Abstract
The treatment landscape for bladder cancer has undergone a rapid evolution in the past five years with the approval of seven new agents. New classes of medications have improved outcomes for many patients who previously had limited treatment options, but there is still much to learn about how to optimize patient selection for these agents and the role of combination therapies. The aims of this review are to discuss these newly approved agents for bladder cancer and to feature promising drugs and combinations—including immune checkpoint inhibitors, targeted therapies, and antibody–drug conjugates—that are in development.
Keywords: urothelial carcinoma, metastatic, muscle-invasive bladder cancer, immunotherapy, targeted therapy, antibody-drug conjugate
Introduction
Bladder cancer is the sixth most common malignancy in the US, where over 80,000 new cases are diagnosed per year 1. Non-muscle-invasive bladder cancer (NMIBC) is typically managed with local therapy, including transurethral resection of bladder tumors (TURBT) and intravesical bacillus Calmette–Guérin (BCG) or chemotherapy. NMIBC has an excellent 5-year overall survival (OS) of 70 to 96% 1. However, there is also a high rate of recurrence and potential for disease progression 2. For muscle-invasive bladder cancer (MIBC), survival outcomes are significantly decreased and treatment, including cystectomy with perioperative chemotherapy or tri-modality therapy (TMT) that includes TURBT, chemotherapy, and radiation therapy, is more aggressive, whereas metastatic disease is generally managed with palliative systemic therapy and has a 5-year OS of about 5% 1.
Platinum-based chemotherapy has been the first-line treatment for metastatic bladder cancer for over 20 years. Overall response rates (ORRs) range from 40 to 50% but this response is generally short-lived 3. Until recently, options for second-line treatment or platinum-ineligible patients have been limited. However, since 2016, seven new agents have been approved by the US Food and Drug Administration (FDA) for locally advanced (LA) or metastatic urothelial carcinoma (mUC) and this has dramatically changed the treatment landscape. The aims of this review are to highlight these newly approved therapies and to discuss promising new treatment strategies for bladder cancer that are on the horizon.
Immune checkpoint inhibitors
The introduction of immunotherapy with agents targeting programmed cell death protein 1 or its ligand (anti-PD-[L]1) marked an important turning point in the management of bladder cancer. Currently, five anti-PD-(L)1 drugs are approved by the FDA for urothelial carcinoma: atezolizumab, avelumab, durvalumab, nivolumab, and pembrolizumab. Following initial success in the mUC setting, numerous trials now use these and other anti-PD-(L)1 agents alone and in combination across the continuum of bladder cancer.
Immunotherapy for metastatic urothelial carcinoma post-platinum
All five of the anti-PD-(L)1 agents for urothelial carcinoma are currently approved by the FDA as treatment for LA/mUC patients who have disease progression during or following platinum-based chemotherapy or within 12 months of neoadjuvant or adjuvant treatment for localized disease with platinum-based chemotherapy. In the trials leading to their approval 4– 8, ORRs across all patients ranged from 15% with atezolizumab in IMvigor210 6 to 21.1% with pembrolizumab in KEYNOTE-045 8. Median OS ranged from 6.5 months with avelumab 4 to 18.2 months with durvalumab 5. Importantly, the phase III KEYNOTE-045 trial of pembrolizumab now has over 2 years of patient follow-up demonstrating a continued OS benefit over second-line chemotherapy with median OS of 10.1 months with pembrolizumab and 7.3 months with chemotherapy (hazard ratio [HR] 0.70, 95% confidence interval [CI] 0.57–0.85) 9. The IMvigor211 study was a similarly designed phase III randomized trial comparing atezolizumab versus chemotherapy. However, the primary endpoint, OS, was tested hierarchically in prespecified populations—that is, IC2/3 (PD-L1 expression on at least 5% of tumor-infiltrating immune cells), followed by IC1/2/3, and then the intention-to-treat population. In the IC2/3 population, there was no significant difference in median OS (atezolizumab 11.1 months versus chemotherapy 10.6 months; HR 0.87, 95% CI 0.63–1.21), precluding further formal statistical analyses in the other prespecified populations and thereby resulting in an overall negative study 10.
Investigation into the use of immunotherapy combinations post-platinum is ongoing, but early data are promising ( Table 1). The phase II CheckMate 032 trial compared nivolumab monotherapy with nivolumab 3 mg/kg plus ipilimumab 1 mg/kg (NIVO3+IPI1) and nivolumab 1 mg/kg plus ipilimumab 3 mg/kg (NIVO1+IPI3) 11. In PD-L1 unselected patients, ORR ranged from 25.6% with nivolumab alone to 38% with NIVO1+IPI3. In patients with PD-L1 expression of at least 1%, ORRs were 26.9% with nivolumab alone but 58.1% with NIVO1+IPI3. Furthermore, median OS was 9.9 months with nivolumab alone and 15.3 months with NIVO1+IPI3 across all patients but was 12.9 and 24.1 months, respectively, in patients with PD-L1 expression of at least 1%. Although grade 3 or 4 treatment-related adverse events were more common with NIVO1+IPI3 compared with nivolumab (39.1% versus 26.9%), these results suggest that combination therapy may provide a significant benefit over monotherapy, particularly for patients whose tumors express PD-L1.
Table 1. Selected trials in locally advanced/metastatic urothelial carcinoma with available results.
