of expression-based, subtype-stratified therapeutic approach as a framework


for prospective hypothesis testing in clinical trials. (From Robertson AG, Kim J, Al-Ahmadie H, et al.: Com-
prehensive molecular characterization of muscle-invasive bladder cancer. Cell Press 171:540–56.e25, 2017.)

mutations was biologically less aggressive compared with the basal- of bladder cancer (Bai et al., 2016). A 2017 follow-up study from
squamous subtype, characterized by squamous differentiation, and the TCGA of 412 muscle-invasive tumors identified 5 expression
expression of CK5, CK6, CK14, and low levels of FOXA1 and subtypes that may stratify to different treatments (Fig. 135.17).
GATA3RNA and protein. In addition, that study identified a subset of patients with a high
Ultimately, grouping bladder cancer into distinct molecular clusters mutational load, driven by an APOBEC-mediated mutagenesis.
may help identify certain patients that may benefit from specific APOEBEC comprises a family of cytidine deaminases that are
therapies. For example, the MD Anderson classification identified a involved in mRNA editing. This APOBEC related subset of patients
distinct subgroup of luminal tumors that they termed “P53-like”, had a 75% 5-year survival, far higher than the typical MIBC patient.
that which were chemoresistant and associated with a poor prognosis
(Choi et al., 2014). In addition, the UNC classification identified a
claudin-low molecular subtype of MIBC enriched for RB1, EP300, HISTOLOGIC VARIANTS OF UROTHELIAL CARCINOMA
and NCOR1, with decreased FGFR3, KDM6A, and PPARG amplifica-
tion or expression. These claudin-low tumors are immune infiltrated Urothelial carcinoma of the bladder had previously been labeled
and actively immune suppressed and may benefit from immune transitional cell carcinoma of the bladder because of its known
checkpoint inhibitors that have been FDA approved for the treatment propensity for cellular differentiation into other tumor types, such
3088 PART XIV  Benign and Malignant Bladder Disorders

as SCC, adenocarcinoma, and clear cell carcinoma. Recently, a wider 60% who undergo transurethral resection with intravesical BCG.
spectrum of histologic variants of urothelial cancer has been identified Similarly, for muscle-invasive disease neoadjuvant chemotherapy
and investigated that includes distinct growth patterns of urothelial does not appear to be effective, and immediate cystectomy is cur-
carcinoma, altered cellular differentiation of stromal reactions, and rently encouraged (Kamat et al., 2007). Molecular profiling of the
unique responses to treatment and external stimuli. The more micropapillary variant suggest that these tumors are characterized by
common histologic variants are discussed here. ERBB2 amplification and activation of miR-296 and RUVBL1 target
genes (Guo et al., 2016; Isharwal et al., 2018; Ross et al., 2014). The
Micropapillary Variant micropapillary variants appear to evolve almost exclusively through
a luminal pathway, displaying enrichment of PPAR-gamma and sup-
Micropapillary urothelial carcinoma was first described in 1994 and pression of p63 genes. Similar to conventional luminal tumors, a
occurs in approximately 0.7% to 2.2% of all urothelial tumors (Amin subset of micropapillary cancers demonstrate activation of wild-type
et al., 1994; Johansson et al., 1999; Fig. 135.18). There is a strong p53, and these represent the most aggressive phenotypes with the
male predominance associated with micropapillary urothelial cancer, worst prognosis (Guo et al., 2016).
with reports of a male-to-female ratio as high as 37:1. Micropapillary
urothelial carcinoma is associated with advanced disease, as more Sarcomatoid Variant
than 50% are stage T3 or T4 at diagnosis; as such 5- and 10-year
overall survival is 51% and 24%, lower than with other common The sarcomatoid variant has a prevalence of 0.3% to 0.6% of all
histologies (Kamat et al., 2007). However, even among lower-stage bladder cancers (Amin, 2009; Moschini et al., 2017a; Wright et al.,
bladder tumors, micropapillary variant is associated with a worse 2007). Historically they were described separately as carcinosarcoma
prognosis because these tumors are associated with a progression rate and sarcomatoid carcinoma, although molecular studies suggest that
from non–muscle-invasive to muscle-invasive disease of 70%, with they have a common clonal origin (Armstrong et al., 2009; Gronau
a high subsequent metastatic rate despite treatment (Kamat et al., et al., 2002; Wright et al., 2007). Patients with the sarcomatoid variant
2007). Histologically, micropapillary urothelial carcinoma exhibit are seen with large infiltrative masses, often at higher stages with a
small tight clusters or thin papillae of atypical cells with eosinophilic 5-year cancer-specific survival as low as 28.4% across all stages (Wang
to clear cytoplasm surrounded by prominent retraction artifact (Watts et al., 2010). Even when controlling for stage they have worse outcomes
and Hansel, 2010). Unlike other histologic subtypes, any amount than pure urothelial tumors. These patients have a poor response
of micropapillary urothelial carcinoma may be present for it to to systemic chemotherapy, and thus immediate radical cystectomy
be defined as micropapillary and clinically managed as such. is often considered when feasible; furthermore, our group and others
The most effective therapy for all stages of micropapillary urothelial have proposed intraoperative radiation therapy (IORT) at the time
carcinoma is surgical resection. Treatment with transurethral resec- of radical cystectomy for these high-risk patients (Kates et al., 2017;
tion and bacillus Calmette-Guérin therapy is ineffective unless the Wang et al., 2010).
tumor is completely resected, and early cystectomy is encouraged
for NMIBC, with a cancer-specific survival of 72% compared with Plasmacytoid Variant
The plasmacytoid variant of urothelial carcinoma is rare, comprising
less than 1% of all urothelial tumors (Lopez-Beltran et al., 2009;
KEY POINTS Moschini et al., 2017b). According to the 2016 WHO classification
• Recent genome-wide expression and sequencing studies of urothelial carcinoma, plasmacytoid urothelial carcinoma now
have identified genes and pathways that are clear drivers in includes the signet ring variant, which is defined by the presence of
urothelial cancer development. cytoplasmic vacuoles with or without intracellular mucin. The
• A molecular taxonomy is emerging broadly defined by plasmacytotic variant microscopically has increased eosinophilic
urothelial cells types. cytoplasm, with eccentrically placed and enlarged hyperchromatic
• The luminal phenotype is characterized by high levels of nuclei. These tumors typically are seen in an advanced stage with
FGFR3, GATA3, KRT20, and PPARG mutations. deep invasion into the bladder muscle and perivesical tissues, and
• The basal phenotype is characterized by squamous with characteristic intraperitoneal metastatic spread. This aggressive
differentiation, and expression of CK5, CK6, CK14, and phenotype is characterized by truncating somatic alterations in the
low levels of FOXA1 and GATA3 RNA and protein CDH1 gene, which occurs in 84% of cases according to one study
expression. (Al-Ahmadie et al., 2016). Although plasmacytoid variant is chemo-
sensitive and thus treated with neoadjuvant chemotherapy whenever
possible, relapses are common with median survival among non-
metastatic patients just 17.7 months (Dayyani et al., 2013; Kaimakliotis
et al., 2014; Ricardo-Gonzalez et al., 2012).

