Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

E U R O P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

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

Platinum Priority – Review – Urothelial Cancer


Editorial by Ashish M. Kamat on pp. 105–106 of this issue

European Association of Urology Guidelines on Muscle-invasive


and Metastatic Bladder Cancer: Summary of the 2020 Guidelines

J. Alfred Witjes a,*, Harman Max Bruins b, Richard Cathomas c, Eva M. Compérat d,
Nigel C. Cowan e, Georgios Gakis f, Virginia Hernández g, Estefania Linares Espinós h,
Anja Lorch i, Yann Neuzillet j, Mathieu Rouanne j, George N. Thalmann k, Erik Veskimäe l,
Maria J. Ribal m, Antoine G. van der Heijden a
a
Department of Urology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands; b Department of Urology, Zuyderland Medical Center,
c d
Sittard-Geleen-Heerlen, The Netherlands; Department of Medical Oncology, Kantonsspital Graubünden, Chur, Switzerland; Department of Pathology,
e
Sorbonne University, Assistance Publique-Hôpitaux de Paris, Hopital Tenon, Paris, France; Department of Radiology, The Queen Alexandra Hospital,
f g
Portsmouth, UK; Department of Urology and Pediatric Urology, University of Würzburg, Würzburg, Germany; Department of Urology, Hospital
Universitario Fundación Alcorcón, Madrid, Spain; Department of Urology, Hospital Universitario La Paz, Madrid, Spain; i Department of Medical Oncology
h

and Hematology, University Hospital Zürich, Zürich, Switzerland; j Department of Urology, Foch Hospital, University of Versailles-Saint-Quentin-en-Yvelines,
Suresnes, France; k Department of Urology, Inselspital, University Hospital Bern, Switzerland; l Department of Urology, Tampere University Hospital, Tampere,
m
Finland; Department of Urology, Hospital Clinic, University of Barcelona, Barcelona, Spain

Article info Abstract

Article history: Context: This overview presents the updated European Association of Urology (EAU)
Accepted March 31, 2020 guidelines for muscle-invasive and metastatic bladder cancer (MMIBC).
Objective: To provide practical evidence-based recommendations and consensus state-
Associate Editor: ments on the clinical management of MMIBC with a focus on diagnosis and treatment.
Evidence acquisition: A broad and comprehensive scoping exercise covering all areas of
James Catto
the MMIBC guideline has been performed annually since its 2017 publication (based on
the 2016 guideline). Databases covered by the search included Medline, EMBASE, and
Keywords: the Cochrane Libraries, resulting in yearly guideline updates. A level of evidence and a
Bladder cancer grade of recommendation were assigned. Additionally, the results of a collaborative
multistakeholder consensus project on advanced bladder cancer (BC) have been incor-
Muscle invasive
porated in the 2020 guidelines, addressing those areas where it is unlikely that
Metastatic prospective comparative studies will be conducted.
Diagnosis Evidence synthesis: Variant histologies are increasingly reported in invasive BC and are
Management relevant for treatment and prognosis. Staging is preferably done with (enhanced)
Cystectomy computerised tomography scanning. Treatment decisions are still largely based on
clinical factors. Radical cystectomy (RC) with lymph node dissection remains the
Multimodality recommended treatment in highest-risk non–muscle-invasive and muscle-invasive
Quality of life nonmetastatic BC, preceded by cisplatin-based neoadjuvant chemotherapy (NAC) for
Follow-up invasive tumours in “fit” patients. Selected men and women benefit from sexuality
Guidelines sparing RC, although this is not recommended as standard therapy. Open and robotic RC
show comparable outcomes, provided the procedure is performed in experienced
European Association of Urology
centres. For open RC 10, the minimum selected case load is 10 procedures per year.
* Corresponding author. Department of Urology, Intern Mail 659, Radboud University Nijmegen
Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands. Tel.: +31 24 36 13 735; Fax: +31 24
35 41 031.
E-mail address: Fred.Witjes@radboudumc.nl (J.A. Witjes).

https://doi.org/10.1016/j.eururo.2020.03.055
0302-2838/© 2020 European Association of Urology. Published by Elsevier B.V. All rights reserved.
E U RO P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4 83

If bladder preservation is considered, chemoradiation is an alternative in well-selected


patients without carcinoma in situ and after maximal resection. Adjuvant chemothera-
py should be considered if no NAC was given. Perioperative immunotherapy can be
offered in clinical trial setting. For fit metastatic patients, cisplatin-based chemotherapy
remains the first choice. In cisplatin-ineligible patients, immunotherapy in Programmed
Death Ligand 1 (PD-L1)-positive patients or carboplatin in PD-L1–negative patients is
recommended. For second-line treatment in metastatic disease, pembrolizumab is
recommended. Postchemotherapy surgery may prolong survival in responders. Quality
of life should be monitored in all phases of treatment and follow-up. The extended
version of the guidelines is available at the EAU website: https://uroweb.org/guideline/
bladder-cancer-muscle-invasive-and-metastatic/.
Conclusions: This summary of the 2020 EAU MMIBC guideline provides updated infor-
mation on the diagnosis and treatment of MMIBC for incorporation into clinical practice.
Patient summary: The European Association of Urology Muscle-invasive and Metastatic
Bladder Cancer (MMIBC) Panel has released an updated version of their guideline, which
contains information on histology, staging, prognostic factors, and treatment of MMIBC.
The recommendations are based on the current literature (until the end of 2019), with
emphasis on high-level data from randomised clinical trials and meta-analyses and on
the findings of an international consensus meeting. Surgical removal of the bladder and
bladder preservation are discussed, as well as the use of chemotherapy and immuno-
therapy in localised and metastatic disease.
© 2020 European Association of Urology. Published by Elsevier B.V. All rights reserved.

1. Introduction the management of advanced and variant bladder cancer


(BC) addressing areas in which it is unlikely that clinical
The current summary of the 2020 European Association of trials and prospective comparative studies will be con-
Urology (EAU) Muscle-invasive and Metastatic Bladder Cancer ducted [4,5]. The methodology used is presented in detail in
(MMIBC) guideline aims at helping urologists to assess the the scientific publications.
evidence-based management of muscle-invasive bladder After publication, the 2018 MIBC guidelines were shared
cancer (MIBC) and to incorporate guideline recommendations with seven patients treated for MIBC. Although they found
into their clinical practice. Guidelines, however, can never the text in general to be informative and instructive, some
replace clinical expertise when making treatment decisions parts were more difficult to understand, amongst others,
for individual patients in a specific context. The guideline due to many abbreviations and methodological elements.
panel consists of an international multidisciplinary group of Sections with less clear evidence were also less compre-
experts from the fields of urology, pathology, radiology, and hensible (eg, “recurrent disease” and “markers”). There was
oncology, whose conflicts of interest can be found online an interest in whether screening for BC is a consideration. In
(https://uroweb.org/guideline/bladder-cancer-muscle- “follow-up”, “quality of life (QoL)”, and “survivorship
invasive-and-metastatic/). aspects”, information should be presented on what patients
A broad and comprehensive literature search, limited to and families might expect after treatment. Finally, lifestyle
English-language publications, covering all sections of the elements would be of relevance, for which patient
MIBC guideline, is performed on a yearly basis. Databases involvement was considered missing.
searched included Medline, EMBASE, and the Cochrane Compared with the 2017 manuscript [6], essential new
Libraries. For the 2020 update, for example, 1899 unique passages are presented in the sections on variant histology,
records were identified and screened for relevance, result- function sparing cystectomy, node dissection, multimodal
ing in the inclusion of 62 new publications. A detailed therapy, surgical volume, chemo- and immunotherapy, and
search strategy is available online: http://uroweb.org/ QoL.
guideline/bladdercancer-muscle-invasive-andmetastatic/? Finally, in some tables, recommendations are presented
type=appendices-publications. that are not supported by text in this summary due to word
Search results are discussed by the panel members, and count limitations. For the background of these recommen-
selected publications are assigned a level of evidence based dations, the reader is referred to the full guideline text.
on a modified Oxford Centre for Evidence-Based Medicine
Levels of Evidence classification [1]. Recommendations are 2. Epidemiology
graded, either “strong” or “weak”, determined by the
balance between desirable and undesirable consequences BC epidemiology still shows a clear male predominance,
of alternative management strategies, the quality of the and incidence and mortality rates differ per European
evidence, and nature and variability of patient values and country, probably due to differences in risk factors,
preferences according to a modified Grades of Recommen- detection, and availability of treatments (Table 1).
dation, Assessment, Development and Evaluation (GRADE) Tobacco smoking intensity is the most well-established
approach [2,3]. risk factor for and directly related to BC, causing 50–65% of
The 2020 MIBC guidelines update includes the findings male cases and 20–30% of female cases [7]. An immediate
of a collaborative multistakeholder consensus project on decrease in the risk of BC was observed in those who
84 E U RO P E A N U RO L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

Table 1 – Summary of evidence and recommendations for improving. Recent meta-analyses suggested BC to have a
epidemiology and risk factors.
relation with urinary calculi and [13], but not with chronic
Summary of evidence LE urinary tract infection (UTI) [14].
Several studies have shown that women present with
Worldwide, BC is the 11th most commonly diagnosed 2a
cancer. more advanced disease and have worse survival rates. Older
Several risk factors associated with BC diagnosis have 3 studies have suggested a delay in diagnosis, smoking, and
been identified. chemical exposure as potential reasons. A more recent
Active and passive tobacco smoking continues to be the 2a
study showed differences in treatment as the main cause
main risk factor, while the exposure-related incidence is
decreasing. [15]; however, the difference disappears after 2 yr of follow-
The increased risk of developing BC in patients 3 up [16].
undergoing EBRT, brachytherapy, or a combination of Genetic susceptibility and familial shared environmental
EBRT and brachytherapy must be considered during
exposure are still under investigation.
patient follow-up. As BC requires time to develop,
patients treated with radiation at a young age are at the
greatest risk and should be followed up closely.
3. Pathology
Recommendations Strength
rating
Approximately 25% of patients with BC present with
Council patients to stop active smoking and avoid passive Strong muscle-invasive or metastatic disease. An adequate (trans-
smoking. urethral) resection should be followed by proper patholog-
Inform workers in potentially hazardous workplaces of Strong
ical handling and documentation of all margins, adjacent
the potential carcinogenic effects of a number of
recognised substances, including duration of exposure tissue, and lymph nodes (LNs) [17].
and latency periods. Protective measures are Morphological variants should be identified and de-
recommended. scribed according to the latest World Health Organization
Do not prescribe pioglitazone to patients with active BC Strong
(WHO) update because of their prognostic relevance
or a history of BC.
[18,19]. Currently, the following differentiations are used:
BC = bladder cancer; EBRT = external beam radiation therapy; LE = level of
evidence.

1 UC (>90% of all cases).


2 UC with partial squamous and/or glandular or tropho-
stopped smoking. The reduction was about 40% within 1– blastic differentiation.
4 yr of quitting smoking and 60% after 25 yr of cessation 3 Micropapillary or microcystic UC.
[8]. Occupational exposure to carcinogenic aromatic amines 4 Nested variant (including large nested variant).
is the second most important risk factor, accounting for 20– 5 Lymphoepithelioma-like.
25% of BC (dyes, rubbers, textiles, paints, and leathers) with 6 Plasmocytoid, signet ring, diffuse.
a latency period of several decades. Older studies have 7 Some UCs with small-cell carcinomas.
reported an increased rate of BC after external beam 8 Sarcomatoid carcinomas.
radiotherapy (EBRT) for gynaecological malignancies and 9 Poorly differentiated.
prostate cancer (PCa). More recently, it was proposed that
modern EBRT modalities for PCa, such as intensity- Other, extremely rare, variants exist, which are not listed
modulated radiotherapy (IMRT), may have lower rates of above.
in-field bladder and rectal secondary malignancies Aside from the guideline recommendations, statements
[9]. However, considering the long latency time, an were added based on the EAU/European Society for Medical
increased risk should be considered during follow-up. Oncology consensus meeting (Table 2) [4,5].
The link between dietary factors and BC remains
unproven in an on-going European prospective study, 4. Diagnostic evaluation of (muscle)-invasive BC
although recently an inverse association between flavo-
noids and lignans, and the risk of aggressive BC tumours has Ultimately, the diagnosis is made by cystoscopy, the
been described [10]. findings of which should be documented in detail, and
Elevated blood pressure and triglycerides are associated histological evaluation of resected tissue. If imaging studies
with increased risks of BC among men, whereas high body show a bladder tumour clearly, diagnostic cystoscopy may
mass index was associated with decreased BC risk [11]. The be omitted (Table 3). Before resection, bimanual examina-
association with diabetes mellitus is inconsistent, but tion under anaesthesia should be done, although interpre-
recently a dose-dependent association with BC was found tation is subjective [20]. Photodynamic diagnosis could be
for the oral hypoglycaemic drug pioglitazone [12], which considered to identify associated carcinoma in situ (CIS),
caused the US Food and Drug Administration (FDA) to since the presence of CIS may lead to a modified treatment
recommend not prescribing pioglitazone in patients with plan (eg, in case of chemoradiation). Transurethral resection
active BC. (TUR) should include a deeper part, which has to be sent in
There is a well-established relationship between schis- separately, and prostatic biopsies in males as well as
tosomiasis and urothelial carcinoma (UC) and squamous bladder neck biopsies in women who might be candidates
cell carcinoma of the bladder, but schistosomiasis control is for orthotopic diversion. For general information on the
E U RO P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4 85

Table 2 – Summary of recommendations and consensus statements for pathology.

Recommendations Strength rating

Record the depth of invasion (categories pT2a and pT2b, pT3a and pT3b, or pT4a and pT4b). Strong
Record margins with special attention paid to the radial margin, prostate, ureter, urethra, peritoneal fat, uterus, and vaginal
top.
Record the total number of LNs, the number of positive LNs, and extranodal spread.
Record lymphatic or blood vessel invasion.
Record the presence of carcinoma in situ.
EAU-ESMO consensus statements [4,5] a
Bladder urothelial carcinoma with small cell neuroendocrine variant should be treated with neoadjuvant chemotherapy
followed by consolidating local therapy.
Muscle-invasive pure squamous cell carcinoma of the bladder should be treated with primary radical cystectomy and
lymphadenectomy.
Muscle-invasive pure adenocarcinoma of the bladder should be treated with primary radical cystectomy and
lymphadenectomy.
Muscle-invasive small cell neuroendocrine variant of bladder urothelial carcinoma should not receive preventive brain
irradiation to avoid brain recurrence.

