Historically, combining platinum-based therapies, such as cisplatin or carboplatin, with immunotherapy has not shown a clear OS advantage over chemotherapy alone. This is particularly true for the combination of these platinum drugs with checkpoint inhibitors in earlier-phase studies. However, recent clinical evidence suggests that the combination of cisplatin and nivolumab, especially as first-line therapy, has shown a significant improvement in OS compared with cisplatin alone. This finding represents a promising development in the optimization of treatment for advanced bladder cancer.
Despite numerous achievements in diagnostics and treatment, there is still an urgent need to develop new therapeutic strategies and optimize existing methods. Due to the dynamic development of oncology in recent years, we have observed an intensification of clinical trials on various forms of bladder cancer therapy, including immunotherapy, targeted therapies, and innovative approaches in chemotherapy and radiotherapy. The literature review from 2020 to 2024 includes both early-phase studies (I–II) and those in advanced stages (III–IV), which allows for a comprehensive understanding of current trends and perspectives in bladder cancer treatment. The presented clinical studies not only provide data on potential therapeutic benefits, but also shed light on possible side effects and limitations that may influence the selection of optimal therapy in everyday clinical practice.
Figure 8 shows the number of clinical trials for bladder cancer published between 2020 and 2024, identified using the PubMed search engine. The search was performed using a combination of keywords, such as “bladder cancer”, “clinical trial”, and “therapy”. Data on the number of clinical trials were counted based on the PubMed search results, limiting them to the years 2020–2024.
3.8.1. Selected Clinical Trials from 2020
The POUT trial, an open-label, randomized, controlled phase 3 clinical trial, was designed to evaluate the efficacy of adjuvant platinum-based chemotherapy in patients with upper urinary tract urothelial carcinoma (UTUC). The trial was conducted at 71 clinical sites in the United Kingdom. It included 261 patients who had undergone radical nephrectomy and had pT2-T4, pN0–N3 M0, or pTany N1–3 M0 UTUC. Participants were randomly assigned to one of two groups: observation, or 4, 21-day cycles of chemotherapy. Randomization was central and based on a random-effect minimization algorithm. The chemotherapy regimen included intravenous cisplatin (70 mg/m
2) or carboplatin (AUC 4.5/AUC 5 for glomerular filtration rate < 50 mL/min) on day 1 of the cycle, and intravenous gemcitabine (1000 mg/m
2) on days 1 and 8 of the cycle. Chemotherapy was initiated within 90 days of surgery. Monitoring included standard cystoscopic, radiological, and clinical examinations. The primary endpoint of the study was disease-free survival (DFS), analyzed according to the intention-to-treat principle using the Peto–Haybittle rule to assess treatment efficacy. The results of the study showed that adjuvant chemotherapy significantly improved disease-free survival (DFS) in patients with UTUC. The hazard ratio (HR) was 0.45 (95% CI 0.30–0.68,
p = 0.0001) with a median follow-up of 30.3 months. The three-year event-free survival was estimated at 71% in the chemotherapy group, compared with 46% in the observation group. Of the 126 patients who started chemotherapy, 44% experienced grade 3 or higher acute adverse events, which was consistent with the expectations for the toxicity of the treatment regimen. In the observation group, only 4% of patients had grade 3 or higher acute adverse events. There were no treatment-related deaths in the study. The findings of the POUT study suggested that gemcitabine- and platinum-based chemotherapy initiated within 90 days after radical nephrectomy should be considered as the new standard of care for patients with locally advanced urothelial cancer of the upper urinary tract, as it significantly improved disease-free survival [
117].
A phase 3 clinical trial in patients with locally advanced or metastatic urothelial cancer was designed to evaluate the efficacy of avelumab maintenance therapy in combination with the best supportive care versus supportive care alone. The trial included patients who had not progressed following first-line platinum-based chemotherapy (gemcitabine plus cisplatin or carboplatin). Participants were randomly assigned to receive avelumab or a control group that received supportive care alone. The primary endpoint of the study was overall survival, assessed both in the overall patient population and in the subgroup with positive PD-L1 status. The study results showed that the addition of avelumab to supportive care significantly prolonged overall survival compared with supportive care alone. In the overall population, the median overall survival was 21.4 months in the avelumab group, compared with 14.3 months in the control group (hazard ratio for death, 0.69). A significant improvement in overall survival was also observed in the PD-L1-positive population, with a one-year survival rate of 79.1% in the avelumab group, compared with 60.4% in the control group (hazard ratio for death, 0.56). The study also showed a benefit in terms of progression-free survival, with a median progression-free survival of 3.7 months in the avelumab group and 2.0 months in the control group in the overall population, and 5.7 months and 2.1 months, respectively, in the PD-L1-positive population. However, the adverse event rates were higher in the avelumab group, at 98.0%, compared with 77.7% in the control group. Grade 3 or higher adverse events occurred in 47.4% of patients in the avelumab group, compared with 25.2% in the control group. The results of this clinical trial indicate a significant benefit of avelumab as maintenance therapy in patients with advanced urothelial cancer who have not progressed after first-line chemotherapy. This therapy prolongs overall survival, especially in the PD-L1-positive population, although it is associated with a higher incidence of adverse events [
118].
The multicenter, randomized HYBRIDBLUE clinical trial, conducted at six German academic centers, compared the efficacy of two surgical techniques for patients with non-muscle-invasive bladder cancer (NMIBC). The aim of the study was to determine whether transurethral en bloc resection with submucosal hydro-dissection (TUEB) was superior to standard transurethral resection of bladder tumors (TURBT) in terms of the quality of resection and the ability to accurately assess histopathological muscle invasion. The study included 305 patients who were randomly assigned to 1 of 2 treatment groups after initial screening: TUEB, in which resection was assisted by hexaminolevulinate (HAL), or TURBT, in which HAL was also used as an aid for visualization of lesions. The primary objective of the study was to compare the proportion of tissue samples that could be reliably assessed for the presence of muscularis propria and the margins of resection (R0 vs. R1), which are crucial for assessing the completeness of the procedure and planning further treatment. Additional aspects included completeness of resection, presence of muscularis propria in the specimens, recurrence rate, and the rate and type of postoperative complications. The final analysis included 115 patients, 56 of whom underwent TUEB and 59 TURBT. The results of the study showed that 86% of patients in the TUEB group had adequate histopathological slides to accurately assess muscle invasion, compared with 63% in the TURBT group (
p = 0.006). Furthermore, complete tumor removal (R0) was confirmed in 57% of patients in the TUEB group, compared with only 9% in the TURBT group (
p < 0.001). There were no major complications (class ≥III) in either group. At 3 months after the procedure, disease recurrence occurred in 3 patients in the TUEB group and 7 in the TURBT group, whereas at 1 year, this number increased to 19 and 11 patients, respectively (
p = 0.33 and
p = 0.08). In summary, the results of this study indicate that TUEB is a safe technique and provides better conditions for histopathological evaluation compared with standard TURBT resection. Although the differences in recurrence rates were not statistically significant, the results suggest that TUEB may reduce the need for early adjuvant resections. However, further studies are needed with recurrence-free survival as the primary endpoint, which will allow for a more precise assessment of the oncological efficacy of both methods [
119].
