Immunotherapy Applications in Urology
PDF
Cite
Share
Request
Review
VOLUME: ISSUE:
P: -

Immunotherapy Applications in Urology

1. Marmara University Faculty of Medicine Department of Urology, İstanbul, Turkiye
No information available.
No information available
Received Date: 22.11.2024
Accepted Date: 26.01.2025
Online Date: 18.04.2025
PDF
Cite
Share
Request

Abstract

Immunotherapy has transformed the management of urological cancers, offering renewed optimism for individuals diagnosed with bladder, kidney, and, to a lesser degree, prostate tumors. Conventional therapies, such as chemotherapy and radiation, frequently demonstrate restricted efficacy. At the same time, immune checkpoint inhibitors (ICIs) have surfaced as promising alternatives, especially in urothelial and renal cell carcinoma (RCC). This review examines immunotherapy mechanisms, focusing on immune checkpoint pathways such as programmed cell death protein 1 and cytotoxic T lymphocyte-associated protein 4, and their contributions to improving immune recognition and eliminating tumor cells. Bacillus Calmette-Guérin immunotherapy is essential for non-muscle-invasive bladder cancer, whereas ICIs have transformed the management of muscle-invasive and metastatic forms of the disease. In RCC, ICIs have markedly enhanced survival outcomes, whether alone or in conjunction with tyrosine kinase inhibitors. Although advancements have been made, the role of immunotherapy in prostate cancer remains in development, with limited efficacy documented thus far. Current research focuses on optimizing combination therapies and identifying biomarkers to improve patient selection. The future of immunotherapy in urology involves its incorporation into earlier treatment stages and its combination with innovative agents, which may enhance outcomes for patients with urological cancers.

Keywords:
Immunotherapy, urooncology, bladder cancer, prostate cancer, renal cancer

Introduction

Immunotherapy has transformed the treatment of numerous malignancies, and its use in urology has profoundly altered the management of urological tumors. Conventional therapies for urological malignancies, including chemotherapy and radiation, frequently provide suboptimal results, whereas immunotherapy presents a promising approach for enhancing outcomes, especially in bladder, kidney, and, to a lesser degree, prostate tumors.

In the field of urothelial carcinoma and renal cell carcinoma (RCC), immune checkpoint inhibitors (ICIs) have become crucial components, utilized either as standalone treatments or in conjunction with other therapies. Notwithstanding these developments, their effectiveness in prostate cancer remains limited. This study examines the basic mechanisms of immunotherapy, its present applications in various urological malignancies, and future prospects that may enhance patient outcomes.

Mechanisms of Immunotherapy in Urological Malignancies

Immunotherapy in urological malignancies improves the body’s capacity by enhancing the immune system’s ability to identify and eliminate malignant cells. The progression of cancer signifies a disruption in the equilibrium between immune surveillance and tumor evasion systems, permitting the unregulated proliferation of aberrant cells. Immunotherapy aims to restore this equilibrium by enhancing the immune system’s ability to identify and eliminate tumor cells that have escaped recognition.

Mechanisms of Tumoral Immune Evasion

The microenvironment of the tumor contains several mechanisms that prevent the immune system from effectively combating the tumor, including T-cell exhaustion. Due to prolonged exposure to antigenic stimuli, exhausted T-cells exhibit a loss of normal T-cell functions, and their effector capacities (e.g., cytokine production and cell killing abilities) are reduced. These cells become resistant to reactivation and express high levels of multiple inhibitory surface molecules, such as cytotoxic T lymphocyte antigen-4 (CTLA-4), programmed death 1 (PD-1), lymphocyte activation gene-3, and T-cell immunoreceptor with Ig and Immunoreceptor Tyrosine-based Inhibitory Motif (ITIM) domains, T-cell immunoglobulin and ITIM domain. These molecules suppress T-cell activation and help tumor cells evade the immune system (1).

Cancer cells bypass immune detection through many strategies, such as diminished tumor antigen production and the secretion of inhibitory chemicals that provoke T-cell anergy or apoptosis (2). Immune checkpoint molecules, including programmed death ligand 1 (PD-L1), are pivotal in this process. The binding of PD-L1 on tumor cells to the PD-1 receptor on T lymphocytes initiates an inhibitory signal that diminishes T-cell activity, hence promoting immune evasion (3).

The CTLA-4/CD80-CD86 association similarly inhibits T-cell activation, facilitating immunological evasion. The introduction of ICIs, including anti-PD-1/PD-L1 and anti-CTLA-4 antibodies, marks a key advancement in the management of many urological cancers. By obstructing these inhibitory pathways, ICIs can rejuvenate T lymphocytes, enabling them to effectively target and eliminate tumor cells (Figure 1).

Advances in Immuno-Oncology

Recent advancements in immuno-oncology have led to medications that accurately target specific immune pathways, improving precision and reducing off-target effects. ICIs have demonstrated notable effectiveness in RCC and bladder cancer, leading to their integration into treatment protocols. However, the benefits of immunotherapy for prostate cancer are still under investigation, producing mixed results so far.

The US Food and Drug Administration has approved ICIs for clinical use in certain genitourinary tumor patients (5). Biomarkers are pivotal in early tumor diagnosis, drug development, disease monitoring, and prognosis evaluation. Many methods exist to detect biomarkers, depending on the laboratory and the material to be analyzed (such as tissue or serum). Polymerase chain reaction is a common method for mRNA or DNA-based analysis. ELISA, Western Blot, or immunohistochemical examination may be preferred for a specific analysis at the protein level. Programmed death ligand-1 (PD-L1) expression is a widely used biomarker to predict response to immunotherapy and is evaluated according to expression levels in tumor cells or immune cells (6). Microsatellite instability (MSI) and high tumor mutational burden are other important biomarkers that indicate that immunotherapy may be effective (7). Additionally, the presence of intratumoral CD8+ T-cells may indicate a strong immune response (8). Selecting patients with higher mutational burden, with specific markers, may increase the likelihood of response to immunotherapy.

