The Advanced Bladder Clinic: Are You Ready?

Published in Everyday Urology - Oncology Insights: Volume 5, Issue 1
Published Date: March 2020

In 2015, cisplatin-ineligible patients with metastatic urothelial carcinoma could not be offered a first-line treatment option capable of prolonging their survival. Also, a paucity of second-line treatments resulted in these same patients usually surviving for only 3 to 6 months after progressing on platinum-treatment. Recent successful registrational trials and U.S. Food and Drug Administration (FDA) approvals have revolutionized this treatment landscape, and fortunately, there are more advances to come. Hence, urologists now have both the opportunity and the impetus to develop advanced bladder cancer clinics to serve bladder cancer patients across the disease spectrum, inclusive of high-risk nonmuscle invasive bladder cancer (NMIBC) and metastatic urothelial bladder cancer (mUC). In this article, I discuss novel therapies, recently approved, cutting edge treatments currently undergoing trial accrual, and practical considerations for implementing an advanced bladder clinic (ABC).

Immune Checkpoint Inhibitors

The discovery that tumor cells inhibit the body’s antitumor immune response by binding programmed cell death 1 (PD-1) receptors on the T-cell surface has facilitated the development of several monoclonal antibodies targeting either the PD receptor (PD-1, PD-2) or its tumor cell ligand, PD-L1. In Phase II/Phase III trials, immune checkpoint inhibitors (CPIs) have demonstrated significantly prolonged survival compared with the standard of care for patients with advanced and metastatic urothelial carcinoma.1-6

To date, five checkpoint inhibitors have received FDA approval for the treatment of advanced and metastatic bladder cancer in the post-platinum setting. Nivolumab (Opdivo®) and pembrolizumab (Keytruda®) target PD-1, while atezolizumab (Tecentriq®), avelumab (Bavencio®), and durvalumab (Imfinzi®) target PD-L1. In 2017, the FDA also approved atezolizumab and pembrolizumab for the first-line treatment of advanced bladder cancer in cisplatin-ineligible patients.7-9

Patient selection

Because urothelial carcinomas express varying levels of PD-L1, not all patients with advanced bladder cancer are appropriate candidates for PD-1/PD-L1 pathway inhibitor therapy. In the KEYNOTE-361 and IMVIGOR-130 trials, for example, pembrolizumab and atezolizumab were associated with lower survival among patients whose urothelial tumors expressed low levels of PD-L1.10

In response to these findings, the FDA now requires the use of companion diagnostic tests to confirm sufficient PD-L1 levels with immunohistochemistry (IHC) testing before patients can receive first-line therapy with pembrolizumab or atezolizumab.10 The Dako PD-L1 IHC 22C3 PharmDx (or 22C3) assay is approved as a companion diagnostic for use with pembrolizumab, while the Ventana PD-L1 (SP142) Assay® is approved for use with atezolizumab. Both IHC assays are relatively inexpensive and can be readily performed in commercial pathology laboratories and academic centers, and urologists can also outsource the IHC assay testing to commercial laboratories.

Earlier-stage use of immune checkpoint inhibitors

First-line PD-1 pathway inhibitor monotherapy induces responses in approximately 25% to 30% of patients with mUC. A subset of these patients show very impressive and durable complete responses with subsequent prolonged survival times,11 which is especially striking when we consider the typical elderly age and comorbidity burden of this patient population. Consequently, there has been keen interest in moving checkpoint inhibitors more proximally within the bladder cancer disease journey, analogous to the experience of approved therapeutics for castration-resistant metastatic prostate cancer, which has led to the rapid evolution of urologic advanced prostate cancer clinic(APC).

Non-muscle invasive bladder cancer

Over the last 40 years, the FDA has approved very few treatments for non-muscle invasive bladder cancer. Thiotepa was approved in 1959, Bacillus Calmette-Guérin (BCG) Tice and BCG Connaught were approved in 1989 and 1990, and valrubicin was approved in 1998.12 Thiotepa is not widely used in contemporary intravesical treatment regimens, and valrubicin has been considered sparingly for radical cystectomy ineligible patients because, mainly due to its efficacy benefit in fewer than 10% of patients treated.13 Hence, intravesical BCG remains the only FDA recommended first-line therapy for NMIBC, although intravesical mitomycin C (MMC) is an intravesical chemotherapeutic used often, albeit off-label bit incorporated within NCCN, AUA, and EAU guidelines, and alongside other generic intravesical chemotherapies, gemcitabine and docetaxel, are receiving enhanced utilization due to the global BCG shortage.

