Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Oncol., 02 December 2025

Sec. Pharmacology of Anti-Cancer Drugs

Volume 15 - 2025 | https://doi.org/10.3389/fonc.2025.1654771

This article is part of the Research TopicTreatment Resistance in Genitourinary CancerView all articles

Overcoming resistance in advanced urothelial carcinoma: mechanisms of escape from antibody-drug conjugates and FGFR3 inhibition

  • 1Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
  • 2Emory University Winship Cancer Institute, Atlanta, GA, United States

For decades, platinum chemotherapy was the mainstay of treating metastatic urothelial carcinoma (mUC). More recently, checkpoint inhibitors (CPI) were an important addition to the armamentarium capable of inducing durable responses for a minority of patients. Management of mUC has changed significantly with the advent of antibody-drug conjugate (ADC) therapies and fibroblast growth factor receptor inhibitors (FGFRi). Enfortumab vedotin, a Nectin-4 targeting ADC, is now the first line therapy of choice in combination with pembrolizumab. Erdafitinib, a pan FGFR1–4 inhibitor, is approved for patients with susceptible FGFR3 alterations. There are multiple other agents in development within both therapeutic classes that hold promise. But most patients will still succumb to their disease, either via primary or secondary resistance. This review looks critically at the approved and pipeline ADC and FGFR-targeting agents of interest in mUC as well as known mechanisms of resistance by which their efficacy is dampened. We propose strategies for overcoming resistance including combination strategies, tumor microenvironment modification, and drug structure modification to maximize efficacy. The progress to date in mUC has been remarkable, but there is still significant work to do in this deadly disease and this review highlights the gap between current available therapeutics and cure that so desperately needs to be closed.

Introduction

Urothelial carcinoma (UC) is the ninth most common cancer in the world with 550,000 new cases annually (1). Historically, platinum-based chemotherapy has been the standard treatment for metastatic UC (mUC). However, more than half of patients with mUC were ineligible for cisplatin therapy and those who received treatment ultimately developed progression of their disease (2, 3). The treatment landscape evolved with the introduction of checkpoint inhibitors (CPI), initially used as monotherapy for cisplatin-ineligible or relapsed patients. Despite these advances, response rates to CPI monotherapy remain modest, ranging from 15 to 21%, with only a small subset of patients achieving a durable benefit (4). In response, CPI was used as maintenance therapy following platinum chemotherapy, and more recently in combination with chemotherapy followed by continued maintenance (58).

The emergence of antibody-drug conjugates (ADCs) and targeted therapies has reshaped the therapeutic paradigm in treating mUC. ADCs consist of three components: an antibody that binds target antigens expressed on tumor cells, a small molecule cytotoxic drug payload, and a linker molecule. After binding to its target tumor antigen, ADCs are endocytosed into tumor cells, processed and unlinked within lysosomes, releasing the payload and leading to cell cycle arrest through direct cytotoxicity. In contrast, targeted therapies act directly on intracellular signaling pathways that drive tumor progression (Figure 1).

Figure 1
Illustration showing mechanisms of antibody-drug conjugates (ADC) resistance. ADC binds to tumor antigen, undergoes endocytosis, forms early endosomes, and reaches lysosomes for payload release. Resistance mechanisms include immune evasion, drug efflux pumps, impaired drug internalization, and antigen resistance or downregulation. FGFR inhibitors (FGFRi) face antigen resistance or bypass signaling. The diagram includes T-cells and molecular signaling pathways like JAK, STAT, and MAPK.

Figure 1. Mechanisms of action for antibody-drug conjugate (left) and FGFR inhibitor (right) therapies and corresponding mechanisms of resistance.

Enfortumab vedotin (EV) is an ADC that targets nectin-4, a cell surface adhesion protein expressed on most urothelial carcinoma cells (9). When combined with pembrolizumab, a PD-1 inhibitor, EV has significantly improved clinical outcomes in patients with treatment-naïve mUC, nearly doubling the progression-free survival (PFS) and overall survival (OS) when compared to platinum-based chemotherapy (10). This combination has since been approved as first-line therapy in mUC regardless of cisplatin eligibility and is the new standard of care for most patients with mUC. Other ADCs currently approved for use in mUC include trastuzumab deruxtecan (T-DXd), a Her2-targeting ADC with a tissue-agnostic approval for HER2-expressing tumors, including UC (11). Sacituzumab govitecan, an anti-Trop-2 ADC that previously held an accelerated FDA approval in mUC based on a promising phase 2, single arm trial later lost this designation after the confirmatory phase 3 study showed no improvement in OS over single-agent chemotherapy. Disitamab vedotin is a HER2-targeting ADC of great interest in the pipeline, as are the Trop-2 targeting datopotamab deruxtecan and sacituzumab tirumotecan which have showed promise in early-phase clinical trials (Table 1).

Table 1
www.frontiersin.org

Table 1. Selected trials and outcomes for antibody-drug conjugates (ADCs) and FGFR inhibitors in advanced UC.

Despite substantial therapeutic advances, most patients with mUC will ultimately experience disease progression either due to lack of initial response (primary resistance) or, more commonly, through the development of resistance after initial sensitivity (secondary resistance) (12, 13). These patterns of limited or transient efficacy are driven by intrinsic and adaptive tumor cell resistance mechanisms. Tumor cells can downregulate or modify target antigen expression, thereby reducing antigen-binding affinity. Following ADC internalization, enhanced lysosomal sequestration and overexpression of efflux pumps such as multidrug resistance 1-(MDR-1) and breast cancer resistance protein (BCRP) prevents ADC trafficking for payload delivery. Lastly, tumor cells can alter their own tumor microenvironment (TME), which can further attenuate therapeutic efficacy (Figure 1). Collectively, these resistance mechanisms highlight the need for combinatorial strategies and biomarker-driven approaches to overcome therapeutic escape and sustain durable responses.

Fibroblast growth factor receptor (FGFR) alterations are present in approximately 20% of mUC and are one of the few molecular targets with an FDA-approved therapeutic for mUC (14, 15). Erdafitinib, an FGFR 1–4 tyrosine kinase inhibitor (TKI), is approved for the treatment of mUC in patients harboring susceptive FGFR3 genetic alterations, underscoring the importance of routine somatic next generation sequencing in all patients with mUC. Several other FGFR targeting agents are under investigation. Ongoing trials continue to refine FGFR-targeting therapies to overcome resistance mechanisms, which highlights it role as a valuable therapeutic option for a subset of patients with mUC. (Table 1; Figure 1).

The advent of these novel therapeutics such as ADCs and FGFR-targeting TKI therapies has significantly expanded the treatment landscape for mUC. However, the targeted approach is not immune from the development of resistance and the key to improved long-term outcomes lies in identifying and circumventing these resistance mechanisms, both primary and secondary. This review explores the biological underpinnings of resistance to ADCs and FGFR inhibitors used to treat mUC and highlights emerging strategies aimed at circumventing these therapeutic challenges. This review provides the first systematic, side-by-side comparison of resistance mechanisms between ADCs and FGFR inhibitors in mUC, highlighting the overlapping and diverging pathways of therapeutic escape. By comparing these resistance patterns, this review can inform future development of combination regimens and drug design strategies to effectively overcome resistance.

Antibody-drug conjugates of interest for treatment of urothelial carcinoma

Enfortumab vedotin

EV is an ADC that is comprised of a human monoclonal antibody against nectin-4 conjugated to monomethyl auristatin E (MMAE), an inhibitor of microtubule formation, via a protease-cleavable linker (16, 17). Nectin-4 is a transmembrane protein that is highly expressed in multiple solid tumors, namely urothelial, gastric, and breast carcinomas, and is a marker of poor prognosis (1820). When EV binds nectin-4, it becomes internalized and releases MMAE which disrupts microtubule formation and leads to cell-cycle arrest in tumor cells. Clinically, EV has shown promising efficacy in treatment of mUC. EV-201 was a single-arm, phase II clinical study of EV monotherapy that demonstrated a 52% overall response rate (ORR) in patients with mUC who had received previous CPI treatment without chemotherapy, highlighting the efficacy of EV in patients with limited treatment options (21, 22). The phase III trial EV-301 showed that EV monotherapy improved medial overall survival (mOS) (12.9 vs. 9.0 months; hazard ratio [HR] 0.70) and median progression-free survival (mPFS) (5.6 vs. 3.7 months; HR 0.62) compared to standard chemotherapy post-platinum and post-CPI over a median follow-up of 24 months. The ORR was 40.6% with EV compared to 17.9% with chemotherapy. Skin toxicity, hyperglycemia, and peripheral neuropathy are adverse events of particular interest when using EV.

EV leapt from the third line to the first line setting after EV-302 showed an improvement in OS in patients treated with EV plus pembrolizumab (EV-P) regardless of cisplatin eligibility (10). A new benchmark for median survival was set at 31.5 months vs 16.1 months with platinum doublet chemotherapy (hazard ratio 0.47; 95% CI 0.38 to 0.58, P<0.001). A total of 67.7% of patients responded to EV-P compared to 44.4% of patients who received chemotherapy; this includes a complete response (CR) rate of 29.1%. The improvement in survival was seen in both cisplatin-eligible and ineligible patients, a finding that has since made cisplatin eligibility criteria, largely in the first-line setting, a relic of the past (23). We know from an updated analysis presented at ASCO 2025 that of patients who achieved a CR, 74.3% of them maintained it 24 months later, indicating the durability of this response and opening the door to a potential cure in mUC (24).

Trastuzumab deruxtecan

Human epidermal growth factor receptor 2 (HER2) is a transmembrane tyrosine kinase inhibitor involved in cell proliferation and differentiation that is overexpressed in many solid tumors including urothelial carcinoma (25, 26). Trastuzumab deruxtecan (T-DXd) is a HER2-directed ADC composed of an anti-HER2 antibody, topoisomerase I inhibitor payload, and a tetrapeptide-based cleavable linker (27, 28). In 2023, DESTINY-PanTumor02, a phase II trial, demonstrated that T-DXd had durable antitumor activity across many HER2 positive tumor types, including bladder carcinoma (n=41). The response rate was 56.3% in patients with UC with HER2 IHC 3+ and 35.0% in those with IHC 2+ (11). The tissue-agnostic approval of this agent in cancers with HER2 IHC 3+ has led to routine testing for this biomarker in the later line setting of mUC. Recent data from a phase Ib, non-randomized study showed that combining T-DXd with nivolumab, a PD-1 inhibitor, had synergistic anti-tumor activity in HER2-positive mUC (29). Pneumonitis remains an adverse event of interest with use of this drug.

Disitamab vedotin

Disitamab vedotin (DV) is also a HER2-targetng ADC with a MMAE payload, like EV. Initially, DV monotherapy demonstrated potent antitumor activity in patients with HER2-positive locally advanced or metastatic UC who had progressed on at least one line of chemotherapy (30). In combined analysis of two-phase II clinical trials, DV monotherapy achieved an ORR of 50.5% of patients with HER2-positive, locally advanced or metastatic UC who had progressed on prior therapies (3133). The median PFS was 5.9 months, and the median OS was 14.2 months. Beyond monotherapy, DV has been evaluated in combination with CPI to enhance antitumor activity (3436). When combined with toripalimab in an HER2 unselected population within a phase Ib/2 trial, the ORR was 76% in treatment-naïve patients, 83.3% in those who were HER2 IHC 3+/2+, and even 33.3% in those who were completely negative for HER2 expression on IHC (32). An ongoing phase 3 study of DV plus pembrolizumab versus chemotherapy in patients with HER2-expressing mUC may further elucidate its role in the treatment of mUC (37).

Sacituzumab govitecan

Sacituzumab govitecan (SG) is an ADC that is composed of an anti-trophoblast cell surface protein (Trop-2) IgG1 monoclonal antibody linked to an SN-38 payload. SN-38 is a metabolite of irinotecan, a topoisomerase I inhibitor (38). The phase II trial TROPHY-U-01 studied the efficacy of SG in patients with mUC who were heavily pretreated with both chemotherapy and immunotherapy. In the single-arm study, SG demonstrated an ORR of 28% with mPFS of 5.4 months and OS of 10.9 months. The study showed that SG had sustained clinical benefit independent of PD-L1 expression or prior response to checkpoint inhibition, leading to the accelerated FDA approval of SG for patients with mUC (39). More recently, TROPiCS-04 was a phase III randomized trial that compared SG to TPC (paclitaxel, docetaxel, or vinflunine) in the same patient population (40). Unfortunately, SG did not result in a statistically significant improvement in OS or PFS when compared to standard chemotherapy. However, the ORR was higher at 23% for SG compared to single-agent chemotherapy (14%). There was an enrichment in neutropenic deaths occurring within the SG-containing arm, most occurring within the first month of treatment, which may have at least in some part contributed to the negative results of this phase 1 study. Only 21% of patients received granulocyte colony stimulating factor (G-CSF) as primary prophylaxis was not mandated. This negative trial led to voluntary withdrawal of the accelerated approval status for SG by the manufacturer. Questions remain about the future of SG in mUC.

Datopotamab deruxtecan

Datopotamab deruxtecan (Dato-DXd) is another ADC that targets Trop-2 with a topoisomerase I inhibitor payload. Trop-2 is overexpressed in several epithelial tumors, including UC, and is a marker of aggressive tumor behavior (41, 42). TROPION-PanTumor01 is an ongoing, phase 1, multicohort study investigating Dato-DXd in multiple tumor types and the early data from mUC was presented in 2025. A total of 40 patients were treated with the drug in the study, resulting in an ORR of 25%, disease control rate (DCR) of 77.5%, and a 6-month duration of response rate of 76.2%. This study was conducted in a heavily pre-treated population with 50% of patients having received at least 3 prior lines of therapy in the metastatic setting (43). Pneumonitis and stomatitis are the most concerning adverse events with this agent (44).

Sacituzumab tirumotecan

Sacituzumab tirumotecan (Sac-TMT) is another Trop-2 targeting ADC linked to a topoisomerase I payload. A phase I trial of this agent was conducted in a patient population previously treated with prior platinum doublet chemotherapy and immunotherapy (45). In the patients who received sac-TMT in the second line (N = 11), the ORR was 45.5%; it was 26.3% in those which received it in the third line and beyond (N = 38). The primary adverse events that occurred were anemia (38.8%) and decreased neutrophil count (28.6%), however there were no treatment-related deaths to date.

FGFR-3 inhibitors for treatment of urothelial carcinoma

Molecular studies of UC continue to identify new oncologic targets for targeted therapy. Among these targets, fibroblast growth factor receptor (FGFR) is an implicated oncogene that potentiates cell proliferation, differentiation, and angiogenesis via activation of the PI3K-AKT, PLC-gamma, STAT, and RAS-MAPK pathways (46). FGFR gene alterations, which have been identified in up to 20% of mUC and as high as 40% of cases arising from upper tract disease, are the biologic target for next-generation pan-FGFR inhibitors (15, 47). Erdafitinib is a potent FGFR 1–4 inhibitor that is now approved for the treatment of mUC in adults with susceptible FGFR3 alterations who have received at least 1 prior line of therapy. Unlike ADCs which are internalized within tumor cells, erdafitinib binds to surface receptor tyrosine kinase (RTK) to downregulate downstream aberrant signaling (48). This mechanism allows erdafitinib to exert its anti-tumor effect without relying on intracellular delivery, offering a complementary strategy to ADC-based therapies.

In a single-arm, phase 2 trial (BLC2001), 40% of patients with mUC with FGFR2/3alterations who progressed on platinum-based chemotherapy had an objective treatment response with erdafitinib monotherapy (49). The mPFS was 5.5 months (95% CI 4.2-6.0) and the mOS was 13.8 months (95% CI 9.8-not reached). THOR was the confirmatory phase 3 trial that compared erdafitinib to chemotherapy after tumor progression following anti-PD-1 or anti-PD-L1 therapy. The study found that erdafitinib resulted in significantly prolonged PFS of 5.6 months compared to 2.7 months in the chemotherapy cohort (HR 0.64; 95% CI, 0.47-0.88; p=0.005) (50). However, when compared to pembrolizumab monotherapy, erdafitinib had similar mean OS in FGFR-altered UC (51). Tolerability is a concern with FGFR inhibitors and patients on erdafitinib struggle with hyperphosphatemia, gastrointestinal symptoms, palmar-plantar erythrodysesthesia syndrome, and onycholysis. Erdafitinib is currently FDA approved for patients with mUC with susceptible FGFR3 genetic alterations after at least one line of prior systemic therapy.

Multiple other FGFR inhibitors have been investigated. A phase I trial of infigratinib, an oral FGFR 1–3 TKI in FGFR3-mutated mUC showed an ORR of 25.4%, mPFS of 3.75 months and mOS of 7.75 months in patients who progressed on at least one line of prior therapy (52). Pemigratinib is an oral GFR1–3 inhibitor that has been studied in a phase II, single-arm study in patients with FGFR3 mutations and other FGFR mutations. There was similar ORR in patients with susceptible FGFR3 mutations who received continuous dosing (23.9%) and intermittent dosing (24.6%). There was minimal activity in patients with other FGF/FGFR mutations other than FGFR3 (53). Rogaratinib is a pan-FGFR inhibitor (1-4) that was studied in a phase II FORT-1 trial that compared single-agent chemotherapy in patients with FGFR1/3 mRNA positive mUC with at least 1 prior line of therapy. The mOS was 8.3 versus 9.8 months (HR 1.11; 95% CI 0.71-1.72, P = 0.76) and the response rates between the arms were similar (20.7% versus 19.3%) (54). A major focus of ongoing FGFR targeting therapy is tolerability and more selective FGFR inhibitors are being investigated. One example is LOXO-435, an isoform-selective, small molecule inhibitor of FGFR3 designed to mitigate off-target effects of FGFR inhibition (55, 56).

Mechanisms of resistance to ADCs and FGFR3 inhibitors

Because the mechanism of action of ADCs involves sequential molecular events to deliver its cytotoxic payload and disrupt DNA production, tumor cells can develop resistance mechanisms at multiple stages. These mechanisms include alteration of its surface antigens to diminish its binding efficacy, failure in drug internalization and trafficking, upregulation of drug-efflux pumps, resistance to cytotoxic payload, activation of bypass signaling pathways, and modulation of the tumor microenvironment (TME) (Figure 1). Further investigation into these mechanisms may provide a better understanding into why patients with mUC have insufficient responses to ADC therapy and FGFR inhibition.

Antigen-related resistance

ADC and FGFR inhibitors target specific antigens on the surface of tumor cells. Therefore, a common proposed mechanism of resistance is tumor cell alteration of its surface antigen expression level. Loganzo et al. studied the mechanisms by which breast cancer cell lines develop resistance to ADCs through persistent drug exposure and found that downregulation of HER2 antigen or increased ABCC1 protein expression were the driving mechanisms (57). This preclinical study suggested that tumor heterogeneity in HER2 expression may correlate to ADC efficacy and thereby to clinical outcomes. These results were corroborated in the KRISTINE and ZEPHIR clinical trials which found that tumors with increased heterogeneity in HER2 expression prior to treatment with ADCs had worse clinical outcomes when compared to patients with tumors of low heterogeneity (58, 59).

To date, several studies have found that NECTIN-4 amplification predicts EV response in mUC. Klümper et al. demonstrated that NECTIN-4 protein expression, independent of gene amplification, is downregulated during metastatic progression of mUC and that its expression correlates with EV response (60). This study suggests that NECTIN-4 amplification can be used as a genomic biomarker for patients with mUC undergoing EV therapy to predict clinical response. Similarly, alteration of target antigen is a common resistance mechanism against FGFR inhibitors. Gatekeeper mutations in FGFR3, such asV555M, following administration of FGFR inhibitors in non-small cell lung cancer can interfere with drug binding, leading to eventual erdafitinib resistance (61, 62).

Impaired drug internalization and trafficking pathways

After binding to its surface antigen, ADCs are internalized into tumor cells via receptor-mediated endocytosis. ADC efficacy is reliant on target-mediated endocytosis to deliver its cytotoxic payload at sufficient concentrations to cause tumor cytotoxicity (63). There are several endocytosis pathways that mediate ADC uptake: clathrin-mediated endocytosis (CME), clathrin-independent endocytosis, and caveolae-mediated endocytosis (64, 65). CME is a receptor-mediated endocytosis process and is the most thoroughly studied in ADC transport. Briefly, the ADC-antigen complex gets internalized through clathrin-coated pits which invaginate and form vesicles with endosomes. The ADC is then trafficked through the endosomal-lysosomal pathway in which the cytotoxic payload is released. Sung et al. developed in vitro ADC-resistant breast cancer cells lines that internalize ADCs into caveolin-1 (CAV1)-coated vesicles which alters their trafficking to lysosomes (66). ADC colocalization into CAV1 vesicles reduced therapy response and was a negative biomarker for patient response. These results underscore the importance of understanding endocytosis pathways as alterations can impact therapeutic outcomes.

Drug-efflux pumps and payload resistance

Another common mechanism of ADC resistance is elimination of the drug by the overexpression of ATP-binding cassette (ABC) transporters, which actively efflux the cytotoxic payloads out of the cancer cells. ABCB1 (P-glycoprotein) and ABCC1 (multidrug resistance-associated protein 1 [MDR1]) are the most well-characterized ABC transporters, which rely on ATP hydrolysis to translocate ADC payloads across cell members and thereby reduce its intracellular concentration. Corbett et al. showed that upregulation of ABCB1 and ABCC1 transporters was associated with resistance to ADC-containing pyrrolobenzodiazepine (PBD) and that inhibiting these transporters could restore drug sensitivity in previously resistant cancer cells (67). Similarly, T-DM1–resistant cells had an increased expression of ABC transporters despite preserved HER2 overexpression, and pharmacologic inhibition of these transporters reinstated T-DM1 responsiveness (68).

Upregulation of drug-efflux pumps correlates to enhanced toxic payload resistance. Studies have found that upregulation of drug efflux transporters such as P-glycoprotein is associated with resistance to MMAE, the payload used in EV. Chang et al. created EV-resistant bladder cancer cell lines in vitro through upregulation of P-glycoprotein and TGF-β genes which led to decreased sensitivity to MMAE (69). They found that resistance to EV was largely attributable to resistance to the payload MMAE rather than downregulation of surface antigen Nectin-4.

Bypass signaling pathways

Activation of alternative signaling pathways is another mechanism of resistance to ADCs and FGFR inhibitors. One prominent mechanism involves the activation of PI3K/AKT/mTOR pathway which promotes tumor cell survival and proliferation. This pathway has been most prominently studied with trastuzumab which showed that PIK3CA mutations resulted in decreased sensitivity to the ADC, and that adding a PI3K inhibitor to trastuzumab had enhanced anti-tumor activity in HER2-positive metastatic breast cancer (70, 71). In mUC, upregulation of the PI3K/AKT/mTOR pathway correlates with increased EV resistance and enhanced tumor cell survival (72). Another critical bypass signaling pathway involves the TGF-β signaling pathway, which can induce the epithelial-mesenchymal transition (EMT) and enhanced tumor metastasis. In urothelial cancer, TGF-β signaling has been implicated in resistance to EV.

Similarly, resistance to FGFR3 inhibitors in urothelial carcinoma can occur through activation of downstream signaling pathways. Hosni et al. demonstrated that adipocyte precursor-derived neuregulin 1 (NRG1) promotes resistance to FGFR inhibition by activating the epidermal growth factor receptor 3 (ERBB3; also known as HER3) signaling pathway, which can bypass the inhibited FGFR3 pathway to sustain tumor cell proliferation (73). In another study, Weickhardt et al. showed that increased expression of phosphorylated ERBB3 is a key resistance mechanism in FGFR3-dependent bladder cancer and that dual targeting of FGFR3 and ERBB3 delayed the reactivation of pERBB3 and enhanced FGFR inhibitor efficacy (74). Better understanding of these signaling pathways is crucial in developing therapies that counteract resistance mechanisms and improve clinical outcomes.

TME and immune evasion

The TME consists of network of stromal cells, immune cells, extracellular matrix, and soluble factors that interact with tumor cells. The TME plays an important role in tumor progression, metastasis, and immune escape which engenders resistance to ADCs. In addition to direct cytotoxicity, ADCs can induce immunogenic cell death (ICD), which is the release of damage-associated molecular proteins (DAMPs) such as calreticulin, ATP, and HMGB1 from dying tumor cells (75, 76). These DAMPs then activate dendritic cells (DCs) which in turn activate T cells to generate a robust antitumor response. Tumor cells can create an immunosuppressive TME phenotype through the recruitment of regulatory T cells (Tregs), MDSCs, and secretion of cytokines such as TGF-B. These changes decrease the effectiveness of immune effector cells, thereby inhibiting ICD and reducing the efficacy of ADCs. Recent studies have found that ADCs conjugated with payloads known to be strong modulators of the immune microenvironment such as pyrrolobenzodiazepine or tubulysin more effectively induce ICD, thereby synergizing with CPI (77, 78).

One significant mechanism involves upregulation of immune checkpoint molecules, such as programmed death-ligand 1 (PD-L1), which inhibits the cytotoxic activity of immune cells. This upregulation of PD-L1 can be a response to the inflammatory response induced by ADCs and creates an immunosuppressive TME, leading to an adaptive resistance mechanism. These changes decrease the effectiveness of immune effector cells, thereby reducing the efficacy of ADCs. As discussed above, combining ADCs or FGFR inhibitors with CPIs can potentially overcome this resistance mechanism.

Ouyang et al. explored the impact of FGFR3 alterations in bladder cancer TME and demonstrated that mutant FGFR3 indirectly induces an immunosuppressive TME by increasing serine synthesis. This activates the PI3K/Akt pathway and suppresses macrophage immunostimulatory functions, shifting them toward an immune-inert phenotype (79). Targeting PI3K in FGFR3 tumors reversed the macrophage phenotype and demonstrated synergistic antitumor activity when combined with erdafitinib. Overcoming these immunosuppressive modulations of TME through direct inhibitors is a potential strategy to enhance the effectiveness of ADC and FGFR3-inhibitors in mUC.

Overcoming resistance to optimize ADCs and FGFR3 inhibitors

Combination therapies

Overcoming tumor resistance to ADCs and FGFR3 inhibitors to increase clinical response rate requires a multifaceted approach. One promising strategy is the use of combination therapies. Combining ADCs with ICI both leverages the cytotoxic effects of ADCs while also activating the host’s immune system to target UC tumor cells. EV was initially approved as a single agent in the third line setting of mUC after prior platinum and prior CPI therapy. EV-302 study, a phase III randomized clinical trial, showed that when combined with pembrolizumab, EV had an overall response rate (ORR) of 67.7% compared to 45.2% in EV monotherapy arm in patients with untreated la/m UC (10, 80). These results highlight the synergistic and not just additive mechanism of action between EV and pembrolizumab and led to a change in the standard of care management of mUC (10, 81). However, despite these promising results, it remains uncertain whether the observed benefits represent true synergy or reflect additive effects from two separate agents. Additionally, overlapping toxicities such as rash, peripheral neuropathy, and immune-related adverse events require careful management and may limit broader applicability in frailer populations.

SC is another ADC which has been studied in combination with CPI. Results of the JAVELIN Bladder Medley trial were presented at ASCO 2025, a phase 2 study investigating maintenance avelumab plus SG versus avelumab alone. The study showed improved PFS but there was no significant difference in OS at the time of this interim analysis and there was toxicity incurrent in the investigational arm consistent with the known profile of SG (82). These findings underscore that not all ADC–ICI combinations yield clear survival advantages, and further data are needed to determine whether improved progression data translate to durable, clinical benefit. There is an enrichment of Trop-2 expression in variant histology and ongoing trials in rare bladder histology are of interest. The SMART trial is investigating SG with or without atezolizumab in locally advanced unresectable or metastatic rare genitourinary cancers including small cell, squamous cell, and adenocarcinoma of the bladder (NCT06161532) (83). The phase 1 DAD trial showed that combination EV and SG is possible. Of the 23 enrolled patients, the response rate was 70% with a manageable safety profile once prophylactic G-CSF was added (84). The DAD-IO trial builds on this concept and is a phase 2 trial of combination EV, SG and pembrolizumab in the first-line setting (85).

Similarly, overcoming resistance to FGFR inhibitors can also be achieved through combination therapies targeting complementary pathways. The BISCAY trial combined durvalumab, a PD-L1 inhibitor, with FGFR inhibitors in mUC but did not show enhanced efficacy over durvalumab monotherapy (86). In contrast, the NORSE study evaluated the combination of erdafitinib with cetrelimab, a PD-1 inhibitor, in FGFR-altered mUC, which had a ORR of 54.5% and a 12-month OS rate of 68%, compared to an ORR of 44.2% and a 12-month OS rate of 56% for erdafitinib monotherapy (87). This study could be interpreted as suggesting that combining FGFR inhibition with ICIs may enhance therapeutic efficacy by reprogramming the TME to support antitumor immunity. Although, it is not clear whether the efficacy is simply additive because of the two classes of agents. Moreover, long-term toxicity profiles of these combinations remain incompletely characterized, particularly given overlapping risks such as hyperphosphatemia, ocular events, and immune-related toxicities. Lastly, targeting bypass signaling pathways that contribute to resistance is another effective strategy. The phosphoinositide 3-kinase (PI3K) pathway has been identified as a key determinant of resistance to FGFR inhibitors (88). Inhibition of PI3K with agents such as BKM120 has been shown to act synergistically with FGFR inhibitors, enhancing their efficacy in urothelial carcinoma cell lines harboring FGFR mutations (8890). However, these findings are largely preclinical, and translating synergistic activity into clinical benefit will require further evaluation of combinatorial dosing and scheduling in future trials.

Targeting the TME

One strategy to overcome resistance is reprogramming the immune-suppressive features within the TME. For instance, MDSCs, tumor-associated macrophages (TAMs), and regulatory T cells (Tregs) facilitate an immunosuppressive TME which limits drug efficacy. Targeting these immune-suppressive cells with immune-modulating therapies such as CSF1R inhibitors can deplete TAMs. Resistance to FGFR inhibitors is mediated by the TME through promotion of TAMs which can secrete factors that activate the FGFR pathway, leading to tumor cell survival. Another approach involves remodeling the extracellular matrix (ECM) which can act as a physical barrier to drug delivery. Enzymatic degradation of the ECM by hyaluronidase can enhance the penetration of DCs within the tumor (91, 92). Normalization of tumor vasculature through anti-angiogenic agents such as vascular endothelial growth factor (VEGF) inhibitors or EGFR tyrosine kinase inhibitors (TKIs), such as osimertinib, may be useful.

Moreover, the TME can influence the expression of drug efflux pumps and enzymes that deactivate the cytotoxic payload. Resistance mechanisms that increase drug excretion by overexpressing drug efflux pumps can be overcome by implementing direct inhibitors of these pumps. Tariquidar, an ATPase inhibitor of P-glycoprotein drug efflux pump, has been used in combination with chemotherapy to increase drug exposure in resistance cancers, including renal cell carcinoma (93, 94). While no studies yet exist combining ADCs with drug efflux pump inhibitors, Cabaud et al. demonstrated that resistance to an anti-nectin 4 ADC could be reversed by tariquidar in preclinical breast cancer model (95).

Novel drug design

One of the most direct strategies to circumvent resistance to ADCs and FGFR inhibitors is structural modification of these drugs to create next-generation compounds capable of overcoming established resistance mechanisms. There are three components of ADCs that can be altered to prevent resistance: antigen targeting, improved linker technology, and payload delivery. Novel bispecific ADCs that target two tumor cell antigens can reduce the likelihood of resistance due to antigen loss in tumor cells with low antigen expression. Dual targeting of HER2 by biparatopic ADCs has been shown to enhance toxin delivery to breast cancer cells with significant intratumor heterogeneity of HER2 expression (96). Preliminary results from a phase I study on a HER2 bispecific ADC, zanidatamad zovodotin, showed an ORR of 28% with disease control rate of 72% across multiple cancer types (97). Additionally, new bispecific ADCs that selectively bind two different antigens, such as Trop2 and Nectin4, are being studied in UC to enhance its selectivity and minimize off target effects (98). In addition, novel technologies like the bicycle drug conjugate (BDC) may be helpful to this end. These molecules are tiny with a short plasma half-life, holding the potential for increased tumor exposure and decreased normal tissue activity. Zelenectide pevedotin is a BDC highly selective for Nectin-4 and linked to MMAE. This agent is currently being studied within the DURAVELO-2 phase 2/3 study of the agent as monotherapy as well as in combination with pembrolizumab versus platinum doublet chemotherapy (99).

Secondly, linker structure can be modified to further optimize ADCs. Recent advancements have led to the development of cleavable linkers that are sensitive to specific TME conditions that ensure the payload is released within tumor cells. Incorporation of hydrophilic XTEN polypeptides in linker design have resulted in ADCs with extended half-lives (100). Lastly, payload delivery can be modified. Li et al. studied how the concentration of payload release affects ADC efficacy and found that the higher rates of payload delivery correlated to enhanced killing of neighboring tumor cells that did not express the target antigen, also known as the bystander effect (101). Creation of dual-drug ADCs, ADCs that deliver two distinct cytotoxic payloads, has shown synergistic antitumor activity in UC mouse models (102104). Ongoing research aims to optimize the potential of dual-drug ADCs to overcome resistance where single-drug ADCs have failed in refractory tumor types.

Development of next generation of FGFR inhibitors is currently ongoing. Resistance to these agents often arise from secondary mutations in the FGFR kinase domain such as gatekeeper mutations like V555M in FGFR3 that reduce drug binding. Futibatinib, an irreversible FGFR1–4 inhibitor, forms covalent bonds with the kinase and maintains efficacy even in patients with solid tumors that have acquired resistance to prior FGFR inhibitors (105). Similar to ADCs, dual-targeted inhibitors are being engineered that can bypass resistance of activating signaling pathways such as EGFR, P13K/AKT, and MAPK pathways. Dual targeting of FGFR3 and ERBB3 showed to overcome resistance of FGFR3-fusion driven bladder cancer (74).

Conclusion and future directions

The therapeutic landscape of mUC has rapidly progressed with the development of ADCs and FGFR inhibitors which offer durable clinical benefit to patients with mUC who have limited treatment options. However, both primary and secondary resistance to these therapeutics limit the depth and durability of response to ADCs and FGFR inhibitors. Intrinsic tumor resistance—namely, antigen downregulation, impaired ADC internalization and trafficking, drug efflux pumps, and bypass signaling pathways—alongside TME-mediated immune suppression all contribute to eventual therapeutic failure. In response, the development of next-generation ADCs and novel combination therapies must be aimed at overcoming these resistance mechanisms by enhancing tumor specificity and mitigating toxicity.

Now that EV and pembrolizumab have become first-line standard-of-care for mUC, the paradigm in this disease has shifted from the old guard of platinum doublet chemotherapy followed by immunotherapy. While outcomes have improved dramatically, most patients will not be cured of their disease even today. A better understanding of resistance mechanisms to these agents and how to overcome them will be essential. Fortunately, we have a wide armamentarium of potential options including biomarker selected and unselected ADC agents and FGFR inhibitors in the pipeline that are poised to change the future of this disease. A proactive approach utilizing combination therapies and novel agents to increase responses and survival is ideal, but attention also remains trained on salvage therapies to recapture response in patients who have experienced progression of their disease.

Author contributions

BW: Visualization, Writing – original draft, Data curation, Writing – review & editing, Resources. MS: Writing – review & editing, Writing – original draft. JC: Writing – review & editing, Writing – original draft. SA: Writing – review & editing, Writing – original draft. SJ: Writing – original draft, Writing – review & editing. VN: Writing – original draft, Writing – review & editing. BC: Writing – review & editing, Writing – original draft. MB: Writing – original draft, Writing – review & editing. JB: Supervision, Writing – review & editing, Writing – original draft, Conceptualization.

Funding

The author(s) declare that no financial support was received for the research, and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2025.1654771/full#supplementary-material

References

1. Richters A, Aben KKH, and Kiemeney L. The global burden of urinary bladder cancer: an update. World J Urol. (2020) 38:1895–904. doi: 10.1007/s00345-019-02984-4

PubMed Abstract | Crossref Full Text | Google Scholar

2. De Santis M, Bellmunt J, Mead G, Kerst JM, Leahy M, Maroto P, et al. Randomized phase II/III trial assessing gemcitabine/carboplatin and methotrexate/carboplatin/vinblastine in patients with advanced urothelial cancer who are unfit for cisplatin-based chemotherapy: EORTC study 30986. J Clin Oncol. (2012) 30:191–9. doi: 10.1200/JCO.2011.37.3571

PubMed Abstract | Crossref Full Text | Google Scholar

3. Loehrer PJ Sr, Einhorn LH, Elson PJ, Crawford ED, Kuebler P, Tannock I, et al. A randomized comparison of cisplatin alone or in combination with methotrexate, vinblastine, and doxorubicin in patients with metastatic urothelial carcinoma: a cooperative group study. J Clin Oncol. (1992) 10:1066–73. C.OMMAS.R.X.X.X. doi: 10.1200/JCO.1992.10.7.1066

PubMed Abstract | Crossref Full Text | Google Scholar

4. Aggen DH and Drake CG. Biomarkers for immunotherapy in bladder cancer: a moving target. J ImmunoTherapy Cancer. (2017) 5:94. doi: 10.1186/s40425-017-0299-1

PubMed Abstract | Crossref Full Text | Google Scholar

5. Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee J-L, Fong L, et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. New Engl J Med. (2017) 376:1015–26. doi: 10.1056/NEJMoa1613683

PubMed Abstract | Crossref Full Text | Google Scholar

6. Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt J, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet. (2017) 389:67–76. doi: 10.1016/S0140-6736(16)32455-2

PubMed Abstract | Crossref Full Text | Google Scholar

7. Powles T, Park SH, Voog E, Caserta C, Valderrama BP, Gurney H, et al. Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. N Engl J Med. (2020) 383:1218–30. doi: 10.1056/NEJMoa2002788

PubMed Abstract | Crossref Full Text | Google Scholar

8. Heijden MSVD, Sonpavde G, Powles T, Necchi A, Burotto M, Schenker M, et al. Nivolumab plus gemcitabine–cisplatin in advanced urothelial carcinoma. New Engl J Med. (2023) 389:1778–89. doi: 10.1056/NEJMoa2309863

PubMed Abstract | Crossref Full Text | Google Scholar

9. Powles T, Bellmunt J, Comperat E, De Santis M, Huddart R, Loriot Y, et al. ESMO Clinical Practice Guideline interim update on first-line therapy in advanced urothelial carcinoma. Ann Oncol. (2024) 35:485–90. doi: 10.1016/j.annonc.2024.03.001

PubMed Abstract | Crossref Full Text | Google Scholar

10. Powles T, Valderrama BP, Gupta S, Bedke J, Kikuchi E, Hoffman-Censits J, et al. Enfortumab vedotin and pembrolizumab in untreated advanced urothelial cancer. N Engl J Med. (2024) 390:875–88. doi: 10.1056/NEJMoa2312117

PubMed Abstract | Crossref Full Text | Google Scholar

11. Meric-Bernstam F, Makker V, Oaknin A, Oh DY, Banerjee S, González-Martín A, et al. Efficacy and safety of trastuzumab deruxtecan in patients with HER2-expressing solid tumors: primary results from the DESTINY-panTumor02 phase II trial. J Clin Oncol. (2024) 42:47–58. doi: 10.1200/JCO.23.02005

PubMed Abstract | Crossref Full Text | Google Scholar

12. Collins DM, Bossenmaier B, Kollmorgen G, and Niederfellner G. Acquired resistance to antibody-drug conjugates. Cancers (Basel). (2019) 11:394. doi: 10.3390/cancers11030394

PubMed Abstract | Crossref Full Text | Google Scholar

13. Gandullo-Sanchez L, Ocana A, and Pandiella A. Generation of antibody-drug conjugate resistant models. Cancers (Basel). (2021) 13:4631. doi: 10.3390/cancers13184631

PubMed Abstract | Crossref Full Text | Google Scholar

14. di Martino E, Tomlinson DC, Williams SV, and Knowles MA. A place for precision medicine in bladder cancer: targeting the FGFRs. Future Oncol. (2016) 12:2243–63. doi: 10.2217/fon-2016-0042

PubMed Abstract | Crossref Full Text | Google Scholar

15. Necchi A, Lo Vullo S, Raggi D, Gloghini A, Giannatempo P, Colecchia M, et al. 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. (2019) 5:853–6. doi: 10.1016/j.euf.2018.02.013

PubMed Abstract | Crossref Full Text | Google Scholar

16. Challita-Eid PM, Satpayev D, Yang P, An Z, Morrison K, Shostak Y, et al. Enfortumab vedotin antibody-drug conjugate targeting nectin-4 is a highly potent therapeutic agent in multiple preclinical cancer models. Cancer Res. (2016) 76:3003–13. doi: 10.1158/0008-5472.CAN-15-1313

PubMed Abstract | Crossref Full Text | Google Scholar

17. Doronina SO, Toki BE, Torgov MY, Mendelsohn BA, Cerveny CG, Chace DF, et al. Development of potent monoclonal antibody auristatin conjugates for cancer therapy. Nat Biotechnol. (2003) 21:778–84. doi: 10.1038/nbt832

PubMed Abstract | Crossref Full Text | Google Scholar

18. Lattanzio R, Ghasemi R, Brancati F, Sorda RL, Tinari N, Perracchio L, et al. Membranous Nectin-4 expression is a risk factor for distant relapse of T1-T2, N0 luminal-A early breast cancer. Oncogenesis. (2014) 3:e118. doi: 10.1038/oncsis.2014.32

PubMed Abstract | Crossref Full Text | Google Scholar

19. Takai Y, Ikeda W, Ogita H, and Rikitake Y. The immunoglobulin-like cell adhesion molecule nectin and its associated protein afadin. Annu Rev Cell Dev Biol. (2008) 24:309–42. doi: 10.1146/annurev.cellbio.24.110707.175339

PubMed Abstract | Crossref Full Text | Google Scholar

20. Zhang Y, Zhang J, Shen Q, Yin W, Huang H, Liu Y, et al. High expression of Nectin-4 is associated with unfavorable prognosis in gastric cancer. Oncol Lett. (2018) 15:8789–95. doi: 10.3892/ol.2018.8365

PubMed Abstract | Crossref Full Text | Google Scholar

21. Rosenberg JE, O'Donnell PH, Balar AV, McGregor BA, Heath EI, Yu EY, et al. Pivotal trial of enfortumab vedotin in urothelial carcinoma after platinum and anti-programmed death 1/programmed death ligand 1 therapy. J Clin Oncol. (2019) 37:2592–600. doi: 10.1200/JCO.19.01140

PubMed Abstract | Crossref Full Text | Google Scholar

22. Yu EY, Petrylak DP, O'Donnell PH, Lee JL, van der Heijden MS, Loriot Y, 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. (2021) 22:872–82. doi: 10.1016/S1470-2045(21)00094-2

PubMed Abstract | Crossref Full Text | Google Scholar

23. Galsky MD, Hahn NM, Rosenberg JE, Sonpavde G, Oh WK, Dreicer R, et al. Defining “cisplatin ineligible” patients with metastatic bladder cancer. J Clin Oncol. (2011) 29:238–8. doi: 10.1200/jco.2011.29.7_suppl.238

Crossref Full Text | Google Scholar

24. Gupta S, Bedke J, Heijden MSVD, Valderrama BP, Kikuchi E, Hoffman-Censits J, et al. Exploratory analysis of responders from the phase 3 EV-302 trial of enfortumab vedotin plus pembrolizumab (EV+P) vs chemotherapy (chemo) in previously untreated locally advanced or metastatic urothelial carcinoma (la/mUC). J Clin Oncol. (2025) 43:4502–2. doi: 10.1200/JCO.2025.43.16_suppl.4502

Crossref Full Text | Google Scholar

25. Patelli G, Zeppellini A, Spina F, Righetti E, Stabile S, Amatu A, et al. The evolving panorama of HER2-targeted treatments in metastatic urothelial cancer: A systematic review and future perspectives. Cancer Treat Rev. (2022) 104:102351. doi: 10.1016/j.ctrv.2022.102351

PubMed Abstract | Crossref Full Text | Google Scholar

26. Yan M, Schwaederle M, Arguello D, Millis SZ, Gatalica Z, and Kurzrock R. HER2 expression status in diverse cancers: review of results from 37,992 patients. Cancer Metastasis Rev. (2015) 34:157–64. doi: 10.1007/s10555-015-9552-6

PubMed Abstract | Crossref Full Text | Google Scholar

27. Nakada T, Sugihara K, Jikoh T, Abe Y, and Agatsuma T. The latest research and development into the antibody-drug conjugate, [fam-] trastuzumab deruxtecan (DS-8201a), for HER2 cancer therapy. Chem Pharm Bull (Tokyo). (2019) 67:173–85. doi: 10.1248/cpb.c18-00744

PubMed Abstract | Crossref Full Text | Google Scholar

28. Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, et al. DS-8201a, A novel HER2-targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a promising antitumor efficacy with differentiation from T-DM1. Clin Cancer Res. (2016) 22:5097–108. doi: 10.1158/1078-0432.CCR-15-2822

PubMed Abstract | Crossref Full Text | Google Scholar

29. Hamilton E, Galsky MD, Ochsenreither S, Del Conte G, Martin M, De Miguel MJ, et al. Trastuzumab deruxtecan with nivolumab in HER2-expressing metastatic breast or urothelial cancer: analysis of the phase ib DS8201-A-U105 study. Clin Cancer Res. (2024) 30:5548–58. doi: 10.1158/1078-0432.CCR-24-1513

PubMed Abstract | Crossref Full Text | Google Scholar

30. Deeks ED. Disitamab vedotin: first approval. Drugs. (2021) 81:1929–35. doi: 10.1007/s40265-021-01614-x

PubMed Abstract | Crossref Full Text | Google Scholar

31. Sheng X, Wang L, He Z, Shi Y, Luo H, Han W, 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. (2024) 42:1391–402. doi: 10.1200/JCO.22.02912

PubMed Abstract | Crossref Full Text | Google Scholar

32. Sheng X, Yan X, Wang L, Shi Y, Yao X, Luo H, 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. (2021) 27:43–51. doi: 10.1158/1078-0432.CCR-20-2488

PubMed Abstract | Crossref Full Text | Google Scholar

33. Yan X, Li J, Xu H, Liu Y, Zhou L, Li S, 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. (2025) 6:100637. doi: 10.1016/j.medj.2025.100637

PubMed Abstract | Crossref Full Text | Google Scholar

34. Wang K, Xu T, Wu J, Yuan Y, Guan X, and Zhu C. Real-world application of disitamab vedotin (RC48-ADC) in patients with breast cancer with different HER2 expression levels: efficacy and safety analysis. Oncologist. (2024) 30:304. doi: 10.1093/oncolo/oyae304

PubMed Abstract | Crossref Full Text | Google Scholar

35. Wei Y, Zhang R, Yu C, Hong Z, Lin L, Li T, et al. Disitamab vedotin in combination with immune checkpoint inhibitors for locally and locally advanced bladder urothelial carcinoma: a two-center's real-world study. Front Pharmacol. (2023) 14:1230395. doi: 10.3389/fphar.2023.1230395

PubMed Abstract | Crossref Full Text | Google Scholar

36. Wen F, Lin T, Zhang P, and Shen Y. RC48-ADC combined with tislelizumab as neoadjuvant treatment in patients with HER2-positive locally advanced muscle-invasive urothelial bladder cancer: a multi-center phase Ib/II study (HOPE-03). Front Oncol. (2023) 13:1233196. doi: 10.3389/fonc.2023.1233196

PubMed Abstract | Crossref Full Text | Google Scholar

37. Galsky MD, Grande E, Necchi A, Drakaki A, Loriot Y, Franco S, et al. Phase 3 study of disitamab vedotin with pembrolizumab vs chemotherapy in patients with previously untreated locally advanced or metastatic urothelial carcinoma that expresses HER2 (DV-001). J Clin Oncol. (2024) 42:TPS4616–TPS4616. doi: 10.1200/JCO.2024.42.16_suppl.TPS4616

Crossref Full Text | Google Scholar

38. Mathijssen RH, van Alphen RJ, Verweij J, Loos WJ, Nooter K, Stoter G, et al. Clinical pharmacokinetics and metabolism of irinotecan (CPT-11). Clin Cancer Res. (2001) 7:2182–94. doi: 10.1200/JCO.2002.11.073

PubMed Abstract | Crossref Full Text | Google Scholar

39. Tagawa ST, Balar AV, Petrylak DP, Kalebasty AR, Loriot Y, Flechon A, et al. TROPHY-U-01: A phase II open-label study of sacituzumab govitecan in patients with metastatic urothelial carcinoma progressing after platinum-based chemotherapy and checkpoint inhibitors. J Clin Oncol. (2021) 39:2474–85. doi: 10.1200/JCO.20.03489

PubMed Abstract | Crossref Full Text | Google Scholar

40. Powles T, Tagawa S, Vulsteke C, Gross-Goupil M, Park SH, Necchi A, et al. Sacituzumab govitecan in advanced urothelial carcinoma: TROPiCS-04, a phase III randomized trial. Ann Oncol. (2025) 36:561–71. doi: 10.1016/j.annonc.2025.01.011

PubMed Abstract | Crossref Full Text | Google Scholar

41. Abbas M, Heitplatz B, Bernemann C, Boegemann M, Trautmann M, Schrader AJ, et al. Immunohistochemical expression of TROP−2 (TACSTD2) on the urothelial carcinoma of the urinary bladder and other types of cancer. Oncol Lett. (2023) 26:527. doi: 10.3892/ol.2023.14114

PubMed Abstract | Crossref Full Text | Google Scholar

42. Okajima D. YS, Yokouchi Y, Fujitani T, Sakurai K, Yamaguchi J, Kitamura M, et al. Preclinical efficacy studies of DS-1062a, a novel TROP2-targeting antibody-drug conjugate with a novel DNA topoisomerase I inhibitor DXd. J Clin Oncol. (2018) 36):e24206. doi: 10.1200/JCO.2018.36.15_suppl.e24206

Crossref Full Text | Google Scholar

43. Meric-Bernstam F, Alhalabi O, Lisberg A, Drakaki A, Garmezy B, Kogawa T, et al. Datopotamab deruxtecan (Dato-DXd) in locally advanced/metastatic urothelial cancer: Updated results from the phase 1 TROPIONPanTumor01 study. J Clin Oncol. (2025) 43:663–3. doi: 10.1200/JCO.2025.43.5_suppl.663

Crossref Full Text | Google Scholar

44. Heist RS, Sands J, Bardia A, Shimizu T, Lisberg A, Krop I, et al. Clinical management, monitoring, and prophylaxis of adverse events of special interest associated with datopotamab deruxtecan. Cancer Treat Rev. (2024) 125:102720. doi: 10.1016/j.ctrv.2024.102720

PubMed Abstract | Crossref Full Text | Google Scholar

45. Ye D, Jiang S, Yuan F, Zhou F, Jiang K, Zhang X, et al. Efficacy and safety of sacituzumab tirumotecan monotherapy in patients with advanced urothelial carcinoma who progressed on or after prior anti-cancer therapies: Report from the phase 1/2 MK-2870–001 study. J Clin Oncol. (2025) 43:796–6. doi: 10.1200/JCO.2025.43.5_suppl.796

Crossref Full Text | Google Scholar

46. Haugsten EM, Wiedlocha A, Olsnes S, and Wesche J. Roles of fibroblast growth factor receptors in carcinogenesis. Mol Cancer Res. (2010) 8:1439–52. doi: 10.1158/1541-7786.MCR-10-0168

PubMed Abstract | Crossref Full Text | Google Scholar

47. Robertson AG, Kim J, Al-Ahmadie H, Bellmunt J, Guo G, Cherniack AD, et al. Comprehensive molecular characterization of muscle-invasive bladder cancer. Cell. (2017) 171:540–556 e25. doi: 10.1016/j.cell.2017.09.007

PubMed Abstract | Crossref Full Text | Google Scholar

48. Perera TPS, Jovcheva E, Mevellec L, Vialard J, De Lange D, Verhulst T, et al. Discovery and pharmacological characterization of JNJ-42756493 (Erdafitinib), a functionally selective small-molecule FGFR family inhibitor. Mol Cancer Ther. (2017) 16:1010–20. doi: 10.1158/1535-7163.MCT-16-0589

PubMed Abstract | Crossref Full Text | Google Scholar

49. Loriot Y, Necchi A, Park SH, Garcia-Donas J, Huddart R, Burgess E, et al. Erdafitinib in locally advanced or metastatic urothelial carcinoma. N Engl J Med. (2019) 381:338–48. doi: 10.1056/NEJMoa1817323

PubMed Abstract | Crossref Full Text | Google Scholar

50. Loriot Y, Matsubara N, Park SH, Huddart RA, Burgess EF, Houede N, et al. Erdafitinib or chemotherapy in advanced or metastatic urothelial carcinoma. N Engl J Med. (2023) 389:1961–71. doi: 10.1056/NEJMoa2308849

PubMed Abstract | Crossref Full Text | Google Scholar

51. Siefker-Radtke AO, Matsubara N, Park SH, Huddart RA, Burgess EF, Ozguroglu M, 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. (2024) 35:107–17. doi: 10.1016/j.annonc.2023.10.003

PubMed Abstract | Crossref Full Text | Google Scholar

52. Lyou Y, Rosenberg JE, Hoffman-Censits J, Quinn DI, Petrylak D, Galsky M, et al. Infigratinib in early-line and salvage therapy for FGFR3-altered metastatic urothelial carcinoma. Clin Genitourin Cancer. (2022) 20:35–42. doi: 10.1016/j.clgc.2021.10.004

PubMed Abstract | Crossref Full Text | Google Scholar

53. Necchi A, Pouessel D, Leibowitz R, Gupta S, Fléchon A, García-Donas J, et al. Pemigatinib for metastatic or surgically unresectable urothelial carcinoma with FGF/FGFR genomic alterations: final results from FIGHT-201. Ann Oncol. (2024) 35:200–10. doi: 10.1016/j.annonc.2023.10.794

PubMed Abstract | Crossref Full Text | Google Scholar

54. Sternberg CN, Petrylak DP, Bellmunt J, Nishiyama H, Necchi A, Gurney H, 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. (2023) 41:629–39. doi: 10.1200/JCO.21.02303

PubMed Abstract | Crossref Full Text | Google Scholar

55. Iyer G, Ebi H, Cook N, Gao X, Kitano S, Matsubara N, et al. A first-in-human phase 1 study of LY3866288 (LOXO-435), a potent, highly isoform-selective FGFR3 inhibitor (FGFR3i) in advanced solid tumors with FGFR3 alterations: Initial results from FORAGER-1. J Clin Oncol. (2025) 43:662. doi: 10.1200/JCO.2025.43.5_suppl.662

Crossref Full Text | Google Scholar

56. Iyer G, Siefker-Radtke A, Milowsky M, Shore N, Gao X, Reimers MA, et al. Abstract CT119: A first-in-human phase 1 study of LOXO-435, a potent, highly isoform-selective FGFR3 inhibitor in advanced solid tumors with FGFR3 alterations (trial in progress). Cancer Res. (2023) 83:CT119–9. doi: 10.1158/1538-7445.AM2023-CT119

Crossref Full Text | Google Scholar

57. Loganzo F, Tan X, Sung M, Jin G, Myers JS, Melamud E, et al. Tumor cells chronically treated with a trastuzumab-maytansinoid antibody-drug conjugate develop varied resistance mechanisms but respond to alternate treatments. Mol Cancer Ther. (2015) 14:952–63. doi: 10.1158/1535-7163.MCT-14-0862

PubMed Abstract | Crossref Full Text | Google Scholar

58. Filho OM, Viale G, Stein S, Trippa L, Yardley DA, Mayer IA, et al. Impact of HER2 heterogeneity on treatment response of early-stage HER2-positive breast cancer: phase II neoadjuvant clinical trial of T-DM1 combined with pertuzumab. Cancer Discov. (2021) 11:2474–87. doi: 10.1158/2159-8290.CD-20-1557

PubMed Abstract | Crossref Full Text | Google Scholar

59. Gebhart G, Lamberts LE, Wimana Z, Garcia C, Emonts P, Ameye L, et al. Molecular imaging as a tool to investigate heterogeneity of advanced HER2-positive breast cancer and to predict patient outcome under trastuzumab emtansine (T-DM1): the ZEPHIR trial. Ann Oncol. (2016) 27:619–24. doi: 10.1093/annonc/mdv577

PubMed Abstract | Crossref Full Text | Google Scholar

60. Klumper N, Ralser DJ, Ellinger J, Roghmann F, Albrecht J, Below E, et al. Membranous NECTIN-4 expression frequently decreases during metastatic spread of urothelial carcinoma and is associated with enfortumab vedotin resistance. Clin Cancer Res. (2023) 29:1496–505. doi: 10.1158/1078-0432.CCR-22-1764

PubMed Abstract | Crossref Full Text | Google Scholar

61. Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PloS Med. (2005) 2:e73. doi: 10.1371/journal.pmed.0020073

PubMed Abstract | Crossref Full Text | Google Scholar

62. Patani H, Bunney TD, Thiyagarajan N, Norman RA, Ogg D, Breed J, et al. Landscape of activating cancer mutations in FGFR kinases and their differential responses to inhibitors in clinical use. Oncotarget. (2016) 7:24252–68. doi: 10.18632/oncotarget.8132

PubMed Abstract | Crossref Full Text | Google Scholar

63. Leyton JV. Improving receptor-mediated intracellular access and accumulation of antibody therapeutics-the tale of HER2. Antibodies (Basel). (2020) 9:32. doi: 10.3390/antib9030032

PubMed Abstract | Crossref Full Text | Google Scholar

64. Hammood M, Craig AW, and Leyton JV. Impact of endocytosis mechanisms for the receptors targeted by the currently approved antibody-drug conjugates (ADCs)-A necessity for future ADC research and development. Pharm (Basel). (2021) 14:674. doi: 10.3390/ph14070674

PubMed Abstract | Crossref Full Text | Google Scholar

65. Kalim M, Chen J, Wang S, Lin C, Ullah S, Liang K, et al. Intracellular trafficking of new anticancer therapeutics: antibody-drug conjugates. Drug Des Devel Ther. (2017) 11:2265–76. doi: 10.2147/DDDT.S135571

PubMed Abstract | Crossref Full Text | Google Scholar

66. Sung M, Tan X, Lu B, Golas J, Hosselet C, Wang F, et al. Caveolae-mediated endocytosis as a novel mechanism of resistance to trastuzumab emtansine (T-DM1). Mol Cancer Ther. (2018) 17:243–53. doi: 10.1158/1535-7163.MCT-17-0403

PubMed Abstract | Crossref Full Text | Google Scholar

67. Corbett S, Huang S, Zammarchi F, Howard PW, van Berkel PH, Hartley JA, et al. The role of specific ATP-binding cassette transporters in the acquired resistance to pyrrolobenzodiazepine dimer-containing antibody-drug conjugates. Mol Cancer Ther. (2020) 19:1856–65. doi: 10.1158/1535-7163.MCT-20-0222

PubMed Abstract | Crossref Full Text | Google Scholar

68. Takegawa N, Nonagase Y, Yonesaka K, Sakai K, Maenishi O, Ogitani Y, et al. DS-8201a, a new HER2-targeting antibody-drug conjugate incorporating a novel DNA topoisomerase I inhibitor, overcomes HER2-positive gastric cancer T-DM1 resistance. Int J Cancer. (2017) 141:1682–9. doi: 10.1002/ijc.30870

PubMed Abstract | Crossref Full Text | Google Scholar

69. Chang K, Lodha. R, Delavan HM, Winebaum J, Porten SP, Feng FY, et al. Mechanisms and strategies to overcome resistance to enfortumab vedotin in bladder cancer. J Clin Oncol. (2024) 42. doi: 10.1200/JCO.2024.42.4_suppl.690

Crossref Full Text | Google Scholar

70. Berns K, Horlings HM, Hennessy BT, Madiredjo M, Hijmans EM, Beelen K, et al. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell. (2007) 12:395–402. doi: 10.1016/j.ccr.2007.08.030

PubMed Abstract | Crossref Full Text | Google Scholar

71. Jain S, Shah AN, Santa-Maria CA, Siziopikou K, Rademaker A, Helenowski I, et al. Phase I study of alpelisib (BYL-719) and trastuzumab emtansine (T-DM1) in HER2-positive metastatic breast cancer (MBC) after trastuzumab and taxane therapy. Breast Cancer Res Treat. (2018) 171:371–81. doi: 10.1007/s10549-018-4792-0

PubMed Abstract | Crossref Full Text | Google Scholar

72. D'Angelo A, Chapman R, Sirico M, Sobhani N, Catalano M, Mini E, et al. An update on antibody-drug conjugates in urothelial carcinoma: state of the art strategies and what comes next. Cancer Chemother Pharmacol. (2022) 90:191–205. doi: 10.1007/s00280-022-04459-7

PubMed Abstract | Crossref Full Text | Google Scholar

73. Hosni S, Kilian V, Klumper N, Gabbia D, Sieckmann K, Corvino D, et al. Adipocyte precursor-derived NRG1 promotes resistance to FGFR inhibition in urothelial carcinoma. Cancer Res. (2024) 84:725–40. doi: 10.1158/0008-5472.CAN-23-1398

PubMed Abstract | Crossref Full Text | Google Scholar

74. Weickhardt AJ, Lau DK, Hodgson-Garms M, Lavis A, Jenkins LJ, Vukelic N, et al. Dual targeting of FGFR3 and ERBB3 enhances the efficacy of FGFR inhibitors in FGFR3 fusion-driven bladder cancer. BMC Cancer. (2022) 22:478. doi: 10.1186/s12885-022-09478-4

PubMed Abstract | Crossref Full Text | Google Scholar

75. Lv Y, Cui X, Li T, Liu C, Wang A, Wang T, et al. Mechanism of action and future perspectives of ADCs in combination with immune checkpoint inhibitors for solid tumors. Clin Exp Med. (2025) 25:139. doi: 10.1007/s10238-025-01655-6

PubMed Abstract | Crossref Full Text | Google Scholar

76. Saini KS, Punie K, Twelves C, Bortini S, de Azambuja E, Anderson S, et al. Antibody-drug conjugates, immune-checkpoint inhibitors, and their combination in breast cancer therapeutics. Expert Opin Biol Ther. (2021) 21:945–62. doi: 10.1080/14712598.2021.1936494

PubMed Abstract | Crossref Full Text | Google Scholar

77. Bauzon M, Drake PM, Barfield RM, Cornali BM, Rupniewski I, and Rabuka D. Maytansine-bearing antibody-drug conjugates induce in vitro hallmarks of immunogenic cell death selectively in antigen-positive target cells. Oncoimmunology. (2019) 8:e1565859. doi: 10.1080/2162402X.2019.1565859

PubMed Abstract | Crossref Full Text | Google Scholar

78. Rios-Doria J, Harper J, Rothstein R, Wetzel L, Chesebrough J, Marrero A, et al. Antibody-drug conjugates bearing pyrrolobenzodiazepine or tubulysin payloads are immunomodulatory and synergize with multiple immunotherapies. Cancer Res. (2017) 77:2686–98. doi: 10.1158/0008-5472.CAN-16-2854

PubMed Abstract | Crossref Full Text | Google Scholar

79. Ouyang Y, Ou Z, Zhong W, Yang J, Fu S, Ouyang N, et al. FGFR3 alterations in bladder cancer stimulate serine synthesis to induce immune-inert macrophages that suppress T-cell recruitment and activation. Cancer Res. (2023) 83:4030–46. doi: 10.1158/0008-5472.CAN-23-1065

PubMed Abstract | Crossref Full Text | Google Scholar

80. O'Donnell PH, Milowsky MI, Petrylak DP, Hoimes CJ, Flaig TW, Mar N, et al. Enfortumab vedotin with or without pembrolizumab in cisplatin-ineligible patients with previously untreated locally advanced or metastatic urothelial cancer. J Clin Oncol. (2023) 41:4107–17. doi: 10.1200/JCO.22.02887

PubMed Abstract | Crossref Full Text | Google Scholar

81. Flaig TW, Spiess PE, Abern M, Agarwal N, Bangs R, Buyyounouski MK, et al. NCCN guidelines(R) insights: bladder cancer, version 3.2024. J Natl Compr Canc Netw. (2024) 22:216–25. doi: 10.6004/jnccn.2024.0024

PubMed Abstract | Crossref Full Text | Google Scholar

82. Hoffman-Censits J, Tsiatas M, Chang P, Kim M, Antonuzzo L, Shin SJJ, et al. Avelumab + sacituzumab govitecan (SG) vs avelumab monotherapy as first-line (1L) maintenance treatment in patients (pts) with advanced urothelial carcinoma (aUC): Interim analysis from the JAVELIN Bladder Medley phase 2 trial. J Clin Oncol. (2025) 2025. 43:4501–1. doi: 10.1016/j.annonc.2025.05.010

PubMed Abstract | Crossref Full Text | Google Scholar

83. Kydd AR, Chandran E, Simon NI, Atiq SO, Wang T-F, Cordes LM, et al. SMART: A phase II study of sacituzumab govitecan (SG) with or without atezolizumab immunotherapy in rare genitourinary (GU) tumors such as small cell, adenocarcinoma, and squamous cell bladder/urinary tract cancer, renal medullary carcinoma (RMC) and penile cancer. J Clin Oncol. (2024) 42:TPS4627–TPS4627. doi: 10.1200/JCO.2024.42.16_suppl.TPS4627

Crossref Full Text | Google Scholar

84. McGregor BA, Sonpavde GP, Kwak L, Regan MM, Gao X, Hvidsten H, et al. The Double Antibody Drug Conjugate (DAD) phase I trial: sacituzumab govitecan plus enfortumab vedotin for metastatic urothelial carcinoma. Ann Oncol. (2024) 35:91–7. doi: 10.1016/j.annonc.2023.09.3114

PubMed Abstract | Crossref Full Text | Google Scholar

85. McGregor BA, Kwak L, Sonpavde GP, Berg SA, Choueiri TK, Bellmunt J, et al. Sacituzumab govitecan (SG) plus enfortumab vedotin (EV) for metastatic urothelial carcinoma (mUC) treatment-experienced (DAD) and with pembrolizumab (P) in treatment naïve UC (DAD-IO). J Clin Oncol. (2024) 42:TPS4618–TPS4618. doi: 10.1200/JCO.2024.42.16_suppl.TPS4618

Crossref Full Text | Google Scholar

86. Powles T, Carroll D, Chowdhury S, Gravis G, Joly F, Carles J, et al. An adaptive, biomarker-directed platform study of durvalumab in combination with targeted therapies in advanced urothelial cancer. Nat Med. (2021) 27:793–801. doi: 10.1038/s41591-021-01317-6

PubMed Abstract | Crossref Full Text | Google Scholar

87. Siefker-Radtke A, Powles T, Moreno V, Won Kang T, Cicin I, Girvin A, et al. Erdafitinib (ERDA) vs ERDA plus cetrelimab (ERDA+CET) for patients (pts) with metastatic urothelial carcinoma (mUC) and fibroblast growth factor receptor alterations (FGFRa): Final results from the phase 2 Norse study. J Clin Oncol. (2023) 41:4504–4504. doi: 10.1200/JCO.2023.41.16_suppl.4504

Crossref Full Text | Google Scholar

88. Zhou Y, Wu C, Lu G, Hu Z, Chen Q, and Du X. FGF/FGFR signaling pathway involved resistance in various cancer types. J Cancer. (2020) 11:2000–7. doi: 10.7150/jca.40531

PubMed Abstract | Crossref Full Text | Google Scholar

89. 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. (2024) 21:312–29. doi: 10.1038/s41571-024-00869-z

PubMed Abstract | Crossref Full Text | Google Scholar

90. Packer LM, Geng X, Bonazzi VF, Ju RJ, Mahon CE, Cummings MC, et al. PI3K inhibitors synergize with FGFR inhibitors to enhance antitumor responses in FGFR2(mutant) endometrial cancers. Mol Cancer Ther. (2017) 16:637–48. doi: 10.1158/1535-7163.MCT-16-0415

PubMed Abstract | Crossref Full Text | Google Scholar

91. Chen M, Chen B, Ge X, Ma Q, and Gao S. Targeted nanodrugs to destroy the tumor extracellular matrix barrier for improving drug delivery and cancer therapeutic efficacy. Mol Pharm. (2023) 20:2389–401. doi: 10.1021/acs.molpharmaceut.2c00947

PubMed Abstract | Crossref Full Text | Google Scholar

92. Huang J, Zhang L, Wan D, Zhou L, Zheng S, Lin S, et al. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct Target Ther. (2021) 6:153. doi: 10.1038/s41392-021-00544-0

PubMed Abstract | Crossref Full Text | Google Scholar

93. Abraham J, Edgerly M, Wilson R, Chen C, Rutt A, Bakke S, et al. A phase I study of the P-glycoprotein antagonist tariquidar in combination with vinorelbine. Clin Cancer Res. (2009) 15:3574–82. doi: 10.1158/1078-0432.CCR-08-0938

PubMed Abstract | Crossref Full Text | Google Scholar

94. Fox E, Widemann BC, Pastakia D, Chen CC, Yang SX, Cole D, et al. Pharmacokinetic and pharmacodynamic study of tariquidar (XR9576), a P-glycoprotein inhibitor, in combination with doxorubicin, vinorelbine, or docetaxel in children and adolescents with refractory solid tumors. Cancer Chemother Pharmacol. (2015) 76:1273–83. doi: 10.1007/s00280-015-2845-1

PubMed Abstract | Crossref Full Text | Google Scholar

95. Cabaud O, Berger L, Crompot E, Adelaide J, Finetti P, Garnier S, et al. Overcoming resistance to anti-nectin-4 antibody-drug conjugate. Mol Cancer Ther. (2022) 21:1227–35. doi: 10.1158/1535-7163.MCT-22-0013

PubMed Abstract | Crossref Full Text | Google Scholar

96. Li JY, Perry SR, Muniz-Medina V, Wang X, Wetzel LK, Rebelatto MC, et al. A biparatopic HER2-targeting antibody-drug conjugate induces tumor regression in primary models refractory to or ineligible for HER2-targeted therapy. Cancer Cell. (2019) 35:948–9. doi: 10.1016/j.ccell.2019.05.010

Crossref Full Text | Google Scholar

97. Jhaveri K, Han H, Dotan E, and Bedard P. Preliminary results from a phase I study using the bispecific, human epidermal growth factor 2 (HER2)-targeting antibody-drug conjugate (ADC) zanidatamab zovodotin (ZW49) in solid cancers. Ann Oncol. (2022) 33:S749–S750. doi: 10.1016/j.annonc.2022.07.589

Crossref Full Text | Google Scholar

98. Wang W, Jing L, Xu M, and Qun Y. VBC103: An innovative Trop2/Nectin4 targeted bispecific antibody drug conjugate (ADC) in bladder urothelial carcinoma (UC), triple-negative breast cancer (TNBC) and beyond. Cancer Res. (2024) 84. doi: 10.1158/1538-7445.AM2024-LB448

Crossref Full Text | Google Scholar

99. Loriot Y, Siefker-Radtke AO, Friedlander TW, Necchi A, Wei AZ, Sridhar SS, et al. A phase 2/3 study of bicycle toxin conjugate zelenectide pevedotin (BT8009) targeting Nectin-4 in patients with locally advanced or metastatic urothelial cancer (la/mUC) (Duravelo-2). J Clin Oncol. (2025) 43:TPS898–8. doi: 10.1200/JCO.2025.43.5_suppl.TPS898

Crossref Full Text | Google Scholar

100. Peng M, Chu X, Peng Y, Li D, Zhang Z, Wang W, et al. Targeted therapies in bladder cancer: signaling pathways, applications, and challenges. MedComm. (2020) 4:e455. doi: 10.1002/mco2.455

PubMed Abstract | Crossref Full Text | Google Scholar

101. Li F, Emmerton KK, Jonas M, Zhang X, Miyamoto JB, Setter JR, et al. Intracellular released payload influences potency and bystander-killing effects of antibody-drug conjugates in preclinical models. Cancer Res. (2016) 76:2710–9. doi: 10.1158/0008-5472.CAN-15-1795

PubMed Abstract | Crossref Full Text | Google Scholar

102. Dan N, Setua S, Kashyap VK, Khan S, Jaggi M, Yallapu MM, et al. Antibody-drug conjugates for cancer therapy: chemistry to clinical implications. Pharm (Basel). (2018) 11:32. doi: 10.3390/ph11020032

PubMed Abstract | Crossref Full Text | Google Scholar

103. Kwon WA, Lee SY, Jeong TY, Kim HH, and Lee MK. Antibody-drug conjugates in urothelial cancer: from scientific rationale to clinical development. Cancers (Basel). (2024) 16:2420. doi: 10.3390/cancers16132420

PubMed Abstract | Crossref Full Text | Google Scholar

104. Yamazaki CM, Yamaguchi A, Anami Y, Xiong W, Otani Y, Lee J, et al. Antibody-drug conjugates with dual payloads for combating breast tumor heterogeneity and drug resistance. Nat Commun. (2021) 12:3528. doi: 10.1038/s41467-021-23793-7

PubMed Abstract | Crossref Full Text | Google Scholar

105. Meric-Bernstam F, Bahleda R, Hierro C, Sanson M, Bridgewater J, Arkenau HT, et al. Futibatinib, an irreversible FGFR1–4 inhibitor, in patients with advanced solid tumors harboring FGF/FGFR aberrations: A phase I dose-expansion study. Cancer Discov. (2022) 12:402–15. doi: 10.1158/2159-8290.CD-21-0697

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: advanced urothelial carcinoma, antibody-drug conjugates, FGFR3 inhibition, drug resistance, immunotherapy combined therapy

Citation: Wummer B, Schwartz M, Ciuro J, Ahmed S, Joshi SS, Narayan VM, Carthon BC, Bilen MA and Brown JT (2025) Overcoming resistance in advanced urothelial carcinoma: mechanisms of escape from antibody-drug conjugates and FGFR3 inhibition. Front. Oncol. 15:1654771. doi: 10.3389/fonc.2025.1654771

Received: 26 June 2025; Accepted: 27 October 2025;
Published: 02 December 2025.

Edited by:

Benyi Li, University of Kansas Medical Center, United States

Reviewed by:

Jayaprakash N. Kolla, Institute of Molecular Genetics (ASCR), Czechia
Xiangwei Wang, Guangdong Medical University, China

Copyright © 2025 Wummer, Schwartz, Ciuro, Ahmed, Joshi, Narayan, Carthon, Bilen and Brown. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jacqueline T. Brown, anRicm93N0BlbW9yeS5lZHU=

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.