Immunotherapy for Bladder Cancer

Non-muscle invasive bladder cancer (NMIBC) Bacillus Calmette-Guérin (BCG) immunotherapy has long been a fundamental treatment for NMIBC, demonstrating more efficacy than transurethral resection of the bladder alone or in conjunction with intravesical chemotherapy in minimizing recurrence (9, 10). A Cochrane review demonstrated the advantage of BCG compared to mitomycin-C in decreasing NMIBC recurrence (11). Moreover, maintenance BCG therapy has been shown to be effective in reducing the risk of progression in high- and intermediate-risk NMIBC (12, 13). Intracavitary treatment poses a potential risk for disseminated BCG infection (in less than 5% of patients) and may cause infusion reactions (14). The presence or absence of side effects does not seem to be a prognostic factor for the efficacy of BCG, and maintenance therapy is not associated with a significant increase in toxicity (15).

Recent data on BCG-unresponsive patients with carcinoma in situ (16), either alone or with concomitant papillary tumors, have shown promising results with new immunotherapies. Systemic pembrolizumab demonstrated a 40% complete response rate in a phase II prospective study, with 48% of responders maintaining their response for up to 12 months (17). Promising results from a phase III multicenter randomized controlled trial (RCT) demonstrated that intravesical nadofaragene firadenovec achieved a 53.4% complete response rate in patients with BCG-unresponsive carcinoma in situ (16). Forty-five percent of responders maintained their response at one year (18). Additional ongoing studies are exploring the use of combination therapies involving intravesical or systemic immunotherapy to enhance treatment outcomes (19, 20).

Muscle-invasive and Metastatic Bladder Carcinoma

In recent years, immunotherapy has emerged as a prominent option for treating muscle-invasive bladder cancer. Traditionally, chemotherapy has remained the first-line treatment for metastatic disease for an extended period; however, it is increasingly being supplanted by immunotherapy approaches. Preliminary studies indicate that the ICI pembrolizumab demonstrates an overall survival advantage of approximately three months compared to second-line chemotherapy. Nevertheless, the current data are insufficient to facilitate its immediate integration into routine clinical practice (21).

The phase III trial Alliance A031501 AMBASSADOR demonstrated that adjuvant pembrolizumab significantly improved disease-free survival (29.6 months vs. 14.2 months; hazard ratio: 0.73, p=0.003) compared to observation in patients with high-risk muscle-invasive urothelial carcinoma after radical surgery. These findings support pembrolizumab as an effective adjuvant therapy in this population. However, pembrolizumab was associated with a higher rate of grade 3 or higher adverse events (50.6% vs. 31.6%) (22).

Nivolumab, a PD-1/PD-L1 checkpoint inhibitor, is recommended as an adjuvant treatment for patients with tumor cell PD-L1 expression ≥1% who are at high risk of recurrence after surgery in non-metastatic pT3-4 urothelial carcinoma and cannot receive cisplatin-based chemotherapy. The CheckMate 274 trial, which indicated significant improvements in disease-free survival (23), supports this recommendation.

The EV-302/KEYNOTE A39 and Checkmate 901 RCTs have recently revised the first-line treatment algorithm in metastatic disease (24, 25). The combination of enfortumab vedotin (EV) and pembrolizumab in metastatic urothelial carcinoma now establishes the new standard of care for patients who are considered eligible for combination therapies. The major eligibility criteria include an Eastern Cooperative Oncology Group performance status of 0-2, a glomerular filtration rate of ≥30 mL/min, and adequate organ function, as determined by the requirements for treatment with EV, and Pembrolizumab. This combination has significantly enhanced progression-free survival (PFS) and overall survival, irrespective of PD-L1 expression. PFS was significantly prolonged with EV+P vs. chemo, reducing the risk of progression or death by 55% (median PFS, 12.5 mo vs. 6.3 mo, respectively). Additionally, severe side effects were found to be lower than those associated with chemotherapy (24).

However, it should be noted that EV has not yet been included in the reimbursement scope of the social security institution in our country. Li et al. (27) evaluated the cost-effectiveness of EV plus pembrolizumab as a first-line treatment for metastatic urothelial carcinoma compared to chemotherapy. While EV plus pembrolizumab improved survival, providing an additional 2.10 life-years (26) and 1.72 quality-adjusted life-years (QALYs), the incremental cost-effectiveness ratio was $558,973 per QALY-well above the willingness-to-pay threshold of $150,000 per QALY. Subgroup analysis suggested that the combination was slightly more cost-effective in cisplatin-ineligible patients, but overall, the therapy is not considered cost-effective from the perspective of U.S. payers (27).

Numerous combinations are currently being studied in various clinical studies. The JAVELIN bladder 100 study evaluated the efficacy of ongoing treatment with the PD-L1 inhibitor avelumab following platinum-gemcitabine chemotherapy. After four to six cycles of platinum-gemcitabine chemotherapy, an increase in overall survival was noted among patients treated with avelumab, with respective survival rates of 21.4 and 14.3 months for those who received and did not receive avelumab (28).

Currently, phase I, II, and III studies indicate that ICIs, including pembrolizumab, nivolumab, atezolizumab, avelumab, and durvalumab, exhibit comparable efficacy and safety in patients who have progressed during or following platinum-based chemotherapy (21, 29-32). Sacituzumab govitecan is a humanized monoclonal antibody that targets trophoblast cell surface antigen 2 (Trop-2). Research indicates that it enhances progression-free and overall survival prior to chemotherapy (33). As a result of new molecules or combinations, it is anticipated that standard treatment algorithms will undergo changes in the near future.

