Next Article in Journal
Focal Boost in Prostate Cancer Radiotherapy: A Review of Planning Studies and Clinical Trials
Next Article in Special Issue
Immune Predictors of Response after Bacillus Calmette–Guérin Treatment in Non-Muscle-Invasive Bladder Cancer
Previous Article in Journal
Pan-Cancer Analysis of Patient Tumor Single-Cell Transcriptomes Identifies Promising Selective and Safe Chimeric Antigen Receptor Targets in Head and Neck Cancer
Previous Article in Special Issue
Immunohistochemical Study of Bladder Cancer Molecular Subtypes and Their Association with PD-L1 Expression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current and Emerging Strategies to Treat Urothelial Carcinoma

by
Berkha Rani
1,
James J. Ignatz-Hoover
1,2,3,
Priyanka S. Rana
1,2,3 and
James J. Driscoll
1,2,3,*
1
Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
2
Division of Hematology & Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
3
Adult Hematologic Malignancies & Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(19), 4886; https://doi.org/10.3390/cancers15194886
Submission received: 29 July 2023 / Revised: 25 September 2023 / Accepted: 25 September 2023 / Published: 8 October 2023
(This article belongs to the Special Issue Targeted Therapy for Bladder Cancer)

Abstract

:

Simple Summary

Urothelial cell carcinoma (UCC) is the ninth most common cancer worldwide and in the US the fourth most common cancer, with ~82,000 new cases (~62,000 men) diagnosed annually leading to ~17,000 deaths/year (~12,000 men). While early-stage cases exhibit more favorable outcomes, the emergence of drug resistance and distant metastasis reduces median overall survival (OS) to 12–15 months. The development of modern genetic and molecular assays to detect high-risk mutations has improved the detection of high-risk disease. Recently, immune therapies have been developed; these demonstrate markedly improved OS rates compared to treatment with chemotherapy alone. However, challenges persist and there remains an urgent, unmet need to develop and advance novel molecular and therapeutic strategies that prevent or overcome drug resistance, to improve patient outcome. Here, we provide an overview of the etiology, diagnostic approach and emerging therapeutic strategies for improving UCC patient quality of life and OS.

Abstract

Urothelial cell carcinoma (UCC, bladder cancer, BC) remains a difficult-to-treat malignancy with a rising incidence worldwide. In the U.S., UCC is the sixth most incident neoplasm and ~90% of diagnoses are made in those >55 years of age; it is ~four times more commonly observed in men than women. The most important risk factor for developing BC is tobacco smoking, which accounts for ~50% of cases, followed by occupational exposure to aromatic amines and ionizing radiation. The standard of care for advanced UCC includes platinum-based chemotherapy and programmed cell death (PD-1) or programmed cell death ligand 1 (PD-L1) inhibitors, administered as frontline, second-line, or maintenance therapy. UCC remains generally incurable and is associated with intrinsic and acquired drug and immune resistance. UCC is lethal in the metastatic state and characterized by genomic instability, high PD-L1 expression, DNA damage-response mutations, and a high tumor mutational burden. Although immune checkpoint inhibitors (ICIs) achieve long-term durable responses in other cancers, their ability to achieve similar results with metastatic UCC (mUCC) is not as well-defined. Here, we discuss therapies to improve UCC management and how comprehensive tumor profiling can identify actionable biomarkers and eventually fulfill the promise of precision medicine for UCC patients.

1. Introduction

1.1. Overview

Urothelial cell carcinoma (UCC, formerly known as transitional cell carcinoma, TCC) is the most common neoplasm of the urinary system. UCC is the most common histologic type of BC and accounts for ~90% of all BC [1,2,3,4]. A number of histologic variants of UCC have been identified, including micropapillary, microcystic, nested, lymphoepithelioma-like, plasmacytoid, sarcomatoid, giant cell, poorly differentiated, lipid rich, clear cell and urothelial carcinoma with divergent differentiation. UCC is defined as the invasion of the cancer cells’ basement membrane or lamina propria or deeper by neoplastic cells of urothelial origin. Invasion is defined as ‘micro invasion’ when the depth of invasion is 2 mm or less [2]. The World Health Organization (WHO) classifies BCs based on differentiation as low grade (grades 1 and 2) or high grade (grade 3) [1]. Distinction between low-grade and high-grade urothelial disease has implications related to risk stratification, patient management and treatment outcome.
Urothelial tumors arise and evolve through divergent genetic and phenotypic pathways [2]. The advent of next-generation sequencing has allowed widespread comprehensive molecular characterization of urothelial tumors and, subsequently, the development of therapies targeting specific molecular pathways implicated in carcinogenesis, e.g., FGFR inhibition, Nectin-4, Trop-2, and HER2 targeting. As these therapies are effective in a second-line setting, they will be advanced in the treatment paradigm to localized and even non-muscle invasive disease. Some tumors progress from urothelial hyperplasia to low-grade, non-invasive superficial papillary tumors. More aggressive variants arise from flat, high-grade carcinoma in situ and progress to invasive tumors or arise de novo as invasive tumors [3,4,5]. These distinct phenotypic variants of urothelial tumors exhibit drastically different biological behavior, responses to treatment and prognoses for patients. The low-grade papillary variant is often multifocal and tends to recur, and infrequently progresses to the muscle invasive stage. In contrast, most invasive variants develop into incurable metastases despite radical cystectomy (RC) and additional modalities. It is evident that UCC variants harbor distinct genetic defects that impact growth control, metastatic potential and treatment decisions, as in Figure 1 in [6]. Low-grade, non-invasive papillary tumors are frequently characterized by activating mutations in HRAS and the fibroblast growth factor receptor 3 gene (FGFR3) [7]. High-grade invasive tumors are characterized by structural and functional defects in the p53 and retinoblastoma protein (Rb) tumor-suppressor pathways [7,8].

1.2. Environmental and Hereditary Risk Factors for Urothelial Cancers

Risk factors for UCC are largely environmental, with smoking by far the most important. Smoking is a major risk factor for BC and accounts for ~50% of cases in both men and women. Current smokers are 4–5 times more likely to develop BC than non-smokers [9,10]. Other environmental risk factors include occupational exposure, radiation, drinking water contaminants, and chronic bladder infections [11]. These factors contribute to an increased risk of developing BC, highlighting the importance of preventative measures, e.g., smoking cessation, and minimizing exposure to harmful substances. The consequences of these risk factors and exposures leads to field changes within the urothelial tract that predispose individuals to the development of recurrent tumors, occurring in new locations in the urothelial tract, a phenomenon referred to as polychronotropism [12]. Genetics are also implicated in an individual’s susceptibility to urothelial BC. Studies have found that individuals with a family history of UCC have a higher relative risk of developing the disease. However, an increased risk cannot be entirely attributed to shared environmental exposures, indicating that genetic factors may play a role in BC development [13]. Hereditary non-polyposis colon cancer syndrome (HNPCC) is caused by mutations in DNA mismatch repair genes and can be identified through microsatellite instability (MSI) or the absence of the corresponding protein by immunostaining. HNPCC is linked to a greater risk of developing BC and upper urinary tract/urothelial cancers (UTUC).

1.3. Novel Approaches for Urothelial Cancers

While clinical management of early-stage UCC has seen significant improvements in the past decade, treatment of locally advanced and mUCC remains difficult [14]. Recently, a number of strategies and therapies have been developed for the treatment of advanced UCC. Noteworthy among these are immune checkpoint inhibitors (ICIs) and antibody-drug conjugates (ADCs) [15]. More traditional approaches, e.g., radiotherapy (RT), as part of trimodal therapy, is an attractive alternative treatment in patients with urothelial muscle-invasive BC (MIBC) [16,17]. Moreover, there is some evidence to suggest the effects of ICIs and RT are potentially synergistic. Photodynamic therapy (PDT) also has merit and is a promising approach. Clinical success can be limited due to the severe hypoxia which promotes ICI resistance [18]. Chemotherapy and hypoxia induce programmed death ligand-1 (PD-L1) overexpression, leading to immunosuppression within the tumor microenvironment (TME). Current approaches do not thoroughly address these defects effectively by a single drug or nano-system. To address this problem, Zhou et al. developed a biomineralization method to construct MB@Bu@MnO2 nanoparticles with a two-step oxygen regulation ability and PD-1/PD-L1 axis cascade-disruption capacity. Manganese dioxide albumin (MnO2@Alb) functions as the drug carrier, butformin (Bu) as a mitochondria-associated oxidative phosphorylation (OXPHOS) disruptor with PD-L1 depression and oxygen reversion ability, and methylene blue (MB) as a PDT drug with programmed cell death protein 1 (PD-1) inhibition capacity. Owing to the tumor-responsive capacity of MB@Bu@MnO2 nanoparticles, Bu and MB were selectively delivered and released in tumors. Hypoxia was reversed by Bu inhibited oxygen consumption, and MnO2 improved oxygen generation. Reactive oxygen species (ROS) generation was enhanced by MB@Bu@MnO2 nanoparticle-mediated PDT owing to reversed tumor hypoxia. The immunosuppression microenvironment was reversed by MB@Bu@MnO2 nanoparticles that enhanced immunogenic cell death and PD-1/PD-L1 axis cascade-disruption, which then promoted T-cell infiltration. Nanosystems could solve the defects of traditional PDT by disrupting the PD-1/PD-L1 axis and reversing the negative effects of hypoxia within the TME [19].

2. Divergent Mechanisms Underlying Urothelial Tumorigenesis and Treatment Resistance

2.1. Invasive and Non-Invasive UCC

The clinical staging of BC requires a multimodal approach that combines histopathology with molecular and imaging studies [20]. Most UCC are a form of non-muscle invasive BC (NMIBC). Urothelial tumors develop along two major, largely independent but somewhat overlapping biological pathways, referred to as papillary and non-papillary or solid [21]. Current management of BC still relies on pathologic staging that does not always reflect the risk for an individual patient. Studies assessing molecular alterations in individual tumors are offering insights into the myriad of cellular pathways that are deregulated in bladder tumorigenesis and progression. Alterations in pathways involved in cell-cycle regulation, apoptosis, cell signaling, angiogenesis and tumor-cell invasion have been shown to influence disease behavior [22]. Noninvasive UCC are further subcategorized into exophytic papillary and carcinoma in situ (CIS) based upon distinct molecular alterations. Exophytic papillary tumors, or Ta tumors, which are a type of noninvasive urothelial cancer, can have varying genetic mutations and are classified as either high or low grade [23,24,25,26,27,28,29]. Low-grade Ta tumors are often associated with mutations involving receptor tyrosine kinase-Ras activation, specifically either HRAS or FGFR3 mutations, while high-grade Ta tumors are typically linked with p16INK4a homozygous deletion and a lower frequency of FGFR3 mutations [30]. In contrast, CIS and invasive tumors have alterations in TP53 and RB1 [31]. Muscle-invasive tumors are characterized by changes in vascular endothelial growth factors (VEGF), cadherins, and matrix metalloproteinases (MMPs) [32].

2.2. Cell Cycle Alterations

UCC is characterized by actionable, genetic alterations in specific molecular pathways that result in uncontrolled cellular proliferation, and that can be pharmacologically targeted [33,34,35]. As illustrated in Table 1, genetic alterations in UCC play a pivotal role in the etiology and progression of the disease. The most well-characterized pathways in UCC are those regulating the cell cycle, particularly the p53 and RB mechanisms, which interact with mediators of apoptosis and gene regulation [7,8,29,36]. Thus, TP53 is encoded on chromosome 17p13.1 and inhibits cell-cycle progression by transcriptionally activating p21WAF1/CIP1. Loss of p21 expression can predict disease progression, and its maintenance can abrogate the effects of altered TP53. Furthermore, TP53 mutations result in p53 inactivation and the rate of TP53 alterations in primary tumors increases progressively from normal urothelium to non-muscle-invasive tumors, to muscle-invasive disease and metastatic nodes [37]; TP53 is genetically altered in 49–54% of cases, while MDM2, which participates in an autoregulatory feedback loop with p53, is genetically altered in 9–11% of cases [34,38]. As a prognostic, TP53 is not clinically established but has been shown to predict recurrence and cancer-specific mortality in muscle-invasive disease.
Rb is a cell-cycle regulatory protein that is encoded by RB1 and that interacts with CDKs and E2F, leading to a transcription of genes required for DNA synthesis, and inactivating RB1 mutations have been confirmed in bladder tumors [7,8,31]. CDKN2A/B, the genes that encode p14 and p16, inhibiting the cyclin-dependent kinases, Cdk4 and Cdk6, are altered in 5–23% of cases of MIBC [25,39]. CCND1 (genetically altered in 10–14% of cases) and CCND3 (genetically altered in 4–11% of cases) encode for cyclins that promote cell-cycle progression through interaction with Cdk4 and Cdk6. CDKN1A, a potent cyclin-dependent kinase inhibitor, is genetically altered in ~14% of cases. How CDK inhibitors, e.g., p21, p16, and p27, negatively regulate CDKs, acting as tumor suppressors is a complex process that involves multiple pathways leading to programmed cell death [38]. Bcl-2, an antiapoptotic member of the Bcl-2 family of proteins, is associated with a poor prognosis in BC patients [37]. A prior study suggested that patients treated with RT for TCC of the bladder that expressed both Bcl-2 and TP53 had worse local control and disease-free, disease-specific OS [25]. CDKN2A is commonly altered in human solid tumors, but prior studies have yielded conflicting evidence regarding the association between CDKN2A genomic alterations and response to ICIs.
Patients harboring CDKN2A genomic alterations were associated with a reduced benefit from ICI therapy and were more likely to have lower PD-L1 expression in tumor-infiltrating immune cells and a less inflammatory immune-TME [39]. Palbociclib, a CDK4/6 inhibitor, was evaluated in a phase II trial of patients with metastatic, platinum-refractory UC with loss of p16 and intact Rb, and was not effective since only 17% of patients achieved PFS at 4 months [40]. Since DNA repair pathways are altered in UC, pre-clinical studies have demonstrated the effectiveness of the combination of CDK 4/6 inhibition with a PARP inhibitor [41]. Trilaciclib, another CDK 4/6 inhibitor, is being evaluated in patients with advanced/metastatic UC receiving chemotherapy followed by avelumab, which is predicted to improve outcomes by protection from myelosuppression of chemotherapy as well as enhancing the immune tumor microenvironment by inducing a transient G1 cell-cycle arrest of hematopoietic cells (NCT04887831). CDKN2A genomic alterations were associated with a reduced benefit from ICI therapy in UC as well as changes in the tumor-immune microenvironment [42,43].
Cell-surface receptors, which play a role in transducing external signals to the nucleus and control gene expression, are deregulated in UCC tumor cells. For example, activating FGFR3 mutations are common in low-grade papillary Ta tumors and BC, leading to increased Ras-mitogen-activated protein kinase (MAPK) [42]. A large cohort of patients with metastatic urothelial cancer were treated with an anti-PD-L1 agent (atezolizumab) and major determinants of clinical outcome were identified [43,44]. Response to treatment was associated with the CD8+ T-effector cell phenotype, with a high neoantigen or tumor mutation burden (TMB), while lack of response was associated with a signature of transforming growth factor β (TGFβ)-signaling in fibroblasts. Other factors that impact cell signaling and gene regulation in BC include the sex hormone receptors, Janus kinase family members, and meiotic recombination 11 (MRE11) [45]. Reduced expression of the estrogen receptor-β (ER-β) has been linked to better progression-free survival (PFS) rates in patients with NIBC, while higher levels have been associated with more advanced tumors. Janus kinase family members, such as STAT3, can predict recurrence and survival in BC patients [46,47,48].

2.3. Influence of Angiogenesis on Invasion and Metastasis

Angiogenesis is influenced by factors secreted from tumor cells that interact with endothelial cells in the stroma. VEGFs, urokinase-type plasminogen activator (uPA), and thrombospondin 1 (TSP-1) all play a role in angiogenesis and UCC progression. Elevated levels of VEGFs and uPA have been associated with poor clinical outcomes, while TSP-1 acts as an inhibitor of angiogenesis and under expression is linked to reduced OS [47,49,50,51,52,53,54]. Reduced expression of E-cadherin has been linked to tumor recurrence and disease progression, and poor OS in BC patients [55]. Several protease families, including matrix metalloproteinases (MMPs), can also modulate the tumor’s ability to disrupt the extracellular matrix and invade neighboring tissue. Overexpression of MMP-2 and MMP-9 is associated with advanced-stage BC and worse OS [56,57].

2.4. Effect of Hypoxia on UCC Invasion and Metastasis

A major aspect in the development of various tumors, including UCC, is growth under hypoxic or/and normoxic conditions that is orchestrated by activated signaling through two major transcription factors, hypoxia-inducible factor 1-alpha (HIF-1α) and HIF-2 [58,59,60]. High immunohistochemical (IHC) expression of HIF-1α in primary UC tumors is associated with higher-grade disease, vascular endothelial growth factor-related angiogenesis, and worse prognosis with regard to disease-free and overall survival in both superficial and invasive disease. Hypoxia-induced autophagy may also propagate chemoresistance to cisplatin via the HIF-1α pathway. Resistance to ICIs presents a major obstacle in mUCC treatment and the clinical importance of hypoxia and immune status has been recently recognized [59]. The hypoxic tumor microenvironment prevents sufficient glucose oxidation, induces oxidative phosphorylation, and promotes the accumulation of lactic acid and adenosine [59,60,61]. These metabolic products lead to disruptions in T-cell ratios, T-cell inactivation, recruits immunosuppressive Tregs and decreases the invasion of antigen-presenting monocytes and dendritic cells to impair the efficacy of ICIs [62,63,64].

2.5. Immune Dysregulation in UCC

UCC is characterized by extensive mutational heterogeneity, which is detectable even in early-stage disease and sets the stage for the evolution of resistance [65,66]. The emergence of treatment-resistant clones is a critical barrier to cure in patients with UCC. Chemotherapy and immunotherapy both act as selective pressures that shape the evolutionary trajectory of UCC throughout the disease course. PD-L1 is a checkpoint protein that impedes immune function, allowing tumor cells to proliferate, and PD-L1 expression on tumors has been linked to advanced stage and grade, disease progression, and worse OS after cystectomy [67,68]. Tumor cells avoid immunity through immune escape, upregulation of immunosuppressive molecules, recruitment of immunosuppressive cells within the TME, and secretion of immunosuppressive signaling molecules, e.g., cytokines. Cisplatin-based chemotherapy has been the standard of care for UCC for three decades. Resistance to PD-1/PD-L1 blockade remains a significant challenge and limitation to clinical application. Defects in the antigen presentation machinery, lack of tumor antigen, T-cell dysfunction, PD-L1 negative tumor cells, non-coding RNAs that regulate tumor immunity, myeloid-derived suppressor cells (MDSCs) activity and immune checkpoints, and the gut microbiome have been implicated in patient response to anti-PD-1/PD-L1 therapy [69,70,71,72]. As a consequence, ICIs have been approved to treat advanced or metastatic BC unreponsive to Bacillus Calmette–Guerin (BCG).

3. Screening, Diagnostic Approach and Staging of Urothelial Carcinoma

Hematuria is the most common symptom in patients with urothelial cancer. Irritative voiding symptoms, such as dysuria, may indicate carcinoma in situ (CIS) or a bladder tumor in at-risk patients, e.g., smokers. Patients are risk-stratified, and intermediate- and high-risk patients are recommended to undergo cystoscopy to confirm the diagnosis and obtain a biopsy [73]. The staging of UCC presents a major challenge as the depth of invasion determined by cystoscopy and Transurethral Resection of Bladder Tumor (TURBT) do not uniformly correlate [74]. Although a TURBT specimen can confirm the presence of muscle-invasive (T2) disease, it cannot provide information on more advanced invasion due to the risk of bladder perforation. Where suboptimal TURBT is performed, there will be detrimental consequences on patient outcomes in regard to undergrading or understaging, increased recurrence or progression. A TURBT specimen should contain muscularis propria, but if absent, a repeated TURBT is often recommended. CT or non-invasive MRI can detect extravesical or nodal disease, and is more reliable if performed with a distended bladder before transurethral resection. FDG-PET/CT imaging may aid in the staging of muscle-invasive disease and detecting metastatic BC [75]. However, its usefulness in local staging is limited by the urinary excretion of FDG [75,76]. For non-muscle-invasive tumors, histologic grading is more relevant since almost all muscle-invasive neoplasms are high grade. Although there is increasing interest in utilizing molecular detection of circulating tumor cells (CTCs) in patients with UCC, the reported diagnostic effectiveness of these methods has been inconsistent. CTC detection assays for UC have low diagnostic sensitivity but high specificity for UC diagnosis. The timing of disease assessment is a crucial consideration in CTC detection since surgical interventions may result in a temporary dissemination of CTCs in the bloodstream, while chemotherapy and other systemic treatments may destroy CTCs or reduce the expression of markers, leading to a conversion of CTC-positive patients to CTC-negative [77]. Additional studies are needed to standardize techniques and determine the best marker combinations for detecting CTCs in UCC and BC patients.

4. Current Treatment of UCC

4.1. Initial Treatment

Initial treatment and prognosis of patients diagnosed with UCC depends on key factors including anatomical site, extent (stage), and histological grade. NMIBC, with the exception of CIS, can be treated by transurethral resection leading to excellent survival outcomes. One successful way of treating NMIBCs is using BCG vaccine intravesical immunotherapy, which decreases the risk of recurrence and progression of NMIBCs [78] Intrauterine injection of BCG causes extensive inflammation in the bladder wall, which targets tumor cells, but BCG intravesical immunotherapy may have short-term immune-stimulating effects [79]. MIBC and UTUC often require RC and/or nephroureterectomy (RNU) [80]. Systemic chemotherapy, that consists of a cisplatin-based regimen for mUCC, generally does not achieve durable responses. Therefore, the treatment outcome of patients with mUCC has been poor, with an OS rate of ~15%. Cisplatin-based neoadjuvant chemotherapy (NAC) is the standard treatment for MIBC, with benefits including downstaging and elimination of micrometastatic disease before RC [81,82]. However, patient eligibility for cisplatin-based NAC is limited by a range of factors, e.g., ECOG, creatinine clearance with hearing loss, neuropathy, and heart failure [83,84]. Tri-modality treatment therapy (TMT) involving TURBT, radiation therapy, and concurrent chemotherapy, is a commonly used alternative. In clinical trials, TMT has demonstrated 5-year OS rates comparable to NAC with RC, which yields rates of 48–65% [85]. MIBC is associated with a higher incidence of distant metastasis compared with NMIBC.

4.2. Bladder Preservation in UCC

TMT has demonstrated safety and efficacy in multiple studies [86,87,88]. Bladder preservation therapies, e.g., partial cystectomy TMT, are worth consideration for MIBC patients unfit for RC, ineligible for chemotherapy, or hesitant to undergo the procedure [84]. Recent studies have shown that TMT may have better cancer-specific survival and OS than RC [84,85,86,87,88,89,90]. Studies are ongoing to explore bladder preservation options for non-muscle-invasive disease (pT2 or lower) and in patients that achieve a complete response (CR) to NAC. Tumor genomic profiling technology may help to identify biomarkers that can predict response to NAC and provide treatment guidelines [91,92,93]. Clinical trials, e.g., RETAIN, are exploring active surveillance and non-surgical local therapy options for MIBC patients with specific genetic alterations and <cT1 disease on restaging TURBT [94,95].

4.3. Surgical

Management of UCC relies heavily on surgical intervention, particularly for early-stage disease. TURBT is the most frequently employed surgical technique for NMIBC and for patients eligible for bladder preservation therapy [91]. In more advanced cases, RC may be required [96,97]. Recent advances in surgical methods have given rise to minimally invasive approaches, e.g., robotic-assisted surgery [98,99]. Moreover, ongoing research efforts aim to develop novel surgical modalities, such as photodynamic therapy (PDT), which employs light to activate a photosensitizing agent and selectively destroy malignant cells while sparing healthy tissue [100].

4.4. Single Agent and Combination Chemotherapy

Patients with mUCC can benefit from a variety of chemotherapy combinations, e.g., MVAC and gemcitabine plus cisplatin [101,102]. In certain cases where combination chemotherapy is not suitable or prior treatment has failed, single-agent chemotherapy options, e.g., platinum compounds (cisplatin, carboplatin), gemcitabine, vinca alkaloids (vinblastine, vinflunine), anthracyclines (doxorubicin, epirubicin), methotrexate, taxanes (paclitaxel, docetaxel, and nanoparticle albumin-bound paclitaxel—nabpaclitaxel), and ifosfamide may be used [103,104]. However, the response to single-agent chemotherapy is brief, and there is no evidence showing improvement in survival with first-line therapy.
Worldwide, ~75% of newly diagnosed UC cases are NMIBC and the current gold standard for treatment is surgical removal by TUR. Due to the high rate (~70%) of recurrence after TUR, patients require an intensive follow-up regime that lasts many years following the initial diagnosis. This lifelong requirement for disease surveillance means UC is associated with the highest cost from diagnosis to death [38,105]. Urine cytology has high sensitivity in detecting high-grade urothelial tumors (84%), but low sensitivity in low-grade tumors (16%). The FDA has now approved six urinary assays for clinical in vitro diagnostic use: BTA-stat, BTA-TRAK, NMP22, NMP22 BladderChek, ImmunoCyt/uCyt+, and UroVysion fluorescence in situ hybridization (FISH). These tests show high sensitivity in the detection of high-grade and late-stage UC, but are unable to detect low-grade malignancies and tend to give false positive results for benign inflammatory conditions. As such, they cannot be used as stand-alone diagnostic tests for UC. Newer tests and biomarkers to stratify treatment are needed. Prior studies analyzed 118, 187 deidentified tumor samples and identified PDL1 amplifications in 843 (0.7%), including more than 100 types of solid tumors. Most PDL1-amplified tumors (84.8%) had a low to intermediate TMB. PDL1 amplification did not always correlate with high-positive PD-L1 expression by immunohistochemical analysis.

5. Emerging Strategies to Treat UCC

5.1. Targeting FGFR

Erdafitinib is a potent FGFR1-4 tyrosine kinase inhibitor. FGFRs are essential in regulating cell proliferation, migration, differentiation, and survival [106]. Up to 20% of patients with advanced UCC have FGFR alterations, and such mutations are even more frequent (37%) in patients with upper-tract urothelial carcinoma. Hence, FGFR-inhibition may be appropriate in patients with luminal I subtype disease, in which immunotherapeutic approaches may be less effective. Erdafitinib has demonstrated activity among patients with locally advanced and unresectable mUCC and with specific FGFR alterations. Erdafitinib has demonstrated activity among patients with locally advanced and unresectable mUCC and with specific FGFR alterations. In an open-label, phase 2 study, 99 patients with locally advanced or mUCC and FGFR alterations who had disease progression after previous chemotherapy, were enrolled. Patients were randomly assigned to receive erdafitinib in either an intermittent or a continuous regimen receiving a median of five cycles. The confirmed response rate was 40%, with a median duration of PFS of 5.5 months and OS of 13.8 months. Responses were prompt and not influenced by the number or type of prior treatments, tumor location or presence of visceral metastasis. Treatment-related adverse events (TRAEs) of ≥grade 3 were reported in 46% of patients, with 13% of patients discontinuing treatment. Common side effects included diarrhea, nausea, vomiting, fatigue, and rash, while more serious AEs were hyperphosphatemia and detachment of retinal pigment epithelium. Table 2 provides a comprehensive overview of completed and ongoing clinical trials, shedding light on the evolving landscape of treatment strategies for UCC.

5.2. Immune Checkpoint Inhibitors

Immunotherapy and targeted treatments are options to address situations where BCG has been unsuccessful, and for cancers that have progressed following cisplatin-based therapies (see Table 3) [107]. High TMB, DNA damage-response mutations, the presence of genomic instability and high PD-L1 expression make UCC suitable for immunotherapy. The treatment landscape for patients with advanced UCC has been significantly changed with the approval of ICIs, e.g., atezolizumab, pembrolizumab, avelumab, and durvalumab. These approvals have opened new treatment options for patients with disease progression after platinum-based chemotherapy, those who are not eligible for first-line cisplatin-based therapy and have PD-L1-positive tumors, and those who are not suitable candidates for platinum-based therapy [108,109]. In 2017, pembrolizumab, a humanized monoclonal antibody targeting PD-1, was approved for patients with advanced UCC who had progressed after cisplatin-based therapy. A phase 3 randomized, controlled trial included 542 patients with advanced UCC randomly assigned to receive pembrolizumab or the investigator’s choice of chemotherapy. The co-primary endpoints were OS and PFS. Results showed that pembrolizumab improved OS compared to chemotherapy, with a median OS of 10.3 months in the pembrolizumab group vs. 7.4 months in the chemotherapy group. The study also found fewer TRAEs of any grade in the pembrolizumab group than the chemotherapy group [110]. Interestingly, patients benefited from pembrolizumab regardless of PD-L1 expression, measured as the combined positive score (CPS), defined as the proportion of PD-L1-expressing tumor and infiltrating immune cells relative to the total number of tumor cells. PD-L1 positivity was defined as having a CPS ≥10%.
Avelumab, another ICI, received approval in 2020 as maintenance therapy for patients with locally advanced or mUCC who did not progress after first-line platinum-based chemotherapy. The results of a phase 3 trial showed that the addition of maintenance avelumab, an anti-programmed death-ligand 1 (PD-L1) monoclonal antibody, to best supportive care, prolonged OS in patients with unresectable locally advanced or mUCC who did not have disease progression with first-line chemotherapy. The study enrolled 700 patients, and OS at 1 year was significantly better in the avelumab group compared to the control group. Avelumab also significantly prolonged OS and PFS in the PD-L1-positive population. The incidence of AEs was higher in the avelumab group but generally manageable [111]. Following demonstration of efficacy, avelumab was incorporated as a treatment option for patients with advanced UCC.
After the success of ICIs in treating mUCC, they have been introduced as adjuvant therapy. In 2021, the FDA approved nivolumab for patients who have undergone complete resection and have high-risk UCC, defined as residual cancer of ≥pT2 or pN+ after receiving neoadjuvant cisplatin-based chemotherapy or ≥pT3 or pN+ without prior chemotherapy. A phase 3, multicenter, double-blind, randomized, controlled trial compared the efficacy of nivolumab vs. placebo in 709 patients with muscle-invasive UCC who had undergone radical surgery. The study showed that nivolumab significantly improved disease-free survival and survival free from recurrence outside the urothelial tract, especially in patients with a tumor PD-L1 expression level of >1% [112]. Atezolizumab, previously used in the treatment of mUCC [113], has been discontinued recently based on a phase III trial which demonstrated that, although the addition of atezlizumab to platinum-based chemotherapy resulted in improved PFS, it did not lead to any OS advantages. Consequently, the FDA revoked its regulatory approval and it is no longer used.
Despite progress in our understanding of the molecular basis of UCC, the heterogeneity of individual tumors and evolutionary pressures imposed by therapy have hampered the ability to effectively eradicate and control the disease [114,115]. Advances in our understanding of molecular mechanisms of tumorigenesis have translated into knowledge-based therapies directed against specific oncogenic signaling targets and fundamental cellular processes [116]. Recently, it was revealed that mitochondria oxidative phosphorylation (OXPHOS) depression can be used as an effective PD-L1 downregulation method. In this method, IR-LND is prepared by conjugating mitochondria-targeted heptamethine cyanine dye IR-68 with mitochondrial complexes I and II depression agent lonidamine (LND), which further self-assembles with albumin (Alb) to form IR-LND@Alb nanoparticles. The PD-L1 expression in tumors is selectively and effectively depressed by IR-LND@Alb nanoparticles. The results suggest that the antitumor efficacy of PD-L1 depression may be superior to conventional anti-PD-L1 antibodies [19,108].

5.3. Antibody-Drug Conjugates

Two antibody-drug conjugates (ADCs) have been approved for the treatment of BC. Enfortumab vedotin targets Nectin-4, which is overexpressed in numerous BCs and is linked to the microtubule inhibitor conjugate monomethyl auristatin E [117,118]. Enfortumab vedotin was evaluated in a global, open-label, phase 3 trial that enrolled 608 patients who had previously received platinum-containing chemotherapy and PD-1/PD-L1 inhibitors [119]. Patients were randomly assigned to receive either enfortumab vedotin or chemotherapy. Results showed that OS was longer in the enfortumab vedotin group than the chemotherapy group, with a median OS of 12.9 vs. 9.0 months, respectively. PFS was longer in the enfortumab vedotin group, with a median PFS of 5.6 months compared to 3.7 months in the chemotherapy group. The incidence of TRAEs was similar in both groups. Subsequently, it was approved by the FDA. Although skin reactions, peripheral neuropathy, and hyperglycemia are common side effects, they are mostly mild to moderate in severity. Sacituzumab govitecan is a new type of ADC that targets Trop-2 through an anti-Trop-2 humanized monoclonal antibody hRS7 IgG1κ combined with SN-38, which is the active metabolite of the topoisomerase 1 inhibitor irinotecan [120,121]. It is recognized for its distinctive toxicity profile that may lead to diarrhea, nausea, vomiting, fatigue, neutropenia, and even severe or life-threatening infusion reactions. The approval of enfortumab vedotin and sacituzumab govitecan was a significant milestone in the treatment of UCC.

5.4. Cellular Vaccines and Oncolytic Viruses

Vaccine therapy activates the immune system to target cancer cells using various methods, such as synthetic peptides and viral vectors. In particular, PANVAC-VF is an example of a vaccine therapy that has shown positive results in clinical trials [122], but further research is needed to optimize its dosing and effectiveness in combination with other treatments. Oncolytic viruses selectively replicate within tumor cells, resulting in their destruction and the release of additional oncolytic viruses and tumor antigens. For instance, CG0070 is an example of an oncolytic virus that specifically targets cells with RB gene mutations, which are common in UCC [123]. Despite showing promising results, the efficacy of vaccine therapy and oncolytic viruses in combination with other therapies needs further investigation.

5.5. CAR-T Therapy

CAR-T therapy is an emerging immunotherapy approach that involves genetically modifying a patient’s T-cells to target and attack cancer cells. In UCC, CAR-T therapy is being investigated as a potential treatment for patients with advanced or metastatic disease who have failed traditional therapies [124,125]. While still in early clinical trials, CAR-T therapy has shown improving responses in some patients with UCC. However, more research is needed to optimize the therapy and determine its long-term safety and efficacy.

5.6. Antiangiogenics

There is currently no established role for antiangiogenic agents in the treatment of advanced or metastatic BC. Ramucirumab and bevacizumab, both VEGF pathway inhibitors, have shown improved PFS but not OS in clinical trials [130,131]. Cabozantinib, a multi-kinase inhibitor including VEGF receptors, is an investigational agent for platinum-refractory mUCC as a single agent or in combination with ICIs, with promising objective response rates [132].

5.7. Herbal Medicines

There is accumulating, emerging information to suggest that herbal medicines could have a beneficial effect on cancer therapy and/or treatment-induced adverse side effects. Historically, natural plant-derived drugs have been considered for cancer treatment. Sanguinarine (SANG), a naturally isolated plant alkaloidal agent, possesses chemopreventive effects [133]. It has been reported that SANG impedes tumor metastasis and development by disrupting a wide range of cell signaling pathways and its molecular targets, e.g., BCL-2, MAPKs, Akt, NF-κB, ROS, and microRNAs. Additional preclinical trials may be required before these agents advance to the clinical stage.

6. Molecular Classification as a Prognostic to Guide Treatment Decisions

UCC are biologically, molecularly and clinically heterogeneous tumors and clinico-pathological factors fail to consistently predict treatment response and patient outcome. Molecular classifiers may help organize patients into less heterogeneous subclasses and stratify treatment decisions. Muscle-invasive BC (MIBC) is a molecularly diverse disease with heterogeneous clinical outcomes [23,134,135,136,137,138]. Table 4 lists a number of biomarkers that may have value in stratifying patient treatment decisions. Recent large-scale transcriptomic profiling studies of MIBC tumors have revealed molecularly distinct clusters. Biomarkers that are prognostic and/or predictive of therapeutic responses can be tested and validated to stratify patient treatment. Several molecular classifications have been proposed, but the molecular, genetic and histologic diversity of the disease impedes clinical application. To achieve an international consensus on MIBC molecular subtypes that reconciles the published classification schemes, the Bladder Cancer Molecular Taxonomy Group developed a classification system using transcriptomes from 1750 patients and a network-based analysis of multiple independent classification systems [24]. The primary molecular classification subdivisions were basal and luminal subtypes, and these subtypes were further subdivided by TCGA into squamous, infiltrated, luminal-papillary, luminal/genomically unstable (GU), and neuronal/small cell carcinoma (SCC) subtypes [23,24,134,135,136,137,138,139]. Each subtype is associated with unique biologic, histologic and genetic characteristics. Hence, treatment strategies may be guided by oncogenic mechanisms, immune and stromal cell infiltration, and histologic and clinical features. This TCGA analysis provided a platform to develop and guide therapies for stratified MIBC patient populations (Figure 1). Additional, prospective TCGA-based analyses are needed to address NMIBC and metastatic disease. In addition, the patient populations used to derive the current primary molecular classification were predominantly of European ancestry and it remains uncertain whether these results are applicable to other ancestral groups. The impact of ethnicity and socioeconomics may impact the biology of the disease. Prospective clinical validation of biomarkers for the six molecular subtypes could guide treatment decision-making. In addition, biomarkers within the tumor, immune cells, adjacent lymph nodes, and the TME could impact patient stratification and treatment decisions for the use of targeted therapies, ICIs, ADCs, cancer vaccines and cellular therapies.
Table 4. Potential Therapeutic Biomarkers to Stratify Urothelial Cancer Treatment. Compiled and adapted from references [120,126,127,128,129,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168].
Table 4. Potential Therapeutic Biomarkers to Stratify Urothelial Cancer Treatment. Compiled and adapted from references [120,126,127,128,129,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168].
ChemotherapyNo. of PatientsPatient Positivity, %Stage of DiseaseComments
DNA Damage Repair Pathways
Nucleotide Excision Repair
ERCC1 Expression, ERCC2 Mutations
100 Patients47Locally advanced and mUCCmRNA obtained from patient tumor biopsies and analyzed by reverse transcriptase polymerase chain reaction (RT-PCR) was used to measure mRNA levels of several DNA repair genes.
Deleterious mutations in an NER pathway helicase, ERRC2, are predictive of cisplatin sensitivity in bladder cancer patients.
DNA repair deficiency phenotype predicts benefit from platinum-based chemotherapy.
Microsatellite Instability (MSI)44 patients40.6NMIBC and MIBCDNA from patient tumor biopsies was used to assess microsatellite sequence length using Polymerase chain reaction (PCR). MSI-H status in UC predicts deep and durable responses to CPI and is associated with inferior chemotherapy responses. CPI should be considered for first-line treatment in this subset of patients [169]
APOBEC mutational signature307 patientsUp to 70mUCCUsing patient tumor biopsies, genomic DNA was isolated and assayed using whole-exome sequencing (WES) and targeted next-generation sequencing (NGS) techniques to assess for APOBEC mutational signature. APOBEC-high are more likely to have mutations in DNA damage response genes (TP53, ATR, BRCA2) and chromatin regulatory genes (ARID1A, MLL, MLL3), potentially leading to a hypermutation phenotype and subsequent enhanced immune response against the tumor. IMvigor-130 trial- mUC patients with APOBEC mutational signature had significantly higher TMB and improved OS with atezolizumab containing regimens in the first-line cisplatin-ineligible scenario [170]
Immunotherapy Markers
PD-L1—IHC40 patients20–72Locally advanced and mUCCPatient tumor biopsies were assayed for PDL1 expression using immunohistochemistry (IHC) and expressed as a combined positive score (CPS, positive tumor cells and immune cells divided by the total number of cells). PD-L1 expression and high (TMB) may predict better responses to ICIs, but patients without these biomarkers may still respond to immunotherapy. Additional caveats include a lack of standardization, tumor heterogeneity and other factors influencing the TME.
PD-L1 IHC
PD-L1 (CD274)—Amplification
TMB
MSI
Genitourinary tumors (0.4%, 10/2420)Protein expression- using a CPS 1 cutoff for UC, the positive prevalence was 83.6% (989/1183)
Prevalence of 0.7% from 1183 patients
TMB- Urothelial carcinoma (36.0%, 426/1183).
MSI-H Prevalence 1.2% of 1183 patients
Locally advanced and mUCCRetrospective pan-cancer analysis of PD-L1 immunohistochemistry and gene amplification, tumor mutation burden and microsatellite instability in 48,782 cases. analysis of all cases in which both PD-L1 IHC (using the DAKO 22C3 IHC assay with either tumor proportion score (TPS) or combined positive score (CPS); or the VENTANA SP142 assay with infiltrating immune cell score (IC)) and comprehensive genomic profiling (CGP) were tested at Foundation Medicine between January 2016 and November 2019. PD-L1 positivity was defined per the CDx indication and tumor proportion score (TPS  ≥  1) for indications without a CDx claim; and TMB positivity is defined as ≥10 mutations/Mb. A total of 48,782 cases were tested for PD-L1 IHC and CGP. Patient tumor biopsies were assayed for PD-L1 amplification using PD-L1 RNA in situ hybridization (RNAish). PD-L1 amplification was detected in only 0.7% of solid tumors. Amplification had a low correlation with PD-L1 IHC and did not correlate with TMB.
Tumor Mutational Burden (TMB)401 patients30.4Locally advanced and mUCCGenomic DNA was isolated from patient tumor biopsies and characterized for mutational burden using NGS. TMB ≥ 10 mutations per megabase was detected in 122 of 401 (30.4%) patients. Total of 191 linked to response to ICI. High TMB correlated with response in certain solid tumor types: melanoma and NSCLC. May correlate mUCC. TMB assessment is multi-factorial.
Inflammatory Gene Signatures Checkmate-275 trial indicated a better response to Nivolumab therapy.
ARID1A mutation +
CXCL13 expression levels
275 patients +
348 patients
50.5%
62%
Locally advanced and mUCCDNA and RNA were isolated from patient tumor biopsies and characterized using NGS. Interrogated CXCL13 expression and ARID1A mutation as a combination biomarker in predicting response to ICT in CheckMate275 and IMvigor210. The combination of the two biomarkers in baseline tumor tissues suggested improved OS compared to either single biomarker. Cumulatively, this study revealed that the combination of CXCL13 plus ARID1A may improve prediction capability for patients receiving ICT.
TRAF2 loss116 patients73% Patient tumor biopsies were analyzed using whole-exome sequencing, RNA-seq, proteomic, and phosphoproteomic analysis to describe TRAF2 status.
Proteomic analysis identified three groups reflecting distinct clinical prognoses and molecular signatures. Immune subtypes of UC tumors revealed a complex immune landscape and suggested that TRAF2 amplification is related to the increased expression of PD-L1. Increased GARS was validated to promote the pentose phosphate pathway by inhibiting activities of PGK1 and PKM2.
CCND1 amplification152 patientsPrimary homogeneous 15%
Primary- heterogeneous 6%
Metastasis- homogeneous 22% Metastasis- heterogeneous 2%
Lymph node (LN)-positive UCC ptsCCND1 and expression of CyclinD1 were evaluated by fluorescence in situ hybridization and immunohistochemistry on patient tumor biopsies obtained from node-positive urothelial bladder cancers.
Targeted Therapies
FGFR alterations87 patients10–30%Locally advanced and mUCCDNA was isolated from patient tumor biopsies and characterized using NGS for FGFR single nucleotide variants, gene fusions or copy number abnormalities. Aberrantly activated through single-nucleotide variants, gene fusions and CNA in 5–10% of all human cancers, frequency increases to 10–30% in UC. Numerous FGFR inhibitors are currently being assessed in preclinical, Phase 1, Phase 2 and Phase 3 clinical trials. Erdafitinib and pemigatinib are currently the only approved inhibitors for use in the treatment of patients with FGFR-altered UC and Cholangiocarcinoma. UC patients with an increased frequency of FGFR3 point mutations tend to respond better to TKI therapy FGFR fusions- Clinical-grade NGS diagnostics to detect FGFR fusions and SNVs using tissue and ctDNA. Rapid identification of patients for targeted therapies and the real-time detection of acquired mutations that signal impending treatment resistance and cancer progression.
NECTIN4169 patients59.7%All NMIBC and MIBC were includedPatient tumor biopsies were analyzed using IHC for Nectin-4 expression.
High expression of Nectin-4 in squamous cell carcinoma and adenocarcinoma may guide treatment with novel Nectin-4-directed ADCs and provide a high-risk patient collective with a new promising therapeutic option.
Nectin-4-directed therapy enfortumab vedotin, an ADC comprised of a fully human monoclonal antibody specific for nectin-4 conjugated through a cleavable linker to the microtubule inhibitor MMAE.
Nectin-4 was not prognostic in histological subtypes of BC [171]
TROP2>1400 patientsUp to 82%Refractory mUCCPatient tumor biopsies were analyzed using IHC for TROP2 expression. Sacituzumab Govitecan (SG) is an ADC targeting TROP2, approved for treatment-refractory mUC. Using gene expression data from four clinical cohorts with >1400 patient samples of muscle-invasive BC and a BC tissue microarray, we found that TROP2 mRNA and protein are highly expressed across basal, luminal, and stroma-rich subtypes, but depleted in the neuroendocrine subtype. High levels of TROP2 in most subtypes were detected except in the neuroendocrine subtype. TROP2 expression is higher than NECTIN4 expression, and cells resistant to enfortumab vedotin, remain sensitive to SG.
Erb2 (Her2, EGFR)128 patients10.5%LN-positive disease and mUCCPatient tumor biopsies were analyzed using IHC for ERBB2 expression. ERBB2 overexpression and amplification were linked with high-grade and high-stage upper-tract urothelial CA (UTUCs) tumors and with tumor progression. Results suggest that ERBB2 is a biomarker for progression in UTUCs.
PI3K/AKT/mTOR/MAPK45 patients~42% in PI3K/AKT/mTOR
17% activating point mutations in PIK3CA
10% overexpression of AKT3
9% with mutations or deletions of TSC1/2
Refractory UCDNA was isolated from patient tumor biopsies and mutations in mTOR genes were assayed by NGS. Limited clinical benefit with targeting this pathway in advanced UC. Phase II, single-arm, non-randomized study with everolimus in refractory UC showed minimal response with median PFS 2.6 months, median OS 8.3 months, and 2 responses seen in 45 patients.
DNA Damage Repair (DDR) Gene Abnormalities19 patientsAlterations in DDR in up to 25%mUCCDNA was isolated from patient tumor biopsies and alterations in DNA damage repair genes were assayed by NGS. Single-agent olaparib showed limited antitumor activity in patients with mUC and DDR alterations. May relate to poorly characterized functional implications of particular DDR alterations and/or cross-resistance with platinum-based chemotherapy [159]
VEGF40 patients82%LN-positive disease and mUCCPatient tumor biopsies were analyzed using IHC for VEGF expression. The proportion of VEGF+ cells were defined to calculate a proportion score. The relative intensity was quantitated into an intensity score. Finally, a total immunostaining score was determined as the product of a proportion score and intensity score. Elevated VEGF correlates with worse outcome. VEGF pathway inhibition attenuates tumor proliferation and invasion. Ramucirumab, a fully humanized monoclonal antibody that binds VEGF receptor 2, has shown benefit both in randomized phase II and III trials. Results with sunitinib, pazopanib, vandetinib, or cabozantinib were not convincing.
Figure 1. Molecular subtype classification of bladder cancer and treatment options. Figure compiled and adapted from Refs. [24,140,141,142,143,144,145,146,147,148,149,150,151,152,171,172,173,174,175,176].
Figure 1. Molecular subtype classification of bladder cancer and treatment options. Figure compiled and adapted from Refs. [24,140,141,142,143,144,145,146,147,148,149,150,151,152,171,172,173,174,175,176].
Cancers 15 04886 g001
Chemotherapy-resistant UCC lacks a uniformly curative therapy, and deciphering how chemotherapy directs selective pressure is a central question with clinical implications [139,177]. Therefore, to identify potential markers that correlated with treatment response, the genetic landscape of chemoradiation-treated UCC samples was analyzed [139]. Whole-exome sequencing (WES) and clonality analysis of 72 UCC samples was performed, including 16 matched sets of primary and advanced tumors prospectively collected before and after chemotherapy. Results showed that chemotherapy-treated UCC is characterized by intra-patient mutational heterogeneity, and that the majority of mutations were not shared. Mutation profiling between primary and recurrent tumors suggested that pre-existing, resistant clonal populations represented the primary mechanism of chemoradiation treatment failure. Cisplatin-based chemotherapy remains the standard of care for patients with muscle-invasive UCC but molecular determinants of response remain incompletely understood [143]. Whole-exome-sequencing was performed on pretreatment tumors and germline DNA was obtained from 50 patients with MIBC who had received neoadjuvant cisplatin-based chemotherapy followed by cystectomy (25 pT0/pTis “responders,” 25 pT2+ “nonresponders”) to identify mutations that had specifically occurred in responders. ERCC2, a nucleotide excision repair gene, was the only significantly mutated gene enriched in the cisplatin responders compared with nonresponders (q < 0.01). Expression of representative ERCC2 mutants in an ERCC2-deficient cell line failed to rescue cisplatin and UV sensitivity compared with wild-type ERCC2. Somatic ERCC2 mutations were shown to correlate with complete responses to cisplatin-based chemosensitivity in muscle-invasive UCC. Inflammation-driven phenotypic plasticity also alters the antigenic landscape of tumor cells, rewiring oncogenic signaling networks to reprogram immune cell functions [178].
A systematic meta-analysis merged gene expression datasets from the GEO repository for 18 datasets and identified 815 differentially expressed genes (DEGs) [171]. The key hub genes were screened for their differential expression in patient urine and blood plasma samples. A three-gene signature model, including COL3A1, FOXM1, and PLK4, was built. The predictive value regarding muscle-invasive patients’ responses to neoadjuvant chemotherapy was analyzed and a six-gene signature model, including ANXA5, CD44, NCAM1, SPP1, CDCA8, and KIF14, was developed. The study identified nine key biomarker genes—ANXA5, CDT1, COL3A1, SPP1, VEGFA, CDCA8, HJURP, TOP2A, and COL6A1—which were differentially expressed in urine or blood, held a prognostic or predictive value, and were immunohistochemically validated. Promising biomarkers may eventually lead to greater personalization of care for patients with BC, but remain investigational.
Five programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) inhibitors are currently approved for treatment of locally advanced or mUCC of the bladder and the upper urinary tract [172]. Due to restrictions by the FDA and EMA, first-line treatment with Atezolizumab and Pembrolizumab in platinum-ineligible patients requires immunohistochemical PD-L1 testing. In the second-line setting, all drugs are approved without PD-L1 testing. The balance of adaptive immunity and pro-tumorigenic inflammation in individual TME is associated with PD-1/PD-L1 resistance in UCC, with the latter linked to a proinflammatory cellular state of myeloid phagocytic cells detectable in tumor and blood [173]. The quantity and spatial distribution of stromal tumor-infiltrating lymphocytes (sTIL) within the TME also predict stages of tumor inflammation, subtypes, and patient survival, and correlate with expression of immune checkpoints in an analysis of 542 patients with MIBC [174]. High sTILs indicated an inflamed subtype with an 80% 5-year DSS, and a lack of immune infiltrates identified an uninflamed subtype with a survival rate of less than 25%. A separate immune evading phenotype with upregulated immune checkpoints was associated with poor survival. Within the TIME are tertiary lymphoid structures (TLS), which can mediate antitumor activity via immune cells. High TLS amounts and close tumor distance correlated significantly with an inflamed phenotype and favorable survival. The uninflamed and evasion phenotypes showed lowest TLS numbers, farthest tumor distances, and shortest survival. High inflammation also correlated with increased neoantigen load and mutational burden.
There is a need in clinical practice to identify biomarkers that can serve as tools for risk assessment, screening and early detection of UCC, as well as for accurate diagnosis, patient prognosis, prediction of response to therapy, and cancer surveillance and monitoring response [175]. Better biomarkers are needed as tools to identify patient subsets since precision oncology is effective only in patients with specific cancer genetic mutations. Systematic, bioinformatic tools represent a valuable approach to identify and validate novel biomarkers with greater sensitivity, specificity, and positive predictive value. Publicly available genome-wide datasets, greater availability of patient pre- and post-treatment samples, analysis of tumor and immune cells, (urinary) extracellular vesicles, and the microbiome are promising sources of biomarkers. Low- or non-invasive methods of obtaining biomarkers also are appealing. Personalized treatment approaches based on individual patient characteristics and tumor biomarkers hold promise to stratify and optimize therapy and improve patient quality-of-life and OS, while reducing treatment-related toxicities. Additional candidate biomarkers to personalize BC care are under investigation, but significant obstacles must be overcome before they can be implemented in clinical practice.

7. Conclusions

Despite advances in diagnostic and therapeutic strategies, mUCC remains a significant challenge. The diagnostic and monitoring techniques used for urologic cancers comprise a group of invasive methodologies that still lack sensitivity and specificity [176]. Early diagnosis is crucial to improve patient outcomes and current treatments offer a range of options to manage UCC and mUCC. The treatment of mUCC has shifted dramatically following the introduction of ICIs [68,179]. Similarly, BCG is one of the most successful immunotherapies and remains the gold standard of care for patients with high-risk NMIBC, with initial response rates of ~70% [180]. Collectively, recent trials indicate that ~30% of mUCC patients demonstrate a response to ICIs. Recently approved ICIs are available as first-line therapy for cisplatin-ineligible patients or as second-line therapy for mUCC patients. Currently, selection of patients with mUCC to receive first-line immunotherapy is informed by expression of PD-L1 [181]. Patients must also be cisplatin-ineligible, but may be carboplatin-eligible if PD-L1 expression is sufficiently high. Patients with mUCC who are ineligible for any platinum-based chemotherapy may also receive immunotherapy in the first line, regardless of PD-L1 expression. High tumor mutational burden and microsatellite instability may be used to select patients for treatment with immunotherapy in cases of advanced BC, with some non-urothelial histologies in which patients have progressed. Promising innovations that may potentially advance precision medicine in UCC include multi-omic approaches, innovative trials designs, cell-free DNA, and machine learning algorithms. Biomarkers may allow for better disease classification, prognostication, and development of more efficacious noninvasive detection and surveillance strategies, as well as for the selection of therapeutic targets which can be used in BC, particularly in a neoadjuvant or adjuvant setting. Clinically relevant and useful biomarkers are also needed to identify high-risk disease, to optimize and personalize ICIs treatment for UCC. At present, alterations of FGFR2/3 are the only genetic feature of mUCC currently used to select patients for targeted therapy [140,182,183]. In addition, erdafitinib is the only targeted therapy specifically approved for FGFR-altered UCC.
Recent studies have demonstrated using incorporating immunotherapies, i.e., ICIs, BCG, cytokines, adoptive T-cell immunotherapy, DCs, and macrophages, has a potential benefit for UCC patients. Nanoparticles, nano-immunotherapy, herbal medicines, and agents that overcome hypoxia-mediated immunosuppression are all potential approaches to augment PD-L1 expression on tumor cells to promote the PD-L1/PD-1 interaction. Antibiotics, fecal microbiota transplantation, and diet regulation are feasible, practical approaches to manipulate the gut microbiome and improve patient responses to immunotherapy. Prevention strategies, e.g., lifestyle modifications, smoking cessation, and chemoprevention, have the potential to reduce the incidence and improve the management and outcome of UCC. Studies that explore the utility of ICIs in the adjuvant or neoadjuvant settings, or in combination with chemotherapy, may reveal strategies to enhance patient response. The treatment paradigm for UCC is rapidly changing and novel biomarkers are emerging to select patients for appropriate therapies. Taken together, these advances offer an opportunity to improve patient survival.

Author Contributions

Conceptualization, J.J.D. and B.R.; writing—original draft preparation, J.J.D. and B.R.; writing—review and editing J.J.D., B.R., J.J.I.-H. and P.S.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Krakhmal, N.V.; Zavyalova, M.V.; Denisov, E.V.; Vtorushin, S.V.; Perelmuter, V.M. Cancer Invasion: Patterns and Mechanisms. Acta Nat. 2015, 7, 17–28. [Google Scholar] [CrossRef]
  2. Katims, A.B.; Reisz, P.A.; Nogueira, L.; Truong, H.; Lenis, A.T.; Pietzak, E.J.; Kim, K.; Coleman, J.A. Targeted Therapies in Advanced and Metastatic Urothelial Carcinoma. Cancers 2022, 14, 5431. [Google Scholar] [CrossRef]
  3. Lenis, A.T.; Lec, P.M.; Chamie, K.; Mshs, M.D. Bladder Cancer: A Review. JAMA 2020, 324, 1980–1991. [Google Scholar] [CrossRef]
  4. Compérat, E.; Amin, M.B.; Cathomas, R.; Choudhury, A.; De Santis, M.; Kamat, A.; Stenzl, A.; Thoeny, H.C.; Witjes, J.A. Current best practice for bladder cancer: A narrative review of diagnostics and treatments. Lancet 2022, 400, 1712–1721. [Google Scholar] [CrossRef]
  5. Akhtar, M.; Al-Bozom, I.; Ben Gashir, M.; Taha, N.M.; Rashid, S.; Al-Nabet, A.D.M.H. Urothelial Carcinoma In Situ (CIS): New Insights. Adv. Anat. Pathol. 2019, 26, 313–319. [Google Scholar] [CrossRef] [PubMed]
  6. Wenzel, M.; Deuker, M.; Nocera, L.; Collà Ruvolo, C.; Tian, Z.; Shariat, S.F.; Saad, F.; Briganti, A.; Becker, A.; Kluth, L.A.; et al. Comparison Between Urothelial and Non-Urothelial Urethral Cancer. Front. Oncol. 2021, 10, 629692. [Google Scholar] [CrossRef] [PubMed]
  7. Hayashi, T.; Fujita, K.; Hayashi, Y.; Hatano, K.; Kawashima, A.; McConkey, D.J.; Nonomura, N. Mutational Landscape and Environmental Effects in Bladder Cancer. Int. J. Mol. Sci. 2020, 21, 6072. [Google Scholar] [CrossRef]
  8. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer Statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef]
  9. Freedman, N.D.; Silverman, D.T.; Hollenbeck, A.R.; Schatzkin, A.; Abnet, C.C. Association between Smoking and Risk of Bladder Cancer among Men and Women. JAMA 2011, 306, 737–745. [Google Scholar] [CrossRef] [PubMed]
  10. Cumberbatch, M.G.; Rota, M.; Catto, J.W.F.; La Vecchia, C. The Role of Tobacco Smoke in Bladder and Kidney Carcinogenesis: A Comparison of Exposures and Meta-Analysis of Incidence and Mortality Risks. Eur. Urol. 2016, 70, 458–466. [Google Scholar] [CrossRef]
  11. Letašiová, S.; Medve’ová, A.; Šovčíková, A.; Dušinská, M.; Volkovová, K.; Mosoiu, C.; Bartonová, A. Bladder cancer, a review of the environmental risk factors. Environ. Health 2012, 11 (Suppl. S1), S11. [Google Scholar] [CrossRef] [PubMed]
  12. Scher, H.I.; Rosenberg, R.E.; Motzer, R.J. Bladder and Renal Cell Carcinomas. In Harrison’s Principles of Internal Medicine; McGraw Hill: New York, NY, USA, 2015; p. 17e. [Google Scholar]
  13. Hubosky, S.G.; Boman, B.M.; Charles, S.; Bibbo, M.; Bagley, D.H. Ureteroscopic management of upper tract urothelial carcinoma (UTUC) in patients with Lynch Syndrome (hereditary nonpolyposis colorectal cancer syndrome). BJU Int. 2013, 112, 813–819. [Google Scholar] [CrossRef] [PubMed]
  14. Shafique, M.A.; Haseeb, A.; Siddiq, M.A.; Mussarat, A.; Rangwala, H.S.; Mustafa, M.S. Current and Emerging Treatments for Urothelial Carcinoma: A Focus on Enfortumab Vedotin. Cancer Manag. Res. 2023, 15, 699–706. [Google Scholar] [CrossRef]
  15. Postow, M.A.; Callahan, M.K.; Wolchok, J.D. Immune Checkpoint Blockade in Cancer Therapy. J. Clin. Oncol. 2015, 33, 1974–1982. [Google Scholar] [CrossRef]
  16. Daro-Faye, M.; Kassouf, W.; Souhami, L.; Marcq, G.; Cury, F.; Niazi, T.; Sargos, P. Combined radiotherapy and immunotherapy in urothelial bladder cancer: Harnessing the full potential of the anti-tumor immune response. World J. Urol. 2021, 39, 1331–1343. [Google Scholar] [CrossRef]
  17. Shah, S.; Zhang, C.A.; Hancock, S.; Fan, A.; Skinner, E.; Srinivas, S. Consolidative Radiotherapy in Metastatic Urothelial Cancer. Clin. Genitourin. Cancer 2017, 15, 685–688. [Google Scholar] [CrossRef] [PubMed]
  18. Zhou, Z.; Liu, Y.; Song, W.; Jiang, X.; Deng, Z.; Xiong, W.; Shen, J. Metabolic reprogramming mediated PD-L1 depression and hypoxia reversion to reactivate tumor therapy. J. Control. Release 2022, 352, 793–812. [Google Scholar]
  19. Liu, Y.; Zhou, Z.; Hou, J.; Xiong, W.; Kim, H.; Chen, J.; Zheng, C.; Jiang, X.; Yoon, J.; Shen, J. Tumor Selective Metabolic Reprogramming as a Prospective PD-L1 Depression Strategy to Reactivate Immunotherapy. Adv. Mater. 2022, 34, e2206121. [Google Scholar] [PubMed]
  20. Adamczyk, P.; Pobłocki, P.; Kadlubowski, M.; Ostrowski, A.; Wróbel, A.; Mikołajczak, W.; Adamowicz, J.; Drewa, T.; Juszczak, K. A Comprehensive Approach to Clinical Staging of Bladder Cancer. J. Clin. Med. 2022, 11, 761. [Google Scholar] [CrossRef]
  21. McConkey, D.J.; Lee, S.; Choi, W.; Tran, M.; Majewski, T.; Lee, S.; Siefker-Radtke, A.; Dinney, C.; Czerniak, B. Molecular genetics of bladder cancer: Emerging mechanisms of tumor initiation and progression. Urol. Oncol. 2010, 28, 429–440. [Google Scholar] [CrossRef]
  22. Mitra, A.P. Molecular substratification of bladder cancer: Moving towards individualized patient management. Ther. Adv. Urol. 2016, 8, 215–233. [Google Scholar] [CrossRef]
  23. Robertson, A.G.; Kim, J.; Al-Ahmadie, H.; Bellmunt, J.; Guo, G.; Cherniack, A.D.; Hinoue, T.; Laird, P.W.; Hoadley, K.A.; Akbani, R.; et al. Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer. Cell 2017, 171, 540–556. [Google Scholar]
  24. Kamoun, A.; de Reyniès, A.; Allory, Y.; Sjödahl, G.; Robertson, A.G.; Seiler, R.; Hoadley, K.A.; Groeneveld, C.S.; Al-Ahmadie, H.; Choi, W.; et al. A Consensus Molecular Classification of Muscle-Invasive Bladder Cancer. Eur. Urol. 2020, 77, 420–433. [Google Scholar] [CrossRef]
  25. Weinstein, J.N.; Akbani, R.; Broom, B.M.; Wang, W.; Verhaak, R.G.W.; McConkey, D.; Lerner, S.; Morgan, M.; Creighton, C.J.; Smith, C.; et al. Comprehensive Molecular Characterization of Urothelial Bladder Carcinoma. Nature 2014, 507, 315–322. [Google Scholar] [CrossRef]
  26. Ren, X.; Guo, S.; Guan, X.; Kang, Y.; Liu, J.; Yang, X. Immunological Classification of Tumor Types and Advances in Precision Combination Immunotherapy. Front. Immunol. 2022, 13, 790113. [Google Scholar] [CrossRef] [PubMed]
  27. Nordentoft, I.; Lamy, P.; Birkenkamp-Demtröder, K.; Shumansky, K.; Vang, S.; Hornshøj, H.; Juul, M.; Villesen, P.; Hedegaard, J.; Roth, A.; et al. Mutational Context and Diverse Clonal Development in Early and Late Bladder Cancer. Cell Rep. 2014, 7, 1649–1663. [Google Scholar] [CrossRef]
  28. Mitra, A.P.; Jordà, M.; Cote, R.J. Pathological Possibilities and Pitfalls in Detecting Aggressive Bladder Cancer. Curr. Opin. Urol. 2012, 22, 397–404. [Google Scholar]
  29. Droller, M.J. FGFR3 and P53 Characterize Alternative Genetic Pathways in the Pathogenesis of Urothelial Cell Carcinoma. J. Urol. 2004, 172, 1911–1914. [Google Scholar] [CrossRef]
  30. Homami, A.; Ataei Kachoei, Z.; Asgarie, M.; Ghazi, F. Analysis of FGFR3 and HRAS genes in patients with bladder cancer. Med. J. Islamic Repub. Iran 2020, 34, 108. [Google Scholar] [CrossRef]
  31. Hoffman-Censits, J.; Choi, W.; Pal, S.; Trabulsi, E.; Kelly, W.K.; Hahn, N.M.; McConkey, D.; Comperat, E.; Matoso, A.; Cussenot, O.; et al. Urothelial Cancers with Small Cell Variant Histology Have Confirmed High Tumor Mutational Burden, Frequent TP53 and RB Mutations, and a Unique Gene Expression Profile. Eur. Urol. Oncol. 2021, 4, 297–300. [Google Scholar] [CrossRef]
  32. Kerzeli, I.K.; Kostakis, A.; Türker, P.; Malmström, P.U.; Hemdan, T.; Mezheyeuski, A.; Ward, D.G.; Bryan, R.T.; Segersten, U.; Lord, M.; et al. Elevated levels of MMP12 sourced from macrophages are associated with poor prognosis in urothelial bladder cancer. BMC Cancer 2023, 23, 605. [Google Scholar] [CrossRef]
  33. Mitra, A.P.; Hansel, D.E.; Cote, R.J. Prognostic Value of Cell-Cycle Regulation Biomarkers in Bladder Cancer. Semin. Oncol. 2012, 39, 524–533. [Google Scholar] [CrossRef] [PubMed]
  34. Hammam, O.; Magdy, M.; Badawy, M.; Osili, K.A.; Kholy, A.E.; LeitHy, T.E. Expression of MDM2 mRNA, MDM2, P53 and P16 Proteins in Urothelial Lesions in the View of the WHO 4th Edition Guidelines as a Molecular Insight towards Personalized Medicine. Maced. J. Med. Sci. 2017, 5, 578–586. [Google Scholar] [CrossRef] [PubMed]
  35. Pan, Q.; Sathe, A.; Black, P.C.; Goebell, P.J.; Kamat, A.M.; Schmitz-Draeger, B.; Nawroth, R. CDK4/6 Inhibitors in Cancer Therapy: A Novel Treatement Strategy for Bladder Cancer. Bladder Cancer 2017, 3, 79–88. [Google Scholar] [CrossRef]
  36. Rivlin, N.; Brosh, R.; Oren, M.; Rotter, V. Mutations in the p53 Tumor Suppressor Gene: Important Milestones at the Various Steps of Tumorigenesis. Genes Cancer 2011, 2, 466–474. [Google Scholar] [CrossRef]
  37. Ong, F.; Moonen, L.M.; Gallee, M.P.; ten Bosch, C.; Zerp, S.F.; Hart, A.A.; Bartelink, H.; Verheij, M. Prognostic factors in transitional cell cancer of the bladder: An emerging role for Bcl-2 and p53. Radiother. Oncol. 2001, 61, 169–175. [Google Scholar] [CrossRef]
  38. Thomas, J.; Sonpavde, G. Molecularly Targeted Therapy towards Genetic Alterations in Advanced Bladder Cancer. Cancers 2022, 14, 1795. [Google Scholar] [CrossRef]
  39. Ross, J.S.; Wang, K.; Al-Rohil, R.N.; Nazeer, T.; Sheehan, C.E.; Otto, G.A.; He, J.; Palmer, G.; Yelensky, R.; Lipson, D.; et al. Advanced urothelial carcinoma: Next-generation sequencing reveals diverse genomic alterations and targets of therapy. Mod. Pathol. 2014, 27, 271–280. [Google Scholar] [CrossRef]
  40. Rose, T.L.; Chism, D.D.; Alva, A.S.; Deal, A.M.; Maygarden, S.J.; Whang, Y.E.; Kardos, J.; Drier, A.; Basch, E.; Godley, P.A.; et al. Phase II trial of palbociclib in patients with metastatic urothelial cancer after failure of first-line chemotherapy. Br. J. Cancer 2018, 119, 801–807. [Google Scholar] [CrossRef] [PubMed]
  41. Klein, F.G.; Granier, C.; Zhao, Y.; Pan, Q.; Tong, Z.; Gschwend, J.E.; Holm, P.S.; Nawroth, R. Combination of Talazoparib and Palbociclib as a Potent Treatment Strategy in Bladder Cancer. J. Pers. Med. 2021, 11, 340. [Google Scholar] [CrossRef] [PubMed]
  42. Dangle, P.P.; Zaharieva, B.; Jia, H.; Pohar, K.S. Ras-MAPK pathway as a therapeutic target in cancer—Emphasis on bladder cancer. Recent Pat. Anticancer Drug Discov. 2009, 4, 125–136. [Google Scholar] [CrossRef]
  43. Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel, E.E., III; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
  44. Van Rhijn, B.W.G.; Zuiverloon, T.C.M.; Vis, A.N.; Radvanyi, F.; Van Leenders, G.J.L.H.; Ooms, B.C.M.; Kirkels, W.J.; Lockwood, G.A.; Boevé, E.R.; Jöbsis, A.C.; et al. Molecular Grade (FGFR3/MIB-1) and EORTC Risk Scores Are Predictive in Primary Non-Muscle-Invasive Bladder Cancer. Eur. Urol. 2010, 58, 433–441. [Google Scholar] [CrossRef] [PubMed]
  45. Laurberg, J.R.; Brems-Eskildsen, A.S.; Nordentoft, I.; Fristrup, N.; Schepeler, T.; Ulhøi, B.P.; Agerbæk, M.; Hartmann, A.; Bertz, S.; Wittlinger, M.; et al. Expression of TIP60 (Tat-Interactive Protein) and MRE11 (Meiotic Recombination 11 Homolog) Predict Treatment-Specific Outcome of Localised Invasive Bladder Cancer. BJU Int. 2012, 110, E1228–E1236. [Google Scholar] [CrossRef]
  46. Ide, H.; Inoue, S.; Miyamoto, H. Histopathological and Prognostic Significance of the Expression of Sex Hormone Receptors in Bladder Cancer: A Meta-Analysis of Immunohistochemical Studies. PLoS ONE 2017, 12, e0174746. [Google Scholar] [CrossRef]
  47. Tuygun, C.; Kankaya, D.; Imamoglu, A.; Sertcelik, A.; Zengin, K.; Oktay, M.; Sertcelik, N. Sex-Specific Hormone Receptors in Urothelial Carcinomas of the Human Urinary Bladder: A Comparative Analysis of Clinicopathological Features and Survival Outcomes According to Receptor Expression. Urol. Oncol. Semin. Orig. Investig. 2011, 29, 43–51. [Google Scholar] [CrossRef]
  48. Boorjian, S.; Ugras, S.; Mongan, N.P.; Gudas, L.J.; You, X.; Tickoo, S.K.; Scherr, D.S. Androgen Receptor Expression Is Inversely Correlated with Pathologic Tumor Stage in Bladder Cancer. Urology 2004, 64, 383–388. [Google Scholar] [CrossRef]
  49. Mitra, A.P.; Pagliarulo, V.; Yang, D.; Waldman, F.M.; Datar, R.H.; Skinner, D.G.; Groshen, S.; Cote, R.J. Generation of a Concise Gene Panel for Outcome Prediction in Urinary Bladder Cancer. J. Clin. Oncol. 2009, 27, 3929–3937. [Google Scholar] [CrossRef] [PubMed]
  50. Goto, T.; Miyamoto, H. The Role of Estrogen Receptors in Urothelial Cancer. Front. Endocrinol. 2021, 12, 643870. [Google Scholar] [CrossRef]
  51. Golus, M.; Bugajski, P.; Chorbińska, J.; Krajewski, W.; Lemiński, A.; Saczko, J.; Kulbacka, J.; Szydełko, T.; Małkiewicz, B. STAT3 and Its Pathways’ Dysregulation-Underestimated Role in Urological Tumors. Cells 2022, 11, 3024. [Google Scholar] [CrossRef] [PubMed]
  52. Crew, J.P.; O’Brien, T.; Bradburn, M.; Fuggle, S.; Bicknell, R.; Cranston, D.; Harris, A.L. Vascular Endothelial Growth Factor Is a Predictor of Relapse and Stage Progression in Superficial Bladder Cancer. Cancer Res. 1997, 57, 5281–5285. [Google Scholar] [CrossRef]
  53. Jaeger, T.M.; Weidner, N.; Chew, K.; Moore, D.H.; Kerschmann, R.L.; Waldman, F.M.; Carroll, P.R. Tumor Angiogenesis Correlates with Lymph Node Metastases in Invasive Bladder Cancer. J. Urol. 1995, 154, 69–71. [Google Scholar] [CrossRef] [PubMed]
  54. Shariat, S.F.; Youssef, R.F.; Gupta, A.; Chade, D.C.; Karakiewicz, P.I.; Isbarn, H.; Jeldres, C.; Sagalowsky, A.I.; Ashfaq, R.; Lotan, Y. Association of Angiogenesis Related Markers With Bladder Cancer Outcomes and Other Molecular Markers. J. Urol. 2010, 183, 1744–1750. [Google Scholar] [CrossRef]
  55. Bryan, R.T. Cell adhesion and urothelial bladder cancer: The role of cadherin switching and related phenomena. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2015, 370, 20140042. [Google Scholar] [CrossRef]
  56. Rodriguez Faba, O.; Palou-Redorta, J.; Fernández-Gómez, J.M.; Algaba, F.; Eiró, N.; Villavicencio, H.; Vizoso, F.J. Matrix Metalloproteinases and Bladder Cancer: What is New? ISRN Urol. 2012, 2012, 581539. [Google Scholar] [PubMed]
  57. Bazargani, S.T.; Clifford, T.; Djaladat, H.; Schuckman, A.; Sadeghi, S.; Dorff, T.; Quinn, D.; Daneshmand, S. Association between Epithelial Tumor Markers’ Trends during the Course of Treatment and Oncological Outcomes in Urothelial Bladder Cancer. Urol. Oncol. Semin. Orig. Investig. 2017, 35, 609–611. [Google Scholar] [CrossRef]
  58. Theodoropoulos, V.E.; Lazaris, A.C.; Sofras, F.; Gerzelis, I.; Tsoukala, V.; Ghikonti, I.; Manikas, K.; Kastriotis, I. Hypoxia-inducible factor 1 alpha expression correlates with angiogenesis and unfavorable prognosis in bladder cancer. Eur. Urol. 2004, 46, 200–208. [Google Scholar] [CrossRef] [PubMed]
  59. Palit, V.; Phillips, R.M.; Puri, R.; Shah, T.; Bibby, M.C. Expression of HIF-1alpha and Glut-1 in human bladder cancer. Oncol. Rep. 2005, 14, 909–913. [Google Scholar] [CrossRef] [PubMed]
  60. Mao, X.; Nanzhang, U.; Xiao, J.; Wu, H.; Ding, K. Hypoxia-Induced Autophagy Enhances Cisplatin Resistance in Human Bladder Cancer Cells by Targeting Hypoxia-Inducible Factor-1α J. Immunol. Res. 2021, 2021, 8887437. [Google Scholar]
  61. Hong, S.; Zhang, Y.; Cao, M.; Lin, A.; Yang, Q.; Zhang, J.; Luo, P.; Guo, L. Hypoxic Characteristic Genes Predict Response to Immunotherapy for Urothelial Carcinoma. Front. Cell Dev. Biol. 2021, 9, 762478. [Google Scholar] [CrossRef]
  62. Krzywinska, E.; Stockmann, C. Hypoxia, Metabolism and Immune Cell Function. Biomedicines 2018, 6, 56. [Google Scholar] [CrossRef]
  63. Ohta, A. A Metabolic Immune Checkpoint: Adenosine in Tumor Microenvironment. Front. Immunol. 2016, 7, 109. [Google Scholar] [CrossRef] [PubMed]
  64. Yuen, V.W.-H.; Wong, C.C.-L. Hypoxia-inducible Factors and Innate Immunity in Liver Cancer. J. Clin. Investig. 2020, 130, 5052–5062. [Google Scholar] [CrossRef] [PubMed]
  65. Matulay, J.T.; Kamat, A.M. Advances in risk stratification of bladder cancer to guide personalized medicine. F1000Research 2018, 7, 1137. [Google Scholar] [CrossRef]
  66. de Jong, J.J.; Liu, Y.; Boorjian, S.A.; Bivalacqua, T.J.; Porten, S.P.; Wheeler, T.; Davicioni, E.; Svatek, R.S.; Boormans, J.L.; Black, P.C.; et al. A Genomic Classifier for Predicting Clinically Aggressive Luminal Bladder Tumors with Higher Rates of Pathological Up Staging. J. Urol. 2020, 204, 239–246. [Google Scholar] [PubMed]
  67. Donin, N.M.; Lenis, A.T.; Holden, S.; Drakaki, A.; Pantuck, A.; Belldegrun, A.; Chamie, K. Immunotherapy for the Treatment of Urothelial Carcinoma. J. Urol. 2017, 197, 14–22. [Google Scholar]
  68. Safarzadeh Kozani, P.; Safarzadeh Kozani, P.; Ahmadi Najafabadi, M.; Yousefi, F.; Mirarefin, S.M.J.; Rahbarizadeh, F. Recent Advances in Solid Tumor CAR-T Cell Therapy: Driving Tumor Cells From Hero to Zero? Front. Immunol. 2022, 13, 795164. [Google Scholar]
  69. Nakanishi, J.; Wada, Y.; Matsumoto, K.; Azuma, M.; Kikuchi, K.; Ueda, S. Overexpression of B7-H1 (PD-L1) Significantly Associates with Tumor Grade and Postoperative Prognosis in Human Urothelial Cancers. Cancer Immunol. Immunother. 2007, 56, 1173–1182. [Google Scholar] [CrossRef]
  70. Ding, X.; Chen, Q.; Yang, Z.; Li, J.; Zhan, H.; Lu, N.; Chen, M.; Yang, Y.; Wang, J.; Yang, D. Clinicopathological and prognostic value of PD-L1 in urothelial carcinoma: A meta-analysis. Cancer Manag. Res. 2019, 11, 4171–4184. [Google Scholar] [CrossRef]
  71. Lyu, X.; Wang, P.; Qiao, Q.; Jiang, Y. Genomic stratification based on microenvironment immune types and PD-L1 for tailoring therapeutic strategies in bladder cancer. BMC Cancer 2021, 21, 646. [Google Scholar] [CrossRef]
  72. Chen, X.; Chen, H.; He, D.; Cheng, Y.; Zhu, Y.; Xiao, M.; Lan, H.; Wang, Z.; Cao, K. Analysis of Tumor Microenvironment Characteristics in Bladder Cancer: Implications for Immune Checkpoint Inhibitor Therapy. Front. Immunol. 2021, 12, 672158. [Google Scholar] [CrossRef]
  73. Kołodziej, A.; Krajewski, W.; Matuszewski, M.; Tupikowski, K. Review of current optical diagnostic techniques for non-muscle-invasive bladder cancer. Central Eur. J. Urol. 2016, 69, 150. [Google Scholar]
  74. Kim, L.H.C.; Patel, M.I. Transurethral resection of bladder tumor (TURBT). Trans. Androl. Urol. 2020, 9, 3056–3072. [Google Scholar] [CrossRef] [PubMed]
  75. Apolo, A.B.; Riches, J.; Schöder, H.; Akin, O.; Trout, A.; Milowsky, M.I.; Bajorin, D.F. Clinical Value of Fluorine-18 2-Fluoro-2-Deoxy-D-Glucose Positron Emission Tomography/Computed Tomography in Bladder Cancer. J. Clin. Oncol. 2010, 28, 3973–3978. [Google Scholar] [CrossRef]
  76. Soubra, A.; Hayward, D.; Dahm, P.; Goldfarb, R.; Froehlich, J.; Jha, G.; Konety, B.R. The Diagnostic Accuracy of 18F-Fluorodeoxyglucose Positron Emission Tomography and Computed Tomography in Staging Bladder Cancer: A Single-Institution Study and a Systematic Review with Meta-Analysis. World J. Urol. 2016, 34, 1229–1237. [Google Scholar] [CrossRef] [PubMed]
  77. Msaouel, P.; Koutsilieris, M. Diagnostic Value of Circulating Tumor Cell Detection in Bladder and Urothelial Cancer: Systematic Review and Meta-Analysis. BMC Cancer 2011, 11, 336. [Google Scholar] [CrossRef]
  78. Shelley, M.; Court, J.B.; Kynaston, H.; Wilt, T.J.; Fish, R.; Mason, M. Intravesical Bacillus Calmette-Guérin in Ta and T1 Bladder Cancer. Cochrane Database Syst. Rev. 2000, 2000, CD001986. [Google Scholar] [CrossRef]
  79. Bevers, R.F.M.; Kurth, K.H.; Schamhart, D.H.J. Role of Urothelial Cells in BCG Immunotherapy for Superficial Bladder Cancer. Br. J. Cancer 2004, 91, 607–612. [Google Scholar] [PubMed]
  80. SEER Cancer Stat Facts: Bladder Cancer. In Surveillance, Epidemiology and End Results Program; National Cancer Institute: Rockville Pike Bethesda, MD, USA, 2018.
  81. Scher, H.I.; Yagoda, A.; Herr, H.W.; Sternberg, C.N.; Morse, M.J.; Sogani, P.C.; Watson, R.C.; Reuter, V.; Whitmore, W.F.; Fair, W.R. Neoadjuvant M-VAC (Methotrexate, Vinblastine, Doxorubicin and Cisplatin) for Extravesical Urinary Tract Tumors. J. Urol. 1988, 139, 474–477. [Google Scholar] [CrossRef]
  82. Park, J.C.; Citrin, D.E.; Agarwal, P.K.; Apolo, A.B. Multimodal Management of Muscle-Invasive Bladder Cancer. Curr. Probl. Cancer 2014, 38, 80–108. [Google Scholar] [CrossRef]
  83. Galsky, M.D.; Hahn, N.M.; Rosenberg, J.; Sonpavde, G.; Hutson, T.; Oh, W.K.; Dreicer, R.; Vogelzang, N.; Sternberg, C.N.; Bajorin, D.F.; et al. Treatment of Patients with Metastatic Urothelial Cancer “Unfit” for Cisplatin-Based Chemotherapy. J. Clin. Oncol. 2011, 29, 2432–2438. [Google Scholar] [PubMed]
  84. Witjes, J.A.; Bruins, H.M.; Cathomas, R.; Compérat, E.M.; Cowan, N.C.; Gakis, G.; Hernández, V.; Linares Espinós, E.; Lorch, A.; Neuzillet, Y.; et al. European Association of Urology Guidelines on Muscle-Invasive and Metastatic Bladder Cancer: Summary of the 2020 Guidelines. Eur. Urol. 2021, 79, 82–104. [Google Scholar]
  85. Chen, R.C.; Shipley, W.U.; Efstathiou, J.A.; Zietman, A.L. Trimodality Bladder Preservation Therapy for Muscle-Invasive Bladder Cancer. JNCCN J. Natl. Compr. Cancer Netw. 2013, 11, 952–960. [Google Scholar] [CrossRef]
  86. Donat, S.M.; Shabsigh, A.; Savage, C.; Cronin, A.M.; Bochner, B.H.; Dalbagni, G.; Herr, H.W.; Milowsky, M.I. Potential Impact of Postoperative Early Complications on the Timing of Adjuvant Chemotherapy in Patients Undergoing Radical Cystectomy: A High-Volume Tertiary Cancer Center Experience. Eur. Urol. 2009, 55, 177–185. [Google Scholar] [CrossRef]
  87. Shabsigh, A.; Korets, R.; Vora, K.C.; Brooks, C.M.; Cronin, A.M.; Savage, C.; Raj, G.; Bochner, B.H.; Dalbagni, G.; Herr, H.W.; et al. Defining Early Morbidity of Radical Cystectomy for Patients with Bladder Cancer Using a Standardized Reporting Methodology. Eur. Urol. 2009, 55, 164–174. [Google Scholar] [CrossRef]
  88. James, N.D.; Hussain, S.A.; Hall, E.; Jenkins, P.; Tremlett, J.; Rawlings, C.; Crundwell, M.; Sizer, B.; Sreenivasan, T.; Hendron, C.; et al. Radiotherapy with or without Chemotherapy in Muscle-Invasive Bladder Cancer. N. Engl. J. Med. 2012, 366, 1477–1488. [Google Scholar] [CrossRef] [PubMed]
  89. Arcangeli, G.; Arcangeli, S.; Strigari, L. A Systematic Review and Meta-Analysis of Clinical Trials of Bladder-Sparing Trimodality Treatment for Muscle-Invasive Bladder Cancer (MIBC). Crit. Rev. Oncol. Hematol. 2015, 94, 105–115. [Google Scholar] [PubMed]
  90. Mak, R.H.; Hunt, D.; Shipley, W.U.; Efstathiou, J.A.; Tester, W.J.; Hagan, M.P.; Kaufman, D.S.; Heney, N.M.; Zietman, A.L. Long-Term Outcomes in Patients with Muscle-Invasive Bladder Cancer after Selective Bladder-Preserving Combined-Modality Therapy: A Pooled Analysis of Radiation Therapy Oncology Group Protocols 8802, 8903, 9506, 9706, 9906, and 0233. J. Clin. Oncol. 2014, 32, 3801–3809. [Google Scholar] [CrossRef]
  91. Griffiths, G. International Phase III Trial Assessing Neoadjuvant Cisplatin, Methotrexate, and Vinblastine Chemotherapy for Muscle-Invasive Bladder Cancer: Long-Term Results of the BA06 30894 Trial. J. Clin. Oncol. 2011, 29, 2171–2177. [Google Scholar] [CrossRef]
  92. Kapoor, A.; Niazi, T.; Noonan, K.; Rendon, R.A.; Alimohamed, N.; Kassouf, W.; Berlin, A.; Chu, W.; Kollmannsberger, C.; So, A.I. 2022 American Society of Clinical Oncology (ASCO) Genitourinary Cancers Symposium: Meeting Highlights. Yosetsu Gakkai Shi/J. Jpn. Weld. Soc. 2022, 16, 125–131. [Google Scholar] [CrossRef]
  93. Plimack, E.R.; Dunbrack, R.L.; Brennan, T.A.; Andrake, M.D.; Zhou, Y.; Serebriiskii, I.G.; Slifker, M.; Alpaugh, K.; Dulaimi, E.; Palma, N.; et al. Defects in DNA Repair Genes Predict Response to Neoadjuvant Cisplatin-Based Chemotherapy in Muscle-Invasive Bladder Cancer. Eur. Urol. 2015, 68, 959–967. [Google Scholar] [CrossRef] [PubMed]
  94. Geynisman, D.M.; Abbosh, P.; Ross, E.A.; Zibelman, M.R.; Ghatalia, P.; Anari, F.; Ansel, K.; Mark, J.R.; Stamatakis, L.; Hoffman-Censits, J.H.; et al. A Phase II Trial of Risk-Enabled Therapy after Initiating Neoadjuvant Chemotherapy for Bladder Cancer (RETAIN). J. Clin. Oncol. 2023, 41, 438. [Google Scholar] [CrossRef]
  95. Geynisman, D.M.; Abbosh, P.; Ross, E.A.; Zibelman, M.R.; Ghatalia, P.; Anari, F.; Ansel, K.; Mark, J.R.; Stamatakis, L.; Hoffman-Censits, J.H.; et al. A Phase II Trial of Risk Enabled Therapy after Initiating Neoadjuvant Chemotherapy for Bladder Cancer (RETAIN BLADDER): Interim Analysis. J. Clin. Oncol. 2021, 39, 397. [Google Scholar] [CrossRef]
  96. Babjuk, M.; Burger, M.; Compérat, E.M.; Gontero, P.; Mostafid, A.H.; Palou, J.; van Rhijn, B.W.G.; Rouprêt, M.; Shariat, S.F.; Sylvester, R.; et al. European Association of Urology Guidelines on Non-Muscle-Invasive Bladder Cancer (TaT1 and Carcinoma In Situ)—2019 Update. Eur. Urol. 2019, 76, 639–657. [Google Scholar]
  97. Chang, S.S.; Bochner, B.H.; Chou, R.; Dreicer, R.; Kamat, A.M.; Lerner, S.P.; Lotan, Y.; Meeks, J.J.; Michalski, J.M.; Morgan, T.M.; et al. Treatment of Non-Metastatic Muscle-Invasive Bladder Cancer: AUA/ASCO/ASTRO/SUO Guideline. J. Urol. 2017, 198, 552–559. [Google Scholar] [CrossRef] [PubMed]
  98. Yu, H.; Friedlander, D.F.; Patel, S.; Hu, J.C. The Current Status of Robotic Oncologic Surgery. CA Cancer J. Clin. 2013, 63, 45–56. [Google Scholar] [CrossRef]
  99. Falagario, U.; Veccia, A.; Weprin, S.; Albuquerque, E.V.; Nahas, W.; Carrieri, G.; Pansadoro, V.; Hampton, L.; Porpiglia, F.; Autorino, R. Robotic-Assisted Surgery for the Treatment of Urologic Cancers: Recent Advances. Expert Rev. Med. Devices 2020, 17, 579–590. [Google Scholar]
  100. Jocham, D.; von Wietersheim, J.; Pflüger, H.; Steiner, H.; Doehn, C.; Büttner, H.; Böhle, A.; Kausch, I. BCG versus Photodynamic Therapy (PDT) for Nonmuscle Invasive Bladder Cancer—A Multicentre Clinical Phase III Study. Aktuelle Urol. 2009, 40, 91–99. [Google Scholar]
  101. Von der Maase, H.; Hansen, S.W.; Roberts, J.T.; Dogliotti, L.; Oliver, T.; Moore, M.J.; Bodrogi, I.; Albers, P.; Knuth, A.; Lippert, C.M.; et al. Gemcitabine and Cisplatin versus Methotrexate, Vinblastine, Doxorubicin, and Cisplatin in Advanced or Metastatic Bladder Cancer: Results of a Large, Randomized, Multinational, Multicenter, Phase III Study. J. Clin. Oncol. 2000, 18, 3068–3077. [Google Scholar] [CrossRef]
  102. De Santis, M.; Bellmunt, J.; Mead, G.; Kerst, J.M.; Leahy, M.; Maroto, P.; Gil, T.; Marreaud, S.; Daugaard, G.; Skoneczna, I.; et al. Randomized Phase II/III Trial Assessing Gemcitabine/Carboplatin and Methotrexate/Carboplatin/Vinblastine in Patients with Advanced Urothelial Cancer Who Are Unfit for Cisplatin-Based Chemotherapy: EORTC Study 30986. J. Clin. Oncol. 2012, 30, 191–199. [Google Scholar] [CrossRef]
  103. Gitlitz, B.J.; Baker, C.; Chapman, Y.; Allen, H.J.; Bosserman, L.D.; Patel, R.; Sanchez, J.D.; Shapiro, R.M.; Figlin, R.A. A Phase II Study of Gemcitabine and Docetaxel Therapy in Patients with Advanced Urothelial Carcinoma. Cancer 2003, 98, 1863–1869. [Google Scholar] [CrossRef] [PubMed]
  104. Witte, R.S.; Elson, P.; Bono, B.; Knop, R.; Richardson, R.R.; Dreicer, R.; Loehrer, P.J. Eastern Cooperative Oncology Group Phase II Trial of Ifosfamide in the Treatment of Previously Treated Advanced Urothelial Carcinoma. J. Clin. Oncol. 1997, 15, 589–593. [Google Scholar] [CrossRef] [PubMed]
  105. Hong, M.; He, G.; Goh, S.; Low, A.W.X.; Tay, K.J.; Lim, T.K.H.; Yeong, J.; Khor, L.Y.; Lim, T.S. Biomarkers for Precision Urothelial Carcinoma Diagnosis: Current Approaches and the Application of Single-Cell Technologies. Cancers 2021, 13, 260. [Google Scholar] [CrossRef]
  106. Loriot, Y.; Necchi, A.; Park, S.H.; Garcia-Donas, J.; Huddart, R.; Burgess, E.; Fleming, M.; Rezazadeh, A.; Mellado, B.; Varlamov, S.; et al. Erdafitinib in Locally Advanced or Metastatic Urothelial Carcinoma. N. Engl. J. Med. 2019, 381, 338–348. [Google Scholar] [CrossRef]
  107. Bedke, J.; Maas, M. Re: Enfortumab Vedotin in Previously Treated Advanced Urothelial Carcinoma. Eur. Urol. 2021, 80, 6017. [Google Scholar]
  108. Zibelman, M.; Ramamurthy, C.; Plimack, E.R. Emerging Role of Immunotherapy in Urothelial Carcinoma—Advanced Disease. Urol. Oncol. Semin. Orig. Investig. 2016, 34, 538–547. [Google Scholar]
  109. Stenehjem, D.D.; Tran, D.; Nkrumah, M.A.; Gupta, S. PD1/PDL1 Inhibitors for the Treatment of Advanced Urothelial Bladder Cancer. Oncol. Targets Ther. 2018, 11, 5973–5989. [Google Scholar]
  110. Bellmunt, J.; de Wit, R.; Vaughn, D.J.; Fradet, Y.; Lee, J.-L.; Fong, L.; Vogelzang, N.J.; Climent, M.A.; Petrylak, D.P.; Choueiri, T.K.; et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N. Engl. J. Med. 2017, 376, 1015–1026. [Google Scholar] [CrossRef]
  111. Powles, T.; Park, S.H.; Voog, E.; Caserta, C.; Valderrama, B.P.; Gurney, H.; Kalofonos, H.; Radulović, S.; Demey, W.; Ullén, A.; et al. Avelumab Maintenance Therapy for Advanced or Metastatic Urothelial Carcinoma. N. Engl. J. Med. 2020, 383, 1218–1230. [Google Scholar] [CrossRef]
  112. Bajorin, D.F.; Witjes, J.A.; Gschwend, J.E.; Schenker, M.; Valderrama, B.P.; Tomita, Y.; Bamias, A.; Lebret, T.; Shariat, S.F.; Park, S.H.; et al. Adjuvant Nivolumab versus Placebo in Muscle-Invasive Urothelial Carcinoma. N. Engl. J. Med. 2021, 384, 2102–2114. [Google Scholar] [CrossRef]
  113. Galsky, M.D.; Arija, J.Á.A.; Bamias, A.; Davis, I.D.; De Santis, M.; Kikuchi, E.; Garcia-del-Muro, X.; De Giorgi, U.; Mencinger, M.; Izumi, K.; et al. Atezolizumab with or without Chemotherapy in Metastatic Urothelial Cancer (IMvigor130): A Multicentre, Randomised, Placebo-Controlled Phase 3 Trial. Lancet 2020, 395, 1547–1557. [Google Scholar] [CrossRef]
  114. Vlachostergios, P.J.; Faltas, B.M. Treatment resistance in urothelial carcinoma: An evolutionary perspective. Nat. Rev. Clin. Oncol. 2018, 15, 495–509. [Google Scholar] [PubMed]
  115. Massari, F.; Santoni, M.; Ciccarese, C.; Brunelli, M.; Conti, A.; Santini, D.; Montironi, R.; Cascinu, S.; Tortora, G. Emerging concepts on drug resistance in bladder cancer: Implications for future strategies. Crit. Rev. Oncol. Hematol. 2015, 96, 81–90. [Google Scholar] [PubMed]
  116. Marusyk, A.; Janiszewska, M.; Polyak, K. Intratumor Heterogeneity: The Rosetta Stone of Therapy Resistance. Cancer Cell. 2020, 37, 471–484. [Google Scholar] [CrossRef]
  117. Padua, T.C.; Moschini, M.; Martini, A.; Pederzoli, F.; Nocera, L.; Marandino, L.; Raggi, D.; Briganti, A.; Montorsi, F.; Necchi, A. Efficacy and toxicity of antibody-drug conjugates in the treatment of metastatic urothelial cancer: A scoping review. Urol. Oncol. 2022, 40, 413–423. [Google Scholar]
  118. Hoimes, C.J.; Flaig, T.W.; Milowsky, M.I.; Friedlander, T.W.; Bilen, M.A.; Gupta, S.; Srinivas, S.; Merchan, J.R.; McKay, R.R.; Petrylak, D.P.; et al. Enfortumab Vedotin Plus Pembrolizumab in Previously Untreated Advanced Urothelial Cancer. J. Clin. Oncol. 2023, 41, 22–31. [Google Scholar] [CrossRef] [PubMed]
  119. Powles, T.; Rosenberg, J.E.; Sonpavde, G.P.; Loriot, Y.; Durán, I.; Lee, J.L.; Matsubara, N.; Vulsteke, C.; Castellano, D.; Wu, C.; et al. Enfortumab Vedotin in Previously Treated Advanced Urothelial Carcinoma. N. Engl. J. Med. 2021, 384, 1125–1135. [Google Scholar] [CrossRef]
  120. 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]
  121. Tagawa, S.T.; Balar, A.V.; Petrylak, D.P.; Kalebasty, A.R.; Loriot, Y.; Fléchon, A.; Jain, R.K.; Agarwal, N.; Bupathi, M.; Barthelemy, P.; 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. J. Clin. Oncol. 2021, 39, 2474–2485. [Google Scholar] [CrossRef]
  122. Petrulio, C.A.; Kaufman, H.L. Development of the PANVACTM-VF Vaccine for Pancreatic Cancer. Expert Rev. Vaccines 2006, 5, 9–19. [Google Scholar]
  123. Grandi, P.; Darilek, A.; Moscu, A.; Pradhan, A.; Li, R. Intravesical Infusion of Oncolytic Virus CG0070 in the Treatment of Bladder Cancer. Methods Mol. Biol. 2023, 2684, 303–317. [Google Scholar] [PubMed]
  124. Kloss, C.C.; Condomines, M.; Cartellieri, M.; Bachmann, M.; Sadelain, M. Combinatorial Antigen Recognition with Balanced Signaling Promotes Selective Tumor Eradication by Engineered T Cells. Nat. Biotechnol. 2013, 31, 71–75. [Google Scholar] [CrossRef]
  125. Zhang, Z.; Li, D.; Yun, H.; Liu, W.; Chai, K.; Tong, J.; Zeng, T.; Gao, Z.; Xie, Y. CAR-T Cells in the Treatment of Urologic Neoplasms: Present and Future. Front. Oncol. 2022, 12, 915171. [Google Scholar] [CrossRef] [PubMed]
  126. Galsky, M.D.; Powles, T.; Li, S.; Hennicken, D.; Sonpavde, G. A phase 3, open-label, randomized study of nivolumab plus ipilimumab or standard of care (SOC) versus SOC alone in patients (pts) with previously untreated unresectable or metastatic urothelial carcinoma (mUC.; CheckMate 901). J. Clin. Oncol. 2018, 36, TPS539. [Google Scholar]
  127. Powles, T.; Gschwend, J.E.; Loriot, Y.; Bellmunt, J.; Geczi, L.; Vulsteke, C.; Abdelsalam, M.; Gafanov, R.; Bae, W.K.; Revesz, J.; et al. Phase 3 KEYNOTE-361 trial: Pembrolizumab (pembro) with or without chemotherapy versus chemotherapy alone in advanced urothelial cancer. J. Clin. Oncol. 2017, 35, TPS4590. [Google Scholar]
  128. Jackson-Spence, F.E.; Powles, T.; Loriot, Y.; Toms, C.; Jovaisaite, A.; Choy, J.; Szabados, B.E.; Nudds, H.; Ackerman, C.; Mousa, K. P126 Trial In Progress: DISCUS: A randomized phase II study comparing 3 vs. 6 cycles of platinum-based chemotherapy prior to maintenance avelumab in advanced urothelial cancer. Eur. Urol. Open Sci. 2022, 42, S219. [Google Scholar] [CrossRef]
  129. Gupta, S.; Ballman, K.V.; Galsky, M.D.; Morris, M.J.; Chen, R.C.; Chan, T.A.; Dercle, L.; Wen, Y.; Sridhar, S.S.; Yen, A.R.; et al. MAIN-CAV: Phase III randomized trial of maintenance cabozantinib and avelumab versus avelumab after first-line platinum-based chemotherapy in patients with metastatic urothelial cancer (mUC) (Alliance A032001). J. Clin. Oncol. 2022, 40, TPS4607. [Google Scholar]
  130. Petrylak, D.; de Wit, R.; Chi, K.N.; Drakaki, A.; Sternberg, C.N.; Nishiyama, H.; Castellano, D.; Hussain, S.; Fléchon, A.; Bamias, A.; et al. Ramucirumab plus Docetaxel versus Placebo plus Docetaxel in Patients with Locally Advanced or Metastatic Urothelial Carcinoma after Platinum-Based Therapy (RANGE): A Randomised, Double-Blind, Phase 3 Trial. Lancet 2017, 390, 2266–2277. [Google Scholar] [CrossRef]
  131. Rosenberg, J.E.; Ballman, K.A.; Halabi, S.; Atherton, P.J.; Mortazavi, A.; Sweeney, C.; Stadler, W.M.; Teply, B.A.; Picus, J.; Tagawa, S.T.; et al. Randomized Phase III Trial of Gemcitabine and Cisplatin with Bevacizumab or Placebo in Patients With Advanced Urothelial Carcinoma: Results of CALGB 90601 (Alliance). J. Clin. Oncol. 2021, 39, 2486–2496. [Google Scholar] [CrossRef]
  132. Apolo, A.B.; Nadal, R.; Tomita, Y.; Davarpanah, N.N.; Cordes, L.M.; Steinberg, S.M.; Cao, L.; Parnes, H.L.; Costello, R.; Merino, M.J.; et al. Cabozantinib in Patients with Platinum-Refractory Metastatic Urothelial Carcinoma: An Open-Label, Single-Centre, Phase 2 Trial. Lancet Oncol. 2020, 21, 1099–1109. [Google Scholar] [CrossRef]
  133. Ullah, A.; Ullah, N.; Nawaz, T.; Aziz, T. Molecular Mechanisms of Sanguinarine in Cancer Prevention and Treatment. Anticancer Agents Med. Chem. 2023, 23, 765–778. [Google Scholar] [PubMed]
  134. Desai, N.B.; Zabor, E.C.; Cha, E.K.; Hreiki, J.; Sfakianos, J.P.; Ramirez, R.; Bagrodia, A.; Rosenberg, J.E.; Bajorin, D.F.; Berger, M.F.; et al. Genomic characterization of response to chemoradiation in urothelial bladder cancer. Cancer 2016, 122, 3715–3723. [Google Scholar] [PubMed]
  135. Hedegaard, J.; Lamy, P.; Nordentoft, I.; Algaba, F.; Høyer, S.; Ulhøi, B.P.; Vang, S.; Reinert, T.; Hermann, G.G.; Mogensen, K.; et al. Comprehensive Transcriptional Analysis of Early-Stage Urothelial Carcinoma. Cancer Cell 2016, 30, 27–42. [Google Scholar] [PubMed]
  136. Fong, M.H.Y.; Feng, M.; McConkey, D.J.; Choi, W. Update on bladder cancer molecular subtypes. Transl. Androl. Urol. 2020, 9, 2881–2889. [Google Scholar] [CrossRef]
  137. Williams, S.B.; Black, P.C.; Dyrskjøt, L.; Seiler, R.; Schmitz-Dräger, B.; Nawroth, R.; Todenhöfer, T.; Kamat, A.M.; Goebell, P.J. Re: Aurélie Kamoun, Aurélien de Reyniès, Yves Allory, et al. A Consensus Molecular Classification of Muscle-invasive Bladder Cancer. Eur. Urol. 2020, 77, 420–433. A Statement from the International Bladder Cancer Network. Eur. Urol. 2020, 77, e105–e106. [Google Scholar] [CrossRef]
  138. Alifrangis, C.; McGovern, U.; Freeman, A.; Powles, T.; Linch, M. Molecular and histopathology directed therapy for advanced bladder cancer. Nat. Rev. Urol. 2019, 16, 465–483. [Google Scholar] [CrossRef] [PubMed]
  139. Faltas, B.M.; Prandi, D.; Tagawa, S.T.; Molina, A.M.; Nanus, D.M.; Sternberg, C.; Rosenberg, J.; Mosquera, J.M.; Robinson, B.; Elemento, O.; et al. Clonal evolution of chemotherapy-resistant urothelial carcinoma. Nat. Genet. 2016, 48, 1490–1499. [Google Scholar] [PubMed]
  140. Mohanty, S.K.; Lobo, A.; Mishra, S.K.; Cheng, L. Precision Medicine in Bladder Cancer: Present Challenges and Future Directions. J. Pers. Med. 2023, 13, 756. [Google Scholar] [CrossRef] [PubMed]
  141. Marzouka, N.A.; Eriksson, P.; Bernardo, C.; Hurst, C.D.; Knowles, M.A.; Sjödahl, G.; Liedberg, F.; Höglund, M. The Lund Molecular Taxonomy Applied to Non-Muscle-Invasive Urothelial Carcinoma. J. Mol. Diagn. 2022, 24, 992–1008. [Google Scholar] [CrossRef] [PubMed]
  142. Bernardo, C.; Eriksson, P.; Marzouka, N.A.; Liedberg, F.; Sjödahl, G.; Höglund, M. Molecular pathology of the luminal class of urothelial tumors. J. Pathol. 2019, 249, 308–318. [Google Scholar] [CrossRef]
  143. Van Allen, E.M.; Mouw, K.W.; Kim, P.; Iyer, G.; Wagle, N.; Al-Ahmadie, H.; Zhu, C.; Ostrovnaya, I.; Kryukov, G.V.; O’Connor, K.W.; et al. Somatic ERCC2 mutations correlate with cisplatin sensitivity in muscle-invasive urothelial carcinoma. Cancer Discov. 2014, 4, 1140–1153. [Google Scholar]
  144. Sarfaty, M.; Teo, M.Y.; Al-Ahmadie, H.; Funt, S.A.; Lee, C.-H.; Aggen, D.H.; Solit, D.B.; Ratna, N.; Regazzi, A.M.; Hechtman, J.F.; et al. Microsatellite instability (MSI-H) in metastatic urothelial carcinoma (mUC): A biomarker of divergent responses to systemic therapy. J. Clin. Oncol. 2020, 38 (Suppl. 6), 566. [Google Scholar]
  145. Sarfaty, M.; Golkaram, M.; Funt, S.A.; Al-Ahmadie, H.; Kaplan, S.; Song, F.; Regazzi, A.; Makarov, V.; Kuo, F.; Ostrovnaya, I.; et al. Novel Genetic Subtypes of Urothelial Carcinoma With Differential Outcomes on Immune Checkpoint Blockade. J. Clin. Oncol. 2023, 41, 3225–3235. [Google Scholar] [CrossRef] [PubMed]
  146. Queiroz, M.M.; de Souza, Z.S.; Gongora, A.B.L.; de Galiza Barbosa, F.; Buchpiguel, C.A.; de Castro, M.G.; de Macedo, M.P.; Coelho, R.F.; Sokol, E.S.; Camargo, A.A.; et al. Emerging biomarkers in metastatic urothelial carcinoma: Tumour mutational burden, PD-L1 expression and APOBEC polypeptide-like signature in a patient with complete response to anti-programmed cell death protein-1 inhibitor. Ecancermedicalscience 2021, 15, 1306. [Google Scholar] [CrossRef] [PubMed]
  147. Goswami, S.; Chen, Y.; Anandhan, S.; Szabo, P.M.; Basu, S.; Blando, J.M.; Liu, W.; Zhang, J.; Natarajan, S.M.; Xiong, L.; et al. ARID1A mutation plus CXCL13 expression act as combinatorial biomarkers to predict responses to immune checkpoint therapy in mUCC. Sci. Transl. Med. 2020, 12, eabc4220. [Google Scholar] [PubMed]
  148. Xu, N.; Yao, Z.; Shang, G.; Ye, D.; Wang, H.; Zhang, H.; Qu, Y.; Xu, F.; Wang, Y.; Qin, Z.; et al. Integrated proteogenomic characterization of urothelial carcinoma of the bladder. J. Hematol. Oncol. 2022, 15, 76. [Google Scholar] [CrossRef]
  149. Seiler, R.; Thalmann, G.N.; Rotzer, D.; Perren, A.; Fleischmann, A. CCND1/CyclinD1 status in metastasizing bladder cancer: A prognosticator and predictor of chemotherapeutic response. Mod. Pathol. 2014, 27, 87–95. [Google Scholar] [CrossRef]
  150. Krook, M.A.; Reeser, J.W.; Ernst, G.; Barker, H.; Wilberding, M.; Li, G.; Chen, H.-Z.; Roychowdhury, S. Fibroblast growth factor receptors in cancer: Genetic alterations, diagnostics, therapeutic targets and mechanisms of resistance. Br. J. Cancer 2021, 124, 880–892. [Google Scholar] [CrossRef] [PubMed]
  151. Rodler, S.; Eismann, L.; Schlenker, B.; Casuscelli, J.; Brinkmann, I.; Sendelhofert, A.; Waidelich, R.; Buchner, A.; Stief, C.; Schulz, G.B.; et al. Expression of Nectin-4 in Variant Histologies of Bladder Cancer and Its Prognostic Value-Need for Biomarker Testing in High-Risk Patients? Cancers 2022, 14, 4411. [Google Scholar] [CrossRef]
  152. Wong, J.L.; Rosenberg, J.E. Targeting nectin-4 by antibody-drug conjugates for the treatment of urothelial carcinoma. Expert Opin. Biol. Ther. 2021, 21, 863–873. [Google Scholar] [CrossRef]
  153. Zimpfer, A.; Kdimati, S.; Mosig, M.; Rudolf, H.; Zettl, H.; Erbersdobler, A.; Hakenberg, O.W.; Maruschke, M.; Schneider, B. ERBB2 Amplification as a Predictive and Prognostic Biomarker in Upper Tract Urothelial Carcinoma. Cancers 2023, 15, 2414. [Google Scholar] [CrossRef]
  154. Mendiratta, P.; Grivas, P. Emerging biomarkers and targeted therapies in urothelial carcinoma. Ann. Transl. Med. 2018, 6, 250. [Google Scholar] [CrossRef] [PubMed]
  155. Tripathi, A.; Grivas, P. The utility of next generation sequencing in advanced urothelial carcinoma. Eur. Urol. Focus 2020, 6, 41–44. [Google Scholar] [CrossRef]
  156. Peng, J.; Sridhar, S.; Siefker-Radtke, A.O.; Selvarajah, S.; Jiang, D.M. Targeting the FGFR Pathway in Urothelial Carcinoma: The Future Is Now. Curr. Treat Opt. Oncol. 2022, 23, 1269–1287. [Google Scholar] [CrossRef]
  157. Doroshow, D.B.; O’Donnell, P.H.; Hoffman-Censits, J.H.; Gupta, S.V.; Vaishampayan, U.; Heath, E.I.; Garcia, P.; Zhao, Q.; Yu, M.; Milowsky, M.I.; et al. Phase II Trial of Olaparib in Patients with Metastatic Urothelial Cancer Harboring DNA Damage Response Gene Alterations. JCO Precis. Oncol. 2023, 7, e2300095. [Google Scholar] [CrossRef] [PubMed]
  158. Tripathi, A.; Lerner, S.P. Poly (ADP-ribose) Polymerase Inhibition in Advanced Urothelial Carcinoma. JCO Precis. Oncol. 2023, 7, e2300293. [Google Scholar] [CrossRef]
  159. Torres-Jiménez, J.; Albarrán-Fernández, V.; Pozas, J.; Román-Gil, M.S.; Esteban-Villarrubia, J.; Carrato, A.; Rosero, A.; Grande, E.; Alonso-Gordoa, T.; Molina-Cerrillo, J. Novel Tyrosine Kinase Targets in Urothelial Carcinoma. Int. J. Mol. Sci. 2021, 22, 747. [Google Scholar] [CrossRef]
  160. Schmidt, A.L.; Siefker-Radtke, A.; McConkey, D.; McGregor, B. Renal Cell and Urothelial Carcinoma: Biomarkers for New Treatments. Am. Soc. Clin. Oncol. Educ. Book 2020, 40, e197–e206. [Google Scholar] [CrossRef]
  161. Narayanan, S.; Srinivas, S. Incorporating VEGF-targeted therapy in advanced urothelial cancer. Ther. Adv. Med. Oncol. 2017, 9, 33–45. [Google Scholar] [CrossRef]
  162. Gallagher, D.J.; Milowsky, M.I.; Gerst, S.R.; Ishill, N.; Riches, J.; Regazzi, A.; Boyle, M.G.; Trout, A.; Flaherty, A.M.; Bajorin, D.F. Phase II study of sunitinib in patients with metastatic urothelial cancer. J. Clin. Oncol. 2010, 28, 1373–1379. [Google Scholar]
  163. Geldart, T.; Chester, J.; Casbard, A.; Crabb, S.; Elliott, T.; Protheroe, A.; Huddart, R.A.; Mead, G.; Barber, J.; Jones, R.J.; et al. SUCCINCT: An open-label, single-arm, non-randomised, phase 2 trial of gemcitabine and cisplatin chemotherapy in combination with sunitinib as first-line treatment for patients with advanced urothelial carcinoma. Eur. Urol. 2015, 67, 599–602. [Google Scholar]
  164. Bellmunt, J.; Powles, T.B.; van der Heijden, M.S.; Galsky, M.D.; He, P.; Wang, X.; Xiao, F.; Jones, F.; Scott, M.; Walker, J.; et al. 708P PD-L1 as a predictor of survival in patients with metastatic urothelial carcinoma (mUC) from the phase III DANUBE trial of durvalumab (D) or durvalumab plus tremelimumab (D + T) versus standard of care chemotherapy (SoC). Ann. Oncol. 2021, 32, S716–S717. [Google Scholar]
  165. Goodman, A.M.; Piccioni, D.; Kato, S.; Boichard, A.; Wang, H.Y.; Frampton, G.; Lippman, S.M.; Connelly, C.; Fabrizio, D.; Miller, V.; et al. Prevalence of PDL1 amplification and preliminary response to immune checkpoint blockade in solid tumors. JAMA Oncol. 2018, 4, 1237–1244. [Google Scholar]
  166. Graf, R.P.; Fisher, V.; Huang, R.S.P.; Hamdani, O.; Gjoerup, O.V.; Stanke, J.; Creeden, J.; Levy, M.A.; Oxnard, G.R.; Gupta, S. Tumor Mutational Burden as a Predictor of First-Line Immune Checkpoint Inhibitor Versus Carboplatin Benefit in Cisplatin-Unfit Patients with Urothelial Carcinoma. JCO Precis. Oncol. 2022, 6, e2200121. [Google Scholar] [CrossRef]
  167. Van der Fels, C.A.M.; Leliveld, A.; Buikema, H.; van den Heuvel, M.C.; 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]
  168. Huang, R.S.P.; Haberberger, J.; Severson, E.; Duncan, D.L.; Hemmerich, A.; Edgerly, C.; Ferguson, N.L.; Williams, E.; Elvin, J.; Vergilio, J.-A.; et al. A pan-cancer analysis of PD-L1 immunohistochemistry and gene amplification, tumor mutation burden and microsatellite instability in 48,782 cases. Mod. Pathol. 2021, 34, 252–263. [Google Scholar]
  169. Koutros, S.; Rao, N.; Moore, L.E.; Nickerson, M.L.; Lee, D.; Zhu, B.; Pardo, L.A.; Baris, D.; Schwenn, M.; Johnson, A.; et al. Targeted deep sequencing of bladder tumors reveals novel associations between cancer gene mutations and mutational signatures with major risk factors. Clin. Cancer Res. 2021, 27, 3725–3733. [Google Scholar] [PubMed]
  170. Hoffman-Censits, J.H.; Lombardo, K.A.; Parimi, V.; Kamanda, S.; Choi, W.; Hahn, N.M.; McConkey, D.J.; McGuire, B.M.; Bivalacqua, T.J.; Kates, M.; et al. Expression of nectin-4 in bladder urothelial carcinoma, in morphologic variants, and nonurothelial histotypes. Appl. Immunohistochem. Mol. Morphol. 2021, 29, 619–625. [Google Scholar]
  171. Sarafidis, M.; Lambrou, G.I.; Zoumpourlis, V.; Koutsouris, D. An Integrated Bioinformatics Analysis towards the Identification of Diagnostic, Prognostic, and Predictive Key Biomarkers for Urinary Bladder Cancer. Cancers 2022, 14, 3358. [Google Scholar] [CrossRef]
  172. Eckstein, M.; Cimadamore, A.; Hartmann, A.; Lopez-Beltran, A.; Cheng, L.; Scarpelli, M.; Montironi, R.; Gevaert, T. PD-L1 assessment in urothelial carcinoma: A practical approach. Ann. Transl. Med. 2019, 7, 690. [Google Scholar] [CrossRef]
  173. Wang, L.; Sfakianos, J.P.; Beaumont, K.G.; Akturk, G.; Horowitz, A.; Sebra, R.P.; Farkas, A.M.; Gnjatic, S.; Hake, A.; Izadmehr, S.; et al. Myeloid Cell-associated Resistance to PD-1/PD-L1 Blockade in Urothelial Cancer Revealed Through Bulk and Single-cell RNA Sequencing. Clin. Cancer Res. 2021, 27, 4287–4300. [Google Scholar] [CrossRef] [PubMed]
  174. Pfannstiel, C.; Strissel, P.L.; Chiappinelli, K.B.; Sikic, D.; Wach, S.; Wirtz, R.M.; Wullweber, A.; Taubert, H.; Breyer, J.; Otto, W.; et al. The tumor immune microenvironment drives a prognostic relevance that correlates with bladder cancer subtypes. Cancer Immunol. Res. 2019, 7, 923–938. [Google Scholar]
  175. Sarhadi, V.K.; Armengol, G. Molecular Biomarkers in Cancer. Biomolecules 2022, 12, 1021. [Google Scholar] [CrossRef]
  176. Lourenço, C.; Constâncio, V.; Henrique, R.; Carvalho, Â.; Jerónimo, C. Urinary Extracellular Vesicles as Potential Biomarkers for Urologic Cancers: An Overview of Current Methods and Advances. Cancers 2021, 13, 1529. [Google Scholar] [CrossRef]
  177. Hölzel, M.; Bovier, A.; Tüting, T. Plasticity of tumour and immune cells: A source of heterogeneity and a cause for therapy resistance? Nat. Rev. Cancer 2013, 13, 365–376. [Google Scholar]
  178. Gupta, S.; Bellmunt, J.; Plimack, E.R.; Sonpavde, G.P.; Grivas, P.; Apolo, A.B.; Pal, S.K.; Siefker-Radtke, A.O.; Flaig, T.W.; Galsky, M.D.; et al. Defining “platinum-ineligible” patients with metastatic urothelial cancer (mUC). J. Clin. Oncol. 2022, 40 (Suppl. S16), 4577. [Google Scholar]
  179. Zhu, A.; Garcia, J.A.; Faltas, B.; Grivas, P.; Barata, P.; Shoag, J.E. Immune Checkpoint Inhibitors and Long-term Survival of Patients With Metastatic Urothelial Cancer. JAMA Netw. Open 2023, 6, e237444. [Google Scholar]
  180. Sun, J.Y.; Zhang, D.; Wu, S.; Xu, M.; Zhou, X.; Lu, X.; Ji, J. Resistance to PD-1/PD-L1 blockade cancer immunotherapy: Mechanisms, predictive factors, and future perspectives. Biomark. Res. 2020, 8, 35. [Google Scholar] [CrossRef]
  181. Yang, Z.; Xu, Y.; Bi, Y.; Zhang, N.; Wang, H.; Xing, T.; Bai, S.; Shen, Z.; Naz, F.; Zhang, Z.; et al. Immune escape mechanisms and immunotherapy of urothelial bladder cancer. J. Clin. Transl. Res. 2021, 7, 485–500. [Google Scholar] [PubMed]
  182. Özdemir, B.C.; Siefker-Radtke, A.O.; Campbell, M.T.; Subudhi, S.K. Current and Future Applications of Novel Immunotherapies in Urological Oncology: A Critical Review of the Literature. Eur. Urol. Focus 2018, 4, 442–454. [Google Scholar] [CrossRef]
  183. Guercio, B.J.; Iyer, G.; Rosenberg, J.E. Developing Precision Medicine for Bladder Cancer. Hematol. Oncol. Clin. N. Am. 2021, 35, 633–653. [Google Scholar] [CrossRef]
Table 1. Frequency of genetic alterations in select genes for MIBC tumors. Compiled and adapted from references [25,29,30,31,32,33,34,35,36,37,38,39].
Table 1. Frequency of genetic alterations in select genes for MIBC tumors. Compiled and adapted from references [25,29,30,31,32,33,34,35,36,37,38,39].
GeneChromosomal
Location
Genetic
Alteration
Frequency Observed in MIBC Tumors
Chromosome
9pDeletion21–30%
9qDeletion17%
Oncogenes
HRAS11p15Activating mutation10–15%
FGFR34p16Activating mutation~50% Overexpression
15% Mutation
PIK3CA3q26Activating mutation25%
MDM212q13Overexpression4% Overexpression
Tumor suppressor genes
TP5317p13Deletion or mutation70%
RB113q14Deletion or mutation37%
PTEN10q23Homozygous deletion or mutationLOH 30–35%
Mutation 17%
CDKN2A9p21Homozygous deletion or methylation or mutationHD 20–30%
LOH ~60%
PTCH9q22Deletion or mutationLOH ~60%
Mutation rare
DBC19q32–33Deletion or methylationLOH ~60%
TSC19q34Deletion or mutationLOH ~60%
Mutation ~15%
Table 2. Completed and Ongoing Trials to Evaluate Novel Therapies for mUCC and MIBC. Compiled and adapted from references [19,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129].
Table 2. Completed and Ongoing Trials to Evaluate Novel Therapies for mUCC and MIBC. Compiled and adapted from references [19,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129].
TrialPatient CharacteristicsRegimenPrimary & Secondary
End Points
Common AEsResults
BLC2001
Phase 2 study in mUCC patients
99 patients with FGFR alteration, who have progressed on chemotherapy or immunotherapyErdafitinib 8 mg in either an intermittent or continuous regimenPrimary end point was ORR and secondary end points were PFS, OS and duration of response.Hyperphosphatemia, Stomatitis and diarrhea.ORR was 40%.
KEYNOTE-045
Phase 3 trial in mUCC
542 patients who recurred or progressed after platinum-based chemotherapyPembrolizumab at a dose of 200 mg every 3 weeks or the investigator’s choice of chemotherapy with paclitaxel, docetaxel, or vinflunineCo-primary endpoints were OS and PFS, among all patients and among patients who had PD-L1 CPS of 10% or more.Pruritus, fatigue, and nauseaOS was 8 vs. 5.2 mos. PFS did not demonstrate a significant difference.
JAVELIN Bladder-100—Phase 3 trial in unresectable locally advanced & mUCC700 patients who completed 1st line chemotherapy without progression.Maintenance avelumab 10 mg/kg IV q2 weekly vs. best supportive carePrimary end point was OS and secondary end points included PFS and safety.Fatigue, pruritus and urinary tract infections.OS at 1 year was 71.3 compared to 58.4%. Median PFS was 3.7 vs. 2.0 mos.
CheckMate-274  Phase 3 trial with MIBC.709 patients with MIBC who had undergone radical cystectomy. Neoadjuvant cisplatin-based chemotherapy before trial entry was allowed.Adjuvant Nivolumab 240 mg IV or placebo q2 weeks for up to 1 year vs.
Placebo.
Primary end point was DFS.
Secondary end point was survival free from recurrence outside the urothelial tract.
Pruritus, fatigue, and diarrheaDFS was 20.8 mos with nivolumab and 10.8 mos with placebo. Patients who were free from recurrence outside the urothelial tract at 6 mos was 77 vs. 63%.
EV-301
Phase 3 trial in locally advanced or mUCC
608 patients who had previously received platinum-containing CHT and had disease progression during or after treatment with a PD-1 or PD-L1 inhibitorEnfortumab vedotin 1.25 mg/kg on days 1, 8, 15 of a 28-day cycle or investigator-chosen CHT on day 1 of a 21-day cycle.The primary end point was overall survival.Alopecia, Peripheral sensory neuropathy, Pruritus.Median OS was 12.8 vs. 8.9 mos.
TROPHY-U- Phase II in mUCC113 patients who previously received platinum-containing CHT and had disease progression during or after treatment with a PD-1 or PD-L1 inhibitorSacituzumab govitecan 10 mg/kg on days 1 and 8 of 21-day cyclesobjective response rate (ORR)
secondary end points were PFS, OS, duration of response, and safety
neutropenia (3%) leukopenia (18%), anemiaORR of 27%
DANUBE-
Phase 3 trial
1032 patients that had received Durvalumab (346), Durvalumab+ Tremelimumab(342), or chemotherapy(344) in patients with untreated, unresectable or locally advance mUCCDurvalumab (1500 mg) IV q4 weeks; Durvalumab (1500 mg)+ Tremelimumab (75 mg) IV q4 weeks for up to 4 doses, followed by durvalumab maintenance (1500 mg) q4 weeks; or SOC chemotherapy (gemcitabine + cisplatin/carboplatin) IV for up to 6 cycles.Co-primary endpoints were OS compared b/w durvalumab and CT in pts whose tumor cells and/or tumor-infiltrating immune cells express high levels of PD-L1 (≥25%) and between durvalumab + tremelimumab and CT regardless of PD-L1 expressionIncreased lipase in the Durvalumab Group and neutropenia in the
chemotherapy group.
Did not meet either of the co-primary endpoints.
Checkmate 901608 patients were randomized to either Nivolumab + Ipilumimab with chemotherapy or chemotherapy alone.Nivolumab 360 mg combined with CHT every 3 weeks or CHT alone.Dual endpoints were PFS and OSPruritis, fatigue, diarrhea, pneumonitisMet its dual primary end points
KEYNOTE-361
Phase 3 trial
1010 patients were randomly assigned to receive Pembrolizumab with CHT or Pembrolizumab alone or CHT alonePembrolizumab 200 mg q3 weeks for a maximum of 35 cycles + IV CHT on D 1 and 8 vs. CHT on day 1 of every 3-week cycle for a maximum of 6 cyclesDual primary endpoints of OS and PFS. Secondary endpoints included duration of response, disease control rate, overall response rate and safetyFatigue, musculoskeletal pain, decreased appetite, constipation, rash, and diarrheaDid not meet either of the endpoints.
DISCUS
(ONGOING)
224 eligible and evaluable patients (112 in each arm) to receive 3 vs. 6 cycles of platinum-based CHT + Avelumab in 1st line of mUCCGemcitabine on D1 and D8 with Carboplatin/Cisplatin on D1 and Avelumab every 2weekly.QoL as measured by the change from baseline in EORTC QLQ-C30 questionnaire GHS/QoL scale scores from baseline to the completion of 6 cycles of treatment
Main-CAV Alliance A032001
(ONGOING)
Maintenance Cabozantinib+Avelumab vs. Avelumab in 1st line mUCC with clinical benefit after platinum-based CHTAvelumab 800 mg IV q2 wk or combination of Avelumab and CABO 40 mg orally daily for up to 2 yrsOS
Table 3. Current and Emerging Therapeutic Options for the Treatment of mUCC. Compiled and adapted from references [19,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,130,131].
Table 3. Current and Emerging Therapeutic Options for the Treatment of mUCC. Compiled and adapted from references [19,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,130,131].
Type of
Treatment
ExamplesMechanism of ActionIndicationsAdverse Events
ChemotherapyGemcitabine, Cisplatin, MethotrexateKills rapidly dividing cancer cellsAdvanced/metastatic stages.
Contraindications include ECOG, hearing disorder, heart failure, peripheral neuropathy and Creatinine Cl <60.
Nausea and vomiting.
Loss of appetite, Hair loss, Mouth sores, Diarrhea, Constipation
Immunotherapy
(PD-1/PD-L1 inhibitors)
Pembrolizumab, AtezolizumabBlocks PD-1/PD-L1 interaction, boosting immune responseAdvanced/metastatic stages. Unresponsive to other treatments.
Indicated as frontline for platinum-ineligible
patients.
Fatigue,
Nausea, Loss of appetite, Fever, Urinary tract infections (UTIs)
FGFR3 InhibitorsErdafitinibInhibits FGFR3, a gene mutation common in UCCAdvanced/metastatic with FGFR3 mutation. Progressed on prior treatmentHyperphosphatemia,
Stomatitis and diarrhea.
Antibody-Drug ConjugatesEnfortumab vedotinTargeted delivery of toxic agents to tumor cells that express Nectin-4Advanced/metastatic stages. Progressed on prior cisplatin-based therapy or immunotherapy.Alopecia, Peripheral sensory neuropathy, pruritus.
CAR-T Cell TherapyCurrently not approved for mUCCAutologous patient T-cells engineered to express a chimeric antigen receptor (CAR)
directed against a cancer cell target. Potential targets include EGFR, MUC1, PD-1, HER2 and PSMA.
Under clinical investigationImmune effector cell-associated neurotoxicity syndrome (ICANS), Cytokine Release Syndrome (CRS)
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

Rani, B.; Ignatz-Hoover, J.J.; Rana, P.S.; Driscoll, J.J. Current and Emerging Strategies to Treat Urothelial Carcinoma. Cancers 2023, 15, 4886. https://doi.org/10.3390/cancers15194886

AMA Style

Rani B, Ignatz-Hoover JJ, Rana PS, Driscoll JJ. Current and Emerging Strategies to Treat Urothelial Carcinoma. Cancers. 2023; 15(19):4886. https://doi.org/10.3390/cancers15194886

Chicago/Turabian Style

Rani, Berkha, James J. Ignatz-Hoover, Priyanka S. Rana, and James J. Driscoll. 2023. "Current and Emerging Strategies to Treat Urothelial Carcinoma" Cancers 15, no. 19: 4886. https://doi.org/10.3390/cancers15194886

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