Trial | Phase | Inclusion criteria | Experimental
arm(s) |
Patients
enrolled |
ORR,
percentage |
DCR,
percentage |
Median
follow-up, months |
mPFS,
months |
mOS,
months |
mDOR,
months |
Status |
---|---|---|---|---|---|---|---|---|---|---|---|
KEYNOTE-045
NCT02256436 |
III | LA/mUC with
progression post- platinum |
Pembrolizumab | 270 | 21.1 | 38.5 | 27.7 | 2.1
HR 0.96 (0.79–1.16) |
10.1
HR 0.70 (0.57–0.85) |
NR | Results
published |
Chemotherapy
(paclitaxel, docetaxel, or vinflunine) |
272 | 11 | 44.9 | 27.7 | 3.3 | 7.3 | 4.4 | ||||
CheckMate
032 NCT01928394 |
I/II | LA/mUC with
progression post- platinum or refused chemo |
Nivolumab 3 mg/kg
(NIVO3) |
78 | 25.6 | 52.5 | 2.8 | 9.9 | 30.5 | Results
published |
|
Nivolumab 3 mg/kg
+ ipilimumab 1 mg/ kg (NIVO3+IPI1) |
104 | 26.9 | 50 | 2.6 | 7.4 | 22.3 | |||||
Nivolumab 1 mg/kg
+ ipilimumab 3 mg/ kg (NIVO1+IPI3) |
92 | 38 | 63 | 4.9 | 15.3 | 22.9 | |||||
IMvigor211
NCT02302807 |
III | LA/mUC with
progression post- platinum (analysis of IC2/3 population) |
Atezolizumab | 116 | 23 | 43 | 2.4
HR 1.01 (0.75–1.34) |
11.1
HR 0.87 (0.63–1.21) |
15.9 | Results
published |
|
Chemotherapy
(paclitaxel, docetaxel, or vinflunine) |
118 | 22 | 54 | 4.2 | 10.6 | 8.3 | |||||
IMvigor130
NCT02807636 |
III | 1L mUC, platinum-
eligible |
Arm A:
atezolizumab + PBC |
451 | 47 | 11.8 | 8.2
HR 0.82 (0.7–0.96) |
16
HR 1.02 (0.83–1.24) |
Preliminary
results presented |
||
Arm B:
atezolizumab monotherapy |
362 | 23 | |||||||||
Arm C: placebo +
PBC |
400 | 44 | 11.8 | 6.3 | 13.4 | ||||||
PIVOT-02
NCT02983045 |
I/II | 1L mUC, cisplatin-
ineligible or refuses |
NKTR-214 +
nivolumab |
34 | 48 | 70 | Preliminary
results presented |
||||
HCRN
GU14-182 NCT02500121 |
II | LA/mUC with at
least SD on 1L PBC |
Maintenance
pembrolizumab |
55 | 23 | 58 | 12.9 | 5.4
HR 0.65 |
22
HR 0.91 (0.52–1.59) |
Results
published |
|
Placebo | 52 | 10 | 39 | 12.9 | 3.0 | 18.7 | |||||
Javelin
Bladder 100 NCT02603432 |
III | LA/mUC with at
least SD on 1L PBC |
Maintenance
avelumab + BSC |
350 | 9.7 | 41.1 | 3.7
HR 0.62 (0.52–0.75) |
21.4
HR 0.69 (0.56–0.86) |
Preliminary
results presented |
||
BSC alone | 350 | 1.4 | 27.4 | 2.0 | 14.3 | ||||||
BLC2001
NCT02365597 |
II | mUC with
progression post- chemotherapy and FGFR2/3 alteration |
Erdafitinib | 101 | 40 | 24 | 5.5 | 11.3 | 6 | Results
published |
|
NCT01004224 | I | LA/mUC with
progression post-platinum or contraindication to PBC and FGFR3 alteration |
Infigratinib | 67 | 25.4 | 64.2 | 3.75 | 7.75 | 5.06 | Results
published |
|
NCT02122172 | II | LA/mUC with
progression post- platinum |
Afatinib | 23 | 8.6 | 39 | 1.4 | 5.3 | Results
published |
||
NCT02236195 | II | LA/mUC with
progression post-platinum and CREBBP or EP300 mutation or deletion |
Mocetinostat | 17 | 11 | 33 | 57 days | 3.5 | Results
published |
||
EV-101
NCT02091999 |
I | Part A: mUC
with progression post-platinum or cisplatin-ineligible Part B: mUC with renal insufficiency Part C: mUC previously treated with anti-PD-(L)1 |
Enfortumab
vedotin Part A: dose escalation Part B/C: dose expansion |
112 | 43 | 16.4 | 5.4 | 12.3 | 7.4 | Part B
completed accrual Part A/C results published |
|
EV-201
NCT03219333 |
II | Cohort 1: LA/mUC
previously treated with anti-PD-(L)1 and PBC Cohort 2: LA/mUC previously treated with anti-PD-(L)1 and cisplatin- ineligible |
Enfortumab
vedotin |
125 | 44 | 72 | 10.2 | 5.8 | 11.7 | 7.6 | Cohort
1 results published Cohort 2 recruiting |
EV-103
NCT03288545 |
Cohort A: 1L mUC,
cisplatin-ineligible |
Cohort A:
Enfortumab vedotin + pembrolizumab |
45 | 73.3 | 93.3 | 11.5 | 12.3 | NR | Cohort
A results presented, additional cohorts recruiting |
||
NCT01631552 | I/II | LA/mUC with
progression after at least 1 prior therapy |
Sacituzumab
govitecan |
45 | 31 | 7.3 | 18.9 | 12.6 | Results
presented |
||
TROPHY-U-01
NCT03547973 |
II | Cohort 1: LA/mUC
with progression after PBC and anti- PD-(L)1 Cohort 2: LA/mUC with progression after anti-PD-(L)1 and platinum- ineligible Cohort 3: LA/mUC with progression after PBC |
Cohort 1+2:
Sacituzumab govitecan Cohort 3: sacituzumab govitecan + pembrolizumab |
35 | 29 | 4.1 | Cohort 1
preliminary results presented Cohort 2+3 recruiting |
1L, first line; BSC, best supportive care; DCR, disease control rate; HR, hazard ratio; IC2/3, PD-L1 expression on at least 5% of tumor-infiltrating immune cells; LA, locally advanced; mDOR, median duration of response; mOS, median overall survival; mPFS, median progression-free survival; mUC, metastatic urothelial carcinoma; NR, not reached; ORR, overall response rate; PBC, platinum-based chemotherapy; SD, stable disease.
There is also a newly established role for anti-PD-(L)1 agents as switch maintenance therapy following completion of first-line platinum-based chemotherapy. The phase III JAVELIN Bladder 100 trial randomly assigned 700 LA/mUC patients whose disease did not progress after first-line platinum-based chemotherapy to receive maintenance avelumab plus best supportive care versus best supportive care alone. At the planned interim analysis, patients receiving maintenance avelumab had a significant improvement in median OS over best supportive care alone (21.4 versus 14.3 months; HR 0.69, 95% CI 0.56–0.86) as well as a progression-free survival (PFS) benefit (3.7 versus 2.0 months; HR 0.62) 12. These results led to the recent FDA approval of avelumab switch maintenance therapy following first-line chemotherapy in patients with mUC. Similarly, the phase II HCRN GU14-182 study enrolled LA/mUC patients who achieved at least stable disease following first-line platinum-based chemotherapy and randomly assigned them to receive maintenance pembrolizumab versus placebo 13. Patients receiving maintenance pembrolizumab demonstrated an improvement in median PFS compared with placebo (5.4 versus 3.0 months; HR 0.65).
First-line immunotherapy for metastatic urothelial carcinoma
In addition to approval for patients who progress following platinum-based chemotherapy, atezolizumab and pembrolizumab are approved in the first-line setting for LA/mUC. Both agents were initially approved as first-line treatment for cisplatin-ineligible patients on the basis of the phase II IMvigor210 14 and KEYNOTE-052 15 trials. Subsequently, the randomized phase III IMvigor130 trial of atezolizumab and KEYNOTE-361 trial of pembrolizumab enrolled platinum-eligible patients with LA/mUC and no prior systemic therapy to receive atezolizumab/pembrolizumab with or without platinum-based chemotherapy versus platinum-based chemotherapy alone 16. In June 2018, interim analyses of these two trials showed that patients with low PD-L1 expression receiving atezolizumab or pembrolizumab monotherapy had decreased survival compared with patients with low PD-L1 expression who received platinum-based chemotherapy, leading to a change in drug approval 16. Currently, atezolizumab and pembrolizumab are indicated as first-line treatment for LA/mUC patients who are cisplatin-ineligible and whose tumors express PD-L1 or patients who are not eligible for any platinum therapy regardless of PD-L1 status. It is important to note that the final analysis of either trial has not been published, but results from IMvigor130 have been presented. Similarly, the phase III DANUBE trial compared durvalumab monotherapy or durvalumab plus tremelimumab versus platinum-based chemotherapy. A press release stated that the study did not meet its primary endpoints for OS in high–PD-L1 patients who received durvalumab or OS in patients who received durvalumab plus tremelimumab regardless of PD-L1 status, but results have not yet been presented or published 17.
