You are viewing a javascript disabled version of the site. Please enable Javascript for this site to function properly.
Go to headerGo to navigationGo to searchGo to contentsGo to footer
In content section. Select this link to jump to navigation

Current Management of Localized Muscle-Invasive Bladder Cancer: A Consensus Guideline from the Genitourinary Medical Oncologists of Canada

Abstract

BACKGROUND:

Despite recent advances in the management of muscle-invasive bladder cancer (MIBC), treatment outcomes remain suboptimal, and variability exists across current practice patterns.

OBJECTIVE:

To promote standardization of care for MIBC in Canada by developing a consensus guidelines using a multidisciplinary, evidence-based, patient-centered approach who specialize in bladder cancer.

METHODS:

A comprehensive literature search of PubMed, Medline, and Embase was performed; and most recent guidelines from national and international organizations were reviewed. Recommendations were made based on best available evidence, and strength of recommendations were graded based on quality of the evidence.

RESULTS:

Overall, 17 recommendations were made covering a broad range of topics including pathology review, staging investigations, systemic therapy, local definitive therapy and surveillance. Of these, 10 (59% ) were level 1 or 2, 7 (41% ) were level 3 or 4 recommendations. There were 2 recommendations which did not reach full consensus, and were based on majority opinion. This guideline also provides guidance for the management of cisplatin-ineligible patients, variant histologies, and bladder-sparing trimodality therapy. Potential biomarkers, ongoing clinical trials, and future directions are highlighted.

CONCLUSIONS:

This guideline embodies the collaborative expertise from all disciplines involved, and provides guidance to further optimize and standardize the management of MIBC.

INTRODUCTION

Urothelial carcinoma of the bladder (UCB) is the most common malignancy of the urinary tract [1], and the 5th most common cancer in Canada, accounting for an estimated 11,800 new cases and 2,500 deaths in 2019 [2]. Most patients present with non-muscle invasive bladder cancer (NMIBC), but 15–25% will have muscle-invasive (≥pT2) bladder cancer (MIBC) [3, 4]. Outcomes of MIBC remain poor [5]. However, increased utilization of perioperative chemotherapy [6] and a growing emphasis on a multidisciplinary and patient-centred approach will likely translate into improved overall outcomes.

Genitourinary Medical Oncologists of Canada (GUMOC) is a non-governmental organization comprised of Canadian medical oncologists specializing in genitourinary cancers. The main objective of this consensus guideline is to summarize the current evidence and develop evidence-based recommendations to promote standardization of care for patients with MIBC in Canada. The target audience of this guideline is any clinician treating MIBC. It may also be referenced by patients, caregivers and regulatory agencies. The recommendations are intended to provide general guidance with a focus on the use of systemic therapy, but are not meant to supersede the clinical discretion of a qualified clinician for individual patients. The guideline is current at the time of publication.

METHODOLOGY

The guideline review committee co-chairs outlined and approved the key topics to be included in this guideline. The target population were patients with MIBC planned for curative intent therapy. The main focus of this guideline was on the use of of systemic therapy given in the perioperative or concurrent settings. The primary outcome of interest was the impact of various approaches on overall survival (OS). Particular topics such as the management of patients who are cisplatin-ineligible and those with variant histologies, which are more controversial topics, were also addressed. Discussions on baseline evaluations optimal local definitive therapy, and surveillance strategies were also included based on multidisciplinary input.

To ensure multidisciplinary representation, Canadian experts in medical oncology, radiation oncology, uro-oncology, and pathology were included on the guideline panel. All authors agreed to disclose any perceived and actual competing interests during the guideline development process. There were no direct financial competing interests specific to any recommendations in this guideline.

Guideline development was based on a comprehensive literature search of PubMed, Medline, and Embase for best available evidence published in the English language since January 1, 1980. The search strategy involved the keywords: “bladder cancer”, “urothelial carcinoma”, “transitional cell carcinoma”, “invasive” and “muscle invasive”. Bibliographies of review articles were searched for relevant articles not captured by our search strategy. Phase III clinical trials published in the form of proceedings from major international oncology conferences (European Society for Medical Oncology [ESMO] and American Society of Clinical Oncology [ASCO]) were also included. Most recent recommendations from ASCO [7, 8], European Association of Urology (EAU) [9, 10], National Comprehensive Network Cancer (NCCN) [11], International Bladder Cancer Network [12, 13], International Consultation on Urological Diseases [ICUD] [14–16], Canadian Urological Association (CUA) [17] and Canadian provincial organizations (British Columbia, Alberta Health Services, Cancer Care Ontario [CCO] [18–20]) were sought for each topic.

Recommendations were drafted by DMJ based on the best available evidence, and sent to all coauthors for written feedback. Each recommendation along with its evidence grading were developed through an iterative consensus process. All coauthors submitted written input. If consensuswas reached, no additional modifications were made. For topics where disagreements were raised, a voting systemwas used to collect written feedback from all coauthors. Expert opinions of the majority (>50% of coauthors) were presented as consensus, with level of consensus (marked by #) and opposing arguments included in full text to ensure a balanced discussion was presented.

To enhance the critical appraisal process, both the ICUD [21] (based on the Oxford Centre for Evidence-Based Medicine Levels of Evidence) and GRADE [22, 23] (the Grading of Recommendations Assessment, Development and Evaluation) classifications were used to rate the quality of the evidence. Strength of the recommendations were graded according to definitions proposed by ICUD [21].

EPIDEMIOLOGY AND RISK FACTORS

According to the 2019 Canadian Cancer Society report, 5-year OS rates for NMIBC, MIBC, regional (node positive), and distant disease were 95%, 69%, 35%, and 5% respectively [2]. The 5-year OS rates of patients with MIBC treated between 1994 and 2008 in Ontario ranges from 30 to 40% [24]. More contemporary data from the US show 5-year OS rates ranging between 50 and 60% [25]. Adverse prognostic factors include lymphovascular invasion [26], hydronephrosis [27, 28], multifocal disease [29], and variant histology [30]. If left untreated, patients with MIBC have a median survival of less than 10 months [31, 32].

Bladder cancer incidence increases with age. It is three times more common in men, and occurs more commonly in developed countries [33]. The median age at diagnosis is 69 years in men and 71 in women [34, 35]. Established risk factors are summarized in Table 2. Smoking is by far the most common and important risk factor accounting for approximately half of all cases [33]. Female gender is often associated with aggressive disease biology [35], advanced stage at presentation [36], and delayed referrals [37]. There is conflicting data however on whether women have inferior survival compared to men after adjusting for age and stage [24, 36, 38–41].

Table 1

Levels of Evidence and Grades of Recommendation

Levels of Evidence
ICUD classification
  Level 1Meta-analysis of RCTs or a good quality RCT
  Level 2Low-quality RCT or meta-analysis of good-quality prospective cohort studies
  Level 3Good-quality retrospective case-control studies or case series
  Level 4Expert opinion based on “first principles” or bench research, not on evidence
GRADE classification
  High qualityFurther research is very unlikely to change our confidence in the estimate of effect
  Moderate qualityFurther research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate
  Low qualityFurther research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate
  Very low qualityAny estimate of effect is very uncertain
Grades of Recommendation (ICUD classification)
  Grade AUsually consistent level 1 evidence
  Grade BConsistent level 2 or 3 evidence or “majority evidence” from RCTs
  Grade CLevel 4 evidence, “majority evidence” from level 2 or 3 studies, expert opinion
Grade DNo recommendation possible because of inadequate or conflicting evidence

RCT, randomized controlled trial.

Table 2

Established risk factors associated withbladder cancer

Established risk factorsRisk estimate [References]
Tobacco smokingHR 2.2 –4.1 [33]
Male genderAge-adjusted IRR 3.8 [293]
Exposure to arsenic in drinking waterRR 3.6 [294]
Occupational exposure to aromatic amines, polycyclic aromatic hydrocarbons, and chlorinated hydrocarbons; for example in paint, dye, metal, and petroleum productsOR 1.0 –3.4 [33, 295]
Chronic hair dye exposure in women with N-acetyltransferase 2 (NAT2) slow acetylation phenotypeOR 7.3 [296]
Lower urinary tract symptoms, chronic urinary retentionRR 1.6 [297]
Prior cyclophosphamide chemotherapyHR 1.4 [298]
Prior pelvic radiotherapy*Age-adjusted IRR 1.6 [299]
Chronic inflammation and urinary schistosomiasis in endemic areas*OR 1.7 [300]

Abbreviations: HR, hazard ratio; IRR, incidence rate ratio; OR, odds ratio; RR, relative risk. *specifically associated with squamous cell carcinoma in addition to urothelial carcinoma of the bladder.

PATHOLOGY

Diagnostic confirmation of muscle invasion (T2) is obtained by transurethral resection of bladder tumor (TURBT) and cystoscopic examination under anesthesia. Inter-observer variation of staging based on TURBT specimens may be considerable [42–44]. Therefore, review by an experienced GU pathologist is advised whenever possible, in line with prior Canadian recommendations [45]. Tumor grade should be reported according to the latest WHO 2016 criteria [46].

Approximately, 10–25% of bladder cancers have a component of variant histology [47–50]. The World Health Organization and International Consultation on Bladder Cancer recognizes more than 10 unique histologic variants in bladder cancer [46, 51], summarized in Table 3. Variant histology includes any malignancy other than pure urothelial histology, such as urothelial with divergent differentiation, urothelial with mixed nonurothelial histology, or pure nonurothelial variant histology. Pure variant histology in particular may confer a distinct clinical behavior and tends to have a high propensity for relapse [52–54]. Variant histology is often challenging to identify on TURBT specimens [48, 55]. As a result, pathology review by an experienced GU pathologist can be informative, and lead to major changes in management [43, 44].

Table 3

World Health Organization’s classification of invasive tumors of the urothelial tract

Pathology
infiltrating urothelial carcinoma
  Nested, including large nested
  Microcystic
  Micropapillary
  Lymphoepithelioma-like
  Plasmacytoid/signet ring cell/diffuse
  Sarcomatoid
  Giant cell
  Poorly differentiated
  Lipid-rich
  Clear cell
Squamous cell neoplasms
Glandular neoplasms, including adenocarcinoma
Urachal carcinoma
Tumours of Mullerian type, including clear cell carcinoma
Neuroendocrine tumors
Melanocytic tumors
Mesenchymal tumors
Urothelial tract hematopoietic and lymphoid tumors
Miscellaneous tumors

STAGING CLASSIFICATION

MIBC is clinically staged according to the Tumor, Node, Metastasis (TNM) system (Table 4). The most recent American Joint Committee on Cancer (AJCC) 8th edition reclassified nodal (cN) staging based on the number and location of involved regional lymph

Box 1.

Recommendations for Pathology Review

Pathology review of TURBT specimens used to diagnose MIBC by dedicated GU pathologists is recommended (Level 3, moderate quality, Grade C).

nodes [56]. Regional lymph nodes including perivesical, obturator, internal and external iliac, or sacral lymph nodes are designated as cN1 or cN2 disease (Table 4). Common iliac nodes are designated as regional (cN3) disease. Although previously classified as stage IV metastatic disease [57], regional lymph node involvement is now designated as stage III disease which has important treatment implications.

Table 4

American Joint Committee on Cancer for Staging of UBC, 8th Edition

Primary Tumor (T)
TxPrimary tumor cannot be assessed
T0No evidence of primary tumor
TaPapillary noninvasive carcinoma
TisCarcinoma in situ
T1Tumor invades lamina propria
T2Tumor invades the muscularis propria
  pT2aTumor invades superficial muscularis propria (inner half)
  pT2bTumor invades deep muscularis propria (outer half)
T3Tumor invades perivesical soft tissue
  pT3aTumor invades perivesical soft tissue microscopically
  pT3bTumor invades perivesical soft tissue macroscopically (extravesical mass)
T4Tumor invades adjacent organs
  T4aTumor invades directly into prostatic stroma, seminal vesicles, uterus, vagina
  T4bTumor invades pelvic wall, abdominal wall
Regional Lymph Node (N)
NxLymph nodes cannot be assessed
N0No lymph node metastasis
N1Single regional lymph node metastasis in the true pelvis (perivesical, obturator, internal and external iliac, or sacral lymph node)
N2Multiple regional lymph node metastasis in the true pelvis
N3Lymph node metastasis to the common iliac lymph nodes
Distant Metastasis (M)
MxDistant metastases cannot be assessed
M0No distant metastasis
M1Distant metastasis
  M1aDistant metastasis limited to lymph nodes beyond the common iliacs
  M1bNon-lymph-node distant metastases
Stage
0Ta or Tis N0M0
  0aTaN0M0
  0isTis N0M0
IT1N0M0
IIT2N0M0
IIIAT3 or T4a N0M0, T1–T4a N1M0
IIIBT1–T4a N2-3M0
IVAT4b N0M0 or Any T any N M1a
IVBAny T any N M1b

DIAGNOSTIC INVESTIGATIONS

Patient evaluation

A full history, physical exam and routine laboratory evaluation (including a complete blood count, kidney function tests, liver function tests, and alkaline phosphatase) should be performed prior to curative therapy. Baseline evaluation should include bladder function, performance and nutritional status, medical comorbidities including hearing impairments, prior operations and procedural complications, current medications, family history and presence of any risk factors.

According to ASCO recommendations, comprehensive geriatric assessment may be considered in patients over 65 years of age to identify vulnerabilities or geriatric impairments not routinely captured by oncology assessments [16, 58]. Smoking cessation should also be discussed, as cigarette smoking can reduce response to chemotherapy, increase surgical complication rates [59], and increase risk of developing a second urothelial primary.

Clinical staging

The goal of staging is to assess the extent of local disease, rule out distant metastases, and accurately select patients for curative-intent therapy. There is considerable variation in staging investigations used to assess MIBC [60]. Inadequate staging is common, and may adversely impact outcomes [61].

Contrast-enhanced computed tomography (CT) is limited in local tumor staging due to an inability to adequately evaluate the depth of bladder wall invasion [62]. Up to half of patients with MIBC are under-staged by CT compared to pathologic staging at the time of cystectomy [61]. Magnetic resonance imaging (MRI) has superior soft tissue contrast resolution [63, 64], however is not routinely performed due to its cost and limited availability. More recently, multiparametric MRI and the Vesical Imaging-Reporting and Data System (VI-RADS) have shown promise in improving detection of muscle invasion [65, 66], however further prospective and multicentre studies are needed. Notably, 2 –4% of patients with UCB will also develop upper tract disease, thus evaluation of the entire urothelial tract with intravenous contrast and delayed images is also important [67].

For nodal staging, contrast-enhanced CT of the abdomen and pelvis is the current standard [68]. For distant staging, data comparing chest CT versus chest xray is lacking, however chest CT offers significantly higher sensitivity in detecting pulmonary malignancy (metastatic or primary) [69–71]. MIBC has a high propensity for distant metastases, and a strong association with smoking history which also predisposes patients to developing primary bronchogenic carcinoma. Chest CT is now endorsed as a routine staging modality for MIBC by EAU [9], ICUD [72], ASCO [8] and CCO [20] (especially in smokers), as well as the CUA [17].

Conventional positron emission tomography (PET) scan is of limited value for assessing local stage due to the pooled activity of excreted 18 fluorodeoxyglucose (FDG) in the bladder, which interferes with visualization of the primary tumor. The role of FDG-PET in distant staging remains undefined, as current landmark trials evaluating curative-intent therapy of MIBC predate its use as a staging modality. Clinical trials testing the utility of FDG-PET are ongoing (NCT02462239). Staging with FDG-PET in addition to conventional CT imaging can lead to change in treatment intent from curative to palliative in 10–20% of patients, however it is unknown whether these changes translate into significant improvement in outcomes [73–75]. Patients with distant disease found on FDG-PET that is occult on conventional imaging should be carefully discussed in a multidisciplinary setting.

Bone scans should be limited to patients with suspicious bony lesions on staging imaging, symptomatic bone pain and/or elevated serum alkaline phosphatase, as routine scintigraphy has been shown to affect therapeutic decision-making in only 1% of MIBC patients [8, 76, 77]. Brain metastases are rare, however baseline brain imaging should be considered in the presence of neurologic symptoms or neuroendocrine variant histology.

Box 2.

Recommendations for Diagnostic Investigations

Complete staging investigations should include CT chest, abdomen and pelvis with intravenous contrast if no contra-indications, or MRI of abdomen and pelvis plus non-contrast CT chest if contrast administration is prohibited (Level 3, moderate quality, Grade C).

SYSTEMIC THERAPY

Neoadjuvant chemotherapy (NAC)

MIBC is a chemo-sensitive disease with high propensity for distant relapse, likely due to presence of micro-metastatic disease at presentation [78–80]. This provides the rationale for using chemotherapy to maximize chance of cure. The goal of neoadjuvant chemotherapy (NAC) is to eradicate micro-metastases and achieve pathologic complete response (pCR), which is associated with improved overall survival.

Neoadjuvant chemotherapy has several advantages over adjuvant chemotherapy including the ability to assess disease response as well as better tolerability due to absence of postoperative complications and/or reduced performance status. Cisplatin-based NAC has a pCR rate of 30 – 40% [81, 82]. The 5-year cancer-specific survival rate for NAC-responders (<ypT2) reaches 90%, compared to 30–40% for non-responders [83–85]. Importantly NAC does not increase surgical morbidity [84, 87, 88].

Select landmark publications of NAC in MIBC are summarized in Table 5. The Advanced Bladder Cancer (ABC) Meta-analysis Collaboration in 2005 reported a significant 5% absolute survival benefit at 5 years [89]. Cisplatin-based combination NAC such as MVAC (methotrexate, vinblastine, doxorubicin and cisplatin), dose-dense (dd)MVAC, and GC (gemcitabine and cisplatin) are strongly recommended, similar to other international guideline recommendations [8, 9, 11]. Single agent platinum has not shown benefit [90]. ddGC is not recommended due to potentially increased rates of cardiovascular toxicity and lack of prospective data [91].

Table 5

Efficacy data in select landmark publications of neoadjuvant chemotherapy in MIBC

Publication, YearPhasenMIBC stageExperimental ArmControl ArmDefinitive Local TherapyMedian FUPCROS
BA06 30894, 1999 [121, 301]III976Clinical T2 grade 3, T3, or T4a and N0 (65%) /Nx, M0Neoadjuvant CMV* q21d×3 cyclesObservationRC or radiation or both8.0 years32.5% vs 12.3%36% vs 30%, HR 0.84 at 10 years, p = 0.037
SWOG 8710, 2003 [84]III317Clinical T2–T4 N0M0Neoadjuvant MVACˆ q28d×3 cyclesObservationRC8.7 years38% vs 15%, p < 0.00157% vs 43% at 5 years, p = 0.06
ABC Meta-analysis, 2003, 2005 [302]3005Clinical T2–T4aNeoadjuvant chemotherapyObservationRC or radiation or both6.2 years50% vs 45%, HR 0.86 at 5 years, p = 0.003
Winquist Meta-analysis, 2004[303]3315Clinical T2–T4aNeoadjuvant chemotherapyObservationRC or radiation or both55% vs 50%, HR 0.90, p = 0.02
Yin Meta-analysis, 2016[170]3285Clinical T2–T4aPlatinum-based neoadjuvant chemotherapyObservationRC or radiation or bothHR 0.87, p = 0.004

pCR, pathologic complete response; RC, radical cystectomy. *Methotrexate 30 mg/m2 and vinblastine 4 mg/m2 day 1/8, cisplatin 100 mg/m2 day 2, and folinic acid 15 mg day 2/9. ˆmethotrexate 30 mg/m2 day 1/15/22, vinblastine 3 mg/m2 day 2/15/22, adriamycin 30 mg/m2 and cisplatin 70 mg/m2 day 2.

The optimal NAC regimen remains controversial. Most commonly used regimens are ddMVAC, MVAC, and GC which are based on level II evidence. Neoadjuvant ddMVAC or accelerated MVAC with G-CSF prophylaxis is associated with a shorter time to surgery than classic MVAC, and a more favorable toxicity profile in two phase II trials [92, 93]. Rates of pCR were 38% and 26% respectively. Although comparative trials are lacking, these results support ddMVAC as the preferred regimen over classic MVAC. GC has only been tested in comparative trials in metastatic UCB, showing similar efficacy and a more favorable safety profile compared to MVAC [94]. Extrapolated to the neoadjuvant setting, GC has become a commonly accepted NAC regimen [81, 95–99]. SWOG S1314 was a phase II trial which randomized MIBC patients to neoadjuvant GC versus ddMVAC [100]. This trial was not designed to compare efficacy of the two regimens but rather to determine the utility of a gene expression model-based biomarker approach in predicting pCR. In this trial, GC and ddMVAC yielded comparable rates of pCR (35% and 32% respectively) and downstaging to ≤pT1 (50% and 56% respectively). Mature OS data is still pending at this time. Other studies have shown neoadjuvant GC have similar pCR rates (20–25%) as MVAC [81, 98, 101] and slightly lower pCR rates than ddMVAC (30–40%) [92, 93, 99, 102, 103]. Survival outcomes of neoadjuvant GC and MVAC/ddMVAC are likely similar [98, 99, 103].

