Next Article in Journal
The Effect of Exercise on Reducing Lymphedema Severity in Breast Cancer Survivors
Next Article in Special Issue
Treatment Modalities for Non-Muscle Invasive Bladder Cancer: An Updated Review
Previous Article in Journal
Improved Metastatic-Free Survival after Systematic Re-Excision Following Complete Macroscopic Unplanned Excision of Limb or Trunk Soft Tissue Sarcoma
Previous Article in Special Issue
PD-L1 Expression in High-Risk Non-Muscle-Invasive Bladder Cancer Is Influenced by Intravesical Bacillus Calmette–Guérin (BCG) Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Revolutionizing Treatment: Breakthrough Approaches for BCG-Unresponsive Non-Muscle-Invasive Bladder Cancer

by
Maciej Jaromin
,
Tomasz Konecki
and
Piotr Kutwin
*
1st Department of Urology, Medical University of Lodz, 93-513 Lodz, Poland
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(7), 1366; https://doi.org/10.3390/cancers16071366
Submission received: 1 March 2024 / Revised: 20 March 2024 / Accepted: 26 March 2024 / Published: 30 March 2024

Abstract

:

Simple Summary

Bladder cancer is a common disease in urological patients. The approach to treatment depends on the severity of the tumor; in this article, we focus on tumors that do not invade the muscle layer of the bladder. Those tumors are resected during an endoscopic procedure (TURBT), but often reoccur. Treating bladder cancer with drugs instead of surgical removal of the bladder (radical cystectomy) is paramount for patients quality of life and overall well-being. The aim of this paper is to review methods of conservative treatment of tumors unresponsive to the typical treatment of choice (BCG instillations).

Abstract

Bladder cancer is the 10th most popular cancer in the world, and non-muscle-invasive bladder cancer (NMIBC) is diagnosed in ~80% of all cases. Treatments for NMIBC include transurethral resection of the bladder tumor (TURBT) and intravesical instillations of Bacillus Calmette-Guérin (BCG). Treatment of BCG-unresponsive tumors is scarce and usually leads to Radical Cystectomy. In this paper, we review recent advancements in conservative treatment of BCG-unresponsive tumors. The main focus of the paper is FDA-approved medications: Pembrolizumab and Nadofaragene Firadenovec (Adstiladrin). Other, less researched therapeutic possibilities are also included, namely: N-803 immunotherapy, TAR-200 and TAR-210 intravesical delivery systems and combined Cabazitaxel, Gemcitabine and Cisplatin chemotherapy. Conservative treatment and delaying radical cystectomy would greatly benefit patients’ quality of life; it is undoubtedly the future of BCG-unresponsive NMIBC.

1. Introduction

According to data from the International Agency for Research on Cancer, in 2020, bladder cancer was ranked 10th most popular cancer in the world—6th in the male population and 15th in the female population. In urology, the bladder is the second most popular location for cancer (after the prostate), accounting for 19% of all diagnosed cancers [1]. It is much more prevalent in men, with only ~20% of overall cases diagnosed in the female population [2]. The most common type of neoplasm in the bladder is Urothelial Carcinoma, which accounts for 90% of all tested specimens [3]. Histopathologically, the most common type is non-muscle-invasive bladder cancer (NMIBC), which contributes to 80–75% of all newly diagnosed cases when detrusor muscle is present in a specimen acquired through TURBT [4].
NMIBC types (according to TNM classification) include Ta, T1 and carcinoma in situ (CIS), regardless of WHO histopathological classification (LG/HG). T2, T3 or T4 tumors are muscle-invasive and are treated differently. According to EAU guidelines, the preferred treatment of NMIBC is a complete resection of the tumor by Transurethral Resection of Bladder Tumor (TURBT) with a mandatory histopathological evaluation of the tumor specimen. A second TURBT, or re-TURBT, is recommended after an incomplete first TURBT if there is no detrusor muscle in the specimen and in all T1 tumors. Non-muscle-invasive bladder cancers have a high recurrence rate, estimated at 30–50% after initial TURBT [5].
After histopathological evaluation and full tumor resection, patients should remain under surveillance and should have proper follow-up treatment, depending on their histopathological diagnosis. EAU guidelines include various disease management methods, such as Office Fulguration and Active Surveillance (both in Ta LG/G1 tumors), Adjuvant Intravesical Chemotherapy or Radical Cystectomy. Nevertheless, the golden standard for post-TURBT treatment of NMIBC are Bacillus Calmette-Guérin (BCG) intravesical instillations.

2. Intravesical BCG Therapy

BCG therapy in urology has an impressive history—the protocol for BCG intravesical installations created in 1972 by Alvaro Morales is used to this date [6,7]. It is proven that combined therapy is superior to TURBT alone as well as TURBT with intravesical chemotherapy, making TURBT with BCG follow-up the golden standard in NMIBC treatment [8,9,10]. BCG instillations are aimed at stopping recurrences or progression of NMIBC after TURBT, mainly in patients with intermediate- and high-risk tumors as well as CIS tumors. For patients with low-risk tumors, BCG therapy is not recommended due to the very low risk of disease progression; office fulguration is a good alternative [11].
Optimal BCG therapy consists of two periods: induction and maintenance. Induction is divided into six weekly BCG intravesical instillations and then three weekly instillations at 3, 6 and 12 months (each installation usually consists of 81 mg of BCG). Other scheduling variants were tested and proved less effective [12,13]. A one-year installation regiment is recommended for patients with intermediate-risk tumors, while for high-risk tumors maintenance may be prolonged to 3 years. Patients included in the very high-risk group are usually offered immediate radical cystectomy, but BCG therapy is recommended if they are unfit or do not consent to the surgical procedure.
EAU defines BCG failure as the detection of any HG tumor during or after BCG therapy, although the definition is not entirely clear and is used differently in various studies. The failure rate of BCG therapy is estimated to be around 40–50% [14,15]. In cases of unresponsive tumors, the method of choice for further treatment is radical cystectomy. In cases of late relapses (>6 months for T1/Ta HG and >12 months for CIS) and LG/G1-G2 tumors, individual situations should be considered, and usually the BCG treatment is repeated in a 1 to 3 year schedule as presented above. Nonetheless, any recurrences during follow-up visits qualify for radical cystectomy.
There are a few predictors of possible failure of BCG therapy, but most of them are known in patients with previous NMIBC history or after histopathological grading. The neutrophil to lymphocyte (NtL) ratio (measured preoperatively from peripheral blood) is a simple metric that has been proven to be associated with recurrence and progression risks. Elevated NtL prognoses a worse recurrence-free rate and progression-free rate after 5 years of initial TURBT [16,17]. It is not entirely clear what the NtL threshold for unfavorable prognosis should be: both NtL > 2.2 and >2.5 were proved to be associated with worse BCG therapy outcomes [18,19].
The tumor microenvironment is a major prognostic factor for BCG response. BCG instillations generate a local immune response by binding to the fibronectin attachment protein (FAP) of the urothelium, activating macrophages and neutrophils through Toll-like receptors (TLR). Activated macrophages promote the secretion of nitric oxide and further recruitment of immune response cells by cytokine signaling pathways. Neutrophils secrete TNF-related apoptosis-inducing ligand (TRAIL), which shows anti-tumor properties; responders to BCG therapy have higher TRAIL urine levels than non-responders [20,21]. High levels of cytotoxic CD8+ T cells, especially their intratumoral presence, predict a good anti-tumor response for BCG and immune checkpoint inhibition. Myeloid-derived suppressor cells in the tumor microenvironment inhibit the proliferation of CD8+ T cells; therefore, their low levels are beneficial to BCG therapy outcomes [22]. The stimulator of interferon genes (STING) protein creates an immunostimulatory environment by recruiting M1 macrophages, effector T cells and T-helper type-1 cells. cGAS-STING is another immune response pathway activated in the tumor microenvironment by BCG. Tumors responding to BCG therapy have higher STING protein expression at baseline, further increased by BCG instillations. In higher-grade tumors, expression of STING protein was decreasing (TaLG—22%, TaHG—15%, T1HG—6.5%), implicating tumor resistance to immune response [23].
BCG therapy may cause adverse effects, either local or systemic. Local side effects include cystitis, epididymo-orchitis, symptomatic granulomatous prostatitis and hematuria. Systemic side effects include arthritis, fever > 38.5 °C for >48 h, BCG sepsis or allergic reactions. Interestingly, an EORTC genito-urinary cancer group study of 1355 patients with a median follow-up time of 7.1 years did not show increased toxicity of ⅓ dose compared to full dose but showed that reduced dose produces suboptimal results [24]. Regarding side effects, the same study reported that 62.8% of patients presented local side effects and 30.6% presented systemic side effects—nonetheless, only 7.8% of patients stopped the BCG therapy due to side effects [25]. Another trial, in which 487 patients received BCG therapy, reported that 20% of patients abandoned the treatment due to side effects [26]. Quinolones are proven to be effective at mitigating side effects after BCG instillations, but they may cause damaging and potentially permanent side effects. The use of quinolones in BSC side effects is still being debated and is not a standard procedure [27].