The NCT03998371 clinical trial prospectively recruited patients with urothelial cancer and cancer-free controls. The aim of the study was to develop a non-invasive diagnostic method for urothelial cancer by low-coverage whole-genome sequencing of DNA from exfoliated urine cells. Analyses were performed using the Illumina HiSeq XTen system, followed by the use of a dedicated bioinformatics workflow, called Urine Exfoliated Cells Copy Number Aberration Detector (UroCAD), to identify chromosome copy number changes. In the discovery phase of the study, urine samples from 126 patients with urothelial cancer and 64 samples from non-cancer individuals were analyzed. Patients with cancer had multiple and characteristic chromosome copy number changes that were absent or much less pronounced in the control group. Based on these changes, a diagnostic model was developed, UroCAD, which included all autosomal chromosome changes. The model achieved an AUC of 0.92, with a 95% confidence interval ranging from 89.4% to 97.3%. At the optimal diagnostic threshold of |Z| ≥ 3.21, the model had a sensitivity, specificity, and accuracy of 82.5%, 96.9%, and 89.0%, respectively. A statistically significant correlation was also found between positive prediction and tumor stage (
p = 0.01). External validation was performed in a further part of the study on 95 participants. The UroCAD test detected urothelial cancers with a sensitivity of 80.4%, a specificity of 94.9%, and an AUC of 0.91. These results were significantly better than those of traditional cytological tests, which had a sensitivity of only 33.9% (
p < 0.001) with comparable specificity (94.9% vs. 100%,
p = 0.49). The study suggests that UroCAD may be an effective and non-invasive diagnostic method for urothelial cancer, with better sensitivity than cytological tests, while maintaining similar specificity. It could potentially be used for both early detection of cancer and monitoring for recurrence, reducing the need for invasive diagnostic procedures, such as cystoscopy, which would significantly reduce the burden on patients [
120].
To optimize the treatment of non-muscle-invasive bladder cancer (NMIBC), an application (APPv) was developed and evaluated in a prospective, double-blind, observational study of therapy compliance. The aim of the study was to assess the compliance of treatment prescriptions for NMIBC, including both overall compliance and adjustment for disease recurrence and progression rates, comparing APPv recommendations with clinical decisions made by urologists based on their experience. The study was conducted in a sample of 100 patients with histologically confirmed NMIBC, both at initial diagnosis and at later stages of disease. The study compared the therapy recommended by the application with the therapy recommended by the urologist to determine the degree of compliance between the two methods. The results showed that in 64% of cases, the treatment recommended by the urologist was consistent with that recommended by the application, as evidenced by a kappa score of 0.55 (
p < 0.0001). Across risk subgroups, agreement was 77% for low-risk patients (kappa 0.55,
p = 0.002), 63% for intermediate-risk patients (kappa 0.52,
p < 0.0001), 17% for high-risk patients (kappa 0.143,
p = 0.014), and 66% for very high-risk patients (kappa 0.71,
p = 0.01). The efficacy analysis showed that patients who received adjuvant intravesical therapy according to the app recommendations remained free from relapses in 89.1% of cases, compared with 61.1% of patients who were noncompliant with the app (
p = 0.0004), translating into a relative risk (RR) of 0.46 (95%CI: 0.25–0.86) versus RR of 2.4 (95%CI: 1.5–3.8,
p = 0.001). Moreover, in the group of patients whose treatment was consistent with the app and urologist’s recommendations, all patients remained free from disease progression, whereas in the group where there was discordance, the percentage was 88.9% (
p = 0.004), with an RR of 1 versus RR of 1.125 (95%CI: 1–1.26,
p = 0.004). In summary, the results of the study suggest that the APPv app can effectively support adherence to treatment recommendations consistent with clinical guidelines, which may translate into better health outcomes in patients with NMIBC [
121].
