NCCN Categories of Evidence and Consensus
Category 1: Based upon high-level evidence, there is uniform NCCN consensus that the intervention is appropriate.
Category 2A: Based upon lower-level evidence, there is uniform NCCN consensus that the intervention is appropriate.
Category 2B: Based upon lower-level evidence, there is NCCN consensus that the intervention is appropriate.
Category 3: Based upon any level of evidence, there is major NCCN disagreement that the intervention is appropriate.
All recommendations are category 2A unless otherwise noted.
Clinical trials: NCCN believes that the best management for any cancer patient is in a clinical trial. Participation in clinical trials is especially encouraged.
Overview
An estimated 79,030 new cases of urinary bladder cancer (60,490 men and 18,540 women) will be diagnosed in the United States in 2017 and approximately 16,870 deaths (12,240 men and 4630 women) will occur.1 Bladder cancer, the sixth most common cancer in the United States,1 is rarely diagnosed in individuals aged <40 years. Given that the median age at diagnosis is 73 years,2 medical comorbidities are a frequent consideration in patient management.
The clinical spectrum of bladder cancer can be divided into 3 categories that differ in prognosis, management, and therapeutic aims. The first category consists of non–muscle-invasive bladder cancer, for which treatment is directed at reducing recurrences and preventing progression to a more advanced stage. The second group encompasses muscle-invasive bladder cancer (MIBC). The goal of therapy is to determine whether the bladder should be removed or if it can be preserved without compromising survival, and to determine whether the primary lesion can be managed independently or if patients are at high risk for distant spread requiring systemic approaches to improve the likelihood of cure. The critical concern for the third group, consisting of metastatic lesions, is how to prolong quantity and maintain quality of life. Numerous agents with different mechanisms of action have antitumor effects on this disease. The goal is how to use these agents to achieve the best possible outcome.
Histology
More than 90% of urothelial tumors originate in the urinary bladder, 8% originate in the renal pelvis, and the remaining 2% originate in the ureter and urethra. Urothelial carcinomas are classified as low- or high-grade as defined by the extent of nuclear anaplasia and architectural abnormalities.
Urothelial (transitional cell) carcinomas are the most common histologic subtype in the United States and Europe and may develop anywhere
transitional epithelium is present, from the renal pelvis to the ureter, bladder, and proximal two-thirds of the urethra. Variant histology is common with higher grades. The fourth edition of the WHO Classification of Tumors has reclassified these histologic subtypes into the following: infiltrating urothelial carcinoma with divergent differentiation; nested, including large nested; microcystic; micropapillary; lymphoepithelioma-like; plasmacytoid/signet ring cell/diffuse; sarcomatoid; giant cell; poorly differentiated; lipid-rich; and clear cell.3 Two review articles highlight the changes between the third and fourth additions of this classification.4,5 The presence of histologic variants in urothelial carcinoma should be documented, because data suggest that the subtype may reflect the risk of disease progression and different genetic origin, and subsequently determine whether a more aggressive treatment approach should be considered (see “Bladder Cancer: Non-Urothelial and Urothelial With Variant Histology” in the complete version of these guidelines, at NCCN.org). In some cases with a mixed histology, systemic treatment may only target cells of urothelial origin and the nonurothelial component can remain.Squamous cell neoplasms of the urothelial tract are a second histologic subtype, which constitute 3% of the urinary tumors diagnosed in the United States. In regions where Schistosoma is endemic, this subtype is more prevalent and may account for up to 75% of bladder cancer cases. The distal third of the urethra is dominated by squamous epithelium. The diagnosis of squamous cell tumors requires the presence of keratinization in the pathologic specimen.6 Squamous cell carcinoma of the bladder is morphologically indistinguishable from squamous cell carcinoma of other sites and generally presents at an advanced stage. The 3 variants within this subtype are pure squamous cell carcinoma, verrucous carcinoma, and squamous cell papilloma.
Other histologic subtypes derived from cells of urothelial origin include glandular neoplasms, epithelial tumors of the upper urinary tract, and tumors arising in a bladder diverticulum. Glandular neoplasms include adenocarcinoma and villous adenoma. Urachal tumors are nonurothelial tumors, most commonly adenocarcinomas, which arise from the urachal ligament and involve the midline/dome of the bladder secondarily.7 Tumors arising within the genitourinary tract but not of urothelial origin (eg, tumors of müllerian type, melanocytic tumors, mesenchymal tumors) are beyond the scope of these guidelines.
Muscle-Invasive Urothelial Bladder Cancer
Additional Workup
Several workup procedures are recommended to accurately determine clinical staging of MIBC. Laboratory studies, such as a complete blood cell count and chemistry profile, including alkaline phosphatase, must be performed, and the patient should be assessed for the presence of regional or distant metastases. This evaluation should include chest imaging and a bone scan in patients with symptoms or clinical suspicion of bone metastasis (eg, elevated alkaline phosphatase, focal bone pain). Imaging studies help assess the extent of tumor spread to lymph nodes or distant organs.8 An abdominal/pelvic CT or MRI is used to assess the local and regional extent of disease. Unfortunately, CT scans, ultrasound, and MRI cannot accurately predict the true depth of invasion.
The overwhelming majority of muscle-invasive tumors are high-grade urothelial carcinomas. Further treatment following initial transurethral resection of bladder tumor (TURBT) is often required for muscle-invasive tumors, although select patients may be treated with TURBT alone.9,10 Different treatment modalities are discussed herein, including radical cystectomy, partial cystectomy, neoadjuvant or adjuvant therapy, bladder-preserving approaches, and chemotherapy for advanced disease.
Radical Cystectomy
Radical surgical treatment of bladder cancer involves a cystoprostatectomy in men and a cystectomy and commonly a hysterectomy in women, followed by the formation of a urinary diversion. This surgery can be performed in an open or robotic manner. Prostatectomy includes removal of the prostate, seminal vesicles, proximal vas deferens, and proximal urethra. Hysterectomy should include removal of the uterus, ovaries, fallopian tubes, urethra, and part of the vagina. Forms of urinary diversion include an ileal conduit or directing urine to an internal urinary reservoir (such as a continent pouch), with drainage to the abdominal wall or the urethra (orthotopic neobladder). Relative contraindications to urethral drainage include Tis in the prostatic ducts or positive urethral margin. Orthotopic diversion or a neobladder provides the closest bladder function to that of a native bladder albeit with an increased risk for nighttime incontinence as well as urinary retention requiring intermittent self-catheterization.
Unfortunately, the accuracy of the staging cystoscopy, examination under anesthesia (EUA), and TURBT is modest, even when combined with cross-sectional imaging, and understaging is frequently encountered. A retrospective study of 778 patients with bladder cancer found that 42% were upstaged following cystectomy.11 A pelvic lymph node dissection (PLND) is considered an integral part of the surgical management of bladder cancer. A more extensive PLND, which may include the common iliac or even lower para-aortic or para-caval nodes, yields more nodes to be examined, increases yield of positive nodes, and may be associated with better survival and a lower pelvic recurrence rate.12–16 Patient factors that may preclude a PLND include severe scarring secondary to previous treatments or surgery, advanced age, or severe comorbidities.
Partial Cystectomy
In <5% of cases, an initial invasive tumor develops in an area of the bladder where an adequate margin of soft tissue and amount of noninvolved urothelium can be removed along with the tumor without compromising continence or significantly reducing bladder capacity. Partial cystectomy is most frequently recommended for lesions that develop on the dome of the bladder and have no associated Tis in other areas of the urothelium. Relative contraindications to this procedure are lesions that occur in the trigone or bladder neck. The requirement for a ureteral reimplantation, however, is not an absolute contraindication. Outcome data on partial cystectomy are varied and, in general, partial cystectomy is not considered the gold-standard surgical treatment of MIBC. Ideal candidates are patients with cancer in a diverticulum or with significant medical comorbidities.
Similar to radical cystectomy, partial cystectomy begins with a laparotomy (intraperitoneal) and resection of the pelvic lymph nodes. Alternatively, partial cystectomy may be safely performed laparoscopically. If the intraoperative findings preclude a partial cystectomy, a radical cystectomy is performed. The decision to recommend adjuvant radiation or chemotherapy is based on the pathologic stage (ie, positive nodes or perivesical tissue involvement) or presence of a positive margin, similar to that for patients who undergo a radical cystectomy.
