ArticlePDF Available

Preclinical analyses of intravesical chemotherapy for prevention of bladder cancer progression

Authors:

Abstract and Figures

There is a critical need to identify treatment options for patients at high risk for developing muscle invasive bladder cancer that avoid surgical removal of the bladder (cystectomy). In the current study, we have performed preclinical studies to investigate the efficacy of intravesical delivery of chemotherapy for preventing progression of bladder cancer. We evaluated three chemotherapy agents, namely cisplatin, gemcitabine, and docetaxel, which are currently in use clinically for systemic treatment of muscle invasive bladder cancer and/or have been evaluated for intravesical therapy. These preclinical studies were done using a geneticallyengineered mouse (GEM) model that progresses from carcinoma in situ (CIS) to invasive, metastatic bladder cancer. We performed intravesical treatment in this GEM model using cisplatin, gemcitabine, and/or docetaxel, alone or by combining two agents, and evaluated whether such treatments inhibited progression to invasive, metastatic bladder cancer. Of the three single agents tested, gemcitabine was most effective for preventing progression to invasive disease, as assessed by several relevant endpoints. However, the combinations of two agents, and particularly those including gemcitabine, were more effective for reducing both tumor and metastatic burden. Our findings suggest combination intravesical chemotherapy may provide a viable bladder-sparing treatment alternative for patients at high risk for developing invasive bladder cancer, which can be evaluated in appropriate clinical trials.
Content may be subject to copyright.
Oncotarget 2013; 4: 269-276269
www.impactjournals.com/oncotarget
www.impactjournals.com/oncotarget/
Oncotarget, February, Vol.4, No 2
Preclinical analyses of intravesical chemotherapy for prevention
of bladder cancer progression
Joan C. Delto
1
, Takashi Kobayashi
1
, Mitchell Benson
1,3
, James McKiernan
1,3
, and
Cory Abate-Shen
1,2,3
1
Department of Urology, Columbia University Medical Center, New York, NY, USA
2
Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
3
Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
Correspondence to: Cory Abate-Shen, email: cabateshen@columbia.edu
Keywords: Non-muscle invasive bladder cancer, intravesical therapy, genetically engineered mouse models, preclinical studies
Received: February 1, 2013 Accepted: February 24, 2013 Published: February 25, 2013
This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use,
distribution, and reproduction in any medium, provided the original author and source are credited.
ABSTRACT:
There is a critical need to identify treatment options for patients at high risk
for developing muscle invasive bladder cancer that avoid surgical removal of the
bladder (cystectomy). In the current study, we have performed preclinical studies
to investigate the ecacy of intravesical delivery of chemotherapy for preventing
progression of bladder cancer. We evaluated three chemotherapy agents, namely
cisplatin, gemcitabine, and docetaxel, which are currently in use clinically for
systemic treatment of muscle invasive bladder cancer and/or have been evaluated
for intravesical therapy. These preclinical studies were done using a genetically-
engineered mouse (GEM) model that progresses from carcinoma in situ (CIS) to
invasive, metastatic bladder cancer. We performed intravesical treatment in this GEM
model using cisplatin, gemcitabine, and/or docetaxel, alone or by combining two
agents, and evaluated whether such treatments inhibited progression to invasive,
metastatic bladder cancer. Of the three single agents tested, gemcitabine was most
eective for preventing progression to invasive disease, as assessed by several
relevant endpoints. However, the combinations of two agents, and particularly those
including gemcitabine, were more eective for reducing both tumor and metastatic
burden. Our ndings suggest combination intravesical chemotherapy may provide a
viable bladder-sparing treatment alternative for patients at high risk for developing
invasive bladder cancer, which can be evaluated in appropriate clinical trials.
INTRODUCTION
Bladder cancer is the fourth most common cancer
in men and the eighth most common overall, with 74,000
new cases diagnosed and an estimated 15,000 deaths in
2012 (1). Notably, distinct subtypes of bladder cancer
have very different patient outcomes (2-5). In particular,
the lethal form is muscle-invasive disease, for which the
precursor is carcinoma in situ (CIS) (6). The primary
treatment for muscle invasive bladder cancer is cystectomy
(surgical removal of the bladder), which is associated with
signicant morbidity; moreover, progression to metastatic
disease has a particularly low 5-year survival (6-9). At the
other end of the spectrum is non-invasive bladder cancer,
which presents as papillary lesions and generally has good
patient prognosis (6, 9). However, non-muscle invasive
bladder cancer can progress to a high-risk disease that
ultimately gives rise to muscle invasive bladder cancer.
Many patients with recurrent non-muscle invasive
bladder cancer are treated with intravesical delivery of
Bacillus Calmette Guerin (BCG), an immunotherapy
regime (10). Although widely used, there can be signicant
adverse reactions to BCG; moreover, 30% of patients
do not respond and even those that respond have a 20%
chance of progression (10, 11). For patients with high-risk
recurrent non-muscle invasive bladder cancer, including
those who have failed BCG therapy, early cystectomy
with urinary diversion is currently the preferred treatment
option (10). However, cystectomy is associated with
signicant morbidity, which severely impacts quality of
Oncotarget 2013; 4: 269-276270
www.impactjournals.com/oncotarget
life, and it may not be a viable option for patients who
are medically unt for surgery. Importantly, patients who
undergo early cystectomy before they progress to invasive
bladder cancer may result in overtreatment.
Thus, there is an urgent need to identify alternative,
bladder-sparing therapies for patients with high-risk
non-muscle invasive bladder cancer. One such option
is intravesical delivery of chemotherapy to prevent
progression to invasive bladder cancer. For example, our
phase I clinical trial using intravesical docetaxel yielded
a 56% complete response rate with 22% durability of
response in three years (12, 13). Other clinical trials
have shown that gemcitabine has promising results for
patients with recurrent non-muscle invasive bladder
cancer in Phase I and Phase II clinical trials (14-16).
Intravesical delivery of selected agents has also been
investigated preclinically in Xenograft models based on
orthotopic implantation of human bladder cancer cells
into immunodecient mouse hosts (17, 18). Although
these clinical and preclinical studies using intravesical
chemotherapy are promising, systemic chemotherapy,
administration of single agents has rarely resulted in
durable long-term remissions. In fact, the most successful
systemic chemotherapy for advanced metastatic bladder
cancer is either a two-drug regimen of gemcitabine
and cisplatin or a four-drug combination regimen of
methotrexate, vinblastine, adriamycin and cisplatin
(MVAC) (8, 9, 19, 20).
We, therefore, reasoned that the design of optimal
intravesical chemotherapy regime(s) for patients with
recurrent non-muscle invasive bladder cancer would
benet from preclinical studies aimed at a direct, side-
by-side comparison of drug regimens involving single
versus double combinations. Toward this end, we have
now performed preclinical studies using a genetically
engineered mouse (GEM) model of progressive
bladder cancer to systematically analyze the efcacy of
chemotherapeutic agents when delivered individually
versus in combination. Our preclinical studies utilize
a GEM model of progressive bladder cancer based on
bladder-specic deletion of two tumor suppressor genes,
p53 and Pten, which are frequently de-regulated in
invasive bladder cancer (21). Following tumor induction
by delivery of Adeno-Cre into the bladder lumen, these
p53
f/f/
; Pten
f/f
mice develop carcinoma in situ (CIS) by 8
weeks, which progresses to invasive bladder cancer with
prevalent metastases by 5 months of age (21, 22). The
bladder tumors from these Adeno-Cre infected p53
f/f/
;
Pten
f/f
mice display similar histologic features as human
muscle invasive bladder cancer, and their metastases arise
in similar tissues, as occurs in humans (21). Our previous
analyses of this GEM model have provided molecular
insights regarding bladder cancer progression and this
model has also provided an effective resource for in vivo
preclinical studies (21, 22). Indeed, we have demonstrated
that bladder tumors arising in these Adeno-Cre infected
p53
f/f/
; Pten
f/f
mice respond to systemic treatment with
rapamycin, while intravesical delivery of rapamycin
prevents progression of CIS to invasive bladder cancer
(21, 22).