Adverse events can affect any organ in the body and range in severity from mild to severe. The most affected organs include the skin, gastrointestinal tract, liver, lungs, thyroid, adrenal glands, and pituitary gland. Other potentially impacted systems include the musculoskeletal, renal, nervous, hematologic, ocular, and cardiovascular systems. Any new symptoms or changes observed during immunotherapy should prompt consideration of a potential connection to the treatment (34).

Immunotherapy in Renal Carcinoma

The majority of immunotherapy studies in RCC focus on clear cell RCC (ccRCC), as it is the most prevalent subtype, accounting for approximately 70-80% of all RCC cases (35). As a result, there is limited knowledge regarding the optimal management of non-clear cell RCC (nccRCC) subtypes. Treatment options for nccRCC remain scarce due to the lack of specific studies focused on these variants. For these reasons, our review primarily focuses on clear cell RCC.

Before the introduction of ICIs, the primary treatments for metastatic RCC included tyrosine kinase inhibitors (TKIs), mTOR inhibitors, and vascular endothelial growth factor (VEGF) inhibitors. CheckMate trials 025 and 214 demonstrated that nivolumab, both alone and in combination with ipilimumab, enhanced overall survival in metastatic ccRCC, resulting in a significant shift in treatment approaches (36, 37).

As a monotherapy, nivolumab has demonstrated superiority over everolimus in terms of overall survival for patients with VEGF-refractory ccRCC. However, no advantage in PFS has been observed in this patient population (38). Currently, no RCTs support the use of single-agent ICIs in metastatic kidney cancer.

To date, numerous combination treatments have been investigated in the context of kidney cancer. Combining immunotherapy with interventions explicitly targeting the VEGF pathway has demonstrated significant efficacy. First-line ICI combination trials for clear-cell RCC are presented in Table 1. The Keynote 426 phase III clinical trial indicates that the combination of pembrolizumab and axitinib outperforms first-line sunitinib in terms of overall survival among treatment-naïve patients, irrespective of PD-L1 expression (39).

A comprehensive five-year analysis of the Keynote 426 study revealed that combination therapy offers a PFS advantage. In the study, for pembrolizumab + axitinib vs. sunitinib, the 60-month overall survival rates were 41.9% vs. 37.1%, and the 60-month PFS rates were 18.3% vs. 7.3%. Furthermore, no significant differences were identified in treatment-related side effects compared to standard treatments (40).

Randomized controlled phase III trials evaluating the combinations of nivolumab with cabozantinib, as well as lenvatinib with pembrolizumab, demonstrated a PFS advantage, compared with sunitinib. These studies assessed efficacy without regard to risk group or PD-L1 status (41, 42).

The COSMIC-313 study is the first RCT aimed at evaluating the efficacy of the cabozantinib-nivolumab-ipilimumab triple combination treatment against the nivolumab-ipilimumab standard treatment combination, with a cohort of 855 patients (22). Although the study has not yet yielded long-term results, initial findings suggest that the triple combination provides a significant advantage in PFS (43).

In light of these findings pertaining to the metastatic stage, new prospective studies are underway to assess the potential impact of immunotherapy, whether administered as neoadjuvant or adjuvant treatment, in patients with localized kidney cancer who are deemed to be at high risk of recurrence. Currently, evaluating PD-L1 expression status is not a standard procedure. Combination therapies, which include immunotherapy, have now been established as the standard treatment for metastatic kidney cancer. It is anticipated that modifications to the treatment algorithm may occur in the future due to numerous RCTs that are currently in progress.

The meta-analysis of 95 RCTs involving 40,552 participants evaluated the risk of renal adverse events (RAE) (11) associated with ICIs. The overall incidence of RAE and acute kidney injury was low, but anti-CTLA-4 monotherapy showed higher toxicity, particularly for grade 3-5 RAE, compared to other ICIs like anti-PD-1 and anti-PD-L1. Combination therapies, such as anti-CTLA-4 plus anti-PD-1 and ICI plus chemotherapy, were associated with higher risks of RAE and AKI compared to monotherapies or traditional therapies, with ICI plus chemotherapy being the most toxic regimen. These findings emphasize the need for careful monitoring of renal function in patients receiving ICI-based treatments (44).

There is certainly a need for studies reporting the cost-effectiveness of immunotherapy. The study evaluated the cost-effectiveness of seven treatment strategies for metastatic renal cell carcinoma, including immunotherapy-TKI combinations and sunitinib, using public-payer costs in the United States. Nivolumab + ipilimumab provided the highest QALYs at 3.6. Still, it was not cost-effective at a willingness-to-pay threshold of $150,000 USD/QALY because of its high incremental cost-effectiveness ratio of $297,465 to $348,516 USD compared to sunitinib. Sunitinib, as the least expensive option, emerged as the most cost-effective treatment, while cost reductions of 22-38% in NI could improve its cost-effectiveness (45).

Immunotherapy in Prostate Cancer

Unlike in bladder and kidney cancers, the use of immunotherapy has not yet gained widespread acceptance in prostate cancer due to limited efficacy.

In the context of castration-resistant prostate cancer (CRPC), sipuleucel-T immunotherapy has undergone extensive investigation. This therapeutic approach involves cultivating the patient’s serum mononuclear cells with the PA2024 fusion protein, which comprises a prostate antigen linked to granulocyte-macrophage colony-stimulating factor. Sipuleucel-T, formulated using the patient’s blood cells, has demonstrated an overall survival advantage of 4.1 months for CRPC patients exhibiting no or minimal symptoms. However, it has not shown an impact on disease progression (46). Many similar prostate cancer vaccine studies have been conducted (47, 48).

