Fibroblast growth factor receptor 3 (FGFR3) is frequently activated by mutation or overexpression, and it is a validated therapeutic target in urothelial carcinoma (UC) of the bladder. However, the role and detailed molecular mechanism of FGFR3 in the immune microenvironment of bladder cancer remain largely unknown. Here, we demonstrate that inhibition of FGFR3 in FGFR3-activated bladder cancer elevates PD-L1 protein levels by affecting its ubiquitination, thereby inhibiting the antitumor activity of CD8+ T cells. Tissue microarray analysis in human UC showed an inverse correlation between FGFR3 and PD-L1. Furthermore, NEDD4, an E3 ubiquitin ligase of the NEDD4 family of proteins, was phosphorylated by FGFR3 activation and served as a regulator of PD-L1 ubiquitination. Mechanistically, NEDD4 interacted with PD-L1 and catalyzed Lys48 (K48)-linked polyubiquitination of PD-L1. In mice bearing NEDD4 knockout bladder cancer, CD8+ T-cell infiltration and antitumor activity were significantly inhibited due to PD-L1 upregulation in bladder cancer cells. Furthermore, multiple FGFR3-activated tumor-bearing mouse models suggested that attenuated CD8+ T-cell–mediated antitumor efficacy following FGFR3-targeted therapy could be rescued by a combination with anti-PD-1 immunotherapy, which leads to effective tumor suppression. This study establishes a key molecular link between targeted therapy and immune surveillance and identifies NEDD4 as a crucial E3 ubiquitin ligase that targets PD-L1 for degradation in FGFR3-activated bladder cancer. These findings may potentially be exploited for combination therapies in UC of the bladder and possibly other malignancies with activated FGFR3.

Significance:

NEDD4 links two important molecules associated with targeted therapy and immune surveillance, providing mechanistic rationale and preclinical support for immuno-targeted combination therapy for FGFR3-activated bladder cancer.

Bladder cancer is one of the most prevalent cancers, with nearly 573,278 new cases and 212,536 deaths annually on a global scale (1). The relative 5-year overall survival rate of advanced-stage, unresectable or metastatic urothelial carcinoma (UC) of the bladder is low (2). Using historical treatment with second-line chemotherapy, the response rate is approximately 10% to 12% and the median overall survival is approximately 7 to 9 months (3–5). FGFR3 mutations, which have a driver role in carcinogenesis, are very common in UC of the bladder and associated with a low immune signature, which is in accordance with the Cancer Genome Atlas (TCGA) luminal-papillary subtype (6–9).

FGFR3 is an actionable target in UC of the bladder, and erdafitinib, a pan-FGFR inhibitor, is the only targeted drug approved by the FDA for bladder cancer treatment. Although the efficacy of erdafitinib in UC of the bladder with prespecified FGFR alterations is up to 40% (the efficacy of immune checkpoint therapy is only 5%), which means there are still more than half of the patients who do not benefit from these new therapies and end up with conventional treatments including chemotherapy and radiotherapy that are less effective with more side effects (10). The UC of the bladder with FGFR alterations that fail to benefit from erdafitinib are usually more aggressive, and the gloomy scenario is that currently no additional work has explored the underlying mechanism.

Currently, targeting immune checkpoints such as PD-1 and PD-L1 has been approved for treating advanced bladder cancer with durable clinical benefit in a subset of patients (3, 11). Because there is possibly no potential cross-resistance between FGFR inhibitors and ICB, the combination strategy has been investigated (12). Recent studies in advanced UC of the bladder showed that the relevance between the mutation status of FGFR3 and the response to PD-1/PD-L1 inhibitors was controversial (13, 14), and the therapeutic effects of immuno-targeted combined therapy were indefinite. Moreover, it is not yet clear which kind of patients may benefit from the combination treatment, and the potential molecular mechanisms are still obscure.

Most studies have demonstrated that the response to anti-PD-1/PD-L1 treatment correlate with PD-L1 expression levels in UC of the bladder, in which high PD-L1 levels usually mean good response to anti-PD-1/PD-L1 treatment (11). Recent studies have revealed a series of mechanisms of PD-L1 regulation by transcriptional and posttranscriptional means in different cancers (15, 16), but the particularity of PD-L1 regulation in bladder cancer has hardly been well elucidated.

Neuronal precursor cell expressed developmentally downregulated 4 (NEDD4) is a HECT family E3 ubiquitin ligase, which functions in substrate recognition and attachment of ubiquitin to substrates (17, 18). The ubiquitin ligase activity of NEDD4 can be promoted by FGFR1 and EGFR activation, which results in the activation of c-Src (19). However, the immunologic regulatory function of NEDD4 remains largely unknown. Here, we demonstrated that NEDD4 could be phosphorylated by FGFR3 activation and functioned as a regulator of PD-L1 ubiquitination to control CD8+ T-cell–mediated immune surveillance in UC of the bladder. The mechanism concerning FGFR3 and PD-L1 links two important molecules and uncovers a novel basis for the application of FGFR3 inhibitors and ICB in UC of the bladder with FGFR3 activation.

Mice

Six- to eight-week-old female C57BL/6 mice were purchased from the Laboratory Animal Center of Shandong University. Six-week-old female BALB/c nude mice (CAnN.Cg-Foxn1nu/Crl) and 4- to 6-week-old Cg-Prkdcscid Il2rgtm1Sug/JicCrl (NOG) mice were purchased from Vital River Laboratory Animal Technology. All mouse strains were maintained under specific pathogen-free conditions. All animal experimental procedures in the study were approved by the Ethics Committee of Qilu Hospital of Shandong University (No. KYLL-2020-264).

Cell culture and drug treatment

Six bladder cancer cell lines (SW780, RT4, MB49, T24, 5637, and UM-UC-3), 293T, SV-HUC-1, SK-NM-C, and KMS-11 were used in this study. The T24, 5637, SW780, RT4, UM-UC-3, 293T, and SK-NM-C cell lines were purchased from ATCC, the MB49 cell line was purchased from Millipore, and the KMS-11 was purchased from JCRB. All cells were cultured in the media supplemented with 10% FBS (Corning), 100 U/mL penicillin and streptomycin (Gibco). All cells used in this study were authenticated by short tandem repeat analysis within 2 years. All newly revived cells were tested free of Mycoplasma contamination by the Mycoalert Detection Kit (Beyotime). The cumulative culture length of the cells between thawing and used in this study was less than 10 passages.

Orthotopic tumor model

The orthotopic tumor model was established by surgical implantation. In short, all the mice were anesthetized with isoflurane (2.5%) and injected with fLuc-MB49 cells or fLuc-SW780 cells (5 × 105) into the bladder using insulin syringes. Several days after implantation, the mice were randomized into different groups according to the tumor burden and administered regularly. When the tumor size reached a threshold, the tumor-bearing mice were sacrificed and the bladder tumors were dissected for further experiments. For in vivo experiments, erdafitinib and infigratinib were formulated in 20% hydroxypropyl-β-cyclodextrin (HP-β-CD) and orally administered at a dose of 12.5 mg/kg twice daily. Anti-PD-1 antibodies used in animal experiments were given by intraperitoneal injection with a dose of 10 mg/kg every 3 days.

Next-generation sequencing

Genomic DNA (gDNA) was extracted from patients' peripheral blood samples and paired frozen tissues or formalin-fixed paraffin embedded sections using a DNeasy Blood & Tissue Kit (Qiagen, 69504) according to the manufacturer's instructions. Raw sequencing data were aligned to the reference human genome (UCSC hg19) through Burrows–Wheeler Aligner. Somatic variants were generated in tumor samples by removing the germline alterations from the matched peripheral blood mononuclear cell gDNA to keep the variants unique to the tumor. Variants were annotated using the ANNOVAR software. A CNV Kit was used to determine the copy-number variations (CNV; https://github.com/etal/cnvkit).The tumor mutation burden of each sample was calculated according to a published and widely used method described by Chalmers and colleagues (20).

Patients

All patients or healthy donors involved in the study fully understood and provided informed consent, and all the procedures involved were approved by the Ethics Committee of the Qilu Hospital of Shandong University (No. KYLL-2019-168) and Jinan Maternity and Child Care Hospital (No. KYLL-2020–275). The PDX samples were obtained from surgical resection from an elderly male patient with high-grade UC of the bladder that had FGFR3-TACC3 gene fusion detected by next-generation sequencing (NGS). Human peripheral blood samples used for CD8+ T-cell isolation were obtained from patients with bladder cancer prior to surgery. Human umbilical cord blood samples used for CD34+ hematopoietic stem cell (HSC) extraction were obtained from consenting donors at Jinan Maternity and Child Care Hospital.

Patient-derived xenograft model establishment

For humanized immune system construction, female NOG mice underwent whole-body irradiation (2.0 Gy), and then huCD34+ HSCs (1 × 105 HSCs per mouse) were injected into the tail vein of irradiated NOG mice. After 8 weeks, the HSCs differentiated into the appropriate immune cells, which were monitored by peripheral blood flow cytometry analysis. Fresh bladder cancer specimens were cut into small pieces (3 × 3 × 3 mm3) and implanted subcutaneously into the flanks of 4- to 6-week-old female NOG mice. After the tumor mass expanded to over quintuple its volume, the xenograft tumor was resected and directly retransplanted for expansion in later serial generations using the same procedure. An orthotopic bladder cancer patient-derived xenograft (PDX) model was established via implantation of a single cell suspension from subcutaneous PDXs into the bladder of mouse with humanized immune system. After the PDX model was established, mice were administered regularly.

Imaging procedure

For bioluminescence living imaging, the anesthetized mice were injected intraperitoneally with d-luciferin (150 mg/kg) and imaged 15 minutes later. Luminescence was detected by an IVIS Lumina system (IVIS Spectrum, PerkinElmer), and image analysis was performed using Living Image Software.

For ultrasound imaging, mouse bladders were catheterized using venous catheters coated with liquid paraffin. Then, phosphate-buffered saline (PBS) was delivered into the lumen using a 1 mL sterile syringe to distend the bladder for imaging. The images were acquired using a Vevo LAZR high-resolution ultrasound system (VisualSonics) with a transducer probe in the B-mode setting.

shRNAs, siRNAs, and CRISPR/Cas9 knockout

Short hairpin RNA (shRNA) constructs targeting FGFR1, FGFR2, FGFR3, and FGFR4 were purchased from GeneChem Co., Ltd., and small-interfering RNAs (siRNA) targeting WWP1, ITCH, MDM2, CBL, and NEDD4 were purchased from Gene Pharma Co., Ltd. Generally, to generate cell lines that stably expressed the targeted genes using shRNAs, cells were seeded into six-well plates at a density of 2 × 105 cells per well. After incubation for 24 hours, cells were transfected with lentiviral particles. Then the transfected cells were selected using puromycin at indicated concentrations before use. For siRNA transfection, the seeded cells were transfected with siRNAs according to the standard protocol. The cells were used in the experiments after 48 hours of transfection. NEDD4 and PD-L1 were knocked out by the CRISPR/Cas9 gene editing system. After transfection, the cells were selected with puromycin and diluted to generate single-cell colonies in a 96-well plate. The transfection efficiency was measured by Western blot analysis.

Cell viability and proliferation assay

Cells were seeded in plates with different treatment. At indicated time points, cell viability was monitored using sulforhodamine B (SRB) assay protocol. The cell viability was calculated (formula = mean ODtreatment / mean ODcontrol × 100%) and the time–response curve was plotted to clarify the antiproliferation effect of the inhibitors (21). For crystal violet staining assay, cells were fixed and stained with 0.5% crystal violet solution, and representative images were acquired using a microscope with a digital camera. For quantitative determination, the stained cells were dissolved in methanol, and the absorbance was measured at 570 nm by a microplate reader (22).

T-cell culture and T-cell–mediated tumor cell killing assay

Human T cells were obtained from human peripheral blood samples, and mouse T cells were derived from mouse spleens. Briefly, human peripheral blood mononuclear cells were isolated from human peripheral blood by Ficoll density gradient centrifugation. Enrichment of CD8+ T cells was performed by magnetic cell sorting (MACS) using the MagCellect Human CD8+ T Cell Isolation Kit (R&D Systems) and MagCellect Mouse CD8+ T Cell Isolation Kit (R&D Systems) following the manufacturer's instructions. The medium used for human T-cell culture was RPMI1640 supplemented with 10% FBS, 100 U/mL penicillin and streptomycin, 100 IU/mL recombinant human IL2, 2 μg/mL antihuman CD3 antibody, and 1 μg/mL antihuman CD28 antibody. Splenic T cells were cultured in complete RPMI1640 medium in the presence of 2 μg/mL antimouse CD3 antibody and 1 μg/mL antimouse CD28 antibody. After stimulation for 5 days, CD8+ T cells were harvested and cocultured with different targeted cells with physical contact. A schematic overview of the procedure is showed in Fig. 3A.

Flow cytometry

Briefly, after different treatments for adequate time periods, cells were collected for further examination. Tumors from the mouse model were dissected out and disrupted to a single-cell suspension by type IV collagenase and DNase.

PD-L1 level determination

For the analysis of PD-L1 levels, cells were harvested and stained with human or mouse anti-PD-L1 antibodies and run on a cytometer for analysis.

Analysis of in vitro T-cell proliferation and activation

To assess the function of CD8+ T cells after treatment, cells were harvested and fixed with 4% paraformaldehyde at 4°C for 1 hour. For permeabilization, the fixed cells were washed twice with PBS and resuspended in 500 μL of Triton X-100 for 15 minutes. Then, the cells were washed and stained with Ki67, TNFα, granzyme B, and perforin antibodies for 30 minutes at 37°C. Fluorescence intensity was analyzed by flow cytometry.

Tumor infiltrating T-cell analysis

In the early stage of treatment, the bladder tumors were removed for tumor infiltrating T-cell analysis. The collected single-cell suspension was stained with the indicated antibodies, then analyzed by flow cytometry. In response to various specific analyses of T-cell activity, CD8+ T cells were sorted out using a magcellect mouse or human CD8+ T Cell Isolation Kit for further analysis.

The above data analysis was carried out using FlowJo or Cytoexpert software. Antibodies used can be found in Supplementary Table S1.

Histology

For hematoxylin and eosin staining, the mice were sacrificed at the endpoint, and the tumor tissue was carefully removed. After fixation with 4% paraformaldehyde, the tissue was embedded and stained using a standard protocol. Images were acquired using a VS120 Olympus microscope with OlyVIA software (Olympus).

Immunofluorescence

Cells were plated on glass coverslips and treated differently. After fixation, the slides were blocked with immunofluorescence blocking fluid and incubated with the primary antibody overnight at 4°C. After washing, the corresponding fluorescent secondary antibodies were added to label the primary antibodies. DAPI was used to stain the nuclear structure. For tumor tissue immunofluorescence analysis, paraffin sections (4 μm) were deparaffinized in xylene and dehydrated in graded ethanol, followed by boiling with 10 mmol/L citrate antigen retrieval solution (Beyotime). Then, the specimens were blocked in goat serum and incubated with primary antibodies at 4°C overnight. After rinsing, the slides were incubated with secondary antibodies at room temperature for 1 hour. Following several washes, the slides were stained with DAPI reagent and mounted with antifade solution. Immunofluorescence was imaged using a laser confocal microscope and analyzed with ZEN Imaging Software by ZEISS.

IHC

IHC was performed following the standard protocol: after xylene and graded ethanol treatment, the sections were subjected to antigen retrieval by boiling in citrate buffer. Then the sections were blocked and incubated with the primary antibodies in a humidified chamber overnight. Immunodetection was visualized with diaminobenzidine (DAB), and the sections were counterstained with hematoxylin. Images were acquired using the scanner described above.

RT-PCR analysis

For quantification of PD-L1 mRNA expression, total RNA of treated cells was extracted using TRizol reagent according to the protocol, and complementary DNAs (cDNA) were synthesized with a cDNA Synthesis Kit. qPCR reaction was conducted with SYBR Green master mix using a fluorescence ratio PCR instrument (Bio-Rad, CFX Connect). The fold change in PD-L1 mRNA expression between different groups was determined by the 2−ΔΔCt method. The PD-L1 primers were as follows: human PD-L1 forward 5′-ATTTGGAGGATGTGCCAGAG-3′ and reverse 5′-CCAGCACACTGAGAATCAACA-3′; human actin forward 5′-GCAAAGACCTGTACGCCAACA-3′ and reverse 5′-TGCATCCTGTCGGCAATG-3′; mouse PD-L1 forward 5′-GCATTATATTCACAGCCTGC-3′ and reverse 5′-CCCTTCAAAAGCTGGTCCTT-3′; mouse actin forward 5′-ACCTTCTACAATGAGCTGCG-3′ and reverse 5′-CTGGATGGCTACGTACATGG-3′.

Coimmunoprecipitation and immunoblotting assay

For coimmunoprecipitation assay, the transfected and treated cells were scraped and lysed in IP buffer containing protease and phosphatase inhibitor cocktails on ice, followed by centrifugation at 14,000 × g for 10 minutes at 4°C to remove insoluble materials. The supernatant of samples was incubated with the protein A/G beads with the indicated antibodies overnight at 4°C. After washing with IP buffer, the beads were eluted by boiling with SDS buffer. The coimmunoprecipitations were detected by immunoblotting with the indicated antibodies (Supplementary Table S1).

For immunoblotting, the protein lysate was run on an SDS-PAGE gel and transferred to a polyvinylidene difluoride membrane. After blocking with nonfat milk, the membranes were incubated with specific primary antibodies at 4°C overnight. After washing, the membranes were stained with HRP-conjugated secondary antibodies for 1 hour at 25°C. Protein band detection was performed with ECL Detection Kits (EMD Millipore), and images were obtained using a chemiluminescence apparatus. Quantification of the band intensity was analyzed using ImageJ software.

Statistical analysis

Statistical differences between groups were determined using two tailed Student t test, one-way ANOVA, and two-way ANOVA. Kaplan–Meier survival curves were used and analyzed by the log-rank (Mantel–Cox) test in mouse experiments using GraphPad Prism Software. The P value in the figure legend indicates *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001 (*, P < 0.05 was regarded as statistically significant).

Data availability statement

The data generated in this study are available upon request from the corresponding author.

FGFR inhibitors attenuate tumor growth and upregulate PD-L1 expression in bladder cancer mouse model

To determine whether the immune system influenced the efficacy of FGFR inhibition, we inoculated MB49 cells into immunocompetent (C57BL/6) and immunodeficient (CAnN.Cg-Foxn1nu/Crl) mouse bladders (Fig. 1A and B). Interestingly, drug intervention with erdafitinib or infigratinib inhibited tumor progression in immunocompetent mice (the inhibition rate was approximately 50%) but exerted a better suppression effect in immunodeficient mice (the inhibition rate was approximately 75%; Fig. 1CJ). These findings suggest the existing relevance between the immune system and the therapeutic effect of FGFR inhibition. We also evaluated the stability of the therapeutic effect, and found that the erdafitinib and infigratinib had persistent tumor suppression effect (Supplementary Figs. S1A–S1F). Moreover, ultrasound imaging and HE staining of tumor tissue further confirmed the tumor inhibition effect of erdafitinib and infigratinib in tumor-bearing mice (Fig. 1K and L). Both erdafitinib and infigratinib administration conspicuously decreased Ki67 and inhibited the downstream FGFR signaling pathways in nude mice, including phosphorylation of ERK1/2, AKT, and MEK. (Fig. 1K and L; Supplementary Fig. S1G; refs. 23, 24).

Figure 1.

Erdafitinib and infigratinib inhibit bladder tumor growth and induce T-cell suppression accompanied with PD-L1 upregulation in mice. A and B, The overview of experimental plan in Luci+ MB49 tumor-bearing C57BL/6 (A) and nude (B) mice model. C and D, Bioluminescence images of Luci+ MB49 tumor-bearing mice model in different groups. E and F, Quantification of bioluminescence intensity (mean ± SD, n = 5). G and H, Macroscopic images of excised bladder tumors. Scale bars, 1 cm. I and J, Tumor weight after erdafitinib and infigratinib treatment (mean ± SD, n = 5). K and L, Ultrasound images of orthotopic bladder tumors (red dash lines), hematoxylin and eosin (H&E)–stained sections of tumors, and immunofluorescence images of the indicated markers for Ki67 (red), cytokine 7 (green), and DAPI (blue) in C57BL/6 and nude mice. Scale bars in ultrasound images, 2 mm, 500 μm for hematoxylin and eosin staining, and 20 μm for immunofluorescence images. M, PD-L1 t-SNE plots of gated flow cytometry data generated by cells in tumor tissue from erdafitinib and infigratinib treatment. Numbers on the right edge represent the proportion of PD-L1 positive cells. N, Plot showing the proportion of PD-L1 positive cells (mean ± SD, n = 3). O, Intratumoral CD3+CD8+ T-cell proportions were analyzed by flow cytometry. P, Plot showing the proportion of tumor-infiltrating CD3+CD8+ T cells in tumor-bearing mice with different treatment (mean ± SD, n = 3). Q, Flow cytometry analysis showing the granzyme B–positive ratio of CD8+ T cells in tumor (mean ± SD, n = 3). R, Statistical diagram showing the granzyme B–positive ratio in CD8+ T cells (left) and granzyme B+ CD8+ T cells (right) in tumor (mean ± SD, n = 3). S, Flow cytometry analysis showing the TNFα-positive ratio of CD8+ T cells in tumor (mean ± SD, n = 3). T, Statistical diagram showing the TNFα-positive ratio of CD3+CD8+ T cells (left) and TNFα+ CD8+ T cells (right) in tumor (mean ± SD, n = 3). **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; one-way ANOVA.

Figure 1.

Erdafitinib and infigratinib inhibit bladder tumor growth and induce T-cell suppression accompanied with PD-L1 upregulation in mice. A and B, The overview of experimental plan in Luci+ MB49 tumor-bearing C57BL/6 (A) and nude (B) mice model. C and D, Bioluminescence images of Luci+ MB49 tumor-bearing mice model in different groups. E and F, Quantification of bioluminescence intensity (mean ± SD, n = 5). G and H, Macroscopic images of excised bladder tumors. Scale bars, 1 cm. I and J, Tumor weight after erdafitinib and infigratinib treatment (mean ± SD, n = 5). K and L, Ultrasound images of orthotopic bladder tumors (red dash lines), hematoxylin and eosin (H&E)–stained sections of tumors, and immunofluorescence images of the indicated markers for Ki67 (red), cytokine 7 (green), and DAPI (blue) in C57BL/6 and nude mice. Scale bars in ultrasound images, 2 mm, 500 μm for hematoxylin and eosin staining, and 20 μm for immunofluorescence images. M, PD-L1 t-SNE plots of gated flow cytometry data generated by cells in tumor tissue from erdafitinib and infigratinib treatment. Numbers on the right edge represent the proportion of PD-L1 positive cells. N, Plot showing the proportion of PD-L1 positive cells (mean ± SD, n = 3). O, Intratumoral CD3+CD8+ T-cell proportions were analyzed by flow cytometry. P, Plot showing the proportion of tumor-infiltrating CD3+CD8+ T cells in tumor-bearing mice with different treatment (mean ± SD, n = 3). Q, Flow cytometry analysis showing the granzyme B–positive ratio of CD8+ T cells in tumor (mean ± SD, n = 3). R, Statistical diagram showing the granzyme B–positive ratio in CD8+ T cells (left) and granzyme B+ CD8+ T cells (right) in tumor (mean ± SD, n = 3). S, Flow cytometry analysis showing the TNFα-positive ratio of CD8+ T cells in tumor (mean ± SD, n = 3). T, Statistical diagram showing the TNFα-positive ratio of CD3+CD8+ T cells (left) and TNFα+ CD8+ T cells (right) in tumor (mean ± SD, n = 3). **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; one-way ANOVA.

Close modal

To better understand the relationship between FGFR inhibition and tumor immunology, we detected the level of PD-L1 and the proportion of CD8+ T cells in tumors treated with erdafitinib and infigratinib by flow cytometry. Unexpectedly, the PD-L1-positive tumor cell ratio increased substantially in the tumors treated with erdafitinib or infigratinib, accompanied by a significant decrease in CD8+ T-cell infiltration in tumor tissues (Fig. 1MP). TNFα and granzyme B were reported to be acknowledged markers for CD8+ T cells activation (25, 26). We found that erdafitinib and infigratinib also decreased the proportion of granzyme B and TNFα positive CD8+ T cells in MB49 tumors (Fig. 1QT). We further evaluated the immune checkpoint PD-L2 expression level of tumors in different groups, as well as PD-1, Tim-3, and LAG-3 expression levels on CD8+ T cells, the results showed that erdafitinib and infigratinib made no obvious effect on these markers in vivo (Supplementary Figs. S2A–S2E). We also detected the proportion of immunosuppressive cells including MDSCs and Tregs in tumors treated with erdafitinib and infigratinib, and no differences were displayed from the results (Supplementary Figs. S2F and S2G). Furthermore, genetic deletion of PD-L1 further improves tumor suppression efficacy of erdafitinib and infigratinib (Supplementary Figs. S2H–S2L). Together, our results in bladder cancer mouse model with different immunologic functions led us to discover that the tumor inhibition effect of FGFR inhibitors was accompanied by PD-L1 upregulation and CD8+ T-cell inhibition.

FGFR3 activation correlates with PD-L1 downregulation in UC of the bladder

FGFR inhibition-mediated PD-L1 upregulation was further verified in multiple bladder cancer cell lines. Strikingly, incubation with erdafitinib and infigratinib induced approximately 40% proliferation inhibition of SW780, RT4, and MB49 cells in which FGFR3 was activated by FGFR3 mutations or high expression (27, 28, 29). In contrast, erdafitinib or infigratinib only induced approximately 20% proliferation inhibition of T24, 5637, and UM-UC-3 cells in which FGFRs were normal (Supplementary Fig. S3A). In addition, cell apoptosis and cell-cycle assessment using flow cytometry revealed that erdafitinib and infigratinib arrested the cell cycle in the G0–G1 stage but did not induce cell apoptosis (Supplementary Figs. S3B and S3C). Interestingly, bladder cancer cell PD-L1 levels were significantly upregulated in line with the strong proliferation inhibition rates in SW780, RT4, and MB49 cells with erdafitinib or infigratinib treatment, in contrast to PD-L1 expression levels in T24, 5637, and UM-UC-3 cells, which have normal FGFR levels, that did not increase obviously (Fig. 2A and B; Supplementary Figs. S4A and S4B). The results also showed that erdafitinib or infigratinib could not affect PD-L1 expression on SV-HUC-1 human normal bladder epithelium cell line (Supplementary Figs. S4A and S4B). Because erdafitinib and infigratinib inhibit several FGFRs, including FGFR1–4, it is of interest to identify which FGFR is involved in the process of FGFR inhibition-mediated PD-L1 upregulation. To this end, using an RNA interference screen of FGFR1–4, we identified FGFR3 as a crucial regulator of PD-L1 in bladder cancer cells. The results revealed that FGFR3 interference could upregulate PD-L1 expression to the greatest extent in SW780 and MB49 bladder cancer cells (Fig. 2C and D; Supplementary Figs. S4C and S4D). Fluorescence confocal analysis showed more obvious results that FGFR3 interference could also inhibit FGFR3 phosphorylation and increase PD-L1 levels in human and mouse bladder cancer cells (Fig. 2E). Furthermore, both erdafitinib and infigratinib upregulated PD-L1 in SW780 and MB49 cells by inhibiting the phosphorylation of FGFR3 (Fig. 2F). In addition, we also verified the basal level of FGFR family proteins and PD-L1 in the panel of multiple bladder cancer cell lines used in this study and we found that SW780, RT4, and MB49 cells had significantly high FGFR3 phosphorylation levels accompanied with relatively low PD-L1 expression (Supplementary Figs. S4E and S4F). Together, our findings offer compelling evidence that FGFR3 plays a critical role in PD-L1 regulation among bladder cancer cells with FGFR3 activation. To verify this phenomenon in other tumors, we selected another two cancer cell lines with FGFR3 activating mutations including Ewing sarcoma cell line SK-N-MC and multiple myelomas cell line KMS-11 (30, 31). The results showed that erdafitinib and infigratinib induced moderate PD-L1 expression in SK-N-MC and KMS-11 cells (Supplementary Fig. S4G). These results provide further evidence that the phenomenon we found in bladder cancer has the potential to be applied to certain tumors. In clinical practice, both chemotherapy and targeted therapy are commonly used in bladder cancer treatment. We found that both cisplatin and paclitaxel could elevate the PD-L1 expression in MB49 and T24 cells (Supplementary Fig. S4H).

Figure 2.

FGFR3 downregulates PD-L1 expression in UC of the bladder in vitro and in vivo. A, Flow cytometry analysis and statistical plots of PD-L1 expression in SW780, RT4, and MB49 cells treated with erdafitinib and infigratinib (mean ± SD, n = 3). B, Western blot representative images show the PD-L1 expression levels in SW780, RT4, MB49 cells with erdafitinib and infigratinib treatment. Actin was used as a control. C, Flow cytometric images and the quantification diagrams of PD-L1 expression levels of SW780 cells after FGFR1–4 knockdown by shRNAs in comparison with nonsilencing and nontargeted control (mean ± SD, n = 3). D, Western blot representative images show the PD-L1 and FGFRs expression in SW780 cells that were treated with FGFR shRNAs. NS, nonsilencing shRNA. E and F, Immunofluorescence microscopy images for p-FGFR3 (red), PD-L1 (green), and DAPI (blue) of SW780 and MB49 cells with two kinds of FGFR3 shRNA treatment (E) or erdafitinib and infigratinib treatment (F). Scale bars, 20 μm. G, Schematic of FGFR3 protein, representative hematoxylin and eosin (H&E) staining, and IHC staining images for PD-L1 expression in WT or FGFR3-mutant UC of the bladder patients. P value was calculated by Pearson χ2; −/+, negative or low expression; ++/+++, medium or high expression. Scale bars, 200 μm in low power field and 100 μm in high power field. H, Tumor stage analysis between the patients with bladder cancer with/without FGFR3 mutation. Data from cBioPortal. ***, P < 0.001; ****, P < 0.0001; one-way ANOVA.

Figure 2.

FGFR3 downregulates PD-L1 expression in UC of the bladder in vitro and in vivo. A, Flow cytometry analysis and statistical plots of PD-L1 expression in SW780, RT4, and MB49 cells treated with erdafitinib and infigratinib (mean ± SD, n = 3). B, Western blot representative images show the PD-L1 expression levels in SW780, RT4, MB49 cells with erdafitinib and infigratinib treatment. Actin was used as a control. C, Flow cytometric images and the quantification diagrams of PD-L1 expression levels of SW780 cells after FGFR1–4 knockdown by shRNAs in comparison with nonsilencing and nontargeted control (mean ± SD, n = 3). D, Western blot representative images show the PD-L1 and FGFRs expression in SW780 cells that were treated with FGFR shRNAs. NS, nonsilencing shRNA. E and F, Immunofluorescence microscopy images for p-FGFR3 (red), PD-L1 (green), and DAPI (blue) of SW780 and MB49 cells with two kinds of FGFR3 shRNA treatment (E) or erdafitinib and infigratinib treatment (F). Scale bars, 20 μm. G, Schematic of FGFR3 protein, representative hematoxylin and eosin (H&E) staining, and IHC staining images for PD-L1 expression in WT or FGFR3-mutant UC of the bladder patients. P value was calculated by Pearson χ2; −/+, negative or low expression; ++/+++, medium or high expression. Scale bars, 200 μm in low power field and 100 μm in high power field. H, Tumor stage analysis between the patients with bladder cancer with/without FGFR3 mutation. Data from cBioPortal. ***, P < 0.001; ****, P < 0.0001; one-way ANOVA.

Close modal

To further validate our findings in clinical UC of bladder specimens, we analyzed the correlations between FGFR3 activating mutations and PD-L1 expression in 98 human UC bladder specimens using gene sequencing and immunohistochemical staining (Fig. 2G; Supplementary Table S2). We found that high PD-L1 expression was detected in 20 (37.7%) of the 53 specimens with wild-type FGFR3 or nonactivating mutations but in only 2 (4.4%) of the 45 specimens with FGFR3 activating mutations (Fig. 2G). These findings supported the concept that FGFR3 activation associated with PD-L1 downregulation had a reliable clinical correlation. A previous study among 1,000 chemotherapy-naive radical cystectomy patients showed that FGFR3 mutations significantly prolonged cancer-specific survival (7). Most less aggressive cases of nonmuscle-invasive bladder cancer are characterized by activating mutations in FGFR3 (32). In addition, the luminal-papillary subtype of UC, which has a higher percentage of alteration in FGFR3 genes, has better prognosis than basal subtype (13). Similarly, we found that FGFR3 mutations were also associated with low-grade and early-stage disease in 476 patients with bladder cancer from the TCGA database (Fig. 2H). These clinical data supported the notion that FGFR3 activating mutation was consistent with low PD-L1 expression and relatively good cancer prognosis.

CD8+ T-cell antitumor activity is inhibited by PD-L1 upregulation in bladder cancer cells with FGFR3 suppression

To our knowledge, the effect of checkpoint inhibition therapy lies in its ability to potentiate T-cell–centered cancer cell immune destruction. Activated T cells can play a powerful role in tumor killing in vivo and in vitro (33). On the basis of this theory, we used cocultivation experiments with bladder cancer cells to evaluate the effects of FGFR3 inhibition or silencing on T-cell responses. We isolated and stimulated human CD8+ T cells from peripheral blood and mouse CD8+ T cells from splenocytes to coculture with bladder cancer cells (Fig. 3A). Consistent with the model in which FGFR3 inhibition by inhibitors and knockdown by shRNA upregulated PD-L1 protein levels in bladder cancer cells, the cocultivation results revealed that PD-L1 upregulation in bladder cancer cells largely protected them from being killed by T cells (Fig. 3B and C). In line with bladder cancer cell resistance to the killing effect of T cells, flow cytometry analysis revealed that the killing effect of T cells was seriously inhibited by upregulation of PD-L1 in tumor cells