You are on page 1of 7

EUROPEAN UROLOGY 64 (2013) 465–471

available at www.sciencedirect.com
journal homepage: www.europeanurology.com

Bladder Cancer

Prospective Evaluation of a Molecular Marker Panel for Prediction


of Recurrence and Cancer-specific Survival After Radical Cystectomy

Yair Lotan a,*, Aditya Bagrodia a, Niccolo Passoni b, Varun Rachakonda a, Payal Kapur c,
Yull Arriaga d, Christian Bolenz e, Vitaly Margulis a, Ganesh V. Raj a,
Arthur I. Sagalowsky a, Shahrokh F. Shariat f,g
a b
Department of Urology, University of Texas Southwestern Medical Center Dallas, TX, USA; Department of Urology, University Vita-Salute San Raffaele,
Milan, Italy; c Department of Pathology, University of Texas Southwestern Medical Center Dallas, TX, USA; d Division of Medical Oncology, University of Texas
e
Southwestern Medical Center Dallas, TX, USA; Department of Urology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany;
f
Department of Urology, Weill Cornell Medical College, New York Presbyterian Hospital, New York, NY, USA; g Division of Medical Oncology, Weill Cornell
Medical College, New York Presbyterian Hospital, New York, NY, USA

Article info Abstract

Article history: Background: Retrospective studies demonstrated that cell cycle–related and prolifera-
Accepted March 24, 2013 tion biomarkers add information to standard pathologic tumor features after radical
Published online ahead of cystectomy (RC). There are no prospective studies validating the clinical utility of
markers in bladder cancer.
print on April 3, 2013 Objective: To prospectively determine whether a panel of biomarkers could identify
patients with urothelial carcinoma of the bladder (UCB) who were likely to experience
Keywords: disease recurrence or mortality.
Design, setting, and participants: Between January 2007 and January 2012, every patient
Bladder cancer
with high-grade bladder cancer, including 216 patients treated with RC and lymphadenec-
Molecular markers tomy, underwent immunohistochemical staining for tumor protein p53 (Tp53); cyclin-
Cystectomy dependent kinase inhibitor 1A (p21, Cip1) (CDKN1A); cyclin-dependent kinase inhibitor
1B (p27, Kip1); antigen identified by monoclonal antibody Ki-67 (MKI67); and cyclin E1.
Intervention: Every patient underwent RC and lymphadenectomy, and marker staining.
Outcome measurements and statistical analysis: Cox regression analyses tested the
ability of the number of altered biomarkers to predict recurrence or cancer-specific
mortality (CSM).
Results and limitations: Pathologic stage among the study population was pT0 (5%), pT1
(35%), pT2 (19%), pT3 (29%), and pT4 (13%); lymphovascular invasion (LVI) was seen in 34%.
The median number of removed lymph nodes was 23, and 60 patients had lymph node
involvement (LNI). Median follow-up was 20 mo. Expression of p53, p21, p27, cyclin E1,
and Ki-67 were altered in 54%, 26%, 46%, 15%, and 75% patients, respectively. In univariable
analyses, pT stage, LNI, LVI, perioperative chemotherapy (CTx), margin status, and number
of altered biomarkers predicted disease recurrence. In a multivariable model adjusting for
pathologic stage, margins, LNI, and adjuvant CTx, only LVI and number of altered bio-
markers were independent predictors of recurrence and CSM. The concordance index of a
baseline model predicting CSM (including pathologic stage, margins, LVI, LNI, and adjuvant
CTx) was 80% and improved to 83% with addition of the number of altered markers.
Conclusions: Molecular markers improve the prediction of recurrence and CSM after RC.
They may identify patients who might benefit from additional treatments and closer
surveillance after cystectomy.
# 2013 European Association of Urology. Published by Elsevier B.V. All rights reserved.

* Corresponding author. Department of Urology, The University of Texas Southwestern Medical


Center at Dallas, 5323 Harry Hines Blvd., Dallas, TX 75390, USA. Tel. +1 214 648 0389;
Fax: +1 214 648 8786.
E-mail address: Yair.lotan@utsouthwestern.edu (Y. Lotan).
0302-2838/$ – see back matter # 2013 European Association of Urology. Published by Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.eururo.2013.03.043
466 EUROPEAN UROLOGY 64 (2013) 465–471