There is also growing evidence to suggest that the clinical benefit of the combination of immune checkpoint inhibitors with chemotherapy in the first-line setting may be limited. Results from IMvigor130 evaluating the combination of atezolizumab plus chemotherapy demonstrated a PFS benefit over chemotherapy alone (8.2 versus 6.3 months; HR 0.82, 95% CI 0.70–0.96) but this benefit was small and of questionable clinical significance 18. One concern regarding the outcome of IMvigor130 is that 40% of patients deemed cisplatin-eligible received carboplatin-based chemotherapy, yet subgroup analysis suggested an overall survival benefit seen only in the cisplatin-treated patients. A recent press release also reported that, in KEYNOTE-361, pembrolizumab plus chemotherapy did not meet the dual primary endpoints for superiority in OS or PFS over chemotherapy alone 19. Similar trials are ongoing to further evaluate first-line immunotherapy plus chemotherapy, including CheckMate901 comparing first-line nivolumab plus ipilimumab or standard-of-care chemotherapy versus standard chemotherapy (NCT03036098) and NILE comparing durvalumab plus chemotherapy with or without tremelimumab with chemotherapy alone (NCT03682068) ( Table 2).
Table 2. Selected ongoing clinical trials for patients with locally advanced/metastatic urothelial carcinoma.
Trial | Phase | Inclusion criteria | Experimental arm(s) | Comparator arm | Status |
---|---|---|---|---|---|
CheckMate901NCT03036098 | III | First-line LA/mUC | Arm A: nivolumab +
ipilimumab Arm C: nivolumab + gemcitabine + cisplatin |
Arm B: gemcitabine
+ cis/carboplatin Arm D: gemcitabine + cisplatin |
Recruiting |
NILE
NCT03682068 |
III | First-line LA/mUC | Arm 1: Durvalumab
+ gemcitabine + cis/ carboplatin Arm 2: Durvalumab + tremelimumab + gemcitabine + cis/ carboplatin |
Gemcitabine +
cis/carboplatin |
Recruiting |
KEYNOTE-361 NCT02853305 | III | First-line LA/mUC | Arm 1: Pembrolizumab
Arm 2: Pembrolizumab + chemotherapy |
Gemcitabine +
cis/carboplatin |
Completed accrual (press
release stating co-primary endpoints not met) |
DANUBE
NCT02516241 |
III | First-line LA/mUC | Arm 1: Durvalumab
Arm 2: Durvalumab + tremelimumab |
Gemcitabine +
cis/carboplatin |
Completed accrual (press
release stating primary endpoints not met) |
PIVOT-10
NCT03785925 |
II | First-line LA/mUC, cisplatin-
ineligible |
NKTR-214 + nivolumab | None | Recruiting |
NCT03473743 | Ib/II | Phase Ib: LA/mUC with FGFR
alteration and any number of prior lines of therapy Phase 2: LA/mUC with FGFR alteration, no prior systemic therapy, and cisplatin-ineligible |
Erdafitinib +
cetrelimab |
None | Recruiting |
NCT02122172 | II | LA/mUC treated with prior
platinum-based chemotherapy and alteration in EGFR, HER2, ERBB3, or ERBB4 |
Afatinib | None | Recruiting |
NCT03854474 | I/II | Arm A: LA/mUC with disease
progression following platinum-based chemotherapy Arm B: LA/mUC with positive PD-L1 expression and cisplatin- ineligible |
Tazemetostat +
pembrolizumab |
None | Undergoing interim
analysis |
EV-103
NCT03288545 |
II | Cohort D: 1L LA/mUC, cisplatin-
eligible Cohort E: 1L LA/mUC, platinum-eligible Cohort G: 1L LA/mUC, platinum-eligible Cohort K: 1L LA/mUC, cisplatin- ineligible |
D: EV + cisplatin
E: EV + carboplatin G: EV + pembrolizumab + cis/carboplatin K (randomized): EV + pembrolizumab |
K (randomized): EV
monotherapy |
Recruiting |
NCT03547973 | II | Cohort 1: LA/mUC with
progression following platinum and anti-PD-(L)1 Cohort 2: LA/mUC cisplatin- ineligible and post anti-PD-(L)1 Cohort 3: LA/mUC with progression following platinum |
1 + 2: sacituzumab
govitecan 3: sacituzumab govitecan + pembrolizumab |
None | Cohort 1 awaiting final
results Cohort 2 and 3 recruiting |
1L, first-line; EV, enfortumab vedotin; LA, locally advanced; mUC, metastatic urothelial cancer.
Immunotherapy for muscle-invasive bladder cancer
Preferred management of MIBC includes neoadjuvant cisplatin-based chemotherapy prior to radical cystectomy (RC), and pathologic complete response (pCR) at cystectomy is associated with increased OS 20. Immunotherapy has not yet been approved in the neoadjuvant setting for MIBC, but some preliminary studies show promise ( Table 3). The phase II PURE-01 study enrolled 50 patients with clinical T2-3bN0M0 MIBC and administered three doses of pembrolizumab prior to RC 21. At cystectomy, 42% of patients had a pCR and 54% of patients had pathologic downstaging to less than pT2. Among patients with a PD-L1 combined positive score (CPS) of at least 10%, 54.3% achieved a pCR and 65.7% were downstaged to less than pT2 whereas only 13.3% and 26.7% of patients with PD-LI of less than 10% achieved these same outcomes. A significant association between tumor mutation burden (TMB) and pCR was also seen. The similar phase II ABACUS trial administered two cycles of neoadjuvant atezolizumab and observed a pCR rate of 31% 22. In contrast to PURE-01, the ABACUS trial found no significant correlation between PD-L1 expression or TMB with pCR or 1-year relapse-free survival rates, but patients with high intraepithelial CD8 + cells had a significantly higher pCR rate compared with those without CD8 + cells (40% versus 20%, P <0.05). These conflicting biomarker results suggest that additional research is needed to clarify the best biomarker for predicting response to immunotherapy in bladder cancer.
Table 3. Selected trials in muscle-invasive bladder cancer with available results.