Neoadjuvant Chemotherapy
One of the most noteworthy issues in treatment is the optimal use of perioperative chemotherapy for MIBC. Data support the role of neoadjuvant chemotherapy before cystectomy for T2, T3, and T4a lesions without nodal involvement.17–22 In a SWOG randomized trial of 307 patients with MIBC, radical cystectomy alone versus 3 (28-day) cycles of neoadjuvant methotrexate, vinblastine, doxorubicin, and cisplatin (MVAC) followed by radical cystectomy were compared. Neoadjuvant chemotherapy increased median survival (77 vs 46 months; P=.06) and lowered the rate of residual disease (15% vs 38%; P<.001) with no apparent increase in treatment-related morbidity or mortality.17 In a meta-analysis of 11 trials involving 3,005 patients, cisplatin-based multiagent neoadjuvant chemotherapy was associated with improved 5-year overall survival (OS) and disease-free survival (DFS; 5% and 9% absolute improvement, respectively).21
Since the neoadjuvant trial with MVAC, the use of dose-dense MVAC (ddMVAC) with growth factor support in the metastatic setting has been shown to have good comparable tolerance with an increased complete response (CR) rate compared with standard (28-day) dosing of MVAC (11% vs 25%; 2-sided P=.006).23 Based on these findings, dd-MVAC has also been investigated in the neoadjuvant setting. In a multicenter prospective phase II trial, patients with cT2 to cT4a tumor staging and N0 or N1 MIBC (n=44) were given 3 cycles of dd-MVAC with pegfilgrastim followed by radical cystectomy and lymph node dissection.24 ddMVAC was anticipated to have a safer profile, a shorter time to surgery, and a similar pathologic CR rate compared with historical control data for neoadjuvant MVAC chemotherapy given in previous studies. Patients receiving ddMVAC had no grade 3 or 4 renal toxicities and no toxicity-related deaths. Grade 1 or 2 treatment-related toxicities were seen in 82% of patients. The median time to cystectomy was 9.7 weeks from start of chemotherapy.24 A separate single-arm phase II study also reported pathologic downstaging in 49% of patients receiving neoadjuvant ddMVAC with a similar safety profile.25 An additional neoadjuvant clinical trial of ddMVAC with bevacizumab reported 5-year survival outcomes of 63% and 64% (OS and disease-specific survival, respectively; median follow-up, 49 months), with pT0N0 and ≤pT1N0 downstaging rates of 38% and 53%, respectively.26 Bevacizumab had no definitive impact on overall outcomes. An international, multicenter, randomized trial (BA06 30894) investigating the effectiveness of neoadjuvant cisplatin, methotrexate, and vinblastine (CMV) in 976 patients showed a 16% reduction in mortality risk (hazard ratio [HR], 0.84; 95% CI, 0.72–0.99; P=.037) at a median follow-up of 8 years.22
The NCCN Panel strengthened the recommendations for neoadjuvant chemotherapy for patients with cT2, cT3, and cT4a bladder cancer without nodal disease and for adjuvant chemotherapy for patients with pT3 or pT4 disease or positive nodes (see cT2, Primary and Adjuvant Treatment [page 1242] and cT3, cT4a, Primary and Adjuvant Treatment [page 1243]). Neoadjuvant chemotherapy followed by radical cystectomy is a category 1 recommendation. Patients with hearing loss or neuropathy, poor performance status, or renal insufficiency may not be eligible for cisplatin-based chemotherapy. If neoadjuvant cisplatin-based chemotherapy cannot be given, neoadjuvant chemotherapy is not recommended. For patients with borderline renal function or minimal dysfunction, a split-dose administration of cisplatin may be considered (category 2B). Although split-dose is a safer alternative, the relative efficacy remains undefined.
Adjuvant Chemotherapy
Data are less clear regarding the role of adjuvant systemic chemotherapy in invasive bladder cancer. Studies have shown that adjuvant chemotherapy may delay recurrences and improve OS,27–29 but no randomized comparisons of adequate sample size have definitively shown a survival benefit, in large part due to poor accrual.30 Clinical trials of adjuvant chemotherapy with cyclophosphamide, doxorubicin, and cisplatin (CAP); MVAC; and methotrexate, vinblastine, epirubicin, and cisplatin (MVEC) regimens have each suggested a survival advantage.31–33 However, methodologic issues call into question the applicability of these studies to all patients with urothelial tumors. In the MVEC trial, patients who experienced relapse in the control arm did not receive chemotherapy, which is not typical of more contemporary treatment approaches. Many of these trials were not randomized, raising the question of selection bias in the analysis of outcomes.
A meta-analysis of 6 trials found a 25% mortality reduction with adjuvant chemotherapy, but the authors pointed out several limitations of the data and concluded that evidence is insufficient for treatment decisions.34 Interestingly, the follow-up analysis included 3 more studies for a total of 9 trials (N=945 patients).29 A 23% risk reduction for death was observed in the updated analysis (HR, 0.77; 95% CI, 0.59–0.99; P=.049) and improved DFS was achieved (HR, 0.66; 95% CI, 0.45–0.91; P=.014). Patients with node-positive disease had an even greater DFS benefit.29 An observational study evaluated 5,653 patients, of which 23% received adjuvant chemotherapy after cystectomy.28 Patients who received adjuvant chemotherapy had an improved OS (HR, 0.70; 95% CI, 0.06–0.76).28 Although evidence for adjuvant therapy is not as strong as for neoadjuvant therapy, the growing body of data support the administration of adjuvant chemotherapy for patients with a high risk for relapse who did not receive neoadjuvant therapy.
The NCCN Guidelines suggest that adjuvant chemotherapy may be given to patients with high-risk pathology who did not receive neoadjuvant chemotherapy, and it is considered a category 2A recommendation. For highly select patients who receive a partial cystectomy, neoadjuvant chemotherapy is a category 2A recommendation, with the option of adjuvant chemotherapy for patients who did not receive neoadjuvant chemotherapy.
A minimum of 3 cycles of a cisplatin-based combination, such as ddMVAC; gemcitabine plus cisplatin (GC); or CMV, may be used in patients undergoing perioperative chemotherapy. Regimen and dosing recommendations are mainly based on studies in advanced disease.17,22,35–37 Carboplatin has not demonstrated a survival benefit and should not be substituted for cisplatin in the perioperative setting. It should be noted that patients with tumors that are ≤pT2 and have no nodal involvement or lymphovascular invasion after cystectomy are considered to have lower risk and are not recommended to receive adjuvant chemotherapy.
Adjuvant Radiation
Patients with locally advanced disease (pT3–4) have high rates of pelvic failure and poor OS after radical cystectomy, pelvic lymph node dissection, and perioperative chemotherapy (pelvic failure, 20%–45% and survival, 10%–50% at 5 years, depending on risk factors factors).38–40 There is an interest in using adjuvant radiation to improve these outcomes, but data are limited and further prospective studies are needed to confirm its benefits. One older randomized study of 236 patients with pT3a to pT4a bladder cancer demonstrated improvement in 5-year DFS and local control compared with surgery alone.41 A more recent randomized phase II trial comparing adjuvant sequential chemotherapy and radiation versus adjuvant chemotherapy alone in 120 patients with locally advanced disease (pT3–4 or node-positive) showed a significant improvement in local control for chemoradiation (3-year local control of 96% vs 69%); however, the improvement in DFS and OS was not significant. Late grade ≥3 gastrointestinal toxicity on the chemoradiation arm was low.42
Although no conclusive data demonstrate improvements in overall survival, it is reasonable to consider adjuvant radiation in patients with pT3/pT4 pN0–2 urothelial bladder cancer after radical cystectomy. Patients meeting these characteristics with positive surgical margins and/or lymph nodes identified in the pelvic dissection have especially high pelvic failure rates (40%–45% by 5 years), and adjuvant radiation is reasonably well tolerated and improves pelvic failure rates. Radiation with a dose range of 45 to 50.4 Gy without concurrent chemotherapy may be used. In patients who have not had prior neoadjuvant chemotherapy, it may be reasonable to sandwich adjuvant radiation between cycles of adjuvant chemotherapy.42 The safety and efficacy of concurrent sensitizing chemotherapy and radiation in the adjuvant setting needs to be further studied.