3. Radical Cystectomy

NMIBC progression to Muscle Invasive Bladder Cancer varies depending on the tumor’s risk group. In very high groups, percentage rates are very high: 1 year’s progression probability is 16%, 5 years—40% and 10 years—53% (according to WHO 2004/2016 risk groups) [28]. Often, T1 tumors are upstaged to muscle-invasive after Radical Cystectomy—a retrospective, multicenter study of 64,675 patients showed a median upstaging rate of 49.7% [29]. Therefore, after diagnosis of a very high-risk tumor, EAU guidelines recommend immediate Radical Cystectomy, and suggest discussing immediate Radical Cystectomy in high-risk group patients. Oncologically, Radical Cystectomy is a very effective procedure, but it comes with the risk of many adverse effects and, in some cases, might be an overtreatment.
A meta-analysis of adverse effects after Radical Cystectomy concluded that the most common are gastrointestinal complications (20%), infectious complications (17%), ileus complications (14%), cardiovascular complications (9%) and respiratory complications (7%). Similar results were reported in a systematic review [30]. Results of a cohort study reporting on 90-day complications post-Radical Cystectomy showed 24.3% of patients exhibited major (Clavien–Dindo grade 1 or 2) complications, and 53.9% exhibited minor (Clavien–Dindo grade 3, 4 or 5) complications. The re-admission rate was 29.6%, and ER visits were 37.9%, both mostly due to infections. Ninety-day mortality was assessed at 2.1% for 30-day mortality and 4.7% for 90-day mortality [31,32]. The overall survival (OS) and cancer-specific survival (CSS) rates vary depending on the examined population and experience of the institution, but they oscillate between 60–30% for OS and 70–40% for CSS [33,34,35].
Radical Cystectomy greatly influences QoL. A prospective study on a group of 842 patients concluded that 1 year after radical cystectomy, 43.1% of patients suffered from high psychological distress (measured by the Questionnaire on Stress in Cancer Patients). Patients also report lowered QoL due to body image, physical function, constipation, diarrhea, financial situation and many more; specific results vary depending on the method of bladder reconstruction. During 12 months of observation after radical cystectomy, patients’ overall QoL improved, which may indicate further progress after this period [36]. In a multinational experiment, an online questionnaire was submitted to 107 patients with NMIBC, showing hypothetical scenarios for disease management. The goal of the study was to analyze attitudes toward radical cystectomy. Results show that patients would accept an increased risk of progression to MIBC and serious side effects (43.8% and 66.1%, respectively) for delaying radical cystectomy for 5 years.
Although such experiments have many limitations, these data clearly shows patients unwillingness to undergo Radical Cystectomy and is aligned with the clinical experience of many physicians. The possible side effects and stress connected with radical cystectomy lead patients to refuse the operation; far more Radical Cystectomies are canceled due to patients lack of consent than medical contraindications. Even if the operation is unavoidable, most patients will choose some form of conservative treatment to delay the procedure, which should improve their QoL and delay complications related to Radical Cystectomy.

4. New Treatment Options

Considering NMIBC treatment, there has been a void between BCG instillations and radical cystectomy, effectively leaving patients with BCG-unresponsive tumors with only one therapeutic option. In recent years, many new approaches and substances have been developed and studied in efforts to enrich standard NMIBC treatment with new non-invasive options. All clinical trials included in this article are listed in Table 1.

5. Intravesical Therapies

5.1. N-803

N-803, also known as ALT-803, is an immunomodulating drug designed for intravesical use. Pharmacokinetically, it is a superagonist of IL-15 receptors—most importantly in CD8+ T and NK cells, promoting their proliferation and activation without broad activation of other proinflammatory pathways [37,38]. Results from a recent clinical trial performed on 160 BCG-unresponsive participants (83 with CIS and 77 with papillary tumors) show great synergistic effects of N-803 and BCG, with a 71% complete response rate for CIS patients in 24.1 months median complete response time. Patients with papillary tumors had a 48% disease-free rate after 24 months. More than 90% of the patients avoided cystectomy two years after BCG + N-803 combined therapy, and none of the patients had any serious adverse effects [39]. Further clinical trials of the BCG + N-803 combination are estimated to end in December 2027 and October 2028.

5.2. Nadofaragene Firadenovec

Nadofaragene firadenovec (or Adstiladrin) is a gene therapy based on non-replicating viral DNA vectors encoding interferon-α2b, promoting the production of interferon-α2b in tumor cells and effectively resulting in apoptosis. Other anti-tumor mechanisms include inducing MHC-I expression, which promotes T cell activity against tumor cells. Adstiladrin is administered intravesically; results from clinical trials (NCT01687244) suggest a recommended dosage of 3 × 1011 viral particles/mL in 75 mL, instilled every 12 weeks [40,41]. Some studies look at factors that predict Adstiladrin response durability. The type I Interferon signaling pathway, cytotoxic response to Interferon-α and transcriptional response might be reliable predictors of tumor resistance to Adstiladrin. After administration of Adstiladrin, anti-adenovirus antibody levels may predict the success of further treatment: a titer >800 was observed in 47% of responders compared to 18% of non-responders; it was also associated with a higher probability of a durable response [42,43].
Adverse effects rates of Adstiladrin are comparable to Pembrolizumab: in a phase 3 clinical trial (NCT02773849), 157 patients were treated with BCG-unresponsive NMIBC. Adverse effects were observed in 66% of patients; grade 3 in 4% of patients; no grade 4 or 5 or death connected to treatment was reported; 3 patients discontinued due to adverse effects [44]. Similarly, in the previously mentioned clinical trial, there were no reports of adverse effects graded 4.5 or treatment-related death; none of the participants withdrew from the trial. Both trials concluded that Adstiladrin is well-tolerated and safe to use in the treatment of NMIBC [41].
In clinical trial NCT01687244, 40 patients were divided into two groups: group A received 1 × 1011 viral particles/mL every 3 months for 1 year, and group B received 3 × 1011 viral particles/mL every 3 months for 1 year. Instillations were administered only to patients with no HG recurrence. Combining results from both groups, 17 patients (42.5%) had no HG recurrence at 9 months (after 3 doses); 14 patients (35%) had no recurrence after 12 months. No patient underwent a cystectomy during the 12-month trial, and the overall survival rate was 100%.
The clinical trial NCT02773849 had a total of 157 patients: group A, including 107 patients with CIS (with or without Ta or T1 tumors), and group B, including 50 participants with Ta HG or T1 without CIS. The dose and regimen of instillations were 3 × 1011 viral particles/mL every 3 months for 1 year. In group A, a complete response was achieved by 53.4% and 24.2% of participants after 3 months and 12 months, respectively. In group B, the median recurrence-free survival was 12.35 months. After 24 months, overall survival was 94.4% in group A, and 93.2% in group B; cystectomy-free survival was 64.6% in group A and 69.8% in group B [44,45,46].
Intravesical instillations are a safe option and are usually easy to perform in an office environment. Most importantly, the safety of the aforementioned drugs is comparable to BCG instillations, which are much safer and better tolerated than Radical Cystectomy. Combining new pharmaceuticals with BCG instillations seems to be a very tempting option, enhancing a proven and well-established therapy with tumor-targeting drugs. It is also important to include new diagnostic methods for tumor microenvironment, NtL ratio or genetic profile of tumors to implement the best possible therapy and regiments. Gathering data will help in recognizing best practices and best combinations of drugs, as well as optimal schedules. Whether monotherapy of BCG instillations should remain the gold standard after first TURBT is up for discussion, but intravesical therapy for BCG-unresponsive tumors is definitely the future of NMIBC treatment.