Trial | Phase | Inclusion criteria | Experimental
arm(s) |
Patients
treated |
Patients
undergoing RC |
pCR,
percentage |
<pT2,
percentage |
Median
follow-up, months |
One-year
RFS, percentage |
Status |
---|---|---|---|---|---|---|---|---|---|---|
PURE-01
NCT02736266 |
II | cT2-3bN0M0 MIBC,
plan for RC, any cisplatin eligibility |
Neoadjuvant
pembrolizumab |
50 | 50 | 42 | 54 | Results
published |
||
ABACUS
NCT02662309 |
II | cT2-4aN0M0 MIBC,
plan for RC, cisplatin- ineligible or refuses |
Neoadjuvant
atezolizumab |
95 | 88 | 31 | 13.1 | 79% | Results
published |
|
BLASST-1
NCT03294304 |
II | cT2-4aN0-1M0 MIBC,
plan for RC, cisplatin- eligible |
Neoadjuvant
nivolumab + gem/cis |
41 | 41 | 49 | 66 | Results
presented |
||
HCRN GU14-188
NCT02365766 |
Ib/II | Cohort 1: cT2-4aN0M0
MIBC, plan for RC, cisplatin-eligible Cohort 2: cT2-4aN0M0 MIBC, plan for RC, cisplatin-ineligible |
Cohort 1:
neoadjuvant pembrolizumab + gem/cis Cohort 2: neoadjuvant pembrolizumab + gemcitabine |
40 | 36 | 44.4 | 60 | 14 | 80% | Cohort 1:
results presented Cohort 2: completed accrual |
cTNM, clinical tumor node metastasis stage; gem/cis, gemcitabine + cisplatin; MIBC, muscle-invasive bladder cancer; pCR, pathologic complete response; <pT2, downstaging to non-muscle-invasive disease; RC, radical cystectomy; RFS, relapse-free survival.
Clinical trials have also combined neoadjuvant immune checkpoint inhibitors plus chemotherapy in MIBC. Recent results from the BLASST-1 trial of neoadjuvant nivolumab with gemcitabine and cisplatin demonstrated a pCR rate of 49% and downstaging to less than pT2 in 65.8% of patients 23. Similarly, results of a phase Ib/II trial of neoadjuvant pembrolizumab plus gemcitabine and cisplatin reported a pCR rate of 44%, and 61% of patients were downstaged to less than pT2 24. With a median follow-up of 14 months, the estimated 12-month relapse-free survival was 80% and OS was 94%. Together, these studies suggest that immunotherapy will likely have a role in the management of patients with MIBC. Trials are under way to further evaluate perioperative immunotherapy as monotherapy, immunotherapy in rational combinations, and chemo-immunotherapy approaches.
For patients who are either not eligible for or not interested in RC, TMT is an alternative treatment option. TMT involves a maximal transurethral bladder tumor resection followed by concurrent chemoradiation. Several trials are investigating the addition of immunotherapy to chemoradiation in patients with MIBC in an attempt to harness the abscopal effect. KEYNOTE-992 is a phase III trial comparing pembrolizumab with chemoradiation versus placebo with chemoradiation, followed by pembrolizumab or placebo every 6 weeks for up to a year, SWOG/NRG 1806 is evaluating the use of chemoradiation with or without atezolizumab, and another trial is evaluating pembrolizumab and gemcitabine with concurrent radiation therapy (NCT02621151). Results from these trials are not yet available but are eagerly anticipated ( Table 4).
Table 4. Selected ongoing clinical trials for patients with muscle-invasive bladder cancer.
Trial | Phase | Inclusion criteria | Experimental arm(s) | Comparator arm | Status |
---|---|---|---|---|---|
KEYNOTE-905
NCT03924895 |
III | Cisplatin-ineligible MIBC, fit
for RC |
Perioperative pembrolizumab + RC
+ PLND |
RC + PLND | Recruiting |
AMBASSADOR
NCT03244384 |
III | Muscle invasive or locally
advanced urothelial carcinoma post-surgery, ineligible for or declines cisplatin |
Adjuvant pembrolizumab | Observation | Recruiting |
CheckMate 274
NCT02632409 |
III | Invasive urothelial cancer
post-surgery at high risk of recurrence |
Adjuvant nivolumab | Placebo | Completed
accrual |
IMvigor010
NCT02450331 |
III | Invasive urothelial cancer
post-surgery at high risk of recurrence |
Adjuvant atezolizumab | Observation | Awaiting final
results (press release stating DFS endpoint not met 26) |
NCT02845323 | II | Cisplatin-ineligible/refusing
MIBC, fit for RC |
Neoadjuvant nivolumab +
urelumab |
Neoadjuvant
nivolumab |
Recruiting |
EV-103
NCT03288545 |
I/II | Cisplatin-ineligible MIBC, fit
for RC |
Cohort H: Neoadjuvant EV
Cohort J: Neoadjuvant EV + pembrolizumab |
Recruiting | |
ENERGIZE | III | Cisplatin-eligible MIBC, fit for
RC |
Arm B: Neoadjuvant nivolumab
+ chemo + placebo followed by adjuvant nivolumab + placebo Arm C: Neoadjuvant nivolumab + chemo + BMS-986205 (IDO inhibitor) followed by adjuvant nivolumab + BMS-986205 |
Arm A:
Gemcitabine + cisplatin followed by RC |
Recruiting |
NCT02690558 | II | Cisplatin-eligible MIBC, fit for
RC |
Neoadjuvant pembrolizumab +
gemcitabine + cisplatin |
Completed
accrual |
|
NEMIO
NCT03549715 |
I/II | Cisplatin-eligible MIBC, fit for
RC |
Arm A: Neoadjuvant durvalumab
+ ddMVAC Arm B: Neoadjuvant durvalumab + tremelimumab + ddMVAC |
Recruiting | |
NIAGARA
NCT03732677 |
III | Cisplatin-eligible MIBC, fit for
RC |
Neoadjuvant durvalumab +
gemcitabine + cisplatin followed by adjuvant durvalumab |
Neoadjuvant
gemcitabine + cisplatin |
Recruiting |
NCT04228042 | I/II | Low-grade UTUC or high-
grade UTUC and cisplatin- ineligible |
Neoadjuvant infigratinib | Recruiting | |
NCT02621151 | II | Localized MIBC, not a
candidate for or declines RC |
Tri-modality therapy with maximal
TURBT and gemcitabine + pembrolizumab concurrent with EBRT |
Recruiting | |
SWOG 1806
NCT03775265 |
III | Localized MIBC | Concurrent chemotherapy +
atezolizumab + radiation |
Concurrent
chemotherapy + radiation |
Recruiting |
KEYNOTE-992
NCT04241185 |
III | Localized MIBC, opting for
bladder preservation |
Pembrolizumab + CRT | Placebo + CRT | Recruiting |
PROOF 302
NCT04197986 |
III | Invasive urothelial carcinoma
with FGFR3 alteration at high risk for recurrence following RC or nephrectomy |
Adjuvant infigratinib | Placebo | Recruiting |
CRT, chemoradiation; ddMVAC, dose-dense methotrexate, vinblastine, doxorubicin, cisplatin; DFS, disease-free survival; EBRT, external beam radiation therapy; EV, enfortumab vedotin; MIBC, muscle-invasive bladder cancer; PLND, pelvic lymph node dissection; RC, radical cystectomy; TURBT, transurethral resection of bladder tumor; UTUC, upper tract urothelial carcinoma.