In the current study, we have performed preclinical
studies using this GEM model to evaluate the efcacy of
intravesical delivery of chemotherapy for prevention of
progression to invasive bladder cancer. We evaluated three
chemotherapy agents, namely cisplatin, gemcitabine, and
docetaxel, which are standardly administered systemically
for treatment of invasive, metastatic bladder cancer (8,
9, 19, 20). We directly compared these agents to assess
their relative efcacy when delivered individually or in
combination for prevention of progression to invasive
bladder cancer. When tested individually, intravesical
delivery of gemcitabine is most effective among the
three agents for delaying progression to muscle-invasive
bladder cancer; however, each combination of two agents
was more effective for reducing both tumor and metastatic
burden. These ndings suggest that patients at high-risk
for developing invasive bladder cancer may be candidates
for combination intravesical chemotherapy.
Figure 1: Study design for preclinical analyses of intravesical chemotherapy.
Tumor induction was initiated by delivery of
Adeno-Cre directly into the bladder lumen of p53
f/f/
; Pten
f/f
female mice at 8 weeks. Six weeks later (at 14 weeks), mice were imaged
using ultrasound (baseline) and then enrolled into one of 8 treatment arms for the preclinical intravesical treatment: vehicle; gemcitabine,
docetaxel or cisplatin as single agents, or cisplatin + docetaxel, gemcitabine + docetaxel, or gemcitabine + cisplatin in combination. The
treatments were continued for 8 weeks during which time the bladders were imaged every two weeks using ultrasound. At the conclusion
of the treatment period (22 weeks) mice were sacriced for analyses including the endpoints indicated.
Oncotarget 2013; 4: 269-276271
www.impactjournals.com/oncotarget
RESULTS
We performed preclinical studies to evaluate the
consequences of intravesical chemotherapy for delaying
progression from CIS to invasive bladder cancer using
a genetically-engineered mouse model that recapitulates
these progression stages. In particular, we initiated
preclinical treatment in p53
f/f/
; Pten
f/f
mice six weeks after
tumor induction with Adeno-Cre (i.e., at 14 weeks of
age; Fig. 1), since, as we have shown previously, by this
time-point these GEM
mice have developed CIS, but have
not progressed to invasive bladder cancer (22). Prior to
initiation of treatment, we conrmed the absence of overt
tumors using ultrasound imaging. Cohorts of mice were
then randomly assigned to the various treatment arms,
which were the Vehicle group, the single agent group
(cisplatin, gemcitabine, or docetaxel), and the combination
group (cisplatin + gemcitabine; gemcitabine + docetaxel;
and cisplatin + docetaxel) (Fig. 1). In initial pilot studies,
we found that the mice were able to tolerate each of these
agents via intravesical delivery, and were able to tolerate
up to two doses of intravesical treatment weekly (data
not shown). Therefore, we performed these preclinical
studies by combining a maximum of two agents for a
biweekly instillation. Each of the single agent and double
combination treatment groups displayed no signicant
weight loss, based on bi-weekly measurements, nor did
they display other overt signs of distress.
Cohorts of mice were treated one time weekly
(for the signal agents) or bi-weekly (for the double
combinations) for a period of 8 weeks (Fig. 1). By this
point following tumor induction (i.e., at 22 weeks), the
vehicle-treated Adeno-Cre-infected p53
f/f/
; Pten
f/f
mice
characteristically develop invasive bladder tumors that
are large in size, have histological features of invasive
bladder cancer, including a high rate of proliferation, and
are highly metastatic (21). Therefore, our analyses focused
on whether intravesical treatment with the individual or
combination chemotherapy regimes prevented progression
to invasive bladder.
Following treatment initiation, the experimental
mice were monitored with ultrasound every two weeks
for detection of tumors (Fig. 1, Fig. 2). During the
8-week treatment period, the vehicle-treated mice
rapidly developed tumors as detected by ultrasound
imaging, as expected (Fig. 2). In contrast, mice treated
with gemcitabine or docetaxel, but not cisplatin, as
single agents developed tumors at a slower rate (Fig.
2). Moreover, mice that received each of the combined
treatments (cisplatin + docetaxel; gemcitabine + docetaxel;
and cisplatin + docetaxel) also displayed a signicant
delay in the formation of overt tumors (p < 0.05; Fig. 2).
These ndings indicate that intravesical chemotherapy
Figure 2: Onset of tumor formation detected by ultrasound imaging. Graphic representation of the percentage of mice tumor-
free over the course of treatment as detected by ultrasound imaging showing the single (top) and combination (bottom) images. The log rank
p-values are indicated for each treatment (compared to Vehicle). Pictures of the ultrasound images are shown in Figure 4.
Oncotarget 2013; 4: 269-276272
www.impactjournals.com/oncotarget
with gemcitabine or docetaxel alone or with the three
combinations of agents delays overt tumor formation in
tumor-prone mice.
At the end of the 8-week treatment period, the
cohorts of experimental mice were sacriced and their
bladders and other tissues were evaluated. As expected,
all of the vehicle-treated mice displayed large bladder
tumors (~2 grams) that were readily evident upon gross
inspection (Table 1, Fig. 3A, 4; Supplementary Fig. 4).
In contrast, mice in each of the treatment groups treated
with intravesical chemotherapy displayed a reduction
in the size of the bladder, although the extent differed,
particularly for mice treated with the single agents (Table
1, Fig. 3A). In particular, the bladder weights of mice
treated with gemcitabine alone were signicantly reduced
(5.4 fold reduced, p = 0.0271), whereas those treated
with docetaxel or cisplatin were reduced in weight (1.8
and 3.73 fold reduced, respectively) the mean values
were not signicantly different compared to the Vehicle-
treated mice (Table 1, Fig. 3A, 4, Supplementary Fig. 1).
In combination, the reduction in bladder size was further
augmented for gemcitabine in combination with either
docetaxel (7.6 fold reduced, p = 0.0194) or cisplatin (6.0
fold reduced, p = 0.0250), and the combination of cisplatin
+ docetaxel also resulted in a signicant reduction
in bladder weight albeit to a lesser extent than the
combinations with gemcitabine (4.53 fold; p = 0.0.0311;
Table 1, Fig. 3A, 4, Supplementary Fig. 1). Notably, by
gross inspection, bladders treated with the combination
agents, and to a lesser extent gemcitabine alone, were
similar in size and appearance to normal bladder (~0.20-
0.30 grams), consistent with the interpretation that there
was minimal tumor volume (Fig. 4, Supplementary Fig. 1).
Therefore, intravesical treatment with either gemcitabine
alone or with the various combinations of agents resulted
in a signicant reduction of tumor burden at the end of the
treatment period.
We evaluated the histopathology of the bladders
following intravesical chemotherapy by examining
multiple H&E sections throughout the bladder using
whole slide imaging (Table 1, Fig. 4). As evident in
representative high power images (Fig. 4), the vehicle
treated mice display invasive bladder cancer as has
been described previously (21). The experimental mice
treated with the single chemotherapy agents displayed
varying degrees of histopathology that were consistent
with their gross bladder phenotypes. In particular, the
bladder epithelium of most of the mice treated with either
gemcitabine or docetaxel was multilayered with areas
of hyperproliferation, reminiscent of to CIS, but did not
display evidence of invasion, whereas the epithelium of
most of the mice treated with cisplatin alone, which had
evident bladder tumors, displayed histological evidence of
invasion (Table 1, Fig. 4). Moreover, the histology of the
bladders of most of the experimental mice treated with
each of the double combinations had some evidence of
CIS, but were otherwise relatively normal in appearance
with little or no evidence of invasion (Table 1, Fig. 4).