EAU = European Association of Urology; ESMO = European Society for Medical Oncology; LN = lymph node.
a
Only statements that met the a priori consensus threshold across all three stakeholder groups are listed (defined as 70% agreement and 15% disagreement, or
vice versa).

Table 3 – Summary of evidence, recommendations, and consensus statements for the primary assessment of presumably invasive bladder
tumours.

Summary of evidence LE

Cystoscopy is necessary for the diagnosis of bladder cancer. 1


Urinary cytology has high sensitivity in high-grade tumours including carcinoma in situ. 2b

Recommendations Strength
rating

Describe all macroscopic features of the tumour (site, size, number, and appearance) and mucosal abnormalities during Strong
cystoscopy. Use a bladder diagram.
Take a biopsy of the prostatic urethra in cases of bladder neck tumour, when bladder carcinoma in situ is present or Strong
suspected, when there is positive cytology without evidence of tumour in the bladder, or when abnormalities of the
prostatic urethra are visible.
Take a biopsy at the time of the second resection, if no biopsy was taken during the initial procedure. Strong
In women undergoing subsequent orthotopic neobladder construction, obtain procedural information (including Strong
histological evaluation) of the bladder neck and urethral margin, either prior to or at the time of cystoscopy.
Specify the grade, depth of tumour invasion, and whether the lamina propria and muscle tissue are present in the specimen Strong
in the pathology report.
EAU-ESMO consensus statements [4,5] a
Differentiating between urachal and nonurachal subtypes of adenocarcinoma is essential when making treatment decisions.

EAU = European Association of Urology; ESMO = European Society for Medical Oncology; LE = level of evidence.
a
Only statements that met the a priori consensus threshold across all three stakeholder groups are listed (defined as 70% agreement and 15% disagreement, or
vice versa).

assessment of bladder tumours, see EAU guidelines on non– Upper urinary tract imaging is best done with CT urography,
muscle-invasive BC (NMIBC) [21]. whereas MR urography should be reserved for patients who
cannot undergo CT urography.
5. Imaging Staging for pulmonary, hepatic, and LN metastases can
best be done with CT [23,24]. Staging of other sites should
Both computerised tomography (CT) and magnetic reso- be done in case of specific complaints.
nance imaging (MRI) may be used for assessment of local There is no consensus yet regarding the role of 18F-
invasion, with the principal aim of detecting T3b disease or fluorodeoxyglucose positron emission tomography/CT for
higher. However, a recent meta-analysis showed 91% metastatic staging (Table 4).
sensitivity and 96% specificity for 3.0-T device MRI
combined with diffusion-weighted imaging to differentiate 6. Comorbidity and radical cystectomy
 T1 tumours from  T2 tumours before surgery [22]. Sen-
sitivity and specificity of CT and MRI to detect LN metastasis Complications related to RC may, amongst others, be related
are low, where pelvic nodes >8 mm and abdominal nodes to pre-existing comorbidity (Table 5). Age above 70 or 80 yr
>10 mm should be regarded as pathologically enlarged. did not appear to be related to complications, but mortality
86 E U RO P E A N U RO L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

Table 4 – Summary of evidence and recommendations for staging in MIBC.

Summary of evidence LE

Imaging as part of staging in MIBC provides information about prognosis and assists in selection of the most appropriate 2b
treatment.
There are currently insufficient data on the use of diffusion-weighted imaging and 18F-fluorodeoxyglucose positron emission
tomography/computed tomography in MIBC to allow for a recommendation to be made.
The diagnosis of upper tract urothelial carcinoma depends on CT urography and ureteroscopy. 2

Recommendations Strength
rating

In patients with confirmed MIBC, use CT of the chest, abdomen, and pelvis as the optimal form of staging. Strong
Perform a CT urography for upper tract evaluation and for staging. Strong
For upper tract evaluation, use diagnostic ureteroscopy and biopsy only in cases where additional information will impact Strong
treatment decisions.
Use magnetic resonance urography when CT urography is contraindicated for reasons related to contrast administration or Strong
radiation dose.
Use CT or MRI for staging locally advanced or metastatic disease in patients in whom radical treatment is considered. Strong
Use CT to diagnose pulmonary metastases. Computed tomography and MRI are generally equivalent for diagnosing local Strong
disease and distant metastases in the abdomen.
CT = computed tomography; LE = level of evidence; MIBC = muscle-invasive bladder cancer; MRI = magnetic resonance imaging.

Table 5 – Summary of evidence and recommendations for comorbidity scales.

Summary of evidence LE

Chronological age is of limited relevance. 3


A comorbidity score developed in particular for the assessment of patients diagnosed with bladder cancer would be helpful. 3

Recommendations Strength
rating

Base the decision on bladder-sparing treatment or radical cystectomy in elderly/frail patients with invasive bladder cancer Strong
on tumour stage and comorbidity.
Assess comorbidity by a validated score, such as the Charlson Comorbidity Index. The American Society of Anesthesiologists Strong
score should not be used in this setting
LE = level of evidence.

was higher in octogenarians than in septuagenarians (4.3% stage and LN status [34]. In addition, other LN-related
vs 2.3%) [25]. Sarcopenia, on the contrary, was an variables investigated include (but not limited to) the
independent predictor for overall survival (OS) and number of (positive) LNs removed, LN density, and
cancer-specific survival (CSS) in case of radical cystectomy extranodal extension. Although LN density is subject to
(RC) [26]. Other risk factors have been described, which surgical and pathological factors, it appeared to be
means that a thorough (geriatric) multidisciplinary assess- independently associated with OS (hazard ratio [HR]:
ment for comorbidity is important. A population-based 1.45) in a meta-analysis [35]. Other prognostic histopatho-
competing risk analysis showed age to carry the highest risk logical variables are lymphovascular invasion [36] and
for other-cause mortality, while local tumour stage was the concomitant CIS in organ-confined disease [37], both with
strongest predictor for decreased CSS [27]. an independent, approximately 1.5-fold increased risk for
The Charlson Comorbidity Index (CCI) can be used to recurrence and CSM. In addition, tumour location (trigone,
assess comorbidity, since it has been studied widely in bladder neck, and prostatic urethra) has been associated
patients with BC and found to be an independent prognostic with decreased survival [34,37]. The value of the neutro-
factor for perioperative mortality [28,29], overall mortality phil-to-lymphocyte ratio remains controversial [38,39].
[30], and cancer-specific mortality (CSM) [31,32]. The age- Recently, much attention was directed to the molecular
adjusted version of the CCI correlated with both CSM and subtypes of BC based on The Cancer Genome Atlas (TCGA)
other-cause mortality [33]. cohort. A consensus meeting on this subject identified six
MIBC molecular classes that reconcile all previously
7. Markers published classification schemes [40]. The molecular
subgroup classes include luminal papillary, luminal non-
In the 2020 guideline, both prognostic and predictive specified, luminal unstable, stroma rich, basal/squamous,
markers are discussed in one paragraph. and neuroendocrine like, each having distinct differentiation
The most important histopathological prognostic vari- patterns, oncogenic mechanisms, tumour microenviron-
ables after RC and lymph node dissection (LND) are tumour ments, and histological and clinical associations. However,
E U RO P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4 87

Table 6 – Summary of evidence and consensus statements for urothelial markers.

Summary of evidence LE

Currently, treatment decisions cannot be based on molecular markers. 3


EAU-ESMO consensus statements [4,5] a
In patients with metastatic disease, genetic profiling should always be done.
Before prescribing a checkpoint inhibitor, tumour mutation burden does not need to be assessed.
In all fit metastatic patients receiving chemotherapy, established prognostic factors for first- and second-line therapy must be considered when making treatment
decisions (Bajorin for first-line and Bellmunt for second-line therapy).
In all fit metastatic patients receiving chemotherapy, established prognostic factors for first- and second-line therapy should be considered when making
treatment decisions (Bajorin for first-line and Bellmunt for second-line therapy).
Before prescribing checkpoint inhibitor therapy, ribonucleic acid subtypes need not be identified.
Before radical cystectomy or chemotherapy, the neutrophil-to-lymphocyte ratio does not need to be assessed.

EAU = European Association of Urology; ESMO = European Society for Medical Oncology; LE = level of evidence.
a
Only statements that met the a priori consensus threshold across all three stakeholder groups are listed (defined as 70% agreement and 15% disagreement, or
vice versa).

Table 7 – Recommendations and consensus statements for treatment failure of non–muscle-invasive bladder cancer.

Recommendations Strength
rating

Discuss immediate radical treatment (RC) with patients at the highest risk of tumour progression (ie, high grade, Strong
multifocality, carcinoma in situ, and tumour size, as outlined in the EAU guidelines for non–muscle-invasive bladder cancer).
Offer RC to patients with BCG-unresponsive tumours. Strong
Offer patients with BCG-unresponsive tumours, who are not candidates for RC due to comorbidities, preservation strategies Weak
(intravesical chemotherapy, chemotherapy and microwave-induced hyperthermia, electromotive administration of
chemotherapy, and intravesical or systemic immunotherapy, preferably within clinical trials).
EAU-ESMO consensus statements [4,5] a
T1 high-grade bladder urothelial cancer with micropapillary histology (established after complete TURBT and/or re-TURBT) should be treated with immediate RC
and lymphadenectomy.
An important determinant for patient eligibility in case of bladder-preserving treatment is absence of carcinoma in situ.

BCG = bacille Calmette-Guérin; EAU = European Association of Urology; ESMO = European Society for Medical Oncology; RC = radical cystectomy;
TURBT = transurethral resection of bladder tumour.
a
Only statements that met the a priori consensus threshold across all three stakeholder groups are listed (defined as 70% agreement and 15% disagreement, or
vice versa).

currently this classification should still be considered as a in selected (high-risk) patients with NMIBC [21]. RC is also
research tool. Clinical predictive markers for a worse strongly recommended in patients with BCG-refractory
response to neoadjuvant chemotherapy (NAC) are secondary tumours, BCG relapse, and BCG-unresponsive tumours,
MIBC (ie, progression after NMIBC) as compared with which are also defined in the NMIBC guideline.
primary MIBC [41], and possibly variant histologies [42].
Several predictive biomarkers have been studied and 9. Neoadjuvant therapy
reported, but, although promising, they are not used currently
in clinical practice (Table 6). In a single-arm phase 2 study NAC with cisplatin-based combinations has been standard
evaluating neoadjuvant pembrolizumab, tumour mutational for decades in patients with resectable N0M0-invasive BC
burden and PD-L1–positive status were associated with a (Fig. 1 and Table 8). The neoadjuvant approach has potential
higher ratio of pT0 disease at RC [43,44]. However, no advantages (a low micrometastasis burden; chemosensi-
significant correlation between PD-L1 expression and pT0 tivity can be assessed, and a good pathological response
rate was observed in another single-arm phase 2 trial means a better prognosis [47] and better chemo tolerability
evaluating neoadjuvant atezolizumab before RC. before surgery, whereas surgical morbidity is not increased
by chemotherapy [48]) and disadvantages (delay of surgery
8. RC in NMIBC in nonresponders; the decision is based on clinical staging
with the risk of overstaging and overtreatment). Finally,
Although bacille Calmette-Guérin (BCG) is demonstrated to only cisplatin-fit patients are candidates. The most recent
prevent progression, this still occurs in up to 19.3% at 5 yr for meta-analysis on the value of NAC used new and updated
T1G3 tumours treated with maintenance BCG [45], and data of 15 randomised controlled trials (RCTs) [49]. Previ-
progression to MIBC has been shown to decrease signifi- ously published data were confirmed showing an 8%
cantly CSS to 35% after 4 yr (Table 7) [46]. One reason for absolute improvement in 5-yr survival with a number
progression and an unfavourable outcome can be initial needed-to-treat of 12.5. Only cisplatin-based combination
understaging. Therefore, EAU NMIBC guidelines support RC chemotherapy resulted in a meaningful benefit [50]. The
88 E U RO P E A N U RO L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

Fig. 1 – Flow chart for the management of T2-T4a N0M0 urothelial bladder cancer.
CT = computed tomography; MRI = magnetic resonance imaging; UUT = upper urinary tract.
E U RO P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4 89

Table 8 – Summary of evidence and recommendations for neoadjuvant therapy.

Summary of evidence LE

Neoadjuvant cisplatin-containing combination chemotherapy improves OS (8% at 5 yr). 1a


Neoadjuvant treatment of responders and especially patients who show complete response (pT0 N0) has a major impact on 2
OS.
Currently, immunotherapy with checkpoint inhibitors as monotherapy, or in different combinations, is being tested in phase
2 and 3 trials. Initial results are promising.
Still no tools are available to select patients who have a higher probability of benefitting from NAC. In the future, genetic
markers, in a personalised medicine setting, might facilitate the selection of patients for NAC and differentiate responders
from nonresponders.
Neoadjuvant chemotherapy has its limitations regarding patient selection, current development of surgical techniques, and 3
current chemotherapy combinations.
Recommendations Strength
rating

Offer NAC for T2-T4a, cN0M0 bladder cancer. In this case, always use cisplatin-based Strong
combination therapy.
Do not offer NAC to patients who are ineligible for cisplatin-based combination Strong
chemotherapy.
Offer only neoadjuvant immunotherapy to patients within a clinical trial setting. Strong

LE = level of evidence; NAC = neoadjuvant chemotherapy; OS = overall survival.

benefit was for distant metastases only, and not for Neoadjuvant immunotherapy using checkpoint inhi-
locoregional control [51]. bitors (PD-1/L1, cytotoxic T-lymphocyte associated pro-
Gemcitabine and cisplatin (GemCis), frequently used in tein 4 [CTLA-4]) is currently tested as immunotherapy
metastatic disease, has not been used in RCTs for NAC, alone or in combination with chemotherapy in several
although pT0/pT1 rates seem to be similar to those achieved on-going phase 3 trials. Research in the neoadjuvant
with methotrexate, vinblastine, adriamycin, and cisplatin setting is done either as monotherapy or in combination
(MVAC) [50]. Dose dense (dd) MVAC demonstrated high with chemotherapy or CTLA-4 checkpoint inhibition.
rates of pathological response, but data are limited [52]. A Updated results of a single-arm phase 2 trial evaluating
phase 2 study with dd GemCis showed high pathological PD-1 inhibitor pembrolizumab have been published with
response rates (<pT2, 45–57%) but with increased vascular a pT0 rate of 37% and pathological response (<pT2) in 55%
toxicity [53]. In case of variant histology, NAC seems to be of patients [43,44].
beneficial in diffuse/plasmacytoid/signet ring cell, mixed
variants, and neuroendocrine/small cell/large cell [18].
While randomised prospective clinical trial data dem- 10. Perioperative radiotherapy
onstrate a clinically relevant OS benefit for NAC, retrospec-
tive data are less clear. A recent retrospective analysis based Data on perioperative radiotherapy are retrospective and
on data from the US National Cancer Database showed that old, not reflecting modern radiotherapy techniques with
only 19% of all patients received NAC before RC [54]. They concurrent chemotherapy (Table 9). Available data suggest
found no clear survival advantage despite propensity score downstaging of the tumour due to preoperative radiother-
adjustment to include patients based on South West apy and longer progression-free survival (PFS), but no
Oncology Group (SWOG) 8710 study criteria [55]. Limita- impact on survival [56].
tions are the retrospective design and the lack of informa- There are also no solid data to defend postoperative
tion on the type of NAC used. radiotherapy, although in locally advanced BC (T3-T4, N0/
Response to NAC cannot be predicted by imaging or N1, M0), the local recurrence rate was reported to decrease
markers yet. with postoperative RT [57].