Bladder Preservation
All bladder-sparing approaches are based on the principle that not all cases require an immediate cystectomy, and the decision to remove the bladder can be deferred until the response to organ-sparing therapy is assessed. Bladder-preserving approaches are reasonable alternatives to cystectomy for patients who are medically unfit for surgery and those seeking an alternative to radical cystectomy. Combined modality chemoradiation therapy as an alternative to immediate cystectomy for MIBC is endorsed by multiple international organizations that have developed evidence-based consensus guidelines and recommendations including the International Consultation on Urologic Diseases-European Association of Urology (ICUD-EAU), UK National Institute for Health and Care Excellence (NICE), and the AUA/ASCO/ASTRO/SUO.43–45 There is an apparent underutilization of aggressive bladder-preserving therapies for noncystectomy candidates, especially the elderly and racial minorities.46,47 Between 23% and 50% of patients with MIBC who are ≥65 years of age receive no treatment or nonaggressive therapy.
With any of the alternatives to cystectomy, there is a concern that bladders that appear to be endoscopically free of tumor based on a clinical assessment (cT0) that includes a repeat TURBT may not be pathologically free of tumor (pT0). Reports have suggested that up to 45% of bladders may be clinically understaged after TURBT.47–49 Conversely, one series reported that all patients who achieved a CR after radiotherapy (RT) with concurrent cisplatin and 5-FU were pT0 on immediate cystectomy.50 Although studies report differing frequencies of residual disease after cytotoxic agents (either radiation or chemotherapy), there is consensus that the rate is lower for patients who present with T2 disease versus T3 disease, which should be considered when proposing a bladder-sparing approach.
The decision to use a bladder-preserving approach is partially based on the location of the lesion, depth of invasion, size of the tumor, status of the “uninvolved” urothelium, and status of the patient (eg, bladder capacity, bladder function, comorbidities). Bladder preservation as an alternative to cystectomy is generally reserved for patients with smaller solitary tumors, negative nodes, no carcinoma in situ, no tumor-related hydronephrosis, and good pretreatment bladder function. Patients who are medically fit for radical cystectomy but who have hydronephrosis are poor candidates for bladder-sparing procedures.51,52 Maximal TURBT with concurrent chemoRT should be given as primary treatment for these patients, with RT alone or TURBT alone reserved for select patients.
For patients who have tumor after reassessment, cystectomy is preferred if feasible. Close cystoscopic observation with TURBT alone, chemotherapy alone, and concurrent chemoRT (if no previous RT) are potential treatment options. When possible, bladder-sparing options should be chosen in the context of clinical trials.
RT With Concurrent Chemotherapy Following TURBT as Primary Treatment for MIBC: Several groups have investigated the combination of concurrent or sequential chemotherapy and RT after TURBT. First, an endoscopic resection that is as complete as possible is performed. Incomplete resection is an unfavorable prognostic factor for the ability to preserve the bladder.53–55
Radiation Therapy Oncology Group protocol 89-03 compared concurrent cisplatin and RT with or without 2 cycles of induction CMV.52 No difference in complete clinical response or 5-year OS was observed between the treatment arms. Other studies also reported no significant survival benefit for neoadjuvant chemotherapy before bladder-preserving chemotherapy with radiation therapy.54,56
Conversely, results from several prospective trials have demonstrated the effectiveness of this approach. In the RTOG 89-03 trial in which 123 patients with clinical stage T2–T4a were treated with RT plus concurrent cisplatin, with or without induction CMV, 5-year OS was approximately 49% in both arms.52 The subsequent RTOG 95-06 trial treated 34 patients with twice-daily irradiation and concurrent cisplatin and 5-FU and reported a 3-year OS rate of 83%.57 In the RTOG 97-06 trial, 47 patients received twice-daily irradiation and concurrent cisplatin, and also received adjuvant chemotherapy with CMV58; the 3-year OS rate was 61%. In the RTOG 99-06 study, 80 patients received twice-daily irradiation plus cisplatin and paclitaxel, followed by adjuvant cisplatin and gemcitabine; the 5-year OS rate was 56%.59 In RTOG 0233, 97 patients received twice-daily radiation with concurrent paclitaxel plus cisplatin or 5-FU plus cisplatin; the 5-year OS was 73%.60 Taken together, the CR rates ranged from 59% to 81%.
Up to approximately 80% of long-term survivors maintain an intact bladder, whereas others ultimately require radical cystectomy.51–59 A combined analysis of survivors from these 4 trials, with a median follow-up of 5.4 years, showed that combined-modality therapy was associated with low rates of late grade 3 toxicity (5.7% genitourinary and 1.9% gastrointestinal).61 No late grade 4 toxicities or treatment-related deaths were recorded.
Chemotherapy Following TURBT as Primary Treatment for MIBC
Chemotherapy alone is considered to be inadequate without additional treatment to the bladder and remains investigational. Studies showed that the proportions of pathologic CR rates in the bladder using neoadjuvant chemotherapy alone were only up to 38%.17 A higher proportion of bladders can be rendered tumor-free and therefore preserved when chemotherapy is combined with concurrent RT.
RT Following TURBT as Primary Treatment for MIBC: RT alone is inferior to RT combined with chemotherapy for patients with an invasive bladder tumor, and is not considered standard for patients who can tolerate combined therapy.62,63 In a randomized trial of 360 patients, RT with concurrent mitomycin C and 5-FU improved the 2-year locoregional DFS rate from 54% (RT alone) to 67% (P=.01), and 5-year OS rate from 35% to 48% (P=.16), without increasing grade 3/4 acute or late toxicity.63 Hence, RT alone is only indicated for those who cannot tolerate a cystectomy or chemotherapy because of medical comorbidities.
TURBT Alone as Primary Treatment for MIBC: TURBT alone may be an option for patients with cT2, cT3, or cT4a disease who are not candidates for cystectomy. TURBT alone may be curative in selected cases that include solitary lesions <2 cm that have minimally invaded the muscle. These cases should also have no associated in situ component, palpable mass, or associated hydronephrosis.64
If considered for TURBT alone, patients should undergo an aggressive re-resection of the site within 4 weeks of the primary procedure to ensure that no residual disease is present. If the repeat TURBT is negative for residual tumor, patients can be managed conservatively with repeat endoscopic evaluations and cytologies every 3 months until a relapse is documented. The stage of the lesion documented at relapse would determine further management decisions.
Treatment of T2, T3, and T4a Tumors
The critical issues in the management and prognosis of these patients are whether a palpable mass is appreciated at EUA and if the tumor has extended through the bladder wall. Tumors that are organ-confined (T2) have a better prognosis than those that have extended through the bladder wall into the perivesical fat (T3) and beyond. T4a tumors involve the prostatic stroma, uterus, or vagina and are typically surgically managed similar to T3 tumors.
Primary surgical treatment for cT2, cT3, and cT4a lesions with no nodal disease seen on abdominal/pelvic CT or MRI scan is a radical cystectomy and pelvic lymphadenectomy. Neoadjuvant chemotherapy is recommended (category 1). If no neoadjuvant cisplatin-based chemotherapy is given, postoperative adjuvant chemotherapy may be considered based on pathologic risk, such as positive nodes or pT3–T4 lesions.
Partial cystectomy along with neoadjuvant cisplatin-based chemotherapy can be considered for cT2 disease with a single tumor in a suitable location and no presence of Tis. Partial cystectomy is not an option for cT3 or cT4a patients. If no neoadjuvant therapy is given, adjuvant RT or chemotherapy based on pathologic risk (ie, positive nodes, positive margin, high-grade lesions, pT3–T4 lesions) may be considered.
Bladder preservation with maximal TURBT followed by concurrent chemoRT may be considered. Candidates for this bladder-sparing approach include patients with tumors that present without hydronephrosis or with tumors that allow a visibly complete or a maximally debulking TURBT. RT with concurrent cisplatin-based chemotherapy or 5-FU plus mitomycin as a radiosensitizer is the most common and well-studied chemoRT method used to treat MIBC.50–54,62,63,65 The following radiosensitizing regimens are recommended: cisplatin plus 5-FU; cisplatin plus paclitaxel; and 5-FU plus mitomycin C. Doublet chemotherapy is generally preferred. Cisplatin alone or low-dose gemcitabine (category 2B) may be considered as alternative regimens.
After a complete TURBT, 60 to 66 Gy of external-beam RT is administered. Two doses of concurrent radiosensitizing chemotherapy may be given on weeks 1 and 4 (although weekly schedules are possible as well). Alternatively, an induction RT dose of 40 to 45 Gy may be given following complete TURBT. The overall tumor status should be reassessed 3 weeks after radiation if 40 to 45 Gy was initially administered, or 2 to 3 months after if the full dose of 60 to 66 Gy was delivered. If no residual tumor is detected, appropriate options include observation or completion of RT up to 66 Gy. If residual disease is present, cystectomy is preferred.