Research indicates that ICIs exhibit minimal efficacy in the treatment of prostate cancer. While some studies demonstrate a response to immunotherapy, the treatment for prostate cancer may require a more tailored approach for each patient. This necessity arises from the substantial variation in mutation burden and spectrum observed among patients with CRPC (49).

MSI arises from the insufficient functionality of DNA repair mechanisms. This deficiency within cancer cells can result in tumors being more readily identified by the immune system, thereby exhibiting an enhanced response to immunotherapy. Although individuals with high MSI in prostate cancer are infrequent, pembrolizumab has received FDA approval for patients with metastatic CRPC and may represent a beneficial supplementary treatment option (50, 51).

Conclusion

Recent advancements in immunotherapy have notably enhanced its application in urology, especially concerning the treatment of bladder and kidney cancers. ICIs are critical elements in the treatment protocols for these malignancies, providing significant enhancements in survival rates. Despite these advancements, the application of immunotherapy in prostate cancer is still limited, necessitating additional research to identify predictive biomarkers and enhance combination strategies for optimal benefit.

Current clinical trials are investigating the application of immunotherapy in both neoadjuvant and adjuvant contexts, with potential outcomes that may broaden its utilization in early-stage cancers. The advancement of knowledge regarding tumor biology and immune interactions is expected to lead to the development of innovative agents and combination therapies, thereby significantly altering the treatment landscape for urological cancers and providing new hope for patients.

Authorship Contributions

Concept: Y.Ş., B.Ş., Design Y.Ş., B.Ş., Data Collection or Processing: Y.Ş., Analysis or Interpretation: Y.Ş., B.Ş., Literature Search: Y.Ş., Writing: Y.Ş., B.Ş.
Conflict of Interest: No conflict of interest was declared by the author.
Financial Disclosure: The author declared that this study received no financial support.