6. Intravenous Therapies

6.1. Cabazitaxel, Gemcitabine and Cisplatin

Cabazitaxel, Gemcitabine and Cisplatin (CGC) are well-known chemotherapeutics used in the treatment of many different neoplasms. Cabazitaxel is mostly used in prostate cancer, while Gemcitabine and Cisplatin are implemented in advanced and metastatic bladder cancer [47,48]. However, combining those three in the intravesical therapy of BCG-unresponsive NMIBC is a novel idea, and only a few clinical trials have been made. One trial was based on a method of sequential administrations of CGC weekly during the induction phase and later monthly or bimonthly during maintenance. During Phase I, 18 patients ineligible for radical cystectomy were enrolled and treated with the aforementioned protocol; the recurrence-free rate at 12 months was 83% and at 24 months, 64%, but for patients receiving the maximum CGC dose (10 out of 18), the recurrence-free rate was 100% and 83% at 12 and 24 months, respectively. No dose-limiting toxicities or adverse effects were observed. During Phase II, 31 patients were enrolled; the completion of this trial is expected to be in December 2024 [49,50]. Another study retrospectively reviewed 107 patients who were receiving Gemtabicine and Docetaxel (with similar induction + maintenance protocols) for 2 years. Results show a recurrence-free rate of 85% and 82% at 12 and 24 months, respectively, and no patient had tumor progression or died of bladder cancer [51]. Data for conjoined intravesical chemotherapy of NMIBC are still scarce, but more studies and results may shed light on a new approach to BCG-unresponsive tumor treatment.

6.2. Pembrolizumab

Programmed cell death 1 (PD-1) protein is a receptor present in T and B cells, activated by its ligand PD-L1. Its main function is to down-regulate the immune response to humans’ own cells, effectively preventing autoimmune reactions. Stimulating the PD-1/PD-L1 pathway is used by cancer cells to avoid immune response, allowing the progression of tumors [52]. BCG is a nonspecific immunotherapeutic, and combining it with an “immune checkpoint” inhibitor may give effective results even in BCG monotherapy-unresponsive tumors, especially if PD-L1 is present in tumor cells [53].
BCG therapy upregulates PD-L1 expression in tumors in vitro and in rat models, possibly due to tumor infiltration of cytotoxic T-cells during the local immune response. This results in impaired T-cell response through the PD-1/PD-L1 pathway, enabling a synergistic effect with PD-1 checkpoint inhibitors. Combining BCG with anti-PD1 antibodies results in increased T-cell activity; both serum INF-y levels and Granzyme B expression in tumor cells were higher after combined therapy compared to monotherapy and the control group. A reduction of myeloid-derived suppressor cells was also observed [54].
Pembrolizumab is a monoclonal IgG4 anti-PD1 antibody administered intravenously. It is important to note that BCG treatment does not impair the effects of Pembrolizumab. A study on 755 patients with Urothelial Carcinoma (20.5% with previous BCG therapy) concluded that previous BCG therapy did not influence overall survival, disease control rate or objective response rate [55].
There are many ongoing trials testing PD-1 inhibitors: Atezolizumab (NCT03799835, NCT04134000, NCT02792192), Durvalumab (NCT03528694, NCT03759496, NCT04106115), Nivolumab (NCT04149574) Sansanlimab (NCT04165317), and more. Soon we may see PD-1 inhibitors as a staple in conservative treatment of NMIBC. Regarding NMIBC, Pembrolizumab is the only drug accepted by the FDA for the treatment of NMIBC and is going to be the main focus of this whole group. To this date, it has been successfully used in melanoma, non-small cell lung cancer and many more neoplasms.
The KEYNOTE-001 clinical trial enrolled 655 patients with advanced or metastatic melanoma; patients received 2 mg/kg every 3 weeks, 10 mg/kg every 3 weeks, or 10 mg/kg every 2 weeks for at least 6 months in case of a complete response. Treatment was discontinued in cases of patients’ decision to withdraw, intolerable toxicity or disease progression. Treatment-related adverse effects were present in 86% of all patients, but only 17% were grade 3–4, and none experienced treatment-related death [56]. A study on Pembrolizumab effects on non-small cell lung cancer reported a 70.9% rate of all adverse effects in a group of 495 patients. The most common included fatigue, rash, decreased appetite and pruritus. In this group, there were only 9.4% of adverse effects above grade 3 [57]. A meta-analysis of the adverse effects of PD-1 inhibitors (pembrolizumab and nivolumab) on 9136 total patients showed only 33 deaths, the leading cause being pneumonitis [58].
The effects of Pembrolizumab on NMIBC are reported in KEYNOTE-057, a phase 2 single-arm clinical trial. A total of 101 patients from 54 facilities with BCG-unresponsive CIS (63.4% had CIS only, 24.9% had CIS + Ta/HG, 11.9% had CIS + T1) were recruited to Cohort A; patients with no CIS but TaT1 tumors were included in Cohort B. All patients were ineligible or refused RC; it is worth mentioning that the number of patients refusing RC was 95%. Participants were administered 200 mg of intravenous Pembrolizumab every 3 weeks for 2 years. Discontinuation of treatment was caused by tumor resistance, recurrence or progression, toxic effects or patients’ decision to withdraw. The median follow-up was 36.4 months; the median number of Pembrolizumab administrations was 7; and the median time of therapy was 4.2 months.
At 3 months, a complete response was observed in 40.6% of patients; for the CIS-only group, 45%, 0%; for the CIS + Ta/HG group, 29.2%; and for the CIS + T1 group, 41.7%. Progression was reported in 9.4% of patients, and stable disease was reported in 47.9%.
Adverse effects were observed in 61% of the patients; 21% of patients had immune-related adverse effects; and 13% of patients experienced adverse effects grade 3 or above, which is consistent with the adverse effects rate reported in other studies [56,57,58]. There were no treatment-related deaths; 6.9% of patients discontinued the treatment due to adverse effects [59,60]. KEYNOTE-676 is an ongoing, phase 3 clinical trial studying the effects of combining Pembrolizumab and BCG in BCG-naive tumors and BCG-unresponsive high-grade tumors. The study is expected to finish in 2028 [61].
Intravesical instillations of Prembolizumab were tested in two clinical trials. A phase-1 clinical trial (NCT03167151) aimed at studying the safety of intravesical administrations of Prembolizumab, dividing 6 patients into three groups (n = 2). Each group received weekly instillations for a total of 6 weeks; each dose was administered in 40 mL infusions retained for 2 h in the bladder. Group 1 started with 50 mg of Prembolizumab in weeks 1 and 2, then proceeded to 100 mg in weeks 3 and 4, and then 200 mg in weeks 5 and 6; group 2 started with 100 mg in weeks 1 and 2 and then 200 mg for weeks 3, 4, 5 and 6; group 3 had a constant dose of 200 mg throughout 6 weeks. During the course of the study, 21 adverse effects were reported, 14 of which were treatment-related. The study does not report any grade 3 or higher adverse effects connected to treatment. Unfortunately, due to delays in recruitment phase 2, the study was abandoned, giving no information on the active intravesical dose of pembrolizumab [62].
The second study inspected the effects of intravesical instillation of Pembrolizumab combined with BCG in nine BCG-unresponsive tumor patients (NCT02808143). The schedule included a pre-induction pembrolizumab instillation (1–2 mg/kg) 2 weeks before the induction phase. During the 5-week induction phase, patients received weekly BCG instillations, combined with Pembrolizumab instillations in weeks 0.2 and 4. After induction, patients received Pembrolizumab instillations every 2 weeks for 8 weeks (4 doses in total), and later Pembrolizumab instillations every 4 weeks for 32 weeks (8 doses in total). There were 22 reported adverse effects, 21 grade 1 or 2 (with 1 incident of serious grade 2 adverse effect) and one grade 5: myasthenia gravis, which resulted in the patient’s death. Overall, only five patients were alive at the end of the study. Among them, the median time of progression-free survival was 35 months. All patients had tumor recurrence, with only one patient having recurrence-free survival for more than a year; progression to muscle-invasive bladder cancer occurred in six out of nine patients [63].
Similarly to intravesical pharmaceuticals, intravenous therapy is a promising tool in NMIBC treatment. Intravenous chemotherapy is a hallmark choice in oncological therapies for many tumors and cancers, including muscle-invasive bladder cancer. In NMIBC, it is not a common choice due to intravesical chemotherapy being less effective in conservative treatment than BCG and less effective than Radical Cystectomy in BCG-unresponsive treatment. Intravenous chemotherapy may seem like an aggressive option for NMIBC, but studies show very good results in recurrence-free and progression-free rates. On the other hand, supporting BCG instillations with “checkpoint inhibitor” immunotherapy seems like a very logical follow-up in patients unresponsive to BCG monotherapy. Intravenous chemotherapy and immunotherapy are likely to become the link between BCG instillations and Radical Cystectomy; similarly to intravesical therapies, more studies and trials are required to establish best practices and regiments.