These ndings further support the efcacy of combination
chemotherapy for prevention of bladder cancer
progression.
As an additional parameter of disease progression,
we evaluated the proliferation of the bladder epithelial
and/or tumor cells of the experimental mice following
treatment with intravesical chemotherapy. As we have
shown previously (21), the vehicle treated mice display
a high rate of proliferation (~40%) (Fig. 3B, Table 1).
Consistent with the tumor size and histological phenotype,
Figure 3: Endpoint analyses for preclinical studies. Summary of data from experimental mice following treatment with Vehicle or
with the single or combination agents as indicated. A) Summary of Bladder weights. B) Summary of cellular proliferation as evaluated by
Ki67 immunostaining of the treated bladders.
Oncotarget 2013; 4: 269-276273
www.impactjournals.com/oncotarget
which was signicantly abrogated by the intravesical
chemotherapy, the single agents, and particularly
gemcitabine and docetaxel, displayed a signicant
reduction in proliferation (17-20%; p = 0.012 to 0.04).
Moreover, mice treated with the combination agents
displayed a more profound reduction in proliferation
(10.9% to 13.7%; p = 0.01 to 0.03). These ndings further
underscore the efcacy of combination intravesical
chemotherapy for prevention of bladder cancer
progression.
Finally, we evaluated whether treatment with
intravesical chemotherapy affected the development of
metastases. As we expected based on previous analyses
(21), at the conclusion of the treatment period the majority
of the vehicle-treated mice (15/18) had prominent overt
metastases to several tissues including lymph nodes, liver,
and pancreas (Table 1). The incidence of metastases was
consistently reduced for the experimental mice treated
with the single agents, although not signicantly. However,
mice treated with each of the combination chemotherapy
regimes displayed a statistically signicant reduction in
metastases (2/12 to 2/15, p = 0.003 to 0.004; Table 1).
Taken together, these ndings demonstrate the efcacy
of intravesical delivery of combination chemotherapy for
prevention of progression to invasive, metastatic bladder
cancer.
DISCUSSION
Our study addresses the need to identify alternative,
bladder-sparing treatment options for patients at risk for
developing invasive bladder cancer. Toward this end, we
investigated the efcacy of intravesical chemotherapy in
a preclinical model of the disease. Several key aspects of
our study advance the ndings of previous clinical studies
(12, 14), as well as preclinical analyses using an orthotopic
xenograft model of invasive bladder cancer (18). First, we
have used a GEM model that reliably exhibits progression
from pre-invasive lesions to overt invasive disease, and
therefore our study has enabled us to specically address
the efcacy of these treatments in the context of disease
progression. Additionally, we have performed a direct,
side-by-side comparison of intravesical delivery of three
chemotherapy agents, namely cisplatin, gemcitabine, and
docetaxel. This has enabled us to compare their efcacy
when delivered alone or in combination. Lastly, we
have evaluated multiple endpoints, which proved to be
important when considering potentially clinically-relevant
endpoints, such as the development of the metastases, in
which the combination treatments outperformed any of the
single agents.
The major conclusion of the current study is that
while the single agents, and particularly gemcitabine,
may be somewhat effective for reducing tumor burden,
Table 1: Summary of treatment endpoints
Bladder Weight Phenotype Proliferation Metastases
Treatment N
Mean
weight
(gr)
SEM
Fold
Change
P value Description
% Ki67
P value
Cases
w/ Mets
P value
Vehicle
PBS
(DMSO in PBS)
22
2.13
(N = 10)
0.64 - -
Large tumors
(18/22); Small
tumors (4/22)
39.1
(N = 4)
-
15/18
(85%)
-
Single Agents
Cisplatin 15
1.15
(N = 7)
0.30 1.85 NS
Small tumors
(12/15); CIS
(3/15)
20.4
(N = 4)
0.038
6/14
(42%)
NS
Gemcitabine 14
0.39
(N = 5)
0.20 5.46 0.0271
Small tumors
(3/14); CIS
(11/14)
17.5
(N = 4)
0.019
4/10
(40%)
NS
Docetaxel 14
0.57
(N = 5)
0.48 3.73 NS
Small tumors
(5/14); CIS
(11/14)
19.2
(N = 4)
0.013
4/10
(40%)
NS
Combinations
Cis + Doce 22
0.47
(N = 13)
0.16 4.53 0.0311
Small tumors
(3/22);
Normal/CIS
(19/22)
10.9
(N = 4)
0.032
2/12
(16%)
0.004
Gem + Doce 23
0.28
(N = 13)
0.18 7.60 0.0194
Small tumors
(2/23);
Normal/CIS
(21/23)
13.7
(N = 4)
0.017
1/13
(8%)
0.001
Cis + Gem 26
0.36
(N = 14)
0.14 6.00 0.0250
Small tumors
(4/26);
Normal/CIS
(22/26)
12.6
(N = 4)
0.011
2/15
(13%)
0.003
Oncotarget 2013; 4: 269-276274
www.impactjournals.com/oncotarget
when considering all of the endpoints examined, the most
effective regime for preventing progression to invasive
bladder cancer is a combination agents, particularly
those including gemcitabine. The further implication
of our ndings is that while intravesical treatment
with gemcitabine, or with the other single agents, may
have an immediate benet for delaying progression,
sustained effects that may impact overall survival, such
as metastases, may require combination treatments. The
additional implication of our study is that more than
one combination is likely to be effective for preventing
progression, which may leave open several options for
patients who do not respond or tolerate one treatment
regime but may benet from an alternative.
We propose that our ndings provide the rationale
for evaluation of combination intravesical chemotherapy
for patients with non-muscle invasive bladder cancer who
are at high risk for progressing to invasive disease. Such
treatment would be particularly effective for management
of patients who are unable to undergo cystectomy, but
may be extended to others who are also at high risk of
progressing to muscle invasion and wish to avoid early
removal of the bladder. Thus, combination intravesical
chemotherapy may provide an alternative to cystectomy
that improves the quality of life.
MATERIALS AND METHODS
Mouse model of progressive bladder cancer
All animal experiments were performed according
to protocols approved by the Institutional Animal Care
and Use Committee at Columbia University Medical
Center. The genetically engineered mouse (GEM)
model of progressive bladder cancer used in this study
was developed and characterized in our laboratory and
has been described previously (21). Briey, these mice
are based on bladder-specic deletion of oxed alleles
of p53 and Pten (i.e., p53
ox/ox
; Pten
ox/ox
) in a C57/Bl6
strain background. Tumors are induced by delivery of
an Adeno-virus expressing Cre recombinase (hereafter
referred to as Adeno-Cre) directly into the bladder lumen
at 8 weeks of age (21). Ultrasound imaging using a
Vevo 2100® Imaging System (Visual Sonics, Toronto,
Ontario, Canada) was performed to detect bladder tumors,
following the instructions of the manufacturer. Following
tumor induction, mice were monitored on a daily basis
for body condition (i.e., muscle tone and weight) and
sacriced when their body condition score was <1.5, as
per guidelines of the Institutional Animal Care and Use
Committee.
Intravesical drug treatment
For these studies we used female Adeno-Cre-
injected p53
ox/ox
; Pten
ox/ox
mice because they are
amenable to intravesical treatment (22). Under anesthesia,
mice were placed in supine position and the external
urinary orice cleansed with betadine. A 24G Jelco
angiocatheter (~10-15 mm in a typical 20 gram female
mouse) was inserted through the urethra to the bladder
lumen. Irrigation with sterile PBS was performed to ensure
proper placement of the catheter tip, and the remaining
urine was aspirated with a 1 cc syringe.