Table 9 – Summary of evidence and recommendations for pre- and postoperative radiotherapy.

Summary of evidence LE

No data exist to support that preoperative RT for operable MIBC increases survival. 2a
Preoperative RT for operable MIBC, using a dose of 45–50 Gy in fractions of 1.8–2 Gy, results in downstaging after 4–6 wk. 2
Limited high-quality evidence supports the use of preoperative RT to decrease local recurrence of MIBC after RC. 3

Recommendations Strength
rating

Do not offer preoperative RT for operable MIBC since it will only result in downstaging, but will not improve survival. Strong
Do not offer preoperative RT when subsequent RC with urinary diversion is planned. Strong

LE = level of evidence; MIBC = muscle-invasive bladder cancer; RC = radical cystectomy; RT = radiotherapy.


90 E U RO P E A N U RO L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

Table 10 – Recommendations for unresectable tumours.

Recommendations Strength rating

Offer radical cystectomy as a palliative treatment to patients with inoperable locally advanced tumours (T4b). Weak
Offer palliative cystectomy to patients with symptoms. Weak

Table 11 – Summary of evidence and recommendations for sexual function–preserving techniques.

Summary of evidence LE

The majority of patients motivated to preserve their sexual function will benefit from sexual function–preserving 2a
techniques.
None of the sexual function–preserving techniques (prostate/capsule/seminal/nerve sparing) have shown to be superior, 3
and no particular technique can be recommended.
Data regarding pelvic organ–preserving radical cystectomy for female patients remain immature. 3

Recommendations Strength
rating

Do not offer sexual function–preserving cystectomy to men as standard therapy for MIBC. Strong
Offer sexual-preserving techniques to men motivated to preserve their sexual function since the majority will benefit. Strong
Select men based on: Strong
 Organ-confined disease
 Absence of any kind of tumour at the level of the prostate, prostatic urethra, or bladder neck
Do not offer pelvic organ–preserving radical cystectomy to women as standard therapy for MIBC. Strong
Offer sexual function–preserving techniques to women motivated to preserve their sexual function since the majority will benefit. Weak
Select women based on: Strong
 Organ-confined disease
 Absence of tumour in bladder neck or urethra

LE = level of evidence; MIBC = muscle-invasive bladder cancer.

11. Radical surgery: introduction quicker bowel recovery compared with patients receiving
placebo, but this drug is, as yet, not approved in Europe
RC is recommended for patients with MIBC stage T2-T4a, [63]. Venous thromboembolism (VTE) prophylaxis may be
N0-Nx, M0, high-risk and unresponsive NMIBC, and implemented as part of an ERAS protocol. Of note, a recent
tumours that cannot be controlled with TUR of the bladder study indicated that patients after NAC had higher VTE rates
(TURB) and intravesical therapy alone (Table 10). Salvage (12% vs 8%; p = 0.002) [64].
cystectomy is indicated in nonresponders to bladder-
sparing treatment and for palliative reasons such as fistula 13. RC: technique and extent
formation, pain, and recurrent visible haematuria.
Studies on the impact of delayed RC on survival In men, standard RC includes removal of the bladder,
outcomes are conflicting. Some studies have suggested a prostate, seminal vesicles, distal ureters, and regional LNs.
delay of 3 mo to be associated with worse survival outcomes In women, standard RC includes removal of the bladder,
[58], whereas others have found this to be safe [59,60]. A entire urethra and adjacent vagina, uterus, distal ureters,
study from the UK suggested that patients with a T2 tumour and regional LNs. Recently, the EAU MMIBC Guideline Panel
could profit from earlier cystectomy [60]. undertook two systematic reviews addressing sexual
function–preserving cystectomy in men and women to
12. Radical surgery: preparation determine functional and oncological outcomes (Table 11)
[65,66].
Bowel preparation is not considered necessary anymore, In men, four sparing techniques have been described:
instead the “fast track” early recovery after surgery (ERAS) prostate and seminal vesicle sparing, prostate capsule
protocol stimulates emotional and physical functioning sparing (after TUR of the prostate or en bloc removal of
scores and bowel recovery with, amongst others, early the adenoma with the bladder), seminal sparing, and nerve-
mobilisation and oralisation, and gastrointestinal stimula- sparing cystectomy. The review included 12 studies with
tion with metoclopramide and chewing gum [61]. Another predominantly open cystectomy and neobladder as diver-
cornerstone of the ERAS protocol is postoperative pain sion. Tumour in the bladder neck, prostatic urethra, and
management, which involves significantly reducing the use prostate were ruled out, except PCa in nerve-sparing
of opioids [62]. Alvimopan has been shown to induce cystectomy. Oncological outcomes were similar to control
E U RO P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4 91

Table 12 – Summary of evidence and recommendations for laparoscopic/robotic-assisted laparoscopic cystectomy.

Summary of evidence LE

RARC has longer operative time (1–1.5 h) and major costs, but shorter length of hospital stay (1–1.5 d) and less blood loss compared with ORC. 1
Retrospective RARC series suffer from a significant stage selection bias as compared with ORC. 1
Grade 3, 90-d complication rate is lower with RARC. 2
Most endpoints, if reported, including intermediate-term oncological endpoint and quality of life, are not different between RARC and ORC. 2
Surgeons experience and institutional volume are considered the key factors for outcome of both RARC and ORC, and not the technique. 2
Recommendations on how to define challenging patients and an experienced RARC surgeon are still under discussion. 3
The use of neobladder after RARC still seems underutilised, and functional results of intracorporeally constructed neobladders should be studied. 4

Recommendations Strength rating

Inform the patient of the advantages and disadvantages of ORC and RARC to allow selection of the proper procedure. Strong
Select experienced centres, not specific techniques, both for RARC and ORC. Strong

LE = level of evidence; ORC = open radical cystectomy; RARC = robotic-assisted radical cystectomy.

groups and within groups with regard to local or metastatic complications, and QoL [71]. The transfusion rate was likely
recurrence, disease-specific survival (DSS), and OS, at a lower after RARC. For other endpoints, outcomes were
median follow-up of 3–5 yr. In case the prostate was spared, uncertain due to study limitations.
the rates of incidental PCa in the intervention group ranged For both techniques, surgeons’ experience and institu-
from 0% to 15%, and no International Society of Urologic tional volume strongly predicted outcome, with 30 cases
Pathologists (ISUP) grade 4 was reported. Postoperative estimated to be enough to achieve proficiency in RARC
potency was significantly better for all techniques as [70]. Oncological results also seem comparable [72],
compared with conventional RC (p < 0.05), without superi- although port-site metastasis (in addition to other meta-
ority of any technique. No major impact was shown with static sites) after RARC was suggested to be under-reported
regard to continence rates for any of the three approaches. [73]. The largest RCT to date is the RAZOR trial [74]. This
However, the overall quality of the evidence was moderate; study showed RARC to be noninferior to open RC in terms of
hence, candidates must be carefully selected, counselled, 2-yr PFS (72.3% vs 71.6%), adverse events (67% vs 69%), and
and closely monitored. QoL. Most series offer extracorporeal reconstruction, which
In women, to prevent pelvic floor disorders (such as is associated with a longer operative time, more blood
prolapse) and sexual dysfunction, a gynaecological history transfusions, but fewer high-grade complications than an
should be obtained and pelvic organ-preserving techniques intracorporeal reconstruction [75]. For laparoscopic RC, a
can be applied, preserving the neurovascular bundle, review came to similar conclusions as described for RARC
vagina, uterus, and/or ovaries. From an oncological point [76].
of view, concomitant malignancy in gynaecological organs
is rare and local recurrences reported after RC are infrequent 15. Lymphadenectomy: role and extent
[67]. In case of an increased risk of hereditary breast or
ovarian cancer, salpingo-oophorectomy should be advised Regional LNs have been shown to consist of all pelvic LNs
[68]. The conclusion of the review, based on retrospective, below the bifurcation of the aorta [77,78]. Mapping studies
low-quality data, was that for well-selected patients, also found that skipping lesions at locations above the
sparing female reproductive organs during RC appears to bifurcation of the aorta, without more distally located LN
be oncologically safe and provides improved functional metastases, is rare [78].
outcomes [66]. The optimal extent of LND has not been established to
date. Standard LND includes nodes up to the common iliac
14. Laparoscopic/robotic-assisted laparoscopic bifurcation, extended LND goes up to the aortic bifurcation
cystectomy including presacral nodes, and a superextended LND
extends cranially to the level of the inferior mesenteric
Most of the available literature on minimal invasive artery. The impact on oncological outcome of the extent of
cystectomy is retrospective with limited numbers. Earlier LND was reviewed [79]. Although the quality of data was
reviews of this literature with a low level of evidence have poor, any LND was better than no LND and a (super)
learned that robotic-assisted laparoscopic RC (RARC), extended was better than a standard LND, but no difference
compared with open RC, has a longer operative time, in outcome was reported between extended and super-
increased costs, less blood loss, a 1–1.5-d shorter hospital extended LND. In a recent prospective phase 3 RCT,
stay, and a lower 90-d complication rate (Table 12) however, an extended LND failed to show a significant
[69,70]. A recent Cochrane review including data from all advantage [80]. In conclusion, an extended LND might have
five published RCTs, however, showed no difference a therapeutic benefit, and if this is the case, it remains to be
between RARC and open RC with regard to the time to seen whether the counted number of removed nodes or the
recurrence, positive surgical margin rate, grade 3–5 template is the most important prognosticator.
92 E U RO P E A N U RO L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

16. Urinary diversion after RC therefore a good option in older and frail patients [87]. QoL
showed equal urinary bother and function to an ileal
Most of the complications after RC and diversion are related conduit [86]. Connection to the skin can be done separately
to the use of bowel for the urinary diversion (Table 13) or with one stoma (after ureteroureteral anastomosis).
[81]. Three alternatives with different segments of the Although the ileal conduit is still the most performed and
intestinal tract are currently used after cystectomy: a well-established option with predictable results, it is not
continent or incontinent abdominal diversion, a urethral without complications such as UTIs, ureteroileal leakage
diversion, and a rectosigmoid diversion. An ileal conduit or and stenosis, stones, and stomal problems [88]. Complica-
an orthotopic neobladder is mostly used. In case a tions increase in frequency with longer follow-up to 94% in
neobladder is planned with the presence of CIS in the those surviving >15 yr [89].
prostatic urethra or bladder neck (females), frozen sections A continent cutaneous urinary diversion can be made
have to be performed, and patients have to undergo regular from different bowel parts. Although it is not done
follow-up cystoscopy and urinary cytology [82]. Positive frequently, patients have been reported to have a well-
LNs do not exclude an orthotopic neobladder. Secondary functioning reservoir with day- and night-time continence
urethral tumours seem to be less common after a approaching 93%, with 15% stomal stenosis and 10% stone
neobladder compared with cutaneous diversions [83]. Cer- formation [90].
tainly, for continent diversions, patients need to be An orthotopic bladder substitution is a safe and reliable
motivated, well informed, and selected. For a neobladder, option in both men and women. Long-term complications
there is no strict age criterion, and in case of N1 disease, it include diurnal and nocturnal incontinence, ureterointest-
still is feasible [84]. Although there are no RCTs comparing inal stenosis, metabolic disorders, and vitamin B12 defi-
different diversions, retrospective data suggest that a ciency. The oncological outcome is not influenced by the
decline in estimated glomerular filtration rate did not differ choice of diversion, although urethral recurrences in
significantly after ileal conduit versus neobladder in neobladder patients seem to be rare [91]. Various forms
patients with preoperative chronic kidney disease [85]. of upper urinary tract reflux protection have been
Ureterocutaneostomy is the simplest form of cutaneous described, without long-term differences in outcome.
diversion with lower operative time, complication rate, and In all, it is not possible to recommend a particular type of
length of hospital stay than ileal conduit [86], and is urinary diversion. However, mostly performed are ileal

Table 13 – Summary of evidence and recommendations for radical cystectomy and urinary diversion.

Summary of evidence LE

For MIBC, RC is the curative treatment of choice. 3


Higher hospital volume likely improves quality of care and reduction in perioperative mortality and morbidity. 3
Radical cystectomy includes removal of regional lymph nodes. 3
There are data to support that extended LND (vs standard or limited LND) improves survival after RC. 3
Radical cystectomy in both sexes must not include removal of the entire urethra in all cases, which may then serve as the outlet for an 3
orthotopic bladder substitution. The terminal ileum and colon are the intestinal segments of choice for urinary diversion.
The type of urinary diversion does not affect oncological outcome. 3
Laparoscopic cystectomy and robotic-assisted laparoscopic cystectomy are feasible but still investigational. Current best practice is open 3
RC.
The use of extended prophylaxis significantly decreases the incidence of venous thromboembolism after RC. 3
In patients aged >80 yr with MIBC, cystectomy is an option. 3
Surgical outcome is influenced by comorbidity, age, previous treatment for bladder cancer or other pelvic diseases, surgeon and hospital 2
volumes of cystectomy, and type of urinary diversion.
Surgical complications of cystectomy and urinary diversion should be reported using a uniform grading system. Currently, the best- 2
adapted grading system for cystectomy is the Clavien grading system.
No conclusive evidence exists as to the optimal extent of LND. 2a

Recommendations Strength rating

Do not delay RC for >3 mo, as it increases the risk of progression and cancer-specific mortality. Strong
Perform at least 10, and preferably >20, RCs per hospital per year. Strong
Before RC, fully inform the patient about the benefits and potential risks of all possible alternatives. The final decision should be based Strong
on a balanced discussion between the patient and the surgeon.
Do not offer an orthotopic bladder substitute diversion to patients who have a tumour in the urethra or at the level of urethral Strong
dissection.
Preoperative bowel preparation is not mandatory. “Fast track” measurements may reduce the time to bowel recovery. Strong
Offer pharmacological prophylaxis, such as low-molecular-weight heparin to RC patients, starting the 1st day after surgery, for a period Strong
of 4 wk.
Offer RC in T2-T4a, N0M0, and high-risk non–muscle-invasive BC. Strong
Perform a lymph node dissection as an integral part of RC. Strong
Do not preserve the urethra if margins are positive. Strong

BC = bladder cancer; LE = level of evidence; LND = lymph node dissection; MIBC = muscle-invasive bladder cancer; RC = radical cystectomy.
E U RO P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4 93

Table 14 – Recommendations and consensus statements for supportive care.