References

1
Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12:492-499.
2
Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3:991-998.
3
Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252-264.
4
Pignot G, Houede N. Immunothérapie en urologie : principes et résultats [Immunotherapy in urology: concept and results]. Prog Urol. 2019;29:922-928.
5
Lorentzen CL, Haanen JB, Met Ö, Svane IM. Clinical advances and ongoing trials on mRNA vaccines for cancer treatment. Lancet Oncol. 2022;23:e450-e458.
6
Patel SP, Kurzrock R. PD-L1 Expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14:847-856.
7
Palmeri M, Mehnert J, Silk AW, Jabbour SK, Ganesan S, Popli P, Riedlinger G, Stephenson R, de Meritens AB, Leiser A, Mayer T, Chan N, Spencer K, Girda E, Malhotra J, Chan T, Subbiah V, Groisberg R. Real-world application of tumor mutational burden-high (TMB-high) and microsatellite instability (MSI) confirms their utility as immunotherapy biomarkers. ESMO Open. 2022;7:100336.
8
Oh DY, Fong L. Cytotoxic CD4+ T cells in cancer: expanding the immune effector toolbox. Immunity. 2021;54:2701-2711.
9
Shelley MD, Kynaston H, Court J, Wilt TJ, Coles B, Burgon K, Mason MD. A systematic review of intravesical bacillus Calmette-Guérin plus transurethral resection vs transurethral resection alone in Ta and T1 bladder cancer. BJU Int. 2001;88:209-216.
10
Böhle A, Jocham D, Bock PR. Intravesical bacillus Calmette-Guerin versus mitomycin C for superficial bladder cancer: a formal meta-analysis of comparative studies on recurrence and toxicity. J Urol. 2003;169:90-95.
11
Schmidt S, Kunath F, Coles B, Draeger DL, Krabbe LM, Dersch R, Kilian S, Jensen K, Dahm P, Meerpohl JJ. Intravesical bacillus Calmette-Guérin versus mitomycin C for Ta and T1 bladder cancer. Cochrane Database Syst Rev. 2020;1:CD011935.
12
Böhle A, Bock PR. Intravesical bacille Calmette-Guérin versus mitomycin C in superficial bladder cancer: formal meta-analysis of comparative studies on tumor progression. Urology. 2004;63:682-686.
13
Sylvester RJ, van der MEIJDEN AP, Lamm DL. Intravesical bacillus Calmette-Guerin reduces the risk of progression in patients with superficial bladder cancer: a meta-analysis of the published results of randomized clinical trials. J Urol. 2002;168:1964-1970.
14
Ramalingam S, Gunasekaran K, Arora H, Muruganandam M, Nagaraju S, Padmanabhan P. Disseminated BCG Infection after intravesical BCG immunotherapy of bladder cancer. QJM. 2021;114:410-411.
15
Sánchez González Á, Rodríguez Faba O, Mosquera L, Sabiote L, Breda A, Palou Redorta J. BCG y cáncer de vejiga: pasado, presente y futuro [BCG and bladder cancer: past, present and future.]. Arch Esp Urol. 2018;71:332-341.
16
Choueiri TK, Motzer RJ, Rini BI, Haanen J, Campbell MT, Venugopal B, Kollmannsberger C, Gravis-Mescam G, Uemura M, Lee JL, Grimm MO, Gurney H, Schmidinger M, Larkin J, Atkins MB, Pal SK, Wang J, Mariani M, Krishnaswami S, Cislo P, Chudnovsky A, Fowst C, Huang B, di Pietro A, Albiges L. Updated efficacy results from the JAVELIN Renal 101 trial: first-line avelumab plus axitinib versus sunitinib in patients with advanced renal cell carcinoma. Ann Oncol. 2020;31:1030-1039.
17
Balar AV, Kamat AM, Kulkarni GS, Uchio EM, Boormans JL, Roumiguié M, Krieger LEM, Singer EA, Bajorin DF, Grivas P, Seo HK, Nishiyama H, Konety BR, Li H, Nam K, Kapadia E, Frenkl T, de Wit R. Pembrolizumab monotherapy for the treatment of high-risk non-muscle-invasive bladder cancer unresponsive to BCG (KEYNOTE-057): an open-label, single-arm, multicentre, phase 2 study. Lancet Oncol. 2021;22:919-930.
18
Boorjian SA, Alemozaffar M, Konety BR, Shore ND, Gomella LG, Kamat AM, Bivalacqua TJ, Montgomery JS, Lerner SP, Busby JE, Poch M, Crispen PL, Steinberg GD, Schuckman AK, Downs TM, Svatek RS, Mashni J Jr, Lane BR, Guzzo TJ, Bratslavsky G, Karsh LI, Woods ME, Brown G, Canter D, Luchey A, Lotan Y, Krupski T, Inman BA, Williams MB, Cookson MS, Keegan KA, Andriole GL Jr, Sankin AI, Boyd A, O’Donnell MA, Sawutz D, Philipson R, Coll R, Narayan VM, Treasure FP, Yla-Herttuala S, Parker NR, Dinney CPN. Intravesical nadofaragene firadenovec gene therapy for BCG-unresponsive non-muscle-invasive bladder cancer: a single-arm, open-label, repeat-dose clinical trial. Lancet Oncol. 2021;22:107-117.
19
Hahn NM, O’Donnell MA, Efstathiou JA, Zahurak M, Rosner GL, Smith J, Kates MR, Bivalacqua TJ, Tran PT, Song DY, Baras AS, Matoso A, Choi W, Smith KN, Pardoll DM, Marchionni L, McGuire B, Grace Phelan M, Johnson BA 3rd, O’Neal T, McConkey DJ, Rose TL, Bjurlin M, Lim EA, Drake CG, McKiernan JM, Deutsch I, Anderson CB, Lamm DL, Geynisman DM, Plimack ER, Hallman MA, Horwitz EM, Al-Saleem E, Chen DYT, Greenberg RE, Kutikov A, Guo G, Masterson TA, Adra N, Kaimakliotis HZ. A phase 1 trial of durvalumab in combination with bacillus Calmette-Guerin (BCG) or external beam radiation therapy in patients with BCG-unresponsive non-muscle-invasive bladder cancer: the hoosier cancer research network GU16-243 ADAPT-BLADDER study. Eur Urol. 2023;83:486-494.
20
Chamie K, Chang SS, Kramolowsky E, Gonzalgo ML, Agarwal PK, Bassett JC, Bjurlin M, Cher ML, Clark W, Cowan BE, David R, Goldfischer E, Guru K, Jalkut MW, Kaffenberger SD, Kaminetsky J, Katz AE, Koo AS, Sexton WJ, Tikhonenkov SN, Trabulsi EJ, Trainer AF, Spilman P, Huang M, Bhar P, Taha SA, Sender L, Reddy S, Soon-Shiong P. IL-15 Superagonist NAI in BCG-unresponsive non-muscle-invasive bladder cancer. NEJM Evid. 2023;2:EVIDoa2200167.