Immunotherapy for non-muscle-invasive bladder cancer
The first FDA approval for immunotherapy in the non-metastatic setting came in January 2020 when pembrolizumab was approved for patients with BCG-unresponsive, high-risk NMIBC with carcinoma in situ (CIS) with or without papillary tumors who are ineligible for or have chosen not to undergo cystectomy. This approval was based on results from cohort A of the KEYNOTE-057 trial showing a 3-month complete response (CR) rate of 40% with 46% of responses lasting at least 12 months 25. Similar results were seen in the phase II SWOG S1605 trial of atezolizumab in BCG-unresponsive NMIBC. Preliminary results from 73 patients showed a 3-month CR rate of 41% and 6-month CR rate of 26% 27. Additional trials evaluating the use of immunotherapy in NMIBC are ongoing; these include the phase III KEYNOTE-676 trial of BCG with or without pembrolizumab (NCT03711032), a phase II trial of gemcitabine plus pembrolizumab (NCT04164082), and the phase II ADAPT-Bladder trial comparing durvalumab monotherapy, durvalumab plus BCG, and durvalumab plus external beam radiation (NCT03317158).
Targeted therapy
One of the most important recent advances in urothelial cancer is the genomic profiling of tumors, which has revealed a number of common genomic alterations 28– 30. In an analysis of 412 MIBCs as part of The Cancer Genome Atlas Project, 58 significantly mutated genes were identified 28. Clinically relevant alterations in MIBC include changes in the genes for cyclin-dependent kinase inhibitor 2a ( CDKN2A), fibroblast growth factor receptor 3 ( FGFR3), erythroblastic oncogene B/human epidermal growth factor receptor 2 ( ERBB2/HER2), and phosphatidylinositol 3-kinase catalytic subunit alpha ( PIK3CA). Additionally, mutations in chromatin-modifying genes are found in up to 83% of patients with urothelial cancer, which has spurred investigation into agents targeting these alterations 31, 32.
FGFR
Erdafitinib is a tyrosine kinase inhibitor of FGFR1–4 and the first targeted therapy approved for mUC. The phase II trial of erdafitinib included 99 patients whose tumor harbored an FGFR3 mutation or FGFR2/3 fusion and who had disease progression following chemotherapy 33. The confirmed ORR was 40% and an additional 39% of patients had stable disease. A total of 22 patients had previously received immunotherapy with only one achieving a response, yet the response rate for erdafitinib for this subgroup was 59%. At a median follow-up of 24 months, the median PFS was 5.5 months (95% CI 4.0–6.0) and the median OS was 11.3 months (95% CI 9.7–15.2) 34. Based on these results, erdafitinib was approved by the FDA in April 2019 for patients with mUC with a susceptible FGFR2/3 alteration following platinum-containing chemotherapy. Multiple ongoing trials—including a phase II study of erdafitinib alone or in combination with cetrelimab, an anti-PD-1 antibody, and as first-line therapy for cisplatin-ineligible patients with mUC and an FGFR alteration (NCT03473743)—are assessing erdafitinib in other clinical scenarios.
In addition to erdafitinib, other FGFR inhibitors are under investigation. An expansion cohort to the phase I trial of infigratinib included 67 mUC patients who progressed on or had contraindications to platinum-based chemotherapy and whose tumor harbored an alteration in FGFR3 35. The confirmed ORR was 25.4%, median duration of response was 5.06 months, median PFS was 3.75 months, and median OS was 7.75 months. A subsequent analysis of this same cohort observed that patients with upper tract urothelial cancer (UTUC) had a confirmed ORR of 50% and a disease control rate (DCR) of 100% but that those with urothelial cancer of the bladder (UCB) had an ORR of 22% and a DCR of 59.3% 36. Additionally, the median PFS and median OS were 8.54 and 21.82 months for those with UTUC and 3.65 and 7.0 months for those with UCB. Prior work has shown that FGFR3 alterations are more common in UTUC than UCB (40% versus 26%) 37 and thus UTUC may be more amenable to FGFR inhibition. Although this study included a small number of patients with UTUC, these initial results certainly warrant further evaluation and a phase I/II trial of neoadjuvant infigratinib for patients with UTUC is planned (NCT04228042). Additionally, a phase III trial comparing adjuvant infigratinib versus placebo in patients with FGFR3 alterations and high risk for disease recurrence is under way (NCT04197986).
Chromatin-modifying genes
Chromatin structure can be modified via many mechanisms, including histone acetylation/deacetylation and histone methylation/demethylation, resulting in regulation of gene transcription. Disruption of this process is implicated in the pathogenesis of urothelial cancer and therefore may be a viable target for new therapies, such as histone deacetylase (HDAC) inhibitors and enhancer of zeste homolog 2 (EZH2) inhibitors 38. Mocetinostat, a class I/IV HDAC inhibitor, was administered to 17 patients with mUC with progression after platinum-based chemotherapy and an inactivating mutation or deletion in CREBBP, EP300 39, or both. The ORR was 11% in stage 1 and so the study was terminated. Although mocetinostat did not appear to be effective in this cohort of patients with mUC, it is possible that a different biomarker is needed to predict patient response.
Pre-clinical studies have demonstrated that EZH2 inhibition induces cell death in models of urothelial cancer 40, 41. Additionally, the response appears to be enhanced when the EZH2 inhibitor tazemetostat is combined with an anti-PD-1 antibody 42. Based on these findings, a phase I/II trial evaluating the combination of tazemetostat plus pembrolizumab in patients with either cisplatin-refractory or cisplatin-ineligible mUC is under way (NCT03854474). Similarly, EZH2 inhibition has been shown to improve the response to anti-CTLA-4 therapy in a murine model of bladder cancer 43. This led to the phase Ib/II ORIOn-E trial of the EZH2 inhibitor CPI-1205 combined with ipilimumab, which includes a cohort of patients with mUC (NCT03525795). This trial is currently closed to accrual, but results have not yet been released. Other agents targeting chromatin modification genes appear promising in pre-clinical studies and are anticipated to move into early-phase clinical trials in the near future.
HER2
Multiple agents targeting HER2, including trastuzumab, lapatinib, and afatinib, have also been tested in patients with urothelial cancer 44. Results thus far have been somewhat mixed, possibly partially owing to inclusion of HER2 unselected patients and the discordance in HER2 classification between immunohistochemistry (IHC), fluorescence in situ hybridization, and molecular characterization. A phase II study of afatinib in HER2 unselected patients with platinum-refractory mUC found an ORR of 8.6% and a median PFS of 1.4 months for the entire cohort 45. However, they also found that 83% (5/6) of patients with HER2 copy number amplification or ERBB3 somatic mutations (or both) achieved a PFS of at least 3 months but that 0% of patients without alterations did. Interestingly, no correlation between IHC for ERBB3, HER2, or EGFR and clinical response to afatinib was seen. Additional trials of HER2 targeted agents, including a follow-up study of afatinib in patients with alterations in EGFR, HER2, ERBB3, or ERBB4 (NCT02122172), are ongoing. It remains to be seen whether this will prove to be a viable treatment option in appropriately selected patients.