Given this limited therapeutic landscape, the single-arm, open-label, Phase II KEYNOTE-057 trial has received global attention for its attempt to achieve clinically meaningful benefit for BCG unresponsive patients with high-risk NMIBC. This study evaluated pembrolizumab (200 mg every 3 weeks) for 24 months or until disease recurrence in 103 cystectomy-ineligible or cystectomy-refusing patients with high-risk, BCG-unresponsive non-muscle invasive bladder cancer with carcinoma in situ (CIS) with or without papillary tumors.14 At the 2019 meeting of the European Society for Molecular Oncology (ESMO), investigators provided an update based on a median follow-up period of 21.1 months.15 The complete response rate was 41%, the median duration of response was 13.5 months, and 57% of complete responses lasted at least 12 months. Most patients with complete responses also reported stable health-related quality of life over time. Based on these findings, in January 2020 the FDA approved pembrolizumab for use in this patient population.16

Several ongoing Phase III trials are further assessing PD-1/PD-L1 pathway inhibitors for the treatment of non-muscle invasive bladder cancer. The global KEYNOTE-676 study (NCT03711032) evaluates pembrolizumab following cystoscopy or transurethral resection in high-risk BCG-unresponsive patients, which have received BCG but with a lesser requirement compared to KEYNOTE-057.17 The primary endpoint is complete response in patients with CIS, and the targeted enrollment is 550 patients. In addition, the POTOMAC trial (NCT03528694) assesses durvalumab in combination with BCG in completely resected, high-risk non-muscle invasive bladder cancer patients that are BCG naïve. Patients are randomly assigned on a 1:1:1 basis to receive induction-only durvalumab plus BCG, induction and maintenance durvalumab plus BCG, or BCG only.18 The primary endpoint is disease-free survival, and the trial aims to enroll 975 patients. Finally, the ALBAN study (NCT03799835) compares one year of BCG plus atezolizumab therapy versus BCG alone in high-risk, completely resected, BCG-naïve non-muscle invasive bladder cancer.19 The primary endpoint is recurrence-free survival, and the estimated enrollment is 614 patients. Additionally, BCG naïve trials are planned in 2020 with other CPIs, also exploring subcutaneous deliveries.

Muscle-invasive bladder cancer

Cisplatin-based chemotherapy significantly prolongs survival when used in the neoadjuvant setting for patients with muscle-invasive bladder cancer prior to radical cystectomy.20 However, approximately 50% of patients are cisplatin-ineligible, and unfortunately, many patients who are eligible for neoadjuvant platinum-based therapy do not receive the treatment.21 There is considerable interest in identifying alternatives which could demonstrate clinical benefit with enhanced tolerability.

The single-arm, open-label, Phase II PURE-01 study evaluated three courses of pembrolizumab preceding radical cystectomy in 114 patients with T2-T4a muscle-invasive bladder cancer (5% N1, 0% T4b).22 A total of 37% of patients had a pathologic complete response (pT0) and 55% were downstaged to pT≤1. Interestingly, 19% of patients presented with predominantly variant tumor histology, for which chemotherapy is of limited efficacy. Among these patients, 86% of those with squamous cell carcinoma were downstaged to pT<1. High PD-L1 expression was predictive of response regardless of tumor histology. Additionally, the single-arm, open-label, multicenter Phase II ABACUS study, which evaluated two cycles of neoadjuvant atezolizumab prior to radical cystectomy in 74 patients with T2-T4bN0 urothelial carcinoma who not fit for or refused cisplatin-based treatment.23 The pathologic complete response rate was 29%, and 39% of patients were downstaged to non-muscle invasive disease.

These study findings have spurred larger trials of PD-1/PD-L1 checkpoint inhibitors for patients with muscle-invasive bladder cancer undergoing radical cystectomy. The randomized, open-label, global Phase III NIAGARA trial (NCT03732677) compares neoadjuvant durvalumab in combination with gemcitabine and cisplatin, followed by adjuvant durvalumab, versus neoadjuvant gemcitabine-cisplatin without adjuvant therapy.24 The targeted enrollment is 1,050 patients, and the co-primary endpoints are event-free survival and pathologic complete response at cystectomy. Another Phase III trial (NCT03661320) with the same co-primary endpoints randomizes patients to one of three treatment regimens: 1) neoadjuvant chemotherapy alone, 2) neoadjuvant chemotherapy plus nivolumab, followed by adjuvant nivolumab, and 3) neoadjuvant chemotherapy plus nivolumab plus the indoleamine-pyrrole 2,3-dioxygenase (IDO) inhibitor BMS-986205. This study aims to enroll 1,200 patients.