7. Intravesical Delivery Systems

TAR 200 is an intravesical delivery system, releasing gemcitabine in a span of over 2 weeks. In a clinical trial, SunRISe-1 (NCT04640623), TAR 200 alone, TAR 200 + systemic Cetrelimab and systemic Cetrelimab alone were evaluated in BCG-unresponsive NMIBC. The total number of participants is estimated to be 200; the total number of TAR-200 doses is 16. The main objective of the study is the evaluation of the complete response rate and disease-free survival up to 5 years. The estimated completion date is July 2027.
A similar trial (NCT05316155) is using an Intravesical Delivery System to study the proper dosage and adverse effects of Erdafitinib, as well as plasma and urine concentration, recurrence-free survival and complete response. A total of 92 patients are divided into five cohorts, each measured for different outcomes. The study is estimated to finish in June 2027.
Erdafinib is a Fibroblast Growth Factor receptor (FGFR) inhibitor, approved on 12 April 2019 by the FDA for patients with locally advanced or metastatic urothelial carcinoma. The FGFR pathway plays an important role in the apoptosis of some cancer cells; overexpression of FGFR is reported in breast cancer, urothelial carcinomas, melanoma, gastric malignancies and more. FGFR inhibitors are directly impacting the FGF/FGFR pathway but also influence the tumor microenvironment indirectly by altering angiogenesis via VEGF inhibition and local immune responses [64]. In muscle-invasive bladder cancer, FGFR1 mRNA was shown to be a predictor of neoadjuvant cisplatin therapy results. Low FGFR1 mRNA levels were correlated with a higher pathological complete response in tumor cells, measured pre- and post-radical cystectomy. The clinical trial NCT02792192 reported 3% complete response, 37% partial response and 39% stable disease (median progression-free survival of 5.5 months). Notably, FGFR2 and FGFR3 gene mutations are especially relevant; mutations FGFR2N549H, FGFR3Y373C;V555M, increase tumor immunity to FGFR-inhibitors [65,66].
Intravesical delivery systems are a novel idea aimed at the slow and constant release of a therapeutic. In theory, a steady influx of drugs may prove crucial in restraining tumor growth and obtaining the maximum effects of administered substances. In addition, patients are exposed to much smaller doses compared to typical intravesical or intravenous therapy, which consequently may limit adverse effects and patient discomfort related to single-dose admission. The results of the above trials are of paramount importance to the future of NMIBC treatment and we will be monitoring them very closely.

8. Radiation Therapy

Radiation therapy is commonly used to treat muscle-invasive bladder cancer but is not recommended in NMIBC therapy. A meta-analysis of two clinical trials established that a dose of 55 Gy in 20 fractions should be the standard radiation therapy for locally invasive bladder cancer. Unfortunately, 751 of the 782 recruited patients had muscle-invasive bladder cancer; no information on the isolated NMIBC group is provided [67]. The Chinese trial ChiCTR2200059970 aims at examining the effects of short-course radiotherapy (5 × 5 Gy over 5 or 10 days) combined with a 12-dose intravenous cycle of the PD-1 antibody Toripalimab. The planned patient population is 55 with high/extremely high-risk NMIBC, with or without previous BCG instillation courses. Trial NCT00981656 featured 34 patients with stage I NMIBC; treatment included concurrent three-dimensional conformal radiation therapy (3DCRT) and radiosensitizing chemotherapy (CT) of cisplatin or a combination of mitomycin with 5-fluorouracil. Primary outcome, the 3-year radical cystectomy free rate, was reported to be at 88.2%, but the percentage of alive patients after 3 years was 69.2%, and no data were published on progression to stage 2 or higher. Data on radiation therapy in NMIBC are very scarce and cluttered; more standardized trials and studies are needed.

9. Discussion

BCG therapy failure is a turning point in the therapy of NMIBC, leaving patients with few options for conservative treatment. For many years, different methods and ideas were developed, but none proved to be reliable—especially in BCG-unresponsive tumors. Intravesical chemotherapy instillations (either single or recurrent) after TURBT were discussed for years but proved less effective than BCG instillations and not beneficial when combined with BCG instillations [68,69]. Ideas for implementing chemohyperthermia have also been studied, but are not yet proven to be effective and adverse effects may be a limiting factor [70].
Many studies report the tumor microenvironment as an important factor in NMIBC therapy. Local immune response, up- and downregulation of PD1/PDL1, VEGF, FGF/FGFR, cGAS-STING signaling pathways and specific gene mutations all contribute to tumor response to particular treatments. Profiling each tumor by its genetic and immunological properties may enable personalized therapy composed of the most efficient pharmaceuticals and delivery methods for each patient. Tumor microenvironment profiling may prove to be as important as histopathological grading for bladder cancer therapy outcomes; more studies and clinical trials are needed.
Therapies presented in this article may seem groundbreaking and revolutionary, but most of them are either still in the phase of clinical trials or very young and not entirely researched. Other limitations include non-standardized outcome measurements, various inclusion/exclusion criteria (e.g., BCG-failure or BCG-unresponsive tumor definitions), a generally small number of patients in trial groups, and heterogeneity in trials of different pharmaceuticals. Safety of patients is most definitely paramount; nonetheless, treatments described in this article usually passed phase 1 of clinical trials well, and none reported alarming toxicity or abnormal amounts of adverse effects. Allowing patients to decide about risks and trade-offs in their therapy is a hallmark of modern medicine; in NMIBC, the decision between conservative and radical treatment seems to be very personal and hard to navigate. Concerns about the effectiveness and safety of novel therapies should be resolved as soon as possible with high-quality studies and trials; in our opinion, both FDA-approved therapeutics (Adstiladrin and Pembrolizumab) are worth mentioning to patients with BCG-unresponsive NMIBC (Table 2).

10. Conclusions

NMIBC treatment is very important in urological oncology since progression to a muscle-invasive tumor results in potential life-threatening complications and quick disease development. Searching for balance between well-tolerated, approachable treatment and satisfying oncological results is the focal point, where new pharmaceuticals and methods should enrich the group of possible choices. For many patients, quality of life and tolerability of the treatment may be as important as clear-cut medical results, leaving the discussion in the realm of percentages, risks and compromises.
A secondary, but ever-important, issue is cost effectiveness. A cohort simulation showed positive results of cost effectiveness with a quality-adjusted life-year ratio [71], but it is far from obvious in different healthcare systems and populations. The price of a single Pebmrolizumab’s dose (lasting 3 weeks) is over $11,000, while one year of Adstiladrin treatment costs $240,000. For most people, such expenses are far beyond their reach, so it is up to insurance companies and healthcare systems to develop a reasonable solution.

