Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Oncology
Join Editorial Board Propose a Special Issue
Print ISSN: 1019-6439 Online ISSN: 1791-2423
Journal Cover
February-2026 Volume 68 Issue 2

Full Size Image

Cover Legend PDF

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
February-2026 Volume 68 Issue 2

Full Size Image

Cover Legend PDF

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Targeted therapies for urothelial carcinoma: From FGFR inhibitors to next‑generation antibody-drug conjugates (Review)

  • Authors:
    • Jinping Du
    • Hao Shen
    • Tongwei Zeng
    • Wei Liu
    • Yongqiang Xie
  • View Affiliations / Copyright

    Affiliations: Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
    Copyright: © Du et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 28
    |
    Published online on: December 22, 2025
       https://doi.org/10.3892/ijo.2025.5841
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Treatment of urothelial carcinoma (UC), particularly in metastatic or cisplatin‑ineligible patients, remains challenging because of limited durable responses to conventional chemotherapy and immune checkpoint inhibitors. Recent advances in targeted therapies, including FGFR inhibitors (for example, erdafitinib) and antibody‑drug conjugates (ADCs) targeting Nectin‑4 (enfortumab vedotin) and HER2 (disitamab vedotin), have reshaped treatment paradigms. FGFR3 alterations, which are present in 20‑40% of patients with advanced UC, predict sensitivity to FGFR tyrosine kinase inhibitors, whereas ADCs demonstrate efficacy across biomarker‑selected and unselected populations. However, clinical implementation is complicated by resistance mechanisms, such as kinase switching, phenotypic plasticity and tumor microenvironment interactions, as well as biomarker heterogeneity. The present review synthesizes current evidence on molecularly guided therapies, contrasts resistance mechanisms between FGFR inhibitors and ADCs, and evaluates strategies to overcome therapeutic limitations. By integrating translational insights and emerging preclinical data, it was aimed to provide a roadmap for optimizing biomarker‑driven approaches, novel combinations and next‑generation agents for the treatment of UC.

Introduction

Urothelial carcinoma (UC) is a neoplastic growth that originates in the urinary tract lining and extends from the renal pelvis to the urethra. It encompasses a range of histological subtypes and anatomical locations, including upper tract UC (UTUC) and primary urethral cancer (1). UTUC accounts for 5-10% of UC cases, whereas primary urethral cancer is even rarer (2). In the United States, it is estimated that there were 87,840 new cases of bladder cancer and 17,840 deaths in 2024, making it the fifth most common cancer in men and the fourteenth in women. The incidence rates of bladder cancer in the US are ~82.3 per 100,000 in men and 21.3 per 100,000 in women (3). In Europe, bladder cancer ranks as the tenth most common cancer, with ~226,000 new cases and 69,000 deaths annually. The incidence rates vary across European countries, with higher rates observed in southern and eastern Europe (3). In Asia, the incidence of UC is lower than that in Western countries, but it still poses a significant health burden, especially in rapidly industrializing regions. For instance, in China, the incidence of bladder cancer is ~13.5 per 100,000, with higher rates in urban areas (3). However, bladder cancer remains a notable health issue in some regions, with incidence rates influenced by factors such as schistosomiasis infection. Globally, the aggressive nature and high recurrence rate of UC make it a challenging disease to manage globally.

Advanced UC, particularly in metastatic or cisplatin-ineligible patients, is associated with poor prognosis (4). The 5-year survival rate for patients with advanced UC is ~10%, highlighting an urgent need for more effective treatment strategies (5). Traditional treatment methods, such as platinum-based chemotherapy, have limitations related to achieving durable responses. For patients with advanced UC being treated with platinum-based chemotherapy, the median OS is typically less than 15 months (6). While the use of immune checkpoint inhibitors (ICIs) has shown promise, their efficacy varies, and numerous patients eventually develop resistance (7). A meta-analysis of 11 trials including 1,630 previously treated patients with UC revealed that the pooled objective response rate (ORR) for single targeted agents was 10.7% (95% CI: 10.7-19.6%), the disease control rate was 33.2% (95% CI: 25-41.4%), and the 1-year OS was 31% (95% CI: 23.6-39.4%) (8). These findings underscore the critical need for novel approaches to improve patient outcomes in this patient population.

Over the past decade, the treatment landscape for UC has evolved significantly with the advent of targeted therapies. Erdafitinib, a fibroblast growth factor receptor (FGFR) inhibitor, and enfortumab vedotin (EV), an antibody-drug conjugates (ADCs) targeting Nectin-4, have demonstrated significant efficacy for the treatment of UC. Erdafitinib achieved a 40% ORR in a phase II trial for patients with FGFR2/3-altered UC (9). EV showed a robust ORR of 43% in a phase I trial, even in patients whose cancer had previously progressed on ICIs (10). These advancements mark a significant shift from traditional chemotherapy and underscore the importance of molecularly guided therapies in improving the prognosis of patients with UC. Ongoing research continues to explore the optimal sequencing of these therapies and the development of next-generation agents to further improve treatment outcomes.

Unlike prior reviews that have separately summarized FGFR inhibitors or ADCs, the present article integrates real-world evidence, biomarker analytics, and resistance biology into a single comparative framework. Recent literature gaps include insufficient head-to-head evaluation of FGFR inhibitors vs. ADCs sequences, limited dissection of convergent resistance pathways (for example, PI3K/AKT re-activation and TGF-β-mediated immune exclusion), and absence of a unified roadmap guiding biomarker-driven combination therapy. By synthesizing 2023-2025 trial data with spatial transcriptomic and urinary circulating-tumor-DNA studies, the present review offers a dynamic, systems-level perspective that links genomic alteration, tumor microenvironment (TME) phenotype, and clinical outcomes; such integration has not been previously accomplished in UC-focused commentaries. Consequently, the manuscript provides clinicians and translational researchers with an evidence-based hierarchy for selecting FGFR or ADCs-based regimens, forecasts emerging resistance mechanisms, and proposes biomarker-adaptive trial designs to accelerate precision medicine implementation.

Molecular biology of UC

In recent years, with advancements in genomics and molecular biology technologies, the molecular landscape of UC has been gradually unveiled. Various molecular alterations and signaling pathways interact synergistically, driving tumor development and progression (11). A deeper understanding of the molecular biology of UC provides a strong foundation for the development of targeted therapies. By identifying key molecular targets and pathways (Fig. 1), researchers can develop more effective therapeutic strategies to improve treatment outcomes and survival rates for patients with UC (12).

Molecular Landscape of UC. Key
molecular targets and pathways discussed in the review are visually
summarized. The molecular landscape of UC with the current targeted
therapies is integrated, highlighting the pathways involved and the
main targets. The image was designed using Figdraw.com. UC, urothelial carcinoma; FGFR,
fibroblast growth factor receptor.

Figure 1

Molecular Landscape of UC. Key molecular targets and pathways discussed in the review are visually summarized. The molecular landscape of UC with the current targeted therapies is integrated, highlighting the pathways involved and the main targets. The image was designed using Figdraw.com. UC, urothelial carcinoma; FGFR, fibroblast growth factor receptor.

Key molecular pathways

UC development and progression involve complex molecular mechanisms. Among them, the Phosphatidylinositol 3-kinase (PI3K)/Protein Kinase B(AKT)/Mammalian Target of Rapamycin (mTOR) pathway plays a significant role (13). This pathway is frequently activated due to mutations in genes such as PIK3CA, PTEN deletion, or AKT1 mutation (14). PI3K catalyzes the production of PIP3, which activates AKT. Activated AKT, in turn, phosphorylates mTOR, promoting cell survival, proliferation and inhibiting apoptosis (14). Studies have shown that PIK3CA mutations are present in ~24-40% of UC cases (15). PTEN deletion occurs in ~17-30% of cases (16). Activation of this pathway contributes to UC progression and chemoresistance. For example, in a particular study, PTEN-deficient mice were more prone to developing UC and displayed more aggressive tumor characteristics, such as increased proliferation rates and higher metastatic potential (17).

The Receptor Tyrosine Kinase (RTK)/Rat Sarcoma (RAS)/V-Raf Murine Sarcoma Viral Oncogene Homolog (RAF)/Mitogen-Activated Protein Kinase Kinase (MEK)/Extracellular Signal-Regulated Kinase (ERK) pathway is another critical signaling pathway (18). Abnormal activation of this pathway can be caused by mutations in FGFR, RAS, or BRAF genes. FGFR gene alterations, including mutations, fusions and amplifications, can lead to ligand-independent receptor activation, subsequently activating downstream signaling pathways (19). RAS mutations are found in ~15-30% of UC cases, and BRAF mutations occur in ~5-10% of cases (20). Activation of this pathway promotes cell proliferation, differentiation and angiogenesis. For example, a study by Zhou et al (21) demonstrated that mutations in this pathway are associated with tumor progression and poor prognosis in patient with UC.

The Tumor Protein 53 (TP53) gene mutation pathway is also crucial in UC. TP53 is a tumor suppressor gene, and its encoded protein plays a pivotal role in cell cycle regulation, DNA repair and apoptosis (22). TP53 mutations are detected in ~40-50% of UC cases. Mutations in this gene lead to loss of its normal function, enabling cells with DNA damage to continue proliferating, thereby promoting tumor development and progression (22). Ecke et al (23) revealed that TP53 mutations are associated with higher tumor grade and stage in UC and are linked to poor patient prognosis.

FGFR alterations

FGFR gene alterations are prevalent in UC. FGFR family includes FGFR1-4. Among them, FGFR3 alterations are the most common in UC, occurring in ~10-30% of cases (24). FGFR3 gene mutations primarily involve point mutations in the tyrosine kinase domain, such as R248C, S249C and Y373C mutations, which lead to receptor constitutive activation (25). FGFR3 gene fusions, such as FGFR3-TACC3 fusions, can also result in abnormal receptor activation. FGFR gene amplifications can increase receptor expression levels, thereby enhancing downstream signaling pathway activation (26). These alterations contribute to tumor cell proliferation and progression. For example, Liu et al (26) showed that FGFR3 mutations are associated with tumor recurrence and progression in non-muscle-invasive bladder cancer. FGFR3 alterations are more common in non-muscle-invasive UC, with a detection rate of ~15-20%, while in muscle-invasive UC, the detection rate is ~5-10%. FGFR2 alterations are relatively rare in UC but are also involved in tumor progression.

Other molecular targets

In addition to the aforementioned molecular pathways and FGFR alterations, other molecular targets also play important roles in UC. TROP2 is a transmembrane glycoprotein that is highly expressed in UC and other solid tumors. Its overexpression is associated with tumor aggressiveness and poor prognosis (27). Sacituzumab govitecan, a TROP2-targeted ADCs, has demonstrated significant efficacy in the treatment of metastatic UC (28). Nectin-4 is another molecule highly expressed in UC. EV, an ADCs targeting Nectin-4, has shown remarkable therapeutic effects in locally advanced or metastatic UC (29). HER-2 is overexpressed in a subset of patients with UC. Trastuzumab deruxtecan, a HER-2-targeted ADCs, has shown promising efficacy in HER-2-overexpressing UC (30). The expression levels of these molecular targets are closely related to tumor biology and patient prognosis.

FGFR pathway-directed therapies: Foundations and clinical implementation

The increasing understanding of FGFR pathway dysregulation in UC has driven the development of targeted therapies. The molecular basis of FGFR abnormalities in UC are addressed in the present section, the clinical data of FGFR inhibitors such as erdafitinib and infigratinib are reviewed, and resistance mechanisms are discussed, aiming to guide optimal use of FGFR-targeted strategies in clinical practice (Fig. 2).

Mechanisms of action and resistance
pathways of FGFR-TKIs and ADCs in urothelial carcinoma. The image
was designed using Figdraw.com. FGFR, fibroblast
growth factor receptor; TKIs, tyrosine kinase inhibitors; ADCs,
antibody-drug conjugates; EMT, epithelial-mesenchymal
transition.

Figure 2

Mechanisms of action and resistance pathways of FGFR-TKIs and ADCs in urothelial carcinoma. The image was designed using Figdraw.com. FGFR, fibroblast growth factor receptor; TKIs, tyrosine kinase inhibitors; ADCs, antibody-drug conjugates; EMT, epithelial-mesenchymal transition.

Molecular basis of FGFR dysregulation in UC

Dysregulation of the FGFR signaling pathway through amplifications, mutations and gene fusions has been implicated in UC (31). FGFR signaling plays crucial roles in tumor cell proliferation, angiogenesis, migration and survival (32) (Table I). FGFR pathway dysregulation is a hallmark of UC, primarily driven by genetic alterations in FGFR3 and, less frequently, FGFR2. These aberrations include activating mutations, gene fusions and amplifications, which constitutively activate downstream signaling cascades such as RAS/MAPK and PI3K/AKT, promoting tumor proliferation and survival (33,34).

Table I

Related research on FGFR dysregulation in urothelial carcinoma.

Table I

Related research on FGFR dysregulation in urothelial carcinoma.

First author/s, yearAberration typesStudy typesMechanism of actionRelationship with prognosis(Refs.)
Ross et al, 2016; Necchi et al, 2019FGFR3 mutations/fusionsGenomic profilingActivating mutations (for example, S249C, R248C) and fusions (for example, *FGFR3-TACC3*) constitutively activate RAS/MAPK and PI3K/AKT pathways.Higher frequency in advanced UC; associated with resistance to platinum chemotherapy (ORR: 28% vs. 45% in wild-type).(33,36)
Kim et al, 2018; Khalid et al, 2020FGFR3 mutationsClinical cohort analysisCo-occurrence with TP53 mutations enhances oncogenicity; promotes ligand-independent kinase activation.Correlates with aggressive MIUC phenotypes; shorter OS in metastatic UC (HR=1.42).(34,40)
Gamallat et al, 2023FGFR3 mutationsSubtype-specific analysisEnriched in large nested variant UC (36%); drives subtype-specific signaling dysregulation.Subtype-specific heterogeneity linked to differential therapeutic responses.(35)
Necchi et al, 2019FGFR3 mRNA overexpressionComparative DNA/RNA analysisDiscordance between DNA mutations and mRNA levels due to alternative splicing or epigenetic regulation.mRNA overexpression correlates with DNA alterations in 65% of cases; prognostic value context-dependent.(36)
Song et al, 2023FGFR2 amplificationsGenomic and microenvironmentalActivates TGF-β-mediated immunosuppression; reduces CD8+ T-cell infiltration.Associated with resistance to ICIs and aggressive phenotypes in advanced UC.(41)
Okato et al, 2024FGFR3-mutant TME Preclinical/translational studyM2 macrophage polarization and immunosuppressive microenvironment; FGFR inhibition enhances anti-PD-1 efficacy.Improved survival in murine models; synergy with immunotherapy in clinical trials.(42)
Mahmoud et al, 2024FGFR3 overexpressionImmunohistochemical analysisProtein overexpression detected in non-metastatic UC; lacks consistent survival correlation.Limited prognostic significance in early-stage UC; potential biomarker for FGFR-targeted therapies.(43)
Shohdy et al, 2019Co-alterations (*CDKN2A/B*)Mechanistic and combination study*CDKN2A/B* deletions enhance dependency on CDK4/6; co-targeting FGFR and CDK4/6 delays resistance.Rationale for combination therapies in FGFR3-altered UC.(39)
Li et al, 2023 FGFR3-mediated resistanceFunctional in vitro studyP4HA2 stabilizes HIF-1α, bypassing FGFR inhibition; metabolic adaptation drives resistance.Identifies novel resistance mechanisms; informs strategies to overcome erdafitinib resistance.(38)

[i] UC, urothelial carcinoma; MIUC, muscle-invasive urothelial carcinoma; OS, overall survival; ORR, objective response rate; TME, tumor microenvironment; ICIs, immune checkpoint inhibitors.

Comprehensive genomic profiling of 295 advanced UC cases revealed FGFR3 alterations in 15% of tumors, predominantly point mutations (for example, S249C, R248C and Y375C) and fusions (for example, FGFR3-TACC3) (33). In muscle-invasive UC, FGFR3 mutations occur in 20% of cases, often co-occurring with TP53 mutations, suggesting a synergistic oncogenic role (34). Notably, large nested variant UC exhibits a higher prevalence of FGFR3 mutations (36%), underscoring subtype-specific molecular heterogeneity (35). At the mRNA level, FGFR3 overexpression correlates with DNA-level mutations in 65% of advanced UC cases, though discordances exist due to alternative splicing or epigenetic regulation (36).

FGFR3 mutations induce ligand-independent dimerization and constitutive kinase activation. Huang et al (37) demonstrated that FGFR3 silencing in UC cell lines suppressed RAS/MAPK pathway activity, impairing invasion and proliferation. However, FGFR-driven tumors often develop resistance via parallel pathways. For instance, P4HA2-mediated HIF-1α stabilization was shown to bypass FGFR inhibition in FGFR3-mutant UC, highlighting metabolic adaptation as a resistance mechanism (38). Additionally, co-alterations in CDKN2A/B (frequently deleted in FGFR3-altered UC) may enhance dependency on CDK4/6, providing a rationale for combination therapies (39).

While FGFR3 mutations were initially associated with non-muscle-invasive UC and favorable prognosis, metastatic FGFR3-altered UC correlates with poorer responses to platinum chemotherapy (ORR: 28 vs. 45% in wild-type) (36). A meta-analysis of 1,574 patients with UC confirmed that FGFR3 mutations independently predict shorter OS in advanced stages (HR=1.42, 95% CI: 1.11-1.82) (40). Conversely, FGFR2 amplifications, observed in 5-8% of UC, are linked to aggressive phenotypes and resistance to ICIs due to TGF-β-mediated immunosuppression (41,42).

Recent studies highlight the immunosuppressive microenvironment in FGFR3-mutant UC, characterized by M2 macrophage infiltration and reduced CD8+ T-cell activity. Preclinical models demonstrated that FGFR inhibition synergizes with anti-PD-1 therapy by reversing immunosuppression, offering a compelling strategy for clinical translation (42). However, the prognostic value of FGFR alterations remains context-dependent; for example, FGFR3 overexpression in non-metastatic UC lacks significant survival correlation (43). Therefore, FGFR dysregulation in UC is mechanistically diverse, influenced by alteration type, coexisting genomic events, and TME factors. Standardized detection methods and functional validation are critical to optimize patient stratification for FGFR-targeted therapies.

Application of FGFR-targeted strategies in the treatment of UC

FGFR-targeted therapies have emerged as a transformative approach in managing advanced UC, particularly for patients with FGFR3 alterations. This section critically evaluates the clinical efficacy, limitations and evolving strategies of FGFR inhibitors, supported by pivotal trials and real-world evidence (Table II).

Table II

Application of FGFR-targeted strategies in the treatment of urothelial carcinoma.

Table II

Application of FGFR-targeted strategies in the treatment of urothelial carcinoma.

First author/s, yearIntervention strategiesTargetsStudy typesSettingSample size (N)Therapeutic effect(Refs.)
Loriot et al, 2019Erdafitinib monotherapyPan-FGFR (FGFR1-4)Phase II BLC2001 trialPost-platinum, FGFR2/3-altered UCN=99ORR: 40%; median PFS: 5.5 months; median OS: 13.8 months in platinum-refractory UC.(44)
Loriot et al, 2023Erdafitinib vs. chemotherapyPan-FGFR (FGFR1-4)Phase III THOR trialFGFR3-altered, post-ICI metastatic UCN=266Improved median OS (12.1 vs. 7.8 months; HR=0.64); ORR: 45.6% vs. 22.4%.(47)
Siefker-Radtke et al, 2024Erdafitinib vs. pembrolizumabPan-FGFR (FGFR1-4)Phase III THOR Cohort 2FGFR-altered, PD-L1-high metastatic UCN=352No OS benefit (10.9 vs. 11.1 months; HR=1.18) in PD-L1-high tumors.(48)
Lyou et al, 2022Infigratinib monotherapyFGFR1-3Phase II trialPost-platinum, FGFR3-altered metastatic UCN=67ORR: 25.4%; median OS: 10.3 months in platinum-refractory UC.(50)
Pal et al, 2022Infigratinib adjuvant therapyFGFR1-3 PROOF-302 trialPhase IIIFGFR3-mutant muscle-invasive UCN=630Preliminary data: 35% reduction in recurrence rates in FGFR3-mutant MIUC.(51)
Sternberg et al, 2023Rogaratinib monotherapyFGFR1-4Phase II/III FORT-1 trialFGFR1/3 mRNA-high metastatic UCN=126ORR: 20.7% vs. 19.3% (chemotherapy); median OS: 8.3 vs. 9.8 months (HR=1.02).(52)
Necchi et al, 2024Pemigatinib monotherapyFGFR1-3Phase II FIGHT-201 trialFGFR-altered, post-platinum metastatic UCN=40ORR: 12%; no survival benefit in UC.(53)
Siefker-Radtke and Loriot, 2022Erdafitinib + pembrolizumabPan-FGFR + PD-1Phase II trialFGFR-altered, ICI-naïve metastatic UCN=58ORR: 47%; median OS: 12.2 months; increased grade ≥3 AEs (e.g., rash, diarrhea).(55)
Necchi et al, 2024Derazantinib + atezolizumabFGFR/CSF1R + PD-L1Phase I/II trialFGFR-altered, post-ICI metastatic UCN=45ORR: 33%; CSF1R inhibition counteracts macrophage-mediated resistance.(56)

[i] ORR, objective response rate; PFS, progression-free survival; OS, overall survival; HR, hazard ratio; MIUC, muscle-invasive urothelial carcinoma; AEs, adverse events.

Erdafitinib, a pan-FGFR inhibitor, is the first FDA-approved FGFR inhibitors for metastatic UC harboring susceptible FGFR2/3 alterations. The phase II BLC2001 trial (N=99) demonstrated an ORR of 40%, median progression-free survival (PFS) of 5.5 months, and median OS of 13.8 months in platinum-refractory patients (44). However, the single-arm design and absence of a control group limit the interpretability of these outcomes, particularly in the context of post-immunotherapy efficacy. Furthermore, the enrichment for FGFR3-altered tumors, without stratification by mutation subtype, may overestimate the true treatment effect, as FGFR3-TACC3 fusions have been associated with more favorable responses than point mutations (45). Hyperphosphatemia, a class-effect toxicity due to FGFR1 inhibition, occurred in 77% of patients but correlated with antitumor efficacy (P=0.02) (46). Long-term follow-up confirmed sustained responses, with a 24-month OS rate of 27% (9). The phase III THOR trial validated erdafitinib's superiority over chemotherapy in FGFR3-altered metastatic UC (Cohort 1: N=266), showing improved median OS (12.1 vs. 7.8 months; HR=0.64) and ORR (45.6% vs. 22.4%) (47). However, in Cohort 2 comparing erdafitinib to pembrolizumab in programmed death-Ligand 1(PD-L1)-high tumors, no OS benefit was observed (10.9 vs. 11.1 months; HR=1.18), underscoring the need for biomarker-driven selection (48). Real-world studies corroborated these findings, with Guercio et al (49) reporting an ORR of 38% and median OS of 11.2 months in 112 patients.

Infigratinib, an FGFR1-3 inhibitor, showed modest activity in a phase II trial (N=67) with an ORR of 25.4% and median OS of 10.3 months in platinum-refractory UC (50). Hyperphosphatemia again emerged as a common adverse event (AE), observed in 73% of patients (46). The phase III PROOF-302 trial is evaluating adjuvant infigratinib in FGFR3-mutant muscle-invasive UC, with preliminary data suggesting a 35% reduction in recurrence rates (51). Besides, Rogaratinib, an FGFR1-4 inhibitor, failed to outperform chemotherapy in the phase II/III FORT-1 trial (N=126) despite selecting patients with high FGFR1/3 mRNA expression. ORR was comparable (20.7 vs. 19.3%), and median OS was shorter (8.3 vs. 9.8 months; HR=1.02), highlighting challenges in biomarker validation (52). Furthermore, Pemigatinib, approved in cholangiocarcinoma, showed limited efficacy in UC (ORR: 12%) in the FIGHT-201 trial (N=40), with no survival benefit (53). Its combination with pembrolizumab was explored in cisplatin-ineligible patients but discontinued due to strategic reasons (54).

Combining FGFR inhibitors with ICIs aims to overcome immunosuppressive microenvironments. Erdafitinib plus pembrolizumab achieved an ORR of 47% and median OS of 12.2 months in a phase II trial (N=58), though grade ≥3 AEs (for example, rash and diarrhea) increased significantly (47). Preclinical models suggest FGFR inhibition reverses M2 macrophage polarization and enhance CD8+ T-cell infiltration, supporting this strategy (55). Derazantinib, a dual FGFR/CSF1R inhibitor, combined with atezolizumab (anti-PD-L1), showed promise in a phase I/II trial (N=45) with an ORR of 33%. CSF1R targeting may counteract tumor-associated macrophage-mediated resistance, offering a novel therapeutic angle (56).

Resistance to FGFR-tyrosine kinase inhibitors (TKIs)

Resistance to FGFR-TKIs remains a critical challenge in UC, driven by diverse molecular mechanisms. Preclinical and clinical studies have identified several pathways contributing to acquired or intrinsic resistance, including compensatory signaling activation, genomic evolution, and TME adaptations. Datta et al (57) demonstrated that Akt activation mediates resistance to BGJ398 (infigratinib) in FGFR3-mutant UC models, highlighting compensatory PI3K/AKT/mTOR pathway activation as a key escape mechanism. This finding aligns with Pettitt et al (58), who observed in vitro resistance to FGFR inhibitors via multiple pathways, including MAPK reactivation and epithelial-mesenchymal transition (EMT), underscoring the heterogeneity of resistance mechanisms. Conversely, Kim et al (59) reported that FGFR1-overexpressing UC cells develop resistance to BGJ398 through sustained MAPK/ERK signaling, which could be reversed by MEK inhibitors, suggesting combinatorial strategies to delay resistance.

Clinical studies further reveal the impact of FGFR alterations on therapeutic outcomes. Rezazadeh Kalebasty et al (60) observed that patients with UC with FGFR mutations exhibited reduced clinical benefit from anti-PD-(L)1 therapies post-FGFR inhibition, suggesting cross-resistance linked to immunosuppressive TME reprogramming. By contrast, Bellmunt et al (61) found that everolimus/pazopanib combination therapy improved outcomes in genomically selected patients with UC, including those with FGFR pathway alterations, though efficacy was limited by mTOR pathway feedback activation. Divergent resistance patterns are also influenced by tumor histology. Brunelli et al (62) identified reduced TROP-2 and NECTIN-4 expression in sarcomatoid UC variants, correlating with poor ADCs responses and potential cross-resistance to FGFR-targeted therapies. Additionally, Audisio et al (63) emphasized the role of clonal evolution under FGFR inhibition, where subpopulations with secondary FGFR2/3 mutations or parallel oncogenic drivers (for example, EGFR) emerge, necessitating longitudinal genomic profiling.

These studies collectively highlight the multifactorial nature of FGFR-TKI resistance. While compensatory signaling (for example, PI3K/AKT and MAPK) and genomic evolution are predominant mechanisms, TME interactions and histological subtypes further modulate therapeutic vulnerability. Future research should prioritize combinatorial approaches (for example, FGFR inhibitors + MEKi/mTORi) and biomarker-driven adaptive therapies to circumvent resistance.

ADCs

ADCs have emerged as a transformative therapeutic class in UC, leveraging tumor-specific antigens to deliver cytotoxic payloads with precision. ADCs have redefined UC treatment, with Nectin-4 and HER2 as validated targets (Fig. 2). Relevant studies have optimized the selection of biomarkers, clarified the pathways of action, and integrated ADCs into multimodal regimens (Table III). Real-world evidence and innovative imaging modalities (for example, Nectin-4 PET) will further personalize ADCs therapy.

Table III

Application of antibody-drug conjugates strategies in the treatment of UC.

Table III

Application of antibody-drug conjugates strategies in the treatment of UC.

First author/s, yearIntervention strategiesTargetsStudy typesSettingSample size (N)Therapeutic effect(Refs.)
Li et al, 2024EV monotherapyNectin-4Phase II trial (China)Post-platinum metastatic UC (Asian cohort)N=60ORR 50%, median PFS 5.8 months in previously treated metastatic UC(66)
Rizzo et al, 2025EV post-ICI failureNectin-4Retrospective study (ARON-2)Post-ICI metastatic UCN=237Median OS 12.1 months vs. chemotherapy 8.3 months (HR=0.62)(67)
Uchimoto et al, 2025BMI impact on EV efficacyNectin-4Cohort study (Japan)Post-ICI metastatic UC (ULTRA-Japan)N=166BMI ≥25: ORR 42.9% vs. 23.1%, OS 14.2 vs. 9.1 months(68)
Mishra et al, 2024Nectin-4 PET-guided dosingNectin-4Translational imaging studyMetastatic UC with heterogeneous Nectin-4 expressionN=12Tracer uptake correlates with EV response (r=0.72, P=0.008)(69)
Jindal et al, 2023TP53/MDM2 biomarker analysisNectin-4Biomarker analysisPost-ICI metastatic UCN=45TP53 wild-type: PFS 6.4 vs. 3.1 months (P=0.03)(70)
Hovelroud et al, 2024EV-associated diabetic ketoacidosisNectin-4Case reportPost-ICI metastatic UCN=1Severe insulin resistance in a nondiabetic patient(71)
Desimpel et al, 2024EV-associated lung toxicityNectin-4Retrospective analysisPost-ICI metastatic UCN=1,892Interstitial lung disease incidence 0.5%(72)
Pipitone et al, 2025EV extravasation managementNectin-4Case report & literature reviewMetastatic UC receiving EVN=1 (case)Protocol for tissue necrosis prevention(73)
Zhu et al, 2025EV + pembrolizumab cost-effectivenessNectin-4Cost-effectiveness modelFirst-line metastatic UCModel-basedIncremental cost-effectiveness ratio>$150,000/QALY; requires price reduction(74)
Sheng et al, 2024Disitamab vedotin monotherapyHER2Phase II trialHER2-positive metastatic UCN=107HER2-positive metastatic UC: ORR 51.2%, median OS 14.2 months(77)
Chen et al, 2023Disitamab vedotin monotherapyHER2Phase II trialHER2-positive locally advanced or metastatic UCN=76HER2-positive metastatic UC: ORR 51.2%, median OS 14.2 months(78)
Yan et al, 2025Disitamab vedotin in HER2-low populationsHER2Phase II trialHER2-negative metastatic UCN=82HER2-negative metastatic UC: ORR 28.6%, median OS 11.8 months(79)
Wang et al, 2025Disitamab vedotin in HER2-null populationsHER2Real-world studyHER2-low/null metastatic UCN=154ORR 18.2%, median PFS 4.1 months(80)
Yao et al, 2025Disitamab vedotin + tislelizumab combinationHER2 + PD-1Phase II trialHER2-expressing, ICI-naïve metastatic UCN=36ORR 58.3%, median PFS 8.5 months(82)
Ge et al, 2025RC48 + PD-1 inhibitorsHER2 + PD-1Real-world studyHER2-expressing metastatic UCN=112ORR 54.5%, median OS 16.1 months(83)
Yang et al, 2025HER2 prognostic value (shorter OS)HER2Retrospective cohortMuscle-invasive UCN=412HER2+ associated with HR=1.58 (P=0.02) in multivariable analysis(84)
Chen et al, 2025HER2 in bladder-preservation therapyHER2Clinical cohortHER2+ muscle-invasive UC treated with bladder-sparing therapyN=127No OS difference in HER2+ MIBC patients(85)
Chou et al, 2022TROP-2 in luminal subtypesTROP-2Molecular subtyping analysisLuminal papillary UC cohortN=120Enriched in luminal papillary UC(86)
Bahlinger et al, 2024FGFR3/Nectin-4 co-alterationsFGFR3Translational studyAdvanced UC with Nectin-4-high expressionN=200FGFR3 mutations in 25% of Nectin-4-high tumors(87)

[i] ORR, objective response rate; PFS, progression-free survival; OS, overall survival; HR, hazard ratio; AEs, adverse events; UC, urothelial carcinoma; MIUC, muscle-invasive urothelial carcinoma; TME, tumor microenvironment; ICIs, immune checkpoint inhibitors; BMI, body mass index; utDNA, urine tumor DNA; PD-L1, programmed death-ligand 1; FGFR, fibroblast growth factor receptor.

Nectin-4-targeted ADCs

Nectin-4, a cell adhesion molecule overexpressed in 60-90% of UCs, has become a pivotal therapeutic target. EV, an ADCs comprising a Nectin-4-directed antibody conjugated to monomethyl auristatin E, received FDA approval based on the EV-301 trial (64). The recent phase III EV-302 trial (NCT04223856) established enfortumab vedotin plus pembrolizumab as a new first-line standard for metastatic UC, demonstrating superior OS and PFS compared with platinum-based chemotherapy (median OS: 31.5 vs. 16.1 months; HR=0.47; median PFS: 12.5 vs. 6.3 months; HR=0.45) (65). Recent studies corroborate its efficacy in diverse clinical settings (66-68). The phase II trial in Chinese patients with previously treated metastatic UC demonstrated an ORR of 50% and median PFS of 5.8 months, validating EV's activity in Asian populations (66). Notably, the aforementioned study lacked central imaging review and biomarker stratification beyond Nectin-4 expression, raising concerns about response assessment heterogeneity. Additionally, the absence of post-progression treatment details introduces potential immortal time bias, particularly in a single arm setting without comparator arm. Real-world data from the ARON-2 retrospective study (n=237) further revealed that EV achieved a median OS of 12.1 months post-ICI failure, outperforming chemotherapy (8.3 months; HR=0.62, P<0.001) (67). Notably, efficacy of EV was influenced by body mass index (BMI): patients with BMI ≥25 had superior tumor response rates (ORR 42.9% vs. 23.1%, P=0.04) and longer OS (14.2 vs. 9.1 months, P=0.02), suggesting metabolic factors may modulate ADCs activity (68).

Biomarker-driven strategies are under exploration. Mishra et al (69) developed a Nectin-4 PET tracer to non-invasively quantify target expression, revealing heterogeneous intratumoral distribution. Higher tracer uptake correlated with EV response (r=0.72, P=0.008), supporting personalized dosing. Additionally, TP53/MDM2 alterations were associated with reduced EV benefit in a biomarker analysis (n=45): TP53 wild-type tumors had longer PFS (6.4 vs. 3.1 months, P=0.03) (70).

EV's toxicity profile remains consistent across studies, with peripheral neuropathy (40-50%), rash (30%) and hyperglycemia (5-10%) as key AEs (66,67). A pharmacovigilance study (n=1,892) identified rare but severe AEs, including diabetic ketoacidosis (0.3%) and interstitial lung disease (0.5%) (71,72). Extravasation management protocols have been proposed following case reports of tissue necrosis (73). In addition, cost-effectiveness analyses weigh EV's benefits against its economic burden. Zhu et al (74) modeled EV + pembrolizumab as first-line therapy for metastatic UC, showing incremental cost-effectiveness ratios exceeding $150,000/QALY, necessitating price reductions for broader adoption.

HER2-directed ADCs

HER2 expression in UC is heterogeneous, with 5-15% classified as HER2-positive (IHC 3+ or 2+/FISH+) and 30-40% as HER2-low (IHC 1+ or 2+ with negative in situ hybridization), consistent with the ASCO-CAP guidelines (75), which are commonly extrapolated to UC in the absence of disease-specific criteria. Disitamab vedotin (DV; RC48), an anti-HER2 ADCs, has shown promise across HER2 expression levels (76). In the combined analysis of the phase II trials (n=107), DV monotherapy achieved an ORR of 51.2% and median OS of 14.2 months in HER2-positive metastatic UC, with grade ≥3 AEs (for example, neutropenia: 20%) deemed manageable (77). Real-world studies reinforce these findings: Chen et al (78) reported an ORR of 48.6% and median PFS of 6.9 months in 76 HER2-positive patients treated with DV ± ICIs.

Intriguingly, DV exhibits activity even in HER2-low/null cohorts. Yan et al (79) conducted a phase II trial (n=82) in HER2-negative metastatic UC, demonstrating an ORR of 28.6% and median OS of 11.8 months, suggesting off-target effects or HER2 detection limitations. Similarly, Wang et al (80) observed ORRs of 24.1% (HER2-low) and 18.2% (HER2-null) in a real-world study (n=154), though responses were less durable (median PFS: 4.1 vs. 5.3 months). These findings challenge traditional HER2 thresholds and advocate for refined scoring systems.

The combination of DV with ICIs has demonstrated synergistic efficacy, representing a significant advancement. In the phase Ib/II RC48-C014 trial (NCT04264936), DV plus the PD-1 inhibitor toripalimab achieved a confirmed ORR of 71.8% and a median PFS of 9.2 months in patients with HER2-expressing (IHC 1+/2+/3+) locally advanced or metastatic UC (81). Notably, efficacy was consistent across HER2-low (IHC 1+ or 2+/FISH-) and HER2-positive (IHC 3+ or 2+/FISH+) subgroups, challenging traditional HER2 positivity thresholds and suggesting broader applicability.

DV-ICI combinations synergize efficacy. Yao et al (82) reported an ORR of 58.3% and median PFS of 8.5 months in 36 ICI-naïve patients with metastatic UC receiving DV + tislelizumab, with immune-related AEs in 22%. Ge et al (83) corroborated these results in a larger cohort (n=112; ORR: 54.5%, median OS: 16.1 months), highlighting enhanced antitumor immunity. While HER2 overexpression correlates with aggressive features (for example, higher grade, nodal metastases), its prognostic value remains contested. Yang et al (84) analyzed 412 UC specimens, finding HER2 positivity (12.6%) associated with shorter OS (HR=1.58, P=0.02) in multivariable analysis. Conversely, Chen et al (85) reported no OS difference in HER2-positive muscle-invasive UC treated with bladder-preservation therapy, underscoring context-dependent roles.

Emerging targets

TROP-2, expressed in 70% of UCs, is under investigation with sacituzumab govitecan. Preclinical data reveal TROP-2 overexpression in sarcomatoid UC (80%) vs. conventional UC (40%), suggesting histology-specific targeting (62). Chou et al (86) identified TROP-2 enrichment in luminal papillary subtypes, potentially guiding patient selection. FGFR3-directed ADCs (for example, erdafitinib combinations) and tissue factor-targeting agents (for example, tisotumab vedotin) are in early trials. Bahlinger et al (87) observed FGFR3 mutations in 25% of Nectin-4-high tumors, advocating dual-target approaches.

Translational insights: Biomarker-driven patient selection

The advent of targeted therapies in UC has underscored the critical role of biomarker-driven patient selection to maximize therapeutic efficacy and minimize toxicity (88). This section evaluates the translational insights from key clinical trials, focusing on FGFR inhibitors and ADCs, and discusses the challenges and opportunities in biomarker validation, heterogeneity, and clinical implementation.

FGFR alterations: From discovery to clinical validation

FGFR alterations, particularly FGFR3 mutations and fusions, have emerged as pivotal oncogenic drivers in UC. These genetic aberrations, identified in 20-40% of metastatic UC and 35-40% of high-risk non-muscle-invasive bladder cancer (NMIBC) tumors, lead to constitutive activation of downstream signaling pathways such as RAS-MAPK and PI3K-AKT, promoting tumor proliferation, survival and angiogenesis (41). Preclinical studies highlighted FGFR3's role in bladder carcinogenesis, spurring the development of selective FGFR inhibitors. Erdafitinib, a first-in-class pan-FGFR-TKI, demonstrated early promise in the phase 2 BLC2001 trial, achieving an ORR of 40% in FGFR-altered metastatic UC, which laid the groundwork for subsequent phase 3 validation (47). The discovery of FGFR3's oncogenic role and its high prevalence in UC established a strong rationale for biomarker-driven therapeutic strategies, positioning FGFR status as a critical predictive biomarker for patient stratification.

The THOR trial (NCT03390504) marked a milestone in the clinical validation of FGFR-targeted therapy. In this phase 3 study, erdafitinib significantly outperformed chemotherapy in patients with FGFR3-altered metastatic UC, demonstrating a median OS of 12.1 months vs. 7.8 months (HR=0.64) and a doubling of PFS (5.6 vs. 2.7 months) (47). Nevertheless, the trial's open-label design may introduce assessment bias, particularly in subjective endpoints such as PFS. Moreover, the exclusion of patients with prior immunotherapy limits the generalizability of these findings to contemporary real-world cohorts, where ICI exposure is now standard of care. These transformative outcomes, coupled with durable responses and manageable toxicity, led to erdafitinib's regulatory approvals in China, USA and other regions, cementing FGFR3 alteration as a validated biomarker for patient selection. However, challenges persist, including tumor heterogeneity, the emergence of resistance mutations (for example, FGFR3 gatekeeper mutations), and the need for standardized biomarker testing protocols (89). Ongoing research focuses on optimizing FGFR inhibitor sequencing, exploring combination therapies with ICIs, and validating liquid biopsy-based FGFR detection to address spatial and temporal heterogeneity in advanced UC (42). These efforts aim to refine precision medicine approaches and extend the benefits of FGFR-targeted therapy to broader patient subsets.

HER2 as an emerging target: Expanding the ADCs landscape

Human epidermal growth factor receptor 2 (HER2) overexpression or amplification is observed in 10-20% of UC and has emerged as a promising target for ADCs (90). Disitamab vedotin (DV), an HER2-targeted ADCs, combined with the PD-1 inhibitor toripalimab, demonstrated unprecedented efficacy in the RC48-C016 trial (NCT04879329), achieving significant OS (11.5 vs. 9.5 months) and PFS (4.2 vs. 2.9 months) benefits over chemotherapy in HER2-expressing metastatic UC, including cisplatin-ineligible patients (81). Notably, efficacy was consistent across HER2-low and HER2-high subgroups, challenging the traditional HER2 positivity thresholds and suggesting broader applicability.

By contrast, EV, a NECTIN-4-directed ADCs, has shown remarkable activity in unselected UC populations (ORR: 67.7%; CR: 29% in EV-302) (91). While EV does not require biomarker preselection, retrospective analyses suggest that NECTIN-4 expression levels correlate with response durability, raising questions about the need for quantitative biomarker thresholds (92). This contrasts with HER2-targeted ADCs, where even low expression may suffice for clinical benefit, as observed in DV trials. Such differences underscore the need for target-specific biomarker frameworks.

Navigating tumor heterogeneity and resistance in biomarker-driven therapies

UC exhibits significant intratumoral heterogeneity, with FGFR and HER2 status varying between primary and metastatic lesions (93). For example, FGFR3 mutations are more common in primary NMIBC, while metastatic sites often acquire additional genomic alterations (for example, TP53 and RB1) (94). Longitudinal studies reveal that FGFR alterations may be lost after BCG therapy or chemotherapy, necessitating repeat biopsies for dynamic biomarker assessment (95). Liquid biopsy approaches [for example, circulating tumor DNA (ctDNA)] are under investigation but require validation for real-time monitoring (96).

Co-mutations (for example, TP53 with FGFR3) may modulate response to targeted therapies. In the THOR trial, patients with FGFR3-TACC3 fusions had superior outcomes compared with those with FGFR3 mutations, suggesting fusion-specific sensitivity (45). Similarly, HER2 amplification often coexists with PI3K/AKT pathway activation, which may confer resistance to DV unless combined with PI3K inhibitors (97). These findings highlight the importance of comprehensive genomic profiling to identify co-targetable pathways.

Biomarker-driven selection has revolutionized UC treatment, yet challenges in standardization, heterogeneity, and resistance persist. FGFR and HER2 inhibitors exemplify the success of precision medicine, while ADCs such as EV demonstrate the potential of target-agnostic approaches. Future research must prioritize biomarker validation, combinatorial strategies and real-world evidence to bridge the gap between trial populations and clinical practice. Recent evidence indicates that urine tumor DNA (utDNA) assays achieve 91.4% sensitivity and 95.1% specificity for detecting UC-associated FGFR3 or TERT mutations, outperforming cytology and enabling longitudinal genotyping without repeated cystoscopy (98). This high diagnostic accuracy underscores the utility of utDNA in capturing tumor-derived genetic material shed into the urine, thereby providing a more comprehensive representation of intratumoral and inter-lesional heterogeneity compared with single-site tissue biopsies. In the prospective TAR-210 trial, real-time utDNA screening for FGFR3 alterations increased trial-enrollment efficiency by 36% and permitted early identification of emergent FGFR3 gate-keeper mutations (99). By circumventing the spatial limitations of tissue biopsies, utDNA offers a non-invasive means to dynamically monitor clonal evolution and adapt therapeutic strategies in response to molecular changes. Analogously, plasma ctDNA panels that cover FGFR2/3, PIK3CA, TP53 and ERBB2 can be performed every 4-6 weeks; rising variant-allele frequencies precede radiological progression by a median of 4.2 months in patients receiving erdafitinib or enfortumab vedotin, providing a lead-time window for therapy adaptation (100). Thus, liquid biopsy platforms already allow dynamic patient selection and early resistance surveillance and are being integrated into adaptive trial designs such as the ULTRA-switch study (100).

Resistance mechanisms and overcoming therapeutic limitations

Resistance to FGFR inhibitors and ADCs in UC is multifactorial, driven by convergent pathways such as kinase switching, persistent downstream signaling and TME interactions (101,102) (Fig. 3). However, emerging strategies, including biomarker-guided combinations, next-generation ADCs and epigenetic modulation, hold promise for overcoming these barriers. Future studies must prioritize longitudinal biomarker validation and innovative trial designs to translate preclinical insights into clinical success.

Schematic of convergent resistance
pathways and novel overcoming strategies for FGFR inhibitors and
ADCs in UC. This figure illustrates the shared and target-specific
resistance mechanisms to FGFR-TKIs and ADCs in UC, alongside
emerging therapeutic strategies to circumvent resistance. The image
was designed using Figdraw.com. FGFR, fibroblast
growth factor receptor; ADCs, antibody-drug conjugates; UC,
urothelial carcinoma.

Figure 3

Schematic of convergent resistance pathways and novel overcoming strategies for FGFR inhibitors and ADCs in UC. This figure illustrates the shared and target-specific resistance mechanisms to FGFR-TKIs and ADCs in UC, alongside emerging therapeutic strategies to circumvent resistance. The image was designed using Figdraw.com. FGFR, fibroblast growth factor receptor; ADCs, antibody-drug conjugates; UC, urothelial carcinoma.

Convergent resistance pathways between FGFR-TKIs and ADCs

Resistance to FGFR inhibitors (for example, erdafitinib) and ADCs (for example, EV) often involves compensatory activation of parallel signaling pathways (103,104). For instance, FGFR inhibition in metastatic UC induces upregulation of platelet-derived growth factor receptor (PDGFR), enabling tumor cells to bypass FGFR dependency via PDGF ligand stimulation (105). Preclinical studies in breast cancer models demonstrated that FGFR inhibitor pemigatinib triggers PDGFRα/β overexpression, reactivating MAPK/ERK signaling and promoting minimal residual disease (MRD) survival (106). Similarly, resistance to HER2-targeted ADCs (for example, disitamab vedotin) may involve MET or HER2 amplification, as observed in non-small cell lung cancer (NSCLC) EGFR-TKI resistance models (107). These findings highlight a shared mechanism of 'kinase switching' across targeted therapies.

Both FGFR inhibitors and ADCs face resistance due to sustained activation of downstream effectors. In FGFR3-altered UC, MEK/ERK and PI3K/AKT pathways remain active despite FGFR inhibition, driven by co-mutations (for example, TP53 and RB1) or epigenetic adaptations (108,109). For ADCs, resistance may arise from defective payload release or upregulation of anti-apoptotic proteins (for example, BCL-2) (110). For example, EV-resistant UC cell lines exhibit increased expression of multidrug resistance transporters, reducing monomethyl auristatin E cytotoxicity (104).

Tumor cells evade targeted therapies through EMT or lineage plasticity (111). FGFR inhibition in UC promotes EMT via STAT3 activation, enhancing metastatic potential (103). ADCs targeting NECTIN-4 or HER2 may similarly encounter resistance due to loss of target expression during phenotypic shifts (112,113). Additionally, stromal interactions in the TME play a pivotal role. Lung fibroblasts secrete PDGF-AA to support MRD survival in FGFR inhibitor-treated models, while cancer-associated fibroblasts shield tumor cells from ADCs penetration by secreting extracellular matrix components (114).

Novel strategies to circumvent resistance

Dual inhibition of FGFR and compensatory pathways (for example, PDGFR or PI3K) has shown promise (115,116). In preclinical UC models, combining erdafitinib with the DNA methyltransferase 1 inhibitor GSK3484862 delayed relapse by suppressing PDGFR upregulation and epigenetic plasticity (90). For ADCs, co-targeting HER2 and MET (for example, trastuzumab + savolitinib) is under investigation in NSCLC, with potential applicability to UC (90).

Bispecific ADCs (for example, targeting HER2 and TROP2) or payload modifications (for example, TOP1 inhibitors) may overcome resistance by broadening target engagement or enhancing cytotoxicity (117). Erdafitinib intravesical delivery systems (TAR-210) improve local efficacy while reducing systemic toxicity, achieving 82% recurrence-free survival in FGFR3-altered NMIBC (99). Similarly, FGFR3-specific degraders (for example, PROTACs) are emerging to address kinase domain mutations (118).

FGFR inhibitors may synergize with ICIs by modulating the TME (119). Erdafitinib increases T-cell infiltration and reduces myeloid-derived suppressor cells in UC models, suggesting enhanced immunogenicity (119). Clinical trials evaluating erdafitinib + pembrolizumab (NCT05316155) are underway, with preliminary data showing durable responses in PD-L1-low populations. Furthermore, histone deacetylase (HDAC) inhibitors reverse resistance-associated epigenetic silencing (120). For example, low-dose decitabine restored FGFR3 expression in erdafitinib-resistant UC cells, re-sensitizing them to therapy. Metabolic reprogramming (for example, valine restriction) combined with HDAC6 inhibitors has shown efficacy in enhancing DNA damage in preclinical models, offering a novel combinatorial approach (120).

In addition, longitudinal assessment of resistance mutations via utDNA enables real-time adaptation of therapy (100). The UI Seek assay, which detects FGFR3/TERT mutations and methylation markers in urine, demonstrated 91.37% sensitivity and 95.09% specificity for UC diagnosis, outperforming traditional cytology (98). In the TAR-210 trial, utDNA-based FGFR3 screening improved patient enrollment by 36%, highlighting its utility in guiding adaptive therapies (121). Longitudinal ctDNA monitoring is being integrated into adaptive trial designs. The ULTRA-ctDNA sub-study (NCT05538680) will trigger crossover to alternative targeted agents or combination regimens as soon as emergent FGFR3 secondary mutations or PI3K/AKT pathway alterations are detected, aiming to prevent clinical relapse rather than merely documenting it post hoc.

Clinical translation of resistance data is now beginning to shape sequencing algorithms. In the multicenter real-world APOLLO study (n=184), patients who progressed on erdafitinib were systematically re-biopsied; 62% of cases acquired PIK3CA or PTEN loss-of-function alterations that were absent at baseline. Subsequent treatment with everolimus plus paclitaxel in these molecularly selected patients yielded a 34% ORR and 8.1-month median PFS, whereas non-selected historical controls achieved only 12% and 4.0 months, respectively (49). Similarly, longitudinal ctDNA surveillance during EV therapy revealed that emergent NECTIN-4 loss or TUBB3 mutations predicted resistance within 4-6 weeks; early addition of taxane-based chemotherapy at the time of molecular progression doubled median time-to-next-treatment compared with waiting for radiological progression (7.3 vs. 3.5 months, HR 0.48, P=0.02) (67).

Consequently, an adaptive 'biomarker-triggered' sequence is being evaluated prospectively in the ULTRA-switch trial (NCT05538680): Upon detection of FGFR3 gatekeeper or PI3K/AKT pathway mutations, patients crossover from erdafitinib to a PI3Kβ inhibitor plus paclitaxel, while NECTIN-4 loss triggers EV-to-taxane switch. Early safety run-in data (n=42) show a 71% clinical benefit rate without additive toxicity, supporting the feasibility of real-time molecular triage.

Challenges and future directions

Resistance to targeted therapies poses a significant challenge in the treatment of UC. With respect to FGFR inhibitors, the mechanisms of resistance include secondary gene mutations and compensatory signaling pathway activation (122). For example, studies have found that FGFR gene mutations can lead to reduced inhibitor binding affinity of the inhibitors to the target, thereby causing resistance. Additionally, the activation of other signaling pathways, such as the EGFR pathway, can compensate for the inhibition of the FGFR pathway, promoting tumor cell survival and proliferation (122,123). Similar resistance mechanisms have been observed in the use of ADCs. Secondary gene mutations can alter the structure of the target antigen, reducing the binding ability of the ADCs (124). Moreover, the upregulation of drug efflux pump expression can decrease the intracellular concentration of the cytotoxic drugs, leading to resistance (124).

To overcome these resistance mechanisms, several strategies are being explored. Combination therapies represent a promising approach. For instance, combining FGFR inhibitors with immunotherapy or chemotherapy may enhance therapeutic efficacy (125). Preclinical studies have shown that the combination of FGFR inhibitors and anti-PD-1 antibodies can synergistically inhibit tumor growth by alleviating immunosuppression in the TME (125,126). The development of next-generation drugs is also crucial. Researchers are working on designing FGFR inhibitors with higher selectivity and affinity to overcome resistance caused by secondary mutations. Furthermore, the use of ctDNA analysis may help in identifying resistance mechanisms and guiding treatment adjustments in a timely manner (127).

Managing the AEs associated with targeted therapies is essential for improving patient quality of life and treatment adherence. FGFR inhibitors are commonly associated with ocular toxicity and hyperphosphatemia. Ocular toxicity can manifest as macular edema, serous retinopathy and blurred vision. Regular ophthalmologic examinations are recommended for early detection and management of these side effects early (128). Hyperphosphatemia can usually be managed through dietary adjustments and the use of phosphate binders. ADCs, on the other hand, can cause peripheral neuropathy and myelosuppression. Peripheral neuropathy may require dose adjustments or the use of neuroprotective agents. Myelosuppression necessitates regular monitoring of blood cell counts and appropriate supportive care. It is crucial to establish standardized management protocols for these AEs to ensure the safe and effective use of targeted therapies in clinical practice (129).

Determining the optimal sequence of targeted therapies, immunotherapy and chemotherapy in UC treatment strategies remains an area of active research. Factors influencing treatment sequence decisions include tumor molecular tumor characteristics, patient performance status and prior treatment history (130). For example, patients with FGFR alterations may benefit from the use of FGFR inhibitors as first-line therapy. However, the optimal timing for introducing immunotherapy or chemotherapy in combination with targeted therapies remains unclear. Further studies are needed to investigate the interactions between different treatment modalities and their impact on patient outcomes. Clinical trials evaluating various treatment sequences are ongoing, and their results will provide valuable insights into the best approaches for optimizing treatment strategies for UC.

Combining targeted therapies with immunotherapy or other treatments offers exciting prospects for UC therapy. Preclinical and early clinical studies have demonstrated that such combinations can increase antitumor activity. For instance, combining FGFR inhibitors with ICIs may improve immune cell infiltration and function in the TME (130). However, combination therapies also present challenges, such as increased toxicity and trial design complexity. Careful consideration of the potential side effects and the development of effective toxicity management strategies are necessary to ensure the safety of combination therapies. Additionally, the design of clinical trials must account for the interactions between different drugs and their pharmacokinetic and pharmacodynamic characteristics. Despite these challenges, the potential benefits of combination therapies make them a promising direction for future research in UC treatment.

As our understanding of the molecular biology of UC continues to deepen, new therapeutic targets and drugs are emerging. In addition to the currently established targets such as FGFR and TROP2, other molecules such as MET, AXL and Wnt/β-catenin are gaining attention. MET amplification and mutations have been identified in a subset of patients with UC and are associated with poor prognosis. Inhibitors targeting MET are being developed and have shown initial promise in preclinical studies. AXL overexpression is linked to tumor progression and resistance to therapy. Drugs targeting AXL may provide new treatment options for patients with UC. The Wnt/β-catenin pathway plays a role in tumor cell proliferation and survival. Modulating this pathway may offer another avenue for therapeutic intervention. These novel targets and drugs, along with advancements in genomics and molecular biology technologies, are expected to further expand the therapeutic landscape for UC and to improve treatment outcomes for patients.

Conclusion

Significant advancements in biomarker-driven approaches and targeted therapies for UC have been made, offering new treatment options and hope for patients. However, challenges such as biomarker heterogeneity, resistance mechanisms, and the need for optimized treatment strategies remain. Future research should focus on improving biomarker validation, developing novel combination therapies, and designing innovative clinical trials to enhance the efficacy and precision of targeted treatments in UC.

Availability of data and materials

Not applicable.

Authors' contributions

JD and YX made significant contributions to the conception of the manuscript, wrote the first version of the manuscript and prepared figures. TZ, HS and WL reviewed the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

The present study was supported by Baiyin City Science and Technology Plan Project (Preclinical study of c-MET targeted therapy in bladder cancer; grant no. 2023-2-14Y).

References

1 

Biasatti A, Bignante G, Ditonno F, Veccia A, Bertolo R, Antonelli A, Lee R, Eun DD, Margulis V, Abdollah F, et al: New insights into upper tract urothelial carcinoma: Lessons learned from the ROBUUST collaborative study. Cancers (Basel). 17:16682025. View Article : Google Scholar : PubMed/NCBI

2 

Nally E, Young M, Chauhan V, Wells C, Szabados B, Powles T and Jackson-Spence F: Upper tract urothelial carcinoma (UTUC): Prevalence, impact and management challenge. Cancer Manag Res. 16:467–475. 2024. View Article : Google Scholar : PubMed/NCBI

3 

Jubber I, Ong S, Bukavina L, Black PC, Compérat E, Kamat AM, Kiemeney L, Lawrentschuk N, Lerner SP, Meeks JJ, et al: Epidemiology of Bladder Cancer in 2023: A Systematic Review of Risk Factors. Eur Urol. 84:176–190. 2023. View Article : Google Scholar : PubMed/NCBI

4 

Moussa MJ, Campbell MT and Alhalabi O: Revisiting treatment of metastatic urothelial cancer: Where do cisplatin and platinum ineligibility criteria stand? Biomedicines. 12:5192024. View Article : Google Scholar : PubMed/NCBI

5 

Rivera C, Jabbal IS, Yaghi M, Landau KS, Muruve N, Saravia D, George TL, Nahleh ZA and Arteta-Bulos R: Five-year survival comparison of different treatment modalities for muscle invasive urothelial carcinoma, squamous cell carcinoma and adenocarcinoma of the bladder: An analysis of the National cancer database. J Clin Oncol. 40(6_suppl): S5752022. View Article : Google Scholar

6 

Gupta S, Andreev-Drakhlin A, Fajardo O, Fassò M, Garcia JA, Wee C and Schröder C: Platinum ineligibility and survival outcomes in patients with advanced urothelial carcinoma receiving first-line treatment. J Natl Cancer Inst. 116:547–554. 2024. View Article : Google Scholar

7 

Mao L, Yang M, Fan X, Li W, Huang X, He W, Lin T and Huang J: PD-1/L1 inhibitors can improve but not replace chemotherapy for advanced urothelial carcinoma: A systematic review and network meta-analysis. Cancer Innov. 2:191–202. 2023. View Article : Google Scholar : PubMed/NCBI

8 

Li XK and Wang WL: The role novel targeted agents in the treatment of previously treated patients with advanced urothelial carcinoma (UC): A meta-analysis. Eur Rev Med Pharmacol Sci. 22:5165–5171. 2018.PubMed/NCBI

9 

Siefker-Radtke AO, Necchi A, Park SH, García-Donas J, Huddart RA, Burgess EF, Fleming MT, Rezazadeh Kalebasty A, Mellado B, Varlamov S, et al: Efficacy and safety of erdafitinib in patients with locally advanced or metastatic urothelial carcinoma: Long-term follow-up of a phase 2 study. Lancet Oncol. 23:248–258. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Yu EY, Petrylak DP, O'Donnell PH, Lee JL, van der Heijden MS, Loriot Y, Stein MN, Necchi A, Kojima T, Harrison MR, et al: Enfortumab vedotin after PD-1 or PD-L1 inhibitors in cisplatin-ineligible patients with advanced urothelial carcinoma (EV-201): A multicentre, single-arm, phase 2 trial. Lancet Oncol. 22:872–882. 2021. View Article : Google Scholar : PubMed/NCBI

11 

Seront E and Machiels JP: Molecular biology and targeted therapies for urothelial carcinoma. Cancer Treat Rev. 41:341–53. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Glaser AP, Fantini D, Shilatifard A, Schaeffer EM and Meeks JJ: The evolving genomic landscape of urothelial carcinoma. Nat Rev Urol. 14:215–229. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Ching CB and Hansel DE: Expanding therapeutic targets in bladder cancer: The PI3K/Akt/mTOR pathway. Lab Invest. 90:1406–1414. 2010. View Article : Google Scholar : PubMed/NCBI

14 

He F, Zhang F, Liao Y, Tang MS and Wu XR: Structural or functional defects of PTEN in urothelial cells lacking P53 drive basal/squamous-subtype muscle-invasive bladder cancer. Cancer Lett. 550:2159242022. View Article : Google Scholar : PubMed/NCBI

15 

Patel VG, McBride RB, Lorduy AC, Castillo-Martin M, Cha EK, Berger MF, Wang L, Oh WK, Zhu J, Cordon-Cardo C, et al: Prognostic significance of PIK3CA mutation in patients with muscle-invasive urothelial carcinoma (UC). J Clin Oncol. 34(15_suppl): e160022016. View Article : Google Scholar

16 

Wang DS, Rieger-Christ K, Latini JM, Moinzadeh A, Stoffel J, Pezza JA, Saini K, Libertino JA and Summerhayes IC: Molecular analysis of PTEN and MXI1 in primary bladder carcinoma. Int J Cancer. 88:620–625. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Tsuruta H, Kishimoto H, Sasaki T, Horie Y, Natsui M, Shibata Y, Hamada K, Yajima N, Kawahara K, Sasaki M, et al: Hyperplasia and carcinomas in Pten-deficient mice and reduced PTEN protein in human bladder cancer patients. Cancer Res. 66:8389–8396. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Imperial R, Toor OM, Hussain A, Subramanian J and Masood A: Comprehensive pancancer genomic analysis reveals (RTK)-RAS-RAF-MEK as a key dysregulated pathway in cancer: Its clinical implications. Semin Cancer Biol. 54:14–28. 2019. View Article : Google Scholar

19 

Degirmenci U, Wang M and Hu J: Targeting aberrant RAS/RAF/MEK/ERK signaling for cancer therapy. Cells. 9:1982020. View Article : Google Scholar : PubMed/NCBI

20 

Kim YS, Lee SC, Hwang IG, Lee SJ and Park SH: Relationship between RAS, BRAF, PIK3CA and location of primary tumor in urothelial carcinoma. Cancer Res. 79(13_Suppl): S12622019. View Article : Google Scholar

21 

Zhou H, Huang HY, Shapiro E, Lepor H, Huang WC, Mohammadi M, Mohr I, Tang MS, Huang C and Wu XR: Urothelial tumor initiation requires deregulation of multiple signaling pathways: implications in target-based therapies. Carcinogenesis. 33:770–780. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Wu G, Wang F, Li K, Li S, Zhao C, Fan C and Wang J: Significance of TP53 mutation in bladder cancer disease progression and drug selection. PeerJ. 7:e82612019. View Article : Google Scholar : PubMed/NCBI

23 

Ecke TH, Sachs MD, Lenk SV, Loening SA and Schlechte HH: TP53 gene mutations as an independent marker for urinary bladder cancer progression. Int J Mol Med. 21:655–661. 2008.PubMed/NCBI

24 

Khalid S, Basulaiman BM, Emack J, Booth CM, Hernandez-Barajas D, Duran I, Smoragiewicz M, Amir E and Vera-Badillo F: FGFR3 mutation as a prognostic indicator in patients with urothelial carcinoma: A systematic review and meta-analysis. J Clin Oncol. 37(7_suppl): S4112019. View Article : Google Scholar

25 

Che X, Liu H, Qin J and Cao S: Landscape of FGFR2/3 alterations in genitourinary cancer. J Clin Oncol. 41(16_suppl): e150472023. View Article : Google Scholar

26 

Liu X, Zhang W, Geng D, He J, Zhao Y and Yu L: Clinical significance of fibroblast growth factor receptor-3 mutations in bladder cancer: a systematic review and meta-analysis. Genet Mol Res. 13:1109–1120. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Zeng P, Chen MB, Zhou LN, Tang M, Liu CY and Lu PH: Impact of TROP2 expression on prognosis in solid tumors: A Systematic Review and Meta-analysis. Sci Rep. 6:336582016. View Article : Google Scholar : PubMed/NCBI

28 

Tagawa ST, Balar AV, Petrylak DP, Kalebasty AR, Loriot Y, Fléchon A, Jain RK, 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. 39:2474–2485. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Rosenberg JE, Powles T, Sonpavde GP, Loriot Y, Duran I, Lee JL, Matsubara N, Vulsteke C, Castellano D, Mamtani R, et al: EV-301 long-term outcomes: 24-month findings from the phase III trial of enfortumab vedotin versus chemotherapy in patients with previously treated advanced urothelial carcinoma. Ann Oncol. 34:1047–1054. 2023. View Article : Google Scholar : PubMed/NCBI

30 

Tagawa ST, Balar AV, Petrylak DP, Rezazadeh A, Loriot Y, Flechon A, Jain RK, Agarwal N, Bupathi M, Barthelemy P, et al: Updated outcomes in TROPHY-U-01 cohort 1, a phase 2 study of sacituzumab govitecan (SG) in patients (pts) with metastatic urothelial cancer (mUC) that progressed after platinum (PT)-based chemotherapy and a checkpoint inhibitor (CPI). J Clin Oncol. 41(6_suppl): S5262023. View Article : Google Scholar

31 

Hall TG, Yu Y, Eathiraj S, Wang Y, Savage RE, Lapierre JM, Schwartz B and Abbadessa G: Preclinical activity of ARQ 087, a novel inhibitor targeting FGFR dysregulation. PLoS One. 11:e01625942016. View Article : Google Scholar : PubMed/NCBI

32 

Dienstmann R, Rodon J, Prat A, Perez-Garcia J, Adamo B, Felip E, Cortes J, Iafrate AJ, Nuciforo P and Tabernero J: Genomic aberrations in the FGFR pathway: Opportunities for targeted therapies in solid tumors. Ann Oncol. 25:552–563. 2014. View Article : Google Scholar

33 

Ross JS, Wang K, Khaira D, Ali SM, Fisher HA, Mian B, Nazeer T, Elvin JA, Palma N, Yelensky R, et al: Comprehensive genomic profiling of 295 cases of clinically advanced urothelial carcinoma of the urinary bladder reveals a high frequency of clinically relevant genomic alterations. Cancer. 122:702–711. 2016. View Article : Google Scholar

34 

Kim YS, Kim K, Kwon GY, Lee SJ and Park SH: Fibroblast growth factor receptor 3 (FGFR3) aberrations in muscle-invasive urothelial carcinoma. BMC Urol. 18:682018. View Article : Google Scholar : PubMed/NCBI

35 

Gamallat Y, Afsharpad M, El Hallani S, Maher CA, Alimohamed N, Hyndman E and Bismar TA: Large, nested variant of urothelial carcinoma is enriched with activating mutations in fibroblast growth factor receptor-3 among other targetable mutations. Cancers (Basel). 15:31672023. View Article : Google Scholar : PubMed/NCBI

36 

Necchi A, Lo Vullo S, Raggi D, Gloghini A, Giannatempo P, Colecchia M and Mariani L: Prognostic effect of FGFR mutations or gene fusions in patients with metastatic urothelial carcinoma receiving first-line platinum-based chemotherapy: Results from a large, single-institution cohort. Eur Urol Focus. 5:853–856. 2019. View Article : Google Scholar

37 

Huang GK, Huang CC, Kang CH, Cheng YT, Tsai PC, Kao YH and Chung YH: Genetic Interference of FGFR3 impedes invasion of upper tract urothelial carcinoma cells by alleviating RAS/MAPK signal activity. Int J Mol Sci. 24:17762023. View Article : Google Scholar : PubMed/NCBI

38 

Li X, Li Y, Liu B, Chen L, Lyu F, Zhang P, He Q, Cheng L, Liu C, Song Y and Xing Y: P4HA2-mediated HIF-1α stabilization promotes erdafitinib-resistance in FGFR3-alteration bladder cancer. FASEB J. 37:e228402023. View Article : Google Scholar

39 

Shohdy KS, Vlachostergios PJ, Abdel-Malek RR and Faltas BM: Rationale for co-targeting CDK4/6 and FGFR pathways in urothelial carcinoma. Expert Opin Ther Targets. 23:83–86. 2019. View Article : Google Scholar

40 

Khalid S, Basulaiman BM, Emack J, Booth CM, Duran I, Robinson AG, Berman D, Smoragiewicz M, Amir E and Vera-Badillo FE: Fibroblast growth factor receptor 3 mutation as a prognostic indicator in patients with urothelial carcinoma: A systematic review and meta-analysis. Eur Urol Open Sci. 21:61–68. 2020. View Article : Google Scholar

41 

Song Y, Peng Y, Qin C, Wang Y, Yang W, Du Y and Xu T: Fibroblast growth factor receptor 3 mutation attenuates response to immune checkpoint blockade in metastatic urothelial carcinoma by driving immunosuppressive microenvironment. J Immunother Cancer. 11:e0066432023. View Article : Google Scholar : PubMed/NCBI

42 

Okato A, Utsumi T, Ranieri M, Zheng X, Zhou M, Pereira LD, Chen T, Kita Y, Wu D, Hyun H, et al: FGFR inhibition augments anti-PD-1 efficacy in murine FGFR3-mutant bladder cancer by abrogating immunosuppression. J Clin Invest. 134:e1692412024. View Article : Google Scholar : PubMed/NCBI

43 

Mahmoud SF, Holah NS, Alhanafy AM and Serag El-Edien MM: Do fibroblast growth factor receptor (FGFR) 2 and 3 proteins play a role in prognosis of invasive urothelial bladder carcinoma? Iran J Pathol. 19:81–88. 2024. View Article : Google Scholar : PubMed/NCBI

44 

Loriot Y, Necchi A, Park SH, 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. 381:338–348. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Parker Kerrigan BC, Ledbetter D, Kronowitz M, Phillips L, Gumin J, Hossain A, Yang J, Mendt M, Singh S, Cogdell D, et al: RNAi technology targeting the FGFR3-TACC3 fusion break-point: An opportunity for precision medicine. Neurooncol Adv. 2:vdaa1322020.

46 

Lyou Y, Grivas P, Rosenberg JE, Hoffman-Censits J, Quinn DI, P Petrylak D, Galsky M, Vaishampayan U, De Giorgi U, Gupta S, et al: Hyperphosphatemia secondary to the selective fibroblast growth factor receptor 1-3 inhibitor infigratinib (BGJ398) is associated with antitumor efficacy in fibroblast growth factor receptor 3-altered advanced/metastatic urothelial carcinoma. Eur Urol. 78:916–924. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Loriot Y, Matsubara N, Park SH, Huddart RA, Burgess EF, Houede N, Banek S, Guadalupi V, Ku JH, Valderrama BP, et al: Erdafitinib or chemotherapy in advanced or metastatic urothelial carcinoma. N Engl J Med. 389:1961–1971. 2023. View Article : Google Scholar : PubMed/NCBI

48 

Siefker-Radtke AO, Matsubara N, Park SH, Huddart RA, Burgess EF, Özgüroğlu M, Valderrama BP, Laguerre B, Basso U, Triantos S, et al: Erdafitinib versus pembrolizumab in pretreated patients with advanced or metastatic urothelial cancer with select FGFR alterations: cohort 2 of the randomized phase III THOR trial. Ann Oncol. 35:107–117. 2024. View Article : Google Scholar

49 

Guercio BJ, Sarfaty M, Teo MY, Ratna N, Duzgol C, Funt SA, Lee CH, Aggen DH, Regazzi AM, Chen Z, et al: Clinical and genomic landscape of FGFR3-altered urothelial carcinoma and treatment outcomes with erdafitinib: A real-world experience. Clin Cancer Res. 29:4586–4595. 2023. View Article : Google Scholar : PubMed/NCBI

50 

Lyou Y, Rosenberg JE, Hoffman-Censits J, Quinn DI, Petrylak D, Galsky M, Vaishampayan U, De Giorgi U, Gupta S, Burris H, et al: Infigratinib in early-line and salvage therapy for FGFR3-Altered metastatic urothelial carcinoma. Clin Genitourin Cancer. 20:35–42. 2022. View Article : Google Scholar :

51 

Pal SK, Somford DM, Grivas P, Sridhar SS, Gupta S, Bellmunt J, Sonpavde G, Fleming MT, Lerner SP, Loriot Y, et al: Targeting FGFR3 alterations with adjuvant infigratinib in invasive urothelial carcinoma: The phase III PROOF 302 trial. Future Oncol. 18:2599–2614. 2022. View Article : Google Scholar : PubMed/NCBI

52 

Sternberg CN, Petrylak DP, Bellmunt J, Nishiyama H, Necchi A, Gurney H, Lee JL, van der Heijden MS, Rosenbaum E, Penel N, et al: FORT-1: Phase II/III study of rogaratinib versus chemotherapy in patients with locally advanced or metastatic urothelial carcinoma selected based on FGFR1/3 mRNA expression. J Clin Oncol. 41:629–639. 2023. View Article : Google Scholar :

53 

Necchi A, Pouessel D, Leibowitz R, Gupta S, Fléchon A, García-Donas J, Bilen MA, Debruyne PR, Milowsky MI, Friedlander T, et al: Pemigatinib for metastatic or surgically unresectable urothelial carcinoma with FGF/FGFR genomic alterations: Final results from FIGHT-201. Ann Oncol. 35:200–210. 2024. View Article : Google Scholar

54 

Gong J, Mita AC, Wei Z, Cheng HH, Mitchell EP, Wright JJ, Ivy SP, Wang V, Gray RC, McShane LM, et al: Phase II study of erdafitinib in patients with tumors with FGFR amplifications: results from the NCI-MATCH ECOG-ACRIN trial (EAY131) subprotocol K1. JCO Precis Oncol. 8:e23004062024. View Article : Google Scholar : PubMed/NCBI

55 

Siefker-Radtke AO and Loriot Y: Erdafitinib for locally advanced or metastatic urothelial carcinoma. Am J Health Syst Pharm. 79:824–825. 2022. View Article : Google Scholar : PubMed/NCBI

56 

Necchi A, Ramlau R, Falcón González A, Chaudhry A, Todenhöfer T, Tahbaz R, Fontana E, Giannatempo P, Deville JL, Pouessel D, et al: Derazantinib alone and with atezolizumab in metastatic urothelial carcinoma with activating FGFR aberrations. JNCI Cancer Spectr. 8:pkae0302024. View Article : Google Scholar : PubMed/NCBI

57 

Datta J, Damodaran S, Parks H, Ocrainiciuc C, Miya J, Yu L, Gardner EP, Samorodnitsky E, Wing MR, Bhatt D, et al: Akt activation mediates acquired resistance to fibroblast growth factor receptor inhibitor BGJ398. Mol Cancer Ther. 16:614–624. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Pettitt GA, Hurst CD, Khan Z, McPherson HR, Dunning MC, Alder O, Platt FM, Black EV, Burns JE and Knowles MA: Development of resistance to FGFR inhibition in urothelial carcinoma via multiple pathways in vitro. J Pathol. 259:220–232. 2023. View Article : Google Scholar :

59 

Kim SH, Ryu H, Ock CY, Suh KJ, Lee JY, Kim JW, Lee JO, Kim JW, Kim YJ, Lee KW, et al: BGJ398, A Pan-FGFR inhibitor, overcomes paclitaxel resistance in urothelial carcinoma with FGFR1 overexpression. Int J Mol Sci. 19:31642018. View Article : Google Scholar : PubMed/NCBI

60 

Rezazadeh Kalebasty A, Benjamin DJ, Loriot Y, Papantoniou D, Siefker-Radtke AO, Necchi A, Naini V, Carcione JC, Santiago-Walker A, Triantos S and Burgess EF: Outcomes of patients with advanced urothelial carcinoma after anti-programmed Death-(ligand) 1 therapy by fibroblast growth factor receptor gene alteration status: An observational study. Eur Urol Open Sci. 47:48–57. 2022. View Article : Google Scholar

61 

Bellmunt J, Lalani AA, Jacobus S, Wankowicz SA, Polacek L, Takeda DY, Harshman LC, Wagle N, Moreno I, Lundgren K, et al: Everolimus and pazopanib (E/P) benefit genomically selected patients with metastatic urothelial carcinoma. Br J Cancer. 119:707–712. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Brunelli M, Gobbo S, Malpeli G, Sirgiovanni G, Caserta C, Munari E, Francesconi S, Caliò A, Martignoni G, Cimadamore A, et al: TROP-2, NECTIN-4 and predictive biomarkers in sarcomatoid and rhabdoid bladder urothelial carcinoma. Pathologica. 116:55–61. 2024. View Article : Google Scholar : PubMed/NCBI

63 

Audisio M, Buttigliero C, Turco F, Delcuratolo MD, Pisano C, Parlagreco E, Di Stefano RF, Di Prima L, Crespi V, Farinea G, et al: Metastatic urothelial carcinoma: Have we take the road to the personalized medicine? Cells. 11:16142022. View Article : Google Scholar : PubMed/NCBI

64 

Heath EI and Rosenberg JE: The biology and rationale of targeting nectin-4 in urothelial carcinoma. Nat Rev Urol. 18:93–103. 2021. View Article : Google Scholar

65 

Gupta S, Loriot Y, Van der Heijden MS, Bedke J, Valderrama BP, Kikuchi E, Fléchon A, Petrylak D, De Santis M, Galsky MD, et al: Enfortumab vedotin plus pembrolizumab versus chemotherapy in patients with previously untreated locally advanced or metastatic urothelial cancer (EV-302): Patient-reported outcomes from an open-label, randomised, controlled, phase 3 study. Lancet Oncol. 26:795–805. 2025. View Article : Google Scholar : PubMed/NCBI

66 

Li S, Shi Y, Dong H, Guo H, Xie Y, Sun Z, Zhang X, Kim E, Zhang J, Li Y, et al: Phase 2 trial of enfortumab vedotin in patients with previously treated locally advanced or metastatic urothelial carcinoma in China. Cancer Med. 13:e703682025. View Article : Google Scholar

67 

Rizzo M, Morelli F, Ürün Y, Buti S, Park SH, Bourlon MT, Grande E, Massari F, Landmesser J, Poprach A, et al: Real-life impact of enfortumab vedotin or chemotherapy in the sequential treatment of advanced urothelial carcinoma: The ARON-2 retrospective experience. Cancer Med. 14:e704792025. View Article : Google Scholar : PubMed/NCBI

68 

Uchimoto T, Iwatsuki K, Komura K, Fukuokaya W, Adachi T, Hirasawa Y, Hashimoto T, Yoshizawa A, Saruta M, Hashimoto M, et al: Association of body mass index and tumor response in metastatic urothelial carcinoma treated with enfortumab vedotin: data from the ULTRA-Japan consortium. Int J Clin Oncol. 30:761–769. 2025. View Article : Google Scholar : PubMed/NCBI

69 

Mishra A, Sharma AK, Gupta K, Banka DR, Johnson BA, Hoffman-Censits J, Huang P, McConkey DJ and Nimmagadda S: Nectin-4 Pet for optimizing enfortumab vedotin dose-response in urothelial carcinoma. bioRxiv [Preprint]. 2024.12.25.630315. 2024.

70 

Jindal T, Zhu X, Bose R, Kumar V, Maldonado E, Deshmukh P, Shipp C, Feng S, Johnson MS, Angelidakis A, et al: Somatic alterations of TP53 and MDM2 associated with response to enfortumab vedotin in patients with advanced urothelial cancer. Front Oncol. 13:11610892023. View Article : Google Scholar : PubMed/NCBI

71 

Hovelroud R, Goh Xiu Ming S, McLeod DSA, Donovan PJ, Ng G and Mungomery M: A case of enfortumab vedotin-associated diabetic ketoacidosis with severe insulin resistance in a nondiabetic woman. JCEM Case Rep. 2:luae2122024. View Article : Google Scholar : PubMed/NCBI

72 

Desimpel G, Zammit F, Lejeune S, Grisay G and Seront E: Lung toxicity occurring during enfortumab vedotin treatment: From a priming case report to a retrospective analysis. Pharmaceuticals (Basel). 17:15472024. View Article : Google Scholar : PubMed/NCBI

73 

Pipitone S, Vitale MG, Baldessari C, Sabbatini R, Dominici M, Porretta Serapiglia C, Ricchi L and Rivasi M: Extravasation of enfortumab vedotin: A case report and literature review on antibody-drug conjugates. Eur J Hosp Pharmejhpharm. 2024-0043232025.Epub ahead of print. View Article : Google Scholar

74 

Zhu Y, Liu K, Zhu H, Li S and Yuan D: Enfortumab vedotin plus pembrolizumab for previously untreated locally advanced or metastatic urothelial carcinoma: A cost-effectiveness analysis. Ther Adv Med Oncol. 17:175883592412955442025. View Article : Google Scholar : PubMed/NCBI

75 

Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, Bilous M, Ellis IO, Fitzgibbons P, Hanna W, et al: Human epidermal growth factor receptor 2 testing in breast cancer: American society of clinical oncology/college of American pathologists clinical practice guideline focused update. Arch Pathol Lab Med. 142:1364–1382. 2018. View Article : Google Scholar : PubMed/NCBI

76 

Sheng X, Yan X, Wang L, Shi Y, Yao X, Luo H, Shi B, Liu J, He Z, Yu G, et al: Open-label, multicenter, phase II Study of RC48-ADC, a HER2-targeting antibody-drug conjugate, in patients with locally advanced or metastatic urothelial carcinoma. Clin Cancer Res. 27:43–51. 2021. View Article : Google Scholar

77 

Sheng X, Wang L, He Z, Shi Y, Luo H, Han W, Yao X, Shi B, Liu J, Hu C, et al: Efficacy and safety of disitamab vedotin in patients with human epidermal growth factor receptor 2-positive locally advanced or metastatic urothelial carcinoma: A combined analysis of two phase II clinical trials. J Clin Oncol. 42:1391–1402. 2024. View Article : Google Scholar

78 

Chen M, Yao K, Cao M, Liu H, Xue C, Qin T, Meng L, Zheng Z, Qin Z, Zhou F, et al: HER2-targeting antibody-drug conjugate RC48 alone or in combination with immunotherapy for locally advanced or metastatic urothelial carcinoma: A multicenter, real-world study. Cancer Immunol Immunother. 72:2309–2318. 2023. View Article : Google Scholar : PubMed/NCBI

79 

Yan X, Li J, Xu H, Liu Y, Zhou L, Li S, Wu X, Tang B, Chi Z, Cui C, et al: Efficacy and safety of DV in HER2-negative and HER2-low locally advanced or metastatic urothelial carcinoma: Results of a phase 2 study. Med. 6:1006372025. View Article : Google Scholar : PubMed/NCBI

80 

Wang D, Cao M, Zhang Y, Bi L, Chen M, Ni M, Zheng Q, Yao K, Liu Z, Yang X, et al: RC48-ADC monotherapy or in combination with immunotherapy for locally advanced or metastatic urothelial carcinoma with HER2 low and null expression: A multicenter, real-world, retrospective study. BMC Cancer. 25:8122025. View Article : Google Scholar : PubMed/NCBI

81 

Zhou L, Yang KW, Zhang S, Yan XQ, Li SM, Xu HY, Li J, Liu YQ, Tang BX, Chi ZH, et al: Disitamab vedotin plus toripalimab in patients with locally advanced or metastatic urothelial carcinoma (RC48-C014): A phase Ib/II dose-escalation and dose-expansion study. Ann Oncol. 36:331–339. 2025. View Article : Google Scholar

82 

Yao JW, Zhong JL, Zhou Q and Guo J: Efficacy and safety of disitamab vedotin in combination with immune checkpoint inhibitors in patients with locally advanced or metastatic urothelial carcinoma. World J Urol. 43:1542025. View Article : Google Scholar : PubMed/NCBI

83 

Ge H, Liu C, Shen C, Hu D, Zhao X and Wang Y, Ge H, Qin R, Ma X and Wang Y: The effectiveness and safety of RC48 alone or in combination with PD-1 inhibitors for locally advanced or metastatic urothelial carcinoma: A multicenter, real-world study. J Transl Med. 23:2432025. View Article : Google Scholar : PubMed/NCBI

84 

Yang M, Yao Y, Wang K, Qi L, Yang B, Khudadad M, Guo Y, Wang Y, Liu Y, Li L, et al: Clinicopathological characteristics and prognostic significance of HER2 status evaluation in patients with urothelial carcinoma: A retrospective single-center experience in China. Virchows Arch. Feb 26–2025.Epub ahead of print. View Article : Google Scholar

85 

Chen Y, Luo F, Zhang T and Li J: Impact of HER2 expression on the prognosis of muscle-invasive bladder cancer patients treated with bladder-preservation comprehensive therapy. Biol Proced Online. 27:22025. View Article : Google Scholar : PubMed/NCBI

86 

Chou J, Trepka K, Sjöström M, Egusa EA, Chu CE, Zhu J, Chan E, Gibb EA, Badura ML, Contreras-Sanz A, et al: TROP2 expression across molecular subtypes of urothelial carcinoma and enfortumab vedotin-resistant cells. Eur Urol Oncol. 5:714–718. 2022. View Article : Google Scholar : PubMed/NCBI

87 

Bahlinger V, Branz A, Strissel PL, Strick R, Lange F, Geppert CI, Klümper N, Hölzel M, Wach S, Taubert H, et al: Associations of TACSTD2/TROP2 and NECTIN-4/NECTIN-4 with molecular subtypes, PD-L1 expression, and FGFR3 mutational status in two advanced urothelial bladder cancer cohorts. Histopathology. 84:863–876. 2024. View Article : Google Scholar : PubMed/NCBI

88 

Katims AB, Reisz PA, Nogueira L, Truong H, Lenis AT, Pietzak EJ, Kim K and Coleman JA: Targeted therapies in advanced and metastatic urothelial carcinoma. Cancers (Basel). 14:54312022. View Article : Google Scholar : PubMed/NCBI

89 

Shvartsbart A, Roach JJ, Witten MR, Koblish H, Harris JJ, Covington M, Hess R, Lin L, Frascella M, Truong L, et al: Discovery of potent and selective inhibitors of wild-type and gatekeeper mutant fibroblast growth factor receptor (FGFR) 2/3. J Med Chem. 65:15433–15442. 2022. View Article : Google Scholar : PubMed/NCBI

90 

Shih CH, Lin YH, Luo HL and Sung WW: Antibody-drug conjugates targeting HER2 for the treatment of urothelial carcinoma: Potential therapies for HER2-positive urothelial carcinoma. Front Pharmacol. 15:13262962024. View Article : Google Scholar : PubMed/NCBI

91 

Khosravanian MJ, Mirzaei Y, Mer AH, Keyhani-Khankahdani M, Abdinia FS, Misamogooe F, Amirkhani Z, Bagheri N, Meyfour A, Jahandideh S, et al: Nectin-4-directed antibody-drug conjugates (ADCs): Spotlight on preclinical and clinical evidence. Life Sci. 352:1229102024. View Article : Google Scholar : PubMed/NCBI

92 

Chu CE, Sjöström M, Egusa EA, Gibb EA, Badura ML, Zhu J, Koshkin VS, Stohr BA, Meng MV, Pruthi RS, et al: Heterogeneity in NECTIN4 expression across molecular subtypes of urothelial cancer mediates sensitivity to enfortumab vedotin. Clin Cancer Res. 27:5123–5130. 2021. View Article : Google Scholar : PubMed/NCBI

93 

Guancial EA, Werner L, Bellmunt J, Bamias A, Choueiri TK, Ross R, Schutz FA, Park RS, O'Brien RJ, Hirsch MS, et al: FGFR3 expression in primary and metastatic urothelial carcinoma of the bladder. Cancer Med. 3:835–844. 2014. View Article : Google Scholar : PubMed/NCBI

94 

Ross JS, Gay LM, Ferry EK, Jacob J, Shapiro O, Hoffman-Censits JH, Millis SZ, Elvin JA, Chung J, Vergilio JA, et al: FGFR3 Driven Metastatic Urothelial Carcinoma of the Urinary Bladder (mUCB): A Comprehensive Genomic Profiling Study. J Clin Oncol. 36(15_suppl): 4531. 2018. View Article : Google Scholar

95 

Redig AJ, Costa DB, Taibi M, Boucher D, Johnson BE, Jänne PA and Jackman DM: Prospective study of repeated biopsy feasibility and acquired resistance at disease progression in patients with advanced EGFR mutant lung cancer treated with erlotinib in a phase 2 trial. JAMA Oncol. 2:1240–1242. 2016. View Article : Google Scholar : PubMed/NCBI

96 

Kang M and Ku JH: Emerging role of liquid biopsy of cell-free tumor DNA for bladder cancer surveillance. Transl Androl Urol. 6:590–592. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Bambury RM and Rosenberg JE: Advanced urothelial carcinoma: Overcoming treatment resistance through novel treatment approaches. Front Pharmacol. 4:32013. View Article : Google Scholar : PubMed/NCBI

98 

Wang P, Shi Y, Zhang J, Shou J, Zhang M, Zou D, Liang Y, Li J, Tan Y, Zhang M, et al: UCseek: Ultrasensitive early detection and recurrence monitoring of urothelial carcinoma by shallow-depth genome-wide bisulfite sequencing of urinary sediment DNA. EBioMedicine. 89:1044372023. View Article : Google Scholar : PubMed/NCBI

99 

Pichler R, van Creij NCH, Subiela JD, Cimadamore A, Caño-Velasco J, Tully KH, Mori K, Contieri R, Afferi L, Mari A, et al: Biological and therapeutic implications of FGFR alterations in urothelial cancer: A systematic review from non-muscle-invasive to metastatic disease. Actas Urol Esp (Engl Ed). 49:5017192025.In English, Spanish. View Article : Google Scholar : PubMed/NCBI

100 

Zhang R, Zang J, Jin D, Xie F, Shahatiaili A, Wu G, Zhang L, Wang L, Zhang Y, Zhao Z, et al: Urinary tumor DNA MRD analysis to identify responders to neoadjuvant immunotherapy in muscle-invasive bladder cancer. Clin Cancer Res. 29:4040–4046. 2023. View Article : Google Scholar : PubMed/NCBI

101 

Benjamin DJ and Mita AC: FGFR-altered urothelial carcinoma: Resistance mechanisms and therapeutic strategies. Target Oncol. 20:1–11. 2025. View Article : Google Scholar

102 

Yajima S and Masuda H: Immune checkpoint inhibitors and antibody-drug conjugates in urothelial carcinoma: Current landscape and future directions. Cancers (Basel). 17:15942025. View Article : Google Scholar : PubMed/NCBI

103 

Katoh M, Loriot Y, Brandi G, Tavolari S, Wainberg ZA and Katoh M: FGFR-targeted therapeutics: Clinical activity, mechanisms of resistance and new directions. Nat Rev Clin Oncol. 21:312–329. 2024. View Article : Google Scholar : PubMed/NCBI

104 

Khoury R, Saleh K, Khalife N, Saleh M, Chahine C, Ibrahim R and Lecesne A: Mechanisms of resistance to antibody-drug conjugates. Int J Mol Sci. 24:96742023. View Article : Google Scholar : PubMed/NCBI

105 

Szklener K, Chmiel P, Michalski A and Mańdziuk S: New directions and challenges in targeted therapies of advanced bladder cancer: The role of FGFR inhibitors. Cancers (Basel). 14:14162022. View Article : Google Scholar : PubMed/NCBI

106 

Khajah MA, Mathew PM and Luqmani YA: Inhibitors of PI3K/ERK1/2/p38 MAPK Show preferential activity against endocrine-resistant breast cancer cells. Oncol Res. 25:1283–1295. 2017. View Article : Google Scholar : PubMed/NCBI

107 

Feldt SL and Bestvina CM: The role of MET in resistance to EGFR inhibition in NSCLC: A review of mechanisms and treatment implications. Cancers (Basel). 15:29982023. View Article : Google Scholar : PubMed/NCBI

108 

Manzano RG, Catalan-Latorre A and Brugarolas A: RB1 and TP53 co-mutations correlate strongly with genomic biomarkers of response to immunity checkpoint inhibitors in urothelial bladder cancer. BMC Cancer. 21:4322021. View Article : Google Scholar : PubMed/NCBI

109 

Ungaro A, Tucci M, Audisio A, Di Prima L, Pisano C, Turco F, Delcuratolo MD, Di Maio M, Scagliotti GV and Buttigliero C: Antibody-Drug conjugates in urothelial carcinoma: A new therapeutic opportunity moves from bench to bedside. Cells. 11:8032022. View Article : Google Scholar : PubMed/NCBI

110 

Wise AR, Maloney S, Hering A, Zabala S, Richmond GE, VanKlompenberg MK, Nair MT and Prosperi JR: Bcl-2 Up-Regulation Mediates Taxane Resistance Downstream of APC Loss. Int J Mol Sci. 25:67452024. View Article : Google Scholar : PubMed/NCBI

111 

Das C, Adhikari S, Bhattacharya A, Chakraborty S, Mondal P, Yadav SS, Adhikary S, Hunt CR, Yadav KK, Pandita S, et al: Epigenetic-metabolic interplay in the DNA damage response and therapeutic resistance of breast cancer. Cancer Res. 83:657–666. 2023. View Article : Google Scholar : PubMed/NCBI

112 

Klümper N, Ralser DJ, Ellinger J, Roghmann F, Albrecht J, Below E, Alajati A, Sikic D, Breyer J, Bolenz C, et al: Membranous NECTIN-4 expression frequently decreases during metastatic spread of urothelial carcinoma and is associated with enfortumab vedotin resistance. Clin Cancer Res. 29:1496–1505. 2023. View Article : Google Scholar :

113 

Qu M, Zhou L, Yan X, Li S, Wu X, Xu H, Li J, Guo J, Zhang X, Li H and Sheng X: Advances in HER2-Targeted treatment for advanced/metastatic urothelial carcinoma. Bladder (San Franc). 10:e212000122023.PubMed/NCBI

114 

Watts TL, Cui R, Szaniszlo P, Resto VA, Powell DW and Pinchuk IV: PDGF-AA mediates mesenchymal stromal cell chemotaxis to the head and neck squamous cell carcinoma tumor microenvironment. J Transl Med. 14:3372016. View Article : Google Scholar : PubMed/NCBI

115 

Chen CH, Changou CA, Hsieh TH, Lee YC, Chu CY, Hsu KC, Wang HC, Lin YC, Lo YN, Liu YR, et al: Dual inhibition of PIK3C3 and FGFR as a new therapeutic approach to treat bladder cancer. Clin Cancer Res. 24:1176–1189. 2018. View Article : Google Scholar

116 

Hosaka K, Yang Y, Seki T, Du Q, Jing X, He X, Wu J, Zhang Y, Morikawa H, Nakamura M, et al: Therapeutic paradigm of dual targeting VEGF and PDGF for effectively treating FGF-2 off-target tumors. Nat Commun. 11:37042020. View Article : Google Scholar : PubMed/NCBI

117 

Schlam I, Moges R, Morganti S, Tolaney SM and Tarantino P: Next-generation antibody-drug conjugates for breast cancer: Moving beyond HER2 and TROP2. Crit Rev Oncol Hematol,. 190:1040902023. View Article : Google Scholar : PubMed/NCBI

118 

Shibata N, Cho N, Koyama H and Naito M: Development of a degrader against oncogenic fusion protein FGFR3-TACC3. Bioorg Med Chem Lett. 60:1285842022. View Article : Google Scholar : PubMed/NCBI

119 

Kato Y, Tabata K, Kimura T, Yachie-Kinoshita A, Ozawa Y, Yamada K, Ito J, Tachino S, Hori Y, Matsuki M, et al: Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS One. 14:e02125132019. View Article : Google Scholar : PubMed/NCBI

120 

Pinkerneil M, Hoffmann MJ, Schulz WA and Niegisch G: HDACs and HDAC inhibitors in urothelial carcinoma-perspectives for an antineoplastic treatment. Curr Med Chem. 24:4151–4165. 2017. View Article : Google Scholar

121 

Jaromin M, Konecki T and Kutwin P: Revolutionizing treatment: Breakthrough approaches for BCG-unresponsive non-muscle-invasive bladder cancer. Cancers (Basel). 16:13662024. View Article : Google Scholar : PubMed/NCBI

122 

Yue S, Li Y, Chen X, Wang J, Li M, Chen Y and Wu D: FGFR-TKI resistance in cancer: Current status and perspectives. J Hematol Oncol. 14:232021. View Article : Google Scholar : PubMed/NCBI

123 

Facchinetti F, Hollebecque A, Braye F, Vasseur D, Pradat Y, Bahleda R, Pobel C, Bigot L, Déas O, Florez Arango JD, et al: Resistance to selective FGFR inhibitors in FGFR-Driven urothelial cancer. Cancer Discov. 13:1998–2011. 2023. View Article : Google Scholar : PubMed/NCBI

124 

Ruder S, Martinez J, Palmer J, Arham AB and Tagawa ST: Antibody-drug conjugates in urothelial carcinoma: Current status and future. Curr Opin Urol. 35:292–300. 2025. View Article : Google Scholar : PubMed/NCBI

125 

Ruan R, Li L, Li X, Huang C, Zhang Z, Zhong H, Zeng S, Shi Q, Xia Y, Zeng Q, et al: Unleashing the potential of combining FGFR inhibitor and immune checkpoint blockade for FGF/FGFR signaling in tumor microenvironment. Mol Cancer. 22:602023. View Article : Google Scholar : PubMed/NCBI

126 

Li R, Linscott J, Catto JWF, Daneshmand S, Faltas BM, Kamat AM, Meeks JJ, Necchi A, Pradere B, Ross JS, et al: FGFR inhibition in urothelial carcinoma. Eur Urol. 87:110–122. 2025. View Article : Google Scholar

127 

Jogo T, Nakamura Y, Shitara K, Bando H, Yasui H, Esaki T, Terazawa T, Satoh T, Shinozaki E, Nishina T, et al: Circulating tumor DNA analysis detects FGFR2 Amplification and concurrent genomic alterations associated with FGFR inhibitor efficacy in advanced gastric cancer. Clin Cancer Res. 27:5619–5627. 2021. View Article : Google Scholar : PubMed/NCBI

128 

Kommalapati A, Tella SH, Borad M, Javle M and Mahipal A: FGFR inhibitors in oncology: Insight on the management of toxicities in clinical practice. Cancers (Basel). 13:29682021. View Article : Google Scholar : PubMed/NCBI

129 

Rugo HS: Management of toxicities from antibody-drug conjugates. Clin Adv Hematol Oncol. 23:96–99. 2025.PubMed/NCBI

130 

Zhang M, Liu C, Tu J, Tang M, Ashrafizadeh M, Nabavi N, Sethi G, Zhao P and Liu P: Advances in cancer immunotherapy: Historical perspectives, current developments, and future directions. Mol Cancer. 24:1362025. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Du J, Shen H, Zeng T, Liu W and Xie Y: Targeted therapies for urothelial carcinoma: From FGFR inhibitors to next‑generation antibody-drug conjugates (Review). Int J Oncol 68: 28, 2026.
APA
Du, J., Shen, H., Zeng, T., Liu, W., & Xie, Y. (2026). Targeted therapies for urothelial carcinoma: From FGFR inhibitors to next‑generation antibody-drug conjugates (Review). International Journal of Oncology, 68, 28. https://doi.org/10.3892/ijo.2025.5841
MLA
Du, J., Shen, H., Zeng, T., Liu, W., Xie, Y."Targeted therapies for urothelial carcinoma: From FGFR inhibitors to next‑generation antibody-drug conjugates (Review)". International Journal of Oncology 68.2 (2026): 28.
Chicago
Du, J., Shen, H., Zeng, T., Liu, W., Xie, Y."Targeted therapies for urothelial carcinoma: From FGFR inhibitors to next‑generation antibody-drug conjugates (Review)". International Journal of Oncology 68, no. 2 (2026): 28. https://doi.org/10.3892/ijo.2025.5841
Copy and paste a formatted citation
x
Spandidos Publications style
Du J, Shen H, Zeng T, Liu W and Xie Y: Targeted therapies for urothelial carcinoma: From FGFR inhibitors to next‑generation antibody-drug conjugates (Review). Int J Oncol 68: 28, 2026.
APA
Du, J., Shen, H., Zeng, T., Liu, W., & Xie, Y. (2026). Targeted therapies for urothelial carcinoma: From FGFR inhibitors to next‑generation antibody-drug conjugates (Review). International Journal of Oncology, 68, 28. https://doi.org/10.3892/ijo.2025.5841
MLA
Du, J., Shen, H., Zeng, T., Liu, W., Xie, Y."Targeted therapies for urothelial carcinoma: From FGFR inhibitors to next‑generation antibody-drug conjugates (Review)". International Journal of Oncology 68.2 (2026): 28.
Chicago
Du, J., Shen, H., Zeng, T., Liu, W., Xie, Y."Targeted therapies for urothelial carcinoma: From FGFR inhibitors to next‑generation antibody-drug conjugates (Review)". International Journal of Oncology 68, no. 2 (2026): 28. https://doi.org/10.3892/ijo.2025.5841
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team