Next Article in Journal
Optimizing Anti-PD1 Immunotherapy: An Overview of Pharmacokinetics, Biomarkers, and Therapeutic Drug Monitoring
Next Article in Special Issue
Redefining Prostate Cancer Precision: Radiogenomics, Theragnostics, and AI-Driven Biomarkers
Previous Article in Journal
“Are You Just Looking to ‘Survive’?”: A Qualitative Study of Importance of Oncology Endpoints Beyond Overall Survival in Early-Stage Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Beyond Visualization: Advanced Imaging, Theragnostics and Biomarker Integration in Urothelial Bladder Cancer

by
Eduardo Albers Acosta
1,*,†,
Lira Pelari Mici
1,†,
Carlos Márquez Güemez
1,
Clara Velasco Balanza
1,
Manuel Saavedra Centeno
1,
Marta Pérez Pérez
1,
Guillermo Celada Luis
1,
Cristina Quicios Dorado
1,
José Daniel Subiela
2,
Rodrigo España Navarro
3,
Patricia Toquero Diez
4,
Nuria Romero Laorden
4 and
Luis San José Manso
1
1
Department of Urology, Hospital Universitario de La Princesa, IIS Princesa, Universidad Autónoma de Madrid, 28006 Madrid, Spain
2
Department of Urology, Hospital Universitario Ramón y Cajal, IRYSIS, Universidad de Alcalá, 28034 Madrid, Spain
3
Department of Urology, Hospital Regional Universitario de Málaga, TAUS CLINICS, 29010 Málaga, Spain
4
Department of Medical Oncology, Hospital Universitario de La Princesa, IIS Princesa, Universidad Autónoma de Madrid, 28006 Madrid, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2025, 17(19), 3261; https://doi.org/10.3390/cancers17193261
Submission received: 5 September 2025 / Revised: 4 October 2025 / Accepted: 7 October 2025 / Published: 8 October 2025

Abstract

Simple Summary

Bladder cancer frequently recurs and may progress. Emerging tools—such as positron emission tomography (PET) tracers beyond fluorodeoxyglucose (FDG), artificial intelligence (AI)-assisted cystoscopy, theragnostics, and liquid biopsy biomarkers—are reshaping how this disease is detected, staged, and treated. This narrative review summarizes recent advances and discusses their clinical implications. Integrating molecular imaging, AI, and biomarker data holds the potential to deliver more precise and genuinely personalized care.

Abstract

Background/Objectives: Bladder cancer is characterized by high recurrence and progression rates, posing a challenge to current diagnostic and treatment strategies. This review aims to provide a comprehensive overview of emerging technologies, including novel PET tracers, AI-assisted cystoscopy, theragnostics, and molecular biomarkers. Methods: We performed a narrative review of the recent literature focusing on innovations in imaging, AI, theragnostics, and biomarker research relevant to bladder cancer diagnosis and management. Results: Several novel PET tracers, such as 68Ga-PSMA and fibroblast activation protein inhibitor (FAPI), demonstrated potential in improving detection sensitivity. AI-enhanced cystoscopy has shown promise in real-time lesion detection, while theragnostic agents enable combined diagnostic and therapeutic applications. Advances in molecular biomarkers, including circulating Tumor DNA (ctDNA) and gene expression signatures, offer new avenues for patient stratification and monitoring. Conclusions: Integration of advanced imaging, AI, theragnostics, and biomarker analysis may transform bladder cancer management, supporting personalized and more effective care strategies.

1. Introduction

Bladder cancer is the tenth most common malignancy worldwide, with approximately 570,000 new cases and 210,000 deaths each year [1]. Despite improvements in surgical and systemic therapies, recurrence and progression remain major clinical challenges, reflecting the biological heterogeneity of the disease [2]. Conventional diagnostic tools, such as computed tomography (CT) and white-light cystoscopy (WLC), are limited in sensitivity, specificity, and tumor characterization, particularly for small or flat lesions, highlighting the need for innovative, non-anatomical approaches [3].
Recent advances in molecular imaging, theragnostic agents, artificial intelligence (AI)-assisted cystoscopy, and biomarker research are reshaping the diagnostic and therapeutic landscape of urothelial carcinoma [4]. These developments enable a deeper understanding of tumor biology, support personalized treatment strategies, and may help overcome the limitations of conventional imaging. This review summarizes key innovations in imaging, theragnostics, and biomarker integration, providing a concise, practice-oriented overview of how these technologies may advance precision oncology in bladder cancer.

2. Materials and Methods

This work is a narrative review aimed at summarizing the latest developments in molecular imaging, theragnostics, and biomarker integration in urothelial cancer. A structured literature search was performed in PubMed, Embase, and the Cochrane Library up to May 2025 using combinations of the keywords “bladder cancer”, “urothelial carcinoma”, “molecular imaging”, “theragnostics”, “PET/CT”, “artificial intelligence”, and “liquid biopsy”. Articles published in English were included if they reported relevant clinical, translational, or imaging-based findings. Priority was given to peer-reviewed original studies, meta-analyses, and high-quality reviews. References for selected papers were also screened for additional relevant publications.
As this was a narrative (non-systematic) review, no formal quantitative analysis or quality assessment was performed. The focus was to provide a critical and integrated synthesis of the most relevant and recent evidence to support clinical and translational understanding.
Two reviewers screened studies independently and resolved discrepancies by consensus. Data extracted comprised study design, population, modality/algorithm, tracers or biomarkers, main outcomes, and clinical implications. Owing to study heterogeneity, findings were synthesized narratively. Key methodological elements and evidence strength are summarized in Table 1.

3. Results

3.1. Advances in Molecular Imaging and Emerging PET Tracers in Bladder Cancer

Positron emission tomography with [18F] fluorodeoxyglucose (FDG) has been a cornerstone in many solid malignancies, but its role in bladder cancer is limited by urinary excretion and low specificity in adjacent tissues [5]. Recent advances have focused on developing radiopharmaceuticals that improve tumor characterization and allow the identification of therapeutic targets.

3.1.1. Fibroblast Activation Protein Inhibitors (FAPI)

Among novel tracers, fibroblast activation protein inhibitor (FAPI) tracers have gained attention due to high tumor uptake and low background signal. In bladder cancer, Gallium-68-labeled FAPI PET/CT has demonstrated utility in lymphatic and haematogenous staging of high-grade tumors, with superior sensitivity for subcentimeter metastases compared with conventional imaging, although urinary excretion still limits primary-tumor assessment [6,7].
Compared with FDG, FAPI consistently shows higher tumor-to-background ratios (5.3–5.6 vs. 1.9–1.95; p = 0.001) and increased detection rates of metastases (~30%), particularly in lymph nodes, lungs and peritoneum, including lesions missed by FDG or CT [8,9]. In a prospective pilot study with [68Ga]FAPI-2286, tracer uptake exceeded FDG (SUVmax 9.9 ± 3.4 vs. 4.2 ± 1.9; p < 0.0001), correlated with lesion size (p = 0.001), and altered surgical planning in three cases [10]. Preoperatively, [68Ga]FAPI-46 PET/CT achieved 95.2% specificity and 93% negative predictive value for nodal staging, supporting its role in tailoring lymphadenectomy [11]. A recent systematic review confirmed these advantages for small nodal and peritoneal metastases while emphasizing the need for optimized protocols for primary-tumor evaluation [7]. Overall, FAPI tracers show consistently higher lesion detectability than FDG and could become valuable tools for nodal and peritoneal staging once larger, standardized studies confirm their performance.

3.1.2. Alternative Modalities and Computational Tools

Beyond tracer innovation, hybrid imaging and AI-based analysis are expanding diagnostic capabilities in bladder cancer.
  • [11C]-Acetate PET/Magnetic Resonance Imaging (MRI): The ACEBIB trial evaluated Carbon-11-labeled acetate ([11C]-acetate) PET/MRI as an alternative tracer for staging and treatment monitoring in bladder cancer. [11C]-acetate targets lipid metabolism, which is upregulated in many tumors, and offers the advantage of minimal urinary excretion compared to FDG, reducing interference in bladder imaging. In this multicenter study, the technique achieved 100% sensitivity for detecting muscle-invasive disease and showed reasonable correlation with response to neoadjuvant chemotherapy. However, its sensitivity for nodal metastases was limited (20%), underscoring the need for further validation before clinical adoption [12].
  • AI applied to PET/FDG: To improve the limited accuracy of FDG PET/CT in nodal staging, machine learning has been explored as a complementary tool. In one study, a random forest model was developed using three objective parameters—Maximum Standardized Uptake Value (SUVmax) of the most avid lymph node, product of diameters of the largest node, and primary tumor size. This model achieved higher diagnostic accuracy than expert consensus in the training cohort (area under curve (AUC) 0.87 vs. 0.73; p = 0.048). Although performance did not hold in the independent validation set (AUC 0.59 vs. 0.64; p = 0.54), interrater agreement was excellent (kappa = 0.66), suggesting that AI-based approaches could provide reproducible, standardized support for lymph node assessment in muscle-invasive bladder cancer [13].
These findings come mainly from small, single-center studies; prospective multicenter validation will be needed before these alternative tracers and AI tools can be adopted in routine practice.

3.1.3. Emerging Molecular Targets for Imaging

Efforts to identify novel imaging targets in bladder cancer have evaluated markers such as Vascular Endothelial Growth Factor (VEGF), Epidermal Growth Factor Receptor (EGFR), and Prostate-Specific Membrane Antigen (PSMA) through immunohistochemistry (IHC) of metastatic lymph nodes. Expression was quantified using a composite immunostaining score (range 0–12) based on staining intensity and proportion of positive cells. VEGF showed moderate to high expression in metastatic nodes (median score 8/12), with acceptable contrast compared to tumor-negative nodes (mean ratio 1.3 ± 0.7), supporting its feasibility for targeted imaging. In contrast, EGFR and PSMA demonstrated only weak staining (median scores 2–3), limiting their potential as imaging targets [14]. VEGF appears feasible for targeted imaging, whereas EGFR and PSMA show limited potential; all remain to be validated in bladder cancer-specific trials.

3.1.4. Nectin-4-Targeted Imaging

As an adhesion molecule overexpressed in urothelial carcinoma, Nectin-4 is of particular interest. In a first-in-human trial, the novel tracer 68Ga-N188 demonstrated strong correlation between PET uptake and IHC expression (SUVmax 8.3 ± 2.4 for strong positive vs. 3.7 ± 1.1 for weak and 1.9 ± 0.4 for negative cases; p < 0.001). ROC analysis confirmed high predictive value, with AUC 0.967, sensitivity 88.1%, and specificity 100% at an SUVmax threshold of 2.85 [15]. However, these encouraging data are still limited to a first-in-human pilot; larger bladder cancer-specific studies are needed to confirm clinical utility.
Table 2 summarizes recent advances in radiopharmaceuticals designed to improve tumor characterization and identify therapeutic targets.
This table shows novel tracers that improve sensitivity for detecting metastatic spread and reduce urinary interference, yet heterogeneity across studies and the lack of large prospective trials still limit their integration into routine clinical practice.

3.2. Enhanced Cystoscopic Evaluation Through AI and Optical Imaging

White-light cystoscopy (WLC) remains the gold standard for bladder cancer diagnosis. While it offers good sensitivity for papillary tumors, its performance in detecting carcinoma in situ and small or flat lesions is suboptimal, contributing to incomplete resections and underdiagnosis [16,17]. Up to 10–20% of tumors may be missed with WLC [18], and incomplete Transurethral Resection of Bladder Tumors (TURBT) has been reported in nearly 50% of cases [19], favoring early recurrence and progression.

3.2.1. Advances in Optical Imaging and the Role of AI

To address these limitations, enhanced optical modalities (imaging techniques that augment conventional white light cystoscopy) have been developed. The most established examples are fluorescence cystoscopy and narrow-band imaging (NBI), both of which improve the visualization of inconspicuous lesions and show superior sensitivity compared to WLC alone. Evidence from two meta-analyses reported sensitivity rates of approximately 90% for fluorescence and NBI, compared to around 75% for WLC [18,20]. However, clinical uptake remains limited due to logistical, economic, and training barriers. More recently, AI has emerged as a complementary tool to improve detection, segmentation, and prognostication, particularly in non-muscle-invasive bladder cancer (NMIBC), where complete TURBT and early diagnosis are critical [3,4,19,20,21].

3.2.2. AI-Based Detection and Real-Time Imaging

Deep learning applied to cystoscopy has markedly improved detection accuracy. Models trained on blue-light images have achieved sensitivities above 91% and specificities over 77% for NMIBC during TURBT [22]. Some groups have gone further by combining optical techniques such as fluorescence and diffuse reflectance spectroscopy with machine learning, allowing real-time characterization of tumor tissue, achieving sensitivity and specificity of 78% and 91%, respectively [23].
Convolutional neural networks (CNNs), which automatically learn image patterns, have shown consistent ability to discriminate tumors from benign lesions, with sensitivities ranging from 85 to 90% and specificities from 89 to 94% in retrospective analyses [24,25]. More advanced architectures, including high-resolution CNN models, have achieved even higher performance, with sensitivities of 91.6–94.8% and precisions of 91.3–94.4%, particularly improving detection of flat lesions such as carcinoma in situ [26]. Their robustness has been confirmed in multicenter evaluations, where one system reported an AUC of 0.96 [27], and another (INF-M01) achieved 97.3% sensitivity, 92.1% specificity, and 93.4% overall accuracy in routine clinical practice, effectively serving as a reliable second observer [28]. These results show that deep learning and combined optical–AI approaches can reach diagnostic accuracies comparable to expert observers and bring real-time tumor characterization within reach of routine TURBT.

3.2.3. Tumor Segmentation and Dual-Function Models

Precise visualization of tumor margins is essential for complete resection. AI-based segmentation models enhance image contrast and boundary clarity, allowing more accurate identification of tumor borders, even in early disease, with reported accuracies of around 91% in retrospective analyses [29]. Systems such as U-Net have shown high precision in outlining lesions during cystoscopy [30], while simplified versions designed for lower computational demands make real-time use feasible in standard clinical settings [31]. More advanced dual-function models can not only delineate the tumor but also classify its type (normal, papillary, or flat), reaching over 91% accuracy. Their boundary detection closely matches expert performance (Intersection over Union (IoU) 0.833; binary accuracy 95.1%), and early evidence suggests they could also support automated tumor grading in a single step [32]. These advances suggest that AI-based segmentation and dual-function models could soon support surgeons in achieving more complete resections and standardized tumor classification in routine practice.

3.2.4. Grading, Biological Behavior, and Real-Time Augmentation

AI systems are also being developed to predict tumor grade and biological behavior directly from cystoscopic images [33]. By analyzing color and texture features, these models have achieved performance comparable to experienced clinicians, with one study reporting a balanced accuracy of 81%, sensitivity of 88%, specificity of 74%, and an AUC of 92% [34,35]. Beyond static classification, AI is increasingly integrated into live cystoscopy, where real-time augmentation platforms enhance visualization under suboptimal lighting, reduce false positives, raise specificity to as high as 98.8%, and minimize interobserver variability [36,37,38]. Altogether, these advances show that AI can help make cystoscopy more accurate and consistent in everyday practice.
An overview of studies combining AI with cystoscopy is presented in Table 3.
This table illustrates the rapid evolution from basic detection systems to advanced segmentation and grading approaches, as well as the methodological diversity that still limits clinical translation.

3.3. Experimental Theragnostic Approaches in Bladder Cancer

3.3.1. From Immunotherapy to Immuno-Theragnostics

Bladder cancer, characterized by high mutational load and immune evasion, has seen immune checkpoint inhibitors (ICIs) such as anti-PD-1 and anti-PD-L1 become essential in advanced disease or cisplatin-ineligible patients [39,40,41,42,43]. Building on this, theragnostic strategies now aim to visualize and treat immune-related targets simultaneously. Immuno-PET allows in vivo imaging of PD-L1 and CD8+ T cells, enabling non-invasive patient selection, monitoring of immune dynamics, and anticipation of response [44,45]. To date, most of these approaches remain preclinical or exploratory in solid tumors other than bladder cancer, with no disease-specific prospective human trials yet available. These early data are promising, but further bladder cancer-specific validation will be needed.

3.3.2. Immune-Related Targets

ICIs are central to NMIBC unresponsive to Bacillus Calmette–Guérin (BCG) and remain integral in muscle invasive bladder cancer (MIBC) and advanced stages [41,42,43]. Response rates are higher in patients with elevated PD-L1 expression, though long-term durability remains a challenge [46,47,48]. For patients refractory to both platinum chemotherapy and ICIs, theragnostic applications are under investigation, linking imaging tracers to therapeutic agents directed against PD-1, PD-L1, and other tumor-associated markers such as Nectin-4 and TROP-2 [49,50]. Radiolabeled antibodies and small-molecule tracers for PD-L1 are in early-phase immuno-PET trials, offering real-time, non-invasive quantification of expression, though none are currently bladder cancer-specific [51,52]. Thus, while conceptually attractive, these strategies remain speculative in urothelial carcinoma.

3.3.3. Nectin-4 and TROP-2: Companion Diagnostics and Antibody-Drug Conjugates

Nectin-4 and TROP-2 are clinically validated therapeutic targets. The Antibody-drug conjugates enfortumab vedotin (anti-Nectin-4) and sacituzumab govitecan (anti-TROP-2) have achieved objective response rates of 31–44% in advanced or heavily pretreated bladder cancer, including patients post-ICI therapy [53,54,55]. Evidence is strongest in advanced MIBC, with no robust support yet for NMIBC or long-term outcomes from phase III studies. Expression patterns support precision medicine: Nectin-4 is most abundant in squamous and plasmacytoid variants, while PD-L1 is more common in sarcomatoid tumors [40,56]. In parallel, PET tracers targeting Nectin-4 have demonstrated strong correlation with immunohistochemistry, but these findings are still limited to preclinical work and exploratory human data, with no validated bladder cancer-specific trials available.

3.4. Liquid Biopsy and Circulating Biomarkers

Liquid biopsy offers minimally invasive, real-time monitoring through circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), Ribonucleic Acid (RNAs), and proteins [57].

3.4.1. CTCs

The detection of CTCs increases with disease burden, reported in 10.3% of NMIBC, 28.6% of MIBC, 40% of nodal disease, and up to 75% of metastatic cases [58]. Reviews and meta-analyses confirm their correlation with tumor stage and grade, with pooled sensitivity of 35% and specificity of 97% [59,60].
Prognostic studies further support their relevance. A 2025 meta-analysis demonstrated that CTC positivity after TURBT significantly predicted recurrence, with a hazard ratio for progression from T1 to T2 of 3.36 (95% CI: 2.68–3.25) [61]. In a prospective cohort of 84 patients, elevated postoperative CTCs correlated with high Ki-67 expression and poorer survival outcomes [62]. Similarly, in a multicenter study of 273 radical cystectomy patients, CTC-negative patients experienced lower relapse and cancer-specific mortality, while CTC-positive patients who received neoadjuvant therapy achieved improved survival, highlighting their potential role as predictive biomarkers [63]. These studies indicate that CTC detection reflects disease burden and progression risk, and may help identify patients most likely to benefit from perioperative therapy.

3.4.2. ctDNA

Because ctDNA turns over rapidly in plasma, it can capture short-term changes in tumor burden and treatment effect, supporting real-time monitoring [64]. In MIBC, a 2025 systematic review showed that preoperative ctDNA positivity was associated with shorter recurrence-free survival and more advanced disease, while postoperative detection correlated with reduced disease-free Survival (DFS) and higher recurrence. Importantly, ctDNA clearance during neoadjuvant chemotherapy (NAC) aligned with treatment response, and longitudinal ctDNA dynamics mirrored efficacy during immunotherapy [65]. A 2024 meta-analysis (16 studies; 1725 patients) reported that elevated baseline ctDNA predicted worse DFS (Hazard Ratio (HR) = 6.26; 95% CI: 3.71–10.58) and overall survival (OS) (HR = 4.23; 95% CI: 2.72–6.57); conversely, on-treatment declines in ctDNA were associated with improved survival, and specific alterations (e.g., FGFR3 mutations) further refined prognostic stratification [66]. Taken together, ctDNA levels and dynamics provide a real-time measure of tumor burden and treatment response, offering a powerful prognostic and monitoring tool once validated in larger cohorts.

3.4.3. RNA Biomarkers

  • mRNA-based assays: Several commercial urine-based assays using messenger RNA (mRNA) expression have demonstrated robust diagnostic performance in bladder cancer. The CxBladder Monitor (genes: CDK1, CXCR2, HOXA13, IGFBP5, MDK) reported 91% sensitivity, 96% negative predictive value, and an AUC of 0.73. The Xpert BC test (ABL1, ANXA10, CRH, IGF2, UPK1B) achieved 84% sensitivity and 91% specificity (AUC 0.872). Similarly, UROBEST reached 80% sensitivity and 94% specificity (AUC 0.91), while the Oncocyte 43-gene panel achieved 90% sensitivity and 82.5% specificity (AUC 0.91) [67]. Together, these data highlight that mRNA assays are among the most advanced biomarker tools, with some already available for clinical use.
  • miRNAs: MicroRNAs (miRNAs), small regulatory RNAs detectable in urine, have also been explored extensively. A 2022 systematic review of 25 studies (N = 4054) identified multiple miRNA candidates with diagnostic potential, though inter-study variability limited consistent signatures [68]. More recently, a 2024 meta-analysis focusing on miR-143 reported sensitivity 0.80, specificity 0.85, and AUC 0.88, underscoring its potential as a reliable non-invasive marker [69]. It shows promising accuracy but still needs consistent validation.
  • lncRNAs: Long non-coding RNAs (lncRNAs), particularly when carried in urinary exosomes, have also been evaluated. A 2021 meta-analysis (1883 bladder cancer patients vs. 1721 controls) demonstrated pooled sensitivity 0.74, specificity 0.76, and AUC 0.83. Importantly, panels of multiple lncRNAs outperformed single markers (AUC 0.86 vs. 0.81), supporting their clinical utility as part of multiparametric approaches [70].
  • Emerging Biomarkers and Multi-Omic Signatures: In addition to circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs), several new biomarkers are under investigation. These include circular RNAs (circRNAs), the RNA demethylase FTO (fat mass and obesity-associated protein, an epitranscriptomic regulator), and distinct protein or peptide signatures—all linked to bladder cancer biology but still requiring further validation [71]. At the genomic level, mutation panels covering genes such as TP53, FGFR3, ERBB2, PIK3CA, ARID1A and FGFR3-ADD1 fusions may help detect minimal residual disease and monitor relapse. These emerging biomarkers could improve early detection and monitoring of bladder cancer once validated.
To improve detection of minimal residual disease, two main approaches are being explored. Tumor-informed panels, designed from each patient’s own tumor sequence, provide higher sensitivity and specificity. In contrast, tumor-agnostic panels use predefined gene sets without prior sequencing of the individual tumor; they are faster and easier to deploy but more prone to false positives [72].
Similarly, fragmentomics refers to the analysis of the size and pattern of circulating DNA fragments. Approaches such as EPIC-seq use these fragmentation patterns to infer gene expression from ctDNA, showing promise even in patients with low tumor burden.
Because bladder cancer is highly heterogeneous, a single biomarker is often not enough. Multi-omic approaches combine different types of biological information such as genetic changes (genomics), gene activity (transcriptomics), proteins (proteomics) and epigenetic marks (epigenomics) to give a more complete picture of the disease and improve risk stratification. Using this kind of analysis, a 2025 study identified sex-specific prognostic signatures: RAD51C, COL22A1 and COL5A2 in females, and DAXX, IKBKB, PDGFRA and PPARG in males, many of which are linked to immune pathways [73].
Other combined approaches also show promise. For example, Surface-Enhanced Raman Spectroscopy (SERS) with urinary miRNAs increased diagnostic accuracy (AUC 0.92–0.97) [74]. Urinary proteins such as elastase, S100A12, p53, and kallikrein-6 have been correlated with tumor stage and immune signaling [75]. Similarly, DNA methylation assays of urinary exfoliated cells achieved 89% sensitivity and 92% specificity for bladder cancer detection [76].
Importantly, systematic reviews confirm that combining biomarkers enhances diagnostic performance, particularly in early-stage or recurrent disease. This principle is being tested in the NEO-BLAST trial, which evaluates a multimodal strategy combining urinary tumor DNA (utDNA), plasma ctDNA, imaging, and repeat TURBT to guide bladder preservation after NAC. Patients with negative ctDNA/utDNA and complete response are followed under active surveillance, while discordant profiles may proceed to cystectomy [77].
Altogether, these findings underscore the growing relevance of multi-marker and multi-omic approaches, which may overcome the limitations of single assays and move clinical practice closer to true precision oncology in bladder cancer.

3.4.4. Integration with Imaging

Radiomics converts imaging data into quantifiable features; while radiogenomics links these patterns to genomic and transcriptomic profiles. A 2025 review found radiogenomic nomograms predicted outcomes with sensitivity 0.83 and specificity 0.815 [78]. Integration with liquid biopsy adds further predictive value: in advanced urothelial carcinoma, changes in ctDNA allele fraction (ΔaVAF) predicted progression 92 days before radiology and improved survival estimation [79].
An overview of landmark studies in biomarkers and imaging integration appears in Table 4.
This table summarizes recent advances aimed at improving diagnostic accuracy, prognostic assessment, and therapeutic decision-making. Most studies highlight the potential of ctDNA and CTCs as clinically relevant biomarkers that could support more personalized and precise management of patients with bladder cancer.

4. Discussion

Molecular imaging in bladder cancer is undergoing a paradigm shift with the development of novel PET tracers, computational tools, and immune-targeted theragnostics. To illustrate the complementary role of these modalities, we have added a summary flowchart (Figure 1) that outlines the diagnostic–therapeutic pathway in bladder cancer, integrating AI-assisted cystoscopy, molecular imaging, theragnostics, and liquid biopsy.
The limitations of FDG in bladder imaging (primarily its urinary excretion) have driven the exploration of alternative tracers such as FAPI. These radiopharmaceuticals provide improved tumor-to-background ratios and enhanced sensitivity for small-volume metastases, particularly in lymph nodes and peritoneal deposits [6,7,8,9]. Beyond improving staging, FAPI-based PET may influence treatment planning by refining surgical strategies and systemic therapy selection. Correlation with treatment response and recurrence risk, coupled with high specificity in nodal assessment [11], positions FAPI as a potential cornerstone in bladder cancer imaging. However, its limited performance in primary tumor assessment, due to urinary excretion, remains a barrier and highlights the need for optimized acquisition protocols [7].
Alongside new tracers, the integration of AI into PET/CT interpretation shows potential to standardize image assessment and reduce interobserver variability, although further validation in prospective, multicenter studies is still required [13]. In parallel, tumor-specific targets such as Nectin-4 and VEGF are emerging as promising avenues for molecular imaging and therapeutic selection. The strong correlation between 68Ga-N188 PET and IHC-determined Nectin-4 expression, with near-perfect specificity, supports its role as a companion diagnostic [15], while VEGF has shown moderate discriminatory ability. Conversely, EGFR and PSMA appear less suitable for bladder cancer imaging [14].
Although results with novel PET tracers are encouraging, most studies are still early-phase or pilot projects with small, heterogeneous cohorts, which limits generalizability and firm conclusions about clinical value. Standardized acquisition protocols, unified interpretation criteria, and head-to-head comparisons across tracers are largely absent. Robust cost-effectiveness data are also lacking. Large, prospective multicenter trials will be essential to validate these tracers, harmonize methodology, and support their integration into routine care.
At the cystoscopic level, WLC remains the standard but is hampered by a miss rate of up to 20% for flat and small lesions and incomplete TURBT rates approaching 50%. Both factors contribute to recurrence and progression [16,17,18,19]. Enhanced optical modalities such as fluorescence cystoscopy and NBI improve detection but face adoption barriers related to logistics, costs, and training [3,18,20]. Recent advances in AI are shifting this paradigm: deep learning models trained on cystoscopic images, particularly under blue-light conditions, now achieve sensitivities above 91% and specificities over 77% for NMIBC detection [22]. Beyond detection, AI tools assist in tumor margin delineation and segmentation [23,24,25,26,27,28,29,30,31,32], provide accurate grading comparable to experts [34,35], and even enhance real-time visualization during TURBT. These platforms reduce false positives and interobserver variability [36,37].
Despite its promise, AI-assisted cystoscopy faces major challenges: limited transparency of algorithms, variable image quality, and small, homogeneous training datasets that restrict generalizability and clinician trust. Data privacy and regulatory barriers hamper creation of large annotated repositories for external validation. Most evidence comes from retrospective, single-center cohorts, and algorithms often lose accuracy when applied to different populations or devices. Successful adoption will require standardized imaging protocols, hardware compatibility, urologist training, cost-effectiveness data, and prospective multicenter trials to demonstrate real-world benefit [33,38].
The field of theragnostics represents a further frontier, particularly relevant in urothelial carcinoma with its variable immune landscape. Immuno-PET enables the in vivo visualization of PD-L1, offering spatially and temporally resolved information beyond the static snapshot of IHC. Although promising for patient selection and monitoring immunotherapy response, no PD-L1 PET tracers have yet been tested in bladder cancer-specific trials. This gap underscores an important research gap. Meanwhile, antibody-drug conjugates targeting Nectin-4 and TROP-2, such as enfortumab vedotin and sacituzumab govitecan, have shown objective response rates of 31–44% in advanced or pretreated populations [53,54]. Expression patterns further support precision approaches: Nectin-4 is enriched in squamous and plasmacytoid variants, while PD-L1 predominates in sarcomatoid tumors [40,56].
Theragnostic strategies and immuno-PET are promising for precision oncology but face significant barriers, including intratumoral heterogeneity, resistance mechanisms, toxicity, cost, and the lack of validated biomarkers to guide patient selection [50,83]. Evidence is largely preclinical or from early-phase, single-center studies, with little bladder cancer-specific validation [42]. Antibody–drug conjugates show activity, but data are mostly limited to phase I/II trials with scarce real-world or long-term outcomes [53,54,55]. Regulatory hurdles for novel radiopharmaceuticals further delay translation, underscoring the need for standardized frameworks and robust, prospective, multicenter studies before routine use.
Parallel advances in biomarkers and liquid biopsy are transforming bladder cancer management toward precision medicine. Multiple analytes—including ctDNA, CTCs, RNAs (miRNAs, lncRNAs, circRNAs), proteins, exosomes, and metabolites—can be detected in blood and urine, providing minimally invasive tools for dynamic monitoring [57,59]. Clinically, biomarkers show promise for diagnosis, particularly in high-grade disease, and for follow-up, serving as adjuncts in NMIBC recurrence detection. They also have potential for prognosis, with markers such as p53, FGFR3, CYFRA 21-1, and ERCC1 being linked to disease progression and treatment response, although routine adoption still requires validation [81,84].
Among circulating biomarkers, ctDNA has emerged as one of the most informative. Its detection predicts recurrence and survival, often anticipating metastatic relapse by up to three months compared to imaging [84,85]. Urinary tumor DNA (utDNA) further complements ctDNA, with high sensitivity for minimal residual disease and the capacity to inform on tumor grade and stage, thereby supporting bladder-sparing strategies [80].
Radiogenomic approaches may strengthen biomarker-imaging integration, while clinical trials such as NEO-BLAST (combining utDNA, plasma ctDNA, imaging, and repeat TURBT) exemplify how multimodal strategies could redefine bladder preservation protocols [77,78]. Recent clinical trials have confirmed that combining imaging-based classifications with biomarker panels can improve treatment decisions in bladder cancer. For example, VI-RADS, an MRI scoring system that classifies the stage and extent of bladder cancer together with molecular profiling, has shown strong predictive value. The PURE-01 study is the most advanced illustration: in patients with muscle-invasive bladder cancer treated with neoadjuvant pembrolizumab, integrating VI-RADS scores (before and after treatment) with genomic profiling allowed the highly accurate prediction of pathological response (pT ≤ 1 N0) and survival [86]. Yet, tumor heterogeneity, limited sensitivity in early stages, and the absence of international consensus on optimal biomarker panels persist as key obstacles [87,88].
Biomarkers and liquid biopsy approaches hold great promise but are still mainly supported by small, observational, single-center studies, limiting generalizability. Differences in study design, sample processing and assay platforms complicate cross-study comparison [86]. Prospective multicenter trials with standardized protocols and predefined endpoints are still needed. Broader implementation also faces regulatory, cost and infrastructure barriers, as well as unequal access to advanced sequencing platforms and specialized training necessary for reproducibility and widespread adoption.

5. Conclusions

Bladder cancer management is entering a transformative era driven by molecular imaging, AI, theragnostics, and biomarker integration. Together, these innovations go beyond conventional visualization, enabling real-time, personalized guidance across diagnosis, treatment, and surveillance.
Next-generation PET tracers, such as FAPI, improve sensitivity for nodal and peritoneal disease, while immune-targeted probes like Nectin-4 and PD-L1 lay the foundation for theragnostic strategies. However, challenges remain in optimizing protocols for primary tumor visualization and expanding access to disease-specific tracers, requiring multicenter validation and cost-effectiveness analyses.
AI-based cystoscopic platforms are rapidly advancing, with deep learning systems achieving expert-level accuracy in detection, segmentation, and grading. These tools have the potential to standardize TURBT, reduce recurrence, and enhance surgical decision-making. Yet, issues of transparency, dataset diversity, and clinical validation still limit translation to practice, highlighting the need for pragmatic implementation studies across diverse healthcare systems.
Liquid biopsy and molecular biomarkers, particularly ctDNA, utDNA, and RNA panels, offer minimally invasive ways to track tumor dynamics and anticipate therapeutic responses earlier than imaging. Their integration with radiomics and multi-omic approaches could redefine risk stratification and patient selection. Still, heterogeneity and a lack of assay standardization remain major obstacles to routine adoption, emphasizing the importance of prospective, biomarker-driven clinical trials.
The convergence of imaging, AI, and molecular biomarkers represents a paradigm shift in uro-oncology. Clinical impact will depend on multicenter validation, international harmonization, and well-designed trials conducted in real-world settings.
Importantly, their combined use offers a holistic framework where complementary strengths of PET tracers, AI-assisted cystoscopy, liquid biopsy, and theragnostic approaches can advance precision oncology in bladder cancer. By bridging these domains, bladder cancer care can move toward true precision medicine—where therapeutic choices are guided by continuous, integrated molecular and imaging insights.

Author Contributions

Conceptualization, E.A.A.; Methodology, E.A.A., L.P.M. and C.M.G.; Investigation, E.A.A., L.P.M., C.M.G., C.V.B., M.S.C., M.P.P., G.C.L., C.Q.D., J.D.S., R.E.N., P.T.D., N.R.L. and L.S.J.M.; Data curation, E.A.A. and L.P.M.; Writing—original draft preparation, E.A.A., L.P.M. and C.M.G.; Writing—review and editing, E.A.A., L.P.M. and C.M.G.; Visualization, M.P.P.; Supervision, E.A.A. and L.P.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The original contributions presented in this study are included in the article; further inquiries can be directed to the corresponding author.

Acknowledgments

The authors acknowledge the use of AI-assisted tools (OpenAI) for minor language editing. The authors also thank the Department of Urology of Hospital Universitario de La Princesa and the Instituto de Investigación Sanitaria—IP for their support during the preparation of this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AIArtificial Intelligence
CTComputed tomography
PETPositron emission tomography
MRIMagnetic resonance imaging
AUCArea under curve
BCaBladder Cancer
BLCBlue Light Cystoscopy
CADComputer-Aided Detection
CTCsCirculating Tumor Cells
ctDNACirculating Tumor DNA
CUETOClub Urológico Español de Tratamiento Oncológico
DxDiagnosis
DFSDisease-free Survival
EORTCEuropean Organisation for Research and Treatment of Cancer
FDGFluorodeoxyglucose
FAPIFibroblast Activation Protein Inhibitor
IoUIntersection over Union
mpMRIMultiparametric magnetic resonance imaging
MIBCMuscle-Invasive Bladder Cancer
NACNeoadjuvant Chemotherapy
NBINarrow Band Imaging
NMIBCNon-Muscle-Invasive Bladder Cancer
OSOverall Survival
PDDPhotodynamic Diagnosis
PFSProgression-Free Survival
PSMAProstate-Specific Membrane Antigen
RFSRecurrence-Free Survival
RNARibonucleic Acid
SoCStandard of Care
SUVmaxMaximum Standardized Uptake Value
T/N/MTumor/Node/Metastasis
TURBTTransurethral Resection of Bladder Tumor
utDNAUrinary Tumor DNA
WLCWhite Light Cystoscopy
18F-FDG:FDG labeled with fluorine-18
68Ga-FAPIGallium-68-labeled fibroblast activation protein inhibitor
68Ga-FAP-2286A specific gallium-68-labeled FAPI radiotracer
68Ga-N188Gallium-68-labeled radiotracer targeting Nectin-4
11C-acetateCarbon-11-labeled acetate
NPVnegative predictive value
IHCImmunohistochemistry
ADCantibody–drug conjugate
MLMachine learning
TISTotal immunostaining score
SeSensitivity
SpSpecificity
GUgenitourinary
CNNConvolutional neural network
ACSAttention mechanism based Cystoscopic images Segmentation model
FPNFeature pyramid network
HRHazard Ratio
lncRNALong Non-Coding RNA
miRNAMicroRNA
mRNAMessenger RNA
MRDMinimal Residual Disease
SERSSurface-Enhanced Raman Spectroscopy
EGFREpidermal growth factor receptor
VEGFVascular Endothelial Growth Factor
BCGBacillus Calmette–Guérin

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. European Association of Urology. EAU Guidelines: Muscle-Invasive and Metastatic Bladder Cancer; European Association of Urology: Arnhem, The Netherlands, 2025; Available online: https://uroweb.org/guidelines (accessed on 14 September 2025).
  3. Remmelink, M.J.; Rip, Y.; Nieuwenhuijzen, J.A.; Ket, J.C.; Oddens, J.R.; de Reijke, T.M.; de Bruin, D.M. Advanced optical imaging techniques for bladder cancer detection and diagnosis: A systematic review. BJU Int. 2024, 134, 890–905. [Google Scholar] [CrossRef]
  4. Shkolyar, E.; Zhou, S.R.; Carlson, C.J.; Chang, S.; Laurie, M.A.; Xing, L.; Bowden, A.K.; Liao, J.C. Optimizing cystoscopy and TURBT: Enhanced imaging and artificial intelligence. Nat. Rev. Urol. 2024, 22, 46–54. [Google Scholar] [CrossRef] [PubMed]
  5. Xue, M.; Liu, L.; Du, G.; Fu, Z. Diagnostic Evaluation of 18F-FDG PET/CT Imaging in Recurrent or Residual Urinary Bladder Cancer: A Meta-Analysis. Urol. J. 2020, 17, 562–567. [Google Scholar] [CrossRef] [PubMed]
  6. Unterrainer, L.M.; Lindner, S.; Eismann, L.; Casuscelli, J.; Gildehaus, F.-J.; Bui, V.N.; Albert, N.L.; Holzgreve, A.; Beyer, L.; Todica, A.; et al. Feasibility of [68Ga]Ga-FAPI-46 PET/CT for detection of nodal and hematogenous spread in high-grade urothelial carcinoma. Eur. J. Nucl. Med. 2022, 49, 3571–3580. [Google Scholar] [CrossRef] [PubMed]
  7. Hagens, M.J.; van Leeuwen, P.J.; Wondergem, M.; Boellaard, T.N.; Sanguedolce, F.; Oprea-Lager, D.E.; Bex, A.; Vis, A.N.; van der Poel, H.G.; Mertens, L.S.; et al. A Systematic Review on the Diagnostic Value of Fibroblast Activation Protein Inhibitor PET/CT in Genitourinary Cancers. J. Nucl. Med. 2024, 65, 888–896. [Google Scholar] [CrossRef]
  8. Novruzov, E.; Dendl, K.; Ndlovu, H.; Choyke, P.L.; Dabir, M.; Beu, M.; Novruzov, F.; Mehdi, E.; Guliyev, F.; Koerber, S.A.; et al. Head-to-head Intra-individual Comparison of [68Ga]-FAPI and [18F]-FDG PET/CT in Patients with Bladder Cancer. Mol. Imaging Biol. 2022, 24, 651–658. [Google Scholar] [CrossRef]
  9. Ortolan, N.; Urso, L.; Zamberlan, I.; Filippi, L.; Buffi, N.M.; Cittanti, C.; Uccelli, L.; Bartolomei, M.; Evangelista, L. Is There a Role for FAPI PET in Urological Cancers? Mol. Diagn. Ther. 2024, 28, 721–725. [Google Scholar] [CrossRef]
  10. Koshkin, V.S.; Kumar, V.; Kline, B.; Escobar, D.; Aslam, M.; Cooperberg, M.R.; Aggarwal, R.R.; de Kouchkovsky, I.; Chou, J.; Meng, M.V.; et al. Initial Experience with 68Ga-FAP-2286 PET Imaging in Patients with Urothelial Cancer. J. Nucl. Med. 2024, 65, 199–205. [Google Scholar] [CrossRef]
  11. Unterrainer, L.M.; Eismann, L.; Lindner, S.; Gildehaus, F.-J.; Toms, J.; Casuscelli, J.; Holzgreve, A.; Kunte, S.C.; Cyran, C.C.; Menold, P.; et al. [68Ga]Ga-FAPI-46 PET/CT for locoregional lymph node staging in urothelial carcinoma of the bladder prior to cystectomy: Initial experiences from a pilot analysis. Eur. J. Nucl. Med. 2024, 51, 1786–1789. [Google Scholar] [CrossRef]
  12. Salminen, A.; Jambor, I.; Merisaari, H.; Ettala, O.; Virtanen, J.; Koskinen, I.; Veskimae, E.; Sairanen, J.; Taimen, P.; Kemppainen, J.; et al. 11C-acetate PET/MRI in bladder cancer staging and treatment response evaluation to neoadjuvant chemotherapy: A prospective multicenter study (ACEBIB trial). Cancer Imaging 2018, 18, 25. [Google Scholar] [CrossRef] [PubMed]
  13. Girard, A.; Dercle, L.; Vila-Reyes, H.; Schwartz, L.H.; Girma, A.; Bertaux, M.; Radulescu, C.; Lebret, T.; Delcroix, O.; Rouanne, M. A machine-learning-based combination of criteria to detect bladder cancer lymph node metastasis on [18F]FDG PET/CT: A pathology-controlled study. Eur. Radiol. 2023, 33, 2821–2829. [Google Scholar] [CrossRef] [PubMed]
  14. van der Fels, C.A.M.; Leliveld, A.; Buikema, H.; Heuvel, M.C.v.D.; de Jong, I.J. VEGF, EGFR and PSMA as possible imaging targets of lymph node metastases of urothelial carcinoma of the bladder. BMC Urol. 2022, 22, 213. [Google Scholar] [CrossRef] [PubMed]
  15. Duan, X.; Xia, L.; Zhang, Z.; Ren, Y.; Pomper, M.G.; Rowe, S.P.; Li, X.; Li, N.; Zhang, N.; Zhu, H.; et al. First-in-Human Study of the Radioligand 68Ga-N188 Targeting Nectin-4 for PET/CT Imaging of Advanced Urothelial Carcinoma. Clin. Cancer Res. 2023, 29, 3395–3407. [Google Scholar] [CrossRef]
  16. Shkolyar, E.; Jia, X.; Chang, T.C.; Trivedi, D.; Mach, K.E.; Meng, M.Q.-H.; Xing, L.; Liao, J.C. Augmented Bladder Tumor Detection Using Deep Learning. Eur. Urol. 2019, 76, 714–718. [Google Scholar] [CrossRef]
  17. Daneshmand, S.; Bazargani, S.T.; Bivalacqua, T.J.; Holzbeierlein, J.M.; Willard, B.; Taylor, J.M.; Liao, J.C.; Pohar, K.; Tierney, J.; Konety, B. Blue light cystoscopy for the diagnosis of bladder cancer: Results from the US prospective multicenter registry. Urol. Oncol. Semin. Orig. Investig. 2018, 36, 361.e1–361.e6. [Google Scholar] [CrossRef]
  18. Russo, G.I.; Sholklapper, T.N.; Cocci, A.; Broggi, G.; Caltabiano, R.; Smith, A.B.; Lotan, Y.; Morgia, G.; Kamat, A.M.; Witjes, J.A.; et al. Performance of Narrow Band Imaging (NBI) and Photodynamic Diagnosis (PDD) Fluorescence Imaging Compared to White Light Cystoscopy (WLC) in Detecting Non-Muscle Invasive Bladder Cancer: A Systematic Review and Lesion-Level Diagnostic Meta-Analysis. Cancers 2021, 13, 4378. [Google Scholar] [CrossRef]
  19. Ben Muvhar, R.; Paluch, R.; Mekayten, M. Recent Advances and Emerging Innovations in Transurethral Resection of Bladder Tumor (TURBT) for Non-Muscle Invasive Bladder Cancer: A Comprehensive Review of Current Literature. Res. Rep. Urol. 2025, 17, 69–85. [Google Scholar] [CrossRef]
  20. Fan, Z.; Shi, H.; Luo, J.; Guo, X.; Wang, B.; Liu, Y.; Yu, J. Diagnostic and therapeutic effects of fluorescence cystoscopy and narrow-band imaging in bladder cancer: A systematic review and network meta-analysis. Int. J. Surg. 2023, 109, 3169–3177. [Google Scholar] [CrossRef]
  21. Zhu, M.; Gu, Z.; Chen, F.; Chen, X.; Wang, Y.; Zhao, G. Application of artificial intelligence in the diagnosis and treatment of urinary tumors. Front. Oncol. 2024, 14, 1440626. [Google Scholar] [CrossRef]
  22. Ali, N.; Bolenz, C.; Todenhöfer, T.; Stenzel, A.; Deetmar, P.; Knoll, T.; Porubsky, S.; Hartmann, A.; Popp, J.; Kriegmair, M.C.; et al. Deep learning-based classification of blue light cystoscopy imaging during transurethral resection of bladder tumors. Sci. Rep. 2021, 11, 11629. [Google Scholar] [CrossRef]
  23. Zlobina, N.V.; Budylin, G.S.; Tseregorodtseva, P.S.; Andreeva, V.A.; Sorokin, N.I.; Kamalov, D.M.; Strigunov, A.A.; Armaganov, A.G.; Kamalov, A.A.; Shirshin, E.A. In vivo assessment of bladder cancer with diffuse reflectance and fluorescence spectroscopy: A comparative study. Lasers Surg. Med. 2024, 56, 496–507. [Google Scholar] [CrossRef]
  24. Mutaguchi, J.; Morooka, K.; Kobayashi, S.; Umehara, A.; Miyauchi, S.; Kinoshita, F.; Inokuchi, J.; Oda, Y.; Kurazume, R.; Eto, M. Artificial Intelligence for Segmentation of Bladder Tumor Cystoscopic Images Performed by U-Net with Dilated Convolution. J. Endourol. 2022, 36, 827–834. [Google Scholar] [CrossRef]
  25. Ikeda, A.; Nosato, H.; Kochi, Y.; Kojima, T.; Kawai, K.; Sakanashi, H.; Murakawa, M.; Nishiyama, H. Support System of Cystoscopic Diagnosis for Bladder Cancer Based on Artificial Intelligence. J. Endourol. 2020, 34, 352–358. [Google Scholar] [CrossRef] [PubMed]
  26. Ye, Z.; Li, Y.; Sun, Y.; He, C.; He, G.; Ji, Z. Leveraging Deep Learning in Real-Time Intelligent Bladder Tumor Detection During Cystoscopy: A Diagnostic Study. Ann. Surg. Oncol. 2025, 32, 3220–3226. [Google Scholar] [CrossRef] [PubMed]
  27. Wu, S.; Chen, X.; Pan, J.; Dong, W.; Diao, X.; Zhang, R.; Zhang, Y.; Zhang, Y.; Qian, G.; Chen, H.; et al. An Artificial Intelligence System for the Detection of Bladder Cancer via Cystoscopy: A Multicenter Diagnostic Study. JNCI J. Natl. Cancer Inst. 2021, 114, 220–227. [Google Scholar] [CrossRef] [PubMed]
  28. Kim, J.; Ham, W.S.; Koo, K.C.; Lee, J.; Ahn, H.K.; Jeong, J.Y.; Baek, S.Y.; Lee, S.J.; Lee, K.S. Evaluation of the Diagnostic Efficacy of the AI-Based Software INF-M01 in Detecting Suspicious Areas of Bladder Cancer Using Cystoscopy Images. J. Clin. Med. 2024, 13, 7110. [Google Scholar] [CrossRef]
  29. Li, L.; Jiang, L.; Yang, K.; Luo, B.; Wang, X. A novel artificial intelligence segmentation model for early diagnosis of bladder tumors. Abdom. Imaging 2024, 50, 3092–3099. [Google Scholar] [CrossRef]
  30. Zhang, Q.; Liang, Y.; Zhang, Y.; Tao, Z.; Li, R.; Bi, H. A comparative study of attention mechanism based deep learning methods for bladder tumor segmentation. Int. J. Med. Inform. 2023, 171, 104984. [Google Scholar] [CrossRef]
  31. Zhao, X.; Lai, L.; Li, Y.; Zhou, X.; Cheng, X.; Chen, Y.; Huang, H.; Guo, J.; Wang, G. A lightweight bladder tumor segmentation method based on attention mechanism. Med. Biol. Eng. Comput. 2024, 62, 1519–1534. [Google Scholar] [CrossRef]
  32. Hwang, W.K.; Jo, S.B.; Han, D.E.; Ahn, S.T.; Oh, M.M.; Park, H.S.; Moon, D.G.; Choi, I.; Yang, Z.; Kim, J.W. Artificial Intelligence-Based Classification and Segmentation of Bladder Cancer in Cystoscope Images. Cancers 2024, 17, 57. [Google Scholar] [CrossRef] [PubMed]
  33. Baana, M.; Arkwazi, M.; Zhao, Y.; Ofagbor, O.; Bhardwaj, G.; Lami, M.; Bolton, E.; Heer, R. Using artificial intelligence for bladder cancer detection during cystoscopy and its impact on clinical outcomes: A protocol for a systematic review and meta-analysis. BMJ Open 2024, 14, e089125. [Google Scholar] [CrossRef] [PubMed]
  34. Yoo, J.W.; Koo, K.C.; Chung, B.H.; Baek, S.Y.; Lee, S.J.; Park, K.H.; Lee, K.S. Deep learning diagnostics for bladder tumor identification and grade prediction using RGB method. Sci. Rep. 2022, 12, 17699. [Google Scholar] [CrossRef] [PubMed]
  35. Lee, J.Y.; Lee, Y.S.; Tae, J.H.; Chang, I.H.; Kim, T.-H.; Myung, S.C.; Nguyen, T.T.; Lee, J.H.; Choi, J.; Kim, J.H.; et al. Selection of Convolutional Neural Network Model for Bladder Tumor Classification of Cystoscopy Images and Comparison with Humans. J. Endourol. 2024, 38, 1036–1043. [Google Scholar] [CrossRef]
  36. Jia, X.; Shkolyar, E.; Laurie, M.A.; Eminaga, O.; Liao, J.C.; Xing, L. Tumor detection under cystoscopy with transformer-augmented deep learning algorithm. Phys. Med. Biol. 2023, 68, 165013. [Google Scholar] [CrossRef]
  37. Chang, T.C.; Shkolyar, E.; Del Giudice, F.; Eminaga, O.; Lee, T.; Laurie, M.; Seufert, C.; Jia, X.; Mach, K.E.; Xing, L.; et al. Real-time Detection of Bladder Cancer Using Augmented Cystoscopy with Deep Learning: A Pilot Study. J. Endourol. 2023, 37, 747–854. [Google Scholar] [CrossRef]
  38. Negassi, M.; Suarez-Ibarrola, R.; Hein, S.; Miernik, A.; Reiterer, A. Application of artificial neural networks for automated analysis of cystoscopic images: A review of the current status and future prospects. World J. Urol. 2020, 38, 2349–2358. [Google Scholar] [CrossRef]
  39. Vitiello, A.; Ferrara, F.; Lasala, R.; Zovi, A. Precision Medicine in the Treatment of Locally Advanced or Metastatic Urothelial Cancer: New Molecular Targets and Pharmacological Therapies. Cancers 2022, 14, 5167. [Google Scholar] [CrossRef]
  40. Sardana, R.; Mishra, S.K.; Williamson, S.R.; Mohanty, A.; Mohanty, S.K. Immune checkpoints and their inhibitors: Reappraisal of a novel diagnostic and therapeutic dimension in the urologic malignancies. Semin. Oncol. 2020, 47, 367–379. [Google Scholar] [CrossRef]
  41. Bednova, O.; Leyton, J.V. Targeted Molecular Therapeutics for Bladder Cancer—A New Option beyond the Mixed Fortunes of Immune Checkpoint Inhibitors? Int. J. Mol. Sci. 2020, 21, 7268. [Google Scholar] [CrossRef]
  42. Wang, Y.; He, M.; Li, J.; Li, L. Immunotherapeutic strategies for invasive bladder cancer: A comprehensive review. Front. Immunol. 2025, 16, 1591379. [Google Scholar] [CrossRef]
  43. Michaud, É.; Mansure, J.J.; Kassouf, W. Integrating novel immunotherapeutic approaches in organ-preserving therapies for bladder cancer. Br. J. Pharmacol. 2024; early view. [Google Scholar] [CrossRef]
  44. Dercle, L.; Sun, S.; Seban, R.-D.; Mekki, A.; Sun, R.; Tselikas, L.; Hans, S.; Bernard-Tessier, A.; Bouvier, F.M.; Aide, N.; et al. Emerging and Evolving Concepts in Cancer Immunotherapy Imaging. Radiology 2023, 306, e239003. [Google Scholar] [CrossRef]
  45. Hughes, D.J.; Subesinghe, M.; Taylor, B.; Bille, A.; Spicer, J.; Papa, S.; Goh, V.; Cook, G.J.R. 18F FDG PET/CT and Novel Molecular Imaging for Directing Immunotherapy in Cancer. Radiology 2022, 304, 246–264. [Google Scholar] [CrossRef] [PubMed]
  46. Necchi, A.; Roumiguié, M.; Kamat, A.M.; Shore, N.D.; Boormans, J.L.; Esen, A.A.; Lebret, T.; Kandori, S.; Bajorin, D.F.; Krieger, L.E.M.; et al. Pembrolizumab monotherapy for high-risk non-muscle-invasive bladder cancer without carcinoma in situ and unresponsive to BCG (KEYNOTE-057): A single-arm, multicentre, phase 2 trial. Lancet Oncol. 2024, 25, 720–730. [Google Scholar] [CrossRef] [PubMed]
  47. Huang, S.; Huang, Y.; Li, C.; Liang, Y.; Huang, M.; Luo, R.; Liang, W. Efficacy and safety of neoadjuvant PD-1 inhibitors or PD-L1 inhibitors for muscle invasive bladder cancer: A systematic review and meta-analysis. Front. Immunol. 2024, 14, 1332213. [Google Scholar] [CrossRef] [PubMed]
  48. Beckabir, W.; Zhou, M.; Lee, J.S.; Vensko, S.P.; Woodcock, M.G.; Wang, H.-H.; Wobker, S.E.; Atassi, G.; Wilkinson, A.D.; Fowler, K.; et al. Immune features are associated with response to neoadjuvant chemo-immunotherapy for muscle-invasive bladder cancer. Nat. Commun. 2024, 15, 4448. [Google Scholar] [CrossRef]
  49. Germanà, E.; Pepe, L.; Pizzimenti, C.; Ballato, M.; Pierconti, F.; Tuccari, G.; Ieni, A.; Giuffrè, G.; Fadda, G.; Fiorentino, V.; et al. Programmed Cell Death Ligand 1 (PD-L1) Immunohistochemical Expression in Advanced Urothelial Bladder Carcinoma: An Updated Review with Clinical and Pathological Implications. Int. J. Mol. Sci. 2024, 25, 6750. [Google Scholar] [CrossRef]
  50. Roviello, G.; Catalano, M.; Nobili, S.; Santi, R.; Mini, E.; Nesi, G. Focus on Biochemical and Clinical Predictors of Response to Immune Checkpoint Inhibitors in Metastatic Urothelial Carcinoma: Where Do We Stand? Int. J. Mol. Sci. 2020, 21, 7935. [Google Scholar] [CrossRef]
  51. Leung, D.; Bonacorsi, S.; Smith, R.A.; Weber, W.; Hayes, W. Molecular Imaging and the PD-L1 Pathway: From Bench to Clinic. Front. Oncol. 2021, 11, 698425. [Google Scholar] [CrossRef]
  52. Abenavoli, E.M.; Linguanti, F.; Calabretta, R.; Bolton, R.C.D.; Berti, V.; Lopci, E. Clinical Application of ImmunoPET Targeting Checkpoint Inhibitors. Cancers 2023, 15, 5675. [Google Scholar] [CrossRef]
  53. Koshkin, V.S.; Henderson, N.; James, M.; Natesan, D.; Freeman, D.; Nizam, A.; Su, C.T.; Khaki, A.R.; Osterman, C.K.; Glover, M.J.; et al. Efficacy of enfortumab vedotin in advanced urothelial cancer: Analysis from the Urothelial Cancer Network to Investigate Therapeutic Experiences (UNITE) study. Cancer 2021, 128, 1194–1205. [Google Scholar] [CrossRef]
  54. Chou, J.; Trepka, K.; Sjöström, M.; Egusa, E.A.; Chu, C.E.; Zhu, J.; Chan, E.; Gibb, E.A.; Badura, M.L.; Contreras-Sanz, A.; et al. TROP2 Expression Across Molecular Subtypes of Urothelial Carcinoma and Enfortumab Vedotin-resistant Cells. Eur. Urol. Oncol. 2022, 5, 714–718. [Google Scholar] [CrossRef] [PubMed]
  55. Loriot, Y.; Petrylak, D.; Kalebasty, A.R.; Fléchon, A.; Jain, R.; Gupta, S.; Bupathi, M.; Beuzeboc, P.; Palmbos, P.; Balar, A.; et al. TROPHY-U-01, a phase II open-label study of sacituzumab govitecan in patients with metastatic urothelial carcinoma progressing after platinum-based chemotherapy and checkpoint inhibitors: Updated safety and efficacy outcomes. Ann. Oncol. 2024, 35, 392–401. [Google Scholar] [CrossRef] [PubMed]
  56. Martini, D.J.; Case, K.B.; Gratz, D.; Pellegrini, K.; Beagle, E.; Schneider, T.; Dababneh, M.; Nazha, B.; Brown, J.T.; Joshi, S.S.; et al. PD-L1 and nectin-4 expression and genomic characterization of bladder cancer with divergent differentiation. Cancer 2024, 130, 3658–3670. [Google Scholar] [CrossRef] [PubMed]
  57. Wan, X.; Wang, D.; Zhang, X.; Xu, M.; Huang, Y.; Qin, W.; Chen, S. Unleashing the power of urine-based biomarkers in diagnosis, prognosis and monitoring of bladder cancer (Review). Int. J. Oncol. 2025, 66, 1–22. [Google Scholar] [CrossRef]
  58. Lu, J.J.; Kakehi, Y.; Takahashi, T.; Wu, X.X.; Yuasa, T.; Yoshiki, T.; Okada, Y.; Terachi, T.; Ogawa, O. Detection of circulating cancer cells by reverse transcription-polymerase chain reaction for uroplakin II in peripheral blood of patients with urothelial cancer. Clin. Cancer Res. 2000, 6, 3166–3171. [Google Scholar]
  59. Lodewijk, I.; Dueñas, M.; Rubio, C.; Munera-Maravilla, E.; Segovia, C.; Bernardini, A.; Teijeira, A.; Paramio, J.M.; Suárez-Cabrera, C. Liquid Biopsy Biomarkers in Bladder Cancer: A Current Need for Patient Diagnosis and Monitoring. Int. J. Mol. Sci. 2018, 19, 2514. [Google Scholar] [CrossRef]
  60. Zhang, Z.; Fan, W.; Deng, Q.; Tang, S.; Wang, P.; Xu, P.; Wang, J.; Yu, M. The prognostic and diagnostic value of circulating tumor cells in bladder cancer and upper tract urothelial carcinoma: A meta-analysis of 30 published studies. Oncotarget 2017, 8, 59527–59538. [Google Scholar] [CrossRef]
  61. Liliana, B.; Zulfiqqar, A.; Mataho, N.L.; Subekti, E. The Use of Circulating Tumor Cells in T1 Stage Non-muscle Invasive Bladder Cancer: A Systematic Review and Meta-Analysis. Urol. Res. Pract. 2025, 50, 343–350. [Google Scholar] [CrossRef]
  62. Liu, J.; Ma, C.; Li, X.; Li, A.; Wang, Z. Circulating tumor cells correlating with Ki-67 predicts the prognosis of bladder cancer patients. Int. Urol. Nephrol. 2022, 55, 309–318. [Google Scholar] [CrossRef] [PubMed]
  63. Beije, N.; de Kruijff, I.; de Jong, J.; Klaver, S.; de Vries, P.; Jacobs, R.; Somford, D.; Slaa, E.T.; van der Heijden, A.; Witjes, J.A.; et al. Circulating tumour cells to drive the use of neoadjuvant chemotherapy in patients with muscle-invasive bladder cancer. ESMO Open 2022, 7, 100416. [Google Scholar] [CrossRef] [PubMed]
  64. Wan, J.C.M.; Massie, C.; Garcia-Corbacho, J.; Mouliere, F.; Brenton, J.D.; Caldas, C.; Pacey, S.; Baird, R.; Rosenfeld, N. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat. Rev. Cancer 2017, 17, 223–238. [Google Scholar] [CrossRef] [PubMed]
  65. Grosso, A.; Cadenar, A.; Pillozzi, S.; Carli, G.; Lipparini, F.; Di Maida, F.; Pichler, R.; Krajewski, W.; Albisinni, S.; Laukhtina, E.; et al. Circulating tumor DNA in muscle-invasive bladder cancer: A systematic review. Actas Urol. Esp. 2025, 49, 501717. [Google Scholar] [CrossRef]
  66. Liu, H.; Chen, J.; Huang, Y.; Zhang, Y.; Ni, Y.; Xu, N.; Zhao, F.; Tang, Y.; Liu, H.; Sun, G.; et al. Prognostic significance of circulating tumor DNA in urothelial carcinoma: A systematic review and meta-analysis. Int. J. Surg. 2024, 110, 3923–3936. [Google Scholar] [CrossRef]
  67. Kavalieris, L.; O’sUllivan, P.; Frampton, C.; Guilford, P.; Darling, D.; Jacobson, E.; Suttie, J.; Raman, J.D.; Shariat, S.F.; Lotan, Y. Performance Characteristics of a Multigene Urine Biomarker Test for Monitoring for Recurrent Urothelial Carcinoma in a Multicenter Study. J. Urol. 2017, 197, 1419–1426. [Google Scholar] [CrossRef]
  68. Grimaldi, A.M.; Lapucci, C.; Salvatore, M.; Incoronato, M.; Ferrari, M. Urinary miRNAs as a Diagnostic Tool for Bladder Cancer: A Systematic Review. Biomedicines 2022, 10, 2766. [Google Scholar] [CrossRef]
  69. Li, J.; Li, H.; Yang, Y.; Sen, Y.; Ye, J. miRNA-143 as a potential biomarker in the detection of bladder cancer: A meta-analysis. Futur. Oncol. 2024, 20, 1275–1287. [Google Scholar] [CrossRef]
  70. Su, Q.; Wu, H.; Zhang, Z.; Lu, C.; Zhang, L.; Zuo, L. Exosome-Derived Long Non-Coding RNAs as Non-Invasive Biomarkers of Bladder Cancer. Front. Oncol. 2021, 11, 719863. [Google Scholar] [CrossRef]
  71. Ren, H.; Zou, L.; Jiang, L.; Zhang, P.; Li, C.; Li, Z.; Niu, H.; Zhang, X.; Liao, H.; Cheng, L.; et al. Prognostic role of circRNAs and RNA methylation enzymes in bladder cancer: A systematic review and meta-analysis of Chinese studies. Urol. Oncol. Semin. Orig. Investig. 2025, 43, 628–638. [Google Scholar] [CrossRef]
  72. Vandekerkhove, G.; Todenhöfer, T.; Annala, M.; Struss, W.J.; Wong, A.; Beja, K.; Ritch, E.; Brahmbhatt, S.; Volik, S.V.; Hennenlotter, J.; et al. Circulating Tumor DNA Reveals Clinically Actionable Somatic Genome of Metastatic Bladder Cancer. Clin. Cancer Res. 2017, 23, 6487–6497. [Google Scholar] [CrossRef] [PubMed]
  73. Wang, Y.; Bhandary, P.; Griffin, K.; Moore, J.H.; Li, X.; Wang, Z.P. Integrative multi-omics study identifies sex-specific molecular signatures and immune modulation in bladder cancer. Front. Bioinform. 2025, 5, 1575790. [Google Scholar] [CrossRef] [PubMed]
  74. Moisoiu, T.; Dragomir, M.P.; Iancu, S.D.; Schallenberg, S.; Birolo, G.; Ferrero, G.; Burghelea, D.; Stefancu, A.; Cozan, R.G.; Licarete, E.; et al. Combined miRNA and SERS urine liquid biopsy for the point-of-care diagnosis and molecular stratification of bladder cancer. Mol. Med. 2022, 28, 39. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, R.; Pagano, I.; Sun, Y.; Murakami, K.; Goodison, S.; Vairavan, R.; Tahsin, M.; Black, P.C.; Rosser, C.J.; Furuya, H. A Diagnostic Gene Expression Signature for Bladder Cancer Can Stratify Cases into Prescribed Molecular Subtypes and Predict Outcome. Diagnostics 2022, 12, 1801. [Google Scholar] [CrossRef]
  76. Pavan, N.; Scaggiante, B. Editorial: Liquid biopsy in the detection and prediction of outcomes in bladder cancer. Front. Oncol. 2024, 14, 1391466. [Google Scholar] [CrossRef]
  77. Lindskrog, S.V.; Strandgaard, T.; Nordentoft, I.; Galsky, M.D.; Powles, T.; Agerbæk, M.; Jensen, J.B.; Alix-Panabières, C.; Dyrskjøt, L. Circulating tumour DNA and circulating tumour cells in bladder cancer—From discovery to clinical implementation. Nat. Rev. Urol. 2025, 22, 590–608. [Google Scholar] [CrossRef]
  78. O’Sullivan, N.J.; Temperley, H.C.; Corr, A.; Meaney, J.F.; Lonergan, P.E.; Kelly, M.E. Current role of radiomics and radiogenomics in predicting oncological outcomes in bladder cancer. Curr. Urol. 2024, 19, 43–48. [Google Scholar] [CrossRef]
  79. Shohdy, K.S.; Villamar, D.M.; Cao, Y.; Trieu, J.; Price, K.S.; Nagy, R.; Tagawa, S.T.; Molina, A.M.; Sternberg, C.N.; Nanus, D.M.; et al. Serial ctDNA analysis predicts clinical progression in patients with advanced urothelial carcinoma. Br. J. Cancer 2022, 126, 430–439. [Google Scholar] [CrossRef]
  80. Lee, J.; Chen, F.; Lopez-Beltran, A.; Necchi, A.; Cimadamore, A.; Spiess, P.E.; Li, R.; Roy-Chowdhuri, S.; Montironi, R.; Golijanin, D.; et al. Urinary Tumor DNA–based Liquid Biopsy in Bladder Cancer Management: A Systematic Review. Eur. Urol. Focus 2025, in press. [Google Scholar] [CrossRef]
  81. Matuszczak, M.; Salagierski, M. Diagnostic and Prognostic Potential of Biomarkers CYFRA 21.1, ERCC1, p53, FGFR3 and TATI in Bladder Cancers. Int. J. Mol. Sci. 2020, 21, 3360. [Google Scholar] [CrossRef]
  82. Lu, Y.; Wu, H.; Jiang, J.; Bao, S.; Ling, C. The value of circulating tumor DNA in the prognostic diagnosis of bladder cancer: A systematic review and meta-analysis. Urol. Int. 2025, 109, 399–411. [Google Scholar] [CrossRef]
  83. Huang, P.; Wang, J.; Yu, Z.; Lu, J.; Sun, Z.; Chen, Z. Redefining bladder cancer treatment: Innovations in overcoming drug resistance and immune evasion. Front. Immunol. 2025, 16, 1537808. [Google Scholar] [CrossRef] [PubMed]
  84. Ahangar, M.; Mahjoubi, F.; Mowla, S.J. Bladder cancer biomarkers: Current approaches and future directions. Front. Oncol. 2024, 14, 1453278. [Google Scholar] [CrossRef]
  85. Christensen, E.; Birkenkamp-Demtröder, K.; Sethi, H.; Shchegrova, S.; Salari, R.; Nordentoft, I.; Wu, H.-T.; Knudsen, M.; Lamy, P.; Lindskrog, S.V.; et al. Early Detection of Metastatic Relapse and Monitoring of Therapeutic Efficacy by Ultra-Deep Sequencing of Plasma Cell-Free DNA in Patients with Urothelial Bladder Carcinoma. J. Clin. Oncol. 2019, 37, 1547–1557. [Google Scholar] [CrossRef]
  86. Brembilla, G.; Basile, G.; Cosenza, M.; Giganti, F.; Del Prete, A.; Russo, T.; Pennella, R.; Lavalle, S.; Raggi, D.; Mercinelli, C.; et al. Neoadjuvant Chemotherapy VI-RADS Scores for Assessing Muscle-invasive Bladder Cancer Response to Neoadjuvant Immunotherapy with Multiparametric MRI. Radiology 2024, 313, e233020. [Google Scholar] [CrossRef]
  87. Kouba, E.; Lopez-Beltran, A.; Montironi, R.; Massari, F.; Huang, K.; Santoni, M.; Chovanec, M.; Cheng, M.; Scarpelli, M.; Zhang, J.; et al. Liquid biopsy in the clinical management of bladder cancer: Current status and future developments. Expert Rev. Mol. Diagn. 2019, 20, 255–264. [Google Scholar] [CrossRef]
  88. Koguchi, D.; Matsumoto, K.; Shiba, I.; Harano, T.; Okuda, S.; Mori, K.; Hirano, S.; Kitajima, K.; Ikeda, M.; Iwamura, M. Diagnostic Potential of Circulating Tumor Cells, Urinary MicroRNA, and Urinary Cell-Free DNA for Bladder Cancer: A Review. Int. J. Mol. Sci. 2022, 23, 9148. [Google Scholar] [CrossRef]
Figure 1. Flowchart illustrates the integration of advanced imaging, AI, theragnostics, and biomarkers across the diagnostic–therapeutic pathway in bladder cancer.
Figure 1. Flowchart illustrates the integration of advanced imaging, AI, theragnostics, and biomarkers across the diagnostic–therapeutic pathway in bladder cancer.
Cancers 17 03261 g001
Table 1. Global methods summary (own elaboration).
Table 1. Global methods summary (own elaboration).
Clinical QuestionDo Novel Molecular Imaging, AI-Assisted Cystoscopy, Theragnostics, and Molecular Biomarkers Improve Detection/Staging/Management vs. SoC in Patients with Bladder Cancer?
Population(1) Humans; (2) Adults; (3) Patients with localized and/or advanced bladder cancer
InterventionNew PET tracers, PET/CT–PET/MRI, radiomics; AI-cystoscopy, NBI; theragnostics; ctDNA/utDNA, CTCs, multi-omics.
ComparisonConventional imaging (CT, MRI, or [18F]FDG PET/CT); expert interpretation alone (for AI studies); standard clinical staging protocols. SoC: WLC ± BLC/NBI (no AI); urine cytology; EORTC/CUETO scores.
OutcomesPrimary: Sensitivity/specificity/AUC, T/N/M accuracy, change-in-management, minimal residual disease (ctDNA/utDNA), RFS/PFS/OS, therapy selection.
Secondary: workflow, inter-reader, safety, cost-effectiveness, feasibility.
Study typesProspective or retrospective clinical studies, systematic reviews, pilot feasibility studies
Databases searchedPubMed, Embase, Cochrane
Search keywordsBladder cancer AND (PET/PET-CT/PET-MRI/FAPI/PSMA/radiomics) OR (cystoscopy AND AI/CAD/BLC/PDD/NBI) OR (theragnostic/radioligand) OR (biomarker/ctDNA/utDNA/CTC).
Manual searchScreening reference lists and journal issues by hand
Inclusion criteriaAdult BCa (NMIBC/MIBC)
Diagnostic/staging & treatment response studies (2015–2025)
Novel PET tracers vs. FDG/conventional imaging
AI applied to PET/CT, MRI, cystoscopy/optical imaging
Quantitative metrics (sens., spec., AUC, SUVmax)
Biomarkers (ctDNA, CTCs, RNA, proteins) for Dx/prognosis/follow-up
Imaging–biomarker integration (radiomics, mpMRI, multimodal)
English/Spanish publications
Exclusion criteriaReviews without patient-level data; case reports, editorials, abstracts without full text, opinion pieces
Abbreviations: AI = Artificial Intelligence; AUC = Area Under the Curve; BCa = Bladder Cancer; BLC = Blue Light Cystoscopy; CAD = Computer-Aided Detection; CT = Computed Tomography; CTCs = Circulating Tumor Cells; CUETO = Club Urológico Español de Tratamiento Oncológico; Dx = Diagnosis; EORTC = European Organisation for Research and Treatment of Cancer; FDG = Fluorodeoxyglucose; FAPI = Fibroblast Activation Protein Inhibitor; MRI = Magnetic Resonance Imaging; mpMRI = Multiparametric MRI; MIBC = Muscle-Invasive Bladder Cancer; NAC = Neoadjuvant Chemotherapy; NBI = Narrow Band Imaging; NMIBC = Non-Muscle-Invasive Bladder Cancer; OS = Overall Survival; PET = Positron Emission Tomography; PDD = Photodynamic Diagnosis; PFS = Progression-Free Survival; PSMA = Prostate-Specific Membrane Antigen; RFS = Recurrence-Free Survival; RNA = Ribonucleic Acid; SoC = Standard of Care; SUVmax = Maximum Standardized Uptake Value; T/N/M = Tumor/Node/Metastasis; TURBT = Transurethral Resection of Bladder Tumor; utDNA = Urinary Tumor DNA; WLC = White Light Cystoscopy.
Table 2. Summary of Key Clinical Studies on Molecular Imaging and PET Tracers in Bladder Cancer.
Table 2. Summary of Key Clinical Studies on Molecular Imaging and PET Tracers in Bladder Cancer.
AuthorsYearStudy TypeMain ObjectivenAdditional Notes
Unterrainer et al. [6]2022ProspectiveDetect nodal/hematogenous spread with 68Ga-FAPI-46 PET/CT15Improved detection over CT; useful for early metastatic staging
Novruzov et al. [8]2022Intra-individual comparisonCompare 68Ga-FAPI vs. 18F-FDG PET/CT in bladder cancer8Superior sensitivity in small nodes/peritoneum
Koshkin et al. [10]2024Prospective
(NCT04621435)
Evaluate 68Ga-FAP-2286 PET/CT in urothelial carcinoma21Altered clinical decisions in 3 patients
Ortolan et al. [9]2024Narrative reviewDiscuss role of FAPI PET in urological cancersEmphasizes diagnostic and theragnostic potential
Unterrainer et al. [11]2024ProspectiveAssess locoregional LN staging with 68Ga-FAPI-46 PET/CT18Useful for surgical planning
Hagens et al. [7]2024Systematic reviewReview diagnostic value of FAPI PET/CT in GU cancers10Encourages protocol optimization
Salminen et al. [12]2018Multicenter prospective
(NCT01918592)
Evaluate 11C-acetate PET/MRI for staging and NAC response22Useful for monitoring response, not nodal staging
Girard et al. [13]2023Retrospective + MLMachine learning model for LN metastasis detection on FDG PET/CT87Objective, reproducible tool
van der Fels et al. [14]2022Immunohistochemical studyAssess VEGF, EGFR, PSMA in nodal mets of bladder cancer48 (LNs)Only VEGF viable for imaging
Duan et al. [15]2023First-in-human
(NCT05321316)
Evaluate 68Ga-N188 PET for Nectin-4 imaging32Promising diagnostic companion for ADCs
Abbreviations: 18F-FDG: FDG labeled with fluorine-18; 68Ga-FAPI: Gallium-68-labeled fibroblast activation protein inhibitor; 68Ga-FAP-2286: A specific gallium-68-labeled FAPI radiotracer; 68Ga-N188: Gallium-68-labeled radiotracer targeting Nectin-4; 11C-acetate: Carbon-11-labeled acetate; PET/CT-MRI: positron emission tomography/magnetic resonance imaging; NAC: neoadjuvant chemotherapy; LN: lymph node (plural: LNs = lymph nodes); SUVmax: maximum standardized uptake value; AUC: area under the curve; NPV: negative predictive value; IHC: immunohistochemistry; ADC: antibody–drug conjugate; ML: machine learning; TIS: Total immunostaining score; Se: Sensitivity; Sp: Specificity; GU: genitourinary; VEGF: Vascular Endothelial Growth Factor; EGFR: Epidermal growth factor receptor.
Table 3. Key Studies on AI-Assisted Cystoscopic Detection and Characterization in Urothelial Bladder Cancer.
Table 3. Key Studies on AI-Assisted Cystoscopic Detection and Characterization in Urothelial Bladder Cancer.
AuthorsYearStudy TypenMain ObjectiveAI/SystemArquitecture/ModelImage Technique
Ye et al. [26]2025Prospective diagnostic accuracy study94Evaluate AI (HRNetV2) for real-time bladder lesion detection during cystoscopy.HRNetV2CNNWLC
Shkolyar et al. [4]2025Review study-Review enhanced imaging and AI use in cystoscopy and TURBT.CystoNet-TCNNWLC
Li et al. [29]2025Retrospective diagnostic study273Develop an AI model (BTS-Net) for bladder tumor segmentation in imaging.BTS-NetSegmentation netWLC
Kim et al. [28]2024Randomized retrospective clinical trial5670Validate AI software INF-M01 for detecting bladder tumors in cystoscopy imagesINF-M01CNNWLC
Zhu et al. [21]2024Narrative review-Review machine learning/deep learning applications in urological cancer diagnosis and treatment.Machine learning and deep learningCNNCT, MRI, US and cystoscopy images
Zlobina et al. [23]2024Observational comparative study21Assess spectroscopy for in vivo bladder cancer detection.Machine learningClassification algorithmsFluorescence and diffuse reflectance spectroscopy
Hwang et al. [32]2024Single-center diagnostic classification and segmentation study.772Use VGG19 to classify bladder lesions and Deeplab v3+ to segment morphological types in cystoscopy images.VGG19 + BeepLab v3+CNN + segmentation netWLC
Lee et al. [35]2024Prospective comparative diagnostic study543Identify the best CNN model (EfficientNetB0) for classifying bladder tumors in cystoscopy images.EfficientNetB0CNNWLC
Zhao el al. [31]2024Diagnostic image-based segmentation study.-Propose NAFF-Net efficient segmentation of bladder tumors in endoscopic imagesNAFF-NetSegmentation netWLC
Jia X et al. [36]2023Prospective pilot diagnostic study67Develop and test CystoNet-T for bladder tumor detection in WLC.CystoNet-TFPNWLC
Chang et al. [37]2023Prospective pilot diagnostic study 50Evaluate real-time CystoNet AI system integrated into live cystoscopy/TURBT video to detect bladder tumors during procedures.CystoNetCNNWLC
Zhang et al. [30]2023Retrospective comparative study-Compare various attention mechanisms to improve bladder tumor segmentation performance in cystoscopic images.Deep learningACSWLC
Mutaguchi et al. [24]2022Retrospective diagnostic segmentation study.120Compare Dilated U-Net vs. standard U-Net for accurate bladder tumor segmentation in cystoscopy images.U-netDilated convolutionWLC
Wu et al. [27]2022Multicenter prospective diagnostic study.10,729Evaluate CAIDS for detecting bladder cancer via cystoscopy, comparing its diagnostic accuracy against standard clinical assessment CAIDS algorithmPyramid Scene Parsing Network (PSPNet)WLC
Yoo et al. [34]2022Retrospective diagnostic study10,991Evaluate Mask R-CNN for tumor detection and grade prediction from WLC and narrow-band images.Deep learningMask R-CNNWLC + NBI
Ali et al. [22]2021Multicenter retrospective diagnostic classification study 216Train and evaluate CNNs to classify malignancy, invasiveness, and grade from BLC.Deep learningCNNBLC
Ikeda et al. [25]2020Retrospective diagnostic study.109Train a GoogLeNet-based CNN to distinguish tumor vs. normal bladder imagesDeep learningCNNWLC
Shkolyar et al. [16]2019Multicenter prospective diagnostic study95Develop and validate CystoNet for automated bladder tumor detection in WLC.CystoNetCNNWLC
Abbreviation: WLC: White-light cystoscopy; CNN: Convolutional neural network; BLC: Blue Light Cystoscopy; ACS: Attention mechanism based Cystoscopic images Segmentation model.; NBI: Narrow Band Images; FPN: Feature pyramid network.
Table 4. Clinical Studies on Biomarkers, Liquid Biopsy, and Imaging Integration in Bladder Cancer.
Table 4. Clinical Studies on Biomarkers, Liquid Biopsy, and Imaging Integration in Bladder Cancer.
AuthorsYearStudy TypeMain ObjectivenKey Statistical FindingsAdditional Notes
Lodewijk et al. [59]2018Narrative reviewTypes, potential and applications of biomarkers in BCa.-Detect a much lower tumor burden Generate significant anxiety and lead to patient overtreatment.
Zhang et al. [60]2017Meta-AnalysisThe prognostic and diagnostic value of CTCs in BCa.30CTCs are an independent predictive indicator of poor outcomes.-
Wan et al. [57]2025ReviewRelevance of urine biomarkers-Advancements and limitations in BCa biomarkers-
Lindskrog et al. [77]2025ReviewPotential of ctDNA and CTC as biomarkers-Several studies have shown clinical potentialCombining biomarkers enhances diagnostic results.
Zulfiqqar et al. [61]2025Systematic Review and Meta AnalysisCTC positivity and recurrence and progression to NMIBC5CTCs enhance prognostic accuracy and therapeutic strategies in NMIBC, CTC positivity after TURBT predicted recurrence
Liu et al. [62]2022ProspectiveThe value of CTCs and Ki-67 in prognosis84Ki-67 high expression associates with high postoperative CTC counts-
Beije et al. [63]2022Multicenter Prospective StudyCTCs to drive the use of NAC in MIBC273CTC+ who received NAC survived longerNegative CTCs alone does not justify withholding NAC.
Grosso et al. [65]2025Systematic ReviewPrognostic role of ctDNA in the perioperative of MIBC8ctDNA+ pre and postoperative associates with poor clinical outcomes.ctDNA could guide NAC management.
Liu et al. [66]2024Meta AnalysisPrognostic significance of ctDNA in BCa1275Strong association of ctDNA dynamic change with survival outcomes.Clinical utility of ctDNA in prognosis
Li et al. [69]2024StudyValue of miRNA-143 in the early detection of BCa4054Sensitivity and specificity were 0.80 and 0.85Coupled with miR-100, it showed better diagnostic power
Vandekerkhove et al. [72]2017StudyctDNA to profile the tumor genome in real-time in BCa51ctDNA provides a practical and cost-effective snapshot of driver gene status in metastatic BCa95% of metastatic patients harboring alterations to TP53, RB1, or MDM2, an 70% in ARID1
Moisoiu et al. [74]2022Retrospective cohortCombined miRNA and SERS for diagnosis and molecular stratification31miRNA profiling synergizes with SERS Discriminate between high-grade and low-grade tumors and between luminal and basal types.
O’Sullivan et al. [78]2025ReviewRadiomics-based nomogram to predict oncological outcomes in BCa Several studies demonstrate the predictive potential of radiogenomicFurther studies are required to validate the results
Shohdy et al. [79]2022Prospective studyctDNA as a dynamic tool for changes in the VAF of genomic alterations53Serial ctDNA analysis predicts disease radiologic progression -
Lee et al. [80]2025ReviewThe current state of utDNA as a marker of BCa-utDNAl improves on most of stages of detection, treatment, and monitoring -
Matuszcz et al. [81]2020ReviewFind the one urine biomarker with the best specificity and sensitivity p53, FGFR3, CYFRA 21-1, and ERCC1 clinical applications-
Yu Lu et al. [82]2025Meta-AnalysisPredictive value of ctDNA detection for disease progression and metastasis risk9ctDNA demonstrates some application value in the prognostic, recurrence and survival.Not enough to replace traditional assessment methods
Abbreviation: BCa = Bladder Cancer; ctDNA = Circulating Tumor DNA; CTCs = Circulating Tumor Cells; NAC = Neoadjuvant chemotherapy; DFS = Disease-Free Survival; HR = Hazard Ratio; IHC = Immunohistochemistry; lncRNA = Long Non-Coding RNA; miRNA = MicroRNA; mRNA = Messenger RNA; MRD = Minimal Residual Disease; NAC = Neoadjuvant Chemotherapy; NMIBC = Non-Muscle-Invasive Bladder Cancer; MIBC = Muscle-Invasive Bladder Cancer; OS = Overall Survival; RFS = Recurrence-Free Survival; SERS = Surface-Enhanced Raman Spectroscopy; utDNA = Urinary Tumor DNA.
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

Albers Acosta, E.; Pelari Mici, L.; Güemez, C.M.; Velasco Balanza, C.; Saavedra Centeno, M.; Pérez Pérez, M.; Celada Luis, G.; Quicios Dorado, C.; Subiela, J.D.; España Navarro, R.; et al. Beyond Visualization: Advanced Imaging, Theragnostics and Biomarker Integration in Urothelial Bladder Cancer. Cancers 2025, 17, 3261. https://doi.org/10.3390/cancers17193261

AMA Style

Albers Acosta E, Pelari Mici L, Güemez CM, Velasco Balanza C, Saavedra Centeno M, Pérez Pérez M, Celada Luis G, Quicios Dorado C, Subiela JD, España Navarro R, et al. Beyond Visualization: Advanced Imaging, Theragnostics and Biomarker Integration in Urothelial Bladder Cancer. Cancers. 2025; 17(19):3261. https://doi.org/10.3390/cancers17193261

Chicago/Turabian Style

Albers Acosta, Eduardo, Lira Pelari Mici, Carlos Márquez Güemez, Clara Velasco Balanza, Manuel Saavedra Centeno, Marta Pérez Pérez, Guillermo Celada Luis, Cristina Quicios Dorado, José Daniel Subiela, Rodrigo España Navarro, and et al. 2025. "Beyond Visualization: Advanced Imaging, Theragnostics and Biomarker Integration in Urothelial Bladder Cancer" Cancers 17, no. 19: 3261. https://doi.org/10.3390/cancers17193261

APA Style

Albers Acosta, E., Pelari Mici, L., Güemez, C. M., Velasco Balanza, C., Saavedra Centeno, M., Pérez Pérez, M., Celada Luis, G., Quicios Dorado, C., Subiela, J. D., España Navarro, R., Toquero Diez, P., Laorden, N. R., & Manso, L. S. J. (2025). Beyond Visualization: Advanced Imaging, Theragnostics and Biomarker Integration in Urothelial Bladder Cancer. Cancers, 17(19), 3261. https://doi.org/10.3390/cancers17193261

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