3.8.2. Selected Clinical Trials from 2023 (Table 9)
A global, randomized, phase 3 clinical trial was conducted to evaluate the efficacy of erdafitinib versus chemotherapy in patients with metastatic urothelial cancer harboring susceptible FGFR3 or FGFR2 alterations. The study population included patients whose disease had progressed after one or two prior therapies, including checkpoint inhibitors, such as anti-PD-1 or anti-PD-L1 antibodies. Participants were randomly assigned to one of two treatment groups: one group received erdafitinib and the other received chemotherapy of the investigator’s choice (docetaxel or vinflunine). The primary objective of the study was to compare overall survival (OS) between the groups. A total of 266 patients were included in the study, of whom 136 were assigned to receive erdafitinib and 130 to receive chemotherapy. The median follow-up time was 15.9 months. The study results showed that the median overall survival was significantly longer in the erdafitinib group (12.1 months) compared with the chemotherapy group (7.8 months). The hazard ratio for death was 0.64 (95% confidence interval 0.47–0.88,
p = 0.005), indicating a 36% reduction in the risk of death in the erdafitinib group. In addition, the median progression-free survival (PFS) was also longer in the erdafitinib group (5.6 months) than in the chemotherapy group (2.7 months), with a hazard ratio for progression or death of 0.58 (95% CI 0.44–0.78,
p < 0.001). In terms of safety, the incidence of grade 3 or 4 adverse events was similar in the two groups, at 45.9% and 46.4%, respectively. In contrast, treatment-related adverse events leading to death were less common in the erdafitinib group (0.7%) than in the chemotherapy group (5.4%). In summary, the results of the study showed that erdafitinib treatment resulted in significantly longer overall survival compared with chemotherapy in patients with metastatic urothelial cancer with FGFR alterations who had previously received checkpoint inhibitors [
122]. A randomized clinical trial was conducted to assess the efficacy of selenium and vitamin E supplementation in preventing recurrence and progression of non-muscle-invasive bladder cancer (NMIBC). The study included 270 patients with newly diagnosed NMIBC, recruited from 10 secondary and tertiary care hospitals in the United Kingdom. Participants were randomly assigned to one of four groups to receive different combinations of supplements: selenium plus placebo, vitamin E plus placebo, selenium plus vitamin E, or placebo plus placebo. Recruitment to the study took place between 17 July 2007 and 10 October 2011, and participation was restricted to a diagnosis of NMIBC (stages Ta, T1, or Tis) and randomization within 12 months of the first transurethral resection of the tumor. The aim of the study was to investigate whether supplementation with selenium and/or vitamin E could reduce the risk of disease recurrence in patients with NMIBC. Patients received oral selenium (200 μg/day selenium-rich yeast), vitamin E (200 IU/day d-alpha-tocopherol), or matching placebo. The mean follow-up time was 5.5 years, and 84% of participants were followed for at least 5 years. The results of the study showed that selenium supplementation had no significant effect on reducing the risk of bladder cancer recurrence. Compared with the control group, there was no statistically significant difference in the relapse-free interval (RFI) in patients receiving selenium. In contrast, vitamin E supplementation was associated with a statistically significant increase in the risk of relapse. The hazard ratio values for vitamin E indicated an increased risk of relapse (1.46; 95% CI 1.02–2.09), suggesting that vitamin E may be potentially harmful for patients with NMIBC. Analyses did not show significant differences in the progression-free interval or overall survival for either group. A total of 1957 adverse events were recorded in the study, 85 of which were considered serious, but none of these events were attributed to the study treatment. Due to slow recruitment of participants, the study was prematurely terminated. The results suggested that vitamin E supplementation may not only not be beneficial but may even pose a risk to patients with NMIBC, which requires further investigation into the biological mechanisms of this phenomenon [
123].
The SWOG S1314 study aimed to investigate the potential role of cell-free DNA (cfDNA) methylation as a predictive biomarker of response to neoadjuvant chemotherapy (NAC) in patients with muscle-invasive bladder cancer (MIBC). The study included 72 patients with MIBC (cT2-T4aN0M0, ≥5 mm viable tumor) who had blood samples collected before and during NAC treatment, as per the protocol. The aim of the study was to determine whether cfDNA methylation signatures in plasma could be associated with pathological response to NAC, as assessed by radical cystectomy. The study utilized the Infinium Methylation EPIC BeadChip technology to measure cfDNA methylation. Based on the analysis of differences in methylation between patients who achieved pathological response (≤pT1N0) and those who did not, a predictive classifier was developed using the Random Forest algorithm. The study results showed that cfDNA methylation level before chemotherapy was associated with treatment response, which allowed the development of a methylation response index (mR-score). Additionally, the mR-score obtained after the first cycle of NAC also showed a similar predictive ability. The study also calculated the fraction of circulating DNA from the bladder based on cfDNA methylation data, which proved to be an additional, independent predictor of treatment response. Combining the mR-score with the fraction of circulating bladder DNA correctly predicted pathological response in 79% of patients based on plasma samples collected both before and after the first cycle of NAC. Although the study provided evidence for the efficacy of cfDNA methylation as a predictive biomarker of response to NAC, its limitations included the relatively small number of participants and low levels of circulating bladder DNA in some cases. Nevertheless, the results of this study represent an important step toward personalizing NAC treatment in patients with bladder cancer, suggesting the potential for using cfDNA methylation as a tool to better tailor therapy to patients’ needs [
124]. Another phase 1 study evaluated the safety, tolerability, and preliminary efficacy of the novel TAR-200 drug delivery system in patients with muscle-invasive bladder cancer who declined or were ineligible for curative-intent therapy. Muscle-invasive bladder cancer presents a significant therapeutic challenge, and many patients, particularly the elderly or frail ones, are unable to undergo standard curative-intent therapies. In response to this unmet need, TAR-200, a system that delivers gemcitabine directly to the bladder continuously over a 21-day dosing cycle, was developed. The study included patients with cT2-cT3bN0M0 urothelial bladder cancer who received four consecutive cycles of therapy for a total of 84 days. The primary endpoints of the study were the assessment of the safety and tolerability of TAR-200 at 84 days. Additionally, clinical complete and partial response rates were assessed, which were verified by cystoscopy, biopsy, and imaging techniques, as well as the duration of response and overall survival. The study included 35 patients, 68.6% of whom were male, with a median age of 84 years. Treatment-related adverse events occurred in 15 patients, 2 of whom required removal of TAR-200. After 3 months of treatment, a complete response was observed in 31.4% of patients and partial response in 8.6%, for a combined response rate of 40.0%. Median overall survival was 27.3 months, and the median duration of response was 14 months. In addition, the progression-free rate at 12 months was 70.5%. The results of this study suggest that TAR-200 is generally safe and well tolerated in older, frail patients and shows promising initial efficacy in a patient population with very limited treatment options [
125].
A study investigating the effect of μ-opioid receptor agonists (MORA) on the progression and metastasis of bladder cancer (BCa) provided important insight into the potential mechanisms underlying these processes and suggested potential therapeutic directions. MORA, commonly used to relieve pain in BC patients both during surgery and for chronic pain management, has been shown to influence the generation of circulating tumor cells (CTCs), which in turn contributes to the enhancement of metastasis. This study used both preclinical models and patient studies to specifically investigate the effect of MORA on neoplastic processes. A novel microfluidic immunocapture microchip was used to detect changes in CTC numbers in both mouse models and BC patients. The results showed a significant increase in the number of CTCs with epithelial and/or mesenchymal features after MORA treatment. Using advanced bioinformatics methods, whole transcriptome sequencing, and molecular biology techniques, the MOR/PI3K/AKT/Slug signaling pathway was identified as a key mechanism responsible for this effect. Activation of this pathway promoted epithelial-to-mesenchymal transition (EMT) of cancer cells, which facilitated their migration and CTC formation. In turn, interventions blocking MOR, Slug, or PI3K inhibited EMT and reduced the number of CTCs, suggesting that the MOR/AKT/Slug axis may be a potential therapeutic target. The results of this study indicated that despite the benefits of the analgesic effects of MORAs, their use may contribute to an increased risk of metastasis by promoting the formation of circulating tumor cells. Therefore, in future clinical interventions, it would be possible to target the EMT-CTC pathway to reduce the negative effects of MORAs without losing their analgesic effects [
126].