Chemotherapy agents (50 µl) were delivered into
Figure 4: Phenotype of mice treated following intravesical treatment. Shown are representative images of bladders from Adeno-
Cre infected p53
f/f/
; Pten
f/f
mice following treatment with Vehicle or with the single or combination agents as indicated. The top panels show
whole mount images of the dissected bladder at the time of dissection. The upper middle panels show representative ultrasound images and
representative H&E images of bladder histology at the conclusion of the treatment. The bottom panels show images of representative of
Ki67-immunostaining for quantication of proliferation.
Oncotarget 2013; 4: 269-276275
www.impactjournals.com/oncotarget
the bladder lumen and a 5-0 silk suture was tied around
the urethral meatus to prevent expulsion; the installation
time was 2 hours. Agents used were as follows: Cis-
Diamineplatinum (II) Dichloride (Cisplatin; #479306)
was purchased from Sigma; Gemcitabine (#NC0063515)
and Docetaxel (#D-1000) were purchased from LC Labs.
Optimal dosages for each agent were estimated based
on prior literature (18, 23-25) and then conrmed in
pilot studies. Cisplatin and Gemcitabine were dissolved
in sterile PBS and diluted to working concentrations of
0.5 mg/ml and 25 mg/ml, respectively. Docetaxel was
reconstituted to a concentration of 12.5 mg/ml in DMSO
and diluted in sterile PBS to 0.5mg/ml. Each drug was
delivered 1 time per week using the following dosage
schedule: Vehicle (PBS) was delivered on Monday and/
or Wednesdays. (Note that the Vehicle group included
mice in which DMSO was diluted into PBS; these were
not appreciably different than the PBS group). Cisplatin
was delivered on Monday; Docetaxel was delivered on
Wednesdays; Gemcitabine was delivered on Friday. When
two agents were delivered, these were given on two days,
rather the same day. To control for the enhanced efcacy
of the combination versus the second treatment, a cohort
of the single agent mice were delivered drug at twice the
dose, which was not appreciably different than the single
dose.
Cohorts of mice were enrolled randomly into
the various treatment arms. The size of the cohorts
was determined using standard power analyses, with
bootstrapping from pilot studies. In particular, based on
the phenotype and response to drug treatment from pilot
studies, we estimated that a minimum of 6 mice would
provide statistical power for analyses; however, each of
experimental groups had a minimum of 10 mice in each
condition. Attrition, due either to death from tumor size or
infection from the catheterization, or other was less than
10% overall; the mice reported on only included those that
survived to the end of the study.
Analyses of mouse phenotypes
Following eight cycles (8 weeks) of drug treatment,
mice were sacriced and autopsied to evaluate the overall
bladder phenotype and to quantify metastatic lesions.
Bladders were harvested and processed for histological
analysis as described (21). Metastases to distant organs
were scored by visual inspection upon sacrice, and
conrmed by histological analyses; a mouse was indicated
to be positive for metastases if we observed a minimum
of two overt lesions. Metastases were observed mainly in
the lymph nodes, pancreas, liver, and GI tract, as we have
reported previously (21). Indications for sacrice prior to
eight cycles of treatment included tumor size of 1.5 cm or
greater, hematuria, or weight loss of greater than 15% of
initial body weight.
Immunohistochemical staining was performed
on parafn-embedded tissues as described (26).
Quantication of cellular proliferation was performed by
immunostaining with Ki67 (Lieca # NCL-Ki67p) using at
least three independent sections on 4 independent mice/
group (26). As we have reported previously, the percentage
of Ki67-positively stained cells in the bladder epithelium
or tumors were in comparison to the unstained cells
(26). Images were captured using a whole slide scanner
(Olympus VS120-S5).
Statistical Analysis
Statistical analysis were performed using Welch
t-test and Fishers Exact test as appropriate. GraphPad
Prism software (Version 4.0) was used for statistical
analysis and to generate data plots.
ACKNOWLEDGEMENTS
Ultrasound imaging was performed in the Herbert
Irving Comprehensive Cancer Center Small Animal
Imaging Shared Resource. This work was supported by
grants CA084294 (to CAS), funding from the Alexander
and Margaret Stewart Trust provided to the Institute for
Cancer Genetics, and from the T.J. Martell Foundation
for Leukemia, Cancer and AIDS Research (to MB). TK
was supported by post-doctoral training grants from the
American Urological Association Foundation and the
American Association for Cancer Research. CAS is an
American Cancer Society Research Professor supported
in part by a generous gift from the F.M. Kirby Foundation.
REFERENCES
1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012.
CA Cancer J Clin. 2012;62:10-29.
2. Goebell PJ, Knowles MA. Bladder cancer or bladder
cancers? Genetically distinct malignant conditions of the
urothelium. Urologic oncology. 2010;28:409-28.
3. McConkey DJ, Lee S, Choi W, Tran M, Majewski T, Lee
S, et al. Molecular genetics of bladder cancer: Emerging
mechanisms of tumor initiation and progression. Urologic
oncology. 2010;28:429-40.
4. Dinney CP, McConkey DJ, Millikan RE, Wu X, Bar-Eli
M, Adam L, et al. Focus on bladder cancer. Cancer cell.
2004;6:111-6.
5. Castillo-Martin M, Domingo-Domenech J, Karni-Schmidt
O, Matos T, Cordon-Cardo C. Molecular pathways of
urothelial development and bladder tumorigenesis. Urologic
oncology. 2010;28:401-8.
6. Kaufman DS, Shipley WU, Feldman AS. Bladder cancer.
Lancet. 2009;374:239-49.
7. Grossman HB, Natale RB, Tangen CM, Speights VO,
Vogelzang NJ, Trump DL, et al. Neoadjuvant chemotherapy
Oncotarget 2013; 4: 269-276276
www.impactjournals.com/oncotarget
plus cystectomy compared with cystectomy alone for
locally advanced bladder cancer. The New England journal
of medicine. 2003;349:859-66.
8. Sternberg CN. Muscle invasive and metastatic bladder
cancer. Annals of oncology : ofcial journal of the
European Society for Medical Oncology / ESMO. 2006;17
Suppl 10:x23-30.
9. Parekh DJ, Bochner BH, Dalbagni G. Supercial and
muscle-invasive bladder cancer: principles of management
for outcomes assessments. Journal of clinical oncology
: ofcial journal of the American Society of Clinical
Oncology. 2006;24:5519-27.
10. Hall MC, Chang SS, Dalbagni G, Pruthi RS, Seigne JD,
Skinner EC, et al. Guideline for the management of
nonmuscle invasive bladder cancer (stages Ta, T1, and Tis):
2007 update. The Journal of urology. 2007;178:2314-30.
11. Zlotta AR, Fleshner NE, Jewett MA. The management of
BCG failure in non-muscle-invasive bladder cancer: an
update. Canadian Urological Association journal = Journal
de l’Association des urologues du Canada. 2009;3:S199-
205.
12. McKiernan JM, Masson P, Murphy AM, Goetzl M,
Olsson CA, Petrylak DP, et al. Phase I trial of intravesical
docetaxel in the management of supercial bladder cancer
refractory to standard intravesical therapy. Journal of
clinical oncology : ofcial journal of the American Society
of Clinical Oncology. 2006;24:3075-80.
13. Laudano MA, Barlow LJ, Murphy AM, Petrylak DP, Desai
M, Benson MC, et al. Long-term clinical outcomes of a
phase I trial of intravesical docetaxel in the management of
non-muscle-invasive bladder cancer refractory to standard
intravesical therapy. Urology. 2010;75:134-7.