Antibody–drug conjugates
Antibody–drug conjugates (ADCs) are a class of cancer therapeutic that link a monoclonal antibody specific for a tumor cell-surface protein with a cytotoxic agent. A number of ADCs have received FDA approval across a wide variety of tumor types, including ado-trastuzumab emtansine for Her2 + breast cancer, brentuximab vedotin for CD30 + Hodgkin’s lymphoma, and most recently enfortumab vedotin (EV) for urothelial cancer 46.
Enfortumab vedotin
EV is an ADC targeting Nectin-4, a cell adhesion molecule highly expressed in nearly all urothelial tumors, conjugated to monomethyl auristatin E, a microtubule-disrupting agent 47. In the dose expansion portion of the EV-101 trial, 112 mUC patients who failed at least one prior therapy received EV 48. The confirmed ORR was 43%, including 5% CR, and the median duration of response was 7.4 months. Subsequently, EV-201 enrolled patients with mUC treated with prior platinum and anti-PD-(L)1 (cohort 1) or treated with prior anti-PD-(L)1 and cisplatin-ineligible (cohort 2) 49. Cohort 1 enrolled 125 patients with a confirmed ORR of 44%, including a 12% CR rate. Responses were seen across subgroups including an ORR of 41% in non-responders to prior anti-PD-(L)1 and 38% in patients with liver metastases. Results for cohort 2 have not yet been released.
EV-103 is an ongoing multi-cohort trial of EV alone or in combination with other therapies and includes cohorts of patients with mUC and localized MIBC. Cohort A evaluated EV plus pembrolizumab as first-line treatment for 45 cisplatin-ineligible patients with LA/mUC 50. The confirmed ORR was 73.3%, including 15.6% CRs, and the DCR was 93.3%. With a median follow-up of 10.4 months, the median duration of response was not yet reached and the median PFS was 12.3 months. These results are extremely encouraging, particularly for cisplatin-ineligible patients who have limited effective treatment options. Additional study cohorts, including EV plus chemotherapy as first-line treatment for mUC and EV alone or with pembrolizumab as neoadjuvant therapy for localized MIBC (NCT03288545), are ongoing.
Sacituzumab govitecan
Sacituzumab govitecan (SG) is an ADC employing an anti-Trop-2 antibody conjugated to SN-38, the active metabolite of irinotecan 51. Trop-2 is a transmembrane glycoprotein that is important for cell growth and tumorigenesis and that is overexpressed in urothelial cancer. The phase I/II basket trial of SG included 45 mUC patients who had received at least one prior line of systemic therapy 52. The ORR was 31%, median duration of response was 12.6 months, median PFS was 7.3 months, and median OS was 18.9 months. The subsequent phase II TROPHY-U-01 trial of SG recently completed accrual for cohort 1, which enrolled 100 mUC patients who progressed after platinum-based therapy and a checkpoint inhibitor 53. Preliminary results from 35 patients demonstrated an ORR of 29% and in light of these data the FDA granted SG fast-track designation for mUC in April 2020 54. Accrual to two additional patient cohorts—a cohort of platinum-ineligible patients who progressed following checkpoint inhibitor therapy and a cohort of immune checkpoint inhibitor-naïve patients who will receive SG plus pembrolizumab—is ongoing.
Summary
The management of bladder cancer has changed dramatically in the past 5 years and is poised to evolve further in the coming years. The approval of seven new drugs during this time has created new options for many patients and in some cases has led to long-term responses. Despite these encouraging successes, however, much work remains to be done.
Considerable excitement has surrounded immune checkpoint inhibitors for bladder cancer, but ORR is still only about 15 to 25% with monotherapy for metastatic disease. Multi-agent therapy, employing different combinations of immunotherapy, chemotherapy, or targeted therapy, may prove to be more efficacious, and further clinical trials testing this strategy are under way. The role of immune checkpoint inhibitors at different disease stages is also undergoing refinement, starting with FDA approval of pembrolizumab in high-risk NMIBC. It is likely only a matter of time until immune checkpoint inhibitors are approved as part of perioperative treatment for patients with MIBC, and evolving data suggest a role for maintenance immunotherapy following induction chemotherapy in metastatic disease.
The discovery of better biomarkers to help select patients who are more likely to respond to certain therapies will also prove important in the years to come. As was seen in the PURE-01 and ABACUS trials, markers such as PD-L1 expression and TMB appear predictive in some cases but not in others, and the possible predictive role of CD8 + cell expression or gene signature expression requires further elucidation. Many mutations are commonly found in bladder cancer, suggesting that targeted therapy has great potential to influence the treatment landscape. We must continue to work to understand which alterations confer susceptibility to targeted inhibition and what is the best method to detect these alterations. As our knowledge of the biological drivers of carcinogenesis and factors influencing treatment response improves, so too will the outcomes of our patients.
Editorial Note on the Review Process
F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).
The referees who approved this article are:
Fumitaka Koga, Department of Urology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
Mototsugu Oya, Department of Urology, Keio University School of Medicine, Tokyo, Japan
Funding Statement
The research was partially supported by a National Service Research Award Post-doctoral Traineeship from the Agency for Healthcare Research and Quality, sponsored by the Cecil G. Sheps Center for Health Services Research at The University of North Carolina (UNC)-Chapel Hill (grant 5T32 HS000032).