1. Introduction 2. Materials and methods

Urothelial carcinoma of the bladder (UCB) is the fifth All procedures described in the present study were undertaken with the
most common cancer worldwide, with an estimated approval and oversight of the Institutional Review Board for the
73 510 cases and 14 880 deaths in the United States for Protection of Human Subjects. All patients were treated at University of
Texas Southwestern Medical Center Dallas, TX, USA.
2012 [1]. Up to 75% of UCB patients are diagnosed with
non–muscle-invasive (NMI) disease, most of which can be
2.1. Patient population
managed by transurethral resection (TUR) and intrave-
sical therapy. However, 30% of patients with high-grade
A prospective study was designed to perform immunohistochemical
NMI UCB will eventually experience disease progression
staging for cyclin E1, Tp53, CDKN1A, p27, and MKI67 on all high-grade
to muscle-invasive (MI) disease [2]. For these patients UCB specimens at our institution starting in January 2007. This study
and the 25% of patients who present with primary MI focused on 216 consecutive patients who underwent RC with bilateral
UCB, radical cystectomy (RC) and bilateral lymphadenec- lymphadenectomy for UCB with curative intent. Preoperative imaging
tomy with or without perioperative chemotherapy (CTx) included chest radiographs and computed tomography scans of the
represents the best chance for cure [3–5]. Unfortunately, abdomen and pelvis. Marker staining was performed on the TUR
>25% of patients classified as having favorable pathologic specimen prior to RC on 36 specimens and the remaining 180 patients
features such as pathologic stages pT1–pT3a without had staining of their RC specimens. Overall, 48 patients (22%) underwent
nodal involvement (pN0) at the time of RC experience neoadjuvant CTx and 29 (13%) underwent adjuvant CTx based on high-
risk features. Neoadjuvant CTx consisted of gemcitabine and cisplatinum
disease recurrence within 5 yr of their surgery. These
in 34 patients (two cycles [n = 2], three cycles [n = 17], four cycles
patients eventually develop metastasis and succumb to
[n = 13], five cycles [n = 2]); methotrexate, vinblastine, doxorubicin, and
their disease [3–5].
cisplatin (MVAC) in nine patients (two cycles [n = 1], three cycles [n = 5],
This variability in outcomes among patients illustrates four cycles [n = 3]); one patient received gemzar/carboplatin for four
the inherent biologic and clinical heterogeneity of UCB. cycles; one patient received ifosfamide and adriamycin for three cycles;
The American Joint Committee on Cancer (AJCC) TNM one patient received ifosfamide/cisplatin/paclitaxel for four cycles; one
staging system, which currently guides UCB management, patient received four cycles of gemcitabine and carboplatin; and one
is limited in its ability to accurately predict an individuals’ received cisplatin-based CTx, but unknown other drugs for three cycles.
risk of disease recurrence and/or UCB-specific mortality Adjuvant therapy consisted of gemcitabine and cisplatinum for three
[6,7]. Even the combination of clinical and pathologic cycles (n = 5), four cycles (n = 17), five cycles (n = 2), or six cycles (n = 1);
variables in mathematical algorithms such as nomograms MVAC was given to two patients (one for three cycles and one for four
cycles); carboplatin and taxol were given to one patient for four cycles;
reaches discrimination for UCB recurrence of only 78%
and one patient was given carboplatin for one cycle followed by gemzar
[6,7].
for four cycles. None of the patients received prior radiation therapy of
Over the last decade, various investigators have shown the pelvis.
that alterations in candidate protein expression profiles All UCB specimens were examined and evaluated by pathologists
may help refine the understanding of the biologic and with expertise in genitourinary diseases according to previously
clinical behavior of UCB, thereby improving the risk described protocols [13,17]. The UCB specimens were staged and graded
stratification and clinical management of UCB patients according to the AJCC 2002 TNM classification and the 1998 World
[8]. The development of emerging biomarkers might be Health Organization/International Society of Urologic Pathology classi-
considered a highly regulated process that is conceptually fication, respectively [18]. Comprehensive clinical and pathologic
similar to therapeutic drug evaluation [9]. parameters of each patient were entered into an institutional review
board-approved prospective database.
Therefore, we have undertaken a phased, systematic
All patients were followed according to a previously described
evaluation and validation of a panel of established biomark-
standardized protocol [3]. Briefly, patients were evaluated postopera-
ers (cyclin E1; tumor protein p53 [Tp53; formerly p53];
tively at least every 3–4 mo for the first year, semiannually for the second
cyclin-dependent kinase inhibitor 1A [p21, Cip1] [CDKN1A; year, and annually thereafter. Follow-up visit consisted on physical
formerly p21]; cyclin-dependent kinase inhibitor 1B (p27, examination and serum chemistry evaluation. Computed tomography
Kip1); and antigen identified by monoclonal antibody Ki-67 scanning of abdomen and pelvis was performed every 6 mo and chest
[MKI67; formerly Ki-67]) selected based on extensive clinical radiography was performed every 3–4 mo in first 2 yr, then semiannually
data supporting their importance in UCB progression and or when clinically indicated.
metastasis [10–12]. As part of this staged approach, we have
established the potential of this panel for UCB prognostica- 2.2. Immunohistochemistry and scoring
tion in retrospective, single-institution studies and in large,
Immunohistochemical stainings on serial sections from the same
multi-institutional studies [11,13–16].
paraffin-embedded tumor block were performed for cyclin E1, Tp53,
In the next phase, we initiated a prospective protocol to
CDKN1A, p27, and MKI67 using the Dako Autostainer (Dako North
test the prognostic value of these biomarkers through
America Inc, Carpinteria, CA, USA). The staining and scoring protocols for
improved staging of patients with high-grade UCB under- all antibodies were previously described in detail [13,15]. Specimens of
going RC. If the additive value of the biomarkers panel is normal human urothelium from patients undergoing cystectomy for
confirmed in this prospective study, we plan to integrate nonmalignant disease served as internal controls. For semiquantitative
this panel in the design of clinical trials of adjuvant scoring, bright-field microscopy imaging coupled with an advanced
therapies. color detection software (Automated Cellular Imaging System, Clarient,
EUROPEAN UROLOGY 64 (2013) 465–471 467