Author Contributions

Conceptualization, M.J. and T.K. data curation, P.K.; writing—original draft preparation, M.J.; writing—review and editing, P.K. and M.J.; supervision, T.K.; project administration, P.K. and M.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Centers for Disease Control and Prevention. Male Urologic Cancers; USCS Data Brief, no 21; Centers for Disease Control and Prevention, US Department of Health and Human Services: Atlanta, GA, USA, 2020. [Google Scholar]
  2. Cook, M.B.; Dawsey, S.M.; Freedman, N.D.; Inskip, P.D.; Wichner, S.M.; Quraishi, S.M.; Devesa, S.S.; McGlynn, K.A. Sex disparities in cancer incidence by period and age. Cancer Epidemiol. Biomark. Prev. 2009, 18, 1174–1182. [Google Scholar] [CrossRef]
  3. Kaseb, H.; Aeddula, N.R. Bladder Cancer. [Updated 2022 October 24]. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. Available online: https://www.ncbi.nlm.nih.gov/books/NBK536923/ (accessed on 2 February 2024).
  4. van Straten, C.G.J.I.; Bruins, M.H.; Dijkstra, S.; Cornel, E.B.; Kortleve, M.D.H.; de Vocht, T.F.; Kiemeney, L.A.L.M.; van der Heijden, A.G. The accuracy of cystoscopy in predicting muscle invasion in newly diagnosed bladder cancer patients. World J. Urol. 2023, 41, 1829–1835. [Google Scholar] [CrossRef] [PubMed]
  5. Tokuyama, N.; Saito, A.; Muraoka, R.; Matsubara, S.; Hashimoto, T.; Satake, N.; Matsubayashi, J.; Nagao, T.; Mirza, A.H.; Graf, H.P.; et al. Prediction of non-muscle invasive bladder cancer recurrence using machine learning of quantitative nuclear features. Mod. Pathol. 2022, 35, 533–538. [Google Scholar] [CrossRef] [PubMed]
  6. Lobo, N.; Brooks, N.A.; Zlotta, A.R.; Cirillo, J.D.; Boorjian, S.; Black, P.C.; Meeks, J.J.; Bivalacqua, T.J.; Gontero, P.; Steinberg, G.D.; et al. 100 years of Bacillus Calmette–Guérin immunotherapy: From cattle to COVID-19. Nat. Rev. Urol. 2021, 18, 611–622. [Google Scholar] [CrossRef] [PubMed]
  7. Morales, A.; Eidinger, D.; Bruce, A.W. Intracavitary Bacillus Calmette-Guerin in the treatment of superficial bladder tumors. J. Urol. 1976, 116, 180–183. [Google Scholar] [CrossRef] [PubMed]
  8. Sylvester, R.J.; Brausi, M.A.; Kirkels, W.J.; Hoeltl, W.; Da Silva, F.C.; Powell, P.H.; Prescott, S.; Kirkali, Z.; van de Beek, C.; Gorlia, T.; et al. Long-Term Efficacy Results of EORTC Genito-Urinary Group Randomized Phase 3 Study 30911 Comparing Intravesical Instillations of Epirubicin, Bacillus Calmette-Guérin, and Bacillus Calmette-Guérin plus Isoniazid in Patients with Intermediate- and High-Risk Stage Ta T1 Urothelial Carcinoma of the Bladder. Eur. Urol. 2010, 57, 766–773. [Google Scholar] [CrossRef] [PubMed]
  9. Thiel, T.; Ryk, C.; Renström-Koskela, L.; Steineck, G.; Schumacher, M.C.; Wiklund, N.P.; de Verdier, P.J. Intravesical BCG treatment causes a long-lasting reduction of recurrence and progression in patients with high-risk non-muscle-invasive bladder cancer. World J. Urol. 2019, 37, 155–163. [Google Scholar] [CrossRef] [PubMed]
  10. Musat, M.G.; Kwon, C.S.; Masters, E.; Sikirica, S.; Pijush, D.B.; Forsythe, A. Treatment Outcomes of High-Risk Non-Muscle Invasive Bladder Cancer (HR-NMIBC) in Real-World Evidence (RWE) Studies: Systematic Literature Review (SLR). Clin. Outcomes Res. 2022, 14, 35–48. [Google Scholar] [CrossRef] [PubMed]
  11. Donat, S.M.; North, A.; Dalbagni, G.; Herr, H.W. Efficacy of office fulguration for recurrent low grade papillary bladder tumors less than 0.5 cm. J. Urol. 2004, 171 Pt 1, 636–639. [Google Scholar] [CrossRef]
  12. Grimm, M.O.; van der Heijden, A.G.; Colombel, M.; Muilwijk, T.; Martínez-Piñeiro, L.; Babjuk, M.M.; Türkeri, L.N.; Palou, J.; Patel, A.; Bjartell, A.S.; et al. Treatment of High-grade Non-muscle-invasive Bladder Carcinoma by Standard Number and Dose of BCG Instillations Versus Reduced Number and Standard Dose of BCG Instillations: Results of the European Association of Urology Research Foundation Randomised Phase III Clinical Trial “NIMBUS”. Eur. Urol. 2020, 78, 690–698. [Google Scholar] [CrossRef]
  13. Kamat, A.M.; Flaig, T.W.; Grossman, H.B.; Konety, B.; Lamm, D.; O’donnell, M.A.; Uchio, E.; Efstathiou, J.A.; Taylor, J.A., III. Consensus statement on best practice management regarding the use of intravesical immunotherapy with BCG for bladder cancer. Nat. Rev. Urol. 2015, 12, 225–235. [Google Scholar] [CrossRef] [PubMed]
  14. Gual Frau, J.; Palou, J.; Rodríguez, O.; Parada, R.; Breda, A.; Villavicencio, H. Failure of Bacillus Calmette-Guérin therapy in non-muscle-invasive bladder cancer: Definition and treatment options. Arch. Esp. Urol. 2016, 69, 423–433. [Google Scholar]
  15. Zlotta, A.R.; Fleshner, N.E.; Jewett, M.A. The management of BCG failure in non-muscle-invasive bladder cancer: An update. Can. Urol. Assoc. J. 2009, 3 (Suppl. S4), S199–S205. [Google Scholar] [CrossRef]
  16. Vartolomei, M.D.; Porav-Hodade, D.; Ferro, M.; Mathieu, R.; Abufaraj, M.; Foerster, B.; Kimura, S.; Shariat, S.F. Prognostic role of pretreatment neutrophil-to-lymphocyte ratio (NLR) in patients with non-muscle-invasive bladder cancer (NMIBC): A systematic review and meta-analysis. Urol. Oncol. 2018, 36, 389–399. [Google Scholar] [CrossRef] [PubMed]
  17. Lee, J.; Yoo, S.; Choo, M.S.; Cho, M.C.; Son, H.; Jeong, H. Prognostic Role of Preoperative Neutrophil-To-Lymphocyte Ratio (NLR) and Recurrence at First Evaluation after Bacillus Calmette–Guérin (BCG) Induction in Non-Muscle-Invasive Bladder Cancer. Diagnostics 2023, 13, 3114. [Google Scholar] [CrossRef] [PubMed]
  18. Li, D.X.; Wang, X.M.; Tang, Y.; Yang, Y.B.; Feng, D.C.; Li, A.; Zhang, F.C.; Bai, Y.J.; Han, P. Prognostic value of preoperative neutrophil-to-lymphocyte ratio in histological variants of non-muscle-invasive bladder cancer. Investig. Clin. Urol. 2021, 62, 641–649. [Google Scholar] [CrossRef] [PubMed]
  19. Ziani, I.; Ibrahimi, A.; Zaoui, Y.; Sayegh, H.E.; Abouqal, R.; Nouini, Y.; Bouziane, A. Role of preoperative neutrophil to lymphocyte ratio in prediction of recurrence, progression, and BCG failure in non-muscle invasive bladder cancer: A retrospective study. Pan Afr. Med. J. 2023, 44, 145. [Google Scholar] [CrossRef] [PubMed]
  20. Cambier, C.J.; Takaki, K.K.; Larson, R.P.; Hernandez, R.E.; Tobin, D.M.; Urdahl, K.B.; Cosma, C.L.; Ramakrishnan, L. Mycobacteria manipulate macrophage recruitment through coordinated use of membrane lipids. Nature 2014, 505, 218–222. [Google Scholar] [CrossRef] [PubMed]