Recommendations Strength rating

Offer radical cystectomy as a palliative treatment to patients with inoperable locally advanced tumours (T4b). Weak
Offer palliative cystectomy to patients with symptoms. Weak
EAU-ESMO consensus statements [4,5] a
Chemoradiation should be given to improve local control in cases of inoperable locally advanced tumours.
In patients with clinical T4 or clinical N + disease (regional), radical chemoradiation can be offered accepting that this may be palliative rather than curative in
outcome.

EAU = European Association of Urology; ESMO = European Society for Medical Oncology.
a
Only statements that met the a priori consensus threshold across all three stakeholder groups are listed (defined as 70% agreement and 15% disagreement, or
vice versa).

orthotopic neobladders and ileal conduits. In selected lower quality, the available evidence suggests that perform-
patients, such as patients with a single kidney, ureterocu- ing >10 RCs per year per hospital reduces 30- and 90-d
taneostomy is an option to consider. mortality. Performing >20 RCs per hospital per year might
even reduce these mortality rates further.
17. Morbidity, survival, and impact of hospital and
surgeon volume on outcome 18. Supportive care

In a large single-centre series, early complications (within In the case of on-going visible haematuria, coagulation
3 mo of surgery) were seen in 58% of patients [81]. Late disorders and anticoagulant drugs must be reviewed
morbidity is usually linked to the type of urinary diversion (Table 14). Intravesical rinsing of the bladder with 1% silver
(see also above). Large studies have shown 5-yr recurrence- nitrate or 1–2% alum can be effective [96]. Instillation of
free survival (RFS) rate to be around 60% and CSS around formalin (2.5–4% for 30 min) is a more effective, but painful,
65% [92,93]. RFS is dependent on nodal involvement and and usually requiring anaesthesia and with a higher risk of,
stage. In a surgery-only study, the 5-yr RFS was 76% in for example, bladder fibrosis [96]. Vesicoureteral reflux
patients with pT1 tumours, 74% in patients with pT2 should be excluded to prevent renal complications. Radia-
tumours, 52% in patients with pT3 tumours, and 36% in tion therapy is another common strategy to control bleeding
patients with pT4 tumours [94]. and pain. Embolisation of specific arteries in the small pelvis
A recent systemic review addressed the impact of has been reported with success rates as high as 90% [96].
hospital and/or surgeon volume on perioperative outcomes
of RC [95]. The results suggest that a higher hospital volume 19. Bladder-sparing treatments for localised disease
is likely associated with lower in-hospital, 30-d, and 90-d
mortality rates. In addition, higher-volume hospitals are Although a thorough TUR is also important in MIBC, the TUR
likely to have lower positive surgical margins, higher LND alone in patients with MIBC can only be a therapeutic option
and neobladder rates, and lower complication rates. For in T2 tumours with a negative restaging TUR. One study
surgeon volume, less evidence is available. In spite of the reported a 77% 15-yr CSS with this approach [97].

Table 15 – Summary of evidence, recommendations, and consensus statements for bladder-sparing treatments.

Summary of evidence LE

External beam radiotherapy alone should be considered only as a therapeutic option when the patient is unfit for cystectomy or as part 3
of a multimodality bladder-preserving approach.
Radiotherapy can also be used to stop bleeding from the tumour when local control cannot be achieved by transurethral manipulation 3
because of extensive local tumour growth.
Complete and partial local responses have been reported with cisplatin-based chemotherapy as primary therapy for locally advanced 2b
tumours in highly selected patients.
Recommendation Strength rating

Do not offer transurethral resection of bladder tumour alone as a curative treatment option as most patients will not benefit. Strong
Do not offer radiotherapy alone as primary therapy for localised bladder cancer. Strong
Do not offer chemotherapy alone as primary therapy for localised bladder cancer. Strong
a
EAU-ESMO consensus statements [4,5]
Radiotherapy alone (single block) is not the preferred radiotherapeutic schedule.
Radiotherapy for bladder preservation should be performed with IMRT and IGRT to reduce side effects.
Dose escalation above standard radical doses to the primary site in case of bladder preservation, either by IMRT or brachytherapy, is not recommended.

EAU = European Association of Urology; ESMO = European Society for Medical Oncology; IGRT = image-guided radiotherapy; IMRT = intensity-modulated
radiotherapy; LE = level of evidence.
a
Only statements that met the a priori consensus threshold across all three stakeholder groups are listed (defined as 70% agreement and 15% disagreement, or
vice versa).
94 E U RO P E A N U RO L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

Current EBRT techniques with soft-tissue matching [4,5,9]. Similar results have been described in a small series
result in superior bladder coverage and a reduced integral of variant histologies [104]. OS was lower than with RC in
dose to the surrounding tissues with a target dose for two recent large retrospective analyses [105,106]. After
curation of 64–66 Gy [98] (Table 15). A complete initial TUR propensity score matching, both analyses show lower
is an important prognostic factor. Late morbidity is low and survival after 2 yr of follow-up, with a significant hazard
reduced even more with IMRT [99]. Still, EBRT alone was ratio (HR) of 1.4–1.5, although one study [105] found lower
shown to be significantly less effective (2-yr OS 42%) than 1–7-yr mortality for MMT (HR: 0.84, 95% confidence
concurrent chemoradiotherapy (2-yr OS 56%, p < 0.001, see interval [CI]: 0.74–0.96). On the contrary, a large systematic
below) in a review of patients treated between 2004 and review found similar OS and DSS for RC and MMT
2013 [100]. [107]. Obvious prognostic factors for MMT are comorbidity,
A recent large retrospective study reported on TURB and age, stage, and downstaging/complete response (CR) after
multiagent chemotherapy [101]. The 2- and 5-yr OS values MMT. Noninvasive recurrences after MMT are found in
were 49% and 32.9% for all patients, and 52.6% and 36.2% for around 25% of patients who can be treated conservatively
cT2 patients, respectively. While these data show that long- [108]. In case of salvage cystectomy, major complication
term survival with intact bladder can be achieved in a rates are not increased. In all, bladder preservation with
subset of patients, it is not recommended for routine use. MMT leads to acceptable outcomes and may be considered a
reasonable treatment option in well-selected patients as
20. Multimodality bladder-preserving treatment compared with RC [103]. There are no definitive data
supporting the benefit of using neoadjuvant or adjuvant
Multimodality treatment (MMT) or trimodality treatment chemotherapy in case of MMT [103]. Whether a node
combines TUR of bladder tumour, radiosensitising chemo- dissection should be performed before MMT, as in RC, also
therapy, and RT (1) with the aim of bladder preservation remains unclear [4,5]. Finally, a bladder-preserving multi-
without compromising oncological outcome in patients fit modality strategy requires very close multidisciplinary
for cystectomy or (2) as an effective alternative for patients cooperation and a high level of patient compliance
unfit or unwilling for cystectomy (Table 16). For optimal (follow-up) [4,5].
results, the TUR/re-TUR should be as radical as possible,
suggesting proper patient selection (T2 tumours, no CIS) 21. Adjuvant therapy
[102]. The different principles of radiation (dose, field, and
schedule) in case of MMT have been described [103]. Used An updated meta-analysis from 2014 with the results of
chemotherapy regimens are cisplatin, mitomycin C plus 5- nine trials with different cisplatin-based combinations and
fluorouracil, gemcitabine, and nicotinamide/carbogen, courses, underpowered, and with significant heterogeneity
without clear preference for a specific radiosensitiser [4,5]. and methodological flaws showed that there remains a very
Five-year CSS and OS rates vary between 50–82% and 36– low level of evidence supporting the use of adjuvant
74%, respectively, with salvage cystectomy rates of 10–30% chemotherapy (Table 17) [109]. Still, the HR for OS was 0.77

Table 16 – Summary of evidence, recommendations, and consensus statements for multimodality treatment.

Summary of evidence LE

In a highly selected patient population, long-term survival rates of multimodality treatment are comparable with those of early 2b
cystectomy.
Recommendations Strength rating
Offer surgical intervention or MMT as a primary curative therapeutic approach since they are more effective than radiotherapy alone. Strong
Offer MMT as an alternative to selected, well-informed, and compliant patients, especially for whom radical cystectomy is not an option. Strong
EAU-ESMO consensus statements [4,5] a
Candidates for curative treatment, such as cystectomy or bladder preservation, should be assessed clinically by at least an oncologist, a urologist, and a neutral
HCP such as a specialist nurse.
An important determinant for patient eligibility in case of bladder-preserving treatment is absence of carcinoma in situ.
An important determinant for patient eligibility in case of bladder preserving treatment is absence or presence of hydronephrosis.
When assessing patient eligibility for bladder preservation, the likelihood of successful debulking surgery should be taken into consideration (optimal debulking).
Bladder urothelial carcinoma with small cell neuroendocrine variant should be treated with neoadjuvant chemotherapy followed by consolidating local therapy.
In case of bladder preservation with radiotherapy, combination with a radiosensitiser is always recommended to improve clinical outcomes, such as cisplatin,
5FU/MMC, carbogen/nicotinamide, or gemcitabine.
Radiotherapy for bladder preservation should be performed with IMRT and IGRT to reduce side effects.
Dose escalation above standard radical doses to the primary site in case of bladder preservation, either by IMRT or by brachytherapy, is not recommended.
In patients with clinical T4 or clinical N+ disease (regional), radical chemoradiation can be offered accepting that this may be palliative rather than curative in
outcome.

EAU = European Association of Urology; ESMO = European Society for Medical Oncology; 5FU = 5-fluorouracil; HCP = healthcare professional; IGRT = image-
guided radiotherapy; IMRT = intensity-modulated radiotherapy; LE = level of evidence; MMT = multimodality treatment.
a
Only statements that met the a priori consensus threshold across all three stakeholder groups are listed (defined as 70% agreement and 15% disagreement, or
vice versa).
E U RO P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4 95

Table 17 – Recommendations and consensus statements for adjuvant therapy.

Recommendations Strength rating

Offer adjuvant cisplatin-based combination chemotherapy to patients with pT3/4 and/or pN + disease if no Strong
neoadjuvant chemotherapy has been given.
Only offer immunotherapy with a checkpoint inhibitor in a clinical trial setting. Strong
EAU-ESMO consensus statements [4,5] a
When adjuvant chemotherapy is offered, patients should be selected based on the result of LND (if done).

EAU = European Association of Urology; ESMO = European Society for Medical Oncology; LND = lymph node dissection.
a
Only statements that met the a priori consensus threshold across all three stakeholder groups are listed (defined as 70% agreement and 15% disagreement, or
vice versa).

(95% CI: 0.59–0.99) and for DFS 0.66 (95% CI: 0.45–0.91), neuropathy, and New York Heart Association (NYHA)
with lower DFS in case of nodal positivity. A recent class III heart failure [118], which comprises over 50% of
publication of the largest RCT (European Organisation for patients with BC. Cisplatin has also been administered in
Research and Treatment of Cancer, EORTC 30994), although small studies to patients with a lower GFR (40–60 ml/
not fully accrued, also showed a significant improvement of min) using different split-dose schedules, in part because
PFS for immediate, compared with deferred, cisplatin-based carboplatin is considered less effective than cisplatin
chemotherapy (HR: 0.54; 95% CI: 0.4–0.73), but no [116]. Gemcitabine and carboplatin (GemCarbo) has long
significant OS benefit [110]. Two other large retrospective been the standard systemic treatment for cisplatin-unfit
database analyses also found a benefit for adjuvant patients based on a phase 2/3 RCT by the EORTC where
chemotherapy versus observation, especially in T3/4 or N GemCarbo was more effective (overall response rate
+ disease [111,112]. In patients with concomitant or pure [ORR] 42% vs 30%) with less toxicity than methotrexate/
variant histology, however, no benefit was found. carboplatin/vinblastine (Fig. 2) [119].
For adjuvant immunotherapy, to date, no data have been The role of first-line immunotherapy is being studied,
published. but currently published data from only two single-arm
phase 2 monotherapy trials in cisplatin-ineligible patients
22. Metastatic disease, first-line systemic therapy are available. Pembrolizumab showed an ORR of 29% and a
CR in 7% of patients [120]. Atezolizumab showed an ORR of
Approximately 50% of patients with MIBC relapse after RC, 23%, a CR in 9%, and median OS of 15.9 mo [121]. The results
depending on the stage. Most relapses are with distant are difficult to interpret due to the missing control arm and
metastases. Standard first-line treatment for fit patients the heterogeneity of the study population with regard to
with a good renal function is cisplatin-based combination PD-L1 status. The toxicity profile, however, was favourable
chemotherapy (Table 18). Most used regimens are GemCis for both drugs. Both drugs are approved by the FDA and
or MVAC or ddMVAC demonstrating median OS of 12–14 mo European Medicines Agency for first-line treatment in
[113]. Neither of the combinations is superior to the other, cisplatin-ineligible patients, but only in case of a positive
but equivalence has not been tested. The long-term survival PD-L1 status.
results have confirmed the efficacy of the two regimens,
with GemCis having less toxicity [114], although tolerability 23. Second-line systemic therapy for metastatic
to MVAC can be improved with granulocyte colony- disease
stimulating factor (G-CSF). High-dose-intensity MVAC
combined with G-CSF is less toxic and has demonstrated The only chemotherapy drug ever approved as second-line
a higher CR rate and better 2-yr survival than standard treatment in Europe was vinflunine, based on a phase 3 RCT,
MVAC in one trial; however, there is no significant showing a modest ORR (8.6%), a clinical benefit with a
difference in median survival between the two regimens favourable safety profile, and a survival benefit in favour of
[115]. Substitution of carboplatin for cisplatin in cisplatin-fit vinflunine, which was, however, only statistically signifi-
patients is associated with inferior outcome and is clearly cant in the per-protocol patient population (not in the
discouraged [116]. intention-to-treat [ITT] population) [122]. A reasonable
Karnofsky performance status (PS) of 80% and presence strategy has been to rechallenge former cisplatin-sensitive
of visceral metastases are independent prognostic factors of patients if progression occurred at least 6–12 mo after first-
poor survival for first-line treatment with MVAC [117]. The line cisplatin-based combination chemotherapy. Currently,
sites of metastases also have an impact on long-term vinflunine is reserved for patients with contraindications to
survival. In LN-only disease, 20.9% of patients were alive at immunotherapy or as third- or later-line treatment. A recent
5 yr compared with only 6.8% of patients with visceral phase 3 RCT evaluated the addition of the angiogenesis
metastases [114]. inhibitor ramucirumab to docetaxel versus docetaxel alone
Cisplatin unfit is defined by at least one of the [123]. The primary endpoint of PFS prolongation was
following criteria: PS > 1, glomerular filtration rate reached, the clinical benefit appears small, and OS data
(GFR) 60 ml/min, grade 2 audiometric loss, peripheral have not yet been reported.
96 E U RO P E A N U RO L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

Table 18 – Summary of evidence, recommendations, and consensus statements for metastatic disease.