CA, USA) was used. Staining intensity and percentage of positive nuclei Table 1 – Clinical and pathologic characteristics of patients
per area measurements were evaluated using 10 random hotspots within
each specimen. Nuclear Tp53 immunoreactivity was considered altered n = 216
when samples demonstrated at least 10% nuclear reactivity. CDKN1A Age at surgery, yr, median (IQR) 70 (62–76)
immunoreactivity was considered altered when samples demonstrated Sex, no. (%)
<10% staining. Nuclear p27 and cyclin E were considered altered when Male 171 (79)
samples demonstrated <30% nuclear reactivity. MKI67 immunoreactivity Female 45 (21)
was considered altered when samples showed >10% nuclear reactivity. Time from TUR to RC, mo, median (IQR) 2.1 (1.3–4.3)
Clinical stage, no. (%)
Staining was done on tissue from the RC or transurethral resection of
cTa/Tis/T1 96 (44)
bladder tumor (TURBT) performed within 3 mo prior to cystectomy.
cT2 85 (39)
Based on the number of altered biomarkers, we used an established cT3/T4 19 (9)
prognostic score (PS) as previously reported [17]. An altered expression Unknown 16 (7)
of none to two biomarkers was considered favorable, whereas an Clinical grade, no. (%)
alteration of at least three biomarkers represented an unfavorable PS. Low 4 (2)
High 201 (93)
Unknown 11 (5)
2.3. Statistical analyses
Concomitant CIS at TUR, no. (%)
No 160 (74)
The Fisher exact test and the Mann-Whitney U test were used to Yes 42 (19)
evaluate associations between biomarker expression and clinicopath- Unknown 14 (6)
ologic parameters. The Kaplan-Meier method was used to explore Pathological stage, no. (%)
pT0 8 (4)
recurrence-free survival (RFS) and cancer-specific survival (CSS), and
pT1 78 (36)
group differences were assessed using the log-rank test. Univariable
pT2 43 (20)
and multivariable Cox regression analyses tested the association pT3 59 (27)
between different prognostic variables and RFS and CSS. A multivari- pT4 28 (13)
able, Cox regression model predicting either RFS or CSS was constructed Pathologic grade, no. (%)
accounting for pathologic stage (pT0 vs pT1 vs pT2 vs pT3–4), soft- 0 8 (4)
Low 2 (1)
tissue margin status, lymphovascular invasion (LVI), lymph node
High 206 (95)
invasion (LNI), adjuvant CTx, and number of altered biomarkers.
Concomitant CIS at final pathology, no. (%)
Furthermore, the change in discrimination resulting from the addition No 120 (56)
to a baseline model (including pathologic stage, margin status, LVI, LNI, Yes 96 (44)
and adjuvant CTx) of the status of each individual biomarker, or the Lymphovascular invasion, no. (%)
number of altered biomarkers, or the prognostic score was quantified No 143 (66)
using the Harell Concordance index [19]. All statistical tests were Yes 73 (34)
Lymph node metastases, no. (%)
performed with the use of Stata v.12 (Stata Corp, College Station, TX,
No 161 (75)
USA). All tests were two-sided with a significance level at 0.05.
Yes 55 (25)
Total removed lymph nodes, no., median (IQR) 23.0 (16.0–32.0)
3. Results Positive lymph nodes, no., median (IQR) 3.0 (1.0–10.0)
Lymph node density, median (IQR) 14.3 (7.7–42.1)
Soft-tissue margin status, no. (%)
3.1. Demographics Negative 200 (93)
Positive 16 (7)
The patient demographics are shown in Table 1. The median Neoadjuvant chemotherapy, no. (%)
No 168 (78)
patient age was 70 yr with most being men (n = 192, 79%).
Yes 48 (22)
Most patients’ cancers were in clinical stage cTa/Tis/T1 Adjuvant chemotherapy, no. (%)
(n = 96, 44%) and cT2 (n = 85, 39%). After RC, there was a No 187 (87)
significant upstaging, with 27% at stage pT3 (n = 59) and 13% Yes 29 (13)