21
Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee JL, Fong L, Vogelzang NJ, Climent MA, Petrylak DP, Choueiri TK, Necchi A, Gerritsen W, Gurney H, Quinn DI, Culine S, Sternberg CN, Mai Y, Poehlein CH, Perini RF, Bajorin DF; KEYNOTE-045 Investigators. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med. 2017;376:1015-1026.
22
Apolo AB, Ballman KV, Sonpavde G, Berg S, Kim WY, Parikh R, Teo MY, Sweis RF, Geynisman DM, Grivas P, Chatta G, Reichert ZR, Kim JW, Bilen MA, McGregor B, Singh P, Tripathi A, Cole S, Simon N, Niglio S, Ley L, Cordes L, Srinivas S, Huang J, Odegaard M, Watt C, Petrylak D, Hoffman-Censits J, Wen Y, Hahn O, Mitchell C, Tan A, Streicher H, Sharon E, Moon H, Woods M, Halabi S, Perez Burbano G, Morris MJ, Rosenberg JE. Adjuvant pembrolizumab versus observation in muscle-invasive urothelial carcinoma. N Engl J Med. 2025;392:45-55.
23
Galsky MD, Bajorin DF, Witjes JA, Gschwend JE, Tomita Y, Nasroulah F, Li J, Collette S, Valderrama BP, Grimm MO, Appleman L, Gravis G, Necchi A, Ye D, Stenner F, Wind-Rotolo M, Zhang J, Ünsal-Kaçmaz K. Disease-free survival analysis for patients with high-risk muscle-invasive urothelial carcinoma from the randomized checkmate 274 trial by PD-L1 combined positive score and tumor cell score. Eur Urol. 2023;83:432-440.
24
Powles T, Valderrama BP, Gupta S, Bedke J, Kikuchi E, Hoffman-Censits J, Iyer G, Vulsteke C, Park SH, Shin SJ, Castellano D, Fornarini G, Li JR, Gümüş M, Mar N, Loriot Y, Fléchon A, Duran I, Drakaki A, Narayanan S, Yu X, Gorla S, Homet Moreno B, van der Heijden MS; EV-302 Trial Investigators. Enfortumab vedotin and pembrolizumab in untreated advanced urothelial cancer. N Engl J Med. 2024;390:875-888.
25
van der Heijden MS, Sonpavde G, Powles T, Necchi A, Burotto M, Schenker M, Sade JP, Bamias A, Beuzeboc P, Bedke J, Oldenburg J, Chatta G, Ürün Y, Ye D, He Z, Valderrama BP, Ku JH, Tomita Y, Filian J, Wang L, Purcea D, Patel MY, Nasroulah F, Galsky MD; CheckMate 901 Trial Investigators. Nivolumab plus gemcitabine-cisplatin in advanced urothelial carcinoma. N Engl J Med. 2023;389:1778-1789.
26
Choueiri TK, Powles T, Burotto M, Escudier B, Bourlon MT, Zurawski B, Oyervides Juárez VM, Hsieh JJ, Basso U, Shah AY, Suárez C, Hamzaj A, Goh JC, Barrios C, Richardet M, Porta C, Kowalyszyn R, Feregrino JP, Żołnierek J, Pook D, Kessler ER, Tomita Y, Mizuno R, Bedke J, Zhang J, Maurer MA, Simsek B, Ejzykowicz F, Schwab GM, Apolo AB, Motzer RJ; CheckMate 9ER Investigators. Nivolumab plus cabozantinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2021;384:829-841.
27
Li A, Wu M, Xie O, Xiang H, Meng K, Tan C, Wang L, Wan X. Cost-effectiveness of first-line enfortumab vedotin in addition to pembrolizumab for metastatic urothelial carcinoma in the United States. Front Immunol. 2024;15:1464092.
28
Powles T, Park SH, Voog E, Caserta C, Valderrama BP, Gurney H, Kalofonos H, Radulović S, Demey W, Ullén A, Loriot Y, Sridhar SS, Tsuchiya N, Kopyltsov E, Sternberg CN, Bellmunt J, Aragon-Ching JB, Petrylak DP, Laliberte R, Wang J, Huang B, Davis C, Fowst C, Costa N, Blake-Haskins JA, di Pietro A, Grivas P. Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. N Engl J Med. 2020;383:1218-1230.
29
Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, Dawson N, O’Donnell PH, Balmanoukian A, Loriot Y, Srinivas S, Retz MM, Grivas P, Joseph RW, Galsky MD, Fleming MT, Petrylak DP, Perez-Gracia JL, Burris HA, Castellano D, Canil C, Bellmunt J, Bajorin D, Nickles D, Bourgon R, Frampton GM, Cui N, Mariathasan S, Abidoye O, Fine GD, Dreicer R. 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.
30
Powles T, Durán I, van der Heijden MS, Loriot Y, Vogelzang NJ, De Giorgi U, Oudard S, Retz MM, Castellano D, Bamias A, Fléchon A, Gravis G, Hussain S, Takano T, Leng N, Kadel EE 3rd, Banchereau R, Hegde PS, Mariathasan S, Cui N, Shen X, Derleth CL, Green MC, Ravaud A. 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;391:748-757.
31
Balar AV, Castellano DE, Grivas P, Vaughn DJ, Powles T, Vuky J, Fradet Y, Lee JL, Fong L, Vogelzang NJ, Climent MA, Necchi A, Petrylak DP, Plimack ER, Xu JZ, Imai K, Moreno BH, Bellmunt J, de Wit R, O’Donnell PH. Efficacy and safety of pembrolizumab in metastatic urothelial carcinoma: results from KEYNOTE-045 and KEYNOTE-052 after up to 5 years of follow-up. Ann Oncol. 2023;34:289-299.
32
Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, Cruz C, Bellmunt J, Burris HA, Petrylak DP, Teng SL, Shen X, Boyd Z, Hegde PS, Chen DS, Vogelzang NJ. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 2014;515:558-562.
33
Loriot Y, Petrylak DP, Rezazadeh Kalebasty A, Fléchon A, Jain RK, Gupta S, Bupathi M, Beuzeboc P, Palmbos P, Balar AV, Kyriakopoulos CE, Pouessel D, Sternberg CN, Tonelli J, Sierecki M, Zhou H, Grivas P, Barthélémy P, Tagawa ST. TROPHY-U-01, a phase II open-label study of sacituzumab govitecan in patients with metastatic urothelial carcinoma progressing after platinum-based chemotherapy and checkpoint inhibitors: updated safety and efficacy outcomes. Ann Oncol. 2024;35:392-401.