The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
[version 1; peer review: 2 approved]
References
- 1. National Cancer Institute: SEER Cancer Stat Facts: Bladder Cancer. Accessed August 31, 2019. Reference Source [Google Scholar]
- 2. van Rhijn BWG, Burger M, Lotan Y, et al. : Recurrence and progression of disease in non-muscle-invasive bladder cancer: from epidemiology to treatment strategy. Eur Urol. 2009;56(3):430–42. 10.1016/j.eururo.2009.06.028 [DOI] [PubMed] [Google Scholar]
- 3. 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(17):3068–77. 10.1200/JCO.2000.18.17.3068 [DOI] [PubMed] [Google Scholar]
- 4. Patel MR, Ellerton J, Infante JR, et al. : Avelumab in metastatic urothelial carcinoma after platinum failure (JAVELIN Solid Tumor): pooled results from two expansion cohorts of an open-label, phase 1 trial. Lancet Oncol. 2018;19(1):51–64. 10.1016/S1470-2045(17)30900-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Powles T, O'Donnell PH, Massard C, et al. : Efficacy and Safety of Durvalumab in Locally Advanced or Metastatic Urothelial Carcinoma: Updated Results From a Phase 1/2 Open-label Study. JAMA Oncol. 2017;3(9):e172411. 10.1001/jamaoncol.2017.2411 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. 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(10031):1909–20. 10.1016/S0140-6736(16)00561-4 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 7. 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(3):312–322. 10.1016/S1470-2045(17)30065-7 [DOI] [PubMed] [Google Scholar]
- 8. Bellmunt J, de Wit R, Vaughn DJ, et al. : Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med. 2017;376(11):1015–26. 10.1056/NEJMoa1613683 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Fradet Y, Bellmunt J, Vaughn DJ, et al. : Randomized phase III KEYNOTE-045 trial of pembrolizumab versus paclitaxel, docetaxel, or vinflunine in recurrent advanced urothelial cancer: results of >2 years of follow-up. Ann Oncol. 2019;30(6):970–6. 10.1093/annonc/mdz127 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 10. Powles T, Durán I, van der Heijden MS, et al. : Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2018;391(10122):748–57. 10.1016/S0140-6736(17)33297-X [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 11. Sharma P, Siefker-Radtke A, de Braud F, et al. : Nivolumab Alone and With Ipilimumab in Previously Treated Metastatic Urothelial Carcinoma: CheckMate 032 Nivolumab 1 mg/kg Plus Ipilimumab 3 mg/kg Expansion Cohort Results. J Clin Oncol. 2019;37(19):1608–16. 10.1200/JCO.19.00538 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 12. Powles T, Park SH, Voog E, et al. : Maintenance avelumab + best supportive care (BSC) versus BSC alone after platinum-based first-line (1L) chemotherapy in advanced urothelial carcinoma (UC): JAVELIN Bladder 100 phase III interim analysis. J Clin Oncol. 2020;38(18_suppl ). 10.1200/JCO.2020.38.18_suppl.LBA1 [DOI] [Google Scholar]
- 13. Galsky MD, Mortazavi A, Milowsky MI, et al. : Randomized Double-Blind Phase II Study of Maintenance Pembrolizumab Versus Placebo After First-Line Chemotherapy in Patients With Metastatic Urothelial Cancer. J Clin Oncol. 2020;38(16):1797–806. 10.1200/JCO.19.03091 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. 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(10064):67–76. 10.1016/S0140-6736(16)32455-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Balar AV, Castellano D, O'Donnell PH, et al. : First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017;18(11):1483–92. 10.1016/S1470-2045(17)30616-2 [DOI] [PubMed] [Google Scholar]
- 16. Suzman DL, Agrawal S, Ning YM, et al. : FDA Approval Summary: Atezolizumab or Pembrolizumab for the Treatment of Patients with Advanced Urothelial Carcinoma Ineligible for Cisplatin-Containing Chemotherapy. Oncologist. 2019;24(4):563–9. 10.1634/theoncologist.2018-0084 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 17. Update on Phase III DANUBE trial for Imfinzi and tremelimumab in unresectable, Stage IV bladder cancer. Accessed July 1, 2020. Reference Source [Google Scholar]
- 18. Grande E, Galsky M, Arranz Arija JA, et al. : IMvigor130: Efficacy and safety from a phase III study of atezolizumab (atezo) as monotherapy or combined with platinum-based chemotherapy (PBC) vs placebo + PBC in previously untreated locally advanced or metastatic urothelial carcinoma (mUC). Ann Oncol. 2019;30(5):v888–v889. 10.1093/annonc/mdz394.047 [DOI] [Google Scholar]
- 19. Merck Provides Update on Phase 3 KEYNOTE-361 Trial Evaluating KEYTRUDA® (pembrolizumab) as Monotherapy and in Combination with Chemotherapy in Patients with Advanced or Metastatic Urothelial Carcinoma. Accessed July 1, 2020. Reference Source [Google Scholar]
- 20. Petrelli F, Coinu A, Cabiddu M, et al. : Correlation of pathologic complete response with survival after neoadjuvant chemotherapy in bladder cancer treated with cystectomy: a meta-analysis. Eur Urol. 2014;65(2):350–7. 10.1016/j.eururo.2013.06.049 [DOI] [PubMed] [Google Scholar]
- 21. Necchi A, Anichini A, Raggi D, 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;36(34):3353–60. 10.1200/JCO.18.01148 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 22. Powles T, Kockx M, Rodriguez-Vida A, et al. : Clinical efficacy and biomarker analysis of neoadjuvant atezolizumab in operable urothelial carcinoma in the ABACUS trial. Nat Med. 2019;25(11):1706–14. 10.1038/s41591-019-0628-7 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 23. Gupta S, Sonpavde G, Weight CJ, 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(6):439 10.1200/JCO.2020.38.6_suppl.439 [DOI] [Google Scholar]
- 24. Kaimakliotis H, Albany C, Hoffman-Censits J, et al. : PD52-03 A multicenter phase 1B/2 study of neoadjuvant pembrolizumab and cisplatin chemotherapy for muscle invasive urothelial cancer. J Clin Oncol. 2019;201(Supplement 4). 10.1097/01.JU.0000556959.45525.89 [DOI] [Google Scholar]
- 25. FDA approves pembrolizumab for BCG-unresponsive, high-risk non-muscle invasive bladder cancer | FDA. Accessed April 11, 2020. Reference Source [Google Scholar]
- 26. Roche - Roche provides an update on Phase III study of Tecentriq in people with muscle-invasive urothelial cancer. Accessed April 15, 2020. Reference Source [Google Scholar]
- 27. Black PC, Tangen C, Singh P, et al. : Phase II trial of atezolizumab in BCG-unresponsive non-muscle invasive bladder cancer: SWOG S1605 (NCT #02844816). J Clin Oncol. 2020;38(15):5022 10.1200/JCO.2020.38.15_suppl.5022 [DOI] [Google Scholar]
- 28. Robertson AG, Kim J, Al-Ahmadie H, et al. : Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer. Cell. 2017;171(3):540–556.e25. 10.1016/j.cell.2017.09.007 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 29. Ross JS, Wang K, Khaira D, et al. : Comprehensive genomic profiling of 295 cases of clinically advanced urothelial carcinoma of the urinary bladder reveals a high frequency of clinically relevant genomic alterations. Cancer. 2016;122(5):702–11. 10.1002/cncr.29826 [DOI] [PubMed] [Google Scholar]
- 30. Knowles MA, Hurst CD: Molecular biology of bladder cancer: new insights into pathogenesis and clinical diversity. Nat Rev Cancer. 2015;15(1):25–41. 10.1038/nrc3817 [DOI] [PubMed] [Google Scholar]
- 31. Gui Y, Guo G, Huang Y, et al. : Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat Genet. 2011;43(9):875–8. 10.1038/ng.907 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Kim PH, Cha EK, Sfakianos JP, et al. : Genomic predictors of survival in patients with high-grade urothelial carcinoma of the bladder. Eur Urol. 2015;67(2):198–201. 10.1016/j.eururo.2014.06.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Loriot Y, Necchi A, Park SH, et al. : Erdafitinib in Locally Advanced or Metastatic Urothelial Carcinoma. N Engl J Med. 2019;381(4):338–48. 10.1056/NEJMoa1817323 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 34. Siefker-Radtke AO, Necchi A, Park SH, et al. : ERDAFITINIB in locally advanced or metastatic urothelial carcinoma (mUC): Long-term outcomes in BLC2001. J Clin Oncol. 2020;38(15_suppl):5015 10.1200/jco.2020.38.15_suppl.5015 [DOI] [Google Scholar]
- 35. Pal SK, Rosenberg JE, Hoffman-Censits JH, et al. : Efficacy of BGJ398, a Fibroblast Growth Factor Receptor 1-3 Inhibitor, in Patients with Previously Treated Advanced Urothelial Carcinoma with FGFR3 Alterations. Cancer Discov. 2018;8(7):812–21. 10.1158/2159-8290.CD-18-0229 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 36. Pal SK, Bajorin D, Dizman N, et al. : Infigratinib in upper tract urothelial carcinoma versus urothelial carcinoma of the bladder and its association with comprehensive genomic profiling and/or cell-free DNA results. Cancer. 2020;126(11):2597–606. 10.1002/cncr.32806 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 37. Audenet F, Isharwal S, Cha EK, et al. : Clonal Relatedness and Mutational Differences between Upper Tract and Bladder Urothelial Carcinoma. Clin Cancer Res. 2019;25(3):967–76. 10.1158/1078-0432.CCR-18-2039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Duex JE, Swain KE, Dancik GM, et al. : Functional Impact of Chromatin Remodeling Gene Mutations and Predictive Signature for Therapeutic Response in Bladder Cancer. Mol Cancer Res. 2018;16(1):69–77. 10.1158/1541-7786.MCR-17-0260 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 39. Grivas P, Mortazavi A, Picus J, et al. : Mocetinostat for patients with previously treated, locally advanced/metastatic urothelial carcinoma and inactivating alterations of acetyltransferase genes. Cancer. 2019;125(4):533–40. 10.1002/cncr.31817 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Ramakrishnan S, Granger V, Rak M, et al. : Inhibition of EZH2 induces NK cell-mediated differentiation and death in muscle-invasive bladder cancer. Cell Death Differ. 2019;26(10):2100–14. 10.1038/s41418-019-0278-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Ler LD, Ghosh S, Chai X, et al. : Loss of tumor suppressor KDM6A amplifies PRC2-regulated transcriptional repression in bladder cancer and can be targeted through inhibition of EZH2. Sci Transl Med. 2017;9(378):eaai8312. 10.1126/scitranslmed.aai8312 [DOI] [PubMed] [Google Scholar]
- 42. Meeks JJ, Shilatifard A, Miller SD, et al. : A pilot study of tazemetostat and MK-3475 (pembrolizumab) in advanced urothelial carcinoma (ETCTN 10183). J Clin Oncol. 2020;38(6_suppl):TPS607 10.1200/JCO.2020.38.6_suppl.TPS607 [DOI] [Google Scholar]
- 43. Goswami S, Apostolou I, Zhang J, et al. : Modulation of EZH2 expression in T cells improves efficacy of anti-CTLA-4 therapy. J Clin Invest. 2018;128(9):3813–8. 10.1172/JCI99760 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 44. Koshkin VS, O’Donnell P, Yu EY, et al. : Systematic Review: Targeting HER2 in Bladder Cancer. Bl Cancer. 2019;5(1):1–12. 10.3233/BLC-180196 [DOI] [Google Scholar]; Faculty Opinions Recommendation
- 45. Choudhury NJ, Campanile A, Antic T, et al. : Afatinib Activity in Platinum-Refractory Metastatic Urothelial Carcinoma in Patients With ERBB Alterations. J Clin Oncol. 2016;34(18):2165–71. 10.1200/JCO.2015.66.3047 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 46. Thomas A, Teicher BA, Hassan R: Antibody-drug conjugates for cancer therapy. Lancet Oncol. 2016;17(6):e254–e262. 10.1016/S1470-2045(16)30030-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Challita-Eid PM, Satpayev D, Yang P, et al. : Enfortumab Vedotin Antibody-Drug Conjugate Targeting Nectin-4 Is a Highly Potent Therapeutic Agent in Multiple Preclinical Cancer Models. Cancer Res. 2016;76(10):3003–13. 10.1158/0008-5472.CAN-15-1313 [DOI] [PubMed] [Google Scholar]
- 48. Rosenberg J, Sridhar SS, Zhang J, et al. : EV-101: A Phase I Study of Single-Agent Enfortumab Vedotin in Patients With Nectin-4-Positive Solid Tumors, Including Metastatic Urothelial Carcinoma. J Clin Oncol. 2020;38(10):1041–9. 10.1200/JCO.19.02044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Rosenberg JE, O'Donnell PH, Balar AV, et al. : Pivotal Trial of Enfortumab Vedotin in Urothelial Carcinoma After Platinum and Anti-Programmed Death 1/Programmed Death Ligand 1 Therapy. J Clin Oncol. 2019;37(29):2592–600. 10.1200/JCO.19.01140 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 50. Rosenberg JE, Flaig TW, Friedlander TW, et al. : Study EV-103: Preliminary durability results of enfortumab vedotin plus pembrolizumab for locally advanced or metastatic urothelial carcinoma. J Clin Oncol. 2020;38(6_suppl):441 10.1200/JCO.2020.38.6_suppl.441 [DOI] [Google Scholar]
- 51. Cardillo TM, Govindan SV, Sharkey RM, et al. : Sacituzumab Govitecan (IMMU-132), an Anti-Trop-2/SN-38 Antibody-Drug Conjugate: Characterization and Efficacy in Pancreatic, Gastric, and Other Cancers. Bioconjug Chem. 2015;26(5):919–31. 10.1021/acs.bioconjchem.5b00223 [DOI] [PubMed] [Google Scholar]
- 52. Tagawa ST, Faltas BM, Lam ET, et al. : Sacituzumab govitecan (IMMU-132) in patients with previously treated metastatic urothelial cancer (mUC): Results from a phase I/II study. J Clin Oncol. 2019;37(7_suppl):354 10.1200/JCO.2019.37.7_suppl.354 [DOI] [Google Scholar]
- 53. Tagawa ST, Balar A, Petrylak DP, et al. : Initial results from TROPHY-U-01: A phase II open-label study of sacituzumab govitecan in patients (Pts) with metastatic urothelial cancer (mUC) after failure of platinum-based regimens (PLT) or immunotherapy. Ann Oncol. 2019;30(Supplement 5):v890–v891. 10.1093/annonc/mdz394.049 [DOI] [Google Scholar]
- 54. FDA Grants Fast Track Designation to Sacituzumab Govitecan for Metastatic Urothelial Cancer. Accessed April 8, 2020. Reference Source [Google Scholar]