In patients with extensive comorbid disease or poor performance status who are noncystectomy candidates, treatment options include concurrent chemoRT, RT alone, or TURBT alone. Based on high-level evidence showing superiority to RT alone, the NCCN Panel recommends chemoRT with cisplatin alone or 5-FU and mitomycin C.62,63 The overall tumor status should be reassessed 2 to 3 months after treatment. If no tumor is evident, the patient should be observed. If tumor is observed, chemotherapy, concurrent chemoRT (if no prior RT), palliative TURBT, or best supportive care may be given.
Treatment of T4b Disease or Positive Nodes
For patients with cT4b disease and negative nodes on abdominal/pelvic CT or MRI scans or biopsy, the primary treatment recommendation includes 2 to 3 courses of chemotherapy with or without RT followed by evaluation with cystoscopy, EUA, TURBT, and imaging of the abdomen and pelvis. If no evidence of tumor is present after primary treatment, consolidation chemotherapy or completion of definitive RT may be considered. If a partial radiation dose of 40 to 45 Gy was given as primary treatment, completion of definitive RT is recommended. Alternatively, adjuvant treatment with chemoRT may be initiated if the patient did not receive prior RT. In general, cT4b disease is considered unresectable. However, in patients with disease that responds to treatment, cystectomy may be an option if the tumor becomes technically resectable.
If residual disease is noted upon evaluation after primary therapy, systemic therapy or cystectomy is recommended. Systemic therapy may include a checkpoint inhibitor, chemoRT (if no prior RT), or a change in chemotherapy. Cystectomy, if feasible, is an option.
For patients with abnormal nodes documented by imaging, a biopsy should be considered, if technically possible, to confirm nodal spread. Patients with positive nodes should receive chemotherapy with or without radiation and should be evaluated with cystoscopy, EUA, TURBT, and abdominal/pelvic imaging. If no residual tumor is detected, patients may receive a radiation boost or a cystectomy. If tumor is still present following primary therapy, these patients should follow treatment of recurrent or persistent disease.
Follow-up
Results from a meta-analysis of 13,185 patients who have undergone cystectomy reported a 0.75% to 6.4% prevalence of upper tract recurrence.66 Surveillance by urine cytology or upper tract imaging detected recurrences in 7% and 30% of cases, respectively.
Follow-up after cystectomy should include urine cytology, liver function tests, creatinine, and electrolytes. Imaging of the chest, upper tracts, abdomen, and pelvis should be conducted at intervals based on the recurrence risk. Patients should be monitored annually for vitamin B12 deficiency if a continent urinary diversion was created. Consider urethral wash cytology for patients with an ileal conduit or continent catheterizable diversion, particularly if Tis was found within the bladder or prostatic urethra. For details of follow-up recommendations, see “Follow-up” in the complete version of these guidelines, at NCCN.org (page BL-E).
Follow-up after a partial cystectomy is similar to that for a radical cystectomy, with the addition of monitoring for relapse in the bladder by serial cytologic examinations and cystoscopies (may include selected mapping biopsy).
For patients who have a preserved bladder, there is a risk for recurrence in the bladder or elsewhere in the urothelial tract and distantly. Imaging studies and laboratory testing should be performed as outlined under post-cystectomy follow-up. Additionally, continued monitoring of the urothelium with cystoscopy and urinary cytologies with or without mapping biopsy is a routine part of the management of all cases in which the bladder is preserved.
Recurrence or Persistent Disease
Metastatic disease or local recurrence may be managed with cystectomy, systemic therapy, or palliative TURBT and best supportive care.
A positive cytology with no evidence of disease in the bladder should prompt retrograde selective washings of the upper tract and a biopsy of the prostatic urethra. If the results are positive, patients are managed as described in the following sections for treatment of upper genitourinary tract tumors or urothelial carcinoma of the prostate.
For patients with a preserved bladder, local recurrence or persistent disease should be evaluated as a new cancer. Recurrences are treated based on the extent of disease at relapse, with consideration of prior treatment. As previously discussed, Tis, Ta, or T1 tumors are generally managed with intravesical bacillus Calmette-Guérin (BCG) therapy or cystectomy. If no response is noted after BCG treatment, a cystectomy is advised. Invasive disease is generally managed with radical cystectomy, and a second attempt at bladder preservation is not advisable. Cystectomy may not be possible in a patient who has undergone a full course of external-beam RT and has bulky residual disease. For these patients, palliative TURBT and best supportive care is advised.
Subsequent-line therapy for metastatic disease or local recurrence includes checkpoint inhibitors, chemotherapy, chemoRT (if no previous RT) or RT (see “Follow-up” in the complete version of these guidelines, at NCCN.org [BL-E], “Recurrent or Persistent Disease” on page 1246, and “Metastatic Urothelial Bladder Cancer,” below).
Metastatic Urothelial Bladder Cancer
Approximately 4% of patients have metastatic disease at the time of diagnosis.2 Additionally, about half of all patients relapse after cystectomy depending on the pathologic stage of the tumor and nodal status. Local recurrences account for approximately 10% to 30% of relapses, whereas distant metastases are more common.
Evaluation of Metastatic Disease
If metastasis is suspected, additional workup to evaluate the extent of the disease is necessary. This includes a chest CT and a bone scan if enzyme levels are abnormal or the patient shows signs or symptoms of skeletal involvement. Central nervous system imaging should be considered. An estimated glomerular filtration rate (GFR) should be obtained to assess patient eligibility for cisplatin. If the evidence of spread is limited to nodes, nodal biopsy should be considered and patients should be managed as previously outlined for positive nodal disease (see “Treatment of cT4b Disease or Positive Nodes,” page 1259, and cT4b, Primary and Adjuvant Treatment, page 1244). Patients who present with disseminated metastatic disease are generally treated with systemic chemotherapy. Management of persistent disseminated disease may involve chemotherapy, radiation, or a combination.
Chemotherapy for Metastatic Disease
The specific chemotherapy regimen recommended partially depends on the presence or absence of medical comorbidities, such as cardiac disease and renal dysfunction, along with the risk classification of the patient based on disease extent. In general, long-term survival with combination chemotherapy alone has been reported only in good-risk patients, defined as those with good performance status, no visceral (ie, liver, lung) or bone disease, and normal alkaline phosphatase or lactic dehydrogenase levels. Poor-risk patients, defined as those with poor performance status or visceral disease, have consistently shown very poor tolerance to multiagent combination programs and few complete remissions, which are prerequisites for cure.
GC67,68 and ddMVAC23,35 are commonly used in combinations that have shown clinical benefit. A large, international, phase III study randomized 405 patients with locally advanced or metastatic disease to GC or standard (28-day) MVAC.37 At a median follow-up of 19 months, OS and time to progression were similar in the 2 arms. Fewer toxic deaths were recorded among patients receiving GC compared with MVAC (1% vs 3%), although this did not reach statistical significance. A 5-year update analysis confirmed that GC was not superior to MVAC in terms of survival (OS, 13.0% vs 15.3% and PFS, 9.8% vs 11.3%, respectively).68 Another large, randomized, phase III trial compared ddMVAC with standard (28-day) MVAC.23,35 At a median follow-up of 7.3 years, 24.6% of patients were alive in the ddMVAC cohort compared with 13.2% in the standard MVAC cohort. There was 1 toxic death in each arm, but less overall toxicity was seen in the dose-dense group. From these data, ddMVAC had improved toxicity and efficacy compared with standard MVAC; therefore, standard (28-day) MVAC is no longer used. Both GC and ddMVAC with growth factor support are category 1 recommendations for metastatic disease. Alternative first-line regimens also include carboplatin or taxane-based regimens (category 2B) or single-agent chemotherapy (category 2B).
The performance status of the patient is a major determinant in the selection of a regimen. Regimens with lower toxicity profiles are recommended in patients with compromised liver or renal status or serious comorbid conditions. In patients who are not cisplatin-eligible, atezolizumab or pembrolizumab are now appropriate first-line options (see “Targeted Therapies,” page 1262). Alternatively, carboplatin may be substituted for cisplatin in the metastatic setting for cisplatin-ineligible patients, such as those with a GFR <60 mL/min. A phase II/III study assessed 2 carboplatin-containing regimens in medically unfit patients (performance status 2).69 The overall response rate was 42% for gemcitabine plus carboplatin and 30% for methotrexate, carboplatin, and vinblastine. However, the response rates dropped to 26% and 20%, respectively, with increased toxicity among patients who were both unfit and had renal impairment (GFR <60 mL/min).