at pT4 (n = 28) when compared to clinical staging at time of IQR = interquartile range; TUR = transurethral resection; RC = radical
TURBT. Concomitant carcinoma in situ was seen in 96 (44%) cystectomy; CIS = carcinoma in situ.
patients and LVI in 73 (34%). The median number of removed
lymph nodes was 23 (interquartile range [IQR]: 16–32) and
LNI was seen in 55 patients (25%). Recurrence was noted in 15%, and 75% patients, respectively. Each marker including
64 patients after cystectomy with a median follow-up of the number of altered markers, with the exception of
20 mo (IQR: 10–37 mo). Of the recurrences, 8 were local, CDKN1A, was associated with recurrence after RC. The
11 were local and distant, and 45 were distant only. RFS at 6, numbers of altered markers, as well as individual markers
12, and 24 mo was 85%, 76%, and 68%, respectively. Overall, p27, Tp53, and MKI67, were associated with cancer-specific
51 patients died of bladder cancer-related causes. CSS rates at mortality (CSM) after RC. Figures 1 and 2 show estimates
6, 12, and 24 mo was 95%, 84%, and 73%, respectively. of disease recurrence-free and cancer-free survival proba-
bilities based on the number of altered markers demon-
3.2. Marker status and univariable analysis strating worse outcomes with increasing number of altered
markers.
Marker expression and association with disease recurrence In univariable analyses for disease recurrence (Table 3),
is shown in Tables 2 and 3. Expression of Tp53, CDKN1A, pT3/T4, positive soft-tissue margins, LNI, LVI, perioperative
p27, cyclin E1, and MKI67 were altered in 54%, 26%, 46%, CTx, and number of altered biomarkers were predictors of
468 EUROPEAN UROLOGY 64 (2013) 465–471

Table 2 – Alterations of cell-cycle makers at radical cystectomy in the overall population and according to recurrence

Overall (n = 216) No recurrence (n = 152) Recurrence (n = 64) p value

Altered biomarkers, no. (%)


0 8 (4) 8 (5) 0 (0) 0.001
1 45 (21) 41 (27) 4 (6)
2 83 (38) 56 (37) 27 (42)
3 63 (29) 39 (26) 24 (38)
4 17 (8) 8 (5) 9 (14)
Altered biomarkers, no. (%)
2 136 (63) 105 (69) 31 (48) 0.004
>2 80 (37) 47 (31) 33 (52)
Cyclin E, no. (%)
Negative 183 (85) 123 (81) 60 (94) 0.017
Positive 33 (15) 29 (19) 4 (6)
CDKN1A, no. (%)
Negative 160 (74) 116 (76) 44 (69) 0.2
Positive 56 (26) 36 (24) 20 (31)
p27, no. (%)
Negative 117 (54) 89 (59) 28 (44) 0.046
Positive 99 (46) 63 (41) 36 (56)
Tp53, no. (%)
Negative 99 (46) 78 (51) 21 (33) 0.013
Positive 117 (54) 74 (49) 43 (67)
MKI67, no. (%)
Negative 54 (25) 53 (35) 1 (2) <0.0001
Positive 162 (75) 99 (65) 63 (98)

CDKN1A = cyclin-dependent kinase inhibitor 1A (p21, Cip1); p27 = cyclin-dependent kinase inhibitor 1B (p27, Kip1); Tp53 = tumor protein p53; MKI67 = antigen
identified by monoclonal antibody Ki-67.

Table 3 – Univariate analysis for recurrence and cancer-specific mortality

Recurrence Cancer-specific mortality

HR IQR p value HR IQR p value

Age 1 0.98–1.03 0.9 1 0.97–1.02 0.8


Sex 1.18 0.66–2.11 0.6 1.51 0.83–2.76 0.2
pT0 0.83 0.20–3.38 0.8 0.91 0.22–3.76 0.9
pT1 0.18 0.09–0.39 <0.0001 0.12 0.04–0.33 <0.0001
pT2 0.44 0.20–0.98 0.043 0.5 0.21–1.16 0.11
pT3–4 6.4 3.65–11.21 <0.0001 6.77 3.54–12.96 <0.0001
Soft-tissue margins 2.53 1.15–5.57 0.022 2.97 1.33–6.65 0.008
Concomitant CIS 1.25 0.76–2.05 0.4 1.46 0.84–2.54 0.2
LVI 5.74 3.40–9.68 <0.0001 6.77 3.70–12.42 <0.0001
LNI 4.58 2.78–7.53 <0.0001 5.38 3.09–9.40 <0.0001
Positive nodes, no. 1.04 1.00–1.07 0.031 1.03 1.00–1.07 0.07
LND 1.02 1.01–1.03 0.001 1.02 1.00–1.03 0.021
Neoadjuvant chemotherapy 1.88 1.09–3.23 0.023 1.64 0.88–3.05 0.12
Adjuvant chemotherapy 2.98 1.74–5.10 <0.0001 3.73 2.10–6.63 <0.0001
Bladder prognosis score 2.41 1.47–3.97 0.001 2.64 1.51–4.59 0.001
Altered biomarkers, no. 1.95 1.49–2.54 <0.0001 1.98 1.48–2.65 <0.0001
Cyclin E 0.35 0.13–0.97 0.044 0.33 0.10–1.05 0.061
CDKN1A 1.57 0.92–2.67 0.1 1.54 0.85–2.79 0.2
p27 1.88 1.14–3.11 0.013 1.99 1.14–3.49 0.016
Tp53 2.16 1.27–3.69 0.005 2.42 1.32–4.42 0.004
MKI67 29.08 4.02–210.29 0.001 2.86  1015 0.00* <0.0001

HR = hazard ratio; IQR = interquartile range; CIS = carcinoma in situ; LVI = lymphovascular invasion; LNI = lymph node invasion; LND = lymph node dissection;
CDKN1A = cyclin-dependent kinase inhibitor 1A (p21, Cip1); p27 = cyclin-dependent kinase inhibitor 1B (p27, Kip1); Tp53 = tumor protein p53; MKI67 = antigen
identified by monoclonal antibody Ki-67.
*
All patients who died of bladder cancer had altered MKI67.

disease recurrence after RC. These same factors were 1.2–4.67; p = 0.013) and the number of altered biomarkers
predictors of CSM with the exception of neoadjuvant CTx use. (HR: 1.83; 95% CI, 1.34–2.52; p = 0.0002) were independent
predictors of disease recurrence, after adjusting for the
3.3. Multivariable analyses effects of for pathologic stage, margin status, LNI, and
adjuvant CTx (Table 4). Only LVI (HR: 2.8; 95% CI, 1.29–6.05;
In a multivariable model of disease recurrence after RC, only p = 0.009) and the number of altered markers (HR: 2.05;
LVI (hazard ratio [HR]: 2.37; 95% confidence interval [CI], 95% CI, 1.42–2.96; p = 0.0001) were independent predictors
EUROPEAN UROLOGY 64 (2013) 465–471 469

Table 4 – Multivariate analysis for recurrence and cancer-specific mortality

Recurrence Cancer-specific mortality

HR 95% CI p value HR 95% CI p value

Pathologic Stage
pT0 Ref – – Ref – –
pT1 0.25 0.05–1.22 0.088 0.17 0.03–0.96 0.044
pT2 0.24 0.05–1.23 0.087 0.23 0.04–1.23 0.086
pT3-T4 1.02 0.23–4.60 1 0.93 0.21–4.11 0.9
Soft-tissue margins 0.57 0.25–1.31 0.2 0.63 0.27–1.46 0.3
LVI 2.37 1.20–4.67 0.013 2.8 1.29–6.05 0.009
LNI 1.51 0.77–2.95 0.2 1.34 0.64–2.81 0.4
Adjuvant chemotherapy 0.79 0.41–1.54 0.5 0.99 0.50–1.99 1
Altered biomarkers. no. 1.83 1.34–2.52 0.0002 2.05 1.42–2.96 0.0001

HR = hazard ratio; CI = confidence interval; Ref = reference; LVI = lymphovascular invasion; LNI = lymph node invasion.

of CSM (all p < 0.01) after adjusting for the effects of The concordance index of a baseline model predicting
pathologic stage, margin status, LNI, and adjuvant CTx. A disease recurrence (including pathologic stage [pT0 vs pT1
separate multivariable analysis adjusting for pathologic vs pT2 vs pT3/T4], soft-tissue margins, LVI, LNI, and
stage, margin status, LNI, and neoadjuvant CTx also found adjuvant CTx) was 79%. The addition of the number of
that LVI and number of altered markers were independent altered markers improved prediction by 3% to 82%. The
predictors of recurrence and CSM, while neoadjuvant CTx addition of MKI67 alone improved prediction of disease
[(Fig._1)TD$IG]
was not an independent predictor of recurrence or CSM. recurrence to 84%, which was more than any other marker
alone, since only one patient with MKI67 negative status
experienced disease recurrence. The concordance index
of the baseline model predicting CSM was 80%. The addition
of the number of altered markers improved prediction by
3% to 83%. The addition of MKI67 alone improved prediction
of CSM to 85%, which was more than any other marker
alone, since every patient who died of UCB had altered
MKI67 status. The addition of Tp53 status alone only added
1% to the prediction of disease recurrence and CSM.

4. Discussion

The utility and importance of biomarkers has been recog-


nized and biomarker discovery efforts are now common in
both academic and industrial settings. Yet despite intensified
interest and investment, few novel biomarkers are used in
Fig. 1 – Kaplan-Meier curve depicting recurrence-free survival according clinical practice [9]. The reasons for this disjunction are
to number of altered biomarkers ( p = 0.004). alt. = altered; manifold and reflect the long and difficult pathway from
BM = biomarker.
candidate biomarker discovery to clinical assay, and the lack
[(Fig._2)TD$IG]
of coherent and comprehensive processes for biomarker
development. The development of various emerging bio-
markers might be considered a highly regulated process that
is conceptually similar to therapeutic drug evaluation,
especially from the point where it enters human testing.
We have adhered to a formal structure and phased
approach to guide the process of biomarker development
and validation [9]. First, we performed single-center,
retrospective, hypothesis-generating, human studies test-
ing the value of the biomarker panel shown in this article
[10–12]. Subsequently, our group found that a marker
panel performed better than individual markers and
improved prediction of disease recurrence and mortality
when modeled together with conventional clinicopatho-
logic features [17]. In the latter study, we found that a
panel of cyclin E1, Tp53, CDKN1A, p27, retinoblastoma
Fig. 2 – Kaplan-Meier curve depicting cancer-specific survival according
to number of altered biomarkers ( p < 0.0001). alt. = altered;
protein (pRb), and MKI67 provided discriminatory infor-
BM = biomarker. mation that significantly improved the ability to predict
470 EUROPEAN UROLOGY 64 (2013) 465–471

recurrence and UCB-specific survival in 191 pT1–pT3 pN0 by knowing the number of altered markers could there be
patients who underwent RC [17]. We recently externally more differentiation of an individual patient’s outcomes.
validated the value of this panel in large, retrospective, We and others have shown that investigating multiple
multicenter studies [15–20]. The current study represents pathways, including downstream effectors (eg, of Tp53),
the culmination of these efforts into a prospective clinical can enhance the predictive power provided by a single
trial of patients with high-grade UCB. There are scant marker [11,12,26]. Furthermore, Tp53 alone only added 1%
prospective data available in oncology, and the clinical to the predictive accuracy of a model using stage, LVI, and
utility of few biomarkers is established [21–25]. LNI.
We found that expression of a higher number of altered Our prospective study has some limitations. There are
biomarkers was an independent predictor of disease issues related to the reliability of immunohistochemical
recurrence and CSM after RC along with LVI, after adjusting biomarker assessment. They are highly dependent on a
for the effects of pathologic stage, margin status, LNI, and range of poorly controlled variables, including choice of
adjuvant CTx. Furthermore, the concordance index of a antibody, antibody dilution, fixation techniques, and
baseline model including pathologic stage, margin status, inconsistency in specimen handling. However, we reduced
LVI, LNI, and adjuvant CTx improved from 79% to 82% after the number of variables in the immunohistochemical
adding the number of altered biomarkers. These findings techniques by using highly standardized protocols, includ-
support prior retrospective studies showing that assess- ing established staining protocols and automated scoring
ment of biomarker status may identify patients at high risk systems based on bright-field microscopy imaging coupled
for cancer recurrence who may benefit from perioperative with advanced color-detection software. These techniques
CTx and closer surveillance. We further confirmed the have been shown to improve experimental standardization
importance of MKI67 in identifying patients at high risk for and staining reproducibility. Furthermore, the prospective
CSM [13,14]. Every patient in our cohort who died of their assessment of each specimen in a consecutive manner
disease had altered MKI67. However, since a large propor- eliminates any biases. One consideration is that not every
tion of patients (65%) without recurrence also had altered marker was equally useful. Cyclin E was only abnormal in a
MKI67, MKI67 positivity alone has limited utility. In small number of patients and CDKN1A was not an
contrast, MKI67 negativity may have significant utility. independent predictor of recurrence or survival. The goal
Prospective studies evaluating the utility of molecular of the trial was not to optimize the most useful set of
biomarkers in surgically treated UCB are still sparse. Two markers but to validate a previously developed panel and
studies in NMI UCB evaluated the utility of Tp53 protein support the principle that a higher cumulative number of
overexpression and Tp53 gene mutation in predicting alterations predicts worse outcomes after cystectomy. It is
recurrence and progression and showed no predictive value likely that other markers will perform equally well or
[22,23]. Another large study in NMI UCB reported that better. Finally, this was not a homogenous population;
mutations in fibroblast growth factor receptor 3 (FGFR3) rather, it reflected a real-world scenario with some patients
were associated with increased rates of disease recurrence, receiving perioperative CTx while others did not. This could
but not disease progression or survival, in patients with affect the performance of the markers in unpredictable
pTa UCB [21]. Regarding patients with advanced disease, ways that may, in part, enhance or diminish their
one companion study to the Southwestern Oncology Group significance.
8710 trial (neoadjuvant CTx plus RC vs RC alone) evaluated
the prognostic role of MKI67, Tp53, and angiogenic changes 5. Conclusions
[25]. Tissue specimens were obtained before neoadjuvant
CTx from 42 patients randomized to the combination- Cell cycle–related and proliferation-related markers at time
treatment arm and 52 randomized to RC alone. Patients of cystectomy improve the prediction of recurrence and
whose tumors had increased MKI67 expression had better CSM after RC. Such a marker panel may help identify
progression-free survival (median: 66 vs 12 mo; p = 0.063) patients who might benefit from additional treatments and
and a nonsignificant increase in overall survival (median: closer surveillance after cystectomy.
73 mo vs 38 mo; p = 0.25). Tp53 expression and microvessel
density were not associated with outcomes. Unfortunately,
Author contributions: Yair Lotan had full access to all the data in the
this study did not include the entire trial population,
study and takes responsibility for the integrity of the data and the
and the small number of patients precluded further accuracy of the data analysis.
subanalyses.
A multicenter, adjuvant CTx study randomized patients Study concept and design: Lotan, Shariat.
with pT1/T2N0M0 disease whose tumors demonstrated Acquisition of data: Lotan, Bagrodia, Rachakonda, Kapur, Arriaga, Bolenz,
Margulis, Raj, Sagalowsky, Shariat.
10% nuclear reactivity on centrally performed immuno-
Analysis and interpretation of data: Lotan, Passoni, Kapur, Shariat.
histochemistry for Tp53 to three cycles of adjuvant MVAC
Drafting of the manuscript: Lotan, Shariat.
versus observation; Tp53-negative patients were observed
Critical revision of the manuscript for important intellectual content: Lotan,
[24]. This study found no difference in recurrence based on Bagrodia, Passoni, Rachakonda, Kapur, Arriaga, Bolenz, Margulis, Raj,
Tp53 status. These results are not surprising considering our Sagalowsky, Shariat.
findings, which demonstrated that patients with only one Statistical analysis: Lotan, Passoni, Shariat.
marker abnormality had favorable outcomes and that only Obtaining funding: Lotan.
EUROPEAN UROLOGY 64 (2013) 465–471 471

Administrative, technical, or material support: None. [12] Shariat SF, Tokunaga H, Zhou J, et al. p53, p21, pRB, and p16
Supervision: Lotan, Margulis, Raj, Sagalowsky, Shariat. expression predict clinical outcome in cystectomy with bladder
Other (specify): None. cancer. J Clin Oncol 2004;22:1014–24.
[13] Margulis V, Shariat SF, Ashfaq R, Sagalowsky AI, Lotan Y. Ki-67 is an
Financial disclosures: Yair Lotan certifies that all conflicts of interest,
independent predictor of bladder cancer outcome in patients trea-
including specific financial interests and relationships and affiliations
ted with radical cystectomy for organ-confined disease. Clin Cancer
relevant to the subject matter or materials discussed in the manuscript
Res 2006;12:7369–73.
(eg, employment/affiliation, grants or funding, consultancies, honoraria,
[14] Margulis V, Lotan Y, Karakiewicz PI, et al. Multi-institutional
stock ownership or options, expert testimony, royalties, or patents filed,
validation of the predictive value of Ki-67 labeling index in patients
received, or pending), are the following: None.
with urinary bladder cancer. J Natl Cancer Inst 2009;101:114–9.
Funding/Support and role of the sponsor: The Simmons Cancer Center [15] Shariat SF, Chromecki TF, Cha EK, et al. Risk stratification of organ
provided funding for this study and was involved in data collection. confined bladder cancer after radical cystectomy using cell cycle
related biomarkers. J Urol 2012;187:457–62.
References [16] Shariat SF, Lotan Y, Karakiewicz PI, et al. P53 predictive value for
pT1-2 N0 disease at radical cystectomy. J Urol 2009;182:907–13.
[1] Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer [17] Shariat SF, Karakiewicz PI, Ashfaq R, et al. Multiple biomarkers
J Clin 2012;62:10–29. improve prediction of bladder cancer recurrence and mortality in
[2] Lotan Y, Svatek RS, Sagalowsky AI. Should we screen for bladder patients undergoing cystectomy. Cancer 2008;112:315–25.
cancer in a high-risk population? A cost per life-year saved analysis. [18] Epstein JI, Amin MB, Reuter VR, Mostofi FK. The World Health
Cancer 2006;107:982–90. Organization/International Society of Urological Pathology consen-
[3] Shariat SF, Karakiewicz PI, Palapattu GS, et al. Outcomes of radical sus classification of urothelial (transitional cell) neoplasms of the
cystectomy for transitional cell carcinoma of the bladder: a con- urinary bladder. Bladder Consensus Conference Committee. Am J
temporary series from the Bladder Cancer Research Consortium. Surg Pathol 1998;22:1435–48.
J Urol 2006;176:2414–22, discussion 2422. [19] Harrell Jr FE, Lee KL, Mark DB. Multivariable prognostic models:
[4] Lotan Y, Gupta A, Shariat SF, et al. Lymphovascular invasion is issues in developing models, evaluating assumptions and adequa-
independently associated with overall survival, cause-specific cy, and measuring and reducing errors. Stat Med 1996;15:361–87.
survival, and local and distant recurrence in patients with negative [20] Shariat SF, Bolenz C, Karakiewicz PI, et al. P53 expression in patients
lymph nodes at radical cystectomy. J Clin Oncol 2005;23:6533–9. with advanced urothelial cancer of the urinary bladder. BJU Int
[5] Stein JP, Lieskovsky G, Cote R, et al. Radical cystectomy in 2009;105:489–95.
the treatment of invasive bladder cancer: long-term results in [21] Hernandez S, Lopez-Knowles E, Lloreta J, et al. FGFR3 and Tp53
1,054 patients. J Clin Oncol 2001;19:666–75. mutations in T1G3 transitional bladder carcinomas: independent
[6] Karakiewicz PI, Shariat SF, Palapattu GS, et al. Nomogram for distribution and lack of association with prognosis. Clin Cancer Res
predicting disease recurrence after radical cystectomy for transi- 2005;11:5444–50.
tional cell carcinoma of the bladder. J Urol 2006;176:1354–61, [22] Lopez-Knowles E, Hernandez S, Kogevinas M, et al., EPICURO Study
discussion 1361–2. Investigators. The p53 pathway and outcome among patients with
[7] Bochner BH, Kattan MW, Vora KC. Postoperative nomogram pre- T1G3 bladder tumors. Clin Cancer Res 2006;12:6029–36.
dicting risk of recurrence after radical cystectomy for bladder [23] Dalbagni G, Parekh DJ, Ben-Porat L, Potenzoni M, Herr HW,
cancer. J Clin Oncol 2006;24:3967–72. Reuter VE. Prospective evaluation of p53 as a prognostic marker
[8] Bolenz C, Lotan Y. Molecular biomarkers for urothelial carcinoma of in T1 transitional cell carcinoma of the bladder. BJU Int 2007;99:
the bladder: challenges in clinical use. Nat Clin Pract Urol 2008; 281–5.
5:676–85. [24] Stadler WM, Lerner SP, Groshen S, et al. Phase III study of molecularly
[9] Bensalah K, Montorsi F, Shariat SF. Challenges of cancer biomarker targeted adjuvant therapy in locally advanced urothelial cancer of
profiling. Eur Urol 2007;52:1601–9. the bladder based on p53 status. J Clin Oncol 2011;29:3443–9.
[10] Shariat SF, Ashfaq R, Sagalowsky AI, Lotan Y. Correlation of cyclin [25] Grossman HB, Natale RB, Tangen CM, et al. Neoadjuvant chemo-
D1 and E1 expression with bladder cancer presence, invasion, therapy plus cystectomy compared with cystectomy alone for
progression, and metastasis. Hum Pathol 2006;37:1568–76. locally advanced bladder cancer. N Engl J Med 2003;349:859–66.
[11] Shariat SF, Karakiewicz PI, Ashfaq R, et al. Multiple biomarkers [26] Shariat SF, Zlotta AR, Ashfaq R, Sagalowsky AI, Lotan Y. Cooperative
improve prediction of bladder cancer recurrence and mortality in effect of cell-cycle regulators expression on bladder cancer devel-
patients undergoing cystectomy. Cancer 2007;112:315–25. opment and biologic aggressiveness. Mod Pathol 2007;20:445–59.

You might also like