Summary of evidence LE

In a first-line setting, PS and the presence or absence of visceral metastases are independent prognostic factors for survival. 1b
In a second-line setting, negative prognostic factors are liver metastasis, PS  1, and low haemoglobin (<10 g/dl). 1b
Cisplatin-containing combination chemotherapy can achieve median survival of up to 14 mo, with long-term DFS reported in 15% of 1b
patients with nodal disease and good PS.
Single-agent chemotherapy provides low response rates of usually short duration. 2a
Carboplatin combination chemotherapy is less effective than cisplatin-based chemotherapy in terms of complete response and survival. 2a
Nonplatinum combination chemotherapy produces substantial responses in first- and second-line settings. 2a
Nonplatinum combination chemotherapy has not been tested against standard chemotherapy in patients who are fit or unfit for 4
cisplatin combination chemotherapy.
There is no defined standard chemotherapy for unfit patients with advanced or metastatic UC. 2b
Postchemotherapy surgery after partial or complete response may contribute to long-term DFS in selected patients. 3
Zoledronic acid and denosumab have been approved for supportive treatment in case of bone metastases of all cancer types including 1b
UC, because they reduce and delay skeletal-related events.
PD-1 inhibitor pembrolizumab has been approved for patients who have progressed during or after previous platinum-based 1b
chemotherapy based on the results of a phase 3 trial.
PD-L1 inhibitor atezolizumab has been approved by the FDA for patients who have progressed during or after previous platinum-based 2a
chemotherapy based on the results of a phase 2 trial.
PD-1 inhibitor nivolumab has been approved for patients who have progressed during or after previous platinum-based chemotherapy 2a
based on the results of a phase 2 trial.
PD-1 inhibitor pembrolizumab has been approved for patients with advanced or metastatic urothelial cancer ineligible for cisplatin- 2a
based first-line chemotherapy based on the results of a phase 2 trial but use of pembrolizumab is restricted to PD-L1–positive patients.
PD-L1 inhibitor atezolizumab has been approved for patients with advanced or metastatic urothelial cancer ineligible for cisplatin- 2a
based first-line chemotherapy based on the results of a phase 2 trial, but the use of atezolizumab is restricted to PD-L1–positive patients.
Recommendations Strength rating

First-line treatment for cisplatin-eligible patients


Use cisplatin-containing combination chemotherapy with GC, MVAC, preferably with G-CSF, HD-MVAC with G-CSF or PCG. Strong
Do not offer carboplatin and nonplatinum combination chemotherapy. Strong
First-line treatment in patients ineligible (unfit) for cisplatin
Offer checkpoint inhibitors such as pembrolizumab or atezolizumab depending on PD-L1–positive status. Strong
Offer carboplatin combination chemotherapy if PD-L1 is negative. Strong
Second-line treatment
Offer the checkpoint inhibitor pembrolizumab to patients progressing during, or after, platinum-based combination chemotherapy for Strong
metastatic disease. Alternatively, offer treatment within a clinical trial setting.
Offer zoledronic acid or denosumab for supportive treatment in case of bone metastases. Weak
Offer only vinflunine to patients for metastatic disease as subsequent-line treatment if immunotherapy or combination chemotherapy, Weak
or FGFR3-inhibitor therapy, or inclusion in a clinical trial is not feasible.
EAU-ESMO consensus statements [4,5] a
Pseudoprogression has not been demonstrated in urothelial cancer.
In patients with advanced/metastatic urothelial cancer who are ineligible for cisplatin-based therapy but with high PD-L1 expression (as per approved drug-
specific methodology), both treatment with an ICI and chemotherapy can be offered.
Since no data exist for cisplatin-ineligible PD-L1–positive patients in order to differentiate between different ICIs (atezolizumab and pembrolizumab), either
agent can be administered.
Enrolment in a clinical trial remains the preferred option for patients with cisplatin-eligible advanced/metastatic urothelial cancer until on-going randomised
trials report in this population.
Treatment with an ICI should be offered to patients with advanced/metastatic urothelial cancer with progression after platinum-based chemotherapy. This
includes tumours that have progressed within a year or following perioperative (cystectomy) chemotherapy.
Once initiated, ICI therapy should be continued until progression of disease in patients with advanced/metastatic urothelial cancer.
In contrast to the first-line setting, the PD-L1 biomarker is not useful for selecting patients for immunotherapy in platinum-refractory metastatic urothelial
cancer.
Carboplatin-based chemotherapy remains a viable first-line treatment option in cisplatin-ineligible, PD-L1–positive patients with metastatic urothelial carcinoma
until data from randomised phase 3 trials of ICIs are available.
Cisplatin-ineligible, immunotherapy-refractory patients with metastatic urothelial carcinoma should be considered for chemotherapy instead of sequencing of
immunotherapy.

DFS = disease-free survival; EAU = European Association of Urology; ESMO = European Society for Medical Oncology; FDA = Food and Drug Administration;
FGFR = fibroblast growth factor receptor; GC = gemcitabine plus cisplatin; G-CSF = granulocyte colony-stimulating factor; HD = high dose; ICI = immune
checkpoint inhibitor; LE = level of evidence; MVAC = methotrexate, vinblastine, adriamycin, and cisplatin; PCG = paclitaxel, cisplatin, gemcitabine; PD-
L1 = programmed death-ligand 1; PS = performance status; UC = urothelial cancer.
a
Only statements that met the a priori consensus threshold across all three stakeholder groups) are listed (defined as 70% agreement and 15% disagreement,
or vice versa).

Most recently, second-line immunotherapy with PD-1/ mab have demonstrated similar efficacy and safety in
PD-L1 checkpoint inhibitors has been established and is still second-line phase 1–3 trials.
being studied as standard second-line therapy. Pembroli- A phase 3 RCT with pembrolizumab, a PD-1 inhibitor,
zumab, nivolumab, atezolizumab, avelumab, and durvalu- showed significant OS benefit over chemotherapy (pacli-
E U RO P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4 97

Paent characteriscs
PS 0-1/ 2/ >2
GFR ≥/< 60 mL/min
Comorbidies

FIT FOR CISPLATIN?


YES NO NO

PS 0-1 and PS 2 or PS ≥ 2 and


GFR ≥ 60 mL/min GFR < 60 mL/min GFR < 60 mL/min
1. Carboplan and
STANDARD GC gemcitabine Monotherapy with either:
MVAC 2. Pembrolizumab 1. Pembrolizumab
HD MVAC (if PD-L1 posive) (if PD-L1 posive)
PCG 3. Atezolizumab 2. Atezolizumab
(if PD-L1 posive) (if PD-L1 posive)
4. Alternate regimens Or
5. Trials or best Best supporve care
supporve care

Second-line treatment
independent of teh me of progression aer first-line treatment

PS 0-1 PS ≥ 2

Progression (independent of the me 1. Consider immunotheraphy


interval aer receiving first-linechemo- 2. Clinical trial
therapy), adequate renal funcon 3. Best supporve care

Standard regimens
1. Pembrolizumab
2. Atezolizumab
3. Nivolumab
Or
1. Clinical trial
2. Combinaon chemotheraphy
3. Monotherapies

Subsequent treatment
1. Chemotherapy
2. Immunotherapy, if not given as
second-line treatment
3. Clinical trail
4. Best supporve care

GC = gemcitabine plus cisplatin; GFR = glomular filtration rate; HD-MVAC = (high-dose) methotrexate,
vinblastine, adriamycin plus cisplatin; PCG = paclitaxel, cisplatin, gemcitabine; PS = performance status.
Fig. 2 – Flow chart for the management of metastatic urothelial cancer.
GC = gemcitabine plus cisplatin; GFR = glomerular filtration rate; HD = high-dose; MVAC = methotrexate, vinblastine, adriamycin, and cisplatin;
PCG = paclitaxel, cisplatin, gemcitabine; PD-L1 = Programmed Death Ligand 1; PS = performance status.

taxel, docetaxel, or vinflunine, 10.3 vs 7.4 mo, HR: 0.73; 95% longer follow-up [125]. In addition, health-related QoL
CI: 0.59–0.91), leading to approval [124]. The benefit was (HRQoL) was stable during pembrolizumab, whereas it
independent of PD-L1 expression levels, but consistent with deteriorated on chemotherapy [126].
98 E U RO P E A N U RO L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

Atezolizumab, a PD-L1 inhibitor, was the first checkpoint Table 19 – Summary of evidence and consensus statements for
postchemotherapy surgery and oligometastatic disease.
inhibitor approved for BC based on the results of a phase
2 trial [127]. A phase 3 RCT compared atezolizumab with Summary of evidence LE
second-line chemotherapy (paclitaxel, docetaxel, or vin-
Postchemotherapy surgery after partial or complete response may 3
flunine). There was no OS benefit for patients with high PD- contribute to long-term DFS in selected patients.
L1 expression, but OS was improved in the ITT population in EAU-ESMO consensus statements [4,5] a

an exploratory analysis (8.6 vs 8.0 mo, HR: 0.85, 95% CI: In a minority of patients with one metastatic lesion, cure is possible after
radical treatment.
0.73–0.99).
In patients with more than two metastatic sites, cure is not possible.
The PD-1 inhibitor nivolumab was also approved based In metachronous OMD, time to relapse is an important prognostic indicator.
on the results of a single-arm phase 2 trial [128]. ORR was Liver is an unfavourable OMD site for curative therapy.
19.6% and OS was 8.74 mo for the entire group. Based on Bone is an unfavourable OMD site for curative therapy.
PET-CT scanning should be included in OMD staging when considering
results of phase 1/2 trials, only durvalumab and avelumab radical treatment.
are currently approved for this indication in the USA. Radical treatment of OMD should be accompanied by adjuvant or
Novel drugs in early-phase clinical research are the neoadjuvant systemic therapy.
fibroblast growth factor receptor (FGFR) inhibitors erdafi-
DFS = disease-free survival; EAU = European Association of Urology;
tinib and rogaratinib demonstrating encouraging response ESMO = European Society for Medical Oncology; LE = level of evidence;
rates in patients with prespecified FGFR alterations OMD = oligometastatic disease; PET-CT = positron emission tomography-
[129,130]. Another promising drug is enfortumab vedotin, computed tomography.
a
Only statements that met the a priori consensus threshold across all three
an antibody-drug conjugate targeting Nectin-4, which is stakeholder groups are listed (defined as 70% agreement and 15%
highly expressed in UC. A phase 2, single-arm study after disagreement, or vice versa).
prior platinum chemotherapy and immunotherapy showed
objective response rates of 44%, including 12% of CRs with a
tolerable safety profile [131].
Patients with metastatic bone disease, irrespective of the orthotopic neobladder is the need for clean intermittent
cancer type, should be considered for bone-targeted catheterisation (62.5%) [139]. In all, HRQoL outcomes are
treatment [132]. Patients treated with zoledronic acid or good, provided there are good patient selection and
denosumab should be informed about possible side effects involvement of patients’ choice [136].
including osteonecrosis of the jaw and hypocalcaemia. On study reported that QoL was better after MMT than
Supplementation with calcium and vitamin D is mandatory. RC [140].

26. Follow-up of MIBC


24. Postchemotherapy surgery and oligometastatic
disease Current oncological surveillance protocols are based on the
patterns of recurrence drawn from retrospective series with
Although some studies suggest a survival benefit and QoL contradictory results, although two series found better
improvement of surgery after partial or complete remission survival for asymptomatic recurrences (Table 21) [141,142].
after chemotherapy, the level of evidence supporting this Contemporary cystectomy has a 5–15% probability of
practice is mainly anecdotal and the respective literature pelvic recurrence that most often occurs within 6–18 mo
old (Table 19). However, these findings have been confirmed after surgery. Pelvic recurrences usually carry a poor
in a recent systematic review including 28 studies [133]. prognosis, despite treatment, with median survival from
4 to 8 mo. Treatment generally involves a combination of
25. Quality of life chemotherapy, radiation, and surgery [143].
Distant recurrence, most likely in LNs, lungs, liver, and
Several questionnaires have been validated for assessing bone, is seen in up to 50% of patients, is stage dependent,
HRQoL in patients with BC. The impact of BC was recently and mainly appears within the first 2 yr after RC. Median
reported to cause a significant decline in HRQoL in all survival of patients with progressive disease treated with
domains (Table 20). In MIBC, physical and social functioning platinum-based chemotherapy is 9–26 mo [144].
were particularly affected [134]. In case of RC, NAC does not New urethral tumours occur in 1.3–13.7%, with a lower
appear to have an impact on QoL in a recent study [135]. The frequency after an orthotopic neobladder (odds ratio [OR]:
impact of the type of diversion remains unclear. One review 0.44) [145]. Risk factors are malignancy in the prostatic
found no difference in overall QoL between continent and urethra in men and bladder neck in women. In case of risk
incontinent diversions [136], with greater improvement in factors, urethral follow-up is indicated (urethral wash or
physical health for incontinent diversions (p = 0.002), but no cytology) since there is a significant survival advantage in
differences in mental or social health. Another recent men with urethral recurrence diagnosed asymptomatically
review, however, showed significantly improved QoL after versus symptomatically [143]. Treatment depends on stage
an orthotopic diversion compared with the QoL after an ileal and the presence of distant metastasis.
conduit [137]. In case of a neobladder, day- and night-time Upper urinary tract UCs (UTUCs) occur in 4–10% of cases
continence significantly improved to, respectively, 92% and and represent the most common sites of late recurrence
51% after 18 mo [138]. A specific problem in women with an [146]. Median OS is 10–55 mo, and 60–67% of patients die of
E U RO P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4 99

Table 20 – Summary of evidence and recommendations for health-related quality of life.

Summary of evidence LE

Compared to noncancer controls, the diagnosis and treatment of bladder cancer have a negative impact on HRQoL. 2a
There is no difference in overall QoL between patients with continent or incontinent diversion. 1a
In most patient groups studied, the overall HRQoL after cystectomy remains good, irrespective of the type of urinary diversion used. 2b
Important determinants of (subjective) QoL are a patient’s personality, coping style, and social support. 3
In patients with platinum-refractory advanced urothelial carcinoma, pembrolizumab may be superior in terms of HRQoL compared with another line of 1b
chemotherapy.
Recommendations Strength rating

Use validated questionnaires to assess HRQoL in patients with muscle-invasive bladder cancer. Strong
Offer a continent urinary diversion unless a patient’s comorbidities, tumour variables, and coping abilities present clear Strong
contraindications.
Preoperative patient information, patient selection, surgical techniques, and careful postoperative follow-up are the cornerstones for Strong
achieving good long-term results.
Provide clear and exhaustive information on all potential benefits and side effects, allowing patients to make informed decisions. Strong
Encourage patients to participate actively in the decision-making process.
HRQoL = health-related quality of life; LE = level of evidence.

Table 21 – Summary of recommendations and consensus statements for specific recurrence sites.

Site of recurrence Summary of evidence LE Recommendation Strength rating

Local recurrence Poor prognosis. Treatment should be 2b Offer radiotherapy, chemotherapy, and Strong
individualised depending on the local possibly surgery as options for
extent of tumour. treatment, either alone or in
combination.
Distant recurrence Poor prognosis. 2b Offer chemotherapy as the first option, Strong
and consider metastasectomy in case
of unique metastasis site.
Upper urinary tract recurrence Risk factors are multifocal disease See EAU guidelines on upper urinary Strong
(NMIBC/CIS or positive ureteral tract urothelial carcinomas.
margins).
Secondary urethral tumour Staging and treatment should be done 3 See EAU guidelines on primary urethral Strong
as for primary urethral tumour. carcinoma.
EAU-ESMO consensus statements [4,5] a
After radical cystectomy with curative intent, regular follow-up is needed.
After radical cystectomy with curative intent, follow-up for the detection of second cancers in the urothelium is recommended.
After radical cystectomy with curative intent, follow-up of the urethra with cytology and/or cystoscopy is recommended in selected patients (eg, multifocality,
carcinoma in situ, and tumour in the prostatic urethra).
After trimodality treatment with curative intent, follow-up for the detection of relapse is recommended every 3–4 mo initially and then, after 3 yr, every 6 mo in
the majority of patients.
After trimodality treatment with curative intent, regular follow-up for the detection of relapse is needed in the majority of patients.
After trimodality treatment with curative intent, follow-up imaging to assess distant recurrence or recurrence outside the bladder is needed.
After trimodality treatment with curative intent, assessment of the urothelium to detect recurrence is recommended every 6 mo in the majority of patients.
After trimodality treatment with curative intent, in addition to a CT scan, other investigations of the bladder are recommended.
In patients with a partial or complete response after chemotherapy for metastatic urothelial cancer, regular follow-up is needed. Imaging studies may be done
according to signs/symptoms.
To detect relapse (outside the bladder) after trimodality treatment with curative intent, CT of the thorax and abdomen is recommended as the imaging method
for follow-up in the majority of patients.
To detect relapse (outside the bladder) after trimodality treatment with curative intent, routine imaging with CT of the thorax and abdomen should be stopped
after 5 yr in the majority of patients.
In patients treated with radical cystectomy with curative intent and who have a neobladder, management of household acids and bases includes regular
measurements of pH and sodium bicarbonate substitution, respectively, according to the measured value.
To detect relapse after radical cystectomy with curative intent, routine imaging with CT of the thorax and abdomen should be stopped after 5 yr in the majority of
patients.
To detect relapse after radical cystectomy with curative intent, a CT of the thorax and abdomen is recommended as the imaging method for follow-up in the
majority of patients.
Levels of LDH and CEA are not essential in the follow-up of patients with urothelial cancer to detect recurrence.
Vitamin B12 levels have to be measured annually in the follow-up of patients treated with radical cystectomy and bowel diversion with curative intent.

CEA = carcinoembryonic antigen; CIS = carcinoma in situ; CT = computed tomography; EAU = European Association of Urology; ESMO = European Society for
Medical Oncology; LDH = lactate dehydrogenase; LE = level of evidence; NMIBC = non–muscle-invasive bladder cancer.
a
Only statements that met the a priori consensus threshold across all three stakeholder groups are listed (defined as 70% agreement and 15% disagreement, or
vice versa).
100 E U RO P E A N U RO L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

metastatic disease [143]. A meta-analysis found that 38% of investigator for phase 2 and 3 trials with Roche, MSD, AstraZeneca, Ipsen,
UTUC recurrence was diagnosed by follow-up investiga- Janssen, Bayer, Novartis, and BMS; is a member of advisory boards for
tions, whereas in the remaining 62%, diagnosis was based Roche, Novartis, Ipsen, MSD, BMS, and Janssen; receives honoraria for
lectures and travel fees from Roche, AstraZeneca, Novartis, and Ipsen;
on symptoms. Risk factors are noninvasive cancer, multifo-
and receives travel fees from MSD. Dr. Y. Neuzillet is a consultant for
cal disease, and positive ureteral or urethral margins.
Astellas, AstraZeneca, Bouchara-Recordati, BMS, Ipsen, Janssen, Medac,
Follow-up schedules are based on low-level evidence. A
MSD, Roche, Sanofi Pasteur, and Sanofi Aventis. Professor Dr. M.J. Ribal
schedule suggested by the EAU Guidelines Panel includes a receives speaker honoraria from Astellas, Janssen, Ipsen, and
CT scan (every 6 mo) until the 3rd year, followed by annual Olympus. Dr. H.M. Bruins, Professor Dr. E.M. Compérat, Dr. N.C. Cowan,
imaging thereafter up to 5 yr. In case of risk factors for Dr. V. Hernàndez, Dr. E. Linares-Espinós, Dr. M. Rouanne, Professor Dr. G.
UTUCs, longer follow-up with CT is indicated [147]. The N. Thalmann, Dr. E. Veskimae, Dr. A.G. van der Heijden have nothing to
length of follow-up is not well known, and depends on stage disclose.
and competing health factors. In case of locally advanced
disease or LN involvement, for example, a higher risk of Funding/Support and role of the sponsor: None.
recurrence for >20 yr was reported [148]. Since data for
follow-up strategies are sparse, a number of key questions References
were included in the recently held consensus project [4,5].
Functional follow-up should be long term to detect and [1] Phillips B, Ball C, Sackett D, et al. Oxford Centre for Evidence-based
Medicine levels of evidence. Updated by Jeremy Howick; March
treat complications related to urinary diversion, which are
2009. https://www.cebm.net/2009/06/oxford-centre-evidence-
seen in up to 54% of patients after 15 yr of follow-up
based-medicine-levels-evidence-march-2009/ [accessed April
[143]. Functional complications include vitamin B12 defi-
24, 2020].
ciency, metabolic acidosis, worsening of renal function, [2] Guyatt GH, Oxman AD, Kunz R, Vist GE, Falck-Ytter Y, Schunemann
urinary infections, urolithiasis, stenosis of ureterointestinal HJ. What is "quality of evidence" and why is it important to
anastomosis, stoma complications in patients with ileal clinicians? BMJ 2008;336:995–8.
conduit, neobladder continence problems, emptying dys- [3] Guyatt GH, Oxman AD, Vist GE, et al. GRADE: an emerging con-
function, and a 21% increased risk of fractures due to chronic sensus on rating quality of evidence and strength of recommenda-
metabolic acidosis and subsequent long-term bone loss tions. BMJ 2008;336:924–6.
[143,149]. [4] Witjes F, Babjuk M, Bellmunt J, et al. EAU-ESMO consensus state-
ments on the management of advanced and variant bladder can-
cer-an international collaborative multi-stakeholder effort: under
Author contributions: J. Alfred Witjes had full access to all the data in the the auspices of the EAU and ESMO Guidelines Committees. Eur
study and takes responsibility for the integrity of the data and the Urol 2020;77:223–50.
accuracy of the data analysis. [5] Horwich A, Babjuk M, Bellmunt J, et al. EAU-ESMO consensus
Study concept and design: Witjes. statements on the management of advanced and variant bladder
Acquisition of data: Witjes, Bruins, Cathomas, Compérat, Cowan, Gakis, cancer-an international collaborative multi-stakeholder effort:
Hernández, Espinós, Lorch, Neuzillet, Rouanne, Thalmann, Veskimäe, under the auspices of the EAU and ESMO Guidelines Committees.
Ribal, van der Heijden. Ann Oncol 2020;77:223–50.
Analysis and interpretation of data: Witjes, Bruins, Cathomas, Compérat, [6] Witjes JA, Lebret T, Comperat EM, et al. Updated 2016 EAU Guide-
Cowan, Gakis, Hernández, Espinós, Lorch, Neuzillet, Rouanne, Thalmann, lines on Muscle-invasive and Metastatic Bladder Cancer. Eur Urol
Veskimäe, Ribal, van der Heijden. 2017;71:462–75.
Drafting of the manuscript: Witjes, van der Heijden. [7] Freedman ND, Silverman DT, Hollenbeck AR, Schatzkin A, Abnet
Critical revision of the manuscript for important intellectual content: CC. Association between smoking and risk of bladder cancer
Witjes, Bruins, Cathomas, Compérat, Cowan, Gakis, Hernández, Espinós, among men and women. JAMA 2011;306:737–45.
Lorch, Neuzillet, Rouanne, Thalmann, Veskimäe, Ribal, van der Heijden. [8] Brennan P, Bogillot O, Cordier S, et al. Cigarette smoking and
Statistical analysis: None. bladder cancer in men: a pooled analysis of 11 case-control studies.
Obtaining funding: None. Int J Cancer 2000;86:289–94.
Administrative, technical, or material support: Witjes. [9] Zelefsky MJ, Housman DM, Pei X, et al. Incidence of secondary cancer
Supervision: None. development after high-dose intensity-modulated radiotherapy and
Other: None. image-guided brachytherapy for the treatment of localized prostate
cancer. Int J Radiat Oncol Biol Phys 2012;83:953–9.
[10] Zamora-Ros R, Sacerdote C, Ricceri F, et al. Flavonoid and lignan
Financial disclosures: J. Alfred Witjes certifies that all conflicts of intake in relation to bladder cancer risk in the European Prospec-
interest, including specific financial interests and relationships and tive Investigation into Cancer and Nutrition (EPIC) study. Br J
affiliations relevant to the subject matter or materials discussed in the Cancer 2014;111:1870–80.
manuscript (eg, employment/affiliation, grants or funding, consultan- [11] Teleka S, Haggstrom C, Nagel G, et al. Risk of bladder cancer by
cies, honoraria, stock ownership or options, expert testimony, royalties, disease severity in relation to metabolic factors and smoking: a
or patents filed, received, or pending), are the following: Professor Dr. J.A. prospective pooled cohort study of 800,000 men and women. Int J
Witjes receives personal fees as an advisor for Roche, Merck, and Cancer 2018;143:3071–82.
BMS. Professor Dr. R. Cathomas received personal fees from Roche, Pfizer, [12] Adil M, Khan RA, Ghosh P, Venkata SK, Kandhare AD, Sharma M.
MSD, BMS, AstraZeneca, Janssen, Astellas, Bayer, Sanofi Aventis, Pioglitazone and risk of bladder cancer in type 2 diabetes mellitus
Debiopharm, Novartis, and Ipsen. Professor Dr. G. Gakis is a company patients: a systematic literature review and meta-analysis of
consultant for Erbe, Ipsen, Pierre Fabre, Medac, and MSD, and receives observational studies using real-world data. Clin Epidemiol Global
research support from IPSEN. Professor Dr. A. Lorch is a principal Health 2018;6:61–8.
E U RO P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4 101

[13] Yu Z, Yue W, Jiuzhi L, Youtao J, Guofei Z, Wenbin G. The risk of in patients treated with radical cystectomy: a population-based
bladder cancer in patients with urinary calculi: a meta-analysis. study. Ann Surg Oncol 2012;19:309–17.
Urolithiasis 2018;46:573–9. [31] Koppie TM, Serio AM, Vickers AJ, et al. Age-adjusted Charlson
[14] Bayne CE, Farah D, Herbst KW, Hsieh MH. Role of urinary tract comorbidity score is associated with treatment decisions and
infection in bladder cancer: a systematic review and meta-analy- clinical outcomes for patients undergoing radical cystectomy for
sis. World J Urol 2018;36:1181–90. bladder cancer. Cancer 2008;112:2384–92.
[15] Patafio FM, Robert Siemens D, Wei X, Booth CM. Is there a gender [32] Bolenz C, Ho R, Nuss GR, et al. Management of elderly patients with
effect in bladder cancer? A population-based study of practice and urothelial carcinoma of the bladder: guideline concordance and
outcomes. Can Urol Assoc J 2015;9:269–74. predictors of overall survival. BJU Int 2010;106:1324–9.
[16] Andreassen BK, Grimsrud TK, Haug ES. Bladder cancer survival: [33] Mayr R, May M, Martini T, et al. Comorbidity and performance
Women better off in the long run. Eur J Cancer 2018;95:52–8. indices as predictors of cancer-independent mortality but not of
[17] Hansel DE, Amin MB, Comperat E, et al. A contemporary update on cancer-specific mortality after radical cystectomy for urothelial
pathology standards for bladder cancer: transurethral resection carcinoma of the bladder. Eur Urol 2012;62:662–70.
and radical cystectomy specimens. Eur Urol 2013;63:321–32. [34] Dutta R, Abdelhalim A, Martin JW, et al. Effect of tumor location on
[18] Veskimae E, Espinos EL, Bruins HM, et al. What is the prognostic survival in urinary bladder adenocarcinoma: a population-based
and clinical importance of urothelial and nonurothelial histologi- analysis. Urol Oncol 2016;34:531.e1–6.
cal variants of bladder cancer in predicting oncological outcomes [35] Ku JH, Kang M, Kim HS, Jeong CW, Kwak C, Kim HS. Lymph node
in patients with muscle-invasive and metastatic bladder cancer? A density as a prognostic variable in node-positive bladder cancer: a
European Association of Urology Muscle Invasive and Metastatic meta-analysis. BMC Cancer 2015;15:447.
Bladder Cancer Guidelines Panel systematic review. Eur Urol Oncol [36] Mathieu R, Lucca I, Roupret M, Briganti A, Shariat SF. The prognos-
2019;2:625–42. tic role of lymphovascular invasion in urothelial carcinoma of the
[19] Comperat EM, Burger M, Gontero P, et al. Grading of urothelial bladder. Nat Rev Urol 2016;13:471–9.
carcinoma and the new “World Health Organisation Classification [37] Kimura S, Mari A, Foerster B, et al. Prognostic value of concomitant
of Tumours of the urinary system and male genital organs 2016”. carcinoma in situ in the radical cystectomy specimen: a systematic
Eur Urol Focus 2019;5:457–66. review and meta-analysis. J Urol 2019;201:46–53.
[20] Ploeg M, Kiemeney LA, Smits GA, et al. Discrepancy between [38] Wu S, Zhao X, Wang Y, et al. Pretreatment neutrophil-lymphocyte
clinical staging through bimanual palpation and pathological ratio as a predictor in bladder cancer and metastatic or unresect-
staging after cystectomy. Urol Oncol 2012;30:247–51. able urothelial carcinoma patients: a pooled analysis of compara-
[21] Babjuk M, Burger M, Zigeuner R. Guidelines on non-muscle-inva- tive studies. Cell Physiol Biochem 2018;46:1352–64.
sive bladder cancer (Ta, T1 and CIS). Arnhem, The Netherlands: [39] Ojerholm E, Smith A, Hwang WT, et al. Neutrophil-to-lymphocyte
European Association of Urology; 2020. ratio as a bladder cancer biomarker: assessing prognostic and
[22] Huang L, Kong Q, Liu Z, Wang J, Kang Z, Zhu Y. The diagnostic value predictive value in SWOG 8710. Cancer 2017;123:794–801.
of MR imaging in differentiating T staging of bladder cancer: a [40] Kamoun A, de Reynies A, Allory Y, et al. A consensus molecular
meta-analysis. Radiology 2018;286:502–11. classification of muscle-invasive bladder cancer. Eur Urol
[23] Heidenreich A, Albers P, Classen J, et al. Imaging studies in meta- 2020;77:420–33.
static urogenital cancer patients undergoing systemic therapy: [41] Pietzak EJ, Zabor EC, Bagrodia A, et al. Genomic differences be-
recommendations of a multidisciplinary consensus meeting of tween "primary" and "secondary" muscle-invasive bladder cancer
the Association of Urological Oncology of the German Cancer as a basis for disparate outcomes to cisplatin-based neoadjuvant
Society. Urol Int 2010;85:1–10. chemotherapy. Eur Urol 2019;75:231–9.
[24] Albers P, Park SI, Niegisch G, et al. Randomized phase III trial of 2nd [42] Motterle G, Andrews JR, Morlacco A, Karnes RJ. Predicting response
line gemcitabine and paclitaxel chemotherapy in patients with to neoadjuvant chemotherapy in bladder cancer. Eur Urol Focus. In
advanced bladder cancer: short-term versus prolonged treatment press. https://doi.org/10.1016/j.euf.2019.10.016.
[German Association of Urological Oncology (AUO) trial AB 20/99]. [43] Necchi A, Anichini A, Raggi D, et al. Pembrolizumab as neoadjuvant
Ann Oncol 2011;22:288–94. therapy before radical cystectomy in patients with muscle-inva-
[25] Haden TD, Prunty MC, Jones AB, Deroche CB, Murray KS, Pokala N. sive urothelial bladder carcinoma (PURE-01): an open-label, sin-
Comparative perioperative outcomes in septuagenarians and gle-arm, phase II study. J Clin Oncol 2018;36:3353–60.
octogenarians undergoing radical cystectomy for bladder can- [44] Necchi A, Raggi D, Gallina A, et al. Updated results of PURE-01 with
cer—do outcomes differ? Eur Urol Focus 2018;4:895–9. preliminary activity of neoadjuvant pembrolizumab in patients
[26] Mayr R, Gierth M, Zeman F, et al. Sarcopenia as a comorbidity- with muscle-invasive bladder carcinoma with variant histologies.
independent predictor of survival following radical cystectomy for Eur Urol 2020;77:439–46.
bladder cancer. J Cachexia Sarcopenia Muscle 2018;9:505–13. [45] Cambier S, Sylvester RJ, Collette L, et al. EORTC nomograms and
[27] Lughezzani G, Sun M, Shariat SF, et al. A population-based com- risk groups for predicting recurrence, progression, and disease-
peting-risks analysis of the survival of patients treated with radical specific and overall survival in non-muscle-invasive stage Ta-T1
cystectomy for bladder cancer. Cancer 2011;117:103–9. urothelial bladder cancer patients treated with 1-3 years of main-
[28] Mayr R, May M, Martini T, et al. Predictive capacity of four tenance bacillus Calmette-Guerin. Eur Urol 2016;69:60–9.
comorbidity indices estimating perioperative mortality after rad- [46] van den Bosch S, Witjes JA. Long-term cancer-specific survival
ical cystectomy for urothelial carcinoma of the bladder. BJU Int in patients with high-risk, non-muscle-invasive bladder cancer
2012;110:E222–7. and tumour progression: a systematic review. Eur Urol
[29] Morgan TM, Keegan KA, Barocas DA, et al. Predicting the proba- 2011;60:493–500.
bility of 90-day survival of elderly patients with bladder cancer [47] Rosenblatt R, Sherif A, Rintala E, et al. Pathologic downstaging is a
treated with radical cystectomy. J Urol 2011;186:829–34. surrogate marker for efficacy and increased survival following
[30] Abdollah F, Sun M, Schmitges J, et al. Development and validation neoadjuvant chemotherapy and radical cystectomy for muscle-
of a reference table for prediction of postoperative mortality rate invasive urothelial bladder cancer. Eur Urol 2012;61:1229–38.
102 E U RO P E A N U RO L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

[48] Sherif A, Holmberg L, Rintala E, et al. Neoadjuvant cisplatinum with standard radical cystectomy in men: a systematic review.
based combination chemotherapy in patients with invasive blad- Urol Oncol 2017;35:539.e17–e.
der cancer: a combined analysis of two Nordic studies. Eur Urol [66] Veskimae E, Neuzillet Y, Rouanne M, et al. Systematic review of the
2004;45:297–303. oncological and functional outcomes of pelvic organ-preserving
[49] Yin M, Joshi M, Meijer RP, et al. Neoadjuvant chemotherapy for radical cystectomy (RC) compared with standard RC in women
muscle-invasive bladder cancer: a systematic review and two-step who undergo curative surgery and orthotopic neobladder substi-
meta-analysis. Oncologist 2016;21:708–15. tution for bladder cancer. BJU Int 2017;120:12–24.
[50] Galsky MD, Pal SK, Chowdhury S, et al. Comparative effectiveness [67] Ali-El-Dein B, Mosbah A, Osman Y, et al. Preservation of the
of gemcitabine plus cisplatin versus methotrexate, vinblastine, internal genital organs during radical cystectomy in selected
doxorubicin, plus cisplatin as neoadjuvant therapy for muscle- women with bladder cancer: a report on 15 cases with long term
invasive bladder cancer. Cancer 2015;121:2586–93. follow-up. Eur J Surg Oncol 2013;39:358–64.
[51] Griffiths G, Hall R, Sylvester R, Raghavan D, Parmar MK. Interna- [68] Temkin SM, Bergstrom J, Samimi G, Minasian L. Ovarian cancer
tional phase III trial assessing neoadjuvant cisplatin, methotrexate, prevention in high-risk women. Clin Obstet Gynecol
and vinblastine chemotherapy for muscle-invasive bladder can- 2017;60:738–57.
cer: long-term results of the BA06 30894 trial. J Clin Oncol [69] Novara G, Catto JW, Wilson T, et al. Systematic review and cumu-
2011;29:2171–7. lative analysis of perioperative outcomes and complications after
[52] Peyton CC, Tang D, Reich RR, et al. Downstaging and survival robot-assisted radical cystectomy. Eur Urol 2015;67:376–401.
outcomes associated with neoadjuvant chemotherapy regimens [70] Wilson TG, Guru K, Rosen RC, et al. Best practices in robot-assisted
among patients treated with cystectomy for muscle-invasive blad- radical cystectomy and urinary reconstruction: recommendations
der cancer. JAMA Oncol 2018;4:1535–42. of the Pasadena Consensus Panel. Eur Urol 2015;67:363–75.
[53] Anari F, O’Neill J, Choi W, et al. Neoadjuvant dose-dense gemci- [71] Rai BP, Bondad J, Vasdev N, et al. Robotic versus open radical
tabine and cisplatin in muscle-invasive bladder cancer: results of a cystectomy for bladder cancer in adults. Cochrane Database Syst
phase 2 trial. Eur Urol Oncol 2018;1:54–60. Rev 2019;4:Cd011903.
[54] Hanna N, Trinh Q-D-D, Seisen T, et al. Effectiveness of neoadjuvant [72] Bochner BH, Dalbagni G, Marzouk KH, et al. Randomized trial
chemotherapy for muscle-invasive bladder cancer in the current comparing open radical cystectomy and robot-assisted laparoscopic
real world setting in the USA. Eur Urol Oncol 2018;1:83–90. radical cystectomy: oncologic outcomes. Eur Urol 2018;74:465–71.
[55] Grossman HB, Natale RB, Tangen CM, et al. Neoadjuvant chemo- [73] Jancke G, Aljabery F, Gudjonsson S, et al. Port-site metastases after
therapy plus cystectomy compared with cystectomy alone for robot-assisted radical cystectomy: is there a publication bias? Eur
locally advanced bladder cancer. N Engl J Med 2003;349:859–66. Urol 2018;73:641–2.
[56] Granfors T, Tomic R, Ljungberg B. Downstaging and survival ben- [74] Parekh DJ, Reis IM, Castle EP, et al. Robot-assisted radical cystec-
efits of neoadjuvant radiotherapy before cystectomy for patients tomy versus open radical cystectomy in patients with bladder
with invasive bladder carcinoma. Scand J Urol Nephrol cancer (RAZOR): an open-label, randomised, phase 3, non-inferi-
2009;43:293–9. ority trial. Lancet 2018;391:2525–36.
[57] Bayoumi Y, Heikal T, Darweish H. Survival benefit of adjuvant [75] Hussein AA, May PR, Jing Z, et al. Outcomes of Intracorporeal
radiotherapy in stage III and IV bladder cancer: results of urinary diversion after robot-assisted radical cystectomy: results
170 patients. Cancer Manage Res 2014;6:459–65. from the International Robotic Cystectomy Consortium. J Urol
[58] Gore JL, Lai J, Setodji CM, Litwin MS, Saigal CS. Mortality increases 2018;199:1302–11.
when radical cystectomy is delayed more than 12 weeks: results [76] Tang K, Li H, Xia D, et al. Laparoscopic versus open radical cystec-
from a Surveillance, Epidemiology, and End Results-Medicare tomy in bladder cancer: a systematic review and meta-analysis of
analysis. Cancer 2009;115:988–96. comparative studies. PLoS One 2014;9:e95667.
[59] Bruins HM, Aben KK, Arends TJ, van der Heijden AG, Witjes AJ. [77] Roth B, Wissmeyer MP, Zehnder P, et al. A new multimodality
The effect of the time interval between diagnosis of muscle- technique accurately maps the primary lymphatic landing sites of
invasive bladder cancer and radical cystectomy on staging and the bladder. Eur Urol 2010;57:205–11.
survival: a Netherlands Cancer Registry analysis. Urol Oncol [78] Dorin RP, Daneshmand S, Eisenberg MS, et al. Lymph node dissec-
2016;34:166.e1–6. tion technique is more important than lymph node count in
[60] Ayres BE, Gillatt D, McPhail S, et al. A delay in radical cystectomy of identifying nodal metastases in radical cystectomy patients: a
&3 months is not associated with a worse clinical outcome. BJU Int comparative mapping study. Eur Urol 2011;60:946–52.
2008;102:1045. [79] Bruins HM, Veskimae E, Hernandez V, et al. The impact of the
[61] Karl A, Buchner A, Becker A, et al. A new concept for early recovery extent of lymphadenectomy on oncologic outcomes in patients
after surgery for patients undergoing radical cystectomy for blad- undergoing radical cystectomy for bladder cancer: a systematic
der cancer: results of a prospective randomized study. J Urol review. Eur Urol 2014;66:1065–77.
2014;191:335–40. [80] Gschwend JE, Heck MM, Lehmann J, et al. Extended versus limited
[62] Xu W, Daneshmand S, Bazargani ST, et al. Postoperative pain man- lymph node dissection in bladder cancer patients undergoing
agement after radical cystectomy: comparing traditional versus radical cystectomy: survival results from a prospective, random-
enhanced recovery protocol pathway. J Urol 2015;194:1209–13. ized trial. Eur Urol 2019;75:604–11.
[63] Lee CT, Chang SS, Kamat AM, et al. Alvimopan accelerates gastro- [81] Hautmann RE, de Petriconi RC, Volkmer BG. Lessons learned from
intestinal recovery after radical cystectomy: a multicenter ran- 1,000 neobladders: the 90-day complication rate. J Urol
domized placebo-controlled trial. Eur Urol 2014;66:265–72. 2010;184:990–4, quiz 1235.
[64] Brennan K, Karim S, Doiron RC, Siemens DR, Booth CM. Venous [82] Roth B, Furrer MA, Giannakis I, et al. Positive pre-cystectomy
thromboembolism and peri-operative chemotherapy for muscle- biopsies of the prostatic urethra or bladder neck do not necessarily
invasive bladder cancer: a population-based study. Bladder Cancer preclude orthotopic bladder substitution. J Urol 2019;201:909–15.
2018;4:419–28. [83] Gerharz EW, Turner WH, Kalble T, Woodhouse CR. Metabolic and
[65] Hernandez V, Espinos EL, Dunn J, et al. Oncological and functional functional consequences of urinary reconstruction with bowel.
outcomes of sexual function-preserving cystectomy compared BJU Int 2003;91:143–9.
E U RO P E A N U R O L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4 103