Taxanes have been shown to be active as both front-line and palliative therapies. Based on these results, several groups are exploring 2- and 3-drug combinations using these agents, with and without cisplatin, as initial therapy. A randomized phase III trial was conducted to compare GC and GC plus paclitaxel in 626 patients with locally advanced or metastatic urothelial cancer.70 The addition of paclitaxel to GC resulted in higher response rates and a borderline OS advantage, which was not statistically significant in the intent-to-treat analysis. Analysis of eligible patients only (92%) resulted in a small (3.2 months) but statistically significant survival advantage in favor of the 3-drug regimen (P=.03); there was no difference in PFS. The incidence of neutropenic fever was substantially higher with the 3-drug combination (13.2% vs 4.3%; P<.001). Panelists feel that the risk of adding paclitaxel outweighs the limited benefit reported from the trial. The alternative regimens, including cisplatin/paclitaxel,71 gemcitabine/paclitaxel,72 cisplatin/gemcitabine/paclitaxel,73 carboplatin/gemcitabine/paclitaxel,74 and cisplatin/gemcitabine/docetaxel,75 have shown modest activity in patients with bladder cancer in phase I–II trials. Category 1 level evidence now supports the use of checkpoint inhibitors in patients with advanced disease previously treated with a platinum-containing regimen (see “Targeted Therapies,” page 1262).
Although current data are insufficient to recommend the above alternative regimens as routine first-line options, non-cisplatin-containing regimens may be considered in patients who cannot tolerate cisplatin because of renal impairment or other comorbidities (see “Principles of Systemic Therapy,” page 1249–1251). Additionally, 2 checkpoint inhibitors, atezolizumab and pembrolizumab, have been FDA-approved for use as a first-line therapy in these patients. Consideration of checkpoint inhibitors must be integrated into the therapeutic planning for all patients with locally advanced and metastatic disease (see “Targeted Therapies,” next column). The NCCN Panel recommends enrollment in clinical trials of potentially less toxic therapies.
Independent of the specific regimen used, patients with metastatic disease are re-evaluated after 2 to 3 cycles of chemotherapy, and treatment is continued for 2 more cycles in patients whose disease responds or remains stable. Chemotherapy may be continued for a maximum of 6 cycles, depending on response. If no response is noted after 2 cycles or if significant morbidities are encountered, a change in therapy is advised, taking into account the patient's current performance status, extent of disease, and specific prior therapy. A change in therapy is also advised for patients who experience systemic relapse after adjuvant chemotherapy.
Studies have shown that surgery or RT may be feasible in highly select cases for patients who show a major partial response in a previously unresectable primary tumor or who have a solitary site of residual disease that is resectable after chemotherapy. In selected series, this approach has been shown to afford a survival benefit. If disease is completely resected, 2 additional cycles of chemotherapy can be considered, depending on patient tolerance.
Clinical trial enrollment is recommended by the NCCN Panel for all patients when appropriate, but is strongly recommended for subsequent-line therapy, because data for locally advanced or metastatic disease treated with subsequent-line therapy are highly variable. The available options depend on what was given as first line. Regimens used in this setting include checkpoint inhibitors, and the following chemotherapies: docetaxel, paclitaxel, gemcitabine, or pemetrexed monotherapy.76–79 Other options include nab-paclitaxel; ifosfamide; methotrexate; ifosfamide, doxorubicin, and gemcitabine; gemcitabine and paclitaxel; GC; and ddMVAC.
Chemoradiotherapy for Metastatic Disease
Chemotherapy is sometimes combined with palliative radiation to treat metastases or pelvic recurrence after cystectomy. However, concurrent chemotherapy is inappropriate if high-dose radiation (>3 Gy fractions) is used. The radiosensitizing chemotherapy regimens remain controversial in this setting. Possible options include cisplatin (category 2A); docetaxel or paclitaxel (category 2B); 5-FU with or without mitomycin C (category 2B); capecitabine (category 3); and low-dose gemcitabine (category 2B). RT alone can also be considered as a subsequent-line therapy for patients with metastatic disease.
Targeted Therapies
Platinum-based chemotherapy has been the standard of care in patients with metastatic disease, with an OS of 9 to 15 months.68,80 However, in patients with disease that relapses after this type of chemotherapy, the median survival is reduced to 5 to 7 months.81 Several new agents, notably checkpoint inhibitors for the treatment of metastatic urothelial carcinoma, have data supporting improved outcomes compared with standard therapies. Cancers with higher rates of somatic mutations have been shown to respond better to checkpoint inhibitors.82–87 Data from The Cancer Genome Atlas rank bladder cancer as the third highest mutated cancer,88,89 suggesting that checkpoint inhibitors may have a substantial impact as a treatment option for this cancer.
The FDA has approved the PD-L1 inhibitors atezolizumab, durvalumab, and avelumab as well as the PD-1 inhibitors nivolumab and pembrolizumab for patients with urothelial carcinoma. Pembrolizumab, atezolizumab, nivolumab, durvalumab, and avelumab are approved for the treatment of locally advanced or metastatic urothelial cell carcinoma that has progressed during or after platinum-based chemotherapy or that has progressed within 12 months of neoadjuvant or adjuvant platinum-containing chemotherapy, regardless of PD-L1 expression levels. Additionally, atezolizumab and pembrolizumab are approved as a first-line treatment option for patients with locally advanced or metastatic urothelial cell carcinoma who are not eligible for cisplatin-containing chemotherapy. All of these approvals as a subsequent treatment option have been based on category 2 level evidence, with the exception of pembrolizumab, which has category 1 level evidence supporting the approval.
Pembrolizumab is a PD-1 inhibitor that has been evaluated as second-line therapy for patients with bladder cancer who previously received platinum-based therapy and subsequently progressed or metastasized.90 An open-label, randomized, phase III trial compared pembrolizumab versus chemotherapy (paclitaxel, docetaxel, or vinflunine) in 542 patients with advanced urothelial carcinoma that recurred or progressed after platinum-based chemotherapy. Data from this trial showed a longer median OS for patients treated with pembrolizumab compared with chemotherapy (10.3 vs 7.4 months; P=.002). In addition, fewer grade 3, 4, or 5 treatment-related adverse events (AEs) occurred in the pembrolizumab-treated patients compared with those treated with chemotherapy (15.0% vs 49.4%).91 Results from this phase III trial have lead the NCCN Panel to assign pembrolizumab a category 1 recommendation as a second-line therapy. A phase II trial evaluated pembrolizumab as a first-line therapy in 370 patients with advanced urothelial carcinoma who were ineligible for cisplatin-based therapy. Data from this study showed an overall response rate of 29%, with 7% of patients achieving a CR. Grade 3 or 4 treatment-related AEs occurred in 18% of patients treated with pembrolizumab.92
Data from a 2-cohort, multicenter, phase II trial evaluated atezolizumab in patients with metastatic disease. Cohort 2 of the trial enrolled 310 patients with metastatic urothelial carcinoma post-platinum treatment and showed a significantly improved overall response rate compared with historical controls (15% vs 10%; P=.0058).93 Follow-up to date suggests these responses may be durable, with ongoing responses recorded in 38 (84%) of 45 responders with a median follow-up of 11.7 months. Although a similar response rate was seen regardless of PD-L1 status of tumor cells, a greater response was associated with increased PD-L1 expression status on infiltrating immune cells in the tumor microenvironment. Grade 3 or 4 treatment-related or immune-mediated AEs occurred in 16% and 5% of patients, respectively. Furthermore, there were no treatment-related deaths in this trial, suggesting good tolerability. In cohort 1 of the same phase II trial, atezolizumab was evaluated as a first-line therapy in 119 patients with locally advanced or metastatic urothelial carcinoma who were ineligible for cisplatin. Data from this study showed an objective response rate (ORR) of 23%, with 9% of patients showing a CR. Median OS was 15.9 months, and grade 3 or 4 treatment-related AEs occurred in 16% of patients.94 A May 2017 press release reported that the phase III IMvigor211 study evaluating atezolizumab compared with chemotherapy in patients with metastatic urothelial carcinoma postplatinum treatment did not meet its primary end point of OS.95 Further examination of the data is ongoing to better understand the results and define the role of atezolizumab as a post-platinum treatment option for metastatic urothelial carcinoma.