Restaging imaging should be performed at the end of NAC prior to local definitive therapy. Restaging cystoscopy can be considered for two indications: 1) to further assess disease status if clinically indicated [86], and 2) to add fiducial markers such as injected lipiodol to facilitate image-guided radiotherapy for patients who are planned to receive bladder-sparing trimodality therapy (TMT) [104, 105]. If using GC or standard dose MVAC, mid-treatment imaging may be used to rule out disease progression during NAC, however it is not standard practice [17]. Locally progressive disease or unacceptable toxicity at any point should trigger a discussion regarding immediate RC. Following NAC, local definitive therapy should occur within 4–6 weeks if possible. Up to 10 weeks between NAC and RC should represent the maximal target time interval limit, as longer intervals may compromise survival outcomes [106–109].

Despite level I evidence, less than 25% of patients receive cisplatin-based NAC [110–115], likely due to age/baseline frailty/comorbidities [97, 116], inability to predict response to NAC at the outset, risk of delay in local definitive therapy in non-responders, and a perceived marginal therapeutic benefit. Significant systematic variation in NAC utilization rates also exist [117]. In settings where a multidisciplinary approach is used, rates of NAC use are higher, up to 50% [118, 119]. This highlights the importance of ongoing multidisciplinary collaboration, patient and provider education. Over the years, NAC utilization rates have steadily increased [6, 120], which is anticipated to translate into improved outcomes.

It is important to note that in patients who are cisplatin-eligible, NAC should also be considered prior to TMT with concurrent chemoradiotherapy (see section 8). The goals of NAC prior to TMT remain similar - to eliminate micro-metastatic disease and achieve pCR. In the BA06 30894 trial, neoadjuvant CMV reduced the risk of death by 20% in patients who received radiation alone and 26% in patients who received RC [121]. In another Danish trial, the addition of NAC to radiotherapy alone in 153 patients improved median OS from 16.3 to 19.2 months, although statistical significance was not reached [122]. One would speculate that patients treated with TMT may also derive similar benefit from NAC. In the large BC2001 phase III trial evaluating concurrent chemoradiotherapy, use of NAC did not impact the benefit of concurrent 5-fluorouracil plus mitomycin, and no significant increase in late toxicity was observed [123]. Radiation Therapy Oncology Group (RTOG) 89-03 was a phase III trial published in 1998 which randomized patients to neoadjuvant CMV (without growth factor support and modern antiemetics at the time) followed by TMT versus TMT alone [124]. The trial was powered to detect a 15% difference in absolute survival, which greatly exceeded the observed survival benefit in RC trials [84]. It closed prematurely after 123 patients were randomized (target accrual was 174 patients) due to increased rates of sepsis and neutropenia. NAC completion rate was only 67% , which significantly limited statistical power [124]. Only two cycles of NAC were used, which likely limited the impact on OS [84, 121]. RTOG 89-03, perhaps not surprisingly, did not detect improved locoregional control, distant control, or OS with the addition of NAC, and dampened earlier enthusiasm for using NAC prior to TMT. Meta-analyses suggest NAC improves survival outcomes regardless of whether patients received TMT or surgery, although differences were not statistically significant [90, 125]. Investigators at the Princess Margaret Cancer Centre and other centres recently reported encouraging outcomes and tolerability of NAC prior to TMT [126, 127]. It is important to note that historically TMT was reserved for patients who were ineligible for RC (and often ineligible for cisplatin-based NAC). By comparison, younger and fitter patients opting for bladder preservation in the contemporary setting are more likely to tolerate and benefit from NAC. While currently there is no proven benefit for NAC prior to TMT, trials are ongoing now evaluating the use of NAC in this setting (NCT03620435, NCT03768570). Further data are warranted to evaluate the use of this approach.

Prior landmark NAC trials excluded patients with lymph node positive disease as stage IV metastatic disease under the previous AJCC staging system [57]. The AJCC 8th Edition now designates N1–3 disease as stage III [56], highlighting their superior outcomes compared to other patients with metastatic disease. Two phase II trials evaluating neoadjuvant ddMVAC included patients with N1 disease [92, 93]. Large retrospective series suggest potential benefit even in N2–N3 disease, yielding pCR rates of 15–27% [128, 129] and an absolute 20% improvement in OS at 3 years [98, 128, 129]. Based on current data, lymph node positive MIBC should be managed with induction systemic therapy, and subsequent local definitive therapy in responders. Cisplatin-based chemotherapy should be given for 4 cycles. However, 6 cycles were administered in previous trials evaluating patients with node only metastatic disease [130]. Based on expert opinion, in select patients with node positive disease, 6 cycles of induction chemotherapy could be considered if a patient is tolerating treatment well and there is ongoing disease response [128, 131]. Whether 6 cycles instead of 4 cycles improves outcomes in node positive MIBC unknown, and requires further study.

Box 3.

Recommendations for Neoadjuvant Systemic Therapy

Neoadjuvant cisplatin-based combination chemotherapy is recommended for cisplatin-eligible patients with cT2-T4aN0* bladder cancer planned for radical cystectomy (Level 1, high quality, Grade A).

Neoadjuvant cisplatin-based combination chemotherapy can be considered prior to trimodality therapy in cisplatin-eligible patients (Level 2, moderate quality, Grade B)#.

*Node positive (N1-3) disease can also be considered for induction chemotherapy and subsequent definitive local therapy. #Level of consensus: Level 2, moderate quality, Grade B – 65%; Level 3/4, low quality, Grade C – 25%; Level 1, high/moderate quality, Grade A – 5%; no response – 5%.

#Level of consensus: Level 2, moderate quality, Grade B –65%; Level 3/4, low quality, Grade C –25%; Level 1, high/moderate quality, Grade A –5%; no response –5%.

Complete clinical response following NAC

pCR at the time of RC is achieved in 30– 40% of patients treated with cisplatin-based NAC [84, 98]. The standard of care for patients who achieve complete clinical response (CR, defined as absence of disease on urinary cytology, TURBT and imaging) following NAC is to proceed with planned local definitive therapy. Retrospective data have reported 5-year disease-free survival reaching 50–80% in these patients opting for surveillance [132–135], however supporting evidence is limited and discrepancy between CR defined by clinical staging and pCR limits the reliability of CR [86, 136]. Ongoing work is exploring a risk adapted approach of selecting certain patients for active surveillance (NCT02710734, NCT03609216). However, such strategies should only be performed in the setting of a clinical trial.

Box 7.

Recommendations for Definitive Local Therapy

Radical cystectomy with bilateral pelvic lymphadenectomy should be offered to patients with resectable MIBC (Level 1, high quality, Grade A).

Although direct comparisons are lacking, trimodality therapy should be offered as an accepted and reasonable alternative to radical cystectomy in select patients ideally meeting the following criteria: small (<5 cm) and unifocal tumor, absence of extensive CIS, no hydronephrosis, good bladder function, no prior pelvic radiotherapy, and compliance for regular cystoscopy surveillance (Level 2, high quality, Grade B).

Patients who are not candidates for or who decline radical cystectomy should be offered trimodality therapy if feasible (Level 2, high quality, Grade B).

Options for radiosensitizing agents in trimodality therapy include cisplatin (either 100 mg/m2 every 3 weeks [Level 1, high quality, Grade A], or 35–40 mg/m2weekly [Level 2, moderate quality, Grade B]), 5-FU with mitomycin (Level 1, high quality, Grade A), or single agent weekly gemcitabine 100 mg/m2 (Level 2, moderate quality, Grade B).

Adjuvant Chemotherapy (AC)

To date, no prospective trial has demonstrated any significant difference in OS comparing NAC to AC in MIBC [137]. AC utilization rates remain low at approximately 20% [6, 120]. About a third of patients may be precluded from AC due to complications from RC and/or reduced performance status [138]. AC trials have historically been difficult to accrue, and were often underpowered, making the overall data in AC less robust than NAC. At least 11 AC trials have been conducted, with only 3 of which demonstrating a similar survival benefit to NAC [139–142] (Appendix 1). While the ABC meta-analyses in 2005 reported insufficient evidence to guide treatment decisions [89], several recent meta-analyses have suggested an OS benefit with AC [143, 144].

Recently, a large retrospective study from the National Cancer Data Base showed potential OS benefit (HR 0.70) in high risk patients (pT3/T4 or node positive disease) [145]. The hazard ratio mirrors data from the Ontario Cancer Registry (HR 0.71) and other reports (HR 0.74–0.77) [146, 147]. Therefore, for patients with high risk disease who did not receive NAC, AC should be considered. Patients should start AC as soon as they are medically fit to do so, ideally within 12 weeks of surgery [113]. However, delay of more than 12 weeks from surgery should not be the sole reason to exclude AC. The benefit of AC in variant histology is unclear, and should be discussed at experienced centres [148].

The use of AC after cisplatin-based NAC is not recommended, given conflicting results from observational series and lack of prospective data [30, 149–154]. Biologically, it is presumed tumor cells resistant to cisplatin-based NAC will also be refractory to AC.

Overall, given the lack of robust data in the adjuvant setting, clinical trial participation is encouraged for patients with high risk MIBC. Trials evaluating adjuvant immune checkpoint inhibitors are underway (Table 7). Adjuvant radiotherapy is an area also requiring further study.

Box 4.

Recommendations for Adjuvant Systemic Therapy

In cisplatin-eligible patients who did not receive cisplatin-based neoadjuvant chemotherapy and have muscle-invasive disease on surgical pathology, adjuvant cisplatin-based chemotherapy should be considered (Level 2, moderate quality, Grade B). Patients with pT3/pT4 or pN+ disease has the highest level of evidence for adjuvant chemotherapy.

Cisplatin-ineligible patients

Standard ineligibility criteria for cisplatin-based chemotherapy were proposed by Galsky et al. in 2011 and are shown in Table 6 [155]. Unfortunately, nearly half of all patients fit for RC are deemed cisplatin-ineligible [116], likely due to baseline frailty and comorbidities inherent to the MIBC patient population, as well as obstructive uropathy from direct disease invasion. Malignant urinary obstruction should be decompressed which may allow more patients to receive cisplatin-based NAC. Percutaneous nephrostomy tube insertion is preferred over stenting, given the latter’s lower success rates [156] and risk of upper tract recurrence associated with stenting [157].

Table 6

Cisplatin-ineligibility criteria in metastatic bladder cancer proposed by Galsky et al. 2011

Any one of the following:
WHO or ECOG PS ≥2, or Karnofsky PS≤60–70%
Creatinine clearance (calculated or measured) <60 mL/min
CTCAE v4 grade ≥2 peripheral neuropathy
CTCAE v4 grade ≥2 audiometric hearing loss
NYHA Class III/IV heart failure

WHO, World Health Organization; ECOG, Eastern Cooperative Oncology Group; PS, performance status; CTCAE, Common Terminology Criteria for Adverse Events; NYHA, New York Heart Association.

Renal function is often a limiting factor for cisplatin-based therapy, and can be estimated bythe Cockcroft Gault, Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) and Modification of Diet in Renal Disease (MDRD) equations. The latter two may be more accurate in patients with cancer [158–161]. Timed urine collections, although preferable, are infrequently utilized due to inconvenience and cost. In patients with impaired renal function (renal clearance ≥50 mL/min), split dose GC (cisplatin 35 mg/m2 on day 1 and day 8) [81, 91, 162–165] and dose reduction (25–50%) of standard GC (cisplatin 70 mg/m2 every 3 weeks) are options, although data supporting these approaches remains limited [166–168]. For patients with baseline renal function <50 ml/min, generally the use of cisplatin-based NAC is not supported by adequate safety data. However multidisciplinary discussion including onco-nephrology at experienced centers and informed discussion with patients are recommended. The risk of permanent renal injury and limited accuracies of existing tools for estimating renal function are important to highlight. Administering cisplatin in patients with renal function of <40 ml/min is not recommended given lack of safety data. Some reports suggest cisplatin-based NAC can be administered to patients undergoing hemodialysis with appropriate dose reduction [169], and these patients should be treated at experienced centers.

Carboplatin-based perioperative chemotherapy should not be offered, given the lack of evidence for a survival benefit [170], unnecessary toxicity and risk of delaying local definitive therapy. Multiple studies have shown inferior outcomes with carboplatin- compared to cisplatin-based chemotherapy in UCB [171–174, 99]. The SWOG S0219 study evaluated neoadjuvant carboplatin, gemcitabine and paclitaxel showed that 60% of patients with clinical T0 disease had residual cancer at cystectomy, and survival rates were only 60% at 2 years [136].

Box 5.

Recommendations for Systemic Therapy in cisplatin-ineligible patients

Patients with calculated renal function of 50–60 ml/min, or measured creatinine clearance of 50–60 ml/min using a 24 hour urine collection, who are otherwise cisplatin-eligible may be considered for neoadjuvant or adjuvant cisplatin-based chemotherapy (Level 2, low quality, Grade C).

In cisplatin-ineligible MIBC patients, definitive local therapy alone (cystectomy or trimodality therapy) or enrollment in a clinical trial is recommended (Level 2, moderate quality, Grade B).

Variant histology

Given the rarity of variant histology, data guiding the management of these tumors are limited to observational studies only. Variant histologies generally have worse prognosis and more upstaging at the time of surgery compared to conventional UCB [175–177]. Pure variant histology may have inferior OS compared to mixed variant histologies treated with RC [54].

The benefit of cisplatin-based NAC in variant histology is not clearly established. Neuroendocrine (or small cell carcinoma) variants have a high tendency for systemic relapse. Tumors with pure, mixed neuroendocrine histology, and neuroendocrine differentiation should be treated with neoadjuvant cisplatin etoposide similar to small cell lung cancer, which leads to pathologic downstaging in 60–80% of patients [178, 179]. Management of neuroendocrine variants is outlined in a separate consensus guideline from GUMOC [180]. Pure non-bilharzial squamous cell carcinomas responds poorly to NAC and radiation, thus should be treated with upfront RC [181–185]. Research is ongoing to identify distinct clinical phenotypes and novel therapeutic targets [186]. On the other hand, urothelial tumors with squamous and glandular differentiation often respond to NAC [187–192]. The SWOG S8710 trial evaluating neoadjuvant MVAC included 59 patients with mixed nonurothelial histologies (such as squamous or glandular differentiation) and showed a significant benefit in OS in this cohort [187]. Many experts on this panel considered these data as moderate or high level evidence supporting the use of NAC in this setting. Bladder adenocarcinoma is rare, and surgery is the main treatment modality for both urachal and non-urachal adenocarcinomas [193–196]. Urachal adenocarcinoma is covered in a separate review by the CUA and GUMOC [197]. Supporting evidence for OS benefit of NAC is limited or has conflicting results in micropapillary [198–202], plasmacytoid [203–208], nested [209, 210], and sarcomatoid variants [177, 211–213], thus recommendations cannot be made in these settings at this time. Local definitive therapy may be the most important component of curative therapy. Data is limited with respect to the benefit of TMT for tumors of variant histology, and therefore no definitive recommendations were made. RC should be considered for these cases. If feasible, multidisciplinary discussion at experienced academic centres and enrollment in clinical trials should be sought for these patients.

DEFINITIVE LOCAL THERAPY

Radical cystectomy and bilateral pelvic lymph node dissection

Following NAC, RC with bilateral pelvic lymph node dissection (PLND) remains the historical standard local definitive therapy approach in patients with MIBC [7, 214–216]. PLND should include removal of pelvic nodes up to the common iliac bifurcation (internal, external, and obturator nodes), although the optimal extent of lymphadenectomy is not established [217–221]. Some authors have proposed thresholds of 10 to 16 removed lymph nodes (as a surrogate for surgical quality) for adequate pathological staging and optimal survival outcomes [45, 218, 222–224]. In patients with pelvic or retroperitoneal node positive disease, the role of postchemotherapy lymph node resection may be limited [131].

Box 6.

Recommendations for Systemic Therapy for Variant Histology

Patients with resectable pure squamous cell carcinoma and adenocarcinoma of the bladder should be considered for upfront surgery (Level 3, low quality, Grade C).

Cisplatin-based neoadjuvant chemotherapy is recommended for resectable pure neuroendocrine, neuroendocrine histology mixed with urothelial carcinoma, and tumors with neuroendocrine differentiation (Level 2, moderate quality, Grade C).

Cisplatin-based neoadjuvant chemotherapy should be considered for urothelial tumors with squamous and glandular differentiation (Level 3, low quality, Grade C).#

#Level of consensus: Level 3, low quality, Grade C –60%; Level 2, moderate/high quality, Grade B –20%; Level 1, high quality, Grade A –15%; no response –5%.

Trimodality therapy (TMT)

Multiple bladder preservation options exist including radical TURBT, radiotherapy alone, and “tetramodal” therapy consisting of TURBT, chemoradiation and partial cystectomy [225]. However TMT is the most favored approach as it offers the highest curative potential with the highest level of supporting data.

TMT consists of maximal TURBT followed by definitive chemoradiotherapy [123, 226, 227], with salvage cystectomy reserved for localized bladder relapse. Radiotherapy is typically given with total doses of 60 Gy (2 Gy/day) or above delivered to the bladder and/or pelvic lymph nodes, or 55 Gy (2.75 Gy/day) delivered to the bladder alone. Concurrent systemic therapy improves local control [123, 228], and possibly OS [229–231], although no standard regimen exists. Concurrent cisplatin was used in the RTOG, National Cancer Institute of Canada (NCIC) and Trans Tasman Radiation Oncology Group (TROG) trials, and is the most commonly used radiosensitizer [124, 227, 228, 230, 232–235]. Concurrent cisplatin can be administered as 35–40 mg/m2 weekly or 100 mg/m2 every 3 weeks. The use of concurrent 5-fluorouacil plus mitomycin C (5-fluorouracil administered as a continuous infusion at 500 mg/m2 daily on days 1 to 5 and 16–20 of radiotherapy, mitomycin administered as an intravenous bolus dose of 12 mg/m2 on day 1) is supported by a large randomized phase III trial [123]. Low dose gemcitabine [236–239] is another alternative especially in more frail patients. Comparative trials are needed to elucidate the optimal radiosensitizer in TMT.

Adequate level 1 evidence directly comparing RC with TMT is lacking after the SPARE trial failed to accrue [240], RC remains the most commonly used treatment approach and the historical standard [241, 242]. For patients who are ineligible for RC, or desire bladder preservation, TMT is the preferred bladder-sparing approach. Radiotherapy alone in the treatment of localized MIBC is only acceptable in extremely frail patients who are ineligible for both RC and TMT.

Ideal candidates for TMT are patients with 1) cT2 with tumors <5 cm [243], 2) solitary tumors without extensive carcinoma in situ (CIS) [244–246], 3) minimal to no hydronephrosis [124, 247], 4) good bladder function [243], 5) completion of maximal TURBT without visible residual tumor [229, 246, 248, 249], and 6) agreeable to long-term surveillance with regular cystoscopy and imaging [243]. TMT is likely equivalent to RC in these patients, as shown by data with long-term follow up [229, 243, 250–255], and up to 89% of patients successfully retain their native bladders [243]. Short-term treatment mortality likely favors TMT, especially in elderly patients [256]. Patients should be carefully selected for TMT (and NAC) through a multidisciplinary approach in experienced centres [257]. A multidisciplinary bladder clinic has been shown to significantly impact treatment selection and has potential to improve patient outcomes [119].

POST-TREATMENT SURVEILLANCE

Surveillance enables early detection of recurrences and curative-intent salvage therapy. It also serves to monitor the development of a second primary and metachronous upper tract malignancy after local definitive therapy which occurs in 5% of patients [258]. Currently, the optimal frequency, modality and duration of surveillance is subject to debate given lack of robust evidence [8, 9,12, 16, 79, 259–261]. Suggested surveillance protocols are included to emphasize the overall importance of surveillance, provide some general guidance and promote care standardization, however these recommendations are based on very limited data. Risk of disease recurrence and patient preference should also be considered. Ultimately prospective clinical trials are needed to elucidate the optimal surveillance approach in MIBC.