The RAIDER study evaluated the acute toxicity of different radiotherapy regimens used in the treatment of bladder cancer, focusing on the comparison of hypofractionated fractionation (55 Gy in 20 fractions) with conventional fractionation (64 Gy in 32 fractions) and the effect of concomitant (chemo)therapy. In the randomized clinical trial, patients with muscle-invasive bladder cancer stage T2–T4a N0 M0 were assigned to one of three groups: standard radiotherapy, standard-dose adaptive radiotherapy, or dose-increased adaptive radiotherapy. Neoadjuvant and concomitant chemotherapy were allowed, which allowed for the analysis of the effect of different treatment regimens on the occurrence of acute adverse events. The study included 345 patients, recruited from 46 centers, who received 20 or 32 fractions of radiotherapy. The median age of participants was 73 years, and 49% of patients had received prior neoadjuvant chemotherapy. Additionally, 71% of patients were treated with concurrent therapy, with the most common being 5-fluorouracil plus mitomycin C. Acute toxicity, graded using the Common Terminology Criteria for Adverse Events (CTCAE), was monitored weekly during radiotherapy and 10 weeks after treatment initiation. The results of the analysis showed that grade 2 or higher gastrointestinal toxicity was significantly higher in patients receiving concurrent therapy compared with radiotherapy alone in the 20-fraction cohort, but similar differences were not observed in the 32-fraction cohort. The highest gastrointestinal toxicity was observed in patients treated with gemcitabine, confirming the significant differences between treatment groups in the 32-fraction cohort. In terms of grade 2 or higher urinary tract toxicity, no significant differences were found between different concomitant therapy regimens in both the 20-fraction and 32-fraction cohorts. This study highlights that acute grade 2 or higher toxicity is a common phenomenon in radiotherapy for bladder cancer, and the adverse event profile varies depending on the concomitant therapy regimen used. In particular, gastrointestinal toxicity seems to be more severe in patients receiving gemcitabine, which indicates the need for further studies to optimize treatment in this group of patients [
127].
Another clinical trial evaluated the safety and efficacy of OncoTherad
® (MRB-CFI-1) nano-immunotherapy in patients with non-muscle-invasive bladder cancer (NMIBC) who had failed to respond to prior treatment with Bacillus Calmette–Guérin (BCG). This single-arm, phase I/II study included 44 patients, of whom 59.1% were BCG-resistant, 31.8% had relapsed disease, and 9.1% were BCG-intolerant. The primary efficacy measures were the pathological complete response (pCR) rate and relapse-free survival (RFS), while secondary measures included the duration of response and safety. After 24 months of follow-up, the pCR rate was 72.7% and the median RFS was 21.4 months. In the group of patients who achieved pCR, the median duration of response was 14.3 months. Of note, no patient developed muscle-invasive cancer or metastatic disease during treatment. Treatment-related adverse events were reported in 77.3% of patients, with mostly grade 1–2 adverse events, indicating good tolerability of the treatment. In terms of the mechanism of action, OncoTherad
® demonstrated the ability to activate the innate immune system via the toll-like receptor 4 (TLR4) signaling pathway, which in turn led to increased activity of the interferon pathway. This activation was crucial for the stimulation of CX3CR1+ CD8 T cells, which was associated with reduced expression of immune checkpoint molecules and reversal of immunosuppression in the bladder tumor microenvironment. The results of the study suggest that OncoTherad
® is a safe and effective treatment option for patients with NMIBC who are BCG-refractory and may also have potential benefits in preventing tumor relapse [
128].
In a population-based study based on the Surveillance, Epidemiology, and End Results (SEER), and external validation using data from The Cancer Genome Atlas (TCGA), prognostic nomograms were developed and validated for patients with bladder cancer (BCa). The aim of the study was to compare the prognostic value of different lymph node metastasis (LNM) indices and to develop prognostic tools that could aid clinical treatment decisions. The study identified 10,093 patients with bladder cancer from the SEER database and 107 patients from the TCGA database. The data were used to create multivariate Cox regression models comparing the predictive performance of indices, such as N classification, number of positive lymph nodes (PLN), lymph node ratio (LNR), and log odds of positive lymph nodes (LODDS). The results of the analysis showed that the LODDS score had a higher prognostic performance than the other indices in terms of both overall survival (OS) and cause-specific survival (CSS). Based on the results, variables such as age, LODDS, and T and M classifications were included in the OS nomograms, while the CSS nomograms included gender, tumor stage, LODDS, and T and M classifications. These nomograms were validated for their prognostic accuracy and clinical utility in three independent cohorts. Furthermore, analysis of the tumor microenvironment revealed significant differences between risk groups, suggesting that different immune profiles may influence patient prognosis. In summary, the study demonstrated that the LODDS score is superior to traditional lymph node metastasis indices in terms of the prognostic value for patients with bladder cancer. The incorporation of LODDS into prognostic nomograms can significantly improve the accuracy of prognostication, thus supporting clinical decision-making [
129]. The next study assessed the performance of radiomic signatures in detecting lymph node (LN) metastases in patients with bladder cancer (BCa), which is crucial for determining the optimal treatment plan. This study investigated the differences in diagnostic performance between manual and automated segmentation of LNs on computed tomography (CT) images. Among 1354 BC patients who underwent radical cystectomy with lymph node dissection, 391 patients were included in the study and had pathological determination of LN status (pN0:
n = 297; pN+:
n = 94). Patients were randomly divided into two groups: training (
n = 274) and test (
n = 117). Pelvic LN segmentation was performed both manually and automatically. From each node, 1004 radiomic features were extracted and used to train a machine learning model to predict lymph node (pN) status based on histopathological results. The results obtained by radiomic analysis based on manual and automated segmentation were compared with the assessment performed by radiologists. The receiver operating characteristic (ROC) curve method was used to assess sensitivity, specificity, and area under the curve (AUC). In the test group, manually segmented lymph nodes achieved an AUC of 0.80 (95% CI 0.69–0.91,
p = 0.64), while automated segmentation achieved an AUC of 0.70 (95% CI 0.58–0.82,
p = 0.17). These results were compared with the radiologist’s assessment, for which the AUC was 0.78 (95% CI 0.67–0.89). Combining the radiomic signature based on manual segmentation with the radiological assessment achieved an AUC of 0.81 (95% CI 0.71–0.92,
p = 0.63). The study showed that the radiomic signature can be an effective tool for distinguishing lymph node status in BC patients, with manual segmentation outperforming the automated approach in terms of diagnostic accuracy. The results suggest that integrating radiomic techniques with traditional radiological assessment can lead to improved disease staging, which is important for further therapeutic decisions [
130].
Another clinical trial aimed to evaluate the therapeutic effect of the combination of Kushen (CKI) and gemcitabine in postoperative patients with non-muscle-invasive bladder cancer (NMIBC) and to investigate the effect of this treatment on selected serum factors and the immune system. The study included 150 patients who were randomly divided into 2 groups. The control group (
n = 75) received standard gemcitabine therapy, which consisted of 0.2 g of gemcitabine once a week for eight weeks after surgery, followed by continuation of treatment in a cycle every two weeks for eight subsequent applications. The observation group (
n = 75) was treated with a CKI injection for 10 days in each of 3 cycles of therapy, in addition to gemcitabine. The study lasted two years, during which blood biochemical parameters, serum factor levels, and changes in T-cell subsets were monitored, as well as the safety of the therapy and the functioning of the immune system. The results indicated that patients in both groups had a significant increase in the levels of interferon-γ, interleukin-2 (IL-2), cell adhesion molecules (CAMs), hepatocyte CAMs, and cysteine proteinase-8 after the end of therapy. This increase was more pronounced in the observation group. At the same time, there was a significant decrease in the levels of tumor necrosis factor-α (TNF-α), C-reactive protein (CRP), IL-6, and other inflammatory and adhesion markers, such as metalloproteinases (MMP-9 and MMP-2), E-cadherin, epithelium-specific CAMs, soluble CAM-1, hepatic CAMs, vascular endothelial growth factor (VEGF), and fibroblast growth factor (FGF). The decrease in these parameters was also more pronounced in the group of patients receiving additional CKIs. Moreover, a significant decrease in the frequency of disease relapses and adverse events was observed in the observation group compared to the control group. The study results suggest that combining CKI therapy with gemcitabine may not only effectively alleviate clinical symptoms and reduce the inflammatory response, but also improve the safety of treatment, reducing the risk of adverse events and improving the overall efficacy of treatment in patients with NMIBC [
131].
Table 9.
Summary of clinical trials in bladder cancer [
117,
118,
119,
120,
121,
122,
123,
124,
125,
126,
127,
128,
129,
130,
131,
132,
133,
134,
135,
136], 2020–2024.
Table 9.
Summary of clinical trials in bladder cancer [
117,
118,
119,
120,
121,
122,
123,
124,
125,
126,
127,
128,
129,
130,
131,
132,
133,
134,
135,
136], 2020–2024.
Year | References | Study Phase | Key Findings |
---|
2020 | [117] | Phase 3 | The POUT trial showed that adjuvant chemotherapy with gemcitabine and platinum initiated within 90 days post-nephrectomy significantly improved disease-free survival (DFS) in upper urinary tract urothelial carcinoma patients compared to observation. |
2020 | [118] | Phase 3 | Avelumab maintenance therapy significantly prolonged overall survival in advanced urothelial cancer patients who did not progress after first-line chemotherapy, particularly in PD-L1-positive patients. |
2020 | [119] | Randomized | The HYBRIDBLUE trial demonstrated that transurethral en bloc resection with submucosal hydro-dissection (TUEB) was superior to standard TURBT for histopathological assessment in non-muscle-invasive bladder cancer. TUEB achieved more accurate muscle invasion assessment and higher rates of complete tumor removal with fewer complications. |
2020 | [120] | Phase 1 | The SAKK 06/14 study of VPM1002BC for non-muscle-invasive bladder cancer patients who failed BCG therapy demonstrated safety and induced a strong immune response. |
2020 | [121] | Phase 1–2 | BioXmark® liquid markers demonstrated high visibility and positional stability during image-guided radiotherapy for invasive bladder cancer, improving treatment precision without related adverse events. |
2020 | [122] | Phase 3 | The use of intraoperative warmed fluids during robot-assisted radical cystectomy reduced the risk of hypothermia, decreased blood transfusions, and shortened hospitalization. |
2020 | [123] | Phase 2 | IMRT in patients with node-positive bladder cancer (NPBC) and high-risk node-negative bladder cancer (NNBC) was feasible, with low toxicity, low recurrence rates, and potential for long-term disease control. |
2020 | [124] | Phase 3 | The KEYNOTE-045 trial demonstrated durable antitumor activity of pembrolizumab in advanced urothelial cancer patients who failed prior platinum-based chemotherapy, with better tolerance and fewer adverse events compared to chemotherapy. |
2020 | [125] | Randomized | High-intensity interval training (HIIT) significantly improved exercise capacity (VO2AT) and reduced blood pressure in older patients before major urological surgery, suggesting better cardiovascular health and potential for improved surgical outcomes. |
2020 | [126] | Prospective | The NCT03998371 trial developed UroCAD, a non-invasive diagnostic method for urothelial cancer based on whole-genome sequencing of exfoliated urine cells, achieving better sensitivity and specificity than traditional cytological tests. |
2021 | [127] | Phase 3 | Nadofaragen firadenovec, a novel gene therapy, achieved a 53.4% complete response rate in BCG-refractory non-muscle-invasive bladder cancer patients within 3 months, offering a new treatment option. |
2021 | [128] | Phase 3 | The GETUG/AFU V05 VESPER trial showed that dose-escalated chemotherapy with dd-MVAC improved local control in muscle-invasive bladder cancer compared to the GC regimen, though it was associated with more severe toxicity. |
2021 | [129] | Observational | SNPs of lncRNA TINCR were associated with increased or decreased risk of bladder cancer, depending on the variant, with polymorphisms affecting the expression of miRNAs, indicating potential regulatory mechanisms. |
2021 | [130] | Randomized | HoLRBT was shown to be as effective as TURBT in treating NMIBC, with fewer complications and shorter catheterization and hospitalization times. |
2021 | [131] | Randomized | En bloc resection with green light laser was superior to traditional TURBT for non-muscular bladder cancer in reducing obturator reflex and blood loss, providing better-quality histopathological samples. |
2021 | [132] | Phase 3 | The VESPER trial compared GC and dd-MVAC chemotherapy regimens, showing that four cycles of cisplatin-based chemotherapy before cystectomy is sufficient for optimal pathological outcomes without significant deterioration in renal function. |
2021 | [133] | Phase 2 | RC48-ADC, a HER2-targeted antibody–drug conjugate, achieved a 51.2% ORR in HER2+ advanced urothelial cancer patients with a median OS of 13.9 months, showing promising efficacy and safety. |
2021 | [134] | Phase 3 | A 14-miRNA hypoxia signature was developed and validated, showing prognostic value in bladder cancer patients receiving carbogen and nicotinamide combined with radiotherapy, improving local recurrence-free survival. |
2021 | [135] | Retrospective | A predictive model for lymph node invasion (LNI) in extended lymph node dissection (PLND) was developed, showing moderate prediction accuracy (AUC 73%) and suggesting the need for better patient selection for extended PLND. |
2021 | [136] | Randomized | Plan-of-the-Day (POD) adaptive radiotherapy in muscle-invasive bladder cancer reduced severe nongenitourinary toxicity (6% vs. 13%) and achieved good local disease control at 3 months (81.3%). |
2022 | [137] | Phase 3 | The BC2001 study indicated that adding chemotherapy (fluorouracil and mitomycin C) to radiotherapy for muscle-invasive bladder cancer improves locoregional control and decreases the need for cystectomy. |
2022 | [138] | Randomized | Diabetic patients using metformin had better overall survival and disease-specific survival compared to non-metformin users and nondiabetic patients with non-muscle-invasive bladder cancer treated with BCG. |
2022 | [139] | Observational | APPv application for NMIBC treatment adherence showed improved treatment outcomes when its recommendations were followed, especially in high-risk and very high-risk patients. |
2022 | [140] | Prospective | The ADXBLADDER test effectively detected high-grade recurrence in low-grade non-muscle-invasive bladder cancer, potentially reducing the need for invasive surveillance cystoscopy. |
2022 | [141] | Randomized | Levofloxacin combined with intravesical BCG therapy reduced the severity of frequent urination and fever while improving long-term progression-free survival and cancer-specific survival in NMIBC patients. |
2022 | [142] | Phase 1/2 | LSAM-DTX showed promising safety, efficacy, and immune response in high-risk non-muscle-invasive bladder cancer patients, with prolonged recurrence-free survival in higher-dose groups and minimal systemic exposure. |
2022 | [143] | Phase 2b | The Optima II trial demonstrated that UGN-102, a chemoablative gel with mitomycin, was a safe and effective non-surgical treatment for NMIBC, with high patient satisfaction and no significant worsening of urinary symptoms. |
2022 | [144] | Randomized | Coated silicone catheters with biofilm-inhibiting technology showed lower bacterial colonization compared to standard Foley catheters in patients after radical cystectomy, though CAUTI rates were similar in both groups. |
2022 | [145] | Prospective | Low serum 25-hydroxyvitamin D levels were significantly associated with bladder cancer, especially the muscle-invasive form, but did not show significant prognostic value for recurrence in NMIBC. |
2022 | [146] | Phase 1 | Oncofid-P-B showed high safety and efficacy in patients with carcinoma in situ (CIS) of the bladder who did not respond to BCG treatment, with 75% achieving complete response (CR) after 12 weeks and 40% maintaining CR after 12 months. |
2023 | [147] | Phase 3 | Erdafitinib significantly improved overall survival and progression-free survival compared to chemotherapy in metastatic urothelial cancer patients with FGFR2/3 alterations who had previously received checkpoint inhibitors. |
2023 | [148] | Randomized | Vitamin E supplementation increased the risk of relapse in NMIBC patients, while selenium supplementation showed no significant effect in preventing recurrence or progression of the disease. |
2023 | [149] | Prospective | cfDNA methylation signatures were predictive of response to neoadjuvant chemotherapy in muscle-invasive bladder cancer, with a methylation response index (mR-score) correctly predicting pathological response in 79% of patients. |
2023 | [150] | Phase 1 | The TAR-200 drug delivery system demonstrated safety, tolerability, and a 40% response rate in patients with muscle-invasive bladder cancer who were ineligible for or declined curative-intent therapies. |
2023 | [151] | Preclinical/Patient study | μ-opioid receptor agonists (MORA) were shown to increase circulating tumor cells in bladder cancer patients, potentially promoting metastasis via the MOR/AKT/Slug signaling pathway. |
2023 | [152] | Randomized | The RAIDER study highlighted that acute grade 2 or higher toxicity is common during radiotherapy for bladder cancer. Gastrointestinal toxicity was more severe in patients receiving concurrent gemcitabine therapy compared to other regimens. No significant differences in urinary tract toxicity were found between regimens. |
2023 | [153] | Phase 1/2 | OncoTherad® (MRB-CFI-1) nanoimmunotherapy demonstrated a 72.7% pathological complete response (pCR) rate in BCG-refractory NMIBC patients, with a median relapse-free survival of 21.4 months. The treatment was well tolerated, with mostly mild to moderate adverse events. The mechanism of action involved activation of the TLR4 and interferon pathways, enhancing CD8+ T-cell activity and reversing immunosuppression in the tumor microenvironment. |
2023 | [154] | Population-based | A study based on SEER and TCGA databases developed and validated prognostic nomograms for bladder cancer patients. The LODDS (log odds of positive lymph nodes) score showed superior prognostic performance over traditional lymph node metastasis indices in predicting both overall survival (OS) and cause-specific survival (CSS). Incorporating LODDS into nomograms significantly improved the accuracy of prognostication and could aid in clinical decision-making. |
2023 | [155] | Diagnostic | A radiomics study assessing manual versus automated lymph node segmentation in bladder cancer patients found that manual segmentation outperformed automated segmentation, achieving an AUC of 0.80 compared to 0.70. Combining manual radiomic analysis with traditional radiological assessment improved lymph node metastasis detection, enhancing diagnostic accuracy and disease staging for further therapeutic decisions. |
2023 | [156] | Randomized | A clinical trial evaluating the combination of Kushen (CKI) and gemcitabine in postoperative NMIBC patients demonstrated a greater reduction in inflammatory markers (TNF-α, CRP, and IL-6) and a more pronounced increase in immune response markers (IFN-γ and IL-2) compared to gemcitabine alone. CKI also improved treatment safety, reduced the frequency of relapses, and alleviated clinical symptoms. |
2024 | [157] | Phase 2 | A phase II study investigating the COXEN gene expression model found that molecular subtypes of muscle-invasive bladder cancer (basal squamous, luminal, and neuronal) were modest predictors of pathological response to neoadjuvant chemotherapy, with limited impact on long-term outcomes, such as progression-free survival (PFS) and overall survival (OS). Further research is needed to understand their role as predictive biomarkers. |
2024 | [158] | Phase 1b | The PrECOG PrE0807 study evaluated the safety and efficacy of neoadjuvant nivolumab, with or without lirilumab, in cisplatin-ineligible patients with muscle-invasive bladder cancer (MIBC). Nivolumab-based neoadjuvant immunotherapy was well tolerated, with a 2-year relapse-free survival of 73–71% and overall survival of 82–89%. The combination therapy significantly increased CD8+ T-cell density, indicating potential immune activation. |
2024 | [159] | Phase 1 | A phase I study combining sacituzumab govitecan (SG) and enfortumab vedotin (EV) for metastatic urothelial cancer showed a 70% objective response rate, including three complete responses. Maximum tolerated doses were determined, and treatment was generally well tolerated with support from G-CSF. These promising results suggest potential for further studies. |
2024 | [160] | Phase 2 | Disitamab vedotin (RC48-ADC) demonstrated a 50.5% objective response rate in patients with HER2-positive locally advanced or metastatic urothelial cancer, with a median progression-free survival of 5.9 months and overall survival of 14.2 months. The treatment was well tolerated with manageable adverse events, making it a promising therapy for this patient population. |
2024 | [161] | Phase 2 | The LUX-Bladder 1 trial evaluated afatinib, an irreversible ErbB family inhibitor, in patients with ERBB1–3 mutations in advanced urothelial cancer. The objective response rate was 5.9–12.5%, and progression-free survival was 9.8–7.8 weeks. Tumors with ERBB2 amplification or the basal-squamous phenotype showed better responses. Common adverse events included diarrhea, with 9.5% of patients experiencing grade 3 events. |
3.8.3. Selected Clinical Trials from 2024
A phase II study evaluated the association of molecular subtypes with pathologic response, progression-free survival (PFS), and overall survival (OS) in patients with muscle-invasive bladder cancer treated with neoadjuvant chemotherapy (NAC). The aim of the study was to use the COXEN (Co-expression Extrapolation) gene expression model to identify patients who would best respond to NAC. The COXEN model included specific results for two chemotherapy regimens: methotrexate, vinblastine, adriamycin, and cisplatin (ddMVAC) and gemcitabine/cisplatin (GC). The study included 237 patients who were randomly assigned to receive 4 cycles of ddMVAC (51% of patients) or GC (49% of patients). Based on transcriptomic data obtained from the Affymetrix platform, patients were divided into molecular subtypes using three classifiers: TCGA (five subtypes), Consensus (six subtypes), and MD Anderson (three subtypes). The aim of the analysis was to assess whether molecular subtypes could predict response to NAC and whether they increased the predictive value of the COXEN model. Of the participants, 155 patients had available gene expression data, had received at least 3 of 4 NAC cycles, and underwent radical cystectomy, which allowed for the assessment of pathological response. The analysis noted that the TCGA classifier divided into three groups (basal squamous, neuronal, and luminal infiltrating) and the COXEN score for the GC regimen showed the highest values of the area under the curve (AUC) for complete pathological response (pT0). In contrast, the Consensus classifier, which had three subtype categories (basal squamous/neuroendocrine, luminal, and stroma-rich), slightly improved the prediction of pathological downstaging (<pT2). The AUC increased from 0.57 (using only stratification factors) to 0.61, suggesting a modest benefit from adding this classifier. Despite these results, no statistically significant associations were observed between molecular subtypes and progression-free survival (PFS) or overall survival (OS). The results of the study suggest that although molecular subtypes may have some prognostic value in terms of pathological response to chemotherapy, they do not show strong correlations with long-term clinical outcomes, such as PFS and OS. The conclusions of the study indicate the need for further research to better understand the role of molecular subtypes as predictive biomarkers in the treatment of bladder cancer [
132].
The PrECOG PrE0807 study is a phase 1b study designed to evaluate the feasibility and safety of neoadjuvant nivolumab, either as monotherapy or in combination with lirilumab, in patients with muscle-invasive bladder cancer (MIBC) who are ineligible for or refuse cisplatin-based chemotherapy. Neoadjuvant cisplatin-based chemotherapy before radical cystectomy (RC) has been shown to improve overall survival in MIBC, but many patients are unable to receive this treatment, creating a need to seek alternative therapies. In the study, patients with cT2-4aN0-1M0 MIBC who were ineligible for cisplatin were administered two doses of nivolumab (480 mg) before RC. Patients were divided into two cohorts: the first received nivolumab alone, and the second received the combination of nivolumab with lirilumab (240 mg). The primary objective of the study was to determine the incidence of grade 3 or higher treatment-related adverse events (TRAEs) according to the Terminology Criteria for Adverse Events (CTCAE) version 5.0. Additionally, the proportion of patients who underwent RC more than 6 weeks after the last dose of treatment, changes in CD8+ T-cell density between transurethral resection of bladder tumor (TURBT) and RC, ypT0N0 and <ypT2N0 rates, and 2-year relapse-free survival (RFS) and overall survival (OS) were analyzed. The study included 43 patients, 41 of whom underwent planned RC. There was no grade 3 TRAE in the nivolumab monotherapy group, whereas grade 3 TRAEs occurred in 7% of patients in the combination group, but all of these events resolved, and no grade 4 or 5 TRAEs were reported. There were also no delays in RC beyond 6 weeks. The ypT0N0 rates were 17% and 21% in the two cohorts, respectively, and the <ypT2N0 rates were 25% and 32%. The two-year relapse-free survival was 73% and 71%, and the two-year overall survival was 82% and 89%, respectively. Importantly, a significant increase in CD8+ T-cell density was observed in the nivolumab plus lirilumab cohort. The study showed that nivolumab-based neoadjuvant immunotherapy was well tolerated and safe in patients with MIBC who were not eligible for cisplatin. Although the ypT0N0 rates were lower than in other studies, the two-year survival rates were comparable to those in other studies of neoadjuvant immunotherapy. These results suggest that nivolumab may be an effective alternative for patients who are ineligible for cisplatin-based chemotherapy, which is currently being studied in another clinical trial (NCT03661320) [
133].
Another phase I study evaluated the safety and efficacy of combining two antibody–drug conjugates (ADCs): sacituzumab govitecan (SG) and enfortumab vedotin (EV), in the treatment of metastatic urothelial cancer (mUC). Both SG and EV are standard monotherapies in the treatment of mUC, but because of their different mechanisms of action and molecular targets, we decided to investigate whether their combination could provide additional clinical benefit. The study included patients with metastatic urothelial cancer who had disease progression despite prior platinum-based therapy and/or immunotherapy. A key inclusion criterion was having an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, meaning that patients had to be in relatively good general condition. The therapy consisted of the administration of a combination of SG and EV on days 1 and 8 of a 21-day cycle. Treatment was continued until disease progression or toxicity that prevented further treatment. The primary objective of the study was to assess the incidence of dose-limiting toxicities (DLTs) during the first treatment cycle, with the aim of establishing the maximum tolerated dose (MTD) for this therapeutic regimen. The study used a Bayesian Optimal Interval model, which allows for flexible dose adjustments depending on adverse events. Response rates (ORR), progression-free survival (PFS), and overall survival (OS) were also assessed. Between May 2021 and April 2023, 24 patients were enrolled in the study, 23 of whom started therapy. The median age was 70 years, and 11 patients had received at least 3 prior lines of therapy. Grade 3 or higher adverse events occurred in 78% of patients (18/23) regardless of the assigned dose, including one fatal event (pneumonia, probably related to enfortumab vedotin). Based on the data obtained, the recommended doses for Phase II are 8 mg/kg for SG and 1.25 mg/kg for EV, respectively, with additional support from granulocyte-stimulating factor (G-CSF). The maximum tolerated doses were established at 10 mg/kg for SG and 1.25 mg/kg for EV. The objective response rate was 70%, with 16 patients achieving a response (95% confidence interval: 47–87%), including 3 complete responses. Three patients had disease progression as their best response. The median follow-up was 14 months, and 9 patients had ongoing responses, including 6 for more than 12 months. In conclusion, the combination of sacituzumab govitecan and enfortumab vedotin in the treatment of metastatic urothelial cancer showed promising results, especially in terms of high response rates and a significant number of complete clinical responses. These results suggest the therapeutic potential of this combination and indicate the need for further studies in subsequent clinical phases [
134].
A clinical trial evaluated the efficacy and safety of disitamab vedotin (DV, RC48-ADC), a novel humanized antibody against human epidermal growth factor receptor 2 (HER2), in patients with locally advanced or metastatic urothelial cancer (UC) that overexpressed HER2. Disitamab vedotin, an antibody conjugate of the cytotoxic monomethyl auristatin E, was evaluated in two open-label, multicenter, single-arm phase II studies (RC48-C005 and RC48-C009). The studies included patients with HER2-positive urothelial cancer that had progressed after at least one line of systemic chemotherapy. HER2 positivity was defined as an immunohistochemical score of 3+ or 2+. Patients received DV at a dose of 2 mg/kg every other week as an intravenous infusion. The primary endpoint was the objective response rate (ORR), which was assessed by an independent expert review committee (BIRC). Additionally, progression-free survival (PFS), overall survival (OS), and the safety profile of the drug were analyzed. A total of 107 patients were included in the study. The ORR was confirmed to be 50.5% (95% CI 40.6–60.3), indicating promising efficacy of the drug. These results were consistent across subgroups, including patients with liver metastases and those previously treated with checkpoint inhibitors (PD-1/PD-L1). The median duration of response was 7.3 months (95% CI 5.7–10.8), and the median PFS and OS were 5.9 months (95% CI 4.3–7.2) and 14.2 months (95% CI 9.7–18.8), respectively. The most common treatment-related adverse events included peripheral sensory neuropathy (68.2%), leukopenia (50.5%), aspartate aminotransferase (AST) elevation (42.1%), and neutropenia (42.1%). Grade 3 or higher adverse events occurred in 54.2% of patients, with the most common being sensory neuropathy (18.7%) and neutropenia (12.1%). The study showed that disitamab vedotin has significant efficacy and an acceptable safety profile in the treatment of patients with locally advanced or metastatic HER2-positive urothelial cancer who had failed prior systemic therapies. The results suggest that DV may be an important treatment option in this group of patients [
135]. A phase II clinical trial (LUX-Bladder 1, NCT02780687) evaluated the efficacy and safety of afatinib, an irreversible ErbB family inhibitor, in patients with advanced urothelial cancer with ERBB1–3 mutations. The study was open-label, single-arm, and participants received afatinib at a dose of 40 mg once daily as a second-line treatment. The primary objective of the study was to assess 6-month progression-free survival (PFS6) in patients with metastatic urothelial cancer. The study included 34 patients in cohort A and 8 patients in cohort B. The results showed that the PFS6 rate was 11.8% in cohort A and 12.5% in cohort B, respectively. The objective response rate (ORR) was 5.9% in cohort A and 12.5% in cohort B, and the disease control rate (DCR) was 50.0% and 25.0%, respectively. The median progression-free survival (PFS) was 9.8 weeks in cohort A and 7.8 weeks in cohort B, and the median overall survival (OS) was 30.1 weeks and 29.6 weeks, respectively. The study observed a partial response in three patients, two of whom had ERBB2 amplification in cohort A and one with EGFR amplification in cohort B. Despite promising results in some patients, phase 2 of the study in cohort A was not conducted due to lack of sufficient antitumor activity at interim analysis. In terms of safety, all patients experienced adverse events, most commonly diarrhea, which occurred in 76.2% of patients, 9.5% of whom had grade 3 diarrhea. Two treatment discontinuations were related to adverse events, and one adverse event resulted in death due to acute coronary syndrome, although this event was not considered treatment related. Biomarker analysis showed that tumors with a basal-squamous phenotype and those with ERBB2 amplification may have a better response to afatinib, suggesting the need for further investigation in these patient subgroups [
136].