A phase II trial in patients with locally advanced or metastatic urothelial carcinoma that progressed after at least 1 platinum-containing regimen reported an overall objective response in 52 of 265 patients (19.6%; 95% CI, 15.0–24.9) after treatment with nivolumab, which was unaffected by PD-1 tumor status.96 Of the 270 patients enrolled in the study, grade 3 or 4 treatment-related AEs were reported in 18%, and 3 resulted from treatment.96 The median OS was 8.74 months (95% CI, 6.05–not yet reached). Based on PD-L1 expression of <1% and ≥1%, OS was 5.95 to 11.3 months, respectively. These data are comparable to the early phase I/II data reporting an ORR of 24% (95% CI, 15.3%–35.4%) that was unaffected by PD-1 tumor status.97 Of the 78 patients enrolled in this study, 2 experienced grade 5 treatment-related AEs, and grade 3 or 4 treatment-related AEs were reported in 22% of patients.97
Early results from a phase I/II multicenter study of durvalumab for 61 patients with PD-L1–positive inoperable or metastatic urothelial bladder cancer whose tumor had progressed during or after one standard platinum-based regimen showed that 46.4% of patients who were PD-L1–positive had disease that responded to treatment; no response was seen in patients who were PD-L1–negative.98 A 2017 update on this study (N=103) showed a 29.5% ORR for PD-L1–high disease and a 7.7% ORR for PD-L1–low/negative disease. The OS rate at 6 months was 68.4% for the PD-L1–high group and 44.7% for the PD-L1–low/negative group. Median duration of response was not yet reached at the time of data cut-off. Grade 3 or 4 treatment-related AEs occurred in 5.2% of treated patients and 3 patients had a grade 3 or 4 immune-mediated AE.99
Avelumab is another PD-L1 inhibitor currently in clinical trials to evaluate its activity in the treatment of bladder cancer. Results from the phase 1b trial for 44 patients with platinum-refractory disease demonstrated an ORR of 18.2% that consisted of 5 CRs and 3 partial responses following treatment with avelumab. The median PFS was 11.6 weeks and median OS was 13.7 months, with a 54.3% OS rate at 12 months. Grade 3 or 4 treatment-related AEs occurred in 6.8% of patients treated with avelumab.100 A recent abstract reported results of the same trial for 241 patients with platinum-refractory metastatic urothelial carcinoma or who were ineligible for cisplatin-based chemotherapy. This study reported an ORR of 17.6%, with 9 CRs and 18 partial responses. Median PFS was 6.4 weeks and median OS was 7.0 months. Grade 3 or 4 treatment-related AEs occurred in 7.5% of patients treated with avelumab, and 2.5% of patients had a grade 3 or 4 immune-related AE.101
The value of checkpoint inhibitors is reflected in the unanimous decision by the NCCN Panel to include pembrolizumab, atezolizumab, nivolumab, durvalumab, and avelumab as second-line systemic therapy options after platinum-based therapy (and, in the case of atezolizumab and pembrolizumab, as first-line therapy options for patients who are not eligible for cisplatin-containing chemotherapy) for locally advanced or metastatic disease (see “Systemic Therapy” on page 1249–1251). With the exception of pembrolizumab as a subsequent treatment option (category 1), the use of checkpoint inhibitors are all category 2A recommendations.
Summary
Urothelial tumors represent a spectrum of diseases with a range of prognoses. After a tumor is diagnosed anywhere within the urothelial tract, the patient remains at risk for developing a new lesion at the same or a different location and with a similar or more advanced stage. Continued monitoring for recurrence is an essential part of management, because most recurrences are superficial and can be treated endoscopically. Within each category of disease, more refined methods to determine prognosis and guide management, based on molecular staging, are under development, with the goal of optimizing each patient's likelihood of cure and chance for organ preservation.
For patients with more extensive disease, newer treatments typically involve combined modality approaches using recently developed surgical procedures or 3-dimensional treatment planning for more precise delivery of RT. Although these are not appropriate in all cases, they offer the promise of an improved quality of life and prolonged survival.
Finally, within the category of metastatic disease, several new agents have been identified that seem superior to those currently considered standard therapies. Checkpoint inhibitors, in particular, have emerged as a new therapy for the treatment of persistent disease. Experts surmise that the treatment of urothelial tumors will evolve rapidly over the next few years, with improved outcomes across all disease stages.
Individual Disclosures for Bladder Cancer Panel
References
- 2.↑
Cancer Stat Facts: Bladder Cancer. Available at: https://seer.cancer.gov/statfacts/html/urinb.html. Accessed July 25, 2017.
- 3.↑
Moch H, Humphrey PA, Ulbright TM, Reuter VE. Tumours of the Urinary Tract. In: WHO Classification of Tumours of the Urinary System and Male Genital Organs, 4th ed. Lyon, France: IARC; 2016.
- 4.↑
Humphrey PA, Moch H, Cubilla AL et al.. The 2016 WHO classification of tumours of the urinary system and male genital organs-part B: prostate and bladder tumours. Eur Urol 2016;70:106–119.
- 5.↑
Athanazio DA, Trpkov K. What is new in genitourinary pathology? Recent developments and highlights of the new 2016 World Health Organization classification of tumors of the urinary system and male genital organs. Applied Cancer Research 2016;36:1.
- 6.↑
Chalasani V, Chin JL, Izawa JI. Histologic variants of urothelial bladder cancer and nonurothelial histology in bladder cancer. Can Urol Assoc J 2009;3:S193–198.
- 7.↑
Siefker-Radtke A. Urachal adenocarcinoma: a clinician's guide for treatment. Semin Oncol 2012;39:619-624.
- 8.↑
Verma S, Rajesh A, Prasad SR et al.. Urinary bladder cancer: role of MR imaging. Radiographics 2012;32:371–387.
- 9.↑
Solsona E, Iborra I, Collado A et al.. Feasibility of radical transurethral resection as monotherapy for selected patients with muscle invasive bladder cancer. J Urol 2010;184:475–480.
- 10.↑
Leibovici D, Kassouf W, Pisters LL et al.. Organ preservation for muscle-invasive bladder cancer by transurethral resection. Urology 2007;70:473–476.
- 11.↑
Shariat SF, Palapattu GS, Karakiewicz PI et al.. Discrepancy between clinical and pathologic stage: impact on prognosis after radical cystectomy. Eur Urol 2007;51:137–149; discussion 149–151.
- 12.↑
Leissner J, Hohenfellner R, Thuroff JW, Wolf HK. Lymphadenectomy in patients with transitional cell carcinoma of the urinary bladder; significance for staging and prognosis. BJU Int 2000;85:817–823.
- 13.
Herr HW, Bochner BH, Dalbagni G et al.. Impact of the number of lymph nodes retrieved on outcome in patients with muscle invasive bladder cancer. J Urol 2002;167:1295–1298.
- 14.
Herr HW, Faulkner JR, Grossman HB et al.. Surgical factors influence bladder cancer outcomes: a cooperative group report. J Clin Oncol 2004;22:2781–2789.
- 15.
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;169:946–950.
- 16.↑
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;112:2401–2408.
- 17.↑
Grossman HB, Natale RB, Tangen CM et al.. Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer. N Engl J Med 2003;349:859–866.
- 18.
Sherif A, Holmberg L, Rintala E et al.. Neoadjuvant cisplatinum based combination chemotherapy in patients with invasive bladder cancer: a combined analysis of two Nordic studies. Eur Urol 2004;45:297–303.
- 19.
Winquist E, Kirchner TS, Segal R et al.. Neoadjuvant chemotherapy for transitional cell carcinoma of the bladder: a systematic review and meta-analysis. J Urol 2004;171:561–569.
- 20.
Vashistha V, Quinn DI, Dorff TB, Daneshmand S. Current and recent clinical trials for perioperative systemic therapy for muscle invasive bladder cancer: a systematic review. BMC Cancer 2014;14:966.
- 21.↑
Advanced Bladder Cancer Meta-analysis Collaboration. Neoadjuvant chemotherapy in invasive bladder cancer: update of a systematic review and meta-analysis of individual patient data. Eur Urol 2005;48:202–205; discussion 205–206.
- 22.↑
Griffiths G, Hall R, Sylvester R et al.. 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;29:2171–2177.