The vast majority of our patients with muscle-invasive bladder cancer would prefer not to undergo cystectomy. Hence, there is considerable interest in developing multi-modal bladder-sparing strategies. In a single-arm, open-label Phase II trial (NCT02621151), patients with >T2 disease receive pembrolizumab prior to maximal transurethral resection of bladder tumor (TURBT), followed by gemcitabine plus three courses of pembrolizumab plus radiation therapy (52 Gy over 4 weeks), followed by transurethral resection of the tumor bed.25 The primary endpoint is bladder-intact disease-free survival. The randomized Phase III S1806 study (NCT03775265) is also accruing patients with T3-3a bladder cancer who receive radiation and chemotherapy with or without atezolizumab.25 The primary endpoint is bladder-intact event-free survival.

Safety considerations

Patients who receive immune checkpoint inhibitors should be actively monitored for toxicities throughout the treatment course. The primary risk of this treatment class is the development of immune-mediated adverse events (IMAEs). Any organ system can be affected, and neuropathies and arthralgias can also occur. The most commonly experienced IMAEs may include pneumonitis, VOLUME 5, ISSUE 1 17 colitis, dermatitis, hepatic transaminitis and thyroiditis. In the KEYNOTE-045 study, each of these four toxicities affected approximately 1% to 6% of patients receiving second-line pembrolizumab for advanced bladder cancer, and 0.4% to 2.3% of events were of grade 3 classification.2

It is essential to educate patients and their caregiver team about the signs and symptoms of immune-mediated adverse events, the fact that they can occur any time during treatment, and the need for prompt reporting to ensure rapid intervention. I recommend developing literature to educate patients on signs and symptoms to watch for and how to report possible toxicities. The American Society of Clinical Oncology (ASCO) recommends taking a graded approach to management of immune-mediated adverse events.26 Treatment can be continued with close monitoring for most grade 1 events, with the exception of certain cardiac, neurologic, and hematologic toxicities. For grade 2 events, it is appropriate to suspend treatment, initiate corticosteroid therapy, and resume checkpoint inhibitor therapy only if the adverse event resolves to grade 1 or less. Grade 3 events often require management with high-dose corticosteroids (prednisone 1 to 2 mg/kg/ day or methylprednisolone 1 to 2 mg/kg/day), which should be tapered over at least 4 to 6 weeks. Grade 4 events should trigger prompt and permanent treatment cessation, unless the patient has an endocrinopathy that is controlled by hormone replacement therapy. Refractory immune-mediated adverse events may require intravenous immunosuppressive therapy with infliximab or mycophenolate, inpatient hospitalization, and multidisciplinary specialty support.

When managing more severe or less common immune-mediated adverse events, it can be advantageous to consult with specialists, such as gastroenterologists, endocrinologists, medical oncologists and other medical specialists. I recommend establishing and maintaining collaborative medical specialty relationships to optimize both the ABC and APC success.

Beyond Checkpoint Inhibition

Other Therapies Several other bladder cancer therapeutic with novel mechanism of action have received FDA approval or are under current FDA review, pending recent completion of a registrational trial. These include immunotoxins, antibody-drug conjugates, inhibitors of fibroblast growth factor receptor (FGFR), and gene therapies.

Immunotoxins 

Immunotoxins and antibody-drug conjugates are proteins consisting of a cytotoxic domain chemically linked to a target-specific binding domain.27 The immunotoxin oportuzumab monatox (Vicinium®) consists of a truncated form of Pseudomonas exotoxin A linked to a humanized antibody fragment targeting epithelial cell adhesion molecule (EpCAM), which is overexpressed on the surface of urothelial carcinoma cells.28 Oportuzumab monatox has a dual mechanism of action that involves both direct cell killing and immune system activation. In a Phase II trial of intravesical oportuzumab monatox for the treatment of BCG-unresponsive CIS or papillary bladder cancer, 16% of patients remained disease-free at 1 year and all patients with a complete response at 1 year remained disease-free for at least 18 months.29 Most adverse events were bladder-specific rather than systemic, and no serious treatment-related adverse events were identified. In a subsequent Phase III trial, patients received a more intensive treatment schedule consisting of intravesical oportuzumab induction therapy twice weekly for 6 weeks and then once weekly for 6 weeks, followed by a maintenance schedule of every other week for up to 2 years.30 In the interim analysis, 68% of patients were recurrence-free at 3 months. Serious adverse events were infrequent.

Antibody-drug conjugates

Antibody-drug conjugates consist of a cytotoxic payload (a cytotoxic antitumor molecule) chemically linked to a monoclonal antibody that targets a tumor cell surface antigen.31 After binding the target antigen, the antibody-drug conjugate undergoes receptor-mediated internalization into the tumor cell, where the chemical linker is degraded by lysosomes, proteases, or reductive agents. The cytotoxic payload is then released and causes microtubule disruption, cell cycle arrest, and apoptosis. These drugs are administered intravenously. The antibody-drug conjugate enfortumab vedotin (PadcevTM) targets Nectin-4, a type 1 transmembrane cell adhesion molecule that is overexpressed in epithelial cancers.

