Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1702 2023-06-16 11:50:21 |
2 layout -1 word(s) 1701 2023-06-19 05:21:12 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Roviello, G.; Catalano, M.; Santi, R.; Palmieri, V.E.; Vannini, G.; Galli, I.C.; Buttitta, E.; Villari, D.; Rossi, V.; Nesi, G. Immune Checkpoint Inhibitors in Urothelial Bladder Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/45713 (accessed on 13 August 2024).
Roviello G, Catalano M, Santi R, Palmieri VE, Vannini G, Galli IC, et al. Immune Checkpoint Inhibitors in Urothelial Bladder Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/45713. Accessed August 13, 2024.
Roviello, Giandomenico, Martina Catalano, Raffaella Santi, Valeria Emma Palmieri, Gianmarco Vannini, Ilaria Camilla Galli, Eleonora Buttitta, Donata Villari, Virginia Rossi, Gabriella Nesi. "Immune Checkpoint Inhibitors in Urothelial Bladder Cancer" Encyclopedia, https://encyclopedia.pub/entry/45713 (accessed August 13, 2024).
Roviello, G., Catalano, M., Santi, R., Palmieri, V.E., Vannini, G., Galli, I.C., Buttitta, E., Villari, D., Rossi, V., & Nesi, G. (2023, June 16). Immune Checkpoint Inhibitors in Urothelial Bladder Cancer. In Encyclopedia. https://encyclopedia.pub/entry/45713
Roviello, Giandomenico, et al. "Immune Checkpoint Inhibitors in Urothelial Bladder Cancer." Encyclopedia. Web. 16 June, 2023.
Immune Checkpoint Inhibitors in Urothelial Bladder Cancer
Edit

Bladder cancer (BC) is the most common malignancy of the genitourinary tract, with high morbidity and mortality rates. Until recently, the treatment of locally advanced or metastatic urothelial BC was based on the use of chemotherapy alone. Since 2016, five immune checkpoint inhibitors (ICIs) have been approved by the Food and Drug Administration (FDA) in different settings, i.e., first-line, maintenance and second-line treatment, while several trials are still ongoing in the perioperative context. Lately, pembrolizumab, a programmed death-1 (PD-1) inhibitor, has been approved for Bacillus Calmette–Guérin (BCG)-unresponsive high-risk non-muscle invasive bladder cancer (NMIBC), using immunotherapy at an early stage of the disease.

urothelial carcinoma bladder cancer PD-1 PD-L1

1. Introduction

Bladder cancer (BC) is the ninth-most common malignancy worldwide, with 83,730 estimated new cases in the USA in 2021 [1] and the seventh-most common cancer in men [2]. Tobacco smoke appears to be the most common risk factor for BC, accounting for approximately 50% of cases [3]. Compared with never smokers, BC risk is three-fold higher in former smokers and over six-fold higher in current smokers, steadily increasing with the number of cigarettes and years smoked [4]. Occupational exposure is responsible for 5–6% of urothelial carcinomas. Among dietary factors, alcohol appears to play a role in the pathogenesis of BC, while the intake of Vitamin D and daily consumption of fruit and vegetables could have a protective effect [5].
At the time of diagnosis, approximately 70% of urothelial carcinomas are superficial, while 30% present with muscle infiltration [2]. Treatment of non-muscle invasive bladder cancer (NMIBC) involves transurethral resection of the bladder tumor (TURBT) followed by intravesical chemotherapy or immunotherapy. Bacillus Calmette–Guérin (BCG) immunotherapy is the gold standard adjuvant treatment for NMIBC with a high risk of progression and is also recommended for intermediate-risk NMIBC [6].
The standard treatment for nonmetastatic muscle invasive bladder cancer (MIBC) (T2–T4, N0, M0) is neoadjuvant cisplatin-based therapy, succeeded by radical cystectomy (RC) and pelvic lymphadenectomy [7]. Patients undergoing RC for MIBC have a high risk of relapse, especially in cases of ≥pT2 disease and/or pathological lymph node involvement. Adjuvant cisplatin-based multi-chemotherapy may be considered for patients fulfilling platinum eligibility criteria that include at least one of the following: Eastern Cooperative Oncology Group (ECOG) performance status of 2, creatinine clearance less than 60 mL/min, grade ≥ 2 hearing loss, grade ≥ 2 neuropathy, and/or New York Heart Association Class III heart failure [8][9].
Cisplatin-containing chemotherapy is the preferred first-line treatment also in metastatic disease. The most commonly used regimens in this setting include a combination of gemcitabine and cisplatin (GC), methotrexate, vincristine, adriamycin and cisplatin (MVAC) every four weeks, or dose-dense (dd) MVAC every two weeks. The median overall survival (OS) rates are 13.8 months, 14.8 months, and 15.5 months for GC, MVAC, and ddMVAC regimens, respectively [10][11]. Outcome is poor for patients who are unfit for platinum chemotherapy or undergo progression after frontline platinum chemotherapy, however, a major milestone in the metastatic setting was the approval of immune checkpoint inhibitors (ICIs) (Table 1).
Table 1. Currently approved ICIs administered in urothelial bladder carcinoma.
Trial Phase FDA Approval No. of Patients ICI Therapy Line of Treatment Previous Platinum Therapy Efficacy Outcomes
IMvigor210 [12] II May 2016 310 Atezolizumab Second line Yes mPFS: 2.1 mo
mOS: 7.9 mo
ORR: 18%
CheckMate-275 [13] II February 2017 265 Nivolumab Second line Yes mPFS: 2.0 mo
mOS: 8.7 mo
ORR: 20%
IMvigor210 [14] II April 2017 123 Atezolizumab First line PD-L1+ platinum ineligible patients No mPFS: 2.7 mo
mOS: 15.9 mo
ORR: 23%
JAVELIN Solid Tumor [15] I May 2017 44 Avelumab Second line Yes mPFS: 11.6 wk
mOS: 13.7 mo
ORR: 18.2%
Study 1108 [16] I/II May 2017 191 Durvalumab Second line Yes mPFS: 1.5 mo
mOS: 18.2 mo
ORR: 18%
KEYNOTE-045 [17] III May 2017 542 Pembrolizumab Second line Yes mPFS: 2.1 mo
mOS: 10.3 mo
ORR: 21%
KEYNOTE-052 [18] II May 2017 370 Pembrolizumab First line PD-L1 + platinum ineligible patients No mPFS: 2.2 mo
mOS: 11.3 mo
ORR: 29%
JAVELIN Bladder 100 [19] III June 2020 700 Avelumab Maintenance therapy Yes mPFS: 3.7 mo
mOS: 21.4 mo
Immune checkpoint inhibitor (ICI); programmed death ligand 1 (PD-L1); median progression-free survival (mPFS); median overall survival (mOS); objective response rate (ORR); months (mo); weeks (wk).
ICIs are monoclonal antibodies directed against cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4), programmed death 1 (PD-1) receptor and programmed death ligand-1 (PD-L1). CTLA-4 is a membrane receptor acting as a major negative regulator of T cell responses through interaction with its ligands, CD80 (B7-1) and CD86 (B7-2), expressed on the surface of antigen-presenting cells. PD-1 is a membrane receptor expressed by T cells, particularly in conditions of chronic antigen exposure, and exerts an inhibitory action on lymphocytes by binding to its two ligands, PD-L1 and PD-L2. PD-L1 is expressed on immune cells, such as T cells, B cells, dendritic cells (DCs) and macrophages [20][21], while PD-L2 is expressed mainly on antigen-presenting cells (APCs), including macrophages and myeloid DCs [22][23]. PD-L1 and PD-L2 have differential functions in immune regulatory processes. Indeed, PD-L1 inhibits T cells in peripheral tissues, whereas PD-L2 suppresses immune T cell activation in lymphoid organs. PD-L2 also inhibits type 2 T-helper (TH2) lymphocytes, but its role is yet to be fully understood [24][25]. By interrupting the ligand/receptor interactions, the anti-CTLA-4 (ipilimumab, tremelimumab) and anti-PD-1 (nivolumab, pembrolizumab)/anti-PD-L1 (atezolizumab, durvalumab, avelumab) antibodies remove T cell inhibition, thus favoring antitumor cytotoxic activity [26] (Figure 1). Characterization of immune checkpoints has furthered development of novel immunotherapeutic agents with clinical activity against a variety of solid tumors, including BC.
Figure 1. Mechanisms of action of ICIs targeting PD-1, PD-L1, and CTLA-4. PD-1 and CTLA-4 are proteins expressed on activated T cells. Their binding to the respective ligands presented on the surface of cancer cells leads to T cell inactivation and prevents tumor cell death. The immune checkpoint blockade ensures the activation of T cells and favors antitumor activity. Created with BioRender.com (accessed on 26 July 2021). PD-1: Programmed cell death-1; PD-L1: Programmed cell death-ligand 1; CTLA-4: Cytotoxic T-lymphocyte-associated antigen 4.
This research current evidence supporting the use of new checkpoint inhibitors in BC, along with information on biomarkers that may predict response to immunotherapy.

2. Non-Muscle Invasive Bladder Cancer (NMIBC)

In approximately 75% of BC patients, the disease is confined to the mucosa (stage Ta, carcinoma in situ) or submucosa (stage T1) [27]. Although TURB alone can eradicate TaT1 tumors completely, they commonly recur and can progress to MIBC, thus necessitating the use of adjuvant treatment. In patients with intermediate-risk tumors, one-year full-dose BCG treatment or chemotherapy instillations for a maximum of one year is recommended. Conversely, full-dose intravesical BCG for one to three years is indicated in patients with high-risk tumors [28].
Therapeutic options for patients with BCG-unresponsive disease include RC, further intravesical therapy, and systemic therapy. A relatively new addition to the landscape of treatment for BCG-unresponsive NMIBC is pembrolizumab. Initial results of the KEYNOTE-057 phase II trial were reported in February 2019 showing a 38.8% (40/102) complete response (CR) rate at 3 months. Following the presentation of these data, pembrolizumab received FDA approval in January 2020 for BCG-unresponsive high-risk NMIBC patients, ineligible for, or refusing RC. Key secondary endpoints were duration of response (DOR) and safety. At a median follow-up of 14 months, 72.5% of patients maintained CR, 25.0% experienced recurrent NMIBC after CR, but none progressed to MIBC. Treatment-related adverse events (AEs) occurred in 63.1% of patients, the most frequent being pruritus, fatigue, diarrhea, hypothyroidism, and maculopapular rash. Grade 3–4 AEs occurred in 12.6% of patients, and one death due to colitis was considered treatment-related [29]. Updated data over a 2-year follow-up were submitted at the 2020 American Society of Clinical Oncology (ASCO) Annual Meeting. The median DOR was 16.2 months, and CR rate was 40.6% with 46.2% of responses longer than 12 months. The median PFS and OS were not reached [30].
At the 2021 ASCO Genitourinary Cancers Symposium, Balar et al. reported additional results with an extended minimum follow-up of 26.3 months [31]. Among those patients achieving CR, 33.3% remained in CR for ≥18 months and 23.1% for ≥24 months as of the data cutoff date. Of the 41.7% patients undergoing cystectomy after discontinuation of pembrolizumab, 35 (88%) had no pathological upstaging to MIBC, three (8%) had evidence of MIBC, and two (5%) had no available pathology data. Safety profile remained consistent with what had been previously reported.
Another phase II trial, SWOG S1605, tested atezolizumab in the same setting. The primary outcome was the pathological complete response (pCR) rate at six months, accomplished through mandatory biopsy. A pCR was observed in 30 (41.1%) patients at 3 months and in 19 (26.0%) at 6 months. The most common AEs were fatigue, pruritus, hypothyroidism, and nausea. Grade 3–5 AEs occurred in 12.3% of patients, and there was one treatment-related death due to myasthenia gravis [32].
Several clinical trials with other ICI agents, both as monotherapy and as part of a combination therapy, are ongoing and in early-stage BC. Particularly relevant are the POTOMAC trial assessing durvalumab plus BCG in BCG-naïve patients, the KEYNOTE-676 study evaluating BCG-associated pembrolizumab in patients with recurrence after induction BCG therapy alone [33], and the NCT03317158 trial establishing the safety of durvalumab as monotherapy and in combination with BCG and external beam radiation therapy (EBRT) in BCG-unresponsive NMIBC patients (Table 2).
Table 2. Ongoing phase II/III trials with active recruitment on ICIs alone or in combination with chemotherapy in different settings of BC treatment.
Trial Phase Allocation No. of Patients Study Populations Line of Treatment Experimental Arms Primary Outcome
NCT02736266 II N/A 90 MIBC neoadjuvant prior to chemoradiation Pembrolizumab pCR
NCT02845323 II randomized 44 MIBC neoadjuvant Nivolumab + Urelumab vs. Nivolumab Immune response (tumor infiltrating CD8+ T cell density)
NCT03520491 II not randomized 45 Cisplatin-ineligible patients with MIBC neoadjuvant Nivolumab and Nivolumab + Ipilimumab No. of patients who proceed to RC-PLND
NCT03472274 II randomized 99 BC patients neoadjuvant Durvalumab and Tremelimumab Antitumor activity
NCT03732677 III randomized 1050 MIBC neoadjuvant/adjuvant Durvalumab + Gemcitabine + Cisplatin neoadjuvant treatment followed by Durvalumab alone for adjuvant treatment EFS
NCT04138628 II randomized 282 Treatment of mBC at the time of biochemical relapse following RC adjuvant Atezolizumab CR
NCT03244384 III randomized 739 Locally advanced and mUC adjuvant Pembrolizumab vs. observation OS, DFS
NCT04223856 III randomized 760 Previously untreated locally advanced or mUC 1st Enfortumab vedotin + Pembrolizumab vs. chemotherapy alone PFS, OS
NCT03036098 III randomized 1290 Unresectable or mUC 1st Nivolumab + Ipilimumab, or SoC chemotherapy vs. SoC Chemotherapy OS, PFS
NCT03682068 III randomized 1434 Unresectable locally advanced or mUC 1st Durvalumab + SoC chemotherapy and Durvalumab + Tremelimumab and SoC Chemotherapy vs. SoC chemotherapy alone OS
NCT03898180 III randomized 694 Locally advanced or mUC 1st Pembrolizumab + Lenvatinib vs. Pembrolizumab +placebo PFS, OS
NCT03697850 II randomized 77 MIBC patients ineligible for RC maintenance therapy Atezolizumab DFS
Not applicable (N/A); number (No.); metastatic urothelial cancer (mUC); metastatic bladder cancer (mBC); standard of care (SoC); best supportive care (BSC); overall survival (OS); progression free survival (PFS); disease free survival (DFS); events free survival (EFS); pathological complete response (pCR); radical cystectomy and pelvic lymph node dissection (RC-PLND); muscle-invasive bladder cancer (MIBC).

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33.
  2. Antoni, S.; Ferlay, J.; Soerjomataram, I.; Znaor, A.; Jemal, A.; Bray, F. Bladder Cancer Incidence and Mortality: A Global Overview and Recent Trends. Eur. Urol. 2017, 71, 96–108.
  3. Cumberbatch, M.G.; Rota, M.; Catto, J.W.; La Vecchia, C. The Role of Tobacco Smoke in Bladder and Kidney Carcinogenesis: A Comparison of Exposures and Meta-analysis of Incidence and Mortality Risks. Eur. Urol. 2016, 70, 458–466.
  4. Polesel, J.; Bosetti, C.; di Maso, M.; Montella, M.; Libra, M.; Garbeglio, A.; Zucchetto, A.; Turati, F.; Talamini, R.; La Vecchia, C.; et al. Duration and intensity of tobacco smoking and the risk of papillary and non-papillary transitional cell carcinoma of the bladder. Cancer Causes Control 2014, 25, 1151–1158.
  5. Cumberbatch, M.G.; Jubber, I.; Black, P.C.; Esperto, F.; Figueroa, J.D.; Kamat, A.M.; Kiemeney, L.; Lotan, Y.; Pang, K.; Silverman, D.T.; et al. Epidemiology of Bladder Cancer: A Systematic Review and Contemporary Update of Risk Factors in 2018. Eur. Urol. 2018, 74, 784–795.
  6. Babjuk, M.; Burger, M.; Compérat, E.M.; Gontero, P.; Mostafid, A.H.; Palou, J.; van Rhijn, B.W.G.; Rouprêt, M.; Shariat, S.F.; Sylvester, R.; et al. European Association of Urology guidelines on non-muscle-invasive bladder cancer (TaT1 and carcinoma in situ)—2019 Update. Eur. Urol. 2019, 76, 639–657.
  7. Ghandour, R.; Singla, N.; Lotan, Y. Treatment options and outcomes in nonmetastatic muscle invasive bladder cancer. Trends Cancer 2019, 5, 426–439.
  8. Kim, D.K.; Lee, J.Y.; Jung, J.H.; Hah, Y.S.; Cho, K.S. Role of adjuvant cisplatin-based chemotherapy following radical cystectomy in locally advanced muscle-invasive bladder cancer: Systematic review and meta-analysis of randomized trials. Investig. Clin. Urol. 2019, 60, 64–74.
  9. Galsky, M.D.; Hahn, N.M.; Rosenberg, J.; Sonpavde, G.; Hutson, T.; Oh, W.K.; Dreicer, R.; Vogelzang, N.; Sternberg, C.N.; Bajorin, D.F.; et al. Treatment of patients with metastatic urothelial cancer “unfit” for cisplatin-based chemotherapy. J. Clin. Oncol. 2011, 29, 2432–2438.
  10. Von der Maase, H.; Hansen, S.W.; Roberts, J.T.; Dogliotti, L.; Oliver, T.; Moore, M.J.; Bodrogi, I.; Albers, P.; Knuth, A.; Lippert, C.M.; et al. Gemcitabine and cisplatin versus methotrexate, vinblastine, doxorubicin, and cisplatin in advanced or metastatic bladder cancer: Results of a large, randomized, multinational, multicenter, phase III study. J. Clin. Oncol. 2000, 18, 3068–3077.
  11. Sternberg, C.N.; de Mulder, P.H.; Schornagel, J.H.; Théodore, C.; Fossa, S.D.; van Oosterom, A.T.; Witjes, F.; Spina, M.; van Groeningen, C.J.; de Balincourt, C.; et al. Randomized phase III trial of high-dose-intensity methotrexate, vinblastine, doxorubicin, and cisplatin (MVAC) chemotherapy and recombinant human granulocyte colony-stimulating factor versus classic MVAC in advanced urothelial tract tumors: European Organization for Research and Treatment of Cancer Protocol no. 30924. J. Clin.Oncol. 2001, 19, 2638–2646.
  12. Rosenberg, J.E.; Hoffman-Censits, J.; Powles, T.; van der Heijden, M.S.; Balar, A.V.; Necchi, A.; Dawson, N.; O’Donnell, P.H.; Balmanoukian, A.; Loriot, Y.; et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: A single-arm, multicentre, phase 2 trial. Lancet 2016, 387, 1909–1920.
  13. Sharma, P.; Retz, M.; Siefker-Radtke, A.; Baron, A.; Necchi, A.; Bedke, J.; Plimack, E.R.; Vaena, D.; Grimm, M.O.; Bracarda, S.; et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): A multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017, 18, 312–322.
  14. Balar, A.V.; Galsky, M.D.; Rosenberg, J.E.; Powles, T.; Petrylak, D.P.; Bellmunt, J.; Loriot, Y.; Necchi, A.; Hoffman-Censits, J.; Perez-Gracia, J.L.; et al. IMvigor210 Study Group. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: A single-arm, multicentre, phase 2 trial. Lancet 2017, 389, 67–76.
  15. Apolo, A.B.; Infante, J.R.; Balmanoukian, A.; Patel, M.R.; Wang, D.; Kelly, K.; Mega, A.E.; Britten, C.D.; Ravaud, A.; Mita, A.C.; et al. Avelumab, an anti-programmed death-ligand 1 antibody, in patients with refractory metastatic urothelial carcinoma: Results from a multicenter, phase Ib study. J. Clin. Oncol. 2017, 35, 2117–2124.
  16. Powles, T.; O’Donnell, P.H.; Massard, C.; Arkenau, H.T.; Friedlander, T.W.; Hoimes, C.J.; Lee, J.L.; Ong, M.; Sridhar, S.S.; Vogelzang, N.J.; 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, e172411.
  17. Bellmunt, J.; de Wit, R.; Vaughn, D.J.; Fradet, Y.; Lee, J.L.; Fong, L.; Vogelzang, N.J.; Climent, M.A.; Petrylak, D.P.; Choueiri, T.K.; et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N. Engl. J. Med. 2017, 376, 1015–1026.
  18. Vuky, J.; Balar, A.V.; Castellano, D.; O’Donnell, P.H.; Grivas, P.; Bellmunt, J.; Powles, T.; Bajorin, D.; Hahn, N.M.; Savage, M.J.; et al. Long-term outcomes in KEYNOTE-052: Phase II study investigating first-line pembrolizumab in cisplatin-ineligible patients with locally advanced or metastatic urothelial cancer. J. Clin. Oncol. 2020, 38, 2658–2666.
  19. Powles, T.; Park, S.H.; Voog, E.; Caserta, C.; Valderrama, B.P.; Gurney, H.; Kalofonos, H.; Radulović, S.; Demey, W.; Ullén, A.; et al. Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. N. Engl. J. Med. 2020, 383, 1218–1230.
  20. Francisco, L.M.; Sage, P.T.; Sharpe, A.H. The PD-1 pathway in tolerance and autoimmunity. Immunol. Rev. 2010, 236, 219–242.
  21. Parry, R.V.; Chemnitz, J.M.; Frauwirth, K.A.; Lanfranco, A.R.; Braunstein, I.; Kobayashi, S.V.; Linsley, P.S.; Thompson, C.B.; Riley, J.L. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell Biol. 2005, 25, 9543–9553.
  22. Riella, L.V.; Paterson, A.M.; Sharpe, A.H.; Chandraker, A. Role of the PD-1 pathway in the immune response. Am. J. Transplant. 2012, 12, 2575–2587.
  23. Momtaz, P.; Postow, M.A. Immunologic checkpoints in cancer therapy: Focus on the programmed death-1 (PD-1) receptor pathway. Pharmgenom. Pers Med. 2014, 7, 357–365.
  24. Massari, F.; Santoni, M.; Ciccarese, C.; Santini, D.; Alfieri, S.; Martignoni, G.; Brunelli, M.; Piva, F.; Berardi, R.; Montironi, R.; et al. PD-1 blockade therapy in renal cell carcinoma: Current studies and future promises. Cancer Treat Rev. 2015, 41, 114–121.
  25. Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr. Opin. Immunol. 2012, 24, 207–212.
  26. Ribas, A. Tumor immunotherapy directed at PD-1. N. Engl. J. Med. 2012, 366, 2517–2519.
  27. Treatment by Cancer Type . . Available online: https://www.nccn.org/guidelines/category_1 (accessed on 1 July 2021).
  28. EAU Guidelines | Uroweb . . Available online: https://uroweb.org/guidelines (accessed on 1 July 2021).
  29. Balar, A.V.; Kulkarni, G.S.; Uchio, E.M.; Boormans, J.; Mourey, L.; Krieger, L.E.M.; Singer, E.A.; Bajorin, D.F.; Kamat, A.M.; Grivas, P.; et al. Keynote 057: Phase II trial of Pembrolizumab (pembro) for patients (pts) with high-risk (HR) nonmuscle invasive bladder cancer (NMIBC) unresponsive to Bacillus Calmette-Guérin (BCG). J. Clin. Oncol. 2019, 37 (Suppl. S7), 350.
  30. Balar, A.V.; Kamat, A.M.; Kulkarni, G.S.; Uchio, E.M.; Boormans, J.L.; Bajorin, D.F.; Roumiguié, M.; Singer, E.A.; Krieger, L.E.M.; Grivas, P.; et al. Pembrolizumab (pembro) for the treatment of patients with Bacillus Calmette-Guérin (BCG) unresponsive, high-risk (HR) non–muscle-invasive bladder cancer (NMIBC): Over two years follow-up of KEYNOTE-057. J. Clin. Oncol. 2020, 38 (Suppl. S15), 5041.
  31. Balar, A.V.; Kamat, A.M.; Kulkami, G.S.; Uchio, E.M.; Boormans, J.L.; Roumiguié, M.; Krieger, L.E.M.; Singer, E.A.; Bajorin, D.F.; Grivas, P.; et al. Pembrolizumab for the treatment of patients with high-risk (HR) non-muscle-invasive bladder cancer (NMIBC) unresponsive to Bacillus Calmette-Guérin: Extended follow-up of KEYNOTE-057 cohort A. J. Clin. Oncol. 2021, 39 (Suppl. S6), 451.
  32. Black, P.C.; Tangen, C.; Singh, P.; McConkey, D.J.; Lucia, S.; Lowrance, W.T.; Koshkin, V.S.; Stratton, K.L.; Bivalacqua, T.; Sharon, E.; et al. Phase II trial of atezolizumab in BCG-unresponsive non-muscle invasive bladder cancer: SWOG S1605 (NCT #02844816). J. Clin. Oncol. 2020, 38 (Suppl. S15), 5022.
  33. Kamat, A.M.; Shore, N.; Hahn, N.; Alanee, S.; Nishiyama, H.; Shariat, S.; Nam, K.; Kapadia, E.; Frenkl, T.; Steinberg, G. KEYNOTE-676: Phase III study of BCG and pembrolizumab for persistent/recurrent high-risk NMIBC. Future Oncol. 2020, 16, 507–516.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , ,
View Times: 388
Revisions: 2 times (View History)
Update Date: 19 Jun 2023
1000/1000
Video Production Service