[84] Lebret T, Herve JM, Yonneau L, et al. After cystectomy, is it justified [101] Audenet F, Waingankar N, Ferket BS, et al. Effectiveness of trans-
to perform a bladder replacement for patients with lymph node urethral resection plus systemic chemotherapy as definitive treat-
positive bladder cancer? Eur Urol 2002;42:344–9, discussion 9. ment for muscle invasive bladder cancer in population level data. J
[85] Gershman B, Eisenberg MS, Thompson RH, et al. Comparative Urol 2018;200:996–1004.
impact of continent and incontinent urinary diversion on long- [102] Giacalone NJ, Shipley WU, Clayman RH, et al. Long-term outcomes
term renal function after radical cystectomy in patients with after bladder-preserving tri-modality therapy for patients with
preoperative chronic kidney disease 2 and chronic kidney disease muscle-invasive bladder cancer: an updated analysis of the Mas-
3a. Int J Urol 2015;22:651–6. sachusetts General Hospital experience. Eur Urol 2017;71:952–60.
[86] Longo N, Imbimbo C, Fusco F, et al. Complications and quality of life [103] Ploussard G, Daneshmand S, Efstathiou JA, et al. Critical analysis of
in elderly patients with several comorbidities undergoing cutane- bladder sparing with trimodal therapy in muscle-invasive bladder
ous ureterostomy with single stoma or ileal conduit after radical cancer: a systematic review. Eur Urol 2014;66:120–37.
cystectomy. BJU Int 2016;118:521–6. [104] Krasnow RE, Drumm M, Roberts HJ, et al. Clinical outcomes of
[87] Berger I, Wehrberger C, Ponholzer A, et al. Impact of the use of patients with histologic variants of urothelial cancer treated with
bowel for urinary diversion on perioperative complications and trimodality bladder-sparing therapy. Eur Urol 2017;72:54–60.
90-day mortality in patients aged 75 years or older. Urol Int [105] Ritch CR, Balise R, Prakash NS, et al. Propensity matched compar-
2015;94:394–400. ative analysis of survival following chemoradiation or radical
[88] Nieuwenhuijzen JA, de Vries RR, Bex A, et al. Urinary diversions cystectomy for muscle-invasive bladder cancer. BJU Int
after cystectomy: the association of clinical factors, complications 2018;121:745–51.
and functional results of four different diversions. Eur Urol [106] Cahn DB, Handorf EA, Ghiraldi EM, et al. Contemporary use trends
2008;53:834–42, discussion 42–4. and survival outcomes in patients undergoing radical cystectomy
[89] Madersbacher S, Schmidt J, Eberle JM, et al. Long-term outcome of or bladder-preservation therapy for muscle-invasive bladder can-
ileal conduit diversion. J Urol 2003;169:985–90. cer. Cancer 2017;123:4337–45.
[90] Wiesner C, Bonfig R, Stein R, et al. Continent cutaneous urinary [107] Fahmy O, Khairul-Asri MG, Schubert T, et al. A systematic review
diversion: long-term follow-up of more than 800 patients with and meta-analysis on the oncological long-term outcomes after
ileocecal reservoirs. World J Urol 2006;24:315–8. trimodality therapy and radical cystectomy with or without
[91] Hautmann RE, Volkmer BG, Schumacher MC, Gschwend JE, Studer neoadjuvant chemotherapy for muscle-invasive bladder cancer.
UE. Long-term results of standard procedures in urology: the ileal Urol Oncol 2018;36:43–53.
neobladder. World J Urol 2006;24:305–14. [108] Sanchez A, Wszolek MF, Niemierko A, et al. Incidence, clinicopatho-
[92] Shariat SF, Karakiewicz PI, Palapattu GS, et al. Outcomes of radical logical risk factors, management and outcomes of nonmuscle inva-
cystectomy for transitional cell carcinoma of the bladder: a con- sive recurrence after complete response to trimodality therapy for
temporary series from the Bladder Cancer Research Consortium. J muscle invasive bladder cancer. J Urol 2018;199:407–15.
Urol 2006;176:2414–22, discussion 22. [109] Leow JJ, Martin-Doyle W, Rajagopal PS, et al. Adjuvant chemother-
[93] Nuhn P, May M, Sun M, et al. External validation of postoperative apy for invasive bladder cancer: a 2013 updated systematic review
nomograms for prediction of all-cause mortality, cancer-specific and meta-analysis of randomized trials. Eur Urol 2014;66:42–54.
mortality, and recurrence in patients with urothelial carcinoma of [110] Sternberg CN, Skoneczna I, Kerst JM, et al. Immediate versus
the bladder. Eur Urol 2012;61:58–64. deferred chemotherapy after radical cystectomy in patients with
[94] Stein JP, Lieskovsky G, Cote R, et al. Radical cystectomy in the pT3-pT4 or N+ M0 urothelial carcinoma of the bladder (EORTC
treatment of invasive bladder cancer: long-term results in 1,054 30994): an intergroup, open-label, randomised phase 3 trial. Lan-
patients. J Clin Oncol 2001;19:666–75. cet Oncol 2015;16:76–86.
[95] Bruins HM, Veskimae E, Hernandez V, et al. The importance of [111] Galsky MD, Stensland KD, Moshier E, et al. Effectiveness of adju-
hospital and surgeon volume as major determinants of morbidity vant chemotherapy for locally advanced bladder cancer. J Clin
and mortality after radical cystectomy for bladder cancer: a sys- Oncol 2016;34:825–32.
tematic review and recommendations by the European Associa- [112] Berg S, D’Andrea D, Vetterlein MW, et al. Impact of adjuvant
tion of Urology Muscle-invasive and Metastatic Bladder Cancer chemotherapy in patients with adverse features and variant
Guideline Panel. Eur Urol Oncol 2020;3:131–44. https://doi.org/10. histology at radical cystectomy for muscle-invasive carcinoma
1016/j.euo.2019.11.005 of the bladder: does histologic subtype matter? Cancer
[96] Ghahestani SM, Shakhssalim N. Palliative treatment of intractable 2019;125:1449–58.
hematuria in context of advanced bladder cancer: a systematic [113] Bellmunt J, Petrylak DP. New therapeutic challenges in advanced
review. Urol J 2009;6:149–56. bladder cancer. Semin Oncol 2012;39:598–607.
[97] Solsona E, Iborra I, Collado A, Rubio-Briones J, Casanova J, Calatrava [114] von der Maase H, Sengelov L, Roberts JT, et al. Long-term survival
A. Feasibility of radical transurethral resection as monotherapy for results of a randomized trial comparing gemcitabine plus cisplatin,
selected patients with muscle invasive bladder cancer. J Urol with methotrexate, vinblastine, doxorubicin, plus cisplatin in
2010;184:475–80. patients with bladder cancer. J Clin Oncol 2005;23:4602–8.
[98] Korpics M, Block AM, Altoos B, et al. Maximizing survival in [115] Sternberg CN, de Mulder P, Schornagel JH, et al. Seven year update
patients with muscle-invasive bladder cancer undergoing curative of an EORTC phase III trial of high-dose intensity M-VAC chemo-
bladder-preserving radiotherapy: the impact of radiotherapy dose therapy and G-CSF versus classic M-VAC in advanced urothelial
escalation. J Radiat Oncol 2017;6:387–95. tract tumours. Eur J Cancer 2006;42:50–4.
[99] Sondergaard J, Holmberg M, Jakobsen AR, Agerbaek M, Muren LP, [116] Galsky MD, Chen GJ, Oh WK, et al. Comparative effectiveness of
Hoyer M. A comparison of morbidity following conformal versus cisplatin-based and carboplatin-based chemotherapy for treat-
intensity-modulated radiotherapy for urinary bladder cancer. Acta ment of advanced urothelial carcinoma. Ann Oncol
Oncol 2014;53:1321–8. 2012;23:406–10.
[100] Korpics MC, Block AM, Martin B, et al. Concurrent chemotherapy is [117] Bajorin DF, Dodd PM, Mazumdar M, et al. Long-term survival in
associated with improved survival in elderly patients with bladder metastatic transitional-cell carcinoma and prognostic factors pre-
cancer undergoing radiotherapy. Cancer 2017;123:3524–31. dicting outcome of therapy. J Clin Oncol 1999;17:3173–81.
104 E U RO P E A N U RO L O GY 7 9 ( 2 0 21 ) 8 2 – 10 4

[118] Galsky MD, Hahn NM, Rosenberg J, et al. A consensus definition of [133] Abufaraj M, Dalbagni G, Daneshmand S, et al. The role of surgery in
patients with metastatic urothelial carcinoma who are unfit for metastatic bladder cancer: a systematic review. Eur Urol
cisplatin-based chemotherapy. Lancet Oncol 2011;12:211–4. 2018;73:543–57.
[119] De Santis M, Bellmunt J, Mead G, et al. Randomized phase II/III trial [134] Smith AB, Jaeger B, Pinheiro LC, et al. Impact of bladder cancer on
assessing gemcitabine/carboplatin and methotrexate/carboplatin/ health-related quality of life. BJU Int 2018;121:549–57.
vinblastine in patients with advanced urothelial cancer who are [135] Feuerstein MA, Goldstein L, Reaves B, et al. Propensity-matched
unfit for cisplatin-based chemotherapy: EORTC study 30986. J Clin analysis of patient-reported outcomes for neoadjuvant chemo-
Oncol 2012;30:191–9. therapy prior to radical cystectomy. World J Urol 2019;37:2401–7.
[120] O’Donnell PH, Plimack ER, Bellmunt J, et al. Pembrolizumab (Pem- [136] Yang LS, Shan BL, Shan BL, et al. A systematic review and meta-
bro; MK-3475) for advanced urothelial cancer: results of a phase IB analysis of quality of life outcomes after radical cystectomy for
study. J Clin Oncol 2015;33:296. bladder cancer. Surg Oncol 2016;25:281–97.
[121] Balar AV, Galsky MD, Rosenberg JE, et al. Atezolizumab as first-line [137] Cerruto MA, D’Elia C, Siracusano S, et al. Systematic review and
treatment in cisplatin-ineligible patients with locally advanced meta-analysis of non RCT’s on health related quality of life after
and metastatic urothelial carcinoma: a single-arm, multicentre, radical cystectomy using validated questionnaires: better results
phase 2 trial. Lancet 2017;389:67–76. with orthotopic neobladder versus ileal conduit. Eur J Surg Oncol
[122] Bellmunt J, Theodore C, Demkov T, et al. Phase III trial of vinflunine 2016;42:343–60.
plus best supportive care compared with best supportive care [138] Clifford TG, Shah SH, Bazargani ST, et al. Prospective evaluation
alone after a platinum-containing regimen in patients with ad- of continence following radical cystectomy and orthotopic
vanced transitional cell carcinoma of the urothelial tract. J Clin urinary diversion using a validated questionnaire. J Urol
Oncol 2009;27:4454–61. 2016;196:1685–91.
[123] Petrylak DP, de Wit R, Chi KN, et al. Ramucirumab plus docetaxel [139] Bartsch G, Daneshmand S, Skinner EC, Syan S, Skinner DG, Penson
versus placebo plus docetaxel in patients with locally advanced or DF. Urinary functional outcomes in female neobladder patients.
metastatic urothelial carcinoma after platinum-based therapy World J Urol 2014;32:221–8.
(RANGE): a randomised, double-blind, phase 3 trial. Lancet [140] Mak KS, Smith AB, Eidelman A, et al. Quality of life in long-term
2017;390:2266–77. survivors of muscle-invasive bladder cancer. Int J Radiat Oncol Biol
[124] Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as second- Phys 2016;96:1028–36.
line therapy for advanced urothelial carcinoma. N Engl J Med [141] Giannarini G, Kessler TM, Thoeny HC, Nguyen DP, Meissner C,
2017;376:1015–26. Studer UE. Do patients benefit from routine follow-up to detect
[125] Fradet Y, Bellmunt J, Vaughn DJ, et al. Randomized phase III recurrences after radical cystectomy and ileal orthotopic bladder
KEYNOTE-045 trial of pembrolizumab versus paclitaxel, doce- substitution? Eur Urol 2010;58:486–94.
taxel, or vinflunine in recurrent advanced urothelial cancer: [142] Boorjian SA, Tollefson MK, Cheville JC, Costello BA, Thapa P, Frank I.
results of & 2 years of follow-up. Ann Oncol 2019;30:970–6. Detection of asymptomatic recurrence during routine oncological
[126] Vaughn DJ, Bellmunt J, Fradet Y, et al. Health-related quality-of-life followup after radical cystectomy is associated with improved
analysis From KEYNOTE-045: a phase III study of pembrolizumab patient survival. J Urol 2011;186:1796–802.
versus chemotherapy for previously treated advanced urothelial [143] Soukup V, Babjuk M, Bellmunt J, et al. Follow-up after surgical
cancer. J Clin Oncol 2018;36:1579–87. treatment of bladder cancer: a critical analysis of the literature.
[127] Powles T, Duran I, van der Heijden MS, et al. Atezolizumab versus Eur Urol 2012;62:290–302.
chemotherapy in patients with platinum-treated locally advanced or [144] Vrooman OP, Witjes JA. Follow-up of patients after curative blad-
metastatic urothelial carcinoma (IMvigor211): a multicentre, open- der cancer treatment: guidelines vs. practice. Curr Opin Urol
label, phase 3 randomised controlled trial. Lancet 2018;391:748–57. 2010;20:437–42.
[128] Sharma P, Retz M, Siefker-Radtke A, et al. Nivolumab in metastatic [145] Fahmy O, Khairul-Asri MG, Schubert T, et al. Urethral recurrence
urothelial carcinoma after platinum therapy (CheckMate 275): a after radical cystectomy for urothelial carcinoma: a systematic
multicentre, single-arm, phase 2 trial. Lancet Oncol 2017;18:312–22. review and meta-analysis. Urol Oncol 2018;36:54–9.
[129] Loriot Y, Necchi A, Park SH, et al. Erdafitinib in locally advanced or [146] Gakis G, Black PC, Bochner BH, et al. Systematic review on the fate
metastatic urothelial carcinoma. N Engl J Med 2019;381:338–48. of the remnant urothelium after radical cystectomy. Eur Urol
[130] Schuler M, Cho BC, Sayehli CM, et al. Rogaratinib in patients with 2017;71:545–57.
advanced cancers selected by FGFR mRNA expression: a phase [147] Picozzi S, Ricci C, Gaeta M, et al. Upper urinary tract recurrence
1 dose-escalation and dose-expansion study. Lancet Oncol following radical cystectomy for bladder cancer: a meta-analysis
2019;20:1454–66. on 13,185 patients. J Urol 2012;188:2046–54.
[131] Rosenberg JE, O’Donnell PH, Balar AV, et al. Pivotal trial of enfor- [148] Stewart-Merrill SB, Boorjian SA, Thompson RH, et al. Evaluation of
tumab vedotin in urothelial carcinoma after platinum and anti- current surveillance guidelines following radical cystectomy and
Programmed Death 1/Programmed Death Ligand 1 therapy. J Clin proposal of a novel risk-based approach. Urol Oncol 2015;33:339.
Oncol 2019;37:2592–600. e1–e.
[132] Aapro M, Abrahamsson PA, Body JJ, et al. Guidance on the use of [149] Gupta A, Atoria CL, Ehdaie B, et al. Risk of fracture after radical
bisphosphonates in solid tumours: recommendations of an inter- cystectomy and urinary diversion for bladder cancer. J Clin Oncol
national expert panel. Ann Oncol 2008;19:420–32. 2014;32:3291–8.

You might also like