The antibody is bound to the microtubule-disrupting cytotoxic payload monomethyl auristatin E (MMAE) by means of a protease-cleavable chemical linker. In the single-arm Phase II EV-201 study, 44% of patients with metastatic urothelial carcinoma who had previously received anti-PD-1/L1 therapy and platinum-based therapy had a confirmed objective response to enfortumab vedotin (EV), 12% of patients had complete responses, and the median duration of response was 7.6 months.32 Based on these findings, the FDA in December 2019 approved EV for the third-line treatment of locally advanced or metastatic urothelial carcinoma in patients who have previously received both anti-PD-1/L1 and platinum-based therapy.33

EV is now being studied as an earlier-line therapy. The Phase 1b EV-103 study (NCT03288545) evaluates its first-line and second-line use alone or in combination with pembrolizumab and/or chemotherapy for patients with locally advanced or metastatic urothelial carcinoma. In February 2020, preliminary durability results were reported at the American Society of Clinical Oncology 2020 Genitourinary Cancers Symposium (ASCO GU 2020).34 After a median follow-up period of 11.5 months, the confirmed investigator-assessed objective response rate was 73.3%, the complete response rate was 15.6%, and median progression-free survival was 12.3 months. Data on overall survival are pending.

Another antibody-drug conjugate in development is sacituzumab govitecan, which targets Trop-2, an epithelial cell-surface glycoprotein that is highly expressed in muscle-invasive bladder cancer.35 A hydrolysable linker binds the antibody to SN-38, the active metabolite of irinotecan. In a Phase I/II basket trial that included 45 patients with metastatic urothelial carcinoma, sacituzumab govitecan induced responses in 31% of patients overall and 23% of prior recipients of checkpoint inhibitor therapy.36 Median progression-free survival was 7.3 months, and median overall survival was 18.9 months. The FDA is currently reviewing a biologics license application for sacituzumab govitecan for the treatment of triple-negative breast cancer. We may soon see a similar application for its therapeutic use in advanced bladder cancer. The near future will bring more trials of antibody-drug conjugates as later-line therapies for advanced and metastatic bladder cancer. As with other novel treatments, there also is the possibility of combining these agents with other novel agents and potentially moving them for earlier application and benefit within the bladder cancer disease spectrum.

Fibroblast growth factor receptor (FGFR) inhibitors

Fibroblast growth factor receptors (FGFR) are a subfamily of receptor tyrosine kinases that include FGFR1, FGFR2, FGFR3, FGFR4.37 Signaling by these receptors helps regulate cell growth, proliferation, differentiation, and survival. Increased expression of FGFR1 and FGFR3 is observed in urothelial carcinoma, and FGFR3 point mutations have been found in approximately 40% to 70% of non-muscle invasive bladder tumors and 15% to 20% of metastatic tumors.38

Erdafitinib (BalversaTM), a tyrosine kinase inhibitor of FGFR1-4, is orally administered and was evaluated in an open-label Phase II trial of 99 patients with locally advanced and unresectable or metastatic urothelial carcinoma with FGFR alterations.39 After a median of five treatment cycles, the confirmed response rate was 40% in the overall cohort and 59% among patients who had previously received immuno-oncologic therapy. Median progression-free survival was 5.5 months, and median overall survival was 13.8 months. In all, 46% of patients had grade 3 or higher adverse events that were considered related to treatment. Based on these data, the FDA in April 2019 granted erdafitinib accelerated approval for the treatment of locally advanced or metastatic bladder cancer with relevant FGFR3 or FGFR2 alterations that has progressed on platinum-based neoadjuvant or adjuvant chemotherapy.40 This is the first and only oral drug approved for the treatment of bladder cancer. Note that the FDA has approved the therascreen FGFR RGQ RT-PCR kit as a companion diagnostic to select candidates for treatment with erdafitinib.41

Combination first-line therapy with erdafitinib plus anti-PD-1 therapy is now being evaluated in a randomized Phase I/II trial (NORSE; NCT03473743) of patients with metastatic or surgically unresectable urothelial carcinoma with selected FGFR alterations. In a similar patient population, a Phase III trial (THOR; NCT03390504) compares second-line or third-line treatment with erdafitinib, pembrolizumab, vinflunine, or docetaxel.

In addition to erdafitinib, the oral selected FGFR[1-3] inhibitor pemigatinib is in Phase II studies for the treatment of recurrent, low or intermediate-risk non-muscle invasive bladder cancer, and for the adjuvant treatment of high-risk, FGFR-altered pT3-T4 and/or pN1-3 bladder cancer in patients who have previously received neoadjuvant cisplatin-based chemotherapy. 