CT of the abdomen and pelvis with or without intravenous contrast together with excretory imaging can be employed to evaluate both upper tract and abdominopelvic recurrences, and is the preferred imaging modality for surveillance. The optimal surveillance chest imaging (chest X ray or CT chest) is unknown.

Following RC, up to 20% of patients develop local recurrence, and 50% develop distant recurrences (most commonly to bone, distant lymph nodes, and lung) [78, 262]. Most recurrences occur within the first 2–3 years. Late recurrences (or development of a new primary) can rarely occur [79, 260, 263], although there is scant data to guide surveillance beyond 5 years. A risk-adapted strategy based on pathological stage can be employed, although further prospective studies are needed for validation [12, 79, 259–261, 264]. Multivariate nomograms including additional prognostic factors may be more accurate in predicting an individual’s survival following RC, compared to pathologic stage alone [265].

Local recurrence after TMT can be either NMIBC or MIBC. Recurrent NMIBC should be managed according to usual guidelines, including TURBT and adjuvant intravesical therapy as indicated. Recurrent MIBC and some higher risk NMIBC can be successfully salvaged with RC. Therefore, all patients require close cystoscopic surveillance post TMT. Based on published TMT surveillance protocols [123, 243, 248, 252] and extrapolating from the NMIBC setting, cystoscopy and urine cytology are generally recommended every 3–6 months for the first 3 years followed by every 6 months for two additional years, and annually thereafter. Delayed local recurrence (or development of a new primary) at 10 years have been reported in up to 10% of patients following TMT, therefore long term cystoscopic surveillance may be warranted [250, 252].

Box 8.

Recommendations for Surveillance

Surveillance following radical cystectomy should include history, physical exam, blood work, and surveillance CT abdomen pelvis and chest imaging (Level 4, very low quality, Grade C).

Based on expert consensus, a suggested surveillance protocol following radical cystectomy includes CT abdomen pelvis and chest imaging every 3–6 months for the first 3 years, every 6 months for 2 additional years, then annually thereafter(Level 4, very low quality, Grade C). A risk based surveillance approach can be considered.

Based on expert consensus, a suggested surveillance protocol following trimodality therapy includes urine cytology, cystoscopy, CT abdomen pelvis and chest imaging every 3–6 months for the first 3 years, every 6 months for 2 additional years, then annually thereafter (Level 4, very low quality, Grade C).

FUTURE DIRECTIONS

Immune checkpoint inhibitors (CPIs) and targeted therapies

The landscape of UCB has changed rapidly in recent years with the use of CPIs, with multiple agents approved since 2016. Pembrolizumab was shown to have a 3-month OS advantage over chemotherapy in the second line metastatic setting by the Keynote 045 phase III trial [266]. In the neoadjuvant setting, pembrolizumab (PURE-01 trial) and atezolizumab (ABACUS trial) have phase II data demonstrating pCR rates of 42% and 29% respectively, with acceptable toxicity profile [267, 268]. In the PURE-01 trial, all treated patients underwent RC. In the ABACUS trial, 2 out of 74 patients treated died prior to RC, 1 was treatment related. Another 3 had clinical deterioration, 1 experienced disease progression prior to RC. Combination CPI with nivolumab and iplimumab was evaluated in the phase Ib trial NABUCCO. Among 24 patients with clinical stage T3/4 or N + MIBC 46% achieved pCR (60% in PD-L1+, and 22% in PD-L1–group), and all underwent RC [269]. Ipilimumab, a CTLA-4 inhibitor, has also been tested as monotherapy [270]. Although 66.7% patients were downstaged at cystectomy, preoperative ipilimumab produced grade 3 toxicity in 4 out of 12 patients, and 2 experienced surgical delays due to toxicity. Durvalumab and tremelimumab was also evaluated as a neoadjuvant regimen in a single arm trial [271]. Among 35 patients, 9 (43%) achieved pCR, 14 (67%) had downstaging, 2 (7%) resulted in surgery delay for >30 days. In a phase Ib/II trial, combination pembrolizumab with chemotherapy was administered in 40 patients prior to RC [272]. There were 5 patients who did not proceed with RC (4 refused, 1 due to adverse event). Downstaging to <T2 disease occurred in 22 patients (61%), and pCR occurred in 16 patients (40%). BLASST-1 is a phase II trial evaluating combination nivolumab, which reported pCR rates of 49% [273]. These results seem to suggest that the addition of immunotherapy to standard of care NAC does not result in synergy with respect to pCR rates, however long term OS data is still awaited and phase III trials are underway. Emerging data suggest neoadjuvant CPI do not adversely affect surgical safety of RC [274].

Combination strategies with targeted therapies are also being investigated. Phase II results have been reported from durvalumab plus olaparib (NEODURVARIB trial) and nintedanib, a tyrosine kinase inhibitor, plus GC (NEO-BLADE trial), with pCR rates 50% and 37% respectively [275, 276]. The NEO-BLADE trial also reported improved OS over GC alone with HR 0.38, p = 0.018. Further randomized trials are required to further establish the role of these combination strategies in the neoadjuvant setting.

Table 7 lists currently active phase III RCTs investigating the safety and efficacy of CPI and targeted therapies in MIBC [277].

Table 7

Currently active and completed trials evaluating checkpoint inhibitors and targeted therapies in muscle invasive bladder cancer (based on search on clinicaltrials.gov on May 6, 2020 for “muscle invasive bladder cancer” and “muscle-invasive bladder cancer” start date “01/01/2010 to 05/06/2020”). Trials with recruiting centres in Canada are marked with **

NeoadjuvantCisplatin eligibleCisplatin ineligible or declinedTrial status
NCT03924895(KEYNOTE 905)**Phase IIINeoadjuvant pembrolizumab 200mg iv every 3 weeks for 3 cycles + adjuvant for 14 cyclesNoYesRecruiting
NCT03924856(KEYNOTE 866)**Phase IIINeoadjuvant GC+/–pembrolizumab 200 mg iv every 3 weeks for 4 dosesYesNoRecruiting
NCT02736266(PURE-01)Phase IINeoadjuvant pembrolizumab 200 mg iv every 3 weeks for 3 dosesYesYesRecruiting; reported
NCT03212651 (PANDORE)Phase IINeoadjuvant pembrolizumab 200 mgNoYesActive, not recruiting
NCT02690558 (LCCC 1520)Phase IINeoadjuvant pembrolizumab 200 mg IV every 3 weeks for 4 cycles + GCYesNoRecruiting
NCT02365766 (HCRN GU14-188)Phase I/IINeoadjuvant pembrolizumab 200 mg IV every 3 weeks for 4 cycles + GC or gemcitabineYesYesActive, not recruiting; reported
NCT03832673 (PECULIAR)Phase IINeoadjuvant pembrolizumab 200 mg IV every 3 weeks for 3 cycles + epacadostat 300 mg BID po every 28 days for 3 cyclesYesYesNot yet recruiting
NCT03978624 (LCCC1827)Phase IINeoadjuvant pembrolizumab 200 mg IV every 3 weeks for 2 doses + entinostat 5 mg po weekly for 3 weeksNoYesRecruiting
NCT03661320 (ENERGIZE)**Phase IIINeoadjuvant chemotherapy +/–nivolumab, BMS-986205; adjuvant nivolumab, BMS-986205YesNoRecruiting
NCT04209114 (CA045-009)**Phase IIINeoadjuvant nivolumab + NKTR-214NoYesRecruiting
NCT03294304 (BLASST-1)Phase IINeoadjuvant nivolumab 360 mg iv every 3 weeks + GC for 4 cyclesYesNoActive, not recruiting; reported
NCT03558087 (HCRN GU16-257)Phase IINeoadjuvant nivolumab 360 mg iv every 3 weeks + GC for 4 cycles. Maintenance nivolumab 240 mg every 2 weeks for up to 8 cyclesYesNoRecruiting
NCT03520491 (MSKCC 18-042)Phase IINeoadjuvant nivolumab 3 mg/kg every 2 weeks for 5 cycles, or nivolumab 1mg/kg + ipilimumab 3 mg/kg every 6 weeks for 2 cycles, or nivolumab 1 mg/kg + ipilimumab 3mg/kg every 3 weeks for 3 cyclesNoYesRecruiting
NCT03387761 (NABUCCO)Phase INeoadjuvant nivolumab, ipilimumab at 1 mg/kg or 3 mg/kgNoYesRecruiting; reported
NCT02845323 (J1682)Phase IINeoadjuvant nivolumab 240 mg iv + urelumab 8mgevery 2 weeks for 2 cyclesNoYesRecruiting
NCT03532451 (PrE0807)Phase INeoadjuvant nivolumab 480 mg iv+/–lirilumab 240 mg IV every 4 weeks for 2 dosesNoYesRecruiting
NCT03518320 (TAR-200-104)Phase INeoadjuvant nivolumab every week for 4 cycles + GemRIS/TAR 200NoYesActive, not recruiting
NCT02662309 (ABACUS)Phase IINeoadjuvant atezolizumab every 3 weeks for 2 cyclesNoYesActive, not recruiting; reported
NCT02451423 (UCSF 14524)Phase IINeoadjuvant atezolizumab 1200 mg every 3 weeks for 2 or 3 dosesNoYesRecruiting
NCT04289779 (ABATE)Phase IINeoadjuvant atezolizumab1200 mg IV every 3 weeks for 3 cycles + cabozantinib 40 mg orally daily for 3 cyclesNoYesRecruiting
NCT02989584 (MSKCC 16-1428)Phase I/IINeoadjuvant atezolizumab + GC or 4 cyclesYesNoRecruiting
NCT03732677 (NIAGARA)**Phase IIINeoadjuvant GC+/–durvalumab, adjuvant durvalumabYesNoRecruiting
NCT03234153 (NITIMIB)Phase IINeoadjuvant durvalumab 1500 mg iv + tremelimumab 75 mg iv every 4 weeks for 4 cyclesNoYesActive, not recruiting
NCT03472274 (DUTRENEO)Phase IINeoadjuvant durvalumab 1500 mg + tremelimumab 75mg every 4 weeks×3 cycles or cisplatin-based chemoYesNoRecruiting
NCT02812420 (NCI-2016-01147)Phase INeoadjuvant durvalumab 1500 mg + tremelimumab 75 mg on weeks 1 and 5NoYesActive, not recruiting; reported
NCT03534492 (NEODURVARIB)Phase IINeoadjuvant durvalumab 1500 mg iv every 4 weeks + olaparib 300 mg BID for up to 2 monthsYesNoCompleted; reported
NCT03773666 (BLASST-2)Phase INeoadjuvant durvalumab every 2 weeks+/–OleclumabNoYesRecruiting
NCT03674424 (AURA)Phase IINeoadjuvant avelumab 10 mg/kg every 2 weeks+/–ddMVAC/GC/GPYesYesRecruiting
NCT03473730 (MDACC 2017-0688)Phase INeoadjuvant daratumumab IV weekly for 4 weeksNoYesRecruiting
NCT04099589 (NCC2121)Phase IINeoadjuvant toripalimab 240 mg injection ever 3 weeks for 2–4 cycles + GCYesNoRecruiting
NCT03288545(EV-103)Phase INeoadjuvant enfortumab vedotin (cohort H)NoYesRecruiting
Neoadjuvant enfortumab vedotin + pembrolizumab (cohort J)
AdjuvantCisplatin eligibleCisplatin ineligible or declinedTrial status
NCT03244384 (AMBASSADOR)Phase IIIAdjuvant pembrolizumab every 3 weeks for up to 18 cycles, or observationNoYesRecruiting
NCT02632409 (Checkmate 274)Phase IIIAdjuvant nivolumabNoYesActive, not recruiting
NCT02450331 (IMvigor010)Phase IIIAdjuvant atezolizumab 1200 mg every 3 weeks up to 1 yearYesYesActive, not recruiting
Trimodality TherapyCisplatin eligibleCisplatin ineligible or declinedTrial status
NCT04241185 (KEYNOTE-992)Phase IIIConcurrent pembrolizumab 400 mg every 6 weeks + cisplatin, 5FU MMC, or gemcitabineYesYesRecruiting
NCT02662062(PCR-MIB)Phase IIConcurrent pembrolizumab 200 mg every 3 weeks + cisplatin, pembrolizumab continued until 12 weeksYesYesRecruiting
NCT02621151 (NYU 15-00220)Phase IIConcurrent pembrolizumab 200 mg every 3 weeks for 3 doses + gemcitabineYesYesRecruiting
NCT02560636(PLUMMB)Phase IConcurrent pembrolizumab 100–200 mg every 3 weeks starting 2 weeks prior to radiotherapy, continued for a maximum of 12 monthsYesYesActive, not recruiting
NCT03993249 (HGCG 0000020479)Phase IIConcurrent nivolumab and standard of care chemoradiotherapyYesYesRecruiting
NCT03844256(CRIMI)Phase I/IIConcurrent nivolumab 480 mg every 4 weeks, or nivolumab 3 mg/kg and ipilimumab 1 mg/kg every 3 weeks, or nivolumab 1 mg/kg and ipilimumab 3 mg/kg every 3 weeks, combined with MMC and capecitabine. Optional nivolumab every 4 weeks for a maximum of 52 weeksYesYesRecruiting
NCT03775265(SWOG S1806)Phase IIIConcurrent atezolizumab ever 3 weeks + chemotherapy (GC or 5FU MMC). Atezolizumab continued for a maximum of 6 monthsYesYesRecruiting
NCT03620435 (ML-39576)**Phase IIConcurrent atezolizumab 1200 mg iv every 3 weeks, continued for a maximum of 1 yearYesYesRecruiting
NCT04186013 (ATEZOBLADDERPRESERVE)Phase IIConcurrent atezolizumab 1200 mg iv every 3 weeks for 6 dosesNoYesRecruiting
NCT03702179 (IMMUNOPRESERVE)Phase IIConcurrent durvalumab 75 mg plus tremelimumab 75 mg every 4 weeks for 3 dosesYesYes+Recruiting
NCT03747419 (DFCI 18-464)Phase IIConcurrent avelumab every 2 weeks for 6 dosesNoYesRecruiting
NCT03617913 (MC1752)Phase IIConcurrent avelumab every 2 weeks for 10 courses + 5FU MMC or cisplatinYesYesActive, not recruiting
NCT04073160 (TRIO Bladder)Phase INeoadjuvant durvalumab 1500 mg plus tremelimumab 75 mg every 4 weeks followed by concurrent durvalumab 1500 mg every 4 weeks, based on molecular subtypes. Durvalumab may be continued for a maximum of 1 yearYesYesNot yet recruiting
NCT03171025(NEXT)Phase IIAdjuvant nivolumab iv 480 mg every 4 weeks for a maximum of 12 months.YesYesRecruiting
NCT03697850(BladderSpar)Phase IIAdjuvant atezolizumab 1200 mg ever 3 weeks for a maximum of 12 months
NCT03768570(CCTG BL13)**Phase IIAdjuvant durvalumab 1500 mg every 4 weeks for a maximum of 12 monthsYesYes+Recruiting

ddMVAC, dose dense methotrexate, vinblastine, doxorubicin, and cisplatin; GC, gemcitabine cisplatin; GP, gemcitabine paclitaxel; 5FU, 5-fluorouracil; MMC, mitomycin. +except poor ECOG and neuropathy ≥Grade 2.

Biomarkers

There is an urgent need to develop predictive biomarkers in MIBC to improve treatment selection [278–281]. In general, molecular subtyping of MIBC reveals basal, luminal (similar to breast cancer), and neuroendocrine-like subtypes [282]. Several molecular classifications exist, and an international consensus was recently published [283]. Basal subtype seems to derive the most benefitfrom NAC [279, 284]. Luminal subtype has lower risk of upstaging at surgery compared with non-luminal tumors [285]. Genomic alterations in DNA-repair pathways including ERCC2, ERBB2, ATM, RB1 and FANCC also seem to enrich response to NAC [281, 286, 287]. A predictive gene expression model (COXEN) that compares a tumor’s gene expression to established signatures which correlate with response failed to predict response to NAC in a prospective trial [100].

With respect to local definitive therapy, low expression of MRE11 (a protein involved in double-stranded DNA damage repair and cell cycle checkpoint) and high expression of TIP60 (tat-interactive protein 60 kDa) have been associated with improved outcomes with RC [288, 289]. Molecular determinants of response to radiotherapy may include miR-23a and miR-27a [290], genomically unstable and squamous cell cancer-like tumor subtypes [291], and tumors with higher immune infiltration [292].

Currently, no predictive biomarker have been rigorously validated for routine clinical use at this time. However, individual molecular testing and biomarker-driven precision oncology hold promise and may become standard of care for MIBC in the future.

SUMMARY

MIBC has seen many treatment advances in the last several years. Improving utilization of cisplatin-based perioperative therapy to address the risk of systemic relapse through a multidisciplinary effort is critical in optimizing outcomes of this lethal disease. The management of cisplatin-ineligible patients remains an area of high unmet need. Many questions still remain unanswered with regards to patient selection, predictive biomarkers, and the role of immunotherapy in MIBC. Enrollment of patients in clinical trials is encouraged whenever possible.

ACKNOWLEDGMENTS

The authors have no acknowledgements.

FUNDING

The authors report no funding.

AUTHOR CONTRIBUTIONS

Conception: DMJ, SSS

Performance of work: DMJ, SSS

Interpretation or analysis of data: all authors

Writing the article: all authors

ETHICAL CONSIDERATIONS

This paper does not present any primary results of the studies it described herein. As such, it is exempt from any requirement for Institutional Review Board approval

CONFLICT OF INTEREST

There are no direct conflict of interests from any authors. Indirect conflicts of interests are as follows.

DMJ: honoraria and/or consulting fees from Bayer.

CC: advisory role for Janssen, Astellas, Pfizer, Ipsen, BMS, Roche, Merck, Bayer, Eisai; Educational travel grants from Pfizer and Sanofi Genzyme.

MK: honoraria and/or consulting fees from Janssen, Ipsen, Astellas, BMS, Merck, AstraZeneca, Bayer; travel support from Novartis.

LAW: advisory boards from Pfizer, BMS, Ipsen, Merck – no personal financial compensation; research funding from Pfizer, BMS, Merck, Roche, Ipsen, AZ – financial compensation to my institution.

GSK: advisory boards for Ferring, Janssen, Bayer, Astellas, Merck, Roche, Thearalase; Investigator for trials from Merck, Astra Zeneca, BMS, Abbvie, Theralase, Sesen Bio.

NSM: consultant/advisory role for Merck, Astellas, Pfizer, Astra Zeneca, Janssen, Sanofi.

PCB: member of an advisory board or equivalent with AbbVie, Asieris, AstraZeneca, Astellas, Bayer, Biosyent, BMS, H3-Biomedicine, Janssen, Merck, Roche, Sanofi, Urogen; member of a Speakers bureau for AbbVie, Biosyent, Janssen, Ferring, TerSera, Pfizer; grant(s) or honorarium received from Decipher Biosciences, iProgen, Sanofi, Bayer; currently participating in or have participated in a clinical trial within the past two years with Genentech, Janssen, BMS, Astellas, Sitka, MDx Health, AstraZeneca; patent shared with Decipher.

SSS: advisory board member for Astellas, AstraZeneca, Bayer, Janssen, Merck, and Roche; has participated in several pharma-supported clinical trials.

REFERENCES

[1] 

Siegel RL , Miller KD , Jemal A . Cancer statistics, 2018. CA Cancer J Clin. (2018) ;68: :7–30. doi:10.3322/caac.21442

[2] 

Canadian Cancer Statistics Advisory Committee. Canadian Cancer Statistics 2019. Can Cancer Soc 2019:cancer.ca/Canadian-Cancer-Statistics-2019-EN.

[3] 

Canadian Cancer Statistics Advisory Committee. Canadian Cancer Statistics. A 2018 Special Report On Cancer Incidenc by Stage. 2018.

[4] 

Boustead GB , Fowler S , Swamy R , Kocklebergh R , Hounsome L . Stage, grade and pathological characteristics of bladder cancer in the UK: British Association of Urological Surgeons (BAUS) Urological Tumour registry. BJU Int. (2014) ;113: :924–30. doi:10.1111/bju.12468

[5] 

Zehnder P , Studer UE , Skinner EC , Thalmann GN , Miranda G , Roth B , et al. Unaltered oncological outcomes of radical cystectomy with extended lymphadenectomy over three decades. BJU Int. (2013) ;112: :51–8. doi:10.1111/bju.12215

[6] 

Booth CM , Karim S , Brennan K , Siemens DR , Peng Y , Mackillop WJ . Perioperative chemotherapy for bladder cancer in the general population: Are practice patterns finally changing? Urol Oncol Semin Orig Investig. (2018) ;36: :89.e13–89.e20. doi:10.1016/j.urolonc.2017.11.015

[7] 

Milowsky MI , Bryan Rumble R , Booth CM , Gilligan T , Eapen LJ , Hauke RJ , et al. Guideline on muscle-invasive and metastatic bladder cancer (European Association of Urology guideline): American Society of Clinical Oncology clinical practice guideline endorsement. J Clin Oncol. (2016) ;34: :1945–52. doi:10.1200/JCO.2015.65.9797

[8] 

Chang SS , Bochner BH , Chou R , Dreicer R , Kamat AM , Lerner P , et al. Treatment Of Non-Metastatic Muscle-Invasive Bladder Cancer: AUA/ASCO/ASTRO/SUO Guideline. J Urol. (2017) ;198: :552–9.

[9] 

Witjes JA , Bruins M , Compérat E , Cowan N , Gakis G , Hernández V , et al. EAU Guidelines on Muscle-Invasive and Metastatic Bladder Cancer. Eur Assoc Urol Guidel. 2018;ISBN 978-9: https://uroweb.org/guideline/bladder-cancer-muscle.

[10] 

Horwich A , Babjuk M , Bellmunt J , Bruins HM , Reijke TM De , Santis M De , et al. EAU – ESMO consensus statements on the management of advanced and variant bladder cancer — an international collaborative multi-stakeholder effort : under the auspices of the EAU and ESMO Guidelines Committees. Ann Oncol. 2019;0:1-31. doi:10.1093/annonc/mdz296

[11] 

Flaig TW , and NCCN Bladder Cancer Panel. Bladder Cancer. NCCN Clin Pract Guidel Oncol. 2019;version 4. https://www.nccn.org/professionals/physician_gls/default.aspx#site [Accessed Sep 22, 2019]

[12] 

Zuiverloon TCM , van Kessel KEM , Bivalacqua TJ , Boormans JL , Ecke TH , Grivas PD , et al. Recommendations for follow-up of muscle-invasive bladder cancer patients: A consensus by the international bladder cancer network. Urol Oncol. 2018. doi:10.1016/j.urolonc.2018.01.014

[13] 

Black PC , Goebell PJ , Kamat AM , Nawroth R , Schmitz-Dräger BJ . Editorial: Managing locally advanced bladder cancer. Third International Bladder Cancer Network seminars series. Urol Oncol Semin Orig Investig. 2018:1-2. doi:10.1016/j.urolonc.2017.11.006

[14] 

Alanee S , Alvarado I , Paari C , Rajeev M , Kenneth K . Update of the International Consultation on Urological Diseases on bladder cancer 2018 : non-urothelial cancers of the urinary bladder. World J Urol. (2019) ;37: :107–14. doi:10.1007/s00345-018-2421-5

[15] 

Fernández MI , Brausi M , Clark PE , Cookson MS , Grossman HB , Khochikar M , et al. Epidemiology, prevention, screening, diagnosis, and evaluation: update of the ICUD–SIU joint consultation on bladder cancer.World J Urol. 2018. doi:10.1007/s00345-018-2436-y

[16] 

Société Internationale d’Urologie. Bladder Cancer: A Joint SIU-ICUD International Consultation. Lisbon, Portugal, October 19, 2017. Montreal: 2017.

[17] 

Kulkarni G , Black P , Sridhar S , Kapoor A , Zlotta AR , Shayegan B , et al. Canadian Urologic Association Guideline: Muscle-Invasive Bladder Cancer. Can Urol Assoc J. (2019) ;13: :230–8.

[18] 

BC Cancer Guidelines: Bladder 2008: http://www.bccancer.bc.ca/health-professionals/cli.

[19] 

Alberta Health Services Guidelines: Muscle Invasive and Locally Advanced/Metastatic Bladder Cancer 2013: https://www.albertahealthservices.ca/assets/info/h.

[20] 

Finelli A , Brown J , Flood T , Kulkarni G , Hotte S . Cancer Care Ontario Bladder Cancer Guideline: An Endorsement of the 2017 American Urological Association Treatment of Non-Metastatic Bladder Cancer: AUA, ASCO, ASTRO, SUO Guideline 2018.

[21] 

Abrams P , Khoury S . International Consultation on Urological Diseases: Evidence-Based Medicine Overview of the Main Steps for Developing and Grading Guideline Recommendations. Neurourol Urodyn. (2010) ;29: :116–8. doi:10.1002/nau

[22] 

Balshem H , Helfand M , Sch HJ , Oxman AD , Kunz R , Brozek J , et al. GRADE guidelines: 3. Rating the quality of evidence. J Clin Epidemiol. (2011) ;64: :401–6. doi:10.1016/j.jclinepi.2010.07.015

[23] 

Guyatt GH , Oxman AD , Vist GE , Kunz R , Falck-Ytter Y , Alonso-Coello P , et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. Bmj. (2008) ;336: :924–6.

[24] 

Booth CM , Siemens DR , Li G , Peng Y , Kong W , Berman DM , et al. Curative Therapy for Bladder Cancer in Routine Clinical Practice: A Poplation-based Outcomes Study. Clin Oncol. (2014) ;26: :506–14. doi:10.1016/j.clon.2014.05.007

[25] 

Cahn DB , Handorf EA , Ghiraldi EM , Ristau BT . Contemporary Use Trends and Survival Outcomes in Patients Undergoing Radical Cystectomy or Bladder-Preservation Therapy for Muscle-Invasive Bladder Cancer. Cancer. (2017) ;123: :4337–45. doi:10.1002/cncr.30900

[26] 

Lotan Y , Gupta A , Shariat SF , Palapattu GS , Vazina A , Karakiewicz PI , et al. Lymphovascular invasion is independently associated with overall survival, cause-specific survival, and local and distant recurrence in patients with negative lymph nodes at radical cystectomy. J Clin Oncol. (2005) ;23: :6533–9. doi:10.1200/JCO.2005.05.516

[27] 

Bartsch GC , Kuefer R , Gschwend JE , de Petriconi R , Hautmann RE , Volkmer BG . Hydronephrosis as a Prognostic Marker in Bladder Cancer in a Cystectomy-Only Series. Eur Urol. (2007) ;51: :690–8. doi:10.1016/j.eururo.2006.07.009

[28] 

Canter D , Guzzo TJ , Resnick MJ , Brucker B , Vira M , Chen Z , et al. Hydronephrosis Is an Independent Predictor of Poor Clinical Outcome in Patients Treated for Muscle-Invasive Transitional Cell Carcinoma With Radical Cystectomy. Urology. (2008) ;72: :379–83. doi:10.1016/j.urology.2008.03.053

[29] 

Holzbeierlein JM , Lopez-Corona E , Bochner BH , Herr HW , Donat SM , Russo P , et al. Partial Cystectomy: a Contemporary Review of the Memorial Sloan-Kettering Cancer Center Experience and Recommendations for Patient Selection. J Urol. (2004) ;172: :878–81. doi:10.1097/01.ju.0000135530.59860.7d

[30] 

Kassouf W , Agarwal PK , Grossman HB , Leibovici D , Munsell MF , Siefker-Radtke A , et al. Outcome of Patients With Bladder Cancer With pN+Disease After Preoperative Chemotherapy and Radical Cystectomy. Urology. (2009) ;73: :147–52. doi:10.1016/j.urology.2008.07.035

[31] 

Martini A , Sfakianos JP , Renstrom-Koskela L , Mortezavi A , Falagario UG , Egevad L , et al. The natural history of untreated muscle-invasive bladder cancer. BJU Int. 2019;July 16:[Epub ahead of print]. doi:10.1111/bju.14872

[32] 

Westergren DO , Gårdmark T , Lindhagen L , Chau A , Malmström P . A Nationwide Population Based Analysis of Patients with Organ Confined, Muscle Invasive Bladder Cancer Not Receiving Curative Intent Therapy in Sweden from 1997 to 2014. J Urol. 2019:doi: 10.1097/JU.0000000000000350. [Epub ahead of print]. doi:10.1097/JU.0000000000000350

[33] 

Burger M , Catto JWF , Dalbagni G , Grossman HB , Herr H , Karakiewicz P , et al. Epidemiology and risk factors of urothelial bladder cancer. Eur Urol. (2013) ;63: :234–41. doi:10.1016/j.eururo.2012.07.033

[34] 

Lynch CF , Cohen MB . Urinary System. Cancer. (1995) ;75: :316–29. doi:10.1002/9781118789568.ch5

[35] 

Scosyrev E , Noyes K , Feng C , Messing E . Sex and Racial Differences in Bladder Cancer Presentation and Mortality in the US. Cancer. (2009) ;115: :68–74. doi:10.1002/cncr

[36] 

Mitra AP , Skinner EC , Schuckman AK , Quinn DI , Dorff TB , Daneshmand S . Effect of gender on outcomes following radical cystectomy for urothelial carcinoma of the bladder: A critical analysis of 1,994 patients. Urol Oncol Semin Orig Investig. (2014) ;32: :52.e1–52.e9. doi:10.1016/j.urolonc.2013.08.007

[37] 

Aziz A , Madersbacher S , Otto W , Mayr R , Comploj E , Pycha A , et al. Comparative Analysis of Gender-Related Differences in Symptoms and Referral Patterns prior to Initial Diagnosis of Urothelial Carcinoma of the Bladder: A Prospective Cohort Study. Urol Int. (2015) ;94: :37–44. doi:10.1159/000363334

[38] 

Krimphove M , Szymaniak J , Marchese M , Tully K , D’Andrea D , Mossanen M , et al. Sex-specific Differences in the Quality of Treatment of Muscle- invasive Bladder Cancer Do Not Explain the Overall Survival. Eur Urol Focus. (2019) ;19: :30156–7. doi:10.1016/j.euf.2019.06.001

[39] 

Santos F , Dragomir A , Kassouf W , Franco E , Aprikian A . Urologist referral delay and its impact on survival after radical cystectomy for bladder cancer. Curr Oncol. (2015) ;22: :e20–26.

[40] 

Soave A , Dahlem R , Hansen J , Weisbach L . Gender-specific outcomes of bladder cancer patients: A stage-specific analysis in a contemporary, homogenous radical cystectomy cohort. Eur J Surg Oncol. (2015) ;41: :368–77. doi:10.1016/j.ejso.2014.03.003

[41] 

Dobruch J , Daneshmand S , Fisch M , Lotan Y , Noon AP , Resnick MJ , et al. Gender and Bladder Cancer: A Collaborative Review of Etiology, Biology, and Outcomes. Eur Urol. (2016) ;69: :300–10. doi:10.1016/j.eururo.2015.08.037

[42] 

Giunchi F , Panzacchi R , Capizzi E , Schiavina R , Brunocilla E , Martorana G , et al. Role of Inter-Observer Variability and Quanti fi cation of Muscularis Propria in the Pathological Staging of Bladder Cancer. Clin Genitourin Cancer. (2016) ;14: :e307–312.

[43] 

Traboulsi SL , Brimo F , Yang Y , Maedler C , Tanguay S , Aprikian AG , et al. Pathology reviewimpacts clinical management of patients with T1 - T2 bladder cancer. Can Urol Assoc J. 2017;11.

[44] 

Luchey AM , Manimala NJ , Dickinson S , Dhillon J , Agarwal G , Lockhart JL , et al. Change in Management Based on Pathologic Second Opinion among Bladder Cancer Patients Presenting to a Comprehensive Cancer Center: Implications for Clinical Practice. Urology. (2016) ;93: :130–4. doi:10.1016/j.urology.2016.01.048

[45] 

Kassouf W , Aprikian A , Saad F , Breau RH , Kulkarni G , Guttman DM , et al. Improving patient journey and quality of care: Summary from the second Bladder Cancer Canada-Canadian Urological Association- Canadian Urologic Oncology Group (BCC-CUA-CUOG) bladder cancer quality of care consensus meeting. CUAJ. (2018) ;12: :E281–97.

[46] 

Humphrey PA , Moch H , Cubilla AL , Ulbright TM , Reuter VE , Catto J . The WHO Classification of Tumours of the Urinary System and Male Genital Organs — Part B: Prostate and Bladder Tumours. Eur Urol. (2016) ;70: :106–19. doi:10.1016/j.eururo.2016.02.028

[47] 

Wasco MJ , Daignault S , Zhang Y , Kunju LP , Kinnaman M , Braun T , et al. Urothelial Carcinoma with Divergent Histologic Differentiation (Mixed Histologic Features) Predicts the Presence of Locally Advanced Bladder Cancer When Detected at Transurethral Resection. Urology. (2007) ;70: :69–74. doi:10.1016/j.urology.2007.03.033

[48] 

Cai T , Tiscione D , Verze P , Pomara G , Racioppi M , Nesi G , et al. Concordance and clinical significance of uncommon variants of bladder urothelial carcinoma in transurethral resection and radical cystectomy specimens. Urology. (2014) ;84: :1141–6. doi:10.1016/j.urology.2014.06.032

[49] 

Rink M , Robinson BD , Green DA , Cha EK , Hansen J , Comploj E , et al. Impact of histological variants on clinical outcomes of patients with upper urinary tract urothelial carcinoma. J Urol. (2012) ;188: :398–404. doi:10.1016/j.juro.2012.04.009

[50] 

Alderson M , Grivas P , Milowsky MI , Wobker SE . Histologic Variants of Urothelial Carcinoma: Morphology, Molecular Features and Clinical Implications. Bl Cancer. 2020:1-16. doi:10.3233/blc-190257

[51] 

Compérat E , Babjuk M , Algaba F , Amin M , Brimo F , Grignon D , et al. SIU –ICUD on bladder cancer: pathology. World J Urol. (2019) ;37: :41–50. doi:10.1007/s00345-018-2466-5

[52] 

Wang G , McKenney JK . Urinary Bladder Pathology. World Health Organization (WHO) Classification and American Joint Committee on Cancer (AJCC) Staging Update. Arch Pathol Lab Med. 2018:doi: 10.5858/arpa.2017-0539-RA. [Epub ahead of print]. doi:10.5858/arpa.201

[53] 

Moschini M , D’Andrea D , Korn S , Irmak Y , Soria F , Compérat E , et al. Characteristics and clinical significance of histological variants of bladder cancer. Nat Rev Urol. (2017) ;14: :651–68. doi: 10.1038/nrurol.2017.125

[54] 

Moschini M , Shariat SF , Lucianò R , D’Andrea D , Foerster B , Abufaraj M , et al. Pure but Not Mixed Histologic Variants Are Associated With Poor Survival at Radical Cystectomy in Bladder Cancer Patients. Clin Genitourin Cancer. (2017) ;15: :e603–7. doi:10.1016/j.clgc.2016.12.006

[55] 

Abd El-Latif A , Watts KE , Elson P , Fergany A , Hansel DE . The sensitivity of initial transurethral resection or biopsy of bladder tumor(s) for detecting bladder cancer variants on radical cystectomy. J Urol. (2013) ;189: :1263–7. doi:10.1016/j.juro.2012.10.054

[56] 

MB Amin, SB Edge, FL Greene, et al, eds. American Joint Commission on Cancer. AJCC Cancer Staging Manual, Eighth Edition. New York: Springer; (2017) .

[57] 

SB Edge, DR Byrd, CC Compton, AG Fritz, FL Greene, A Trotti, eds. American Joint Committee on Cancer. AJCC Cancer Staging Manual, Seventh Edition. New York: Springer; (2010) .

[58] 

Mohile SG , Dale W , Somerfield MR , Hurria A . Practical Assessment and Management of Vulnerabilities in Older Patients Receiving Chemotherapy: ASCO Guideline for Geriatric Oncology Summary. J Oncol Pract. (2018) ;14: :442–6. doi:10.1200/JOP.18.00180

[59] 

Boeri L , Soligo M , Frank I , Boorjian SA , Thompson RH , Tollefson M , et al. Cigarette smoking is associated with adverse pathological response and increased disease recurrence amongst patients with muscle-invasive bladder cancer treated with cisplatin-based neoadjuvant chemotherapy and radical cystectomy: a single-centre experien. BJU Int. (2019) ;122: :1011–9. doi:10.1111/bju.14612

[60] 

Mcinnes MDF , Siemens DR , Mackillop WJ , Peng Y , Wei S , Schieda N , et al. Utilisation of preoperative imaging for muscle-invasive bladder cancer: a population-based study. BJU Int. (2016) ;117: :430–8. doi:10.1111/bju.13034

[61] 

Bostrom PJ , van Rhijn BWG , Fleshner N , Finelli A , Jewett M , Thoms J , et al. Staging and Staging Errors in Bladder Cancer. Eur Urol Suppl. (2010) ;9: :2–9. doi:10.1016/j.eursup.2010.01.005

[62] 

Malayeri AA , Pattanayak P , Apolo AB . Imaging muscle-invasive and metastatic urothelial carcinoma. Curr Opin Urol. (2015) ;25: :441–8. doi:10.1097/MOU.0000000000000208

[63] 

Rajesh A , Sokhi HK , Fung R , Mulcahy KA , Bankart MJG . Bladder cancer: Evaluation of staging accuracy using dynamic MRI. Clin Radiol. (2011) ;66: :1140–5. doi:10.1016/j.crad.2011.05.019

[64] 

Tekes A , Kamel I , Imam K , Szarf G , Schoenberg M , Nasir K , et al. Dynamic MRI of Bladder Cancer: Evaluation of Staging Accuracy. AJR. (2005) ;184: :121–7.

[65] 

Barchetti G , Simone G , Ceravolo I , Salvo V , Campa R , Giudice F Del , et al. Multiparametric MRI of the bladder: inter-observer agreement and accuracy with the Vesical Imaging-Reporting and Data System (VI-RADS) at a single reference center. Eur Radiol. (2019) ;29: :5498–506.

[66] 

Wang H , Li S , Chen L . Multiparametric MRI for Bladder Cancer: Validation of VI-RADS for the Detection of Detrusor Muscle Invasion. Radiology.. (2019) ;291: :668–74.

[67] 

Vikram R , Sandler CM , Ng CS . Imaging and staging of transitional cell carcinoma: Part 1, lower urinary tract. Am J Roentgenol. (2009) ;192: :1481–7. doi:10.2214/AJR.08.1318

[68] 

American College of Radiology. ACR Appropriateness Criteria® Pretreatment Staging of Muscle-Invasive Bladder Cancer. 2017. https://acsearch.acr.org/docs/69370/Narrative/ [Accessed Aug 27, 2018].

[69] 

Davis D . CT Evaluation in Patients for Pulmonary with Extrathoracic Metastases Malignancy. Radiology. (1991) ;180: :1–12.

[70] 

del Ciello A , Franchi P , Contegiacomo A , Cicchetti G , Bonomo L , Larici AR . Missed lung cancer: when, where, and why? Diagn Interv Radiol. (2017) ;23: :118–26. doi:10.5152/dir.2016.16187

[71] 

Hafeez S , Huddart R . Advances in bladder cancer imaging. BMC Med. (2013) ;11: :1–10.

[72] 

Fernández MI , Brausi M , Clark PE , Cookson MS , Grossman HB , Khochikar M , et al. Epidemiology, prevention, screening, diagnosis, and evaluation: update of the ICUD - SIU joint consultation on bladder cancer. World J Urol. (2019) ;37: :3–13. doi:10.1007/s00345-018-2436-y

[73] 

Apolo AB , Riches J , Schöder H , Akin O , Trout A , Milowsky MI , et al. Clinical value of fluorine-18 2-fluoro-2-deoxy-D-glucose positron emission tomography/computed tomography in bladder cancer. J Clin Oncol. (2010) ;28: :3973–8. doi:10.1200/JCO.2010.28.7052

[74] 

Kollberg P , Almquist H , Bläckberg M , Cronberg C , Gudjonsson S , Kleist J , et al. [18F] Fluorodeoxyglucose –positron emission tomography / computed tomography improves staging in patients with high-risk muscle-invasive bladder cancer scheduled for radical cystectomy. Scand J Urol. (2015) ;49: :296–301. doi:10.3109/21681805.2014.990053

[75] 

Mertens LS , Fioole-bruining A , Vegt E , Vogel W V , Rhijn BW Van , Horenblas S . Impact of 18 F-fluorodeoxyglucose (FDG) -positron-emission tomography / computed tomography (PET/CT) on management of patients with carcinoma invading bladder muscle. BJU Int. (2013) ;112: :729–34. doi:10.1111/bju.12109

[76] 

Brismar J , Gustafson T . Bone scintigraphy in staging of bladder carcinoma. Acta Radiol. (1988) ;29: :251–2. doi:10.1177/028418518802900223

[77] 

Eberhardt SC , Oto A , Akin O , Alexander LF , Allen BC , Coakley F V , et al. ACR Appropriateness Criteria â Pretreatment Staging of Muscle-Invasive Bladder Cancer. J Am Coll Radiol. (2018) ;15: :S150–159. doi:10.1016/j.jacr.2018.03.020

[78] 

Mari A , Campi R , Tellini R , Gandaglia G , Albisinni S , Abufaraj M , et al. Patterns and predictors of recurrence after open radical cystectomy for bladder cancer: a comprehensive review of the literature. World J Urol. (2018) ;36: :157–70. doi:10.1007/s00345-017-2115-4

[79] 

Yafi FA , Aprikian AG , Fradet Y , Chin JL , Izawa J , Rendon R , et al. Surveillance guidelines based on recurrence patterns after radical cystectomy for bladder cancer: the Canadian Bladder Cancer Network experience. BJU. (2012) ;110: :1317–23. doi:10.1111/j.1464-410X.2012.11133

[80] 

Moschini M , Karnes RJ , Sharma V , Gandaglia G , Fossati N , Dell’Oglio P , et al. Patterns and prognostic significance of clinical recurrences after radical cystectomy for bladder cancer: A 20-year single center experience. Eur J Surg Oncol. (2016) ;42: :735–43. doi:10.1016/j.ejso.2016.02.011

[81] 

Dash A , Pettus IV JA , Herr HW , Bochner BH , Dalbagni G , Donat SM , et al. A role for neoadjuvant gemcitabine plus cisplatin in muscle-invasive urothelial carcinoma of the bladder: A retrospective experience. Cancer. (2008) ;113: :2471–7. doi:10.1002/cncr.23848

[82] 

Waingankar N , Jia R , Marqueen KE , Audenet F , Sfakianos JP , Mehrazin R , et al. The impact of pathologic response to neoadjuvant chemotherapy on conditional survival among patients with muscle-invasive bladder cancer. Urol Oncol Semin Orig Investig. (2019) ;37: :572.e21–572.e28. doi:10.1016/j.urolonc.2019.04.027

[83] 

Buttigliero C , Tucci M , Vignani F , Scagliotti G V . , Di Maio M. Molecular biomarkers to predict response to neoadjuvant chemotherapy for bladder cancer. Cancer Treat Rev. (2017) ;54: :1–9. doi:10.1016/j.ctrv.2017.01.002

[84] 

Grossman HB , Natale RB , Tangen CM , Speights VO , Vogelzang NJ , Trump DL , et al. Neoadjuvant Chemotherapy plus Cystectomy Compared with Cystectomy Alone for Locally Advanced Bladder Cancer. N Engl J Med. (2003) ;349: :859–66.

[85] 

Zargar H , Zargar-shoshtari K , Lotan Y , Spiess PE , Black P . Final Pathological Stage after Neoadjuvant Chemotherapy and Radical Cystectomy for Bladder Cancer d Does pT0 Predict Better Survival than pTa / Tis / T1 ? J Urol. (2016) ;195: :886–93. doi:10.1016/j.juro.2015.10.133

[86] 

Reese AC , Ball MW , Gandhi N , Gorin MA , Netto GJ , Bivalacqua TJ , et al. The Utility of an Extensive Postchemotherapy Staging Evaluation in Patients Receiving Neoadjuvant Chemotherapy for Bladder Cancer. Urology. (2014) ;84: :358–64. doi:10.1016/j.urology.2014.03.040

[87] 

Sherif A , Holmberg L , Rintala E , Mestad O , Nilsson J . Neoadjuvant Cisplatinum Based Combination Chemotherapy in Patients with Invasive Bladder Cancer: A Combined Analysis of Two Nordic Studies. Eur Urol. (2004) ;45: :297–303. doi:10.1016/j.eururo.2003.09.019

[88] 

Gandaglia G , Popa I , Abdollah F , Schiffmann J , Shariat SF , Briganti A , et al. The Effect of Neoadjuvant Chemotherapy on Perioperative Outcomes in Patients Who Have Bladder Cancer Treated with Radical Cystectomy: A Population-based Study. Eur Urol. (2014) ;66: :561–8. doi:10.1016/j.eururo.2014.01.014

[89] 

Advanced Bladder Cancer (ABC) Meta-Analysis Collaborators . Adjuvant chemotherapy in invasive bladder cancer: A systematic review and meta-analysis of individual patient data. Eur Urol. (2005) ;48: :189–99. doi:10.1016/j.eururo.2005.04.005

[90] 

Advanced Bladder Cancer (ABC) Meta-Analysis Collaborators . Neoadjuvant chemotherapy in invasive bladder cancer: a systematic review and meta-analysis. Lancet. (2003) ;361: :1927–34. doi:10.1016/S0140-6736(03)13580-5

[91] 

Anari F , O’Neill J , Choi W , Chen DYT , Haseebuddin M , Kutikov A , et al. Neoadjuvant Dose-dense Gemcitabine and Cisplatin in Muscle-Invasive Bladder Cancer: Results of a Phase 2 Trial. Eur Urol Oncol. (2018) ;1: :54–60. doi:10.1016/j.euo.2018.02.007

[92] 

Plimack ER , Hoffman-Censits JH , Viterbo R , Trabulsi EJ , Ross EA , Greenberg RE , et al. Accelerated methotrexate, vinblastine, doxorubicin, and cisplatin is safe, effective, and efficient neoadjuvant treatment for muscle-invasive bladder cancer: Results of a multicenter phase II study with molecular correlates of response and toxicity. J Clin Oncol. (2014) ;32: :1895–901. doi:10.1200/JCO.2013.53.2465

[93] 

Choueiri TK , Jacobus S , Bellmunt J , Qu A , Appleman LJ , Tretter C , et al. Neoadjuvant dose-dense methotrexate, vinblastine, doxorubicin, and cisplatin with pegfilgrastim support in muscle-invasive urothelial cancer: Pathologic, radiologic, and biomarker correlates. J Clin Oncol. (2014) ;32: :1889–94. doi:10.1200/JCO.2013.52.4785

[94] 

Von Der Maase H , Sengelov L , Roberts JT , Ricci S , Dogliotti L , Oliver T , et al. Long-term survival results of a randomized trial comparing gemcitabine plus cisplatin, with methotrexate, vinblastine, doxorubicin, plus cisplatin in patients with bladder cancer. J Clin Oncol. (2005) ;23: :4602–8. doi:10.1200/JCO.2005.07.757

[95] 

Yeshchina O , Badalato GM , Wosnitzer MS , Hruby G , Roychoudhury A , Benson MC , et al. Relative efficacy of perioperative gemcitabine and cisplatin versus methotrexate, vinblastine, adriamycin, and cisplatin in the management of locally advanced urothelial carcinoma of the bladder. Urology. (2012) ;79: :384–90. doi:10.1016/j.urology.2011.10.050

[96] 

Gandhi NM , Baras A , Munari E , Faraj S , Reis LO , Liu JJ , et al. Gemcitabine and cisplatin neoadjuvant chemotherapy for muscle-invasive urothelial carcinoma: Predicting response and assessing outcomes. Urol Oncol Semin Orig Investig. (2015) ;33: :204e1–7. doi:10.1016/j.urolonc.2015.02.011

[97] 

Apolo AB , Kim JW , Bochner BH , Steinberg SM , Bajorin DF , Kevin Kelly W , et al. Examining the management of muscle-invasive bladder cancer by medical oncologists in the United States. Urol Oncol Semin Orig Investig. (2014) ;32: :637–44. doi:10.1016/j.urolonc.2013.12.012

[98] 

Zargar H , Espiritu PN , Fairey AS , Mertens LS , Dinney CP , Mir MC , et al. Multicenter assessment of neoadjuvant chemotherapy for muscle-invasive bladder cancer. Eur Urol. (2015) ;67: :241–9. doi:10.1016/j.eururo.2014.09.007

[99] 

Peyton CC , Tang D , Reich RR , Azizi M , Chipollini J , Pow-sang JM , et al. Downstaging and Survival Outcomes Associated With Neoadjuvant Chemotherapy Regimens Among Patients Treated With Cystectomy for Muscle-Invasive Bladder Cancer. JAMA Oncol. 2018:doi: 10.1001/jamaoncol.2018.3542. [Epub ahead of print]. doi:10.1001/jamaoncol.2018.3542

[100] 

Flaig TW , Tangen CM , Daneshmand S , Alva AS , Lerner SP , Scott M . SWOG S1314: A randomized phase II study of co-expression extrapolation (COXEN) with neoadjuvant chemotherapy for localized, muscle-invasive bladder cancer. J Clin Oncol. 2019;37:no. 15 suppl (May 20, 2019) 4506-4506. doi:10.1200/JCO.2019.37.15

[101] 

Kim HS , Jeong CW , Kwak C , Kim HH , Ku JH . Pathological T0 following cisplatin-based neoadjuvant chemotherapy for muscle-invasive bladder cancer: A network meta-analysis. Clin Cancer Res. (2016) ;22: :1086–94. doi:10.1158/1078-0432.CCR-15-1208

[102] 

Zargar H , Shah JB , van Rhijn BW , Daneshmand S , Bivalacqua TJ , Spiess PE , et al. Neoadjuvant Dose Dense MVAC versus Gemcitabine and Cisplatin in Patients with cT3-4aN0M0 Bladder Cancer Treated with Radical Cystectomy. J Urol. (2018) ;199: :1452–8. doi:10.1016/j.juro.2017.12.062

[103] 

Galsky MD , Pal SK , Chowdhury S , Harshman LC , Crabb SJ , Wong YN , et al. Comparative effectiveness of gemcitabine plus cisplatin versus methotrexate, vinblastine, doxorubicin, plus cisplatin as neoadjuvant therapy for muscle-invasive bladder cancer. Cancer. (2015) ;121: :2586–93. doi:10.1002/cncr.29387

[104] 

Hafeez S , Warren-oseni K , Dcr T , Mcnair HA , Hansen VN , Jones K , et al. Prospective Study Delivering Simultaneous Integrated High-dose Tumor Boost (< /=70 Gy) With Image Guided Adaptive Radiation Therapy for Radical Treatment of Localized Muscle-Invasive Bladder Cancer. Int J Radiat Oncol Biol Phys. (2016) ;94: :1022–30. doi:10.1016/j.ijrobp.2015.12.379

[105] 

Beulens AJW , Toorn P Van Der , Wildt MJ De , Scheepens WA . High-precision Bladder Cancer Irradiation in the Elderly: Clinical Results for a Plan-of-the-day Integrated Boost Technique with Image Guidance Using Lipiodol Markers. Eur Urol Oncol. (2018) ;2: :39–46. doi:10.1016/j.euo.2018.08.012

[106] 

Alva AS , Tallman CT , He C , Hussain MH , Hafez K , Montie JE , et al. Efficient Delivery of Radical Cystectomy After Neoadjuvant Chemotherapy for Muscle-Invasive Bladder Cancer. A Multidisciplinary Approach. Cancer. (2012) ;118: :44–53. doi:10.1002/cncr.26240

[107] 

Mmeje CO , Benson CR , Nogueras-gonz GM , Jayaratna IS , Gao J , Siefker-radtke AO , et al. Determining the optimal time for radical cystectomy after neoadjuvant chemotherapy. BJU Int. (2018) ;122: :89–98. doi:10.1111/bju.14211

[108] 

Boeri L , Soligo M , Frank I , Boorjian SA , Thompson RH , Tollefson M , et al. Delaying Radical Cystectomy After Neoadjuvant Chemotherapy for Muscle-invasive Bladder Cancer is Associated with Adverse Survival Outcomes. Eur Urol Oncol. (2019) ;2: :390–6.

[109] 

Gore JL , Lai J , Setodji CM , Litwin MS , Saigal CS , the Urologic Diseases in America Project. Mortality increases when radical cystectomy is delayed more than 12 weeks: results from a SEER-medicare analysis. Cancer. (2009) ;115: :988–96. doi:10.1002/cncr.24052.MORTALITY

[110] 

Fedeli U , Fedewa S a , Ward EM . Treatment of muscle invasive bladder cancer: evidence from the National Cancer Database, 2003 to 2007. J Urol. (2011) ;185: :72–8. doi:10.1016/j.juro.2010.09.015

[111] 

Gore JL , Litwin MS , Lai J , Yano EM , Madison R , Setodji C , et al. Use of radical cystectomy for patients with invasive bladder cancer. J Natl Cancer Inst. (2010) ;102: :802–11. doi:10.1093/jnci/djq121

[112] 

Williams SB , Huo J , Chamie K , Hu JC , Giordano SH , Hoffman KE , et al. Underutilization of Radical Cystectomy Among Patients Diagnosed with Clinical Stage T2 Muscle-invasive Bladder Cancer. Eur Urol Focus. (2017) ;3: :258–64. doi:10.1016/j.euf.2016.04.008

[113] 

Booth CM , Siemens DR , Peng Y , Tannock IF , Mackillop WJ . Delivery of perioperative chemotherapy for bladder cancer in routine clinical practice. Ann Oncol. (2014) ;25: :1783–8. doi:10.1093/annonc/mdu204

[114] 

Huo J , Ray-zack MD , Shan Y , Chamie K , Boorjian SA , Kerr P , et al. Discerning Patterns and Quality of Neoadjuvant Chemotherapy Use Among Patients with Muscle-invasive Bladder Cancer. Eur Urol Oncol. 2018:doi: 10.1016/j.euo.2018.07.009. [Epub ahead of print]. doi:10.1016/j.euo.2018.07.009

[115] 

Miles BJW , Fairey AS , Eliasziw M , Estey EP . Referral and treatment rates of neoadjuvant chemotherapy in muscle-invasive bladder cancer before and after publication of a clinical practice guideline. Can Urol Assoc J. (2010) ;4: :263–7.

[116] 

Thompson RH , Boorjian SA , Kim SP , Cheville JC , Thapa P , Tarrel R , et al. Eligibility for neoadjuvant/adjuvant cisplatin-based chemotherapy among radical cystectomy patients. BJU Int. (2014) ;113: :17–21. doi: 10.1111/bju.12274

[117] 

Karim S , Siemens DR , Mackillop WJ , Krzyzanowska MK , Brennan K , Booth CM . Estimating the optimal perioperative chemotherapy utilization rate for muscle-invasive bladder cancer. Cancer Med. (2019) ;00: :1–14. doi:10.1002/cam4.2449

[118] 

Hsu T , Black PC , Chi KN , Canil CM , Eigl BJ , Kulkarni G , et al. Treatment of muscle-invasive bladder cancer in Canada: A survey of genitourinary medical oncologists and urologists. J Can Urol Assoc. (2014) ;8: :309–16. doi:10.5489/cuaj.2111

[119] 

Diamantopoulos LN , Winters BR , Grivas P , Ngo SD , Zeng J , Hsieh AC , et al. Bladder Cancer Multidisciplinary Clinic (BCMC) Model Influences Disease Assessment and Impacts Treatment Recommendations. Bl Cancer. (2019) ;5: :289–98. doi:10.3233/BLC-190239

[120] 

Reardon ZD , Patel SG , Zaid HB , Stimson CJ , Resnick MJ , Keegan KA , et al. Trends in the Use of perioperative chemotherapy for localized and locally advanced muscle-invasive bladder cancer: A sign of changing tides. Eur Urol. (2015) ;67: :165–70. doi:10.1016/j.eururo.2014.01.009

[121] 

International collaboration of trialists. International phase III trial assessing neoadjuvant cisplatin, methotrexate, and vinblastine chemotherapy for muscle-invasive bladder cancer: Long-term results of the BA06 30894 trial. J Clin Oncol. (2011) ;29: :2171–7. doi:10.1200/JCO.2010.32.3139

[122] 

Sengeløv L , Maase H Von Der , Lundbeck F , Barlebo H , Engelholm SA , Krarup T , et al. Neoadjuvant Chemotherapy with Cisplatin and Methotrexate in Patients with Muscle-Invasive Bladder Tumours Neoadjuvant Chemotherapy with Cisplatin and Methotrexate in Patients with Muscle-Invasive Bladder Tumours. Acta Oncol (Madr). (2009) ;41: :447–56. doi:10.1080/028418602320405041

[123] 

James N , Hussain S , Hall E . Radiotherapy with or without chemotherapy in muscle-invasive bladder cancer. N Engl J Med. 2012:1477-88. doi:10.1016/j.eururo.2012.12.008

[124] 

Shipley WU , Winter KA , Kaufman DS , Lee WR , Heney NM , Tester WR , et al. Phase III trial of neoadjuvant chemotherapy in patients with invasive bladder cancer treated with selective bladder preservation by combined radiation therapy and chemotherapy: initial results of Radiation Therapy Oncology Group 89-03. J Clin Oncol. (1998) ;16: :3576–83. doi:10.1200/jco.1998.16.11.3576

[125] 

Fahmy O , Khairul-Asri MG , Schubert T , Renninger M , Malek R , Kübler H , et al. A systematic review and meta-analysis on the oncological long-term outcomes after trimodality therapy and radical cystectomy with or without neoadjuvant chemotherapy for muscle-invasive bladder cancer. Urol Oncol Semin Orig Investig. (2018) ;36: :43–53. doi:10.1016/j.urolonc.2017.10.002

[126] 

Jiang DM , Jiang H , Chung PWM , Zlotta AR , Fleshner NE , Bristow RG , et al. Neoadjuvant Chemotherapy Before Bladder- Sparing Chemoradiotherapy in Patients With Nonmetastatic Muscle-Invasive Bladder Cancer. Clin Genitourin Cancer. (2019) ;17: :38–45. doi:10.1016/j.clgc.2018.09.021

[127] 

Hafeez S , Horwich A , Omar O , Mohammed K , Thompson A , Kumar P , et al. Selective organ preservation with neo-adjuvant chemotherapy for the treatment of muscle invasive transitional cell carcinoma of the bladder. Br J Cancer. (2015) ;112: :1626–35. doi:10.1038/bjc.2015.109

[128] 

Zargar-Shoshtari K , Zargar H , Lotan Y , Shah JB , Van Rhijn BW , Daneshmand S , et al. A Multi-Institutional Analysis of Outcomes of Patients with Clinically Node Positive Urothelial Bladder Cancer Treated with Induction Chemotherapy and Radical Cystectomy. J Urol. (2016) ;195: :53–9. doi:10.1016/j.juro.2015.07.085

[129] 

Hermans TJN , Fransen van de Putte EE , Horenblas S , Meijer RP , Boormans JL , Aben KKH , et al. Pathological downstaging and survival after induction chemotherapy and radical cystectomy for clinically node-positive bladder cancer—Results of a nationwide population-based study. Eur J Cancer. (2016) ;69: :1–8. doi:10.1016/j.ejca.2016.09.015

[130] 

von der Maase H , Hansen S , Roberts J , Dogliotti L , Oliver T , Moore M , et al. Gemcitabine and Cisplatin Versus Methotrexate, Vinblastine, Doxorubicin, and Cisplatin in Advanced or Metastatic Bladder Cancer: Results of a Large, Randomized, Multinational, Multicenter, Phase III Study. J Clin Oncol. (2000) ;17: :3068–77.

[131] 

Necchi A , Mariani L , Vullo S Lo , Yu EY , Woods ME , Wong Y-N , et al. Lack of effectiveness of postchemotherapy lymphadenectomy in bladder cancer patients with clinical evidence of metastatic pelvic or retroperitoneal lymph nodes only: a propensity score-based analysis. Eur Urol Focus. (2019) ;5: :242–9. doi:10.1016/j.euf.2017.05.006.Lack

[132] 

Herr HW . Outcome of Patients Who Refuse Cystectomy after Receiving Neoadjuvant Chemotherapy for Muscle-Invasive Bladder Cancer. Eur Urol. (2008) ;54: :126–32. doi:10.1016/j.eururo.2007.12.031

[133] 

Robins D , Matulay J , Lipsky M , Meyer A , Ghandour R , DeCastro G , et al. Outcomes Following Clinical Complete Response to Neoadjuvant Chemotherapy for Muscle-invasive Urothelial Carcinoma of the Bladder in Patients Refusing Radical Cystectomy. Urology. (2018) ;111: :116–21. doi:10.1016/j.urology.2017.09.003

[134] 

Mazza P , Moran GW , Li G , Robins DJ , Matulay JT , Herr HW , et al. Conservative Management Following Clinical Complete Response to Neoadjuvant Chemotherapy of Muscle Invasive Bladder Cancer: Contemporary Outcomes of a Multi-Institutional Cohort Study. J Urol. 2018.

[135] 

Meyer A , Ghandour R , Bergman A , Castaneda C , Wosnitzer M , Hruby G , et al. The Natural History of Clinically Complete Responders to Neoadjuvant Chemotherapy for Urothelial Carcinoma of the Bladder. J Urol. (2014) ;192: :696–701. doi:10.1016/j.juro.2014.03.078

[136] 

deVere White RW , Lara PN , Goldman B , Tangen CM , Smith DC , Wood DP , et al. A Sequential Treatment Approach to Myoinvasive Urothelial Cancer: A Phase II Southwest Oncology Group Trial (S0219). J Urol. (2009) ;181: :2476–81. doi:10.1016/j.juro.2009.01.115

[137] 

Millikan R , Dinney C , Swanson D , Sweeney P , Ro JY , Smith TL , et al. Integrated Therapy for Locally Advanced Bladder Cancer: Final Report of a Randomized Trial of Cystectomy Plus Preoperative and Postoperative M-VAC. J Clin Med. (2001) ;19: :4005–13.

[138] 

Donat SM , Shabsigh A , Savage C , Cronin AM , Bochner BH , Dalbagni G , et al. Potential Impact of Postoperative Early Complications on the Timing of Adjuvant Chemotherapy in Patients Undergoing Radical Cystectomy: A High-Volume Tertiary Cancer Center Experience. (2009) ;55: :177–86. doi:10.1016/j.eururo.2008.07.018

[139] 

Skinner DG , Daniels JR , Russell CA , Lieskovsky G , Boyd SD , Nichols P , et al. The role of adjuvant chemotherapy following cystectomy for invasive bladder cancer: a prospective comparative trial. J Urol. (1991) ;145: :457–9. doi:https://doi.org/10.1016/S0022-5347(17)38368-4

[140] 

Stockle M , Meyenburg W , Wellek S , Voges GE , Rossmann M , Gertenbach U , et al. Adjuvant polychemotherapy of nonorgan-confined bladder cancer after radical cystectomy revisited: long-term results of a controlled prospective study and further clinical experience. J Urol. (1995) ;153: :47–52. doi:10.1097/00005392-199501000-00019

[141] 

Paz-Ares L , Solsona E , Esteban E , Saez A , Gonzalez-Larriba J , Anton A , et al. Randomized phase III trial comparing adjuvant paclitaxel/gemcitabine/cisplatin (PGC) to observation in patients with resected invasive bladder cancer: Results of the Spanish Oncology Genitourinary Group (SOGUG) 99/01 Study. J Clin Oncol. 2010:no. 18 suppl. DOI: 10.1200/jco.2010.28.18_suppl.lb. doi:10.1200/jco.2010.28.18

[142] 

Sternberg CN , Skoneczna I , Kerst JM , Albers P , Fossa SD , Agerbaek M , et al. Immediate versus deferred chemotherapy after radical cystectomy in patients with pT3-pT4 or N+M0 urothelial carcinoma of the bladder (EORTC 30994): An intergroup, open-label, randomised phase 3 trial. Lancet Oncol. (2015) ;16: :76–86. doi:10.1016/S1470-2045(14)71160-X

[143] 

Leow JJ , Martin-Doyle W , Rajagopal PS , Patel CG , Anderson EM , Rothman AT , et al. Adjuvant chemotherapy for invasive bladder cancer: A 2013 updated systematic review and meta-analysis of randomized trials. Eur Urol. (2014) ;66: :42–54. doi:10.1016/j.eururo.2013.08.033

[144] 

Kim HS , Jeong CW , Kwak C , Kim HH , Ku JH . Adjuvant chemotherapy for muscle-invasive bladder cancer: A systematic review and network meta-analysis of randomized clinical trials. Oncotarget. (2017) ;8: :81204–14. doi:10.18632/oncotarget.20979.

[145] 

Galsky MD , Stensland KD , Moshier E , Sfakianos JP , McBride RB , Tsao CK , et al. Effectiveness of adjuvant chemotherapy for locally advanced bladder cancer. J Clin Oncol. (2016) ;34: :825–32. doi:10.1200/JCO.2015.64.1076

[146] 

Booth CM , Siemens DR , Li G , Peng Y , Tannock IF , Kong W , et al. Perioperative Chemotherapy for Muscle-Invasive Bladder Cancer. A Population-Based Outcome Study. Cancer. (2014) ;120: :1630–8. doi:10.1002/cncr.28510

[147] 

Booth CM , Tannock IF , Cancer M . Benefits of Adjuvant Chemotherapy for Bladder Cancer. JAMA Oncol. (2015) ;1: :727–8.doi: 10.1001/jamaoncol.2015.1210.Conflict

[148] 

Berg S , D’Andrea D , Vetterlein MW , Cole AP , Fletcher SA , Krimphove MJ , et al. Impact of Adjuvant Chemotherapy in Patients With Adverse Features and Variant Histology at Radical Cystectomy for Muscle-Invasive Carcinoma of the Bladder: Does Histologic Subtype Matter? Cancer. (2019) ;125: :1449–58. doi:10.1002/cncr.31952

[149] 

Seisen T , Jamzadeh A , Leow JJ , Rouprêt M , Cole AP , Lipsitz SR , et al. Adjuvant chemotherapy vs observation for patients with adverse pathologic features at radical cystectomy previously treated with neoadjuvant chemotherapy. JAMA Oncol. (2018) ;4: :225–9. doi:10.1001/jamaoncol.2017.2374

[150] 

Zargar-Shoshtari K , Kongnyuy M , Sharma P , Fishman MN , Gilbert SM , Poch MA , et al. Clinical role of additional adjuvant chemotherapy in patients with locally advanced urothelial carcinoma following neoadjuvant chemotherapy and cystectomy. World J Urol. (2016) ;34: :1567–73. doi:10.1007/s00345-016-1825-3

[151] 

Parker WP , Habermann EB , Day CN , Zaid HB , Frank I , Thompson RH , et al. Adverse Pathology After Neoadjuvant Chemotherapy and Radical Cystectomy: The Role of Adjuvant Chemotherapy. Clin Genitourin Cancer. (2018) ;16: :64–71.e5. doi:10.1016/j.clgc.2017.07.010

[152] 

Sui W , Lim EA , Decastro GJ , Mckiernan JM . Use of Adjuvant Chemotherapy in Patients with Advanced Bladder Cancer after Neoadjuvant Chemotherapy. Bl Cancer. (2017) ;3: :181–9. doi:10.3233/BLC-170107

[153] 

Martinez Chanza N , Werner L , Plimack E , Yu EY , Alva AS , Crabb SJ , et al. Incidence, Patterns, and Outcomes with Adjuvant Chemotherapy for Residual Disease After Neoadjuvant Chemotherapy in Muscle-invasive Urinary Tract Cancers. Eur Urol Oncol. 2019:doi: 10.1016/j.euo.2018.12.013. [Epub ahead of print]. doi:10.1016/j.euo.2018.12.013

[154] 

Pederzoli F , Bandini M , Briganti A , Plimack ER , Yu EY , Bamias A , et al. Incremental Utility of Adjuvant Chemotherapy in Muscle-invasive Bladder Cancer: Quantifying the Relapse Risk Associated with Therapeutic Effect. Eur Urol. 2019:Eur Urol. 2019 Jul 11. pii: S0302-2838(19)30522-6. doi:10.1016/j.eururo.2019.06.032

[155] 

Galsky M , Hahn N , Rosenberg J , Sonpavde G , Hutson T , Oh W , et al. A consensus definition of patients with metastatic urothelial carcinoma who are unfit for cisplatin?based chemotherapy. Lancet Oncol. (2011) ;12: :211–4. doi:10.1016/S1470

[156] 

Hsu L , Li H , Pucheril D , Hansen M , Littleton R , Peabody J , et al. Use of percutaneous nephrostomy and ureteral stenting in management of ureteral obstruction. World J Nephrol. (2016) ;5: :172. doi:10.5527/wjn.v5.i2.172

[157] 

Kiss B , Furrer MA , Wuethrich PY , Burkhard FC , Thalmann GN , Roth B . Stenting Prior to Cystectomy is an Independent Risk Factor for Upper Urinary Tract Recurrence. J Urol. (2017) ;198: :1263–8. doi:10.1016/j.juro.2017.06.020

[158] 

Levey AS , Bosch JP , Lewis JB , Greene T . A More Accurate Method To Estimate Glomerular Filtration Rate from Serum Creatinine: A New Prediction Equation. Ann Intern Med. (1999) ;130: :461–70.

[159] 

Dash A , Galsky MD , Vickers AJ , Serio AM , Koppie TM , Dalbagni G , et al. Impact of Renal Impairment on Eligibility for Adjuvant Cisplatin-Based Chemotherapy in Patients With Urothelial Carcinoma of the Bladder. Cancer. (2006) ;107: :506–13. doi:10.1002/cncr.22031

[160] 

Janowitz T , Williams EH , Marshall A , Ainsworth N , Thomas PB , Sammut SJ , et al. New Model for Estimating Glomerular Filtration Rate in Patients With Cancer. J Clin Oncol. (2017) ;35: :2798–805. doi:10.1200/JCO.2017.72.7578

[161] 

Kidney Disease: Improving Global Outcomes (KDIGO) CKDWork Group. KDIGO 2012 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Kidney Int Suppl. (2013) ;3: :1–150.

[162] 

Hussain SA , Palmer DH , Lloyd B , Collins SI , Barton D , Ansari J , et al. A study of split-dose cisplatin-based neo-adjuvant chemotherapy in muscle-invasive bladder cancer. Oncol Lett. (2012) ;3: :855–9. doi:10.3892/ol.2012.563

[163] 

Morales-Barrera R , Bellmunt J , Suárez C , Valverde C , Guix M , Serrano C , et al. Cisplatin and gemcitabine administered every two weeks in patients with locally advanced or metastatic urothelial carcinoma and impaired renal function. Eur J Cancer. (2012) ;48: :1816–21. doi:10.1016/j.ejca.2012.04.002

[164] 

Koshkin VS , Barata PC , Rybicki LA , Zahoor H , Almassi N , Redden AM , et al. Feasibility of Cisplatin-Based Neoadjuvant Chemotherapy in Muscle-Invasive Bladder Cancer Patients With Diminished Renal Function. Clin Genitourin Cancer. (2018) ;16: :e879–92. doi:10.1016/j.clgc.2018.02.002

[165] 

Osterman C , Babu D , Geynisman D , Lewis B , Somer R , Balar A , et al. Efficacy of Split Schedule Versus Conventional Schedule Neoadjuvant Cisplatin-Based Chemotherapy for Muscle-Invasive Bladder Cancer. Oncologist. (2019) ;45: :688–90. doi:10.1634/theoncologist.2018-0561

[166] 

Kintzel PE , Dorrt RT . Anticancer guidelines drug renal toxicity and elimination for altered renal function. Cancer Treat Rev. (1995) ;21: :33–64.

[167] 

Cisplatin, Cancer Care Ontario 2018. https://www.cancercareontario.ca/en/drugformulary/drugs/cisplatin.

[168] 

Cisplatin, BC Cancer Agency 2016. http://www.bccancer.bc.ca/drug-database-site/drugindex/cisplatin_monograph_1jul2016.pdf.

[169] 

Janus N , Thariat J , Boulanger H , Deray G . Proposal for dosage adjustment and timing of chemotherapy in hemodialyzed patients. Ann Oncol. 2010:1395-403. doi:10.1093/annonc/mdp598

[170] 

Yin M , Joshi M , Meijer RP , Glantz M , Holder S , Harvey HA , et al. Neoadjuvant Chemotherapy for Muscle-Invasive Bladder Cancer: A Systematic Review and Two-Step Meta-Analysis. Oncologist. (2016) ;21: :708–15. doi:10.1634/theoncologist.2015-0440

[171] 

Bellmunt J , Ribas A , Eres N , Albanell J , Almanza C , Bermejo B , et al. Carboplatin-based versus cisplatin-based chemotherapy in the treatment of surgically incurable advanced bladder carcinoma. Cancer. (1997) ;80: :1966–72. doi: 10.1002/(SICI)1097-0142(19971115)80:10<1966::AID-CNCR14>3.0.CO;2-W

[172] 

Petrioli R , Frediani B , Manganelli A , Barbanti G , De Capua B , De Lauretis A , et al. Comparison between a cisplatin?containing regimen and a carboplatin?containing regimen for recurrent or metastatic bladder cancer patients. A randomized phase II study. Cancer. (1996) ;77: :344–51. doi:10.1002/(SICI)1097

[173] 

Dogliotti L , Cartení G , Siena S , Bertetto O , Martoni A , Bono A , et al. Gemcitabine plus Cisplatin versus Gemcitabine plus Carboplatin as First-Line Chemotherapy in Advanced Transitional Cell Carcinoma of the Urothelium: Results of a Randomized Phase 2 Trial. Eur Urol. (2007) ;52: :134–41. doi:10.1016/j.eururo.2006.12.029

[174] 

Galsky MD , Chen GJ , Oh WK , Bellmunt J , Roth BJ , Petrioli R , et al. Comparative effectiveness of cisplatin-based and carboplatin-based chemotherapy for treatment of advanced urothelial carcinoma. Ann Oncol. (2012) ;23: :406–10. doi:10.1093/annonc/mdr156

[175] 

Black PC , Brown GA , Dinney CPN . The impact of variant histology on the outcome of bladder cancer treated with curative intent. Urol Oncol Semin Orig Investig. (2009) ;27: :3–7. doi:10.1016/j.urolonc.2007.07.010

[176] 

Royce TJ , Lin CC , Gray PJ , Shipley WU , Jemal A , Efstathiou JA . Clinical characteristics and outcomes of nonurothelial cell carcinoma of the bladder: Results from the National Cancer Data Base. Urol Oncol Semin Orig Investig. (2017) ;36: :78.e1–78.e12. doi:10.1016/j.urolonc.2017.10.013

[177] 

Cohen AJ , Packiam V , Nottingham C , Steinberg G , Smith ND , Patel S . Upstaging of nonurothelial histology in bladder cancer at the time of surgical treatment in the National Cancer Data Base. Urol Oncol Semin Orig Investig. (2017) ;35: :34.e1–34.e8. doi:10.1016/j.urolonc.2016.08.002

[178] 

Lynch SP , Shen Y , Kamat A , Grossman HB , Shah JB , Millikan RE , et al. Neoadjuvant chemotherapy in small cell urothelial cancer improves pathologic downstaging and long-term outcomes: Results from a retrospective study at the md anderson cancer center. Eur Urol. (2013) ;64: :307–13. doi:10.1016/j.eururo.2012.04.020

[179] 

Siefker-Radtke AO , Kamat AM , Grossman HB , Williams DL , Qiao W , Thall PF , et al. Phase II clinical trial of neoadjuvant alternating doublet chemotherapy with ifosfamide/doxorubicin and etoposide/cisplatin in small-cell urothelial cancer. J Clin Oncol. (2009) ;27: :2592–7. doi:10.1200/JCO.2008.19.0256

[180] 

Moretto P , Wood L , Emmenegger U , Blais N , Mukherjee SD , Winquist E , et al. Management of small cell carcinoma of the bladder: Consensus guidelines from the Canadian Association of Genitourinary Medical Oncologists (CAGMO). Can Urol Assoc J. (2013) ;7: :E44–56.

[181] 

Dotson A , May A , Davaro F , Johar S , Sameer R , Zachary S . Squamous cell carcinoma of the bladder: poor response to neoadjuvant chemotherapy. Int J Clin Oncol. (2019) ;24: :706–11. doi:10.1007/s10147-019-01409-x

[182] 

Kassouf W , Spiess PE , Siefker-radtke A , Swanson D , Grossman HB , Munsell MF , et al. Outcome and Patterns of Recurrence of Nonbilharzial Pure Squamous Cell Carcinoma of the Bladder. Cancer. 2007:764-9. doi:10.1002/cncr.22853

[183] 

Izard JP , H MP, Siemens DR , D M, Mackillop WJ , Ch B, et al. Outcomes of squamous histology in bladder cancer: A population-based study. Urol Oncol Semin Orig Investig. (2015) ;33: :425.e7–425.313.

[184] 

Quilty P , Duncan W . Radiotherapy for Squamous Carcinoma of the Urinary Bladder. Int J Radiat Oncol Biol Phys. (1986) ;12: :861–5.

[185] 

Zahoor H , Elson P , Stephenson A , Haber GP , Kaouk J , Fergany A , et al. Patient Characteristics, Treatment Patterns and Prognostic Factors in Squamous Cell Bladder Cancer. Clin Genitourin Cancer. (2018) ;16: :e437–42. doi:10.1016/j.clgc.2017.10.005

[186] 

Koshkin VS , Garcia JA , Reynolds J , Elson P , Magi-Galluzzi C , McKenney JK , et al. Transcriptomic and protein analysis of small-cell bladder cancer (SCBC) Identifies prognostic biomarkers and DLL3 as a relevant therapeutic target. Clin Cancer Res. (2018) ;16: :e437–42. doi:10.1158/1078-0432.CCR-18-1278

[187] 

Scosyrev E , Ely BW , Messing EM , Speights VO , Grossman HB , Wood DP , et al. Do mixed histological features affect survival benefit from neoadjuvant platinum-based combination chemotherapy in patients with locally advanced bladder cancer? A secondary analysis of Southwest Oncology Group-Directed Intergroup Study (S8710). BJU Int. (2010) ;108: :693–700. doi:10.1111/j.1464-410X.2010.09900

[188] 

Kaimakliotis H , Monn M , Cho J , Pedrosa J , Hahn N , Albany C , et al. Neoadjuvant chemotherapy in urothelial bladder cancer: impact of regimen and variant histology. Futur Oncol. (2016) ;12: :1795–804. doi:10.2217/fon-2016-0056

[189] 

Mitra AP , Bartsch CC , Bartsch G , Miranda G , Skinner EC , Daneshmand S . Does presence of squamous and glandular differentiation in urothelial carcinoma of the bladder at cystectomy portend poor prognosis? An intensive case-control analysis. Urol Oncol Semin Orig Investig. (2014) ;32: :117–27. doi:10.1016/j.urolonc.2012.08.017

[190] 

Zargar-Shoshtari K , Sverrisson EF , Sharma P , Gupta S , Poch MA , Pow-Sang JM , et al. Clinical Outcomes after Neoadjuvant Chemotherapy and Radical Cystectomy in the Presence of Urothelial Carcinoma of the Bladder with Squamous or Glandular Differentiation. Clin Genitourin Cancer. (2016) ;14: :82–8. doi:10.1016/j.clgc.2015.08.006

[191] 

Kim SP , Frank I , Cheville JC , Thompson RH , Weight CJ , Thapa P , et al. The impact of squamous and glandular differentiation on survival after radical cystectomy for urothelial carcinoma. J Urol. (2012) ;188: :405–9. doi:10.1016/j.juro.2012.04.020

[192] 

Stensland KD , Zaid H , Broadwin M , Sorcini A , Canes D , Galsky M , et al. Comparative Effectiveness of Treatment Strategies for Squamous Cell Carcinoma of the Bladder. Eur Urol Oncol. 2018:doi: 10.1016/j.euo.2018.11.003. [Epub ahead of print]. doi:10.1016/j.euo.2018.11.003

[193] 

Davaro F , Schaefer J , May A , Raza J , Siddiqui S , Hamilton Z . Invasive non - urachal adenocarcinoma of the bladder : analysis of the National Cancer Database. World J Urol. 2018:[Epub ahead of print]. doi:10.1007/s00345-018-2411-7

[194] 

Yu B , Zhou J , Cai H , Xu T , Xu Z , Zou Q , et al. Neoadjuvant chemotherapy for primary adenocarcinomas of the urinary bladder: a single-site experience. BMC Urol. (2015) ;15: :1–4.

[195] 

Siefker-radtke A . Urachal Adenocarcinoma: A Clinician’s Guide for Treatment. Semin Oncol. (2012) ;39: :619–24.

[196] 

Patel SG , Weiner AB , Keegan K , Morgan T . Oncologic outcomes in patients with nonurothelial bladder cancer. Indian J Urol. (2018) ;34: :39–44. doi:10.4103/iju.IJU

[197] 

Hamilou Z , North S , Canil C , Wood L , Hotte S , Sridhar S , et al. Management of urachal cancer: A review by the Canadian Urological Association and Genitourinary Medical Oncologists of Canada. Can Urol Assoc J. 2019.

[198] 

Fernández MI , Williams SB , Willis DL , Slack RS , Dickstein RJ , Parikh S , et al. Clinical risk stratification in patients with surgically resectable micropapillary bladder cancer. BJU Int. (2017) ;119: :684–91. doi: 10.1111/bju.13689

[199] 

Meeks JJ , Taylor JM , Matsushita K , Herr HW , Donat SM , Bochner BH , et al. Pathological response to neoadjuvant chemotherapy for muscle-invasive micropapillary bladder cancer. BJU Int. (2013) ;111: :325–30. doi:10.1111/j.1464-410X.2012.11751.x

[200] 

Ghoneim IA , Miocinovic R , Stephenson AJ , Garcia JA , Gong MC , Campbell SC , et al. Neoadjuvant systemic therapy or early cystectomy? Single-center analysis of outcomes after therapy for patients with clinically localized micropapillary urothelial carcinoma of the bladder. Urology. (2011) ;77: :867–70. doi:10.1016/j.urology.2010.11.043

[201] 

Abufaraj M , Foerster B , Schernhammer E , Moschini M , Kimura S , Hassler MR , et al. Micropapillary Urothelial Carcinoma of the Bladder: A Systematic Review and Meta-analysis of Disease Characteristics and Treatment Outcomes. Eur Urol. (2019) ;75: :649–58. doi:10.1016/j.eururo.2018.11.052

[202] 

Sui W , Matulay JT , James MB , Onyeji IC , Theofanides MC , RoyChoudhury A , et al. Micropapillary bladder cancer: Insights from the national cancer database. Bl Cancer. (2016) ;2: :415–23. doi:10.3233/BLC-160066

[203] 

Li Q , Assel M , Benfante NE , Pietzak EJ , Herr HW , Donat M , et al. The Impact of Plasmacytoid Variant Histology on the Survival of Patients with Urothelial Carcinoma of Bladder after Radical Cystectomy. Eur Urol Focus. (2019) ;5: :104–8. doi:10.1016/j.euf.2017.06.013

[204] 

Keck B , Wach S , Stoehr R , Kunath F , Bertz S , Lehmann J , et al. Plasmacytoid variant of bladder cancer defines patients with poor prognosis if treated with cystectomy and adjuvant cisplatin-based chemotherapy. BMC Cancer. (2013) ;13: :1–8.

[205] 

Gunaratne DA , Krieger LEM , Maclean F , Vaux KJ , Chalasani V . Neoadjuvant Chemotherapy With Gemcitabine and Cisplatin for Plasmacytoid Urothelial Bladder Cancer: A Case Report and Review of the Literature. Clin Genitourin Cancer. (2016) ;14: :e103–5. doi:10.1016/j.clgc.2015.08.009

[206] 

Dayyani F , Czerniak BA , Sircar K , Munsell MF , Randall E . Plasmacytoid Urothelial Carcinomas - A Chemo-sensitive Cancer with Poor Prognosis, and Peritoneal Carcinomatosis. J Urol. (2014) ;189: :1656–61. doi:10.1016/j.juro.2012.11.084.Plasmacytoid

[207] 

Cockerill PA , Cheville JC , Boorjian SA , Blackburne A , Thapa P , Tarrell RF , et al. Outcomes Following Radical Cystectomy for Plasmacytoid Urothelial Carcinoma: Defining the Need for Improved Local Cancer Control. Urology. (2017) ;102: :143–7. doi:10.1016/j.urology.2016.09.053

[208] 

Diamantopoulos LN , Khaki AR , Grivas P , Gore JL , Schade GR , Hsieh AC , et al. Plasmacytoid Urothelial Carcinoma: Response to Chemotherapy and Oncologic Outcomes. Bl Cancer. (2020) ;6: :71–81. doi:10.3233/BLC-190258

[209] 

Wasco MJ , Daignault S , Bradley D , Shah RB . Nested variant of urothelial carcinoma: a clinicopathologic and immunohistochemical study of 30 pure and mixed cases. Hum Pathol. (2010) ;41: :163–71. doi:10.1016/j.humpath.2009.07.015

[210] 

Venyo AK . Nested Variant of Urothelial Carcinoma. Adv Urol. Epub Jan. (2014) ;2014: :Epub 2014 Jan 22. doi:10.1155/2014/192720

[211] 

Sui W , Matulay JT , Onyeji IC , Theofanides MC , James MB , Roychoudhury A , et al. Contemporary treatment patterns and outcomes of sarcomatoid bladder cancer. World J Urol. (2017) ;35: :1055–61. doi:10.1007/s00345-016-1962-8

[212] 

Malla M , Wang JF , Trepeta R , Feng A , Wang J . Sarcomatoid Carcinoma of the Urinary Bladder. Clin Genitourin Cancer. (2016) ;14: :366–72. doi:10.1016/j.clgc.2016.03.004

[213] 

Vetterlein MW , Wankowicz SAM , Seisen T , Lander R . Neoadjuvant Chemotherapy Prior to Radical Cystectomy for Muscle-Invasive Bladder Cancer With Variant Histology. Cancer. (2017) ;123: :4346–55. doi:10.1002/cncr.30907

[214] 

Griffiths G . International phase III trial assessing neoadjuvant cisplatin, methotrexate, and vinblastine chemotherapy for muscle-invasive bladder cancer: Long-term results of the BA06 30894 trial. J Clin Oncol. (2011) ;29: :2171–7.

[215] 

Freiha F , Reese J , Torti FM . A randomized trial of radical cystectomy versus radical cystectomy plus cisplatin, vinblastine and methotrexate chemotherapy for muscle invasive bladder cancer. J Urol. (1996) ;155: :495–500. doi:10.1200/JCO.2010.32.3139

[216] 

Wishnow K , Ayala AJ , Levinson AK , Logothetis CJ , Johnson DE , Swanson DA , et al. Stage B (P2/3A/N0) Transitional Cell Carcinoma Of Bladder Highly Curable By Radical Cystectomy. Urology. (1992) ;39: :12–6. doi:10.1016/S0022-5347(01)66430-9

[217] 

Abol-Enein H , Tilki D , Mosbah A , El-Baz M , Shokeir A , Nabeeh A , et al. Does the extent of lymphadenectomy in radical cystectomy for bladder cancer influence disease-free survival? A prospective single-center study. Eur Urol. (2011) ;60: :572–7. doi:10.1016/j.eururo.2011.05.062

[218] 

Leissner J , Ghoneim MA , Abol-Enein H , Thuroff JW , Franzaring L , Fisch M , et al. Extended Radical Lymphadenectomy in Patients With Urothelial Bladder Cancer:: Results of a Prospective Multicenter Study. J Urol. (2004) ;171: :139–44. doi:10.1097/01.ju.0000102302.26806.fb

[219] 

Mills RD , Turner WH , Fleischmann A , Mark-Walder R , Thalmann GN , Studer UE . Pelvic Lymph Node Metastases From Bladder Cancer: Outcome in 83 Patients After Radical Cystectomy and Pelvic Lymphadenectomy. J Urol. (2001) ;166: :19–23. doi:10.1016/S0022-5347(05)66067-3

[220] 

Hwang E , Sathianathen N , Imamura M , Kuntz G , Risk M , Dahm P . Extended versus standard lymph node dissection for urothelial carcinoma of the bladder in patients undergoing radical cystectomy (Review). Cochrane Database Syst Rev. 2019:1-31. doi:10.1002/14651858.CD013336.www.cochranelibrary.com

[221] 

Gschwend E , Heck MM , Lehmann J , Rubben H , Albers P , Wolffe JM , et al. Extended Versus Limited Lymph Node Dissection in Bladder Cancer Patients Undergoing Radical Cystectomy: Survival Results from a Prospective, Randomized Trial. Eur Urol. (2019) ;75: :604–11. doi:10.1016/j.eururo.2018.09.047

[222] 

Herr HW , Bochner BH , Dalbaghi G , Donat SM , Reuter VE , Bajorin DF . Impact of the Number of Lymph Nodes Retrieved on Outcome in Patients With Muscle Invasive Bladder Cancer. J Urol. (2002) ;167: :1295–8. doi:10.1016/S0022-5347(05)65284-6

[223] 

Wright JL , Lin DW , Porter MP . The association between extent of lymphadenectomy and survival among patients with lymph node metastases undergoing radical cystectomy. Cancer. (2008) ;112: :2401–8. doi:10.1002/cncr.23474

[224] 

Konety BR , Joslyn SA , O’Donnell MA . Extent of Pelvic Lymphadenectomy and Its Impact On Outcome in Patients Diagnosed With Bladder Cancer: Analysis of Data From the Surveillance, Epidemiology and End Results Program Data Base. J Urol. (2003) ;169: :946–50. doi:10.1097/01.ju.0000052721.61645.a3

[225] 

Kijima T , Tanaka H , Koga F , Masuda H . Selective tetramodal bladder-preservation therapy, incorporating induction chemoradiotherapy and consolidative partial cystectomy with pelvic lymph node dissection for muscle-invasive bladder cancer: oncological and functional outcomes of 107 patients. BJU Int. (2019) ;124: :242–50. doi:10.1111/bju.14736

[226] 

Bellmunt J , Orsola A , Leow JJ , Wiegel T , De Santis M , Horwich A . Bladder cancer: ESMO practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. (2014) ;25: :iii40–8. doi:10.1093/annonc/mdu223

[227] 

Gogna NK , Matthews JHL , Turner SL , Mameghan H , Duchesne GM , Spry N , et al. Efficacy and tolerability of concurrent weekly low dose cisplatin during radiation treatment of localised muscle invasive bladder transitional cell carcinoma: A report of two sequential Phase II studies from the Trans Tasman Radiation Oncology Group. Radiother Oncol. (2006) ;81: :9–17. doi:10.1016/j.radonc.2006.09.001

[228] 

Coppin CM , Gospodarowicz MK , James K , Tannock IF , Zee B , Carson J , et al. Improved local control of invasive bladder cancer by concurrent cisplatin and preoperative or definitive radiation. The National Cancer Institute of Canada Clinical Trials GrouJ Clin Oncol. (1996) ;14: :2901–7. doi:10.1016/j.radonc.2006.09.001

[229] 

Rödel C , Grabenbauer GG , Kühn R , Papadopoulos T , Dunst J , Meyer M , et al. Combined-modality treatment and selective organ preservation in invasive bladder cancer: Long-term results. J Clin Oncol. (2002) ;20: :3061–71. doi:10.1200/JCO.2002.11.027

[230] 

Ghate K , Brennan K , Karim S , Siemens DR , Mackillop WJ , Booth CM . Concurrent chemoradiotherapy for bladder cancer: Practice patterns and outcomes in the general population. Radiother Oncol. (2018) ;127: :136–42. doi:10.1016/j.radonc.2017.12.009

[231] 

Hoskin PJ , Rojas AM , Bentzen SM , Saunders MI . Radiotherapy With Concurrent Carbogen and Nicotinamide in Bladder Carcinoma. J Clin Oncol. (2010) ;28: :4912–8. doi:10.1200/JCO.2010.28.4950

[232] 

Mitin T , Hunt D , Shipley WU , Kaufman DS , Uzzo R , Wu CL , et al. Transurethral surgery and twice-daily radiation plus paclitaxel-cisplatin or fluorouracil-cisplatin with selective bladder preservation and adjuvant chemotherapy for patients with muscle invasive bladder cancer (RTOG 0233): A randomised multicentre phase. Lancet Oncol. (2013) ;14: :863–72. doi:10.1016/S1470-2045(13)70255-9

[233] 

Hagan MP , Winter KA , Kaufman DS , Wajsman Z , Zietman AL , Heney NM , et al. RTOG 97- Initial report of a phase I-II trial of selective bladder conservation using TURBT, twice-daily accelerated irradiation sensitized with cisplatin, and adjuvant MCV combination chemotherapy. Int J Radiat Oncol Biol Phys. (2003) ;57: :665–72. doi:10.1016/S0360-3016(03)00718-1

[234] 

Kaufman DS , Winter KA , Shipley WU , Heney NM , Wallace HJ , Toonkel LM , et al. Phase I-II RTOG Study (99-06) of Patients With Muscle-Invasive Bladder Cancer Undergoing Transurethral Surgery, Paclitaxel, Cisplatin, and Twice-daily Radiotherapy Followed by Selective Bladder Preservation or Radical Cystectomy and Adjuvant Chemotherapy. Urology. (2009) ;73: :833–7. doi:10.1016/j.urology.2008.09.036

[235] 

Tester W , Caplan R , Heaney J , Venner P , Whittington R , Byhardt R , et al. Neoadjuvant combined modality program with selective organ preservation for invasive bladder cancer: Results of Radiation Therapy Oncology Group phase II trial 8802. J Clin Oncol. (1996) ;14: :119–26. doi:10.1200/JCO.1996.14.1.119

[236] 

Choudhury A , Swindell R , Logue JP , Elliott PA , Livsey JE , Wise M , et al. Phase II study of conformal hypofractionated radiotherapy with concurrent gemcitabine in muscle-invasive bladder cancer. J Clin Oncol. (2011) ;29: :733–8. doi:10.1200/JCO.2010.31.5721

[237] 

Coen JJ , Zhang P , Saylor PJ , Lee CT , Wu C , Parker W . Bladder Preservation With Twice-a-Day Radiation Plus Fluorouracil / Cisplatin or Once Daily Radiation Plus Gemcitabine for Muscle-Invasive Bladder Cancer : NRG / RTOG 0712 — A Randomized Phase II Trial. J Clin Oncol. (2019) ;37: :44–51. doi:10.1200/JCO.18.00537

[238] 

Caffo O , Thompson C , Santis M De , Kragelj B , Hamstra DA , Azria D , et al. Concurrent gemcitabine and radiotherapy for the treatment of muscle-invasive bladder cancer: A pooled individual data analysis of eight phase I - II trials. Radiother Oncol. (2016) ;121: :193–8. doi:10.1016/j.radonc.2016.09.006

[239] 

Thompson C , Joseph N , Sanderson B , Logue J , Wylie J , Elliott T , et al. Tolerability of Concurrent Chemoradiation Therapy With Gemcitabine (GemX), With and Without Prior Neoadjuvant Chemotherapy, in Muscle Invasive Bladder Cancer. Int J Radiat Oncol Biol Phys. (2017) ;97: :732–9. doi:10.1016/j.ijrobp.2016.11.040

[240] 

Huddart RA , Birtle A , Maynard L , Beresford M , Blazeby J , Donovan J , et al. Clinical and patient-reported outcomes of SPARE - a randomised feasibility study of selective bladder preservation versus radical cystectomy. BJU Int. (2017) ;120: :639–50. doi:10.1111/bju.13900

[241] 

Wettstein MS , Rooprai JK , Pazhepurackel C , Wallis CJD , Klaassen Z , Uleryk EM , et al. Systematic review and meta-analysis on trimodal therapy versus radical cystectomy for muscle-invasive bladder cancer: Does the current quality of evidence justify definitive conclusions? PLoS One. (2019) ;14: :e0216255.

[242] 

Williams SB , Shan Y , Jazzar U , Mehta HB , Baillargeon JG , Huo J , et al. Comparing Survival Outcomes and Costs Associated With Radical Cystectomy and Trimodal Therapy for Older Adults With Muscle-Invasive Bladder Cancer. JAMA Surg. (2018) ;153: :881–9. doi:10.1001/jamasurg.2018.1680

[243] 

Kulkarni GS , Hermanns T , Wei Y , Bhindi B , Satkunasivam R , Athanasopoulos P , Sridhar SS . Propensity Score Analysis of Radical Cystectomy Versus Bladder-Sparing Trimodal Therapy in the Setting of a Multidisciplinary Bladder Cancer Clinic. J Clin Oncol. (2017) ;35: :2299–307. doi:10.1200/JCO.2016.69.2327

[244] 

Ploussard G , Daneshmand S , Efstathiou JA , Herr HW , James ND , Rödel CM , et al. Critical analysis of bladder sparing with trimodal therapy in muscle-invasive bladder cancer: A systematic review. Eur Urol. (2014) ;66: :120–37. doi:10.1016/j.eururo.2014.02.038

[245] 

Gakis G , Efstathiou J , Lerner S , Cookson M , Keegan K . ICUD-EAU International Consultation on Bladder Cancer Non-muscle-invasive urothelial carcinoma of the bladder. Eur Urol. (2013) ;63: :36–44. doi:10.1016/j.eururo.2012.08.061

[246] 

Efstathiou JA , Spiegel DY , Shipley WU , Heney NM , Kaufman DS , Niemierko A , et al. Long-term outcomes of selective bladder preservation by combined-modality therapy for invasive bladder cancer: The MGH experience. Eur Urol. (2012) ;61: :705–11. doi:10.1016/j.eururo.2011.11.010

[247] 

Shipley WU , Kaufman DS , Zehr E , Heney NM , Lane SC , Thakral HK , et al. Selective bladder preservation by combined modality protocol treatment: Long-term outcomes of 190 patients with invasive bladder cancer. Urology. (2002) ;60: :62–7. doi:10.1016/S0090-4295(02)01650-3

[248] 

Buchser D , Zapatero A , Rogado J , Talaya M , Vidales CM de , Arellano R , et al. Long-term Outcomes and Patterns of Failure Following Trimodality Treatment With Bladder Preservation for Invasive Bladder Cancer. Urology. (2019) ;124: :183–90. doi:10.1016/j.urology.2018.07.058

[249] 

Perdona S , Autorino R , Damiano R , Sio M De , Morrica B , Gallo L , et al. Bladder-sparing, Combined-modality Approach for Muscle-invasive Bladder Cancer. A Multi-institutional, Long-term Experience. Cancer. (2008) ;112: :75–83. doi:10.1002/cncr.23137

[250] 

Mak RH , Hunt D , Shipley WU , Efstathiou JA , Tester WJ , Hagan MP , et al. Long-term outcomes in patients with muscle-invasive bladder cancer after selective bladder-preserving combined-modality therapy:Apooled analysis of radiation therapy oncology group protocols 8802, 8903, 9506, 9706, 9906, and 0233. J Clin Oncol. (2014) ;32: :3801–9. doi:10.1200/JCO.2014.57.5548

[251] 

Vashistha V , Wang H , Mazzone A , Liss MA , Svatek RS , Schleicher M , et al. Radical Cystectomy Compared to Combined Modality Treatment for Muscle-Invasive Bladder Cancer: A Systematic Review and Meta-Analysis. Int J Radiat Oncol Biol Phys. (2017) ;97: :1002–20. doi:10.1016/j.ijrobp.2016.11.056.

[252] 

Giacalone NJ , Shipley WU , Clayman RH , Niemierko A , Drumm M , Heney NM , et al. Long-term Outcomes After Bladder-preserving Tri-modality Therapy for Patients with Muscle-invasive Bladder Cancer: An Updated Analysis of the Massachusetts General Hospital Experience. Eur Urol. (2017) ;71: :952–60. doi:10.1016/j.eururo.2016.12.020

[253] 

García-Perdomo HA , Montes-Cardona CE , Guacheta M , Castillo DF , Reis LO . Muscle-invasive bladder cancer organ-preserving therapy: systematic review and meta-analysis. World J Urol. 2018:1-12. doi:10.1007/s00345-018-2384-6

[254] 

Arcangeli G , Strigari L , Arcangeli S . Radical cystectomy versus organ-sparing trimodality treatment in muscle-invasive bladder cancer: A systematic review of clinical trials. Crit Rev Oncol Hematol. (2015) ;95: :387–96. doi:10.1016/j.critrevonc.2015.04.006

[255] 

Gofrit ON , Nof R , Meirovitz A , Pode D , Frank S , Katz R , et al. Radical cystectomy vs. chemoradiation in T2-4aN0M0 bladder cancer: A case-control study. Urol Oncol Semin Orig Investig. (2015) ;33: :19.e1–19.e5. doi:10.1016/j.urolonc.2014.09.014

[256] 

Haque W , Verma V , Aghazadeh M , Darcourt J , Butler E , Teh B . Short-term Mortality Associated with Definitive Chemoradiotherapy Versus Radical Cystectomy for Muscle-invasive Bladder Cancer. Clin Geni. 2019:doi: 10.1016/j.clgc.2019.06.015. [Epub ahead of print]. doi:10.1016/j.clgc.2019.06.015

[257] 

Fischer-Valuck BW , Rudra S , Gabani P , Brenneman R , Mueller R , Chin W , et al. Impact of Facility Radiation Patient Volume on Overall Survival in Patients with Muscle Invasive Bladder Cancer Undergoing Trimodality Bladder Preservation Therapy. Bl Cancer. 2019;Prepress:1-10. doi:10.3233/BLC-190233

[258] 

Sanderson KM , Cai J , Miranda G , Skinner DG , Stein JP . Upper Tract Urothelial Recurrence Following Radical Cystectomy for Transitional Cell Carcinoma of the Bladder: An Analysis of 1,069 Patients With 10-Year Followup. J Urol. (2007) ;177: :2088–94. doi:10.1016/j.juro.2007.01.133

[259] 

Schroeck FR , Smith N , Shelton JB . Implementing risk-aligned bladder cancer surveillance care. Urol Oncol Semin Orig Investig. (2018) ;36: :257–64. doi:10.1016/j.urolonc.2017.12.016

[260] 

Stewart-merrill SB , D M, Boorjian SA , D M, Thompson RH , D M, et al. Evaluation of current surveillance guidelines following radical cystectomy and proposal of a novel risk-based approach. Urol Oncol Semin Orig Investig. (2015) ;33: :339.e1–339.e8. doi:10.1016/j.urolonc.2015.04.017

[261] 

Cagiannos I , Morash C . Surveillance strategies after definitive therapy of invasive bladder cancer. CUAJ. (2009) ;3: :237–42.

[262] 

Honma I , Masumori N , Sato E , Takayanagi A , Takahashi A , Itoh N , et al. Local recurrence after radical cystectomy for invasive bladder cancer: An analysis of predictive factors. Urology. (2004) ;64: :744–8. doi:10.1016/j.urology.2004.05.003

[263] 

Yoo SH , Kim H , Kwak C , Kim HH , Jung JH , Ku JH . Late Recurrence of Bladder Cancer following Radical Cystectomy : Characteristics and Outcomes. Urol Int. 2019;1-6. doi:10.1159/000502656

[264] 

Kusaka A , Hatakeyama S , Hosogoe S , Hamano I . Risk-stratified surveillance and cost effectiveness of follow-up after radical cystectomy in patients with muscle-invasive bladder cancer. Oncotarget. (2017) ;8: :65492–505.

[265] 

Shariat SF , Karakiewicz PI , Palapattu GS , Lotan Y , Rogers CG , Amiel GE , et al. Outcomes of Radical Cystectomy for Transitional Cell Carcinoma of the Bladder: A Contemporary Series From the Bladder Cancer Research Consortium. J Urol. (2006) ;176: :2414–22. doi:10.1016/j.juro.2006.08.004

[266] 

Bellmunt J , de Wit R , Vaughn DJ , Fradet Y , Lee J-L , Fong L , et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med. (2017) ;376: :1015–26. doi:10.1056/NEJMoa1613683

[267] 

Necchi A , Anichini A , Raggi D , Briganti A , Massa S , Lucianò R , et al. Pembrolizumab as Neoadjuvant Therapy Before Radical Cystectomy in Patients With Muscle-Invasive Urothelial Bladder Carcinoma (PURE-01): An Open-Label, Single-Arm, Phase II Study. J Clin Oncol. 2018:JCO.18.01148. doi:10.1200/JCO.18.01148

[268] 

Powles T , et al. Aphase II study investigating the safety and efficacy of neoadjuvant atezolizumab in muscle invasive bladder cancer (ABACUS), n.d., p. 2018 ASCO Annual Meeting Presentation June 3, 2018.

[269] 

Heijden M Van Der , Dijk N Van , Smit L , Hendricksen K , Feijter D , Bekers E , et al. Pre-operative ipilimumab and nivolumab in locoregionally advanced, stage III, urothelial cancer (NABUCCO). Ann Oncol. (2019) ;30: :v356–402. doi:10.1093/annonc/mdz249

[270] 

Carthon BC , Wolchok JD , Yuan J , Kamat A , Ng Tang DS , Sun J , et al. Preoperative CTLA-4 blockade: Tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin Cancer Res. (2010) ;16: :2861–71. doi:10.1158/1078-0432.CCR-10-0569

[271] 

Gao J , Siefker-Radtke AO , Navai N , Campbell MT , Tidwell RS , Guo C , et al. A pilot presurgical study evaluating anti-PD-L1 durvalumab (durva) plus anti-CTLA-4 tremelimumab (treme) in patients (pts) with high-risk muscle-invasive bladder carcinoma (MIBC) who are ineligible for cisplatin-based neoadjuvant chemotherapy (NAC). J Clin Oncol 37, 2019, p. suppl; abstr 4551.

[272] 

Hoimes CJ , Albany C , Hoffman-Censits J , Fleming MT , Trabulsi E , Picus J , et al. A Phase IB/2 Study of Neoadjuvant Pembrolizumab and Chemotherapy for Locally Advanced Urothelial Cancer. Ann Oncol. (2018) ;29: :(suppl_8).

[273] 

Gupta S , Sonpavde G , Weight CJ , McGregor BA , Gupta S , Maughan BL , et al. Results from BLASST-1 (Bladder Cancer Signal Seeking Trial) of nivolumab, gemcitabine, and cisplatin in muscle invasive bladder cancer (MIBC) undergoing cystectomy. J Clin Oncol. (2020) ;38: :439–439. doi:10.1200/jco.2020.38.6_suppl.439

[274] 

Briganti A , Gandaglia G , Scuderi S , Gallina A , Colombo R , Necchi A , et al. Surgical Safety of Radical Cystectomy and Pelvic Lymph Node Dissection Following Neoadjuvant Pembrolizumab in Patients with Bladder Cancer : Prospective Assessment of Perioperative Outcomes from the PURE-01 Trial. Eur Urol. 2019. doi:10.1016/j.eururo.2019.12.019

[275] 

Rodriguez-Moreno JF , de Velasco G , Fernandez IB , Carlos A-F , Fernandez R , Vazquez-Estevez S , et al. Impact of the combination of durvalumab (MEDI4736) plus olaparib (AZD2281) administered prior to surgery in the molecular profile of resectable urothelial bladder cancer: NEODURVARIB Trial. J Clin Oncol. (2020) ;38: :(suppl 6; abstr 542).

[276] 

Hussain SA , Lester JF , Jackson R , Gornall M , Elliott A , Crabb SJ , et al. Phase II randomized placebo-controlled neoadjuvant trial of nintedanib or placebo with gemcitabine and cisplatin in locally advanced muscle invasive bladder cancer (NEO-BLADE). J Clin Oncol. (2020) ;38: :438–438. doi:10.1200/jco.2020.38.6_suppl.438

[277] 

Nadal R , Apolo AB . Overview of Current and Future Adjuvant Therapy for Muscle-Invasive Urothelial Carcinoma. Curr Treat Options Oncol. (2018) ;19: :36. doi:10.1007/s11864-018-0551-z

[278] 

Choi W , Porten S , Kim S , Willis D , Plimack ER , Hoffman-Censits J , et al. Identification of Distinct Basal and Luminal Subtypes of Muscle-Invasive Bladder Cancer with Different Sensitivities to Frontline Chemotherapy. Cancer Cell. (2014) ;25: :152–65. doi:10.1016/j.ccr.2014.01.009

[279] 

Seiler R , Ashab HAD , Erho N , van Rhijn BWG , Winters B , Douglas J , et al. Impact of Molecular Subtypes in Muscle-invasive Bladder Cancer on Predicting Response and Survival after Neoadjuvant Chemotherapy. Eur Urol. (2017) ;72: :544–54. doi:10.1016/j.eururo.2017.03.030

[280] 

Robertson AG , Kim J , Al-Ahmadie H , Bellmunt J , Guo G , Cherniack AD , et al. Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer. Cell. (2017) ;171: :540–556.e25. doi: 10.1016/j.cell.2017.09.007

[281] 

Plimack ER , Dunbrack RL , Brennan TA , Andrake MD , Zhou Y , Serebriiskii IG , et al. Defects in DNA Repair Genes Predict Response to Neoadjuvant Cisplatin-based Chemotherapy in Muscle-invasive Bladder Cancer. Eur Urol. (2015) ;68: :959–67. doi:10.1016/j.eururo.2015.07.009

[282] 

da Costa JB , Gibb EA , Bivalacqua TJ , Liu Y , Oo HZ , Miyamoto DT , et al. Molecular Characterization of Neuroendocrine- like Bladder Cancer. Clin Cancer Res. (2019) ;25: :3908–20. doi:10.1158/1078-0432.CCR-18-3558

[283] 

Kamoun A , Reynie‘s A de , Allory Y , Sjödahl G , Robertson A , Seiler R , et al. A Consensus Molecular Classification of Muscle-invasive Bladder Cancer. Eur Urol. 2019;September:[Epub ahead of print]. doi:10.1016/j.eururo.2019.09.006

[284] 

McConkey DJ , Choi W , Ochoa A , Siefker-Radtke A , Czerniak B , Dinney CPN . Therapeutic Opportunities in the Intrinsic Subtypes of Muscle-Invasive Bladder Cancer. Hematol Oncol Clin North Am. (2015) ;29: :377–94. doi:10.1016/j.hoc.2014.11.003

[285] 

Lotan Y , Boorjian SA , Zhang J , Bivalacqua TJ , Porten SP , Wheeler T , et al. Molecular Subtyping of Clinically Localized Urothelial Carcinoma Reveals Lower Rates of Pathological Upstaging at Radical Cystectomy Among Luminal Tumors. Eur Urol. (2019) ;76: :200–6. doi:10.1016/j.eururo.2019.04.036

[286] 

Groenendijk FH , De Jong J , Fransen Van De Putte EE , Michaut M , Schlicker A , Peters D , et al. ERBB2 Mutations Characterize a Subgroup of Muscle-invasive Bladder Cancers with Excellent Response to Neoadjuvant Chemotherapy. Eur Urol. (2016) ;69: :384–8. doi:10.1016/j.eururo.2015.01.014

[287] 

Van Allen EM , Mouw KW , Kim P , Iyer G , Wagle N , Al-Ahmadie H , et al. Somatic ERCC2 mutations correlate with cisplatin sensitivity in muscle-invasive urothelial carcinoma. Cancer Discov. (2014) ;4: :1140–53. doi:10.1158/2159-8290.CD-14-0623

[288] 

Choudhury A , Nelson LD , Teo MTW , Chilka S , Bhattarai S , Johnston CF , et al. MRE11 Expression Is Predictive of Cause-Specific Survival following Radical Radiotherapy for Muscle-Invasive Bladder Cancer. Cancer Res. (2010) ;70: :7017–26. doi:10.1158/0008-5472.CAN-10-1202

[289] 

Laurberg JR , Brems-eskildsen AS , Nordentoft I , Fristrup N , Schepeler T , Ulh BP , et al. Expression of TIP60 (tat-interactive protein) and MRE11 (meiotic recombination 11 homolog) predict treatment-specific outcome of localised invasive bladder cancer. BJU Int. (2012) ;Dec: :E1228–36. doi:10.1111/j.1464-410X.2012.11564.x

[290] 

Meng W , Efstathiou J , Singh R , Mcelroy J , Volinia S , Cui R , et al. MicroRNA Biomarkers for Patients With Muscle- Invasive Bladder Cancer Undergoing Selective Bladder-Sparing Trimodality Treatment. Int J Radiat Oncol Biol Phys. (2019) ;104: :197–206. doi:10.1016/j.ijrobp.2018.12.028

[291] 

Tanaka H , Yoshida S . Impact of Immunohistochemistry-Based Subtypes in Muscle-Invasive Bladder Cancer on Response to Chemoradiation Therapy. Int J Radiat Oncol Biol Phys. (2018) ;102: :1408–16. doi: 10.1016/j.ijrob2018.06.030

[292] 

Efstathiou JA , Mouw KW , Gibb EA , Liu Y , Wu C , Drumm MR , et al. Impact of Immune and Stromal Infiltration on Outcomes Following Bladder-Sparing Trimodality Therapy for Muscle-Invasive Bladder Cancer. Eur Urol. (2019) ;76: :59–68. doi:10.1016/j.eururo.2019.01.011

[293] 

Antoni S , Ferlay J , Soerjomataram I , Znaor A , Jemal A , Bray F . Bladder Cancer Incidence and Mortality: A Global Overview and Recent Trends. Eur Urol. (2017) ;71: :96–108. doi:10.1016/j.eururo.2016.06.010

[294] 

Fernández MI , López JF , Vivaldi B , Coz F . Long-term impact of arsenic in drinking water on bladder cancer health care and mortality rates 20 years after end of exposure. J Urol. (2012) ;187: :856–61. doi: 10.1016/j.juro.2011.10.157

[295] 

Rushton L , Bagga S , Bevan R , Brown TP , Cherrie JW , Holmes P , et al. Occupation and cancer in Britain. Br J Cancer. (2010) ;102: :1428–37. doi:10.1038/sj.bjc.6605637

[296] 

Koutros S , Silverman DT , Baris D , Zahm SH , Morton LM , Colt JS , et al. Hair dye use and risk of bladder cancer in the New England bladder cancer study. Int J Cancer. (2011) ;129: :2894–904. doi:10.1002/ijc.26245

[297] 

Zhou J , Kelsey KT , Smith S , Giovannucci E , Michaud DS . Lower Urinary Tract Symptoms and Risk of Bladder Cancer in Men: Results From the Health Professionals Follow-up Study. Urology. (2015) ;85: :1312–8. doi:10.1016/j.urology.2015.02.024

[298] 

MacKenzie T , Zens MS , Ferrara A , Schned A , Karagas MR . Diabetes and risk of bladder cancer. Cancer. (2011) ;117: :1552–6. doi:10.1002/cncr.25641

[299] 

Abern MR , Dude AM , Tsivian M , Coogan CL . The characteristics of bladder cancer after radiotherapy for prostate cancer. Urol Oncol Semin Orig Investig. (2013) ;31: :1628–34. doi:10.1016/j.urolonc.2012.04.006

[300] 

Bedwani R , Renganathan E , Kwhsky F El , Braga C , Seif HHA , Azm TA , et al. Schistosomiasis and the risk of bladder cancer in Alexandria, Egypt. Br J Cancer. (1998) ;77: :1186–9.

[301] 

International collaboration of trialists. Neoadjuvant cisplatin, methotrexate, and vinblastine chemotherapy for muscle-invasive bladder cancer: a randomised controlled trial. Lancet. (1999) ;354: :533–40. doi:10.1016/S0140-6736(99)02292-8

[302] 

Advanced Bladder Cancer (ABC) Meta-Analysis Collaborators . Neoadjuvant Chemotherapy in Invasive Bladder Cancer: Update of a Systematic Review and Meta-Analysis of Individual Patient Data. Eur Urol. (2005) ;48: :202–6. doi:10.1016/j.eururo.2005.04.006

[303] 

Winquist E , Kirchner TS , Segal R , CHIN J , LUKKA H . Neoadjuvant Chemotherapy for Transitional Cell Carcinoma of the Bladder: A Systematic Review and Meta-Analysis. J Urol. (2004) ;171: :561–9. doi: 10.1097/01.ju.0000090967.08622.33

[304] 

Studer UE , Bacchi M , Biedermann C , Jaeger P , Kraft R , Mazzucchelli L , et al. Adjuvant cisplatin chemotherapy following cystectomy for bladder cancer: Results of a prospective randomized trial. J Urol. (1994) ;152: :81–4. doi:10.1016/S0022-5347(17)32822-7

[305] 

Lehmann J , Franzaring L , Thüroff J , Wellek S , Stöckle M . Complete long-term survival data from a trial of adjuvant chemotherapy vs control after radical cystectomy for locally advanced bladder cancer. BJU Int. (2006) ;97: :42–7. doi:10.1111/j.1464-410X.2006.05859.x

[306] 

Bono AV , Benvenuti C , Gibba A , Guazzeri S , Cosciani-Cunico S AG . Adjuvant chemotherapy in locally advanced bladder cancer. Final analysis of a controlled multicentre study. Acta Urol Ita. (1997) ;11: :5–8. doi:10.1002/14651858.CD006018/references

[307] 

Cognetti F , Ruggeri EM , Felici A , Gallucci M , Muto G , Pollera CF , et al. Adjuvant chemotherapy with cisplatin and gemcitabine versus chemotherapy at relapse in patients with muscle-invasive bladder cancer submitted to radical cystectomy: An italian, multicenter, randomized phase iii trial. Ann Oncol. (2012) ;23: :695–700. doi:10.1093/annonc/mdr354

[308] 

Stadler WM , Lerner SP , Groshen S , Stein JP , Shi SR , Raghavan D , et al. Phase III study of molecularly targeted adjuvant therapy in locally advanced urothelial cancer of the bladder based on p53 status. J Clin Oncol. (2011) ;29: :3443–9. doi:10.1200/JCO.2010.34.4028

Appendices

APPENDIX 1

APPENDIX 1

Select publications of adjuvant chemotherapy in MIBC

Publication, YearPhasenInclusion CriteriaExperimental ArmControl ArmMedian FUOSDFS
Skinner 1991 [139]III91pT3/T4 or pN+Cisplatin, doxorubicin, cyclophosphamide q28d×4 cyclesObservation14.5 years4.3 vs 2.4 years, HR 0.75, p = 0.0062HR 0.73, p = NS
Studer 1994 [304]III77M0Cisplatin q28d×3 cyclesObservation5.75 years5y OS 57% vs 54%, p = NSHR 1.02, p = NS
Stockle 1995 [140, 305]III49pT3b/T4a or pN+Methotrexate, vinblastine, cisplatin plus doxorubicin or epirubicinObservation14.8 years10y OS 26.9% vs 17.4%, HR 2.52, p = 0.00710y DFS 43.7% vs 13.0%, HR 2.84, p = 0.002
Freiha 1996 [215]III55pT3b/T4a or pN+Cisplatin, methotrexate, vinblastine q21d×4 cyclesObservation5.08 years63 vs 36 months, HR 0.78, p = 0.3237 vs 12 months, HR 46, p = 0.01
Bono 1997 [306]III93pT2-T4a, pN0Cisplatin methotrexate×4 cyclesObservation3.45 yearsHR 0.75, p = NSHR 0.65, p = NS
SOGUG 99/01 2010 [141]III142pT3/T4 (77%) or pN+(70%)Paclitaxel, gemcitabine, cisplatin q21d×4 cyclesObservation30 months5y OS 60% vs 31%, p < 0.0009p < 0.0001
Cognetti 2011 [307]III194pT2G3, pT3/T4, or N+GC q28d×4 cyclesObservation35 months5y OS 43.4% vs 53.7%, HR 1.29, p = 0.2442.3% vs 37.2%, HR 1.08, p = 0.70
Stadler 2011 [308]III114pT1/T2 N0M0MVAC×3 cyclesObservation64.8 monthsp = 0.89p = 0.62
Sternberg 2015 [142]III284pT3/T4 or N+GC or ddMVAC with GCSF×4 cyclesSame chemo×6 cycles at relapse7.0 years5y OS 53.6% vs 47.7%, HR 0.78, p = 0.135y DFS 47.6% vs 31.8%, HR 0.54, p < 0.0001
ABC Meta-analysis 2005 [89]491Clinical T2–T4aAdjuvant chemotherapyObservation5.2 yearsAbsolute improvement 9% at 3 years, HR 0.75, p = 0.019Absolute improvement 12% at 3 years, HR 0.68, p = 0.004
Leow Meta-analysis 2014 [143]945Clinical T2–T4aAdjuvant cisplatin-based chemotherapyObservationRange 30–69 monthsHR 0.77, p = 0.049HR 0.66, p = 0.014
Kim Meta-analysis 2017 [144]1546MIBC and RCAdjuvant cisplatin-based chemotherapyObservationRange 30–168 monthsHR 0.79, p = 0.004HR 0.64, p = 0.002

NS, nonsignificant; GC, gemcitabine cisplatin; ddMVAC, dose dense MVAC; GCSF, granulocyte stimulating factors.