Nested Variant
The nested variant is a rare but aggressive cancer that has a male-
to-female ratio of 6:1 and can be confused with benign lesions such
as Von Brunn nests that are located in the lamina propria, cystitis
cystica, and inverted papillomas (Fig. 135.19). Although the nested
variant is typically upstaged at the time of cystectomy and found to
be locally advanced at the time of presentation, stage-matched
comparisons between nested variant and pure urothelial carcinoma
suggest similar rates of recurrence and survival (Beltran et al., 2014;
Linder et al., 2013).

Urothelial Carcinoma With Divergent Differentiation


Urothelial carcinoma with squamous differentiation is present in
up to 16% to 22% of patients with bladder cancer, although these
may be underestimates, and studies suggest that up to 60% of bladder
Fig. 135.18.  Micropapillary urothelial cancer. cancers have squamous elements (Black et al., 2009; Shanks and
Chapter 135  Tumors of the Bladder 3089

Fig. 135.19.  Nested variant of urothelial cancer. Fig. 135.20.  Small cell carcinoma of the bladder. Tumor composed of sheets
and nests of basophilic-appearing tumor cells with high nucleocytoplasmic
ratio. Histologic features resemble small cell carcinoma seen in the lung. Tumor
Iczkowski, 2009). The under-reporting of squamous differentiation cells stain and show diffuse and strong immunoreactivity with neuroendocrine
and the percent of the tumor involved make it difficult to assess markers chromogranin (inset) and synaptophysin.
how this divergent histology affects clinical outcomes (Shah et al.,
2013). Squamous differentiation is associated with the basal molecular
subtype of bladder cancer, which is seen at a more advanced stage
and is more focally aggressive (Choi et al., 2014). These tumors may
be associated with recurrence and progression for patients with
localized and advanced stages (Li et al., 2018; Minato et al., 2017).
Glandular differentiation comprises small tubular or gland-like
spaces in otherwise conventional urothelial carcinoma and is present
in approximately 10% of patients with urothelial bladder cancer.
Similar to squamous differentiation, glandular differentiation is
associated with a high stage at presentation and with clinical to
pathological upstaging at the time of radical cystectomy; however,
their clinical behavior does not appear to be different from conven-
tional urothelial carcinoma, with similar rates of recurrence-free and
overall survival (Kim et al., 2012; Mitra et al., 2014).

Nonurothelial Malignancy
Small Cell
Small cell carcinoma (Fig. 135.20) is a rare, poorly differentiated
neuroendocrine neoplasm that primarily arises in the lung but can Fig. 135.21.  Squamous cell cancer with associated urothelial cancer.
occur in extrapulmonary sites, including the prostate and bladder.
Although patients typically are seen with hematuria, there have been
reports of paraneoplastic syndromes associated with small cell urothelial carcinoma, and there may be a role for adjuvant chemo-
carcinoma including hypercalcemia, Cushing syndrome, and sensory therapy. In addition, historically small cell was thought to be a
neuropathy. Small cell carcinoma of the bladder has a high somatic nonsurgical entity and most appropriate for chemoradiation, whereas
mutational burden driven by an APOBEC-mediated mutational some groups still advocate for trimodal therapy. The combination
process, and TP53, RB1, and TERT promoter mutations are frequently of chemotherapy with cystectomy is associated with the highest
present (Chang et al., 2018). Evolutionary studies have suggested response rates in published series, and further investigation into the
that small cell carcinoma of the bladder may represent a dedifferentia- safety of chemoradiation in small cell carcinoma is warranted (Bryant
tion from urothelial carcinoma, rather than having a shared lineage et al., 2016; Siefker-Radtke et al., 2004).
from small cell carcinoma of the lung.
Small cell carcinoma of the bladder is clinically aggressive and Squamous Cell Cancer
chemosensitive. As such, platinum-based chemotherapy is recom-
mended as the initial treatment regardless of the stage of presentation. Chronic infection with S. haematobium or other bacteria leads to SCC of
A variety of chemotherapy regimens have been reported, but cisplatin the bladder (Fig. 135.21). In countries with endemic schistosomiasis,
and etoposide are the current treatment of choice based on the SCC thus makes up the majority of bladder cancer and presents a
standard regimen for small cell carcinoma of the lung. Siefker-Radtke unique growth pattern and treatment algorithm. In the United States
et al. (2004) reported that patients with primary small cell treated and Europe SCC is more uncommon, occurring in 2% to 5% of all
initially with cystectomy observed a 36% 5-year disease-free survival, bladder cancers (Rausch et al., 2014). The central factor contributing
which was not improved with adjuvant chemotherapy. However, to the development of SCC is chronic inflammation of the urinary
patients who underwent neoadjuvant chemotherapy observed a tract. Historically patients with spinal cord injury requiring chronic
cancer-specific survival of 78%. However, Kaushik et al. (2015) have catheterization had an incidence of SCC of 2.3%; however, this
demonstrated, in a separate study, that the aggressive features of incidence rate has declined to 0.39% in contemporary series likely
small cell may be related to a high stage at diagnosis. Further, when because of the trend toward intermittent self-catheterization. This
matched stage-for-stage, small cell responds similarly to conventional supports the concept that chronic infection and foreign bodies can
3090 PART XIV  Benign and Malignant Bladder Disorders

ruling out adenocarcinomas from other organs is recommended. At


our center we have our patients undergo a colonoscopy whenever
an adenocarcinoma is identified. Most primary bladder adenocar-
cinomas present with muscle invasive disease, and radical cystectomy
is the standard treatment. Currently there is no level 1 data to support
the use of chemotherapy or radiation therapy in the neoadjuvant
or adjuvant setting. When chemotherapy is delivered the traditional
MVAC or GC regimens used for conventional urothelial carcinoma
are of little benefit. Often a regimen including 5-flourouracil and
cisplatin is used based on experience with adenocarcinoma at other
organ sites (Dadhania et al., 2015).

Urachal Adenocarcinoma
Urachal adenocarcinoma is a rare cancer, accounting for approxi-
mately one-third of all bladder adenocarcinomas, that arises from
the allantoic remnant that connects the bladder to the umbilical cord
during embryogenesis. Although urachal remnants are typically lined
by urothelial cells, urachal cancer is almost always adenocarcinoma
Fig. 135.22.  Adenocarcinoma of the bladder. Transurethral resection of in origin. The 2016 WHO classification introduced criteria for the
the prostate fragment showing extensive involvement by primary bladder diagnosis of urachal adenocarcinoma based on the work by Gopalan
adenocarcinoma. Note presence of cribriform and glandular architecture of et al. (2009): the tumor should be located in the bladder dome or
tumor (inset). anterior abdominal wall, with the epicenter of the tumor occurring in
the bladder wall; the tumor could not have widespread cystitis cystica,
and there should be a thorough investigation for a secondary source
lead to bladder cancer formation. Data regarding the carcinogenesis for the adenocarcinoma. Urachal adenocarcinomas are difficult to stage
of SCC are largely limited to schistosoma-induced SCC, detailed using standard TNM staging because nearly all are muscle invasive
earlier in this chapter. because of their anatomic location. Staging of urachal carcinoma
Despite having a lower incidence of nodal and metastatic disease, thus follows the system developed by Sheldon et al. (1984). Stage
SCC often is seen initially at more advanced local stages than I tumors are carcinomas confined to the urachal mucosa, stage II
conventional urothelial carcinoma. Radical cystectomy is the mainstay tumors are confined to the urachus, stage III tumors are urachal tumors
treatment for SCC, and there is no clear consensus regarding the that extend locally into the bladder (IIIA), abdominal wall (IIIB),
use of neoadjuvant chemotherapy before surgery. Preoperative radia- or peritoneum (IIIC), and stage IV tumors are metastatic (Sheldon
tion therapy was extensively studied in the 1970s and 1980s, but et al., 1984). The standard treatment for urachal adenocarcinoma
was associated with worse survival outcomes and thus should only is en bloc resection of the bladder dome, urachal ligament, and
be used if the patient is not undergoing cystectomy (Swanson et al umbilicus (Siefker-Radtke et al., 2003).
1990; Johnson et al 1976; Costello et al 1984). Adjuvant and
intraoperative radiation therapy have also been proposed but there
is no clear consensus on how or when it should be used. There is KEY POINTS
limited data regarding the molecular biology of SCC, particularly • Eighty percent of urothelial carcinomas contain some
that which is not associated with schistosomiasis. Fibroblast growth mixed differentiation, most commonly squamous cell.
factor (FGF2) has been shown to be associated with aggressive • Altered growth patterns, particularly micropapillary and
pathologic features and worse outcomes after radical cystectomy nested variants, carry a poor prognosis, even for non–
(Youssef et al 2015). muscle-invasive disease.
• Neoadjuvant chemotherapy appears to be active in mixed
Adenocarcinoma tumors containing adenocarcinoma or squamous cell
cancer but not against altered growth patterns such as in
Adenocarcinoma (Fig. 135.22) may be a primary cancer arising from micropapillary and nested urothelial cancer.
the bladder urothelium with a pure glandular phenotype, or may • Small cell carcinoma of the bladder should be treated as
be a secondary cancer from a distant metastasis, or more commonly metastatic disease with institution of chemotherapy
from direct extension from the colon, prostate, endometrium, or followed by either radiation therapy or surgery for
cervix. Although the urachus is not an anatomic component of the elimination of the local disease.
urinary bladder, urachal adenocarcinomas share similar pathologic
and clinical features to bladder adenocarcinoma. Risk factors for
adenocarcinoma are a history of bladder exstrophy, schistosomiasis,
and chronic irritation or obstruction. Because secondary bladder REFERENCES
adenocarcinomas are more common than primary adenocarcinomas, The complete reference list is available online at ExpertConsult.com.
Chapter 135  Tumors of the Bladder 3090.e1

Cappellen D, De Oliveira C, Ricol D, et al: Frequent activating mutations of


REFERENCES FGFR3 in human bladder and cervix carcinomas, Nat Genet 23:18–20,
Ahmad I, Morton JP, Singh LB, et al: β-Catenin activation synergizes with 1999.
PTEN loss to cause bladder cancer formation, Oncogene 30:178–189, 2011. Chang MT, Penson A, Desai NB, et al: Small-cell carcinomas of the bladder
Al-Ahmadie HA, Iyer G, Lee BH, et al: Frequent somatic CDH1 loss-of-function and lung are characterized by a convergent but distinct pathogenesis, Clin
mutations in plasmacytoid variant bladder cancer, Nat Genet 48:356–358, Cancer Res 24:1965–1973, 2018.
2016. Chappidi MR, Kates M, Tosoian JJ, et al: Evaluation of gender-based disparities
Alishahi S, Byrne D, Goodman CM, et al: Haematuria investigation based in time from initial haematuria presentation to upper tract urothelial
on a standard protocol: emphasis on the diagnosis of urological malignancy, carcinoma diagnosis: analysis of a nationwide insurance claims database,
J R Coll Surg Edinb 47:422–427, 2002. BJU Int 120:2017.
Al-Zalabani AH, Stewart KFJ, Wesselius A, et al: Modifiable risk factors for Cheng L, Cheville JC, Neumann RM, et al: Flat intraepithelial lesions of the
the prevention of bladder cancer: a systematic review of meta-analyses, urinary bladder, Cancer 88:625–631, 2000.
Eur J Epidemiol 31:811–851, 2016. Cheng L, Cheville JC, Neumann RM, et al: Natural history of urothelial
American Cancer Society: Cancer facts & figures 2017, 2017 (website): https:// dysplasia of the bladder, Am J Surg Pathol 23:443–447, 1999.
www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/ Choi JB, Lee EJ, Han K-D, et al: Estimating the impact of body mass index
cancer-facts-figures-2017.html. on bladder cancer risk: stratification by smoking status, Sci Rep 8:947,
American Urological Association: Five more things physicians and patients should 2018.
question. Choosing Wisely, 2015 (website): http://www.choosingwisely.org/ Choi W, Porten S, Kim S, et al: Identification of distinct basal and luminal
wp-content/uploads/2015/02/AUA-Choosing-Wisely-List.pdf. subtypes of muscle-invasive bladder cancer with different sensitivities to
Amin MB, Edge SB: American Joint Committee on Cancer. AJCC cancer staging frontline chemotherapy, Cancer Cell 25:152–165, 2014.
manual, 8th ed, 2017, Springer International Publishing. Chou R, Gore JL, Buckley D, et al: Urinary biomarkers for diagnosis of bladder
Amin MB, Ro JY, el-Sharkawy T, et al: Micropapillary variant of transitional cancer a systematic review and meta-analysis, Ann Intern Med 163(12):922–
cell carcinoma of the urinary bladder. Histologic pattern resembling ovarian 932, 2015.
papillary serous carcinoma, Am J Surg Pathol 18:1224–1232, 1994. Chrouser K, Leibovich B, Bergstralh E, et al: Bladder cancer risk following
Amin MB: Histological variants of urothelial carcinoma: diagnostic, therapeutic primary and adjuvant external beam radiation for prostate cancer, J Urol
and prognostic implications, Mod Pathol 22:96–118, 2009. 174:107–110, 2005.
Antoni S, Ferlay J, Soerjomataram I, et al: Bladder cancer incidence and Cohn JA, Vekhter B, Lyttle C, et al: Sex disparities in diagnosis of bladder
mortality: a global overview and recent trends, Eur Urol 71:96–108, 2017. cancer after initial presentation with hematuria: a nationwide claims-based
Armstrong AB, Wang M, Eble JN, et al: TP53 mutational analysis supports investigation, Cancer 120:555–561, 2014.
monoclonal origin of biphasic sarcomatoid urothelial carcinoma (carci- Costello AJ, Tiptaft RC, England HR, et al: Squamous cell carcinoma of
nosarcoma) of the urinary bladder, Mod Pathol 22:113–118, 2009. bladder, Urology 23:234–236, 1984.
Aydin Mungan N, Kiemeney LALM, Van Dijck JAAM, et al: Gender differences Cowan ML, Rosenthal DL, VandenBussche CJ: Improved risk stratification
in stage distribution of bladder cancer, Urology 55(3):368–371, 2000. for patients with high-grade urothelial carcinoma following application
Badalato G, Patel T, Hruby G, et al: Does the presence of muscularis propria of the Paris System for Reporting Urinary Cytology, Cancer Cytopathol
on transurethral resection of bladder tumour specimens affect the rate of 125:427–434, 2017.
upstaging in cT1 bladder cancer?, BJU Int 108:1292–1296, 2011. Cumberbatch MG, Rota M, Catto JWF, et al: The role of tobacco smoke in
Bagnardi V, Rota M, Botteri E, et al: Alcohol consumption and site-specific bladder and kidney carcinogenesis: a comparison of exposures and meta-
cancer risk: a comprehensive dose–response meta-analysis, Br J Cancer analysis of incidence and mortality risks, Eur Urol 70:458–466, 2016.
112:580–593, 2015. Cumberbatch MGK, Cox A, Teare D, et al: Contemporary occupational
Bai Y, Ni M, Cooper B, et al: Claudin-low bladder tumors are immune carcinogen exposure and bladder cancer, JAMA Oncol 1:1282, 2015.
infiltrated and actively immune suppressed, BMC Genomics 15:2016. Curtis S: MIlken Institute 2016 Report on Bladder Cancer. MIlken Inst, 2016.
Baithun SI, Rogers HS, Martin JE, et al: Primary dysplasia of bladder, Lancet Czerniak B, Dinney C, McConkey D: Origins of bladder cancer, Annu Rev
1:483, 1988. Pathol 11:149–174, 2016.
Beltran AL, Cheng L, Montironi R, et al: Clinicopathological characteristics Dadhania V, Czerniak B, Guo CC: Adenocarcinoma of the urinary bladder,
and outcome of nested carcinoma of the urinary bladder, Virchows Arch Am J Clin Exp Urol 3:51–63, 2015.
465:199–205, 2014. Damrauer JS, Hoadley KA, Chism DD, et al: Intrinsic subtypes of high-grade
Benson RC, Swanson SK, Farrow GM: Relationship of leukoplakia to urothelial bladder cancer reflect the hallmarks of breast cancer biology, Proc Natl
malignancy, J Urol 131:507–511, 1984. Acad Sci U S A 111:3110–3115, 2014.
Bernardo C, Cunha MC, Santos JH, et al: Insight into the molecular basis Daneshmand S, Patel S, Lotan Y, et al: Efficacy and Safety of blue light flexible
of Schistosoma haematobium-induced bladder cancer through urine cystoscopy with hexaminolevulinate in the surveillance of bladder cancer:
proteomics, Tumor Biol 37:11279–11287, 2016. a Phase III, Comparative, multicenter study, J Urol 199:1158–1165, 2018.
Black PC, Brown GA, Dinney CPN: The impact of variant histology on the Daugherty SE, Lacey JV, Pfeiffer RM, et al: Reproductive factors and menopausal
outcome of bladder cancer treated with curative intent, Urol Oncol 27:3–7, hormone therapy and bladder cancer risk in the NIH-AARP Diet and
2009. Health Study, Int J Cancer 133:462–472, 2013.
Boice JD, Day NE, Andersen A, et al: Second cancers following radiation Davis R, Jones JS, Barocas DA, et al: Diagnosis, evaluation and follow-up of
treatment for cervical cancer. An international collaboration among cancer asymptomatic microhematuria (AMH) in adults: AUA Guideline, J Urol
registries, J Natl Cancer Inst 74:955–975, 1985. 188:2473–2481, 2012.
Bray F, Ren JS, Masuyer E, et al: Global estimates of cancer prevalence for 27 Dayyani F, Czerniak BA, Sircar K, et al: Plasmacytoid urothelial carcinoma,
sites in the adult population in 2008, Int J Cancer 132:1133–1145, 2013. a chemosensitive cancer with poor prognosis, and peritoneal carcinomatosis,
Breen V, Kasabov N, Kamat AM, et al: A holistic comparative analysis of J Urol 189:1656–1661, 2013.
diagnostic tests for urothelial carcinoma: a study of Cxbladder Detect, Dinney CPN, McConkey DJ, Millikan RE, et al: Focus on bladder cancer,
UroVysion® FISH, NMP22® and cytology based on imputation of multiple Cancer Cell 6:111–116, 2004.
datasets, BMC Med Res Methodol 15:45, 2015. Donat SM, Genega EM, Herr HW, et al: Mechanisms of prostatic stromal
Bryant CM, Dang LH, Stechmiller BK, et al: Treatment of small cell carcinoma invasion in patients with bladder cancer: clinical significance, J Urol
of the bladder with chemotherapy and radiation after transurethral resection 165:1117–1120, 2001.
of a bladder tumor, Am J Clin Oncol 39:69–75, 2016. Dy GW, Gore JL, Forouzanfar MH, et al: Global burden of urologic cancers,
Burger M, Catto JWF, Dalbagni G, et al: Epidemiology and risk factors of 1990-2013, Eur Urol 71:437–446, 2017.
urothelial bladder cancer, Eur Urol 63:234–241, 2013. Edwards TJ, Dickinson AJ, Natale S, et al: A prospective analysis of the
Burger M, Grossman HB, Droller M, et al: Photodynamic diagnosis of diagnostic yield resulting from the attendance of 4020 patients at a protocol-
non–muscle-invasive bladder cancer with hexaminolevulinate cystoscopy: driven haematuria clinic, BJU Int 97:301–305, 2006.
a meta-analysis of detection and recurrence based on raw data, Eur Urol Esrig D, Freeman JA, Elmajian DA, et al: Transitional cell carcinoma involving
64:846–854, 2013. the prostate with a proposed staging classification for stromal invasion, J
Buteau A, Seideman CA, Svatek RS, et al: What is evaluation of hematuria Urol 156:1071–1076, 1996.
by primary care physicians? Use of electronic medical records to assess Faurschou M, Mellemkjaer L, Voss A, et al: Prolonged risk of specific malignan-
practice patterns with intermediate follow-up, Urol Oncol 32:128–134, cies following cyclophosphamide therapy among patients with granulo-
2014. matosis with polyangiitis, Rheumatology 54:1345–1350, 2015.
Cancer Genome Atlas Research Network TCGAR: Comprehensive molecular Ferlay J, Soerjomataram I, Ervik M, et al: GLOBOCAN 2012 v1.1, Cancer
characterization of urothelial bladder carcinoma, Nature 507:315–322, Incidence and Mortality Worldwide: IARC CancerBase No. 11 [Internet]. Lyon,
2014. Fr. Int. Agency Res. Cancer, 2014. Available from: http://globocan.iarc.fr.
3090.e2 PART XIV  Benign and Malignant Bladder Disorders

Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, et al: Cancer incidence and Karagas MR, Park S, Warren A, et al: Gender, smoking, glutathione-S-transferase
mortality patterns in Europe: estimates for 40 countries in 2012, Eur J variants and bladder cancer incidence: a population-based study, Cancer
Cancer 49:1374–1403, 2013. Lett 219(1):63–69, 2005.
Fitzmaurice C, Allen C, Barber RM, et al: Global, regional, and national Karakiewicz PI, Benayoun S, Zippe C, et al: Institutional variability in the
cancer incidence, mortality, years of life lost, years lived with disability, accuracy of urinary cytology for predicting recurrence of transitional cell
and disability-adjusted life-years for 32 cancer groups, 1990 to 2015, JAMA carcinoma of the bladder, BJU Int 97:997–1001, 2006.
Oncol 3:524, 2017. Kates M, Chappidi MR, Brant A, et al: High dose-rate intra-operative radiation
Garcia-Closas M, Malats N, Silverman D, et al: NAT2 slow acetylation, GSTM1 therapy during high risk genitourinary surgery: initial observations and a
null genotype, and risk of bladder cancer: results from the Spanish Bladder proposal for its study in bladder cancer, Bladder Cancer 3:191–199, 2017.
Cancer Study and meta-analyses, Lancet 366:649–659, 2005. Kaushik D, Frank I, Boorjian SA, et al: Long-term results of radical cystectomy
Garg T, Pinheiro LC, Atoria CL, et al: Gender disparities in hematuria evaluation and role of adjuvant chemotherapy for small cell carcinoma of the bladder,
and bladder cancer diagnosis: a population based analysis, J Urol Int J Urol 22:549–554, 2015.
192:1072–1077, 2014. Kavalieris L, O’Sullivan PJ, Suttie JM, et al: A segregation index combining
Gildea JJ, Herlevsen M, Harding MA, et al: PTEN can inhibit in vitro organo- phenotypic (clinical characteristics) and genotypic (gene expression) biomark-
typic and in vivo orthotopic invasion of human bladder cancer cells even ers from a urine sample to triage out patients presenting with hematuria
in the absence of its lipid phosphatase activity, Oncogene 23:6788–6797, who have a low probability of urothelial carcinoma, BMC Urol 15:23, 2015.
2004. Khadra MH, Pickard RS, Charlton M, et al: A prospective analysis of 1,930
Golka K, Reckwitz T, Kempkes M, et al: N -Acetyltransferase 2 (NAT2) and patients with hematuria to evaluate current diagnostic practice, J Urol
Glutathione S-Transferase µ (GSTM1) in bladder-cancer patients in a highly 163:524–527, 2000.
industrialized area, Int J Occup Environ Health 3:105–110, 1997. Kiemeney LALM: Hereditary bladder cancer, Scand J Urol Nephrol 42:110–115,
Gopalan A, Sharp DS, Fine SW, et al: Urachal carcinoma, Am J Surg Pathol 2008.
33:659–668, 2009. Kim PH, Sukhu R, Cordon BH, et al: Reflex fluorescence in situ hybridization
Gronau S, Menz CK, Melzner I, et al: Immunohistomorphologic and molecular assay for suspicious urinary cytology in patients with bladder cancer with
cytogenetic analysis of a carcinosarcoma of the urinary bladder, Virchows negative surveillance cystoscopy, BJU Int 114(3):354–359, 2014.
Arch 440:436–440, 2002. Kim SP, Frank I, Cheville JC, et al: The impact of squamous and glandular
Grossfeld GD, Litwin MS, Wolf JS, et al: Evaluation of asymptomatic micro- differentiation on survival after radical cystectomy for urothelial carcinoma,
scopic hematuria in adults: the American Urological Association best J Urol 188:405–409, 2012.
practice policy–part II: patient evaluation, cytology, voided markers, imaging, Kleinerman RA, Boice JD, Storm HH, et al: Second primary cancer after
cystoscopy, nephrology evaluation, and follow-up, Urology 57:604–610, treatment for cervical cancer. An international cancer registries study, Cancer
2001. 76:442–452, 1995.
Guo CC, Dadhania V, Zhang L, et al: Gene expression profile of the clinically Knowles MA, Hurst CD: Molecular biology of bladder cancer: new insights
aggressive micropapillary variant of bladder cancer, Eur Urol 70:611–620, into pathogenesis and clinical diversity, Nat Rev Cancer 15:25–41, 2015.
2016. Koebnick C, Michaud D, Moore SC, et al: Body mass index, physical activity,
Hall EJ, Wuu C-S: Radiation-induced second cancers: the impact of 3D-CRT and bladder cancer in a large prospective study, Cancer Epidemiol Biomarkers
and IMRT, Int J Radiat Oncol Biol Phys 56:83–88, 2003. Prev 17:1214–1221, 2008.
Hartge P, Harvey EB, Linehan WM, et al: Unexplained excess risk of bladder Koshiaris C, Aveyard P, Oke J, et al: Smoking cessation and survival in lung,
cancer in men, J Natl Cancer Inst 82:1636–1640, 1990. upper aero-digestive tract and bladder cancer: cohort study, Br J Cancer
Health NC for CDP and HP (US) O on S and: The Health Consequences of 117:1224–1232, 2017.
Smoking—50 Years of Progress [Internet]. Heal. Consequences Smoking—50 Kunju LP, Lee CT, Montie J, et al: Utility of cytokeratin 20 and Ki-67 as
Years Prog. A Rep. Surg. Gen. Centers for Disease Control and Prevention markers of urothelial dysplasia, Pathol Int 55:248–254, 2005.
(US), 2014 (website): http://www.ncbi.nlm.nih.gov/pubmed/24455788. Leal J, Luengo-Fernandez R, Sullivan R, et al: Economic burden of bladder
(Cited 26 April 2018). cancer across the European Union, Eur Urol 69:438–447, 2016.
Hemelt M, Yamamoto H, Cheng KK, et al: The effect of smoking on the male Li G, Hu J, Niu Y: Squamous differentiation in pT1 bladder urothelial car-
excess of bladder cancer: a meta-analysis and geographical analyses, Int J cinoma predicts poor response for intravesical chemotherapy, Oncotarget
Cancer 124:412–419, 2009. 9:217–223, 2018.
Ho C-H, Sung K-C, Lim S-W, et al: Chronic indwelling urinary catheter increase Li P, Chen J, Miyamoto H: Androgen receptor signaling in bladder cancer,
the risk of bladder cancer, even in patients without spinal cord injury, Cancers (Basel) 9(2):2017. http://www.ncbi.nlm.nih.gov/pubmed/28241422.
Medicine (Baltimore) 94:e1736, 2015. (online journal).
Hodges KB, Lopez-Beltran A, Davidson DD, et al: Urothelial dysplasia and Linder BJ, Frank I, Cheville JC, et al: Outcomes following radical cystectomy
other flat lesions of the urinary bladder: clinicopathologic and molecular for nested variant of urothelial carcinoma: a matched cohort analysis, J
features, Hum Pathol 41:155–162, 2010. Urol 189:1670–1675, 2013.
Holyoake A, O’Sullivan P, Pollock R, et al: Development of a multiplex RNA Liu J-J, Jones JS, Rao PK: Urinalysis in the evaluation of hematuria, J Am
urine test for the detection and stratification of transitional cell carcinoma Med Assoc 315:2726, 2016.
of the bladder, Clin Cancer Res 14:742–749, 2008. Longo TA, Brousell SC, Inman BA: Urine cytology and existing urinary biomark-
Isharwal S, Huang H, Nanjangud G, et al: Intratumoral heterogeneity of ers for bladder cancer. In Hansel D, Lerner S, editors: Precision molecular
ERBB2 amplification and HER2 expression in micropapillary urothelial pathology of bladder cancer, Switzerland, 2018, Springer, pp 137–155.
carcinoma, Hum Pathol 77:63–69, 2018. Lopez-Beltran A, Requena MJ, Montironi R, et al: Plasmacytoid urothelial
Islami F, Stoklosa M, Drope J, et al: Global and regional patterns of tobacco carcinoma of the bladder, Hum Pathol 40:1023–1028, 2009.
smoking and tobacco control policies, Eur Urol Focus 1:3–16, 2015. Lopez-Knowles E, Hernández S, Malats N, et al: PIK3CA Mutations are an
Jamal A, King BA, Neff LJ, et al: Current cigarette smoking among adults early genetic alteration associated with FGFR3 mutations in superficial
— United States, 2005–2015, MMWR Morb Mortal Wkly Rep 65:1205–1211, papillary bladder tumors, Cancer Res 66:7401–7404, 2006.
2016. Lough T, Luo Q, O’Sullivan P, et al: Clinical utility of Cxbladder Monitor
Jiang X, Castelao JE, Groshen S, et al: Urinary tract infections and reduced for patients with a history of urothelial carcinoma: a physician–patient
risk of bladder cancer in Los Angeles, Br J Cancer 100:834–839, 2009. real-world clinical data analysis, Oncol Ther 1–13, 2018. http://link.springer.
Johansson SL, Borghede G, Holmäng S: Micropapillary bladder carcinoma: com/10.1007/s40487-018-0059-5. (online journal).
a clinicopathological study of 20 cases, J Urol 161:1798–1802, 1999. Majewski T, Lee S, Jeong J, et al: Understanding the development of human
Johnson AM, O’Connell MJ, Miyamoto H, et al: Androgenic dependence of bladder cancer by using a whole-organ genomic mapping strategy, Lab
exophytic tumor growth in a transgenic mouse model of bladder cancer: Invest 88:694–721, 2008.
a role for thrombospondin-1, BMC Urol 8:2008. Malats N, Real FX: Epidemiology of bladder cancer, Hematol Oncol Clin North
Johnson DE, Schoenwald MB, Ayala AG, et al: Squamous cell carcinoma of Am 29:177–189, 2015.
the bladder, J Urol 115:542–544, 1976. Maxwell JP, Wang C, Wiebe N, et al: Long-term outcome of primary papillary
Kaimakliotis HZ, Monn MF, Cheng L, et al: Plasmacytoid bladder cancer: urothelial neoplasm of low malignant potential (PUNLMP) including
variant histology with aggressive behavior and a new mode of invasion PUNLMP with inverted growth, Diagn Pathol 10:3, 2015.
along fascial planes, Urology 83:1112–1116, 2014. McBride OW, Swan DC, Santos E, et al: Localization of the normal allele of
Kamat AM, Dinney CPN, Gee JR, et al: Micropapillary bladder cancer, Cancer T24 human bladder carcinoma oncogene to chromosome 11, Nature
110:62–67, 2007. 300:773–774, 1982.
Kamat AM, Witjes JA, Brausi M, et al: Defining and treating the spectrum of Messing EM, Teot L, Korman H, et al: Performance of urine test in patients
intermediate risk nonmuscle invasive bladder cancer, J Urol 192:305–315, monitored for recurrence of bladder cancer: a multicenter study in the
2014. United States, J Urol 174:1238–1241, 2005.
Chapter 135  Tumors of the Bladder 3090.e3

Michaud DS, Platz EA, Giovannucci E: Gonorrhoea and male bladder cancer Roswall N, Olsen A, Christensen J, et al: Micronutrient intake and risk of
in a prospective study, Br J Cancer 96:169–171, 2007. urothelial carcinoma in a prospective Danish Cohort, Eur Urol 56:764–770,
Minato A, Fujimoto N, Kubo T: Squamous differentiation predicts poor 2009.
response to cisplatin-based chemotherapy and unfavorable prognosis in Rushton L, Bagga S, Bevan R, et al: Occupation and cancer in Britain, Br J
urothelial carcinoma of the urinary bladder, Clin Genitourin Cancer Cancer 102:1428–1437, 2010.
15:1063–1067, 2017. Sanchez-Carbayo M, Socci ND, Charytonowicz E, et al: Molecular profiling
Mitra AP, Bartsch CC, Bartsch G, et al: Does presence of squamous and of bladder cancer using cDNA microarrays: defining histogenesis and
glandular differentiation in urothelial carcinoma of the bladder at cystectomy biological phenotypes, Cancer Res 62:6973–6980, 2002.
portend poor prognosis? An intensive case-control analysis, Urol Oncol Sauter G: Tumors of the urinary system in: World Health Organisation classification
32:117–127, 2014. of tumors. Pathol Genet Tumors Urin Syst Male Genit Organs, 110–123,
Miyamoto H, Yang Z, Chen Y-T, et al: Promotion of bladder cancer development 2004.
and progression by androgen receptor signals, J Natl Cancer Inst 99:558–568, Scosyrev E, Golijanin D, Wu G, et al: The burden of bladder cancer in
2007. men and women: analysis of the years of life lost, BJU Int 109:57–62,
Montironi R, Lopez-Beltran A, Scarpelli M, et al: 2004 World Health Organiza- 2012.
tion classification of the noninvasive urothelial neoplasms: inherent Scosyrev E, Noyes K, Feng C, et al: Sex and racial differences in bladder cancer
problems and clinical reflections, Eur Urol Suppl 8(5):453–457, 2009. presentation and mortality in the US, Cancer 115:68–74, 2009.
Montironi R, Lopez-Beltran A: The 2004 WHO Classification of bladder Semins MJ, Schoenberg MP: A case of florid cystitis glandularis, Nat Clin
tumors: a summary and commentary, Int J Surg Pathol 13:143–153, 2005. Pract Urol 4:341–345, 2007.
Moschini M, D’Andrea D, Korn S, et al: Characteristics and clinical significance Shah RB, Montgomery JS, Montie JE, et al: Variant (divergent) histologic
of histological variants of bladder cancer, Nat Rev Urol 14:651–668, 2017a. differentiation in urothelial carcinoma is under-recognized in community
Moschini M, Dell’Oglio P, Luciano R, et al: Incidence and effect of variant practice: impact of mandatory central pathology review at a large referral
histology on oncological outcomes in patients with bladder cancer treated hospital, Urol Oncol 31:1650–1655, 2013.
with radical cystectomy, Urol Oncol 35:335–341, 2017b. Shanks JH, Iczkowski KA: Divergent differentiation in urothelial carcinoma
Mungan M, Canda A, Tuzel E, et al: Risk Factors for mucosal prostatic urethral and other bladder cancer subtypes with selected mimics, Histopathology
involvement in superficial transitional cell carcinoma of the bladder, Eur 54:885–900, 2009.
Urol 48:760–763, 2005. Sheldon CA, Clayman RV, Gonzalez R, et al: Malignant urachal lesions, J
Naito S, Algaba F, Babjuk M, et al: The Clinical Research Office of the Urol 131:1–8, 1984.
Endourological Society (CROES) multicentre randomised trial of narrow Siefker-Radtke AO, Dinney CP, Abrahams NA, et al: Evidence supporting
band imaging–assisted transurethral resection of bladder tumour (TURBT) preoperative chemotherapy for small cell carcinoma of the bladder: a
versus conventional white light imaging–assisted TURBT in primary retrospective review of the M. D. Anderson cancer experience, J Urol
non–muscle-invasive bladder cancer patients: trial protocol and 1-year 172:481–484, 2004.
results, Eur Urol 70:506–515, 2016. Siefker-Radtke AO, Gee J, Shen YU, et al: Multimodality management of
Network TCGAR: Comprehensive molecular characterization of urothelial urachal carcinoma: the M. D. Anderson Cancer Center Experience, J Urol
bladder carcinoma, Nature 507:315–322, 2014. 169:1295–1298, 2003.
Nieder AM, Porter MP, Soloway MS: Radiation therapy for prostate cancer Siegel RL, Miller KD, Jemal A: Cancer statistics, 2018, CA Cancer J Clin 68:7–30,
increases subsequent risk of bladder and rectal cancer: a population based 2018.
cohort study, J Urol 180:2005–2010, 2008. Sjodahl G, Lauss M, Lövgren K, et al: A molecular taxonomy for urothelial
O’Sullivan P, Sharples K, Dalphin M, et al: A multigene urine test for the carcinoma, Clin Cancer Res 18:3377, 2012.
detection and stratification of bladder cancer in patients presenting with Smeulders N, Woodhouse CR: Neoplasia in adult exstrophy patients, BJU
hematuria, J Urol 188:741–747, 2012. Int 87:623–628, 2001.
Ozbey I, Aksoy Y, Polat Ö, et al: Squamous metaplasia of the bladder: findings Sokolova IA, Halling KC, Jenkins RB, et al: The development of a multitarget,
in 14 patients and review of the literature, Int Urol Nephrol 31:457–461, multicolor fluorescence in situ hybridization assay for the detection of
1999. urothelial carcinoma in urine, J Mol Diagn 2:116–123, 2000.
Paner GP, Stadler WM, Hansel DE, et al: Updates in the eighth edition of Staack A, Schlechte H, Sachs M, et al: Clinical value of vesical leukoplakia
the tumor-node-metastasis staging classification for urologic cancers, Eur and evaluation of the neoplastic risk by mutation analyses of the tumor
Urol 73:560–569, 2018. suppressor gene TP53, Int J Urol 13:1092–1097, 2006.
Planz B, Jochims E, Deix T, et al: The role of urinary cytology for detection Sun J-W, Zhao L-G, Yang Y, et al: Obesity and risk of bladder cancer: a
of bladder cancer, Eur J Surg Oncol 31:304–308, 2005. dose-response meta-analysis of 15 cohort studies, PLoS ONE 2015. (Website):
Platt FM, Hurst CD, Taylor CF, et al: Spectrum of phosphatidylinositol 3-kinase http://dx.plos.org/10.1371/journal.pone.0119313. Koul HK, editor, Public
pathway gene alterations in bladder cancer, Clin Cancer Res 15:6008–6017, Library of Science.
2009. Svatek RS, Hollenbeck BK, Holmäng S, et al: The economics of bladder
Pode D, Shapiro A, Wald M, et al: Noninvasive detection of bladder cancer cancer: costs and considerations of caring for this disease, Eur Urol
with the BTA stat test, J Urol 161:443–446, 1999. 66:253–262, 2014.
Puzio-Kuter AM, Castillo-Martin M, Kinkade CW, et al: Inactivation of p53 Svatek RS, Shariat SF, Novara G, et al: Discrepancy between clinical and
and Pten promotes invasive bladder cancer, Genes Dev 23:675–680, 2009. pathological stage: external validation of the impact on prognosis in an
Raitanen M-P, Aine R, Rintala E, et al: Differences between local and review international radical cystectomy cohort, BJU Int 107:898–904, 2010.
urinary cytology in diagnosis of bladder cancer. an interobserver multicenter Swanson DA, Liles A, Zagars GK: Preoperative irradiation and radical cystectomy
analysis, Eur Urol 41:284–289, 2002. for stages T2 and T3 squamous cell carcinoma of the bladder, J Urol
Rausch S, Lotan Y, Youssef RF: Squamous cell carcinogenesis and squamous 143:37–40, 1990.
cell carcinoma of the urinary bladder: a contemporary review with focus Tan WS, Sarpong R, Khetrapal P, et al: Can renal and bladder ultrasound
on nonbilharzial squamous cell carcinoma, Urol Oncol 32:32.e11–32.e16, replace CT urogram in patients investigated for microscopic hematuria?,
2014. J Urol 2018. http://linkinghub.elsevier.com/retrieve/pii/S0022534718430455.
Ricardo-Gonzalez RR, Nguyen M, Gokden N, et al: Plasmacytoid carcinoma (online journal).
of the bladder: a urothelial carcinoma variant with a predilection for Toma M, Friedrich M, Hautmann S, et al: Comparison of the ImmunoCyt
intraperitoneal spread, J Urol 187:852–855, 2012. test and urinary cytology with other urine tests in the detection and surveil-
Rieder JMD, Parsons JK, Gearhart JP, et al: Primary squamous cell carcinoma lance of bladder cancer, World J Urol 22:145–149, 2004.
in unreconstructed exstrophic bladder, Urology 67:199.e1–199.e2, 2006. Tomlinson DC, L’Hôte CG, Kennedy W, et al: Alternative splicing of fibroblast
Rodriguez Pena MDC, Tregnago AC, Eich M-L, et al: Spectrum of genetic growth factor receptor 3 produces a secreted isoform that inhibits fibroblast
mutations in de novo PUNLMP of the urinary bladder, Virchows Arch growth factor–induced proliferation and is repressed in urothelial carcinoma
471:761–767, 2017. cell lines, Cancer Res 65:10441–10449, 2005.
Ros MM, Bas Bueno-de-Mesquita HB, Büchner FL, et al: Fluid intake and Travis LB, Curtis RE, Glimelius B, et al: Bladder and kidney cancer following
the risk of urothelial cell carcinomas in the European Prospective Investiga- cyclophosphamide therapy for non-Hodgkin’s lymphoma, J Natl Cancer
tion into Cancer and Nutrition (EPIC), Int J Cancer 128:2695–2708, 2011. Inst 87:524–531, 1995.
Rosenthal DL, Wojcik EM, Kurtycz DFI: The Paris System for reporting urinary van der Post RS, Kiemeney LA, Ligtenberg MJL, et al: Risk of urothelial bladder
cytology. Springer Nature, 2017 (website): https://link.springer.com/content/ cancer in Lynch syndrome is increased, in particular among MSH2 mutation
pdf/10.1007/978-3-319-22864-8.pdf. carriers, J Med Genet 47:464–470, 2010.
Ross JS, Wang K, Gay LM, et al: A high frequency of activating extracellular van Oers JMM, Adam C, Denzinger S, et al: Chromosome 9 deletions are
domain ERBB2 (HER2) mutation in micropapillary urothelial carcinoma, more frequent thanFGFR3 mutations in flat urothelial hyperplasias of the
Clin Cancer Res 20:68–75, 2014. bladder, Int J Cancer 119:1212–1215, 2006.

You might also like