Gene therapy

Nadofaragene firadenovec (rad-FNα/syn3; Adstiladrin®) is an investigational gene therapy for the treatment of high-grade, BCG-unresponsive, non-muscle invasive bladder cancer.42 It consists of a replication-deficient recombinant adenovirus gene transfer vector that delivers the human IFNα2b gene directly into urothelial cell nuclei, thereby increasing endogenous production of alfa-2b within the bladder wall.

Nadofaragene firadenovec has demonstrated substantial efficacy for the treatment of BCG unresponsive high-risk NMIBC. In a single-arm, Phase II trial of patients meeting this description who were cystectomy-ineligible, 35% of all patients and 50% of patients with papillary-only (Ta/T1) disease were alive and free from high-grade recurrence 12 months after receiving nadofaragene firadenovec.43 Furthermore, 79% of patients who responded to treatment remained recurrence-free at approximately two years.

In a subsequent single-arm Phase III trial of 157 patients with BCG-unresponsive non-muscle invasive bladder cancer, nadofaragene firadenovec (administered intravesically once every 3 months, with up to four doses in 12 months) achieved complete responses in 53% of patients with CIS, of whom 46% remained free from high-grade recurrence at 12 months.44 Among patients with high-grade Ta/T1 disease alone, 44% were free from high-grade recurrence at 12 months. Toxicities were considered acceptable, and the treatment schedule was regarded as very patient and clinic friendly.

Summary

After several decades of limited therapeutic progress for bladder cancer management, therapies for advanced urothelial carcinoma have expanded remarkably over the last several years. Five anti-PD-1/L1 checkpoint inhibitors are now FDA-approved for use in the second line, and two are approved as first-line treatments for cisplatin-ineligible patients. Ongoing studies of combination regimens, as well as of PD-L1 expression and other potential biomarkers of treatment response, are expected to further enhance outcomes and achieve broader indications. However, the greatest impact is likely to come from the use of checkpoint inhibitors for the management of earlier-stage disease, including the treatment of high-risk NMIBC-BCG naïve patients, the neoadjuvant treatment of muscle-invasive bladder cancer prior to radical cystectomy, and as a component of multimodal bladder-sparing strategies.

In addition to PD-1/PD-L1 pathway inhibitors, the antibody-drug conjugate enfortumab vedotin and the FGFR inhibitor erdafitinib have received accelerated FDA approval for advanced urothelial carcinoma. Other ongoing registrational trials are expected to further broaden our treatment options, as novel treatments and combinations continue to shift into earlier-stage and earlier-line use. Remarkably, we may soon have an approved immuno-oncologic agent and two new intravesical therapies for the management high-risk BCG-unresponsive NMIBC.

These novel bladder cancer therapeutics exhibit unique risk profiles that markedly differ from chemotherapy and require appropriate training for detection and management. Education of patients, families, and the clinical care team is vital to ensure that adverse effects are promptly detected, reported, and managed. Furthermore, the diversity of the treatment landscape and the advent of potential combined and multi-modal therapies mandates collaboration and coordination amongst urologists, medical and radiation oncologists, pathologists, medical specialists and nursing expertise in order to effectively deliver care across the bladder cancer disease spectrum. For dedicated urologists and their physician colleagues, these advances provide an exciting and long-awaited opportunity to initiate and expand the Advanced Bladder Cancer Clinic.

Written by: Neal Shore, MD, Editor-in-Chief, Everyday Urology - Oncology Insights, UroToday.com, Medical Director, The Carolina Urologic Research Center, Myrtle Beach, South Carolina

References:

1. Apolo, Andrea B., Jeffrey R. Infante, Ani Balmanoukian, Manish R. Patel, Ding Wang, Karen Kelly, Anthony E. Mega et al. "Avelumab, an anti–programmed death-ligand 1 antibody, in patients with refractory metastatic urothelial carcinoma: results from a multicenter, phase ib study." Journal of Clinical Oncology 35, no. 19 (2017): 2117.
2. Bellmunt, Joaquim, Ronald De Wit, David J. Vaughn, Yves Fradet, Jae-Lyun Lee, Lawrence Fong, Nicholas J. Vogelzang et al. "Pembrolizumab as second-line therapy for advanced urothelial carcinoma." New England Journal of Medicine 376, no. 11 (2017): 1015-1026.
3. Powles, Thomas, Ignacio Durán, Michiel S. Van Der Heijden, Yohann Loriot, Nicholas J. Vogelzang, Ugo De Giorgi, Stéphane Oudard 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." The Lancet 391, no. 10122 (2018): 748-757.
4. Powles, Thomas, Peter H. O'Donnell, Christophe Massard, Hendrik-Tobias Arkenau, Terence W. Friedlander, Christopher J. Hoimes, Jae Lyun Lee 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 oncology 3, no. 9 (2017): e172411-e172411.
5. Rosenberg, Jonathan E., Jean Hoffman-Censits, Tom Powles, Michiel S. Van Der Heijden, Arjun V. Balar, Andrea Necchi, Nancy Dawson 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." The Lancet 387, no. 10031 (2016): 1909-1920.
6. Sharma, Padmanee, Margitta Retz, Arlene Siefker-Radtke, Ari Baron, Andrea Necchi, Jens Bedke, Elizabeth R. Plimack et al. "Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial." The Lancet Oncology 18, no. 3 (2017): 312-322.
7. Balar, Arjun V., Daniel Castellano, Peter H. O'Donnell, Petros Grivas, Jacqueline Vuky, Thomas Powles, Elizabeth R. Plimack 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." The Lancet Oncology 18, no. 11 (2017): 1483-1492.
8. Balar, Arjun V., Matthew D. Galsky, Jonathan E. Rosenberg, Thomas Powles, Daniel P. Petrylak, Joaquim Bellmunt, Yohann Loriot 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." The Lancet 389, no. 10064 (2017): 67-76.
9. Suzman, Daniel L., Sundeep Agrawal, Yang‐min Ning, V. Ellen Maher, Laura L. Fernandes, Stella Karuri, Shenghui Tang et al. "FDA Approval Summary: Atezolizumab or Pembrolizumab for the Treatment of Patients with Advanced Urothelial Carcinoma Ineligible for Cisplatin‐Containing Chemotherapy." Oncologist 24, no. 4 (2019).
10. US Food and Drug Administration. (2018). FDA alerts health care professionals and oncology Clinical investigators about an efficacy issue identified in clinical trials for some patients taking Keytruda (pembrolizumab) or Tecentriq (atezolizumab) as monotherapy to treat urothelial cancer with low expression of PD-L1. FDA https://www. fda. gov/Drugs/DrugSafety/ucm608075. htm.
11. O'Donnell, Peter H., Petros Grivas, Arjun Vasant Balar, Joaquim Bellmunt, Jacqueline Vuky, Thomas Powles, Elizabeth R. Plimack et al. "Biomarker findings and mature clinical results from KEYNOTE-052: First-line pembrolizumab (pembro) in cisplatin-ineligible advanced urothelial cancer (UC)." (2017): 4502-4502.
12. Jarow, Jonathan, V. Ellen Maher, Shenghui Tang, Amna Ibrahim, Geoffrey Kim, Rajeshwari Sridhara, and Richard Pazdur. "Development of systemic and topical drugs to treat non-muscle invasive bladder cancer." Bladder Cancer 1, no. 2 (2015): 133-136.
13. Steinberg, Gary, Robert Bahnson, Stanley Brosman, Richard Middleton, Z. E. V. Wajsman, Michael Wehle, and VALRUBICIN STUDY GROUP‡. "Efficacy and safety of valrubicin for the treatment of Bacillus Calmette-Guerin refractory carcinoma in situ of the bladder." The Journal of urology 163, no. 3 (2000): 761-767.
14. De Wit, Ronald, Girish S. Kulkarni, Edward M. Uchio, Laurence Eliot Miles Krieger, Joost L. Boormans, Mathieu Roumiguié, Eric A. Singer et al. "Pembrolizumab (pembro) for patients (pts) with high-risk (HR) non–muscle invasive bladder cancer (NMIBC) unresponsive to Bacillus Calmette-Guérin (BCG): Updated follow-up from KEYNOTE-057." (2019): 4530-4530.
15. de Wit, R., G. S. Kulkarni, E. Uchio, J. L. Boormans, M. Roumiguié, L. E. M. Krieger, E. A. Singer et al. "916P Health-related quality of life (HRQoL) and updated follow-up from KEYNOTE-057: Phase II study of pembrolizumab (pembro) for patients (pts) with high-risk (HR) non-muscle invasive bladder cancer (NMIBC) unresponsive to bacillus calmette-guérin (BCG)." Annals of Oncology 30, no. Supplement_5 (2019): mdz249-015.
16.  Research, Center for Drug Evaluation and. “FDA Approves Pembrolizumab for BCG-Unresponsive, High-Risk Non-Muscle Invasive Bladder Cancer.” FDA, January 8, 2020. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-bcg-unresponsive-high-risk-non-muscle-invasive-bladder-cancer.
17. Kamat, Ashish M., Neal D. Shore, Noah M. Hahn, Shaheen Alanee, Hiroyuki Nishiyama, Shahrokh Shariat, Kijoeng Nam, Ekta Kapadia, Tara L. Frenkl, and Gary D. Steinberg. "Keynote-676: Phase 3 study of bacillus calmette-guerin (BCG) with or without pembrolizumab (pembro) for high-risk (HR) non–muscle invasive bladder cancer (NMIBC) that is persistent or recurrent following BCG induction." (2019): TPS502-TPS502.
18. De Santis, Maria, Ramil Abdrashitov, Axel Hegele, Margaret Kolb, Suzanne Parker, Juan Palou Redorta, Hiroyuki Nishiyama, Feng Xiao, Ashok Kumar Gupta, and Neal D. Shore. "A phase III, randomized, open-label, multicenter, global study of durvalumab and bacillus calmette-guérin (BCG) versus BCG alone in high-risk, BCG-naïve non-muscle-invasive bladder cancer (NMIBC) patients (POTOMAC)." (2019): TPS500-TPS500.
19. Roupret, M., Neuzillet, Y., Bertaut, A., Pignot, G., Houede, N., Champiat, S., ... & Loriot, Y. (2019). ALBAN: An open label, randomized, phase III trial, evaluating efficacy of atezolizumab in addition to one year BCG (bacillus Calmette-Guerin) bladder instillation in BCG-naive patients with high-risk nonmuscle invasive bladder cancer (AFU-GETUG 37).
20. Grossman, H. Barton, Ronald B. Natale, Catherine M. Tangen, V. O. Speights, Nicholas J. Vogelzang, Donald L. Trump, Ralph W. deVere White et al. "Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer." New England Journal of Medicine 349, no. 9 (2003): 859-866.
21. Necchi, Andrea, Andrea Anichini, Daniele Raggi, Alberto Briganti, Simona Massa, Roberta Lucianò, Maurizio Colecchia 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." Journal of Clinical Oncology 36, no. 34 (2018): 3353-3360.
22. Necchi, Andrea, Daniele Raggi, Andrea Gallina, Russell Madison, Maurizio Colecchia, Roberta Lucianò, Rodolfo Montironi et al. "Updated results of PURE-01 with preliminary activity of neoadjuvant pembrolizumab in patients with muscle-invasive bladder carcinoma with variant histologies." European Urology 77, no. 4 (2020): 439-446.
23. Castellano D. ESMO 2018: ABACUS, A phase II Study Investigating the Safety and Efficacy of Neoadjuvant Atezolizumab in Muscle Invasive Bladder Cancer. ESMO 2018 Congress. Munich, Germany2018.
24. Powles, Thomas, Joshua J. Meeks, Matt D. Galsky, Michiel Simon Van Der Heijden, Hiroyuki Nishiyama, Hikmat Al-Ahmadie, Ashok Kumar Gupta et al. "A phase III, randomized, open label, multicenter, global study of efficacy and safety of durvalumab in combination with gemcitabine+ cisplatin (G+ C) for neoadjuvant treatment followed by durvalumab alone for adjuvant treatment in muscle-invasive bladder cancer (MIBC)(NIAGARA)." (2019): TPS4592-TPS4592.
25. Grivas, Petros, Alexandra Drakaki, Terence W. Friedlander, and Guru Sonpavde. "Conceptual Framework for Therapeutic Development Beyond Anti–PD-1/PD-L1 in Urothelial Cancer." American Society of Clinical Oncology Educational Book 39 (2019): 284-300.
26. Brahmer, Julie R., Christina Lacchetti, Bryan J. Schneider, Michael B. Atkins, Kelly J. Brassil, Jeffrey M. Caterino, Ian Chau et al. "Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline." Journal of clinical oncology: official journal of the American Society of Clinical Oncology 36, no. 17 (2018): 1714.
27. Dosio, Franco, Paola Brusa, and Luigi Cattel. "Immunotoxins and anticancer drug conjugate assemblies: the role of the linkage between components." Toxins 3, no. 7 (2011): 848-883.
28. Agarwal, Piyush K., and Cora N. Sternberg. "Clinical Trials Corner." Bladder cancer (Amsterdam, Netherlands) 2, no. 4 (2016): 469.
29. Kowalski, Mark, Jacinthe Guindon, Louise Brazas, Celine Moore, Joycelyn Entwistle, Jeannick Cizeau, Michael AS Jewett, and Glen C. MacDonald. "A phase II study of oportuzumab monatox: an immunotoxin therapy for patients with noninvasive urothelial carcinoma in situ previously treated with bacillus Calmette-Guerin." The Journal of urology 188, no. 5 (2012): 1712-1718.
30. Dickstein R, Wu N, Curt D, Franks M, Wolk F. VISTA, Phase 3 Trial Of Vicinium, An Epcam-Targeted Pseudomonas Exotoxin, In Bcg-Unresponsive Non-Muscle Invasive Bladder Cancer. Global Congress on Bladder Cancer; 2018; Madrid.
31. Nejadmoghaddam, Mohammad-Reza, Arash Minai-Tehrani, Ramin Ghahremanzadeh, Morteza Mahmoudi, Rassoul Dinarvand, and Amir-Hassan Zarnani. "Antibody-drug conjugates: possibilities and challenges." Avicenna journal of medical biotechnology 11, no. 1 (2019): 3.
32. Rosenberg, Jonathan E., Peter H. O’Donnell, Arjun V. Balar, Bradley A. McGregor, Elisabeth I. Heath, Evan Y. Yu, Matthew D. Galsky et al. "Pivotal trial of enfortumab vedotin in urothelial carcinoma after platinum and anti-programmed death 1/programmed death ligand 1 therapy." Journal of Clinical Oncology 37, no. 29 (2019): 2592.
33. Research, Center for Drug Evaluation and. “FDA Grants Accelerated Approval to Enfortumab Vedotin-Ejfv for Metastatic Urothelial Cancer.” FDA, December 20, 2019. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-enfortumab-vedotin-ejfv-metastatic-urothelial-cancer.
34. Rosenberg JE. Study EV-103: Preliminary Durability Results of Enfortumab Vedotin Plus Pembrolizumab for Locally Advanced or Metastatic Urothelial Carcinoma. 2020 Genitourinary Cancers Symposium. San Francisco, CA2020.
35. Bardia, Aditya, Ingrid A. Mayer, Linda T. Vahdat, Sara M. Tolaney, Steven J. Isakoff, Jennifer R. Diamond, Joyce O’Shaughnessy et al. "Sacituzumab govitecan-hziy in refractory metastatic triple-negative breast cancer." New England Journal of Medicine 380, no. 8 (2019): 741-751.
36. Tagawa, Scott T., Bishoy Morris Faltas, Elaine Tat Lam, Philip James Saylor, Aditya Bardia, Julio Hajdenberg, Alicia K. Morgans et al. "Sacituzumab govitecan (IMMU-132) in patients with previously treated metastatic urothelial cancer (mUC): Results from a phase I/II study." (2019): 354-354.
37. Tiong, Kai Hung, Li Yen Mah, and Chee-Onn Leong. "Functional roles of fibroblast growth factor receptors (FGFRs) signaling in human cancers." Apoptosis 18, no. 12 (2013): 1447-1468.
38. Rodriguez-Vida, Alejo, Matilde Saggese, Simon Hughes, Sarah Rudman, Simon Chowdhury, Neil R. Smith, Peter Lawrence et al. "Complexity of FGFR signalling in metastatic urothelial cancer." Journal of hematology & oncology 8, no. 1 (2015): 119.
39.Loriot, Yohann, Andrea Necchi, Se Hoon Park, Jesus Garcia-Donas, Robert Huddart, Earle Burgess, Mark Fleming et al. "Erdafitinib in locally advanced or metastatic urothelial carcinoma." New England Journal of Medicine 381, no. 4 (2019): 338-348.
40. Highlights of Prescribing Information: Balversa. 2019. at https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212018s000lbl.pdf.)
41. US Food and Drug Administration. "List of cleared or approved companion diagnostic devices (in vitro and imaging tools)." US Food and Drug Administration (2015).
42. FerGene Announces Pivotal Phase 3 Study of Nadofaragene Firadenovec Met Its Primary Endpoint With More Than Half of Patients With High-Grade Non-Muscle Invasive Bladder Cancer (CIS ± Ta/T1) Achieving a Complete Response at Three Months. 2019. (Accessed March 3, 2020, at https://www.businesswire.com/news/home/20191205005851/en/ FerGene-Announces-Pivotal-Phase-3-Study-Nadofaragene.)
43. Shore, Neal D., Stephen A. Boorjian, Daniel J. Canter, Kenneth Ogan, Lawrence I. Karsh, Tracy M. Downs, Leonard G. Gomella et al. "Intravesical rAd–IFNα/Syn3 for patients with high-grade, bacillus calmette-guerin–refractory or relapsed non–muscle-invasive bladder cancer: a phase II randomized study." Journal of Clinical Oncology 35, no. 30 (2017): 3410.
44. Boorjian, Stephen A., Colin PN Dinney, and SUO Clinical Trials Consortium. "Safety and efficacy of intravesical nadofaragene firadenovec for patients with high-grade, BCG unresponsive nonmuscle invasive bladder cancer (NMIBC): Results from a phase III trial." (2020): 442-442.

Related Content:

Download: Everyday Urology: Volume 5, Issue 1