34
Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 2018;378:158-168.
35
Moch H, Amin MB, Berney DM, Compérat EM, Gill AJ, Hartmann A, Menon S, Raspollini MR, Rubin MA, Srigley JR, Hoon Tan P, Tickoo SK, Tsuzuki T, Turajlic S, Cree I, Netto GJ. The 2022 World Health Organization classification of tumours of the urinary system and male genital organs-part a: renal, penile, and testicular tumours. Eur Urol. 2022;82:458-468.
36
Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, Castellano D, Choueiri TK, Gurney H, Donskov F, Bono P, Wagstaff J, Gauler TC, Ueda T, Tomita Y, Schutz FA, Kollmannsberger C, Larkin J, Ravaud A, Simon JS, Xu LA, Waxman IM, Sharma P; CheckMate 025 Investigators. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373:1803-1813.
37
Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, Powles T, Donskov F, Neiman V, Kollmannsberger CK, Salman P, Gurney H, Hawkins R, Ravaud A, Grimm MO, Bracarda S, Barrios CH, Tomita Y, Castellano D, Rini BI, Chen AC, Mekan S, McHenry MB, Wind-Rotolo M, Doan J, Sharma P, Hammers HJ, Escudier B; CheckMate 214 Investigators. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378:1277-1290.
38
Motzer RJ, Escudier B, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Plimack ER, Procopio G, McDermott DF, Castellano D, Choueiri TK, Donskov F, Gurney H, Oudard S, Richardet M, Peltola K, Alva AS, Carducci M, Wagstaff J, Chevreau C, Fukasawa S, Tomita Y, Gauler TC, Kollmannsberger CK, Schutz FA, Larkin J, Cella D, McHenry MB, Saggi SS, Tannir NM. Nivolumab versus everolimus in patients with advanced renal cell carcinoma: updated results with long-term follow-up of the randomized, open-label, phase 3 checkmate 025 trial. Cancer. 2020;126:4156-4167.
39
Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, Pouliot F, Alekseev B, Soulières D, Melichar B, Vynnychenko I, Kryzhanivska A, Bondarenko I, Azevedo SJ, Borchiellini D, Szczylik C, Markus M, McDermott RS, Bedke J, Tartas S, Chang YH, Tamada S, Shou Q, Perini RF, Chen M, Atkins MB, Powles T; KEYNOTE-426 Investigators. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380:1116-1127.
40
Plimack ER, Powles T, Stus V, Gafanov R, Nosov D, Waddell T, Alekseev B, Pouliot F, Melichar B, Soulières D, Borchiellini D, McDermott RS, Vynnychenko I, Chang YH, Tamada S, Atkins MB, Li C, Perini R, Molife LR, Bedke J, Rini BI. Pembrolizumab plus axitinib versus sunitinib as first-line treatment of advanced renal cell carcinoma: 43-month follow-up of the phase 3 KEYNOTE-426 study. Eur Urol. 2023;84:449-454.
41
Erratum: nivolumab plus cabozantinib vs sunitinib for first-line treatment of advanced renal cell carcinoma (aRCC): 3-year follow-up from the phase 3 checkmate 9 ER trial. J Clin Oncol. 2023;41:3767.
42
Motzer R, Alekseev B, Rha SY, Porta C, Eto M, Powles T, Grünwald V, Hutson TE, Kopyltsov E, Méndez-Vidal MJ, Kozlov V, Alyasova A, Hong SH, Kapoor A, Alonso Gordoa T, Merchan JR, Winquist E, Maroto P, Goh JC, Kim M, Gurney H, Patel V, Peer A, Procopio G, Takagi T, Melichar B, Rolland F, De Giorgi U, Wong S, Bedke J, Schmidinger M, Dutcus CE, Smith AD, Dutta L, Mody K, Perini RF, Xing D, Choueiri TK; CLEAR Trial Investigators. Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma. N Engl J Med. 2021;384:1289-1300.
43
Choueiri TK, Powles T, Albiges L, Burotto M, Szczylik C, Zurawski B, Yanez Ruiz E, Maruzzo M, Suarez Zaizar A, Fein LE, Schutz FA, Heng DYC, Wang F, Mataveli F, Chang YL, van Kooten Losio M, Suarez C, Motzer RJ; COSMIC-313 Investigators. Cabozantinib plus nivolumab and ipilimumab in renal-cell carcinoma. N Engl J Med. 2023;388:1767-1778.
44
Liu K, Qin Z, Xu X, Li T, Ge Y, Mao H, Xing C. Comparative risk of renal adverse events in patients receiving immune checkpoint inhibitors: a bayesian network meta-analysis. Front Oncol. 2021;11:662731.
45
Yoo M, Nelson RE, Cutshall Z, Dougherty M, Kohli M. Cost-effectiveness analysis of six immunotherapy-based regimens and sunitinib in metastatic renal cell carcinoma: a public payer perspective. JCO Oncol Pract. 2023;19:e449-e456.
46
Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, Xu Y, Frohlich MW, Schellhammer PF; IMPACT Study Investigators. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411-422.
47
Simons JW, Sacks N. Granulocyte-macrophage colony-stimulating factor-transduced allogeneic cancer cellular immunotherapy: the GVAX vaccine for prostate cancer. Urol Oncol. 2006;24:419-424.
48
Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bilhartz DL, Wyand M, Manson K, Panicali DL, Laus R, Schlom J, Dahut WL, Arlen PM, Gulley JL, Godfrey WR. Overall survival analysis of a phase II randomized controlled trial of a poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol. 2010;28:1099-1105.
49
Fong L, Kwek SS, O’Brien S, Kavanagh B, McNeel DG, Weinberg V, Lin AM, Rosenberg J, Ryan CJ, Rini BI, Small EJ. Potentiating endogenous antitumor immunity to prostate cancer through combination immunotherapy with CTLA4 blockade and GM-CSF. Cancer Res. 2009;69:609-615.
50
Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: an overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol. 2018;62:29-39.
51
Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, Biedrzycki B, Donehower RC, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Duffy SM, Goldberg RM, de la Chapelle A, Koshiji M, Bhaijee F, Huebner T, Hruban RH, Wood LD, Cuka N, Pardoll DM, Papadopoulos N, Kinzler KW, Zhou S, Cornish TC, Taube JM, Anders RA, Eshleman JR, Vogelstein B, Diaz LA Jr. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509-2520.
52
Ljungberg B, Albiges L, Abu-Ghanem Y, Bedke J, Capitanio U, Dabestani S, Fernández-Pello S, Giles RH, Hofmann F, Hora M, Klatte T, Kuusk T, Lam TB, Marconi L, Powles T, Tahbaz R, Volpe A, Bex A. European association of urology guidelines on renal cell carcinoma: the 2022 update. Eur Urol. 2022;82:399-410.
53
Powles T, Plimack ER, Soulières D, Waddell T, Stus V, Gafanov R, Nosov D, Pouliot F, Melichar B, Vynnychenko I, Azevedo SJ, Borchiellini D, McDermott RS, Bedke J, Tamada S, Yin L, Chen M, Molife LR, Atkins MB, Rini BI. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol. 2020;21:1563-1573.
54
Plimack ER, Powles T, Stus V, Gafanov R, Nosov D, Waddell T, Alekseev B, Pouliot F, Melichar B, Soulières D, Borchiellini D, McDermott RS, Vynnychenko I, Chang YH, Tamada S, Atkins MB, Li C, Perini R, Molife LR, Bedke J, Rini BI. Pembrolizumab plus axitinib versus sunitinib as first-line treatment of advanced renal cell carcinoma: 43-month follow-up of the phase 3 KEYNOTE-426 study. Eur Urol. 2023;84:449-454.
55
Haanen JBAG, Larkin J, Choueiri TK, Albiges L, Rini BI, Atkins MB, Schmidinger M, Penkov K, Michelon E, Wang J, Mariani M, di Pietro A, Motzer RJ. Extended follow-up from JAVELIN Renal 101: subgroup analysis of avelumab plus axitinib versus sunitinib by the international metastatic renal cell carcinoma database consortium risk group in patients with advanced renal cell carcinoma. ESMO Open. 2023;8:101210.
56
Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, Venugopal B, Kollmannsberger C, Negrier S, Uemura M, Lee JL, Vasiliev A, Miller WH Jr, Gurney H, Schmidinger M, Larkin J, Atkins MB, Bedke J, Alekseev B, Wang J, Mariani M, Robbins PB, Chudnovsky A, Fowst C, Hariharan S, Huang B, di Pietro A, Choueiri TK. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380:1103-1115.
57
Motzer RJ, Powles T, Atkins MB, Escudier B, McDermott DF, Alekseev BY, Lee JL, Suarez C, Stroyakovskiy D, De Giorgi U, Donskov F, Mellado B, Banchereau R, Hamidi H, Khan O, Craine V, Huseni M, Flinn N, Dubey S, Rini BI. Final overall survival and molecular analysis in IMmotion151, a phase 3 trial comparing atezolizumab plus bevacizumab vs sunitinib in patients with previously untreated metastatic renal cell carcinoma. JAMA Oncol. 2022;8:275-280.
58
Rini BI, Powles T, Atkins MB, Escudier B, McDermott DF, Suarez C, Bracarda S, Stadler WM, Donskov F, Lee JL, Hawkins R, Ravaud A, Alekseev B, Staehler M, Uemura M, De Giorgi U, Mellado B, Porta C, Melichar B, Gurney H, Bedke J, Choueiri TK, Parnis F, Khaznadar T, Thobhani A, Li S, Piault-Louis E, Frantz G, Huseni M, Schiff C, Green MC, Motzer RJ; IMmotion151 Study Group. Atezolizumab plus bevacizumab versus sunitinib in patients with previously untreated metastatic renal cell carcinoma (IMmotion151): a multicentre, open-label, phase 3, randomised controlled trial. Lancet. 2019;393:2404-2415.
59
Motzer RJ, McDermott DF, Escudier B, Burotto M, Choueiri TK, Hammers HJ, Barthélémy P, Plimack ER, Porta C, George S, Powles T, Donskov F, Gurney H, Kollmannsberger CK, Grimm MO, Barrios C, Tomita Y, Castellano D, Grünwald V, Rini BI, McHenry MB, Lee CW, McCarthy J, Ejzykowicz F, Tannir NM. Conditional survival and long-term efficacy with nivolumab plus ipilimumab versus sunitinib in patients with advanced renal cell carcinoma. Cancer. 2022;128:2085-2097.
60
Motzer RJ, Powles T, Burotto M, Escudier B, Bourlon MT, Shah AY, Suárez C, Hamzaj A, Porta C, Hocking CM, Kessler ER, Gurney H, Tomita Y, Bedke J, Zhang J, Simsek B, Scheffold C, Apolo AB, Choueiri TK. Nivolumab plus cabozantinib versus sunitinib in first-line treatment for advanced renal cell carcinoma (CheckMate 9ER): long-term follow-up results from an open-label, randomised, phase 3 trial. Lancet Oncol. 2022;23:888-898.
61
Motzer RJ, Porta C, Eto M, Powles T, Grünwald V, Hutson TE, Alekseev B, Rha SY, Merchan J, Goh JC, Lalani AA, De Giorgi U, Melichar B, Hong SH, Gurney H, Méndez-Vidal MJ, Kopyltsov E, Tjulandin S, Gordoa TA, Kozlov V, Alyasova A, Winquist E, Maroto P, Kim M, Peer A, Procopio G, Takagi T, Wong S, Bedke J, Schmidinger M, Rodriguez-Lopez K, Burgents J, He C, Okpara CE, McKenzie J, Choueiri TK; CLEAR Trial Investigators. Lenvatinib plus pembrolizumab versus sunitinib in first-line treatment of advanced renal cell carcinoma: final prespecified overall survival analysis of CLEAR, a phase III study. J Clin Oncol. 2024;42:1222-1228.
62
Choueiri TK, Eto M, Motzer R, De Giorgi U, Buchler T, Basappa NS, Méndez-Vidal MJ, Tjulandin S, Hoon Park S, Melichar B, Hutson T, Alemany C, McGregor B, Powles T, Grünwald V, Alekseev B, Rha SY, Kopyltsov E, Kapoor A, Alonso Gordoa T, Goh JC, Staehler M, Merchan JR, Xie R, Perini RF, Mody K, McKenzie J, Porta CG. Lenvatinib plus pembrolizumab versus sunitinib as first-line treatment of patients with advanced renal cell carcinoma (CLEAR): extended follow-up from the phase 3, randomised, open-label study. Lancet Oncol. 2023;24:228-238.