  21. Kresowik, T.P.; Griffith, T.S. Bacillus Calmette-Guerin immunotherapy for urothelial carcinoma of the bladder. Immunotherapy 2009, 1, 281–288. [Google Scholar] [CrossRef]
  22. Ibrahim, O.M.; Pandey, R.K.; Chatta, G.; Kalinski, P. Role of tumor microenvironment in the efficacy of BCG therapy. Trends Res. 2020, 3. [Google Scholar] [CrossRef]
  23. Lombardo, K.A.; Obradovic, A.; Singh, A.K.; Liu, J.L.; Joice, G.; Kates, M.; Bishai, W.; McConkey, D.; Chaux, A.; Eich, M.L.; et al. BCG invokes superior STING-mediated innate immune response over radiotherapy in a carcinogen murine model of urothelial cancer. J. Pathol. 2022, 256, 223–234. [Google Scholar] [CrossRef] [PubMed]
  24. Oddens, J.; Brausi, M.; Sylvester, R.; Bono, A.; van de Beek, C.; van Andel, G.; Gontero, P.; Hoeltl, W.; Turkeri, L.; Marreaud, S.; et al. Final results of an EORTC-GU cancers group randomized study of maintenance bacillus Calmette-Guérin in intermediate- and high-risk Ta, T1 papillary carcinoma of the urinary bladder: One-third dose versus full dose and 1 year versus 3 years of maintenance. Eur. Urol. 2013, 63, 462–472. [Google Scholar] [CrossRef] [PubMed]
  25. Brausi, M.; Oddens, J.; Sylvester, R.; Bono, A.; van de Beek, C.; van Andel, G.; Gontero, P.; Turkeri, L.; Marreaud, S.; Collette, S.; et al. Side effects of Bacillus Calmette-Guérin (BCG) in the treatment of intermediate- and high-risk Ta, T1 papillary carcinoma of the bladder: Results of the EORTC genito-urinary cancers group randomised phase 3 study comparing one-third dose with full dose and 1 year with 3 years of maintenance BCG. Eur. Urol. 2014, 65, 69–76. [Google Scholar] [CrossRef]
  26. Avan der Meijden, A.P.; Sylvester, R.J.; Oosterlinck, W.; Hoeltl, W.; Bono, A.V. Maintenance Bacillus Calmette-Guerin for Ta T1 Bladder Tumors Is Not Associated with Increased Toxicity: Results from a European Organisation for Research and Treatment of Cancer Genito-Urinary Group Phase III Trial. Eur. Urol. 2003, 44, 429–434. [Google Scholar] [CrossRef] [PubMed]
  27. Liedberg, F.; Xylinas, E.; Gontero, P. Quinolone Prophylaxis in Conjunction with Bacillus Calmette-Guérin Instillations for Bladder Cancer: Time To Reconsider the Evidence and Open the Quinolone Box? Eur. Urol. Focus 2023. [Google Scholar] [CrossRef] [PubMed]
  28. Sylvester, R.J.; Rodríguez, O.; Hernández, V.; Turturica, D.; Bauerová, L.; Bruins, H.M.; Bründl, J.; van der Kwast, T.H.; Brisuda, A.; Rubio-Briones, J.; et al. European Association of Urology (EAU) Prognostic Factor Risk Groups for Non-muscle-invasive Bladder Cancer (NMIBC) Incorporating the WHO 2004/2016 and WHO 1973 Classification Systems for Grade: An Update from the EAU NMIBC Guidelines Panel. Eur. Urol. 2021, 79, 480. [Google Scholar] [CrossRef]
  29. Flitcroft, M.; Kothari, A.; Shah, N.; Knoll, K.; Johnson, S. Facility level variation in pathologic upstaging of bladder cancer after radical cystectomy. J. Urol. 2023, 209, e828. [Google Scholar] [CrossRef]
  30. Katsimperis, S.; Tzelves, L.; Tandogdu, Z.; Ta, A.; Geraghty, R.; Bellos, T.; Manolitsis, I.; Pyrgidis, N.; Schulz, G.B.; Sridhar, A.; et al. Complications After Radical Cystectomy: A Systematic Review and Meta-analysis of Randomized Controlled Trials with a Meta-regression Analysis. Eur. Urol. Focus 2023, 9, 920–929. [Google Scholar] [CrossRef] [PubMed]
  31. Djaladat, H.; Katebian, B.; Bazargani, S.T.; Miranda, G.; Cai, J.; Schuckman, A.K.; Daneshmand, S. 90-Day complication rate in patients undergoing radical cystectomy with enhanced recovery protocol: A prospective cohort study. World J. Urol. 2017, 35, 907–911. [Google Scholar] [CrossRef]
  32. Dybowski, B.; Ossoliński, K.; Ossolińska, A.; Peller, M.; Bres-Niewada, E.; Radziszewski, P. Impact of stage and comorbidities on five-year survival after radical cystectomy in Poland: Single centre experience. Cent. Eur. J. Urol. 2015, 68, 278–283. [Google Scholar] [CrossRef]
  33. Ploussard, G.; Shariat, S.F.; Dragomir, A.; Kluth, L.A.; Xylinas, E.; Masson-Lecomte, A.; Rieken, M.; Rink, M.; Matsumoto, K.; Kikuchi, E.; et al. Conditional Survival After Radical Cystectomy for Bladder Cancer: Evidence for a Patient Changing Risk Profile over Time. Eur. Urol. 2014, 66, 361–370. [Google Scholar] [CrossRef] [PubMed]
  34. Crettenand, F.; M’Baya, O.; Grilo, N.; Valerio, M.; Dartiguenave, F.; Cerantola, Y.; Roth, B.; Rouvé, J.D.; Blanc, C.; Lucca, I. ERAS® protocol improves survival after radical cystectomy: A single-center cohort study. Medicine 2022, 101, e30258. [Google Scholar] [CrossRef] [PubMed]
  35. Bahlburg, H.; Hellmann, T.; Tully, K.; Butea-Bocu, M.C.; Reike, M.; Roghmann, F.; Noldus, J.; Müller, G. Psychosocial distress and quality of life in patients after radical cystectomy—One year follow-up in 842 German patients. J. Cancer Surviv. 2023. [Google Scholar] [CrossRef]
  36. Chen, W.; Liu, N.; Yuan, Y.; Zhu, M.; Hu, X.; Hu, W.; Wang, S.; Wang, C.; Huang, B.; Xing, D. ALT-803 in the treatment of non-muscle-invasive bladder cancer: Preclinical and clinical evidence and translational potential. Front. Immunol. 2022, 13, 1040669. [Google Scholar] [CrossRef] [PubMed]
  37. Chamie, K.; Chang, S.S.; Gonzalgo, M.; Kramolowsky, E.V.; Sexton, W.J.; Bhar, P.; Reddy, S.K.; Soon-Shiong, P. Final clinical results of pivotal trial of IL-15RαFc superagonist N-803 with BCG in BCG-unresponsive CIS and papillary nonmuscle-invasive bladder cancer (NMIBC). J. Clin. Oncol. 2022, 40, 4508. [Google Scholar] [CrossRef]
  38. Rhode, P.R.; Egan, J.O.; Xu, W.; Hong, H.; Webb, G.M.; Chen, X.; Liu, B.; Zhu, X.; Wen, J.; You, L.; et al. Comparison of the Superagonist Complex, ALT-803, to IL15 as Cancer Immunotherapeutics in Animal Models. Cancer Immunol. Res. 2016, 4, 49–60. [Google Scholar] [CrossRef]
  39. Navai, N.; Benedict, W.F.; Zhang, G.; Abraham, A.; Ainslie, N.; Shah, J.B.; Grossman, H.B.; Kamat, A.M.; Dinney, C.P. Phase 1b Trial to Evaluate Tissue Response to a Second Dose of Intravesical Recombinant Adenoviral Interferon α2b Formulated in Syn3 for Failures of Bacillus Calmette-Guerin (BCG) Therapy in Nonmuscle Invasive Bladder Cancer. Ann. Surg. Oncol. 2016, 23, 4110–4114. [Google Scholar] [CrossRef]
