Advancing therapeutic frontiers in urothelial carcinoma: targeted strategies and clinical implications
Introduction
Background
Advanced urothelial carcinoma (aUC) presents a formidable challenge in the medical landscape due to its aggressive nature and widespread prevalence. For decades, platinum-based chemotherapy has served as the primary treatment modality for aUC, achieving a median overall survival (mOS) of 14–15 months (1). However, recent advances in the molecular understanding of urothelial carcinoma and the development of new targeted therapies have changed this scenario and allowed the development of new molecules to treat this disease.
Rationale and knowledge gap
Initially, immune checkpoint inhibitors (ICIs) have exhibited efficacy as monotherapy in patients who have progressed on platinum-based chemotherapy (2). Additionally, targeted therapy with antibody-drug conjugates (ADCs) and tyrosine kinase inhibitors (TKIs) have shown efficacy in multiple trials for aUC post platinum-based chemotherapy (3). The success of these novel treatment options in pre-treated patients has spurred the development of clinical trials evaluating these drugs, alone or in combination, in the frontline setting. Initial evaluations of ICIs against first-line platinum-based chemotherapy, as monotherapy or in combination with chemotherapy, yielded disappointing results in phase 3 trials such as Keynote 361 (4), Imvigor 130 (5), and Danube (6). Conversely, ICIs used as a maintenance strategy after 4–6 cycles of platinum-based chemotherapy for patients who did not progress during chemotherapy demonstrated an OS gain in the Javelin 100 phase 3 trial, with a mOS of 21.4 months for maintenance avelumab vs. 14.3 months for the control arm [hazard ratio (HR) =0.69; P=0.001] (7). Recently, the CheckMate 901 trial demonstrated an OS gain for the combination of nivolumab with cisplatin and gencitabine in untreated aUC patients eligible for receiving cisplatin, with a mOS of 21.7 months for the combination compared with 18.9 months for the control arm (HR =0.78; P=0.02) (8).
ADCs and TKIs are also being evaluated in combination with ICIs, with the combination of enfortumab vedotin (EV) with pembrolizumab, assessed in the large phase trial EV 302 (9), demonstrating impressive and unprecedented results in aUC. The mOS was 31.5 months for the EV-combination arm compared to 16.1 months for the control arm composed of platinum-based chemotherapy (HR =0.47; P<0.001) (9). Despite these significant advances, we still have a lot to learn. For example: how to best select patients for these treatments and how to further improve the effectiveness.
Objective
This article aims to delve into this novel targeted therapy, particularly ADCs and novel TKIs, and review the latest data on these treatment strategies in aUC.
Biomarkers for targeted therapies
The predictive value of biomarkers in aUC remains modest, with only a few biomarkers approved for clinical use, such as fibroblast growth factor receptor (FGFR) (10). Several biomarkers are under review in urothelial cancers, including human epidermal growth factor receptor 2 (HER2), DNA damage response (DDR) proteins, and poly(ADP-ribose) polymerase (PARP), with potential to guide therapies like PARP inhibitors (iPARPs), HER2 inhibitors, and ADCs. However, none of these biomarkers have undergone prognostic validation to date.
The development of molecular and genetic markers is a crucial area that can enhance clinical decision-making. Promising biomarkers serving as predictive and/or prognostic tools include FGFR mutations, epidermal growth factor receptor (EGFR) expression or mutations, HER2 expression, programmed death ligand-1 (PD-L1), tumor mutational burden (TMB), excision repair cross-complementing protein 1 and 2 (ERCC1 and ERCC2), PARP expression, DNA repair mutations, and circulating tumor DNA (ctDNA). FGFR 2/3 alterations have shown to be a valuable predictive biomarker, as demonstrated in the phase III THOR trial, indicating superior survival outcomes, median progression-free survival (mPFS) of 5.6 vs. 2.7 months and mOS of 12.1 vs. 7.8 months, and higher response rates for erdafitinib compared to single-agent chemotherapy (11). Increased expression of phosphorylated epidermal growth factor receptor 3 (pERBB3) appears to be a critical adaptive resistance mechanism to FGFR inhibitors. Combination therapy with a pan-ERBB inhibitor and an FGFR inhibitor may potentially overcome this resistance (12).
The human epidermal growth factor receptor (HER) family plays a significant role in carcinogenesis, cell proliferation, invasion, and metastasis (13). Both HER1/EGFR and HER2 (ErbB-2) are potential targets, with HER2 amplification being linked to unfavorable outcomes in aUC (14). The phase II evaluation of the EGFR-targeted agent cetuximab yielded disappointing results (15), while single-agent monoclonal antibodies or oral small-molecule inhibitors targeting HER2 have also shown limited clinical benefit (16,17). However, anti-HER2/3 ADCs present a promising avenue, with overall response rates reaching as high as 51% in aUC (18).
PD-L1 is a predictive biomarker for immunotherapy response in various cancer types; however, its predictive value in aUC appears inconsistent, with conflicting data from different studies. For instance, the phase II Imvigor210 trial demonstrated similar response rates between PD-L1(+) and PD-L1(−) subgroups when treated with atezolizumab (19). The phase III JAVELIN Bladder 100 trial revealed a 1-year survival enhancement with avelumab maintenance therapy (71.3% vs. 58.4%) irrespective of PD-L1 expression. While the overall benefit appears more pronounced in the PD-L1-positive group, it remains significant in both populations (7).
Urothelial carcinoma exhibits a notably high TMB (20), which consistently predicts response to ICIs in several aUC trials. For instance, in the CheckMate 275 study, nivolumab demonstrated improved PFS, OS, and objective response rate (ORR) in high-TMB patients compared to the overall study population (21). Similar associations between higher TMB and increased response rate were observed in the IMvigor 210 and PURE-01 trials evaluating atezolizumab and pembrolizumab, respectively (22,23).
Biomarkers associated with DDR pathways and PARP expression or activity hold potential as prognostic and predictive markers, although their value has not been consistently demonstrated. Reduced DNA repair capacity due to mutations or inactivation in these repair mechanisms has been linked to heightened sensitivity to cisplatin-based chemotherapy and iPARPs (24-26).
ADCs
The ideal systemic anti-cancer treatment should maximize its effect on malignant cells while sparing healthy cells from off-target toxicity, a concept envisioned by Paul Ehrlich in 1907 (27). Traditional chemotherapy and targeted therapies, such as TKIs, have yet to achieve the “magic bullet” ideal. To address this challenge, ADCs were developed to deliver therapeutic agents specifically to tumor cells expressing certain antigens.
ADCs comprise four components: a target antigen, a monoclonal antibody, a linker molecule binding the payload to the antibody, and the cytotoxic payload. Upon antibody binding and internalization via endocytosis, the payload is released in a bioactive form (28). Various antigen targets have been identified in urothelial carcinoma, prompting exploration of ADCs in aUC patients. Table 1 summarizes the results of clinical trials evaluating ADCs for aUC patients.
Table 1
Study [year] | Phase | N | Population | Drug | Median OS | Median PFS | Objective response rate | Median DOR | Target expression |
---|---|---|---|---|---|---|---|---|---|
Rosenberg et al., EV-101 [2020] (29) | I | 155 patients with mUC | Nectin-4-expressing solid tumors including mUC with ≥1 prior chemotherapy regimen and/or PD-(L)1 inhibitor or cisplatin ineligible | EV | 12.3 mo (95% CI: 9.3–15.3), OS rate at 1 year was 51.8% | 5.4 mo (95% CI: 5.1–6.3) | 43% (95% CI: 33.6–52.6%)—data regarding patients treated with EV 1.25 mg/kg | 7.4 mo (95% CI: 5.6–9.6) | IHC H-score of Nectin-4 expression (median H-score of 290) |
Rosenberg et al., EV-201 Cohort 1 [2019] (30) | II | 125 | Locally advanced or mUC previously treated with platinum chemotherapy and PD-1 or PD-L1 inhibitors | EV | 11.7 mo (95% CI: 9.1–not reached) | 5.8 mo (95% CI: 4.9–7.5) | 44% (95% CI: 35.1–46.2%) | 7.6 mo (95% CI: 4.93–7.46) | Patients enrolled regardless of target expression |
Rosenberg et al., EV-201 Cohort 2 [2019] (30) | II | 91 | Cisplatin-ineligible, locally advanced or mUC previously treated with PD-1 or PD-L1 inhibitors | EV | 14.7 mo (95% CI: 10.51–18.20) | 5.8 mo (95% CI: 5.03–8.28) | 52% (95% CI: 41–62%) | 10.9 mo (95% CI: 5.78–not reached) | Patients enrolled regardless of target expression |
Rosenberg et al., EV-301 long term outcomes [2023] (31) | III | 608 (301 EV, 307 chemotherapy) | Locally advanced or mUC with prior platinum chemotherapy and PD-1/L1 inhibitor treatment | EV vs. chemotherapy (docetaxel, paclitaxel, or vinflunine) | 12.91 mo for EV vs. 8.94 mo for chemotherapy (HR =0.70; 95% CI: 0.58–0.85; P<0.001) | 5.55 mo for EV vs. 3.71 mo for chemotherapy (HR =0.62; 95% CI: 0.51–0.75; P<0.001) | 40.6% for EV vs. 17.9% for chemotherapy (P<0.001) | 7.62 mo (95% CI: 5.66–11.17) for EV vs. 8.21 mo (95% CI: 5.68–9.56) for chemotherapy | Patients enrolled regardless of target expression |
O’Donnell et al., EV-103 Cohort K [2023] (32) | Ib/II | 151 (77 EV + pembrolizumab and 74 EV monotherapy) | Previously untreated, cisplatin-ineligible, locally advanced or mUC | EV + pembrolizumab or EV alone | 22.3 mo (95% CI: 9.09–not reached) for EV + pembrolizumab and 21.7 mo (95% CI: 5.21–not reached) for EV alone | Not reached (95% CI: 8.31–not reached) for EV + pembrolizumab and 8 mo (95% CI: 6.05–10.35) for EV alone | 64.5% (95% CI: 52.7–75.1%) for EV + pembrolizumab and 45.2% (95% CI: 33.5–57.3%) for EV alone | Not reached (95% CI: 0.25–not reached) for EV + pembrolizumab and 13.2 mo (95% CI: 6.14–15.97) for EV alone | Patients enrolled regardless of target expression |
Powles et al., EV-302/KEYNOTE-A39 [2023] (33) | III | 886 (442 EV + pembrolizumab, 444 chemotherapy) | Previously untreated locally advanced or mUC eligible for cisplatin- or carboplatin-containing chemotherapy | EV + pembrolizumab vs. gemcitabine with cisplatin or carboplatin | 31.5 mo for EV + pembrolizumab vs. 16.1 mo for chemotherapy (HR =0.47; 95% CI: 0.38–0.58; P<0.001) | 12.5 mo for EV + pembrolizumab vs. 6.3 mo for chemotherapy (HR =0.45; 95% CI: 0.38–0.54; P<0.001) | 67.7% EV + pembrolizumab and 44.4% chemotherapy (P<0.001) | Not reported | Patients enrolled regardless of target expression |
Meric-Bernstam et al., DESTINY-PanTumor02 [2024] (34) | II | 268 (41 with bladder cancer) | HER2-expressing locally advanced or metastatic cancers after ≥1 systemic treatment or without alternative treatments | Trastuzumab-deruxtecan | 21.1 mo (95% CI: 15.3–29.6), 12.8 mo (95% CI: 11.2–15.1) for bladder cancer | 6.9 mo (95% CI: 5.6–8), 7 mo (95% CI: 4.2–9.7) for bladder cancer | 37.1% (95% CI: 31.3–43.2%), 39% (95% CI: 24.2–55.5%) for bladder cancer | 11.3 mo (95% CI: 9.6–17.8) | HER2 expression by IHC classified as 3+ or 2+ |
Galsky et al., [2022] (35) | Ib/II | 34 | HER2 low and HER2 positive locally advanced or mUC with prior platinum-based chemotherapy | Trastuzumab deruxtecan + nivolumab | 11 mo (95% CI: 7.2–NE) | 6.9 mo (95% CI: 2.7–14.4) | 36.7% (95% CI: 19.9–56.1%) | Not reported | HER2 expression by IHC classified as 3+, 2+ or 1+ |
Loriot et al., TROPHY-U-01 Cohort 1 [2024] (36) | II | 113 | Patients with locally advanced or mUC with disease progression after platinum-based chemotherapy and CPIs | Sacituzumab govitecan | 10.9 mo (95% CI: 9.0–13.8) | 5.4 mo (95% CI: 3.5–7.2) | 27% (95% CI: 19.5–36.6%) | 7.2 mo (95% CI: 4.7–8.6) | Patients enrolled regardless of target expression |
Grivas et al., TROPHY-U-01 Cohort 3 [2024] (37) | II | 41 | mUC with prior after platinum chemotherapy-I-naive | Sacituzumab govitecan + pembrolizumab | 12.7 mo (95% CI: 10.7–not estimable) | 5.3 mo (95% CI: 3.4–10.2) | 41% (95% CI: 26.3–57.9%) | 11.1 mo (95% CI: 4.8–not estimable) | Patients enrolled regardless of target expression |
de Vries et al., KAMELEON [2023] (38) | II | 20 (13 mUC and 7 with locally advanced or metastatic pancreatic cancer or cholangiocarcinoma) | HER2-positive locally advanced or mUC, pancreatic cancer or cholangiocarcinoma | T-DM1 | 7.03 mo (95% CI: 3.75–not evaluable) for UC | 2.20 mo (95% CI: 1.18–4.30) for UC | 38.5% (90% CI: 16.57–64.52%) for UC | 3.3 mo (95% CI: 2.83–5.52) for UC | HER2 expression by IHC in ≥30% of stained cells |
Sheng et al., pooled analysis of RC48-C005 and RC48-009 trials [2024] (39) | II | 107 | HER2-positive locally advanced or mUC after ≥1 systemic chemotherapy | Disitamab vedotin | 5.9 mo (95% CI: 4.3–7.2) | 14.2 mo (95% CI: 9.7–18.8) | 50.5% (95% CI: 40.6–60.3%) | 7.3 mo (95% CI: 5.7–10.8) | HER2 expression by IHC classified as 3+ or 2+ |
Petrylak et al., [2024] (40) | I | 93 | mUC with or without prior chemotherapy or CPI therapy | Sirtratumab vedotin | Not reported | 16 weeks (95% CI: 13.71–23.14) | 18.3% (95% CI: 11–27.6%) | 24.71 weeks (95% CI: 16–nor estimable) | Patients enrolled regardless of target expression |
Xu et al., [2022] (41) | II | 19 | mUC, HER2 negative ≥1 systemic therapy | Disitamab vedotin | 16.4 mo (95% CI: 7.1–21.7) | 5.5 mo (95% CI: 3.9–6.8) | 26.3% (95% CI: 9.1–51.2%) | Not reported | HER2 expression by IHC classified as 0 or 1+ |
Sheng et al., [2023] (42) | Ib/II | 41 | Locally advanced or mUC with or without prior systemic therapy | Disitamab vedotin + toripalimab | Not reported (2-year OS rate of 63.2%) | 9.2 mo (95% CI: 5.7–10.3) | 73.2% (95% CI: 57.1–85.8%) | Not reported | Patients enrolled regardless of target expression |
McGregor et al., DAD trial [2024] (43) | I | 24 | Patients with locally advanced or mUC with disease progression after platinum-based chemotherapy and/or CPIs | Sacituzumab govitecan + EV | Not reported (immature data), 12 mo OS: 86% (95% CI: 61–95%) | Not reported (immature data), 12 mo PFS: 41% (95% CI: 18–62%) | 70% (95% CI: 47–87%) | Not reported | Patients enrolled regardless of target expression |
ADCs, antibody-drug conjugates; mUC, metastatic urothelial carcinoma; OS, overall survival; PFS, progression-free survival; DOR, duration of response; PD-(L)1, programmed death (ligand)-1; EV, enfortumab vedotin; mo, months; CI, confidence interval; IHC, immunohistochemistry; HR, hazard ratio; HER2, human epidermal growth factor receptor 2; NE, not evaluable; CPIs, checkpoint inhibitors; T-DM1, trastuzumab emtansine; UC, urothelial carcinoma.
Targeting Nectin-4
EV is an ADC consisting of monomethyl auristatin E (MMAE), which destabilizes microtubule formation, linked to a protease-cleavable linker and a fully human IgG1 kappa antibody targeting the junction protein Nectin-4 (44). Poliovirus receptor-related protein 4 (PVRL4), also known as Nectin-4, is a type I transmembrane polypeptide and a member of the immunoglobulin-like nectin family. While functioning as an adhesion molecule in normal cells, it exhibits low expression in normal tissues but is highly expressed in certain tumors. In bladder, for instance, we have data showing overall positive expression rates of 83% (45). This elevated expression makes Nectin-4 a promising therapeutic target in urothelial carcinoma.
In the phase I study EV-101, EV demonstrated an ORR of 43% in heavily pretreated patients with aUC (29). To further evaluate its efficacy, the phase II trial EV-201 was conducted, comprising two distinct cohorts. Cohort 1 included 125 patients with aUC who had experienced disease progression or recurrence after platinum-based chemotherapy or immunotherapy. In this cohort, an ORR of 44% was observed, with 15 complete responses (12%) (30). For context, single-agent chemotherapy following platinum and ICI progression typically yields lower response rates, approximately around 14% (46). Based on the results from cohort 1 of the EV-201 trial, EV received accelerated approval from the United States Food and Drug Administration (FDA) in 2019 for progressive aUC refractory to platinum-based chemotherapy and PD-1/PD-L1 inhibitors.
The phase III trial EV-301 compared EV, dose of 1.25 mg/kg on days 1, 8, and 15 of each 28-day, with physician’s choice chemotherapy (either taxmen or vinflunine) in 608 patients with aUC who progressed after platinum-based chemotherapy and ICIs. EV demonstrated a significant improvement in mOS by 3.91 months (12.88 vs. 8.97 months, HR =0.70; P=0.001) and longer PFS with 5.55 vs. 3.71 months (HR =0.62; P<0.001) (44). Long-term outcomes confirmed EV’s efficacy, leading to FDA approval for aUC in patients who progressed after first-line systemic treatment (31).
In the first-line setting, EV alone or with pembrolizumab was evaluated in Cohort K of the phase Ib/II study EV-103 for cisplatin-ineligible patients. The trial randomly assigned 151 patients in a 1:1 ratio to two arms without performing any statistical comparison between them. EV plus pembrolizumab demonstrated an ORR of 64.5% [95% confidence interval (CI): 52.7–75.1%], with mOS of 22.3 months (95% CI: 19.09–not reached), while EV monotherapy showed an ORR of 45.2% (95% CI: 33.5–57.33%) (32). The FDA granted accelerated approval for the combination therapy.
The phase III trial EV-302/KEYNOTE-A39 further assessed the synergistic effect of EV and ICIs. This study compared the combination of EV plus pembrolizumab with platinum-based chemotherapy (gemcitabine associated with cisplatin or carboplatin) in previously untreated patients with aUC. The final outcome analysis was finished in August 2023 with 886 patients recruited. The experimental treatment significantly improved mPFS (12.5 vs. 6.3 months, HR =0.45; 95% CI: 0.38–0.54; P<0.001) and mOS (31.5 vs. 16.1 months, HR =0.47; 95% CI: 0.38–0.58; P<0.001). The combination showed a better response rate with an ORR of 67.7% vs. 44.4% for the chemotherapy arm (P<0.001) (9). This study highlights the association of EV and pembrolizumab as the new potential standard of care in first line.
Treatment-related adverse events (TRAEs) were notable, with over 50% grade 3 side effects in EV studies. Common TRAEs included alopecia, peripheral sensory neuropathy, pruritus, and fatigue. Hyperglycemia was observed, particularly in diabetic and overweight patients, with 19 reported cases in the EV-301 study, including one death (9,44).
Targeting TROP2
Sacituzumab govitecan (SG) is a therapeutic agent consisting of an IgG1 kappa antibody fused with SN-38, the active metabolite of irinotecan, a topoisomerase I inhibitor (36). The antibody targets TROP2, a transmembrane calcium signal transducer expressed in healthy multi-stratified epithelial and trophoblast cells, as well as in various carcinomas. TROP2 is involved in intracellular signaling pathways governing cell proliferation, migration, and adhesion. Higher TROP2 expression is particularly evident in invasive bladder cancer compared to non-invasive bladder cancer and normal urothelium. Moreover, increased TROP2 expression correlates positively with disease severity, suggesting its potential as both a prognostic marker and a therapeutic target (47-49).
The TROPHY-U-01 trial, a phase II study comprising multiple cohorts, investigated the efficacy of SG, dose of 10 mg/kg SG intravenously (IV) on D1 and D8 every 3 week (Q3W), in aUC. In Cohort 1, consisting of 113 patients who had previously received platinum-based chemotherapy and anti-PD-1 or anti-PD-L1 therapy, SG demonstrated an ORR of 27% (95% CI: 19.5–36.6%), with six confirmed complete responses. mPFS was 5.4 months (95% CI: 3.5–7.2), and mOS was 10 months (95% CI: 9–13.8) (36). Based on these findings, SG received accelerated approval from the FDA for aUC patients who had previously received platinum-based chemotherapy with PD-1/PD-L1 inhibitors in 2021. The phase III Tropics-04 study aims to validate these results in patients who progressed after platinum-based chemotherapy and PD-1/PD-L1 inhibitors (NCT04527991).
In Cohort 3, comprising 61 patients with platinum-refractory aUC who had not undergone immunotherapy, SG plus pembrolizumab demonstrated an ORR of 34% and a 6-month PFS of 47% (37). Cohort 2 is evaluating SG after first-line immunotherapy only, and Cohorts 4 and 5 are investigating its combination with cisplatin, with or without avelumab, or induction therapy with the anti-PD-1 antibody zimberelimab, followed by maintenance therapy with avelumab or zimberelimab.
The most common grade ≥3 TRAEs observed in the TROPHY-U-01 trial were myelotoxicity-related, including neutropenia (35%, including 10% febrile neutropenia), leucopenia (18%), anemia (14%), and diarrhea (10%) (37).
The Double Antibody Drug Conjugate (DAD) study investigated the combination of SG and EV in patients with aUC who had prior progression on platinum-based chemotherapy and/or ICIs. This phase I trial enrolled 24 patients to assess the feasibility and toxicity of the combined treatment. Results showed an impressive ORR of 70% (95% CI: 47–87%), with 86% of patients alive after 12 months, and 41% experiencing no disease progression during this period. However, notable toxicities included diarrhea (87%) and neuropathy (57%), while myelotoxicity was observed in 35% of patients with grade 3 anemia and neutropenia. Based on the toxicity profile, the recommended phase II doses were determined as SG 10 mg and EV 1.25 mg/kg, both administered on days 1 and 8 every 21 days. The DAD-IO trial will further evaluate the combination of this antibody conjugate doublet, either alone or in combination with pembrolizumab (43).
Targeting SLITRK6
Sirtratumab vedotin (SV) is a novel ADC that combines an IgG2 antibody targeting the transmembrane protein SLITRK6 with MMAE via a protease-cleavable linker. The SLITRK family, consisting of six members, is primarily expressed in the central nervous system, where they regulate neuronal cell growth and synaptic development (50,51). Pre-clinical study have demonstrated SLITRK6 expression in 88% of tested samples, with 100% expression observed in metastatic bladder tumors (51).
The safety and pharmacokinetic profile of SV were assessed in a phase I dose-escalation, IV at six levels (0.10 to 1.25 mg/kg), trial involving 93 patients with aUC. The trial reported an ORR of 18.3%, with 17 partial responses in the total study population. The median duration of response (DOR) was 15 weeks, and the mPFS was 16 weeks. The most significant TRAEs included fatigue (54.8%), nausea (37.6%), and reduced appetite (35.5%). Notably, there were 27 cases of ocular toxicity, two of which were grade 3 (40). Despite these preliminary findings, comprehensive efficacy data for SV remains to be further explored.
Targeting HER2
HER2 is a type I transmembrane protein belonging to the ERBB family, playing a crucial role in activating signaling pathways associated with tumorigeneris (13), making it a significant therapeutic target in breast and gastric cancers (52,53). In urothelial cancer, HER2 is the third most prevalent malignancy with overexpression rates ranging from 6% to 12.4% (54,55). Despite this prevalence, the efficacy of HER2-targeting agents in urothelial carcinoma remains under investigation.
Trastuzumab emtansine (T-DM1), the first ADC to receive FDA approval, combines an anti-HER2 humanized antibody with the anti-microtubule agent DM-1 via a non-cleavable linker. In a basket trial involving 58 patients, including 4 with urothelial cancer, the ORR was 26%, with a median DOR of 8 months. However, none of the urothelial cancer patients responded to T-DM1 (56). The KAMELEON study, a single-arm, non-randomized phase II trial, aimed to evaluate T-DM1’s (2.4 mg/kg once weekly or 3.6 mg/kg Q3W) efficacy in bladder cancer, cholangiocarcinoma, and pancreatic cancer. Among the 20 enrolled patients, 13 had urothelial carcinoma, with an ORR of 38.5%. However, due to recruitment issues, the study was prematurely terminated without meeting its primary endpoints (38).
The novel ADC disitamab vedotin (DV) combines a new-generation humanized anti-HER2 antibody, hertuzumab, with MMAE via a cathepsin-cleavable linker (57). Its efficacy and safety, at a dose of 2 mg/kg IV every 2 weeks (Q2W), were evaluated in two phase II single-arm studies recruiting patients with HER2-positive aUC that had progressed after at least one line of systemic treatment. The combined analysis of 107 patients showed an ORR of 50.5% (95% CI: 40.6–60.3%), with a higher response rate observed in patients with 3+ in IHC or with 2+ and fluorescence in situ hybridization (FISH) +. The median DOR was 7.3 months (95% CI: 5.7–10.8). mPFS was 5.9 months (95% CI: 4.3–7.2), and mOS was 14.2 months (95% CI: 9.7–18.8) (39). DV was also evaluated in patients with aUC HER2 negative (IHC 0 or 1+) and ≥1 prior systemic treatment. Notably, all patients with HER2 0 in IHC had stable disease. The ORR was 26.3% (95% CI: 9.1–51.2%). mPFS and mOS were 5.5 months (95% CI: 3.9–6.8) and 16.4 months (95% CI: 7.1–21.7 months), respectively (41). The main TRAEs reported in the combined analysis of phase II trials with DV were peripheral neuropathy (68.2%, 18.7% of them grade 3), leucopenia (50.5%), and aspartate aminotransferase (AST) increase (42.1%). Neutropenia was reported in 42.1% of the patients, with 12.1% of them grade 3 (39).
DV was also evaluated in combination with ICIs. Preliminary results from a phase Ib/II trial accessed the safety and activity of the combination of DV (2 mg/kg IV Q2W) and the anti-PD-1 toripalimab (3 mg/kg Q2W) in patients with aUC. The study showed promising efficacy, with an ORR of 73.2% (95% CI: 57.1–85.8%) (42). An ongoing phase II trial is assessing DV with or without pembrolizumab in patients with HER2-expressing aUC, with three cohorts focusing on different treatment lines and HER2 expression levels (NCT04879329).
Trastuzumab deruxtecan (T-Dxd) combines an anti-HER2 antibody with a topoisomerase I inhibitor payload via a cleavable linker. Its higher drug-to-antibody ratio (DAR) (8:1) and cytotoxic effect, explain the efficacy of this ADC in scenarios such as HER2-low metastatic breast cancer (58,59). The phase II basket trial DESTINY-PanTumor02 evaluated T-Dxd (5.4 mg/kg Q3W) in patients with metastatic tumors refractory to at least one line of systemic treatment and a HER2 expression of 3+ or 2+ in IHC. Two hundred sixty-eight patients were enrolled, 41 of them with bladder cancer. After a median follow-up of 12.75 months, the ORR for the entire population was 37.1% (95% CI: 31.3–43.2%), mPFS was 6.9 months (95% CI: 5.6–8), and mOS was 21.1 months (95% CI: 15.3–29.6). The bladder cancer cohort reported an ORR of 39% (95% CI: 24.2–55.5%), with one complete response. mPFS in the bladder cancer cohort was 7 months (95% CI: 4.2–9.7), and mOS was 12.8 months (95% CI: 11.2–15.1) (34).
The combination of T-Dxd at 5.4 mg/kg and nivolumab 360 mg IV Q3W, was investigated in a phase Ib/II trial with 34 patients with HER2-positive aUC. The primary analysis showed an ORR of 36.7% (95% CI: 19.9–56.1%) with 4 complete responses. The median DOR was 13.1 months (95% CI: 4.1–not evaluable). Drug-related pneumonitis occurred in 23.5% of the patients (35).
TKIs
In the context of aUC, immunotherapy, cytotoxic chemotherapy, and, more recently, ADCs represent fundamental components of systemic treatment. However, there remains a critical need to enhance therapeutic outcomes. The exploration of combination therapies and the strategic sequencing of these treatments are pivotal endeavors.
Protein kinases play critical regulatory roles in signaling pathways and cellular mechanisms, including proliferation, differentiation, survival, and apoptosis (60). Tyrosine kinases, found in nuclear and membrane-bound forms, act as transmembrane receptors, often upregulated in tumors, crucial for cancer initiation and progression. TKI strategies show effectiveness in targeted tumor therapies (60).
For aUC, the presence of mutations or overexpression of tyrosine kinases significantly accelerates tumor proliferation and progression (61). Given the considerable heterogeneity in behavior and aggressiveness of aUC, gene expression profiling and molecular pathway analysis emerge as promising modalities for evaluating patient prognosis and guiding treatment selection (61). Table 2 summarizes the results of clinical trials evaluating TKIs for aUC patients.
Table 2
Study [year] | Phase | N | Population | Drug | Median overall survival | Median progression-free survival | Objective response rate | Median DOR | Target expression |
---|---|---|---|---|---|---|---|---|---|
Marandino et al., BLC2001 [2019] (62) | II | 99 | Locally advanced or mUC with FGFR alterations and progression during or after at least one course of chemotherapy or within 12 months after neoadjuvant or adjuvant chemotherapy | Erdafitinib | 13.8 mo (95% CI: 9.8–NR) | 5.5 mo (95% CI: 4.2–6.0) | 40% (95% CI: 31–50%) | 5.6 mo (95% CI: 4.2–7.2) | FGFR3 mutation or FGFR2/3 fusion |
Loriot et al., THOR Cohort 1 [2023] (11) | III | 266 (136 erdafitinib + 130 chemotherapy) | Unresectable or mUC with FGFR3/2 alterations previously treated with one or two treatments that included an anti-PD-(L)1 | Erdafitinib vs. chemotherapy (docetaxel or vinflunine) | 12.1 mo for erdafitnib vs. 7.8 mo for chemotherapy (HR =0.64; 95% CI: 0.47–0.88; P=0.005) | 5.6 mo for erdafitnib vs. 2.7 mo for chemotherapy (HR =0.58; 95% CI: 0.44–0.78; P<0.001) | 45.6% for edafitinib vs. 11.5% for chemotherapy (relative benefit, 3.94; 95% CI: 2.37–6.57; P<0.001) | 4.9 mo (95% CI: 3.8–7.5) for erdafitinib vs. 5.6 mo (95% CI: 2.1–6.0) for chemotherapy | FGFR3 mutation or FGFR2/3 fusion |
Siefker-Radtke et al., THOR Cohort 2 [2024] (63) | III | 351 (175 erdafitinib + 176 pembrolizumab) | Unresectable or mUC with FGFR3/2 alterations previously treated with 1 treatment not containing an anti-PD-(L)1 agent | Erdafitinib vs. pembrolizumab | 10.9 mo for erdafitnib vs. 11.1 mo for pembrolizumab (HR =1.18; 95% CI: 0.90–1.50) | 4.4 mo for erdafitinib vs. 2.7 mo for pembrolizumab (HR =0.88; 95% CI: 0.70–1.10) | 40% for erdafitinib vs. 21.6% for pembrolizumab, relative risk of 1.85 (95% CI: 1.32–2.39; P<0.001) | 4.3 mo (95% CI: 3.7–6.9) for erdafitinib vs. 14.4 mo (95% CI: 7.4–27.8) for pembrolizumab | FGFR3 mutation or FGFR2/3 fusion |
Lyou et al., [2022] (64) | I | 67 | mUC with prior platinum-based chemotherapy, and FGFR3 mutation/fusion | Infigratinib | 7.93 mo (95% CI: 2.4–NE) for first line and 7.75 mo (95% CI: 5.72–13.67) for subsequent lines | 3.65 mo (95% CI: 0.95–7.43) for first line and 3.75 mo (95% CI: 3.09–5.42) for subsequent lines | 25.4% (95% CI: 15.5–37.5%) | Not reported | FGFR3 mutation or FGFR2/3 fusion |
Sternberg et al., FORT1-1 [2023] (65) | II/III | 174 (87 rogaratinib + 88 chemotherapy) | Locally advanced or mUC with ≥1 prior platinum-containing regimen with FGFR1/3 mRNA-positive | Rogaratinib vs. chemotherapy (docetaxel, paclitaxel or vinflunine) | 8.3 mo for rogaratinib vs. 9.8 mo (HR =1.11; 95% CI: 0.71–1.72; P=0.67) | 4.9 mo for rogaratinib vs. 5.8 mo for chemotherapy (HR =1.23; 95% CI: 0.84–1.80; P=0.86) | 20.7% for rogaratinib (95% CI: 12.7–30.7%) vs. 19.3% for chemotherapy (95% CI: 11.7–29.1%) | 4.9 mo (95% CI: 3.5–9.1) for rogaratinib vs. 5.8 mo (95% CI: 3.5–7.7) for chemotherapy | FGFR1/3 mRNA-positive |
Powles et al., NORSE [2021] (66) | II | 87 (44 edafitinib + cetrelimab and 43 erdafitinib) | mUC with no prior systemic therapy, cisplatin ineligible and FGFR mutation/fusion | Edafitinib + cetrelimab vs. erdafitnib | 20.8 mo (95% CI: 5.12–NE) for edafitinib + cetrelimab vs. 16.2 mo (95% CI: 8.3–NE) for erdafitnib | 10.9 mo (95% CI: 5.45 to 13.73) for edafitinib + cetrelimab vs. 5.6 mo (95% CI: 4.3 to 7.36) for erdafitnib | 54.5% (95% CI: 38.8–69.6%) for edafitinib + cetrelimab vs. 44.2% (95% CI: 29.1–60.1%) for erdafitnib | 11.1 mo (95% CI: 8.7–NE) for edafitinib + cetrelimab vs. 9.7 mo (95% CI: 4.6–NE) for erdafitnib | FGFR mutation/fusion |
Rosenberg et al., FORT-2 [2021] (67) | Ib/II | 26 mUC | Advanced solid tumors, including UC (cisplatin-ineligible, advanced/metastatic with no prior systemic therapy), with FGFR1-3 mRNA overexpression | Rogaratinibe + atezolizumab | – | – | 44% (95% CI: 24–65%) | Not reached | FGFR1-3 mRNA overexpression |
Apolo et al., Cohort 1 [2020] (68) | II | 68 (49 cohort 1) | mUC with prior after platinum chemotherapy, platinum-refractory | Cabozantinib | 8.1 mo (95% CI: 5.2–10.3) | 3.7 mo (95% CI: 3.1–6.4) | 19.1% (95% CI: 8.6–34.1%) | – | Patients enrolled regardless of target expression |
Apolo et al., [2020] (69) | I | 54 (15 with mUC) | mUC and other genitourinary malignances | Cabozantinib + nivolumab and cabozantinib + nivolumab + ipilimumab | 25.4 mo (95% CI: 5.7–41.6 mo) for patients with mUC | 12.8 mo (95% CI: 1.8–24.1 mo) for patients with mUC | 38.5% (95% CI: 13.9–68.4%) for patients with mUC | Not reached for patients with mUC | Patients enrolled regardless of target expression |
Girardi et al., [2022] (70) | I | 30 | mUC that progressed on checkpoint inhibition | Cabozantinib plus nivolumab | 10.4 mo (95% CI: 5.8–19.5 mo) | 3.6 mo (95% CI: 2.1–5.5 mo) | 16.0% (95% CI: 4.5–36.1%) | 33.5 mo (95% CI: 3.7–33.5) | Patients enrolled regardless of target expression |
Pal et al., COSMIC Cohort 2 [2020] (71) | Ib | 30 | Locally advanced or mUC with disease progression after platinum-containing chemotherapy | Cabozantinib plus atezolizumab | – | 5.4 mo (range, 0.0+ to 17.3+) | 27% (80% CI: 16–40%) | Not reached | Patients enrolled regardless of target expression |
Pal et al., COSMIC Cohort 3 [2022] (72) | Ib | 30 | Locally advanced or mUC with no prior therapy and cisplatin-ineligible | Cabozantinib plus atezolizumab | 14.3 mo (95% CI: 8.6–NE) | 5.6 mo (95% CI: 3.1–11.1) | 20% (95% CI: 8–39%) | 7.1 mo (95% CI: 2.8–NE) | Patients enrolled regardless of target expression |
Pal et al., COSMIC Cohort 4 [2022] (72) | Ib | 30 | Locally advanced or mUC with no prior therapy and cisplatin-based chemo eligible | Cabozantinib plus atezolizumab | 13.5 mo (95% CI: 7.8–23.2) | 7.8 mo (95% CI: 1.6–13.8) | 30% (95% CI: 15–39%) | NE (95% CI: 7.2–NE) | Patients enrolled regardless of target expression |
Pal et al., COSMIC Cohort 5 [2022] (72) | Ib | 31 | Locally advanced or mUC with one prior ICI and no prior VEGFR-TKI therapy | Cabozantinib plus atezolizumab | 8.2 mo (95% CI: 5.5–9.8) | 3.0 mo (95% CI: 5.5–23.2) | 10% (95% CI: 2–26%) | 4.1 mo (95% CI: 2.6–NE) | Patients enrolled regardless of target expression |
Study [year] | Phase | N | Population | Drug | Median overall survival | Median progression-free survival | Objective response rate | Median DOR | Target expression |
Jain et al., PemCa [2024] (73) | II | 36 | Locally advanced or mUC, cisplatin-ineligible (including patient refusal of cisplatin), treatment naïve, and no prior PD-1/L1 therapy | Cabozantinib plus pembrolizumab | 17.1 mo (95% CI: 12.6–NE) | 7.6 mo (95% CI: 5.3–12.6) | 45.7% (95% CI: 30.5–61.8%) | 14.7 mo | Patients enrolled regardless of target expression |
Msaouel et al., Cohort 1 + 3 [2024] (74) | II | 67 | Locally advanced or mUC refractory to therapy with checkpoint inhibitors and treated with platinum | Sitravatinib plus nivolumab | 8.6 mo (95% CI: 5.7–16.0) | 3.9 mo (95% CI: 3.1–5.5) | 14.9% (95% CI: 7.4–25.7%) | 9.6 mo (95% CI: 3.8–NE) | Patients enrolled regardless of target expression |
Msaouel et al., Cohort 5 [2024] (74) | II | 53 | Locally advanced or mUC naïve checkpoint inhibitors and treated with platinum | Sitravatinib plus nivolumab | 13.4 mo (95% CI: 5.8–21.3) | 3.9 mo (95% CI: 3.5–5.7) | 32.1% (95% CI: 19.9–46.3%) | 7.3 mo (95% CI: 5.5–13.0) | Patients enrolled regardless of target expression |
Msaouel et al., Cohort 7 [2024] (74) | II | 56 | Locally advanced or mUC refractory to therapy with checkpoint inhibitors and antibody-drug conjugate and treated with platinum | Sitravatinib plus nivolumab | 9.0 mo (95% CI: 7.4–10.8) | 3.7 mo (95% CI: 2.2–5.5) | 5.4% (95% CI: 1.1–14.9%) | 7.4 mo (95% CI: 5.6–NE) | Patients enrolled regardless of target expression |
Sadeghi et al., [2023] (75) | II | 70 | mUC that recurred or progressed after platinum-based chemotherapy | sEphB4-HSA and pembrolizumab | 14.6 mo (95% CI: 9.2–21.5) | 4.1 mo (95% CI: 1.5–5.7) | 41% (95% CI: 29–54%) | Not reached | Patients enrolled regardless of target expression |
TKIs, tyrosine kinase inhibitors; UC, urothelial carcinoma; DOR, duration of response; mUC, metastatic urothelial carcinoma; FGFR, fibroblast growth factor receptor; mo, months; CI, confidence interval; NR, not reached; PD-(L)1, programmed death (ligand)-1; HR, hazard ratio; NE, not estimable; VEGFR, vascular endothelial growth factor receptor; ICI, immune checkpoint inhibitor.
FGFR
The FGFR family is made of four genes: FGFR1–4 (76). They regulate angiogenesis in adults and in embryonic development, as well as cell migration, survival and proliferation (77). Molecular alterations, such as mutations and fusions, are prevalent in aUC and are related to carcinogenesis (78). These molecular alterations can be found in up to 20% of patients with bladder aUC and 37% in those with upper tract urothelial carcinoma (79,80).
In 2019, erdafitinib, an FGFR1–4 TKI, became the first targeted therapy approved for aUC, based on the BLC2001 study, which demonstrated its efficacy in chemotherapy-refractory patients with FGFR molecular alterations (62). A 24-month follow-up confirmed its efficacy, with an ORR of 40% (95% CI: 31–50%) and a mPFS of 5.5 months (95% CI: 4.2–6.0). Among these patients, 46% experienced TRAEs of grade 3 or higher. Hyperphosphatemia (77%), stomatitis (58%), diarrhea (51%), and dry mouth (46%) were the most common TRAEs of any grade. Common TRAE of grade 3 or higher were stomatitis (10%), hyponatremia (10%) and asthenia (7%) (62).
The Phase 3 THOR study assessed erdafitinib (8 mg orally once daily) vs. standard chemotherapy (Cohort 1) or pembrolizumab (Cohort 2) in aUC patients with susceptible FGFR3/2 alterations. In Cohort 1, patients must have progressed during or after one or two prior systemic therapy regimens including an ICI. Patients were randomized to erdafitinib or chemotherapy (docetaxel or vinflunine). Erdafitinib significantly increased OS with a median of 12.1 vs. 7.8 months for chemotherapy (HR =0.64; P=0.005) and improved mPFS (5.6 vs. 2.7 months, HR =0.58; 95% CI: 0.44–0.78). The erdafitinib group had a higher ORR compared to chemotherapy (45.6% vs. 11.5%; P<0.001). Grade 3 and 4 TRAE occurred in 45.9% of patients receiving erdafitinib. The most common grade 3 TRAEs were palmar-plantar erythrodysesthesia syndrome (9.6%), stomatitis (8.1%), and onycholysis (5.9%) in the erdafitinib group. Hyperphosphatemia of any grade occurred in 80% of patients receiving erdafitinib (11).
Cohort 2 evaluated aUC patients who progressed on one prior line of systemic treatment and had not received ICIs (81). Patients were randomized to erdafitinib or pembrolizumab (200 mg IV Q3W). There were no significant differences in OS (HR =1.18; 95% CI: 0.92–1.51; P=0.18) or PFS (HR =0.88; 95% CI: 0.70–1.10) between erdafitinib and pembrolizumab. mOS for erdafitinib was 10.9 months (95% CI: 9.2–12.6) vs. 11.1 months for pembrolizumab (95% CI: 9.7–13.6), and mPFS 4.4 months (95% CI: 4.1–5.5) and 2.7 months (95% CI: 1.6–3.0), respectively. The ORR was higher with erdafitinib at 40% vs. 21% for pembrolizumab. Hyperphosphatemia (72.8%), stomatitis (45.1%), and diarrhea (44.5%) were the most common TRAEs of any grade in the erdafitinib group (63).
Other FGFR inhibitors are currently under investigation. In a phase I trial, infigratinib, a selective FGFR 1–3 TKI, was administered orally once daily in a 28-day cycle (21 days on, 7 days off) at a starting dose of 125 mg per day. Eligible patients had received prior platinum-based chemotherapy or were ineligible for such treatment. Of the 67 enrolled patients, 13 (19.4%) received infigratinib as first-line therapy due to ineligibility for platinum-based chemotherapy. The ORR was 30.8% (95% CI: 9.1–61.4%) for first-line patients and 24.1% (95% CI: 13.5–37.6%) for second-line or later patients. Among all 67 patients, there were no significant differences in mPFS and OS times between early-line and salvage scenarios. The most common TRAEs, of any degree, were increased creatinine (40.3%), fatigue (38.8%), and hyperphosphatemia (38.8%) (64).
In the phase II/III randomized, open-label FORT-1 trial, rogaratinib (an FGFR1–4 inhibitor) at a dose of 800 mg orally twice daily was compared with second-line chemotherapy (either a taxane or vinflunine) in 175 eligible patients. Results indicated that rogaratinib did not improve OS or PFS despite showing similar antitumor activity. The ORRs were 20.7% (95% CI: 12.7–30.7%) with rogaratinib and 19.3% (95% CI: 11.7–29.1%) with chemotherapy. mOS was 8.3 months (95% CI: 6.5–not estimable) with rogaratinib and 9.8 months (95% CI: 6.8–not estimable; HR =1.11; P=0.67) with chemotherapy. mPFS was 2.7 months (95% CI: 1.6–4.6) with rogaratinib and 3.2 months (95% CI: 2.7–4.4) (HR =1.23; P=0.86). Grade 3 TRAE occurred in 43% of patients receiving rogaratinib and 39% of patients receiving chemotherapy. Six patients (7.0%) receiving rogaratinib had grade ≥2 retinal disorders. Gastrointestinal toxicities were among the most frequently reported adverse effects, with hyperphosphatemia being the second most frequent adverse effect associated with rogaratinib (65).
Evaluations of combination regimens, such as FGFR inhibitors with immunotherapy, present new therapeutic prospects. The phase 2 NORSE study investigated erdafitinib (8 mg orally once daily) alone or in combination with the anti-PD-1 cetrelimab (240 mg IV Q2W) in first-line cisplatin-ineligible metastatic urothelial cancer (mUC) patients with FGFR alterations (mutations/fusions). Out of 87 enrolled patients, results favored erdafitinib plus cetrelimab over erdafitinib alone. The combination demonstrated an ORR of 54.5% (95% CI: 38.8–69.6%) vs. 44% (95% CI: 29.1–60.1%) for erdafitinib alone. mPFS was 10.97 months (95% CI: 5.45–13.63) vs. 5.62 months (95% CI: 4.34–7.36) with erdafitinib alone. Median DOR was 11.1 months (95% CI: 8.77–NE) vs. 9.72 months (95% CI: 4.60–NE) (66).
Similarly, the FORT-2 study reported promising outcomes for rogaratinib plus atezolizumab combination therapy, although caution is advised due to a potential increase in grade 3 and 4 adverse events. In the FORT-2 phase Ib/II trial, the safety and efficacy of first line rogaratinib (600 mg oral twice daily) plus atezolizumab (1,200 mg IV Q3W) were analyzed in cisplatin-ineligible patients with FGFR 1–3 mRNA overexpression. The overall disease control rate was 68% (95% CI: 47–85%), with an ORR of 44% (95% CI: 24–65%), including four complete responses. The most frequent TRAEs were nausea (42%), hyperphosphatemia (58%), and diarrhea (65%). Grade 3/4 TRAE included elevated lipase without pancreatitis (19%), elevated amylase (12%), rash (2.8%), and syncope (2.8%). The maximum tolerable dose of rogaratinib was established at 600 mg twice daily (67).
Vascular endothelial growth factor receptor (VEGFR)
Angiogenesis plays a significant role in carcinogenesis of most cancers including urothelial carcinoma. The role of VEGFR in aUC has been extensively investigated (82). C-MET, a cell surface tyrosine kinase receptor primarily expressed in epithelial cells (83), is linked to a poorer prognosis and shorter OS in patients with muscle-invasive bladder cancer (84). Cabozantinib, a non-selective TKI targeting c-Met, VEGFR2, FLT3, KIT, AXL, and RET, is being explored in aUC, showing promise both alone and in combination with ICIs (68).
A phase II trial at the National Cancer Institute assessed cabozantinib in platinum-refractory aUC and rare genitourinary histologies. Cohort 1 comprised platinum-refractory aUC patients with measurable disease, while Cohorts 2 and 3 were exploratory. Cabozantinib at 60 mg/day yielded a clinical benefit of 64.3%, with an ORR of 19.1% and mPFS and mOS of 3.7 and 8.1 months, respectively. Grade 3–4 TRAEs were fatigue (9%), hypertension (7%), proteinuria (6%) and hypophosphatemia (6%). The study showed that cabozantinib has innate and adaptive immunomodulatory qualities that may combat tumor-induced immunosuppression, according to exploratory translational assessments (68). This justifies integrating cabozantinib with ICIs.
In another phase I study, cabozantinib plus nivolumab and cabozantinib plus nivolumab and ipilimumab were evaluated in aUC and other genitourinary malignancies. Recommended phase 2 doses were established at cabozantinib 40 mg/day plus nivolumab 3 mg/kg and cabozantinib 40 mg/day, nivolumab 3 mg/kg and ipilimumab 1 mg/kg. The ORR in aUC patients was 38.5%, with a mPFS of 12.8 months and mOS of 25.4 months. Grade 3 or 4 TRAEs occurred in 87% of patients receiving cabozantinib and nivolumab plus ipilimumab and 75% in patients treated with cabozantinib and nivolumab and fatigue (10% and 17%, respectively), diarrhea (7% and 4%, respectively) and hypertension (10% and 21%, respectively) were the most common (69). Subsequently, a study assessed cabozantinib plus nivolumab in aUC patients who progressed on previous ICIs, reporting an ORR of 16.0% and mPFS and mOS of 3.6 and 10.4 months, respectively (70).
The COSMIC-021 trial evaluated cabozantinib 40 mg daily and atezolizumab 1,200 mg IV Q3W in solid tumors, showing an ORR of 27% in aUC patients previously treated with platinum-based chemotherapy (Cohort 2). Cohorts 3, 4, and 5 investigated this combination in locally advanced or metastatic urothelial carcinoma, with ORRs ranging from 10% to 30% and mOS ranging from 8.2 to 14.3 months (72) (Table 2). Additionally, cabozantinib is under evaluation with avelumab in the MAIN-CAV trial for maintenance therapy after platinum-based chemotherapy.
In patients with cisplatin-ineligible and PD-L1-positive aUC, defined as having a combined positive score (CPS) of ≥10, the LEAP-011 trial, a randomized phase III study, evaluated the efficacy of first-line treatment with pembrolizumab (200 mg IV Q3W) plus the broad-spectrum tyrosine-kinase inhibitor lenvatinib (20 mg orally) compared to pembrolizumab plus placebo. The study found no significant differences in mPFS and mOS between the two groups. mPFS was 4.5 months in the combination arm vs. 4.0 months in the pembrolizumab arm (HR =0.90; 95% CI: 0.72–1.14), and mOS was 11.8 vs. 12.9 months, respectively (HR =1.14; 95% CI: 0.87–1.48). However, the combination arm showed increased toxicity, with grade 3 or higher TRAEs occurring in 51% of patients compared to 27% in the control arm (85).
The phase II PemCa trial investigated pembrolizumab (200 mg IV Q3W) plus cabozantinib (40 mg orally once daily) as first-line therapy in aUC patients ineligible for cisplatin-based chemotherapy. Patients with PD-L1 CPS ≥10 or ineligible for platinum-based chemotherapy, irrespective of PD-L1 status, were eligible. Out of 36 patients, 22% were PD-L1 positive. The ORR was 46% (95% CI: 30.5–61.8%), with a median follow-up of 14.3 months. mPFS was 7.6 months (95% CI: 5.3–12.6) and mOS 17.1 months (95% CI: 12.6–not estimable). Grade ≥3 TRAEs occurred in 50% of patients, with fatigue (63.9%), diarrhea (58.3%), and palmar-plantar erythrodysesthesia syndrome (41.7%) being the most common (73).
Sitravatinib, a multi-TKI targeting various receptors, including VEGFR and MET, was assessed in a phase 2 multi-cohort study in combination with nivolumab for aUC patients. Eight cohorts were enrolled based on prior treatments and tested the combination of sitravatinib (120 mg once daily or 100 mg once daily) and nivolumab (240 mg every 2 weeks or 480 mg every 4 weeks IV), depending on prior treatment with ICIs, platinum-based chemotherapy, or ADC. mPFS varied from 3.5 to 7.8 months, while the ORR varied amongst the groups from 0% to 33% (Table 2). Cohort 5, comprising patients who had previously received platinum-based chemotherapy but no ICI, exhibited the highest ORR of 32.1% (95% CI: 19.9–46.3%) with a mPFS of 3.9 months (95% CI: 3.5–5.7). Cohort 2, which evaluated 23 patients, who had previously received immunotherapy but were not eligible for platinum, had an ORR of 21.7% and the longest mPFS of 7.79 months (95% CI: 3.94–14.62). The most frequent TRAEs were diarrhea (55.3%), fatigue (50.8%), decreased appetite (38.5%), dysphonia (34.8%) and vomiting (29.9%) (74).
XL092 (zanzalintinib), an oral inhibitor targeting MET, VEGFR2, and TAM kinases, is under investigation for advanced solid tumors, including urothelial carcinoma, after one prior platinum-based regimen and no prior immunotherapy. Preclinical studies showed anti-tumor activity and synergy with immunotherapy agents. A phase 1b study (NCT05176483) will assess XL092’s safety and clinical activity alone and in combination therapies (86).
EPH
The relationship between cell migration and neoplastic transformation is closely associated with Eph receptor tyrosine kinases and their ephrin ligands. Particularly, EphB4, which interacts with EphrinB2, plays a pivotal role in angiogenesis. Inhibiting EphB4 could potentially induce apoptosis and impede tumor growth (87). A phase II study evaluated EphB4 inhibition using soluble EphB4-human serum albumin (sEphB4-I) in combination with pembrolizumab in patients with aUC who had relapsed or progressed after platinum-based therapy. The study reported a mOS of 14.6 months (95% CI: 9.2–21.5), with patients expressing Ephrin-B2 showing an OS of 21.5 months (95% CI: 12.4–not reached) and a complete response rate of 24% (11 of 46; 95% CI: 12–36%). The most common TRAE of any grade were hypertension (74%), fatigue (44%), and anemia (17%), with 46% of patients experiencing grade 3 or 4 hypertension (75). Currently, a phase II study is ongoing to further investigate this combination in aUC, including cohorts for first-line treatment and those previously treated, with a focus on patients exhibiting a positive biomarker for EphrinB2 (NCT04486781).
iPARPs
Up to 25% of aUC patients carry mutations in various genes involved in the DDR, including BRCA1, BRCA2, ERCC2, and ATM, as revealed by extensive genome sequencing studies. One strategy to impede the repair of single-strand DNA breaks is through pharmacological inhibition of PARP. Consequently, iPARPs induce excessive DNA damage, leading to cancer cell death in the context of homologous recombination deficiency (HRD) (88,89).
The ATLAS study assessed the efficacy of the iPARP rucaparib (600 mg orally twice daily) as monotherapy in 97 previously treated aUC patients. HRD-positive tumors were identified in 21% of patients, HRD-negative tumors in 30%, and HRD status undetermined in 48%. mPFS did not significantly differ between HRD-positive and HRD-negative tumors (1.4 vs. 1.8 months). The three most common any-grade TRAEs were anemia (36.1%), nausea (42.3%), and asthenia/fatigue (57.7%) (90).
Initial data from the BAYOU trial, a randomized phase II study, compared an ICI with an iPARP. This trial evaluated durvalumab (1,500 mg IV every 4 weeks) in combination with olaparib (300 mg orally twice daily) or placebo for untreated patients with aUC ineligible for platinum-based chemotherapy. A total of 154 patients were randomized. In the intention-to-treat (ITT) group, there was no significant difference in mPFS between the combination and durvalumab alone arms. mPFS was 4.2 months (95% CI: 3.6–5.6) in the combination group vs. 3.5 months (95% CI: 1.9–5.1) in the durvalumab alone group (HR =0.94; P=0.78). In the subgroup of patients with homologous recombination repair gene mutations (HRRm), mPFS was 5.6 months (95% CI: 1.9–8.1) for the combination group vs. 1.8 months (95% CI: 1.7–2.2) for durvalumab monotherapy (HR =0.18; 95% CI: 0.06–0.47). Grade 3 or 4 TRAEs occurred in 18% and 9% of patients, respectively (25).
Targeting DDR signaling pathways may potentially be a successful strategy in patients with aUC, although the results of studies testing iPARPs that did not meet the primary endpoint, and the current outcomes with these drugs, introduce uncertainty. Nonetheless, alternative approaches utilizing these drugs are under investigation, such as the NICARAGUA study (NCT03397394), which is exploring niraparib plus cabozantinib in unselected patients with treatment-refractory aUC.
Conclusions
In this article, we explore the latest therapeutic advancements in the treatment of aUC, a challenging disease due to its aggressive nature and widespread prevalence. Traditionally, platinum-based chemotherapy has been the mainstay treatment modality for decades. However, in recent years, we have witnessed the emergence of new therapeutic options such as ICIs and targeted therapy with ADCs, and TKIs. Recent clinical trials have demonstrated the efficacy of these approaches, both as monotherapy and in combination, in patients with platinum-refractory aUC. Promising results from trials such as EV-302/KEYNOTE-A39 have highlighted the combination of EV with pembrolizumab as a new standard of care in the first-line setting. Additionally, targeted therapies such as ADCs targeting TROP2 and HER2 and TKIs for receptors like FGFR, VEGFR and c-MET are also emerging as promising strategies. While these advancements have significantly improved treatment outlooks, challenges remain, such as managing TRAE and identifying predictive biomarkers of response. However, it is undeniable that we are witnessing a transformation in the treatment paradigm of aUC, providing patients with new hopes and opportunities for prolonged survival and improved quality of life.
Acknowledgments
Funding: None.
Footnote
Peer Review File: Available at https://cco.amegroups.com/article/view/10.21037/cco-24-67/prf
Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://cco.amegroups.com/article/view/10.21037/cco-24-67/coif). D.M.G. reports that he has taken part as invites speaker for Pfizer, Merck, BMS Brazil, Jansen oncology, Bayer, Adium Brazil and Ipsen. He has received support from Pfizer, Ipsen and Adium for meeting registration, accommodation and/or travel expense. Additionally, he has received consulting fees from BMS Brazil and Jansen Oncology and has received payment for expert testimony from Jansen Oncology. The other authors have no conflicts of interest to declare.
Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.
References
- von der Maase H, Hansen SW, Roberts JT, et al. Gemcitabine and cisplatin versus methotrexate, vinblastine, doxorubicin, and cisplatin in advanced or metastatic bladder cancer: results of a large, randomized, multinational, multicenter, phase III study. J Clin Oncol 2000;18:3068-77. [Crossref] [PubMed]
- Balar AV, Castellano DE, Grivas P, et al. Efficacy and safety of pembrolizumab in metastatic urothelial carcinoma: results from KEYNOTE-045 and KEYNOTE-052 after up to 5 years of follow-up. Ann Oncol 2023;34:289-99. [Crossref] [PubMed]
- Padua TC, Moschini M, Martini A, et al. Efficacy and toxicity of antibody-drug conjugates in the treatment of metastatic urothelial cancer: A scoping review. Urol Oncol 2022;40:413-23. [Crossref] [PubMed]
- Powles T, Csőszi T, Özgüroğlu M, et al. Pembrolizumab alone or combined with chemotherapy versus chemotherapy as first-line therapy for advanced urothelial carcinoma (KEYNOTE-361): a randomised, open-label, phase 3 trial. Lancet Oncol 2021;22:931-45. [Crossref] [PubMed]
- Galsky MD, Arija JÁA, Bamias A, et al. Atezolizumab with or without chemotherapy in metastatic urothelial cancer (IMvigor130): a multicentre, randomised, placebo-controlled phase 3 trial. Lancet 2020;395:1547-57. [Crossref] [PubMed]
- Powles T, van der Heijden MS, Castellano D, et al. Durvalumab alone and durvalumab plus tremelimumab versus chemotherapy in previously untreated patients with unresectable, locally advanced or metastatic urothelial carcinoma (DANUBE): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol 2020;21:1574-88. [Crossref] [PubMed]
- Powles T, Park SH, Voog E, et al. Avelumab Maintenance Therapy for Advanced or Metastatic Urothelial Carcinoma. N Engl J Med 2020;383:1218-30. [Crossref] [PubMed]
- van der Heijden MS, Sonpavde G, Powles T, et al. Nivolumab plus Gemcitabine-Cisplatin in Advanced Urothelial Carcinoma. N Engl J Med 2023;389:1778-89. [Crossref] [PubMed]
- Powles T, Valderrama BP, Gupta S, et al. Enfortumab Vedotin and Pembrolizumab in Untreated Advanced Urothelial Cancer. N Engl J Med 2024;390:875-88. [Crossref] [PubMed]
- Benjamin DJ, Hsu R. Treatment approaches for FGFR-altered urothelial carcinoma: targeted therapies and immunotherapy. Front Immunol 2023;14:1258388. [Crossref] [PubMed]
- Loriot Y, Matsubara N, Park SH, et al. Erdafitinib or Chemotherapy in Advanced or Metastatic Urothelial Carcinoma. N Engl J Med 2023;389:1961-71. [Crossref] [PubMed]
- Weickhardt AJ, Lau DK, Hodgson-Garms M, 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. [Crossref] [PubMed]
- Moasser MM. The oncogene HER2: its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene 2007;26:6469-87. [Crossref] [PubMed]
- Fleischmann A, Rotzer D, Seiler R, et al. Her2 amplification is significantly more frequent in lymph node metastases from urothelial bladder cancer than in the primary tumours. Eur Urol 2011;60:350-7. [Crossref] [PubMed]
- Hussain M, Daignault S, Agarwal N, et al. A randomized phase 2 trial of gemcitabine/cisplatin with or without cetuximab in patients with advanced urothelial carcinoma. Cancer 2014;120:2684-93. [Crossref] [PubMed]
- Powles T, Huddart RA, Elliott T, et al. Phase III, Double-Blind, Randomized Trial That Compared Maintenance Lapatinib Versus Placebo After First-Line Chemotherapy in Patients With Human Epidermal Growth Factor Receptor 1/2-Positive Metastatic Bladder Cancer. J Clin Oncol 2017;35:48-55. [Crossref] [PubMed]
- Bedard PL, Li S, Wisinski KB, et al. Phase II Study of Afatinib in Patients With Tumors With Human Epidermal Growth Factor Receptor 2-Activating Mutations: Results From the National Cancer Institute-Molecular Analysis for Therapy Choice ECOG-ACRIN Trial (EAY131) Subprotocol EAY131-B. JCO Precis Oncol 2022;6:e2200165. [Crossref] [PubMed]
- Sheng X, Yan X, Wang L, 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. [Crossref] [PubMed]
- Balar AV, Galsky MD, Rosenberg JE, 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. [Crossref] [PubMed]
- Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature 2013;500:415-21. [Crossref] [PubMed]
- Galsky MD, Saci A, Szabo PM, et al. Nivolumab in Patients with Advanced Platinum-resistant Urothelial Carcinoma: Efficacy, Safety, and Biomarker Analyses with Extended Follow-up from CheckMate 275. Clin Cancer Res 2020;26:5120-8. [Crossref] [PubMed]
- Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 2016;387:1909-20. [Crossref] [PubMed]
- Necchi A, Anichini A, Raggi D, et al. Pembrolizumab as Neoadjuvant Therapy Before Radical Cystectomy in Patients With Muscle-Invasive Urothelial Bladder Carcinoma (PURE-01): An Open-Label, Single-Arm, Phase II Study. J Clin Oncol 2018;36:3353-60. [Crossref] [PubMed]
- Teo MY, Bambury RM, Zabor EC, et al. DNA Damage Response and Repair Gene Alterations Are Associated with Improved Survival in Patients with Platinum-Treated Advanced Urothelial Carcinoma. Clin Cancer Res 2017;23:3610-8. [Crossref] [PubMed]
- Rosenberg JE, Park SH, Kozlov V, et al. Durvalumab Plus Olaparib in Previously Untreated, Platinum-Ineligible Patients With Metastatic Urothelial Carcinoma: A Multicenter, Randomized, Phase II Trial (BAYOU). J Clin Oncol 2023;41:43-53. [Crossref] [PubMed]
- Garje R, Vaddepally RK, Zakharia Y. PARP Inhibitors in Prostate and Urothelial Cancers. Front Oncol 2020;10:114. [Crossref] [PubMed]
- Strebhardt K, Ullrich A. Paul Ehrlich's magic bullet concept: 100 years of progress. Nat Rev Cancer 2008;8:473-80. [Crossref] [PubMed]
- Tolcher AW. The Evolution of Antibody-Drug Conjugates: A Positive Inflexion Point. Am Soc Clin Oncol Educ Book 2020;40:1-8. [Crossref] [PubMed]
- Rosenberg J, Sridhar SS, Zhang J, et al. EV-101: A Phase I Study of Single-Agent Enfortumab Vedotin in Patients With Nectin-4-Positive Solid Tumors, Including Metastatic Urothelial Carcinoma. J Clin Oncol 2020;38:1041-9. [Crossref] [PubMed]
- Rosenberg JE, O'Donnell PH, Balar AV, 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. [Crossref] [PubMed]
- Rosenberg JE, Powles T, Sonpavde GP, 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 2023;34:1047-54. [Crossref] [PubMed]
- O'Donnell PH, Milowsky MI, Petrylak DP, 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. [Crossref] [PubMed]
- Powles TB, Perez Valderrama B, Gupta S, et al. LBA6 EV-302/KEYNOTE-A39: Open-label, randomized phase III study of enfortumab vedotin in combination with pembrolizumab (EV+P) vs chemotherapy (Chemo) in previously untreated locally advanced metastatic urothelial carcinoma (la/mUC). Ann Oncol 2023;34:abstr S1340.
- Meric-Bernstam F, Makker V, Oaknin 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. [Crossref] [PubMed]
- Galsky MD, Del Conte G, Foti S, et al. Primary analysis from DS8201-A-U105: A phase 1b, two-part, open-label study of trastuzumab deruxtecan (T-DXd) with nivolumab (nivo) in patients (pts) with HER2-expressing urothelial carcinoma (UC). J Clin Oncol 2022;40:abstr 438.
- Loriot Y, Petrylak DP, Rezazadeh Kalebasty A, et al. TROPHY-U-01, a phase II open-label study of sacituzumab govitecan in patients with metastatic urothelial carcinoma progressing after platinum-based chemotherapy and checkpoint inhibitors: updated safety and efficacy outcomes. Ann Oncol 2024;35:392-401. [Crossref] [PubMed]
- Grivas P, Pouessel D, Park CH, et al. Sacituzumab Govitecan in Combination With Pembrolizumab for Patients With Metastatic Urothelial Cancer That Progressed After Platinum-Based Chemotherapy: TROPHY-U-01 Cohort 3. J Clin Oncol 2024;42:1415-25. [Crossref] [PubMed]
- de Vries EGE, Rüschoff J, Lolkema M, et al. Phase II study (KAMELEON) of single-agent T-DM1 in patients with HER2-positive advanced urothelial bladder cancer or pancreatic cancer/cholangiocarcinoma. Cancer Med 2023;12:12071-83. [Crossref] [PubMed]
- Sheng X, Wang L, He Z, 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. [Crossref] [PubMed]
- Petrylak DP, Eigl BJ, George S, et al. Phase I Dose-Escalation Study of the Safety and Pharmacokinetics of AGS15E Monotherapy in Patients with Metastatic Urothelial Carcinoma. Clin Cancer Res 2024;30:63-73. [Crossref] [PubMed]
- Xu H, Sheng X, Zhou L, et al. A phase II study of RC48-ADC in HER2-negative patients with locally advanced or metastatic urothelial carcinoma. J Clin Oncol 2022;40:abstr 4519.
- Sheng X, Zhou L, Yang K, et al. Disitamab vedotin, a novel humanized anti-HER2 antibody-drug conjugate (ADC), combined with toripalimab in patients with locally advanced or metastatic urothelial carcinoma: An open-label phase 1b/2 study. J Clin Oncol 2023;41:abstr 4566.
- McGregor BA, Sonpavde GP, Kwak L, 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. [Crossref] [PubMed]
- Powles T, Rosenberg JE, Sonpavde GP, et al. Enfortumab Vedotin in Previously Treated Advanced Urothelial Carcinoma. N Engl J Med 2021;384:1125-35. [Crossref] [PubMed]
- Challita-Eid PM, Satpayev D, Yang P, 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. [Crossref] [PubMed]
- Petrylak DP, de Wit R, Chi KN, et al. Ramucirumab plus docetaxel versus placebo plus docetaxel in patients with locally advanced or metastatic urothelial carcinoma after platinum-based therapy (RANGE): a randomised, double-blind, phase 3 trial. Lancet 2017;390:2266-77. [Crossref] [PubMed]
- Trerotola M, Cantanelli P, Guerra E, et al. Upregulation of Trop-2 quantitatively stimulates human cancer growth. Oncogene 2013;32:222-33. [Crossref] [PubMed]
- Stepan LP, Trueblood ES, Hale K, et al. Expression of Trop2 cell surface glycoprotein in normal and tumor tissues: potential implications as a cancer therapeutic target. J Histochem Cytochem 2011;59:701-10. [Crossref] [PubMed]
- Avellini C, Licini C, Lazzarini R, et al. The trophoblast cell surface antigen 2 and miR-125b axis in urothelial bladder cancer. Oncotarget 2017;8:58642-53. [Crossref] [PubMed]
- Morlet T, Rabinowitz MR, Looney LR, et al. A homozygous SLITRK6 nonsense mutation is associated with progressive auditory neuropathy in humans. Laryngoscope 2014;124:E95-103. [Crossref] [PubMed]
- Morrison K, Challita-Eid PM, Raitano A, et al. Development of ASG-15ME, a Novel Antibody-Drug Conjugate Targeting SLITRK6, a New Urothelial Cancer Biomarker. Mol Cancer Ther 2016;15:1301-10. [Crossref] [PubMed]
- Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001;344:783-92. [Crossref] [PubMed]
- Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010;376:687-97. [Crossref] [PubMed]
- Iyer G, Al-Ahmadie H, Schultz N, et al. Prevalence and co-occurrence of actionable genomic alterations in high-grade bladder cancer. J Clin Oncol 2013;31:3133-40. [Crossref] [PubMed]
- Yan M, Schwaederle M, Arguello D, et al. HER2 expression status in diverse cancers: review of results from 37,992 patients. Cancer Metastasis Rev 2015;34:157-64. [Crossref] [PubMed]
- Li BT, Shen R, Buonocore D, et al. Ado-Trastuzumab Emtansine for Patients With HER2-Mutant Lung Cancers: Results From a Phase II Basket Trial. J Clin Oncol 2018;36:2532-7. [Crossref] [PubMed]
- Shi F, Liu Y, Zhou X, et al. Disitamab vedotin: a novel antibody-drug conjugates for cancer therapy. Drug Deliv 2022;29:1335-44. [Crossref] [PubMed]
- Barroso-Sousa R, Tolaney SM. Clinical Development of New Antibody-Drug Conjugates in Breast Cancer: To Infinity and Beyond. BioDrugs 2021;35:159-74. [Crossref] [PubMed]
- Modi S, Jacot W, Yamashita T, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Low Advanced Breast Cancer. N Engl J Med 2022;387:9-20. [Crossref] [PubMed]
- Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med 2005;353:172-87. [Crossref] [PubMed]
- Soria F, Krabbe LM, Todenhöfer T, et al. Molecular markers in bladder cancer. World J Urol 2019;37:31-40. [Crossref] [PubMed]
- Marandino L, Raggi D, Giannatempo P, et al. Erdafitinib for the treatment of urothelial cancer. Expert Rev Anticancer Ther 2019;19:835-46. [Crossref] [PubMed]
- Siefker-Radtke AO, Matsubara N, Park SH, 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. [Crossref] [PubMed]
- Lyou Y, Rosenberg JE, Hoffman-Censits J, et al. Infigratinib in Early-Line and Salvage Therapy for FGFR3-Altered Metastatic Urothelial Carcinoma. Clin Genitourin Cancer 2022;20:35-42. [Crossref] [PubMed]
- Sternberg CN, Petrylak DP, Bellmunt J, 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. [Crossref] [PubMed]
- Powles TB, Chistyakov V, Beliakouski V, et al. LBA27 Erdafitinib (ERDA) or ERDA plus cetrelimab (CET) for patients with metastatic or locally advanced urothelial carcinoma (mUC) and Fibroblast Growth Factor Receptor alterations (FGFRa): First phase (Ph) II results from the NORSE study. Ann Oncol 2021;32:S1303. [Crossref]
- Rosenberg JE, Gajate P, Morales-Barrera R, et al. Safety and efficacy of rogaratinib in combination with atezolizumab in cisplatin-ineligible patients (pts) with locally advanced or metastatic urothelial cancer (UC) and FGFR mRNA overexpression in the phase Ib/II FORT-2 study. J Clin Oncol 2021;39:abstr 4521.
- Apolo AB, Nadal R, Tomita Y, et al. Cabozantinib in patients with platinum-refractory metastatic urothelial carcinoma: an open-label, single-centre, phase 2 trial. Lancet Oncol 2020;21:1099-109. [Crossref] [PubMed]
- Apolo AB, Nadal R, Girardi DM, et al. Phase I Study of Cabozantinib and Nivolumab Alone or With Ipilimumab for Advanced or Metastatic Urothelial Carcinoma and Other Genitourinary Tumors. J Clin Oncol 2020;38:3672-84. [Crossref] [PubMed]
- Girardi DM, Niglio SA, Mortazavi A, et al. Cabozantinib plus Nivolumab Phase I Expansion Study in Patients with Metastatic Urothelial Carcinoma Refractory to Immune Checkpoint Inhibitor Therapy. Clin Cancer Res 2022;28:1353-62. [Crossref] [PubMed]
- Pal SK, Agarwal N, Loriot Y, et al. Cabozantinib in combination with atezolizumab in urothelial carcinoma previously treated with platinum-containing chemotherapy: Results from cohort 2 of the COSMIC-021 study. J Clin Oncol 2020;38:abstr 5013.
- Pal SK, Agarwal N, Singh P, et al. Cabozantinib (C) in combination with atezolizumab (A) in urothelial carcinoma (UC): Results from Cohorts 3, 4, 5 of the COSMIC-021 study. J Clin Oncol 2022;40:abstr 4504.
- Jain RK, Swami U, Bilen MA, et al. Cabozantinib plus pembrolizumab as first-line therapy for cisplatin-ineligible advanced urothelial carcinoma (PemCab). J Clin Oncol 2024;42:abstr 539.
- Msaouel P, Sweis RF, Bupathi M, et al. A Phase 2 Study of Sitravatinib in Combination with Nivolumab in Patients with Advanced or Metastatic Urothelial Carcinoma. Eur Urol Oncol 2024;7:933-43. [Crossref] [PubMed]
- Sadeghi S, Quinn D, Dorff T, et al. EphrinB2 Inhibition and Pembrolizumab in Metastatic Urothelial Carcinoma. J Clin Oncol 2023;41:640-50. [Crossref] [PubMed]
- Xie Y, Su N, Yang J, et al. FGF/FGFR signaling in health and disease. Signal Transduct Target Ther 2020;5:181. [Crossref] [PubMed]
- Turner N, Grose R. Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer 2010;10:116-29. [Crossref] [PubMed]
- Haugsten EM, Wiedlocha A, Olsnes S, et al. Roles of fibroblast growth factor receptors in carcinogenesis. Mol Cancer Res 2010;8:1439-52. [Crossref] [PubMed]
- Knowles MA, Hurst CD. Molecular biology of bladder cancer: new insights into pathogenesis and clinical diversity. Nat Rev Cancer 2015;15:25-41. [Crossref] [PubMed]
- Li Q, Bagrodia A, Cha EK, et al. Prognostic Genetic Signatures in Upper Tract Urothelial Carcinoma. Curr Urol Rep 2016;17:12. [Crossref] [PubMed]
- Loriot Y, Matsubara N, Huddart RA, et al. 2362MO Erdafitinib (erda) vs chemotherapy (chemo) in patients (pts) with advanced or metastatic urothelial cancer (mUC) with select FGFR alterations (FGFRalt): Subgroups from the phase III THOR study. Ann Oncol 2023;34:abstr S1201.
- Elayat G, Punev I, Selim A. An Overview of Angiogenesis in Bladder Cancer. Curr Oncol Rep 2023;25:709-28. [Crossref] [PubMed]
- Organ SL, Tsao MS. An overview of the c-MET signaling pathway. Ther Adv Med Oncol 2011;3:S7-19. [Crossref] [PubMed]
- Kim YW, Yun SJ, Jeong P, et al. The c-MET Network as Novel Prognostic Marker for Predicting Bladder Cancer Patients with an Increased Risk of Developing Aggressive Disease. PLoS One 2015;10:e0134552. [Crossref] [PubMed]
- Matsubara N, de Wit R, Balar AV, et al. Pembrolizumab with or Without Lenvatinib as First-line Therapy for Patients with Advanced Urothelial Carcinoma (LEAP-011): A Phase 3, Randomized, Double-Blind Trial. Eur Urol 2024;85:229-38. [Crossref] [PubMed]
- Choueiri TK, McGregor BA, Shah NJ, et al. A phase 1b study (STELLAR-002) of XL092 administered in combination with nivolumab (NIVO) with or without ipilimumab (IPI) or bempegaldesleukin (BEMPEG) in patients (pts) with advanced solid tumors. J Clin Oncol 2022;40:abstr TPS4600.
- Xia G, Kumar SR, Stein JP, et al. EphB4 receptor tyrosine kinase is expressed in bladder cancer and provides signals for cell survival. Oncogene 2006;25:769-80. [Crossref] [PubMed]
- Robertson AG, Kim J, Al-Ahmadie H, et al. Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer. Cell 2017;171:540-556.e25. [PubMed]
- Teo MY, Seier K, Ostrovnaya I, et al. Alterations in DNA Damage Response and Repair Genes as Potential Marker of Clinical Benefit From PD-1/PD-L1 Blockade in Advanced Urothelial Cancers. J Clin Oncol 2018;36:1685-94. [Crossref] [PubMed]
- Grivas P, Loriot Y, Morales-Barrera R, et al. Efficacy and safety of rucaparib in previously treated, locally advanced or metastatic urothelial carcinoma from a phase 2, open-label trial (ATLAS). BMC Cancer 2021;21:593. [Crossref] [PubMed]