14. Dalbagni G, Russo P, Bochner B, Ben-Porat L, Sheinfeld
J, Sogani P, et al. Phase II trial of intravesical gemcitabine
in bacille Calmette-Guerin-refractory transitional cell
carcinoma of the bladder. Journal of clinical oncology
: ofcial journal of the American Society of Clinical
Oncology. 2006;24:2729-34.
15. Laufer M, Ramalingam S, Schoenberg MP, Haiseld-
Wolf ME, Zuhowski EG, Trueheart IN, et al. Intravesical
gemcitabine therapy for supercial transitional cell
carcinoma of the bladder: a phase I and pharmacokinetic
study. Journal of clinical oncology : ofcial journal of the
American Society of Clinical Oncology. 2003;21:697-703.
16. Witjes JA, van der Heijden AG, Vriesema JL, Peters
GJ, Laan A, Schalken JA. Intravesical gemcitabine: a
phase 1 and pharmacokinetic study. European urology.
2004;45:182-6.
17. Hadaschik BA, Black PC, Sea JC, Metwalli AR, Fazli L,
Dinney CP, et al. A validated mouse model for orthotopic
bladder cancer using transurethral tumour inoculation
and bioluminescence imaging. BJU international.
2007;100:1377-84.
18. Hadaschik BA, ter Borg MG, Jackson J, Sowery RD, So
AI, Burt HM, et al. Paclitaxel and cisplatin as intravesical
agents against non-muscle-invasive bladder cancer. BJU
international. 2008;101:1347-55.
19. Dumez H, Martens M, Selleslach J, Guetens G, De Boeck
G, Aerts R, et al. Docetaxel and gemcitabine combination
therapy in advanced transitional cell carcinoma of the
urothelium: results of a phase II and pharmacologic study.
Anti-cancer drugs. 2007;18:211-8.
20. Sternberg CN, Yagoda A, Scher HI, Watson RC, Geller
N, Herr HW, et al. Methotrexate, vinblastine, doxorubicin,
and cisplatin for advanced transitional cell carcinoma of the
urothelium. Efcacy and patterns of response and relapse.
Cancer. 1989;64:2448-58.
21. Puzio-Kuter AM, Castillo-Martin M, Kinkade CW, Wang
X, Shen TH, Matos T, et al. Inactivation of p53 and Pten
promotes invasive bladder cancer. Genes & development.
2009;23:675-80.
22. Seager CM, Puzio-Kuter AM, Patel T, Jain S, Cordon-
Cardo C, Mc Kiernan J, et al. Intravesical delivery of
rapamycin suppresses tumorigenesis in a mouse model
of progressive bladder cancer. Cancer Prev Res (Phila).
2009;2:1008-14.
23. Brocks CP, Buttner H, Bohle A. Inhibition of tumor
implantation by intravesical gemcitabine in a murine model
of supercial bladder cancer. The Journal of urology.
2005;174:1115-8.
24. Mugabe C, Matsui Y, So AI, Gleave ME, Heller M, Zeisser-
Labouebe M, et al. In vitro and in vivo evaluation of
intravesical docetaxel loaded hydrophobically derivatized
hyperbranched polyglycerols in an orthotopic model of
bladder cancer. Biomacromolecules. 2011;12:949-60.
25. Nativ O, Dalal E, Laufer M, Sabo E, Aronson M.
Antineoplastic effect of gemcitabine in an animal model of
supercial bladder cancer. Urology. 2004;64:845-8.
26. Kinkade CW, Castillo-Martin M, Puzio-Kuter A, Yan J,
Foster TH, Gao H, et al. Targeting AKT/mTOR and ERK
MAPK signaling inhibits hormone-refractory prostate
cancer in a preclinical mouse model. The Journal of clinical
investigation. 2008;118:3051-64.

Supplementary resource (1)

... These studies further identified therapeutic targets downstream of the aforementioned mutations. Yang and colleagues [62] identified that the simultaneous inactivation of p53 and activation of Kras induced the quick formation of spindle-cell sarcoma in the soft tissues adjacent to the bladder but the slow formation of urothelial hyperplasia inside the bladder. These results strongly show that the effect of oncogene regulation to produce either hyperplasia or carcinogenesis greatly depends on the type of tissue. ...
... These results strongly show that the effect of oncogene regulation to produce either hyperplasia or carcinogenesis greatly depends on the type of tissue. Most importantly, studies with GEM models that recapitulate disease progression allowed the preclinical validation of the key pathways involved in urothelial cancer progression from non muscle invasive (NMI) to muscle invasive (MI) disease [59,60,62,63] and paved the way to clinical trials for intra-vesical delivery of single or combinatorial chemotherapeutics to prevent disease progression in high-risk patients with early-stage disease [64]. ...
... These models are easy to manage, easy to establish, cost-effective, and are widely used in mechanistic studies as well as in evaluating the efficacy of novel therapeutic agents [82]. Several transplantable models are used in complementary ways to study different aspects of tumor growth and metastasis, as well as the effect of tumor-stromal interactions [62]. ...
Article
Full-text available
Background: Bladder cancer (urothelial cancer of the bladder) is the most common malignancy affecting the urinary system with an increasing incidence and mortality. Mouse models of bladder cancer should possess a high value of reproducibility, predictability, and translatability to allow mechanistic, chemo-preventive, and therapeutic studies that can be furthered into human clinical trials. Objectives: To provide an overview and resources on the origin, molecular and pathological characteristics of commonly used animal models in bladder cancer. Methods: A PubMed and Web of Science search was performed for relevant articles published between 1980 and 2021 using words such as: "bladder" and/or "urothelial carcinoma" and animal models. Animal models of bladder cancer can be categorized as autochthonous (spontaneous) and non-autochthonous (transplantable). The first are either chemically induced models or genetically engineered models. The transplantable models can be further subclassified as syngeneic (murine bladder cancer cells implanted into immunocompetent or transgenic mice) and xenografts (human bladder cancer cells implanted into immune-deficient mice). These models can be further divided-based on the site of the tumor-as orthotopic (tumor growth occurs within the bladder) and heterotopic (tumor growth occurs outside of the bladder).
... 27 The sequential multi-agent chemotherapy regimen in this phase I clinical trial was based on our preclinical studies of intravesical treatments in a genetically engineered mouse model of bladder cancer. 14,30 Our preclinical study of single and combination regimens of intravesical cisplatin, gemcitabine and docetaxel showed that mice receiving any 2-drug combination had a statistically significant delay in overt tumor formation, as well as reduced development of overt metastases compared to the single agent treatments. 14 The participants in this trial represent a particularly high risk group. ...
... 14,30 Our preclinical study of single and combination regimens of intravesical cisplatin, gemcitabine and docetaxel showed that mice receiving any 2-drug combination had a statistically significant delay in overt tumor formation, as well as reduced development of overt metastases compared to the single agent treatments. 14 The participants in this trial represent a particularly high risk group. All were categorized as BCG unresponsive except for 1. ...
Article
Purpose: For patients with BCG unresponsive or recurrent/relapsing non-muscle invasive bladder cancer (NMIBC), multi-agent intravesical trials have been limited. The goal of this study was to investigate the safety of intravesical cabazitaxel, gemcitabine, and cisplatin (CGC) in the salvage setting. Materials and methods: This was a dose-escalation, drug-escalation trial for patients with BCG unresponsive or recurrent/relapsing NMIBC who declined or were ineligible for radical cystectomy. All patients underwent a 6-week induction regimen of sequentially administered cabazitaxel, gemcitabine and cisplatin. Complete response was defined as no cancer on post-induction TURBT and negative urine cytology, while partial response allowed for positive cytology. Responders continued with maintenance cabazitaxel and gemcitabine monthly for the first year and bimonthly for the second year. Results: Eighteen patients were enrolled. Mean age was 71 years, median follow-up was 27.8 months (16.3 - 46.9), and mean number of previous rounds of intravesical therapies prior to trial enrollment was 3.7. Nine patients (50%) had received intravesical chemotherapy after BCG, and seven (39%) were previously treated in a Phase I clinical trial setting. At enrollment, 6/18 (33%) subjects had T1 disease, and 13/18 (72%) had CIS. There were no dose-limiting toxicities. Initial partial and complete response rates were 94% and 89%, respectively. At one year, recurrence-free survival (RFS) was 0.83 (0.57 - 0.94), and at two years estimated RFS was 0.64 (0.32, 0.84). Conclusions: In this high risk and highly pre-treated cohort of BCG unresponsive or recurrent/relapsing NMIBC subjects, combination intravesical cabazitaxel, gemcitabine, and cisplatin was a well-tolerated and potentially effective regimen.
... 35 Other combination chemotherapies in NMIBC has proven superior efficacy than single agent, particularly in the BCG-failure category. 36 However, chemotherapy such as mitomycin C in association with BCG instillation was not superior to BCG monotherapy in the treatment of CIS. 37 Also, combination of gemcitabine and mitomycin C was tested and has demonstrated a RFS rates of 38% at 2 years post treatment. ...
Article
Introduction: Bacillus Calmette-Guérin (BCG) failure occurs in approximately 40% of patients with non-muscle-invasive bladder cancer (NMIBC) within two years. We describe our institutional experience with sequential intravesical gemcitabine and docetaxel (gem/doce) as salvage therapy post-BCG failure in patients who were not candidates for or declined radical cystectomy (RC). Methods: We retrospectively reviewed BCG-failure patients with NMIBC who received gem/doce from April 2019 through October 2022 at the CHU de Québec–Université Laval. Patients received at least five weekly intravesical instillations according to published protocols. Patients who responded to gem/doce had maintenance instillations monthly for up to two years. Primary outcome was progression-free survival (PFS). Secondary outcomes included recurrence-free survival (RFS), cystectomy-free survival (CFS), cancer-specific survival (CSS), overall survival (OS), and treatment adverse events. Survival probabilities were estimated using the Kaplan-Meier method from the first gem/doce instillation. Results: Thirty-five patients with a median age of 78 years old were included in the study. The median followup time was 21 months (interquartile range 10–29). More than 25% of patients received two or more prior BCG induction treatments. Overall/MIBC PFS estimates at one year were 85%/88% and 60%/70% at two years. Adverse events occurred in 37% of the patients, but only two patients didn’t complete the treatment due to intolerance. Three patients underwent radical cystectomy due to cancer progression. OS was 94% at two years. Conclusions: With 60% of PFS at two years, gem/doce appears to be a safe and well-tolerated option for BCG failure patients. Further studies are needed to justify widespread use.
... In clinical studies, doxorubicin (Dox), cisplatin (CDDP) and cisplatin analogs are widely used highly water-soluble anti-cancer drugs [57]. Considering intravesical chemotherapy of NMIBCs, the CDDP-based treatment has proven to be less effective than other drugs [58]. Possibly, the small, neutral CDDP molecule is poorly adsorbed and has low penetration into the bladder urothelium as a result of its poor adsorptive properties. ...
Article
Full-text available
Bladder cancer (BC) is the most frequently occurring cancer of the urinary system, with non-muscle-invasive bladder cancer (NMIBC) accounting for 75–85% of all the bladder cancers. Patients with NMIBC have a good survival rate but are at high risk for tumor recurrence and disease progression. Intravesical instillation of antitumor agents is the standard treatment for NMIBC following transurethral resection of bladder tumors. Chemotherapeutic drugs are broadly employed for bladder cancer treatment, but have limited efficacy due to chemo-resistance and systemic toxicity. Additionally, the periodic voiding of bladder and low permeability of the bladder urothelium impair the retention of drugs, resulting in a weak antitumoral response. Chitosan is a non-toxic and biocompatible polymer which enables better penetration of specific drugs to the deeper cell layers of the bladder as a consequence of temporarily abolishing the barrier function of urothelium, thus offering multifaceted biomedical applications in urinary bladder epithelial. Nowadays, the rapid development of nanoparticles significantly improves the tumor therapy with enhanced drug transport. This review presents an overview on the state of chitosan-based nanoparticles in the field of intravesical bladder cancer treatment.
... However, given its role as the mainstay of treatment for metastatic urothelial carcinoma, interest has recently rebounded in the form of pre-clinical and clinical trials [32,40,41]. Investigators are now using cisplatin as part of sequential multidrug intravesical regimens with MMC and doxorubicin or gemcitabine and cabazitaxel with promising initial results [32,[42][43][44][45]. As such, we report its solubility profiles under various physical states to aid in dose determination of future studies. ...
Article
Full-text available
BACKGROUND: Nearly 70% of all new cases of bladder cancer are non-muscle invasive disease, the treatment for which includes transurethral resection followed by intravesical = therapy. Unfortunately, recurrence rates approach 50%, in part due to poor intravesical drug delivery. Hyperthermia is frequently used as an adjunct to intravesical chemotherapy to improve drug delivery and response to treatment. OBJECTIVE: To assess the solubility profile of intravesical chemotherapies under varying conditions of pHand temperature. METHODS: Using microplate laser nephelometry we measured the solubility of three intravesical chemotherapy agents (mitomycin C, gemcitabine, and cisplatin) at varying physical conditions. Drugs were assessed at room temperature (23◦C), body temperature (37◦C), and 43◦C, the temperature used for hyperthermic intravesical treatments. To account for variations in urine pH, solubility was also investigated at pH 4.00, 6.00, and 8.00. RESULTS: Heat incrementally increased the solubility of all three drugs studied. Conversely, pH largely did not impact solubility aside for gemcitabine which showed slightly reduced solubility at pH 8.00 versus 6.00 or 4.00. Mitomycin C at the commonly used 2.0 mg/mL was insoluble at room temperature, but soluble at both 37 and 43◦C. CONCLUSIONS: Hyperthermia as an adjunct to intravesical treatment would improve drug solubility, and likely drug delivery as some current regimens are insoluble without heat. Improvements in solubility also allow for testing of alternative administration regimens to improve drug delivery or tolerability. Further studies are needed to confirm that improvements in solubility result in increased drug delivery.
... In one study, they compared the results of instilling intravesical cisplatin, gemcitabine, and/or docetaxel alone or by combining two agents. 35 They found that gemcitabine (pyrimidine analog that inhibits cell growth and induces apoptosis) was the single most effective agent at preventing progression to invasive disease. However, a combination of two agents, especially those that included gemcitabine, was significantly more effective at reducing tumor burden. ...
Article
Non-muscle invasive bladder cancer (NMIBC) is a common and burdensome malignancy. A substantial proportion of patients with intermediate- and high-risk disease will progress to invasive bladder cancer and are at a significant risk for metastasis and death. Bacillus Calmette-Guerin (BCG) therapy for selected cases has been the standard of care for nearly 40 years. Unfortunately, a world-wide shortage has made BCG challenging to obtain. Furthermore, recurrences and progressions do occur. With the US Food and Drug Administration creating a clear path to drug approval for novel treatments, many therapies have been tested, including intravesical cytotoxic chemotherapy, intravesical immunotherapy, systemic immunotherapy, and novel agents, such as gene therapy and targeted therapy. In this review, we highlight ongoing clinical trials.
Article
Unraveling the multifaceted cellular and physiological processes associated with metastasis is best achieved by using in vivo models that recapitulate the requisite tumor cell-intrinsic and -extrinsic mechanisms at the organismal level. We discuss the current status of mouse models of metastasis. We consider how mouse models can refine our understanding of the underlying biological and molecular processes that promote metastasis, and we envisage how the application of new technologies will further enhance investigations of metastasis at single-cell resolution in the context of the whole organism. Our view is that investigations based on state-of-the-art mouse models can propel a holistic understanding of the biology of metastasis, which will ultimately lead to the discovery of new therapeutic opportunities.
Article
To study the progression of bladder cancer from non–muscle-invasive to muscle-invasive disease, we have developed a novel toolkit that uses complementary approaches to achieve gene recombination in specific cell populations in the bladder urothelium in vivo, thereby allowing us to generate a new series of genetically engineered mouse models (GEMM) of bladder cancer. One method is based on the delivery of adenoviruses that express Cre recombinase in selected cell types in the urothelium, and a second uses transgenic drivers in which activation of inducible Cre alleles can be limited to the bladder urothelium by intravesicular delivery of tamoxifen. Using both approaches, targeted deletion of the Pten and p53 tumor suppressor genes specifically in basal urothelial cells gave rise to muscle-invasive bladder tumors. Furthermore, preinvasive lesions arising in basal cells displayed upregulation of molecular pathways related to bladder tumorigenesis, including proinflammatory pathways. Cross-species analyses comparing a mouse gene signature of early bladder cancer with a human signature of bladder cancer progression identified a conserved 28-gene signature of early bladder cancer that is associated with poor prognosis for human bladder cancer and that outperforms comparable gene signatures. These findings demonstrate the relevance of these GEMMs for studying the biology of human bladder cancer and introduce a prognostic gene signature that may help to stratify patients at risk for progression to potentially lethal muscle-invasive disease. Significance Analyses of bladder cancer progression in a new series of genetically engineered mouse models has identified a gene signature of poor prognosis in human bladder cancer.
Article
Non-muscle-invasive bladder cancer (NMIBC) remains one of the most common malignancies and is associated with considerable treatment costs. Patients with intermediate-risk or high-risk disease can be treated with intravesical BCG, but many of these patients will experience tumour recurrence, despite adequate treatment. Standard of care in these patients is radical cystectomy with urinary diversion, but this approach is associated with considerable morbidity and lifestyle modification. As an alternative, perioperative intravesical chemotherapy is recommended for low-risk papillary NMIBC, and induction intravesical chemotherapy is an option for patients with intermediate-risk NMIBC and BCG-unresponsive NMIBC. However, poor pharmaceutical absorption and drug washout during normal voiding can limit sustained drug concentrations in the urothelium, which reduces efficacy, and small-molecule chemotherapeutic agents can be absorbed through the urothelium into the bloodstream, leading to systemic adverse effects. Several novel drug delivery methods - including hyperthermia, mechanical sustained released devices and nanoparticle drug conjugation - have been developed to overcome these limitations. These novel methods have the potential to be combined with established chemotherapeutic agents to change the paradigm of NMIBC treatment.
Article
Full-text available
Urothelial cancer has served as one of the most important sources of information about the mutational events that underlie the development of human solid malignancies. Although "field effects" that affect the entire bladder mucosa appear to initiate disease, tumors develop along 2 distinct biological "tracks" that present vastly different challenges for clinical management. Recent whole genome methodologies have facilitated even more rapid progress in the identification of the molecular mechanisms involved in bladder cancer initiation and progression. Specifically, whole organ mapping combined with high resolution, high throughput SNP analyses have identified a novel class of candidate tumor suppressors ("forerunner genes") that localize near more familiar tumor suppressors but are disrupted at an earlier stage of cancer development. Furthermore, whole genome comparative genomic hybridization (CGH) and mRNA expression profiling have demonstrated that the 2 major subtypes of urothelial cancer (papillary/superficial and non-papillary/muscle-invasive) are truly distinct molecular entities, and in recent work our group has discovered that muscle-invasive tumors express molecular markers characteristic of a developmental process known as "epithelial-to-mesenchymal transition" (EMT). Emerging evidence indicates that urothelial cancers contain subpopulations of tumor-initiating cells ("cancer stem cells") but the phenotypes of these cells in different tumors are heterogeneous, raising questions about whether or not the 2 major subtypes of cancer share a common precursor. This review will provide an overview of these new insights and discuss priorities for future investigation.
Article
Full-text available
Up to 40% of patients with non-muscle-invasive bladder cancer (NMIBC) will fail intravesical bacillus Calmette-Guérin (BCG) therapy. There is unfortunately no current gold standard for salvage intravesical therapy after appropriate BCG treatment. Indeed, outcomes are at best suboptimal. The vast majority of low-grade NMIBC are prone to recur but very rarely progress. Failure after intravesical BCG in these patients is usually superficial and low-grade. At the other end of the spectrum, failure to respond to BCG in high-risk T1 bladder cancer and/or carcinoma in situ (CIS or TIS) is more problematic, since those tumours often have the potential to progress to muscle invasion. In these cases, radical cystectomy remains the mainstay after BCG failure. With appropriate selection, certain patients who "fail" BCG (but with favourable risk factors) can be managed with intravesical regimens, including repeated BCG, BCG plus cytokines, intravesical chemotherapy, thermochemotherapy or new immunotherapeutic modalities. In this review, reasons explaining BCG failure, how to define BCG failure, optimal risk stratification and prediction of response and management of BCG failures are discussed.
Article
Full-text available
Early-stage bladder cancer occurs as two distinct forms: namely, low-grade superficial disease and high-grade carcinoma in situ (CIS), which is the major precursor of muscle-invasive bladder cancer. Although the low-grade form is readily treatable, few, if any, effective treatments are currently available for preventing progression of nonmuscle-invasive CIS to invasive bladder cancer. Based on our previous findings that the mammalian target of Rapamycin (mTOR) signaling pathway is activated in muscle-invasive bladder cancer, but not superficial disease, we reasoned that suppression of this pathway might block cancer progression. To test this idea, we performed in vivo preclinical studies using a genetically engineered mouse model that we now show recapitulates progression from nonmuscle-invasive CIS to muscle-invasive bladder tumors. We find that delivery of Rapamycin, an mTOR inhibitor, subsequent to the occurrence of CIS effectively prevents progression to invasive bladder cancer. Furthermore, we show that intravesical delivery of Rapamycin directly into the bladder lumen is highly effective for suppressing bladder tumorigenesis. Thus, our findings show the potential therapeutic benefit of inhibiting mTOR signaling for treatment of patients at high risk of developing invasive bladder cancer. More broadly, our findings support a more widespread use of intravesical delivery of therapeutic agents for treatment of high-risk bladder cancer patients, and provide a mouse model for effective preclinical testing of potential novel agents.
Article
Of 133 patients with advanced urothelial tract cancer given methotrexate (MTX), vinblastine (VBL), Adriamycin (ADR) (doxorubicin; Adria Laboratories, Columbus, OH), and cisplatin (DDP) (M-VAC regimen), significant tumor regression occurred in 72% ± 8% of 121 with transitional cell carcinoma (TCC) evaluable for response. Complete remission (CR) was achieved in 36% ± 9% of patients, of whom 11% required the addition of surgical resection of residual disease. Although 68% of CR patients have relapsed, CR median survival will exceed 38 months compared with 11 months for partial (36%) and minor (6%) responders, and 8 months for nonresponders: 2-year and 3-year survivals were 68% and 55%, respectively, versus 0% to 7% for the remaining patients. Sixteen percent of responders developed brain lesions, half of whom had no systemic relapse at the time of progression. Three patients with non-TCC histologies did not respond. In 32 patients who had pathologic restaging, the clinical (T) understaging (T < pathologic [P] restaging) error was 35%. Although all metastatic sites showed evidence of tumor regression, CR was noted more frequently in lung, in intraabdominal lymph nodes and masses, and in bone (24% to 35%); the rate for hepatic lesions was 15%. There were 52% of 21 N3–4Mo patients who achieved CR versus 33% of 100 with No-+M+ lesions. Toxicity was significant with 4 (3%) drug-related deaths, 25% incidence of nadir sepsis, 58% ⩾ 3+ myelosuppression, and 49% with mucositis. Responsiveness of metastasis in various sites, patterns of relapse, and the usefulness of the new CR response criteria are reported, as is the current status of cisplatin and methotrexate combination regimens. Cancer 64:2448–2458, 1989.
Article
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths expected in the United States in the current year and compiles the most recent data on cancer incidence, mortality, and survival based on incidence data from the National Cancer Institute, the Centers for Disease Control and Prevention, and the North American Association of Central Cancer Registries and mortality data from the National Center for Health Statistics. A total of 1,638,910 new cancer cases and 577,190 deaths from cancer are projected to occur in the United States in 2012. During the most recent 5 years for which there are data (2004-2008), overall cancer incidence rates declined slightly in men (by 0.6% per year) and were stable in women, while cancer death rates decreased by 1.8% per year in men and by 1.6% per year in women. Over the past 10 years of available data (1999-2008), cancer death rates have declined by more than 1% per year in men and women of every racial/ethnic group with the exception of American Indians/Alaska Natives, among whom rates have remained stable. The most rapid declines in death rates occurred among African American and Hispanic men (2.4% and 2.3% per year, respectively). Death rates continue to decline for all 4 major cancer sites (lung, colorectum, breast, and prostate), with lung cancer accounting for almost 40% of the total decline in men and breast cancer accounting for 34% of the total decline in women. The reduction in overall cancer death rates since 1990 in men and 1991 in women translates to the avoidance of about 1,024,400 deaths from cancer. Further progress can be accelerated by applying existing cancer control knowledge across all segments of the population, with an emphasis on those groups in the lowest socioeconomic bracket.
Article
The objective of this study was to evaluate the tolerability, to establish a dosing regimen, and to evaluate the efficacy of intravesical docetaxel (DTX) formulations in a mouse model of bladder cancer. DTX in commercial formulation (Taxotere, DTX in Tween 80) or loaded in hyperbranched polyglycerols (HPGs) was evaluated. The synthesis and characterization of HPGs with hydrophobic cores and derivatized with methoxy poly(ethylene glycol) in the shell and further functionalized with amine groups (HPG-C(8/10)-MePEG and HPG-C(8/10)-MePEG-NH(2)) is described. Intravesical DTX in either commercial or HPGs formulations (up to 1.0 mg/mL) was instilled in mice with orthotopic bladder cancer xenografts and was well tolerated with no apparent signs of local or systemic toxicities. Furthermore, a single dose of intravesical DTX (0.5 mg/mL) loaded in HPGs was significantly more effective in reducing the tumor growth in an orthotopic model of bladder cancer than the commercial formulation of Taxotere. In addition, DTX-loaded HPG-C(8/10)-MePEG-NH(2) was found to be more effective at lower instillation dose than DTX (0.2 mg/mL)-loaded HPG-C(8/10)-MePEG. Overall, our data show promising antitumor efficacy and safety in a recently validated orthotopic model of bladder cancer. Further research is warranted to evaluate its safety and efficacy in early phase clinical trials in patients refractory to standard intravesical therapy.
Article
Despite the fact that the current histopathologic classification for bladder cancer has led to improved concepts for the clinical management of the disease, key questions with regard to assessment of risk for recurrence and/or progression to invasive disease remain. In addition, response to specific therapies cannot be predicted accurately. Bladder tumors comprise a heterogeneous group with respect to both histopathology and clinical behavior. Thus, it is anticipated that a thorough knowledge and interpretation of the molecular alterations involved in tumor development and progression will lead to greater prognostic and predictive power. This may not only lead to better comprehension of the biology of the disease, but may also lead to the development of novel individualized therapies. Novel means of stratification are urgently needed to provide a new subclassification of urothelial lesions. This review discusses and summarizes the genetic alterations that have been reported in bladder cancer and relates these to the current 2-pathway model for tumor development. The molecular pathogenesis of high-grade noninvasive papillary tumors and of T1 tumors is not yet clear, and possibilities are discussed.
Article
Bladder cancer is the fifth most common human malignancy and the second most frequently diagnosed genitourinary tumor after prostate cancer. The majority of malignant tumors arising in the urinary bladder are urothelial carcinomas. Clinically, superficial bladder tumors (stages Ta and Tis) account for 75% to 85% of neoplasms, while the remaining 15% to 25% are invasive (T1, T2–T4) or metastatic lesions at the time of initial presentation. Several studies have revealed that distinct genotypic and phenotypic patterns are associated with early vs. late stages of bladder cancer. Early superficial disease appears to segregate into 2 main pathways: (1) superficial papillary bladder tumors, which are characterized by gain-of-function mutations affecting oncogenes such as H-RAS, FGFR3, and PI3K, and deletions of the long arm of chromosome 9 (9q); (2) Carcinoma in situ, a “flat” high grade lesion considered to be a precursor of invasive cancer, is characterized by loss-of-function mutations affecting tumor suppressor genes, such as p53, RB, and PTEN. Based on these data, a model for bladder tumor progression has been proposed in which 2 separate genetic pathways characterize the evolution of early bladder neoplasms. Several molecular markers have been correlated with tumor stage, but the rationale for these 2 well-defined genetic pathways still remains unclear.
Article
To report the long-term clinical outcomes and durability of response after treatment with induction intravesical docetaxel. Most novel agents used to treat bacillus Calmette-Guerin refractory high-grade non-muscle-invasive (NMI) bladder cancer are evaluated only after short follow-up periods. Our previously published phase I trial demonstrated that docetaxel is a safe agent for intravesical therapy with minimal toxicity and no detectable systemic absorption. We sought to determine long-term clinical outcomes after treatment with intravesical docetaxel. Eighteen patients with recurrent Ta (n = 7), T1 (n = 5), and Tis (n = 6) transitional cell carcinoma who experienced treatment failure with at least 1 prior intravesical therapy completed the phase I trial. Docetaxel was administered as 6 weekly intravesical instillations using a dose-escalation model terminated at 0.75 mg/mL. Efficacy was evaluated by interval cystoscopy with biopsies when indicated, cytology, and computed tomography imaging. Follow-up consisted of quarterly cystoscopy, cytology, computed tomography, and biopsy when indicated. With a median follow-up of 48.3 months, 4 patients (22%) have demonstrated a complete durable response and currently remain disease-free without further treatment. Three patients (17%) had a partial response, defined as a single NMI recurrence with no further therapy for bladder cancer. Eleven patients (61%) failed treatment, and required another intervention. One patient developed stage progression. No delayed toxicities were noted. The median disease-free survival time was 13.3 months. After 4 years of follow-up without maintenance therapy, intravesical docetaxel has demonstrated the ability to prevent recurrence in a select number of patients with refractory NMI bladder cancer and warrants further investigation.
Article
Bladder cancer is a heterogeneous disease, with 70% of patients presenting with superficial tumours, which tend to recur but are generally not life threatening, and 30% presenting as muscle-invasive disease associated with a high risk of death from distant metastases. The main presenting symptom of all bladder cancers is painless haematuria, and the diagnosis is established by urinary cytology and transurethral tumour resection. Intravesical treatment is used for carcinoma in situ and other high grade non-muscle-invasive tumours. The standard of care for muscle-invasive disease is radical cystoprostatectomy, and several types of urinary diversions are offered to patients, with quality of life as an important consideration. Bladder preservation with transurethral tumour resection, radiation, and chemotherapy can in some cases be equally curative. Several chemotherapeutic agents have proven to be useful as neoadjuvant or adjuvant treatment and in patients with metastatic disease. We discuss bladder preserving approaches, combination chemotherapy including new agents, targeted therapies, and advances in molecular biology.