  40. Boorjian, S.A.; Alemozaffar, M.; Konety, B.R.; Shore, N.D.; Gomella, L.G.; Kamat, A.M.; Bivalacqua, T.J.; Montgomery, J.S.; Lerner, S.P.; Busby, J.E.; et al. 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. [Google Scholar] [CrossRef]
  41. Shore, N.D.; Boorjian, S.A.; Canter, D.J.; Ogan, K.; Karsh, L.I.; Downs, T.M.; Gomella, L.G.; Kamat, A.M.; Lotan, Y.; Svatek, R.S.; 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. J. Clin. Oncol. 2017, 35, 3410–3416, Erratum in J. Clin. Oncol. 2019, 37, 2187. [Google Scholar] [CrossRef]
  42. Mitra, A.P.; Narayan, V.M.; Mokkapati, S.; Miest, T.; Boorjian, S.A.; Alemozaffar, M.; Konety, B.R.; Shore, N.D.; Gomella, L.G.; Kamat, A.M.; et al. Antiadenovirus Antibodies Predict Response Durability to Nadofaragene Firadenovec Therapy in BCG-unresponsive Non-muscle-invasive Bladder Cancer: Secondary Analysis of a Phase 3 Clinical Trial. Eur. Urol. 2022, 81, 223–228. [Google Scholar] [CrossRef]
  43. Green, J.L.; Osterhout, R.E.; Klova, A.L.; Merkwirth, C.; McDonnell, S.R.P.; Zavareh, R.B.; Fuchs, B.C.; Kamal, A.; Jakobsen, J.S. Molecular characterization of type I IFN-induced cytotoxicity in bladder cancer cells reveals biomarkers of resistance. Mol. Ther. Oncolytics 2021, 23, 547–559. [Google Scholar] [CrossRef] [PubMed]
  44. Shuckman, A.K.; Lotan, Y.; Boorjian, S.A.; Cilwa, K.E.; Dinney, C.P.N. Efficacy of intravesical nadofaragene firadenovec for patients with carcinoma in situ (CIS), BCG-unresponsive non-muscle invasive bladder cancer (NMIBC): Longer-term follow-up from the phase III trial [abstract no. MP16-01]. J. Urol. 2021, 206 (Suppl. S3), E296. [Google Scholar] [CrossRef]
  45. Lotan, Y.; Schuckman, A.K.; Boorjian, S.A.; Cilwa, K.E.; Dinney, C.P.N. Phase III trial of intravesical nadofaragene firadenovec in patients with high-grade, BCG-unresponsive, non-muscle invasive bladder cancer: Two year follow-up in the TA/T1 cohort [abstract no. MP16-02]. J. Urol. 2021, 206 (Suppl. S3), E296. [Google Scholar]
  46. Abidi, A. Cabazitaxel: A novel taxane for metastatic castration-resistant prostate cancer-current implications and future prospects. J. Pharmacol. Pharmacother. 2013, 4, 230–237. [Google Scholar] [CrossRef] [PubMed]
  47. 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. [Google Scholar] [CrossRef] [PubMed]
  48. DeCastro, G.J.; Sui, W.; Pak, J.S.; Lee, S.M.; Holder, D.; Kates, M.M.; Virk, R.K.; Drake, C.G.; Anderson, C.B.; James, B.; et al. A Phase I Trial of Intravesical Cabazitaxel, Gemcitabine and Cisplatin for the Treatment of Nonmuscle Invasive bacillus Calmette-Guérin Unresponsive or Recurrent/Relapsing Urothelial Carcinoma of the Bladder. J. Urol. 2020, 204, 247–253. [Google Scholar] [CrossRef] [PubMed]
  49. DeCastro, G.J.; Anderson, C.B.; Abate-Shen, C.; Gray, J.; Ingram, J.; Stein, M.N.; McKiernan, J.M. Phase 2 trial of intravesical cabazitaxel, gemcitabine, and cisplatin (CGC) for the treatment of non-muscle invasive BCG unresponsive urothelial carcinoma of the bladder. J. Clin. Oncol. 2023, 41, TPS4616. [Google Scholar] [CrossRef]
  50. McElree, I.M.; Steinberg, R.L.; Martin, A.C.; Richards, J.; Mott, S.L.; Gellhaus, P.T.; Nepple, K.G.; O’Donnell, M.A.; Packiam, V.T. Sequential Intravesical Gemcitabine and Docetaxel for bacillus Calmette-Guérin-Naïve High-Risk Nonmuscle-Invasive Bladder Cancer. J. Urol. 2022, 208, 589–599. [Google Scholar] [CrossRef] [PubMed]
  51. Tang, Q.; Chen, Y.; Li, X.; Long, S.; Shi, Y.; Yu, Y.; Wu, W.; Han, L.; Wang, S. The role of PD-1/PD-L1 and application of immune-checkpoint inhibitors in human cancers. Front. Immunol. 2022, 13, 964442. [Google Scholar] [CrossRef]
  52. Bedke, J.; Black, P.C.; Szabados, B.; Guerrero-Ramos, F.; Shariat, S.F.; Xylinas, E.; Brinkmann, J.; Blake-Haskins, J.A.; Cesari, R.; Redorta, J.P. Optimizing outcomes for high-risk, non-muscle-invasive bladder cancer: The evolving role of PD-(L)1 inhibition. Urol. Oncol. Semin. Orig. Investig. 2023, 41, 461–475. [Google Scholar] [CrossRef]
  53. Taoka, R.; Kobayashi, T.; Hidaka, Y.; Abe, H.; Ito, K.; Kojima, T.; Kato, M.; Kanda, S.; Hatakeyama, S.; Matsui, Y.; et al. Impact of prior intravesical bacillus Calmette-Guerin therapy on the effectiveness of pembrolizumab for patients with metastatic urothelial carcinoma. Urol. Oncol. Semin. Orig. Investig. 2022, 40, 107.e1–107.e9. [Google Scholar] [CrossRef]
  54. Wang, Y.; Liu, J.; Yang, X.; Liu, Y.; Liu, Y.; Li, Y.; Sun, L.; Yang, X.; Niu, H. Bacillus Calmette-Guérin and anti-PD-L1 combination therapy boosts immune response against bladder cancer. OncoTargets Ther. 2018, 11, 2891–2899. [Google Scholar] [CrossRef] [PubMed]
  55. Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.J.; Kefford, R.; Wolchok, J.D.; Hersey, P.; Joseph, R.; Weber, J.S. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann. Oncol. 2019, 30, 582–588. [Google Scholar] [CrossRef] [PubMed]
  56. Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L.; et al. KEYNOTE-001 Investigators. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, D.Y.; Salem, J.E.; Cohen, J.V.; Chandra, S.; Menzer, C.; Ye, F.; Zhao, S.; Das, S.; Beckermann, K.E.; Ha, L.; et al. Fatal Toxic Effects Associated wth Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol. 2018, 4, 1721–1728, Erratum in JAMA Oncol. 2018, 4, 1792. [Google Scholar] [CrossRef] [PubMed]
  58. Balar, A.V.; Kamat, A.M.; Kulkarni, 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 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, Erratum in Lancet Oncol. 2021, 22, e347. [Google Scholar] [CrossRef] [PubMed]
  59. Deininger, S.; Törzsök, P.; Mitterberger, M.; Pallauf, M.; Oswald, D.; Deininger, C.; Lusuardi, L. From Interferon to Checkpoint Inhibition Therapy—A Systematic Review of New Immune-Modulating Agents in Bacillus Calmette–Guérin (BCG) Refractory Non-Muscle-Invasive Bladder Cancer (NMIBC). Cancers 2022, 14, 694. [Google Scholar] [CrossRef] [PubMed]
  60. 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. [Google Scholar] [CrossRef] [PubMed]
  61. Available online: https://clinicaltrials.gov/study/NCT03711032?cond=Non-muscle%20Invasive%20Bladder%20Cancer&term=keynote-676&rank=1 (accessed on 2 February 2024).