- 23.↑
Sternberg CN, de Mulder P, Schornagel JH et al.. Seven year update of an EORTC phase III trial of high-dose intensity M-VAC chemotherapy and G-CSF versus classic M-VAC in advanced urothelial tract tumours. Eur J Cancer 2006;42:50–54.
- 24.↑
Plimack ER, Hoffman-Censits JH, Viterbo R 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;32:1895–1901.
- 25.↑
Choueiri TK, Jacobus S, Bellmunt J 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;32:1889–1894.
- 26.↑
McConkey DJ, Choi W, Shen Y et al.. A prognostic gene expression signature in the molecular classification of chemotherapy-naive urothelial cancer is predictive of clinical outcomes from neoadjuvant chemotherapy: a phase 2 trial of dose-dense methotrexate, vinblastine, doxorubicin, and cisplatin with bevacizumab in urothelial cancer. Eur Urol 2016;69:855–862.
- 27.↑
Millikan R, Dinney C, Swanson D et al.. Integrated therapy for locally advanced bladder cancer: final report of a randomized trial of cystectomy plus adjuvant M-VAC versus cystectomy with both preoperative and postoperative M-VAC. J Clin Oncol 2001;19:4005–4013.
- 28.↑
Galsky MD, Stensland KD, Moshier E et al.. Effectiveness of adjuvant chemotherapy for locally advanced bladder cancer. J Clin Oncol 2016;34:825–832.
- 29.↑
Leow JJ, Martin-Doyle W, Rajagopal PS 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.
- 30.↑
Hussain MH, Wood DP, Bajorin DF et al.. Bladder cancer: narrowing the gap between evidence and practice. J Clin Oncol 2009;27:5680–5684.
- 31.↑
Lehmann J, Franzaring L, Thuroff J et al.. Complete long-term survival data from a trial of adjuvant chemotherapy vs control after radical cystectomy for locally advanced bladder cancer. BJU Int 2006;97:42–47.
- 32.
Stockle M, Wellek S, Meyenburg W et al.. Radical cystectomy with or without adjuvant polychemotherapy for non-organ-confined transitional cell carcinoma of the urinary bladder: prognostic impact of lymph node involvement. Urology 1996;48:868–875.
- 33.↑
Skinner DG, Daniels JR, Russell CA et al.. The role of adjuvant chemotherapy following cystectomy for invasive bladder cancer: a prospective comparative trial. J Urol 1991;145:459–464; discussion 464–457.
- 34.↑
Adjuvant chemotherapy in invasive bladder cancer: a systematic review and meta-analysis of individual patient data Advanced Bladder Cancer (ABC) Meta-analysis Collaboration. Eur Urol 2005;48:189–199; discussion 199–201.
- 35.↑
Sternberg CN, de Mulder PH, Schornagel JH et al.. Randomized phase III trial of high-dose-intensity methotrexate, vinblastine, doxorubicin, and cisplatin (MVAC) chemotherapy and recombinant human granulocyte colony-stimulating factor versus classic MVAC in advanced urothelial tract tumors: European Organization for Research and Treatment of Cancer Protocol no. 30924. J Clin Oncol 2001;19:2638–2646.
- 36.
Dash A, Pettus JA 4th, Herr HW et al.. A role for neoadjuvant gemcitabine plus cisplatin in muscle-invasive urothelial carcinoma of the bladder: a retrospective experience. Cancer 2008;113:2471–2477.
- 37.↑
von der Maase H, Hansen SW, Roberts JT 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;18:3068–3077.
- 38.↑
Novotny V, Froehner M, May M et al.. Risk stratification for locoregional recurrence after radical cystectomy for urothelial carcinoma of the bladder. World J Urol 2015;33:1753–1761.
- 39.
Ku JH, Kim M, Jeong CW et al.. Risk prediction models of locoregional failure after radical cystectomy for urothelial carcinoma: external validation in a cohort of korean patients. Int J Radiat Oncol Biol Phys 2014;89:1032–1037.
- 40.↑
Christodouleas JP, Baumann BC, He J et al.. Optimizing bladder cancer locoregional failure risk stratification after radical cystectomy using SWOG 8710. Cancer 2014;120:1272–1280.
- 41.↑
Zaghloul MS, Awwad HK, Akoush HH et al.. Postoperative radiotherapy of carcinoma in bilharzial bladder: improved disease free survival through improving local control. Int J Radiat Oncol Biol Phys 1992;23:511–517.
- 42.↑
Zaghloul MS, Christodouleas JP, Smith A et al.. Adjuvant sandwich chemotherapy and radiation versus adjuvant chemotherapy alone for locally advanced bladder cancer [abstract]. Int J Radiat Oncol Biol Phys 2016;96:Abstract S94.
- 43.↑
Gakis G, Efstathiou J, Lerner SP et al.. ICUD-EAU International Consultation on Bladder Cancer 2012: radical cystectomy and bladder preservation for muscle-invasive urothelial carcinoma of the bladder. Eur Urol 2013;63:45–57.
- 44.
Bladder cancer: diagnosis and management. National Institute for Health and Care Excellence Web site. Available at: https://www.nice.org.uk/guidance/ng2. Accessed August 7, 2017.
- 45.↑
Chang SS, Bochner BH, Chou R et al.. Treatment of non-metastatic muscle-invasive bladder cancer: AUA/ASCO/ASTRO/SUO guideline. J Urol 2017;198:552–559.
- 46.↑
Fedeli U, Fedewa SA, Ward EM. Treatment of muscle invasive bladder cancer: evidence from the National Cancer Database, 2003 to 2007. J Urol 2011;185:72–78.
- 47.↑
Gray PJ, Fedewa SA, Shipley WU et al.. Use of potentially curative therapies for muscle-invasive bladder cancer in the United States: results from the National Cancer Data Base. Eur Urol 2013;63:823–829.
- 48.
Herr HW, Bajorin DF, Scher HI. Neoadjuvant chemotherapy and bladder-sparing surgery for invasive bladder cancer: ten-year outcome. J Clin Oncol 1998;16:1298–1301.
- 49.↑
Splinter T, Denis L. Restaging procedures, criteria of response, and relationship between pathological response and survival. Semin Oncol 1990;17:606–612.
- 50.↑
Housset M, Maulard C, Chretien Y et al.. Combined radiation and chemotherapy for invasive transitional-cell carcinoma of the bladder: a prospective study. J Clin Oncol 1993;11:2150–2157.
- 51.↑
Shipley WU, Kaufman DS, Zehr E et al.. Selective bladder preservation by combined modality protocol treatment: long-term outcomes of 190 patients with invasive bladder cancer. Urology 2002;60:62–67; discussion 67–68.
- 52.↑
Shipley WU, Winter KA, Kaufman DS 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;16:3576–3583.
- 53.↑
Rodel C, Grabenbauer GG, Kuhn R et al.. Combined-modality treatment and selective organ preservation in invasive bladder cancer: long-term results. J Clin Oncol 2002;20:3061–3071.
- 54.↑
Efstathiou JA, Spiegel DY, Shipley WU et al.. Long-term outcomes of selective bladder preservation by combined-modality therapy for invasive bladder cancer: the MGH experience. Eur Urol 2012;61:705–711.
- 55.↑
Giacalone NJ, Shipley WU, Clayman RH 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;71:952–960.
- 56.↑
Zapatero A, Martin De Vidales C, Arellano R et al.. Long-term results of two prospective bladder-sparing trimodality approaches for invasive bladder cancer: neoadjuvant chemotherapy and concurrent radio-chemotherapy. Urology 2012;80:1056–1062.
- 57.↑
Kaufman DS, Winter KA, Shipley WU et al.. The initial results in muscle-invading bladder cancer of RTOG 95-06: phase I/II trial of transurethral surgery plus radiation therapy with concurrent cisplatin and 5-fluorouracil followed by selective bladder preservation or cystectomy depending on the initial response. Oncologist 2000;5:471–476.
- 58.↑
Hagan MP, Winter KA, Kaufman DS et al.. RTOG 97-06: 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;57:665–672.
- 59.↑
Kaufman DS, Winter KA, Shipley WU 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;73:833–837.
- 60.↑
Mitin T, Hunt D, Shipley WU 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 2 trial. Lancet Oncol 2013;14:863–872.
- 61.↑
Efstathiou JA, Bae K, Shipley WU et al.. Late pelvic toxicity after bladder-sparing therapy in patients with invasive bladder cancer: RTOG 89-03, 95-06, 97-06, 99-06. J Clin Oncol 2009;27:4055–4061.
- 62.↑
Coppin CM, Gospodarowicz MK, James K 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 Group. J Clin Oncol 1996;14:2901–2907.
- 63.↑
James ND, Hussain SA, Hall E et al.. Radiotherapy with or without chemotherapy in muscle-invasive bladder cancer. N Engl J Med 2012;366:1477–1488.
- 64.↑
Herr HW. Conservative management of muscle-infiltrating bladder cancer: prospective experience. J Urol 1987;138:1162–1163.
- 65.↑
Mak RH, Zietman AL, Heney NM et al.. Bladder preservation: optimizing radiotherapy and integrated treatment strategies. BJU Int 2008;102:1345–1353.
- 66.↑
Picozzi S, Ricci C, Gaeta M et al.. Upper urinary tract recurrence following radical cystectomy for bladder cancer: a meta-analysis on 13,185 patients. J Urol 2012;188:2046–2054.
- 67.↑
Kaufman D, Raghavan D, Carducci M et al.. Phase II trial of gemcitabine plus cisplatin in patients with metastatic urothelial cancer. J Clin Oncol 2000;18:1921–1927.
- 68.↑
von der Maase H, Sengelov L, Roberts JT 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;23:4602–4608.
- 69.↑
De Santis M, Bellmunt J, Mead G et al.. Randomized phase II/III trial assessing gemcitabine/carboplatin and methotrexate/carboplatin/vinblastine in patients with advanced urothelial cancer “unfit” for cisplatin-based chemotherapy: phase II—results of EORTC study 30986. J Clin Oncol 2009;27:5634–5639.
- 70.↑
Bellmunt J, von der Maase H, Mead GM et al.. Randomized phase III study comparing paclitaxel/cisplatin/gemcitabine and gemcitabine/cisplatin in patients with locally advanced or metastatic urothelial cancer without prior systemic therapy: EORTC Intergroup Study 30987. J Clin Oncol 2012;30:1107–1113.
- 71.↑
Burch PA, Richardson RL, Cha SS et al.. Phase II study of paclitaxel and cisplatin for advanced urothelial cancer. J Urol 2000;164:1538–1542.
- 72.↑
Meluch AA, Greco FA, Burris HA 3rd et al.. Paclitaxel and gemcitabine chemotherapy for advanced transitional-cell carcinoma of the urothelial tract: a phase II trial of the Minnie pearl cancer research network. J Clin Oncol 2001;19:3018–3024.
- 73.↑
Bellmunt J, Guillem V, Paz-Ares L et al.. Phase I-II study of paclitaxel, cisplatin, and gemcitabine in advanced transitional-cell carcinoma of the urothelium. Spanish Oncology Genitourinary Group. J Clin Oncol 2000;18:3247–3255.
- 74.↑
Hussain M, Vaishampayan U, Du W et al.. Combination paclitaxel, carboplatin, and gemcitabine is an active treatment for advanced urothelial cancer. J Clin Oncol 2001;19:2527–2533.
- 75.↑
Pectasides D, Glotsos J, Bountouroglou N et al.. Weekly chemotherapy with docetaxel, gemcitabine and cisplatin in advanced transitional cell urothelial cancer: a phase II trial. Ann Oncol 2002;13:243–250.
- 76.↑
Lorusso V, Pollera CF, Antimi M et al.. A phase II study of gemcitabine in patients with transitional cell carcinoma of the urinary tract previously treated with platinum. Italian Co-operative Group on Bladder Cancer. Eur J Cancer 1998;34:1208–1212.
- 77.
McCaffrey JA, Hilton S, Mazumdar M et al.. Phase II trial of docetaxel in patients with advanced or metastatic transitional-cell carcinoma. J Clin Oncol 1997;15:1853–1857.
- 78.
Papamichael D, Gallagher CJ, Oliver RT et al.. Phase II study of paclitaxel in pretreated patients with locally advanced/metastatic cancer of the bladder and ureter. Br J Cancer 1997;75:606–607.
- 79.↑
Vaughn DJ, Broome CM, Hussain M et al.. Phase II trial of weekly paclitaxel in patients with previously treated advanced urothelial cancer. J Clin Oncol 2002;20:937–940.
- 80.↑
De Santis M, Bellmunt J, Mead G et al.. Randomized phase II/III trial assessing gemcitabine/carboplatin and methotrexate/carboplatin/vinblastine in patients with advanced urothelial cancer who are unfit for cisplatin-based chemotherapy: EORTC study 30986. J Clin Oncol 2012;30:191–199.
- 81.↑
Bellmunt J, Theodore C, Demkov T et al.. Phase III trial of vinflunine plus best supportive care compared with best supportive care alone after a platinum-containing regimen in patients with advanced transitional cell carcinoma of the urothelial tract. J Clin Oncol 2009;27:4454–4461.
- 82.↑
Garon EB, Rizvi NA, Hui R et al.. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015;372:2018–2028.
- 83.
Brahmer J, Reckamp KL, Baas P et al.. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med 2015;373:123–135.
- 84.
Postow MA, Chesney J, Pavlick AC et al.. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med 2015;372:2006–2017.
- 85.
Larkin J, Chiarion-Sileni V, Gonzalez R et al.. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med 2015;373:23–34.
- 86.
Wolchok JD, Kluger H, Callahan MK et al.. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013;369:122–133.
- 87.↑
Le DT, Uram JN, Wang H et al.. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:2509–2520.
- 88.↑
Alexandrov LB, Nik-Zainal S, Wedge DC et al.. Signatures of mutational processes in human cancer. Nature 2013;500:415–421.
- 89.↑
Cancer Genome Atlas Research N. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 2014;507:315–322.
- 90.↑
Plimack ER, Bellmunt J, Gupta S et al.. Safety and activity of pembrolizumab in patients with locally advanced or metastatic urothelial cancer (KEYNOTE-012): a non-randomised, open-label, phase 1b study. Lancet Oncol 2017;18:212–220.
- 91.↑
Bellmunt J, de Wit R, Vaughn DJ et al.. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 2017;376:1015–1026.
- 92.↑
O'Donnell PH, Grivas P, Balar AV et al.. Biomarker findings and mature clinical results from KEYNOTE-052: first-line pembrolizumab (pembro) in cisplatin-ineligible advanced urothelial cancer (UC) [abstract]. J Clin Oncol 2017;35(Suppl):Abstract 4502.
- 93.↑
Rosenberg JE, Hoffman-Censits J, Powles T et al.. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 2016;387:1909–1920.
- 94.↑
Balar AV, Galsky MD, Rosenberg JE et al.. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet 2017;389:67–76.
- 95.↑
Roche provides update on phase III study of TECENTRIQ® (atezolizumab) in people with previously treated advanced bladder cancer. Available at: http://www.roche.com/media/store/releases/med-cor-2017-05-10.htm. Accessed July 26, 2017.
- 96.↑
Sharma P, Retz M, Siefker-Radtke A et al.. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol 2017;18:312–322.
- 97.↑
Sharma P, Bono P, Kim JW et al.. Efficacy and safety of nivolumab monotherapy in metastatic urothelial cancer: results from the phase I/II CheckMate 032 study [abstract]. J Clin Oncol 2016;34(Suppl):Abstract 4501.
- 98.↑
Massard C, Gordon MS, Sharma S et al.. Safety and efficacy of durvalumab (MEDI4736), an anti-programmed cell death ligand-1 immune checkpoint inhibitor, in patients with advanced urothelial bladder cancer. J Clin Oncol 2016;34:3119–3125.
- 99.↑
Powles T, O'Donnell PH, Massard C et al.. Updated efficacy and tolerability of durvalumab in locally advanced or metastatic urothelial carcinoma [abstract]. J Clin Oncol 2017;35(Suppl);Abstract 286.
- 100.↑
Apolo AB, Infante JR, Balmanoukian A et al.. Avelumab, an anti-programmed death-ligand 1 antibody, in patients with refractory metastatic urothelial carcinoma: results from a Mmulticenter, phase Ib study. J Clin Oncol 2017;35:2117–2124.
- 101.↑
Patel MR, Ellerton JA, Infante JR et al.. Avelumab in patients with metastatic urothelial carcinoma: pooled results from two cohorts of the phase 1b JAVELIN Solid Tumor trial [abstract]. J Clin Oncol 2017;35(Suppl):Abstract 330.