  62. Woodcock, V.K.; Chen, J.L.; Purshouse, K.; Butcher, C.; Collins, L.; Haddon, C.; Verrall, G.; Elhussein, L.; Roberts, C.; Tarlton, A. PemBla: A Phase 1 study of intravesical pembrolizumab in recurrent non-muscle-invasive bladder cancer. BJUI Compass 2023, 4, 322–330. [Google Scholar] [CrossRef] [PubMed]
  63. Meghani, K.; Cooley, L.F.; Choy, B.; Kocherginsky, M.; Swaminathan, S.; Munir, S.S.; Svatek, R.S.; Kuzel, T.; Meeks, J.J. First-in-human Intravesical Delivery of Pembrolizumab Identifies Immune Activation in Bladder Cancer Unresponsive to Bacillus Calmette-Guérin. Eur. Urol. 2022, 82, 602–610. [Google Scholar] [CrossRef]
  64. Kommalapati, A.; Tella, S.H.; Borad, M.; Javle, M.; Mahipal, A. FGFR Inhibitors in Oncology: Insight on the Management of Toxicities in Clinical Practice. Cancers 2021, 13, 2968. [Google Scholar] [CrossRef]
  65. Ecke, T.H.; Voβ, P.C.; Schlomm, T.; Rabien, A.; Friedersdorff, F.; Barski, D.; Otto, T.; Waldner, M.; Veltrup, E.; Linden, F.; et al. Prediction of Response to Cisplatin-Based Neoadjuvant Chemotherapy of Muscle-Invasive Bladder Cancer Patients by Molecular Subtyping including KRT and FGFR Target Gene Assessment. Int. J. Mol. Sci. 2022, 23, 7898. [Google Scholar] [CrossRef] [PubMed]
  66. Benjamin, D.J.; Hsu, R. Treatment approaches for FGFR-altered urothelial carcinoma: Targeted therapies and immunotherapy. Front. Immunol. 2023, 14, 1258388. [Google Scholar] [CrossRef] [PubMed]
  67. Choudhury, A.; Porta, N.; Hall, E.; Song, Y.P.; Owen, R.; MacKay, R.; West, C.M.L.; Lewis, R.; Hussain, S.A.; James, N.D. BC2001 and BCON investigators. Hypofractionated radiotherapy in locally advanced bladder cancer: An individual patient data meta-analysis of the BC2001 and BCON trials. Lancet Oncol. 2021, 22, 246–255. [Google Scholar] [CrossRef] [PubMed]
  68. Cui, J.; Wang, W.; Chen, S.; Chen, P.; Yang, Y.; Guo, Y.; Zhu, Y.; Chen, F.; Shi, B. Combination of Intravesical Chemotherapy and Bacillus Calmette-Guerin Versus Bacillus Calmette-Guerin Monotherapy in Intermediate- and High-risk Nonmuscle Invasive Bladder Cancer: A Systematic Review and Meta-analysis. Medicine 2016, 95, e2572. [Google Scholar] [CrossRef] [PubMed]
  69. Huang, D.; Jin, Y.H.; Weng, H.; Huang, Q.; Zeng, X.T.; Wang, X.H. Combination of Intravesical Bacille Calmette-Guerin and Chemotherapy vs. Bacille Calmette-Guerin Alone in Non-muscle Invasive Bladder Cancer: A Meta-Analysis. Front. Oncol. 2019, 9, 121. [Google Scholar] [CrossRef] [PubMed]
  70. Tan, W.S.; Prendergast, A.; Ackerman, C.; Yogeswaran, Y.; Cresswell, J.; Mariappan, P.; Phull, J.; Hunter-Campbell, P.; Lazarowicz, H.; Mishra, V.; et al. Adjuvant Intravesical Chemohyperthermia Versus Passive Chemotherapy in Patients with Intermediate-risk Non-muscle-invasive Bladder Cancer (HIVEC-II): A Phase 2, Open-label, Randomised Controlled Trial. Eur. Urol. 2023, 83, 497–504. [Google Scholar] [CrossRef]
  71. Joshi, M.; Atlas, S.J.; Beinfeld, M.; Chapman, R.H.; Rind, D.M.; Pearson, S.D.; Touchette, D.R. Cost-Effectiveness of Nadofaragene Firadenovec and Pembrolizumab in Bacillus Calmette-Guérin Immunotherapy Unresponsive Non-Muscle Invasive Bladder Cancer. Value Health 2023, 26, 823–832. [Google Scholar] [CrossRef]
Table 1. Clinical trials for conservative treatment of NMIBC. * trial completed with posted results, ** status unknown.
Table 1. Clinical trials for conservative treatment of NMIBC. * trial completed with posted results, ** status unknown.
TrialTreatmentEstimated Completion DatePhaseRoute of AdministrationNumber of PatientsPrimary Outcome Measures
NCT01687244Adstiladrin2016 r. *Phase 2Intravesical40Dose and safety assessment:
75 mL of rAd-IFN Dose 3 × 1011 Vps/mL
NCT02773849Adstiladrin2023 r. *Phase 3Intravesical157Complete response after
3 months: 53.4%
NCT03167151Pembrolizumab2019 r. *Phase 1 + 2Intravenous6Safety assessment: 200 mg iv.
Dose
NCT02808143Pembrolizumab + BCG2023 r. *Phase 1Intravenous9Safety assessment: 1/2 mg/kg
Pembrolizumab + 50 mg BGC
NCT04640623Cetrelimab2027 r.Phase 2Intravesical200Complete response and
disease-free survival:
results not posted
NCT05316155Erdafitinib2024 r.Phase 1Intravesical112Safety assessment:
Results not posted
NCT03799835Atezolizumab2028 r.Phase 3Intravenous516Recurrence-free survival after
24 months: results not posted
NCT04134000Atezolizumab2024 r.Phase 1Intravenous40Safety assessment of BCG+
atezolizumab:
results not posted
NCT02792192Atezolizumab
/Atezolizumab + BCG
2020 r. *Phase 1/2Intravenous24Complete response after
6 months: 33.3%/41.7%
NCT03528694Durvalumab + BCG2025 r.Phase 3Intravenous1018Disease-free survival:
results not posted
NCT03759496Durvalumab2022 r. **Phase 2 Intravesical39Safety assessment:
results not posted
NCT04106115Durvalumab2029 r.Phase 1 + 2Intravenous64Safety assessment + disease-free survival:
results not posted
NCT04149574Nivolumab2024 r.Phase 3Intravenous13Event-free survival:
results not posted
NCT04165317Sasanlimab
/sansalimab + BCG
2026 r.Phase 3Intravenous1070Event-free survival:
results not posted
Table 2. Comparison of Pembrolizumab and Adstiladrin.
Table 2. Comparison of Pembrolizumab and Adstiladrin.
PembrolizumabAdstiladrin
FDA approval date16 October 202016 December 2022
MechanismMonoclonal IgG4 anti-PD1 antibodyViral DNA vectors, encoding Interferon-α2b
Administration methodIntravenousIntravesical
ScheduleEvery 3 weeksEvery 12 weeks
Adverse effectsGrade 1 and 2: ~60%Grade 1 and 2: 60–70%
Grade 3: ~15%Grade 3: 4%
Severe: ~10%Severe: none
Complete responseAfter 3 months: 41%After 3 months: 53%
After 12 months: 19%After 12 months: 24%
Cost per year$187,000$240,000
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jaromin, M.; Konecki, T.; Kutwin, P. Revolutionizing Treatment: Breakthrough Approaches for BCG-Unresponsive Non-Muscle-Invasive Bladder Cancer. Cancers 2024, 16, 1366. https://doi.org/10.3390/cancers16071366

AMA Style

Jaromin M, Konecki T, Kutwin P. Revolutionizing Treatment: Breakthrough Approaches for BCG-Unresponsive Non-Muscle-Invasive Bladder Cancer. Cancers. 2024; 16(7):1366. https://doi.org/10.3390/cancers16071366

Chicago/Turabian Style

Jaromin, Maciej, Tomasz Konecki, and Piotr Kutwin. 2024. "Revolutionizing Treatment: Breakthrough Approaches for BCG-Unresponsive Non-Muscle-Invasive Bladder Cancer" Cancers 16, no. 7: 1366. https://doi.org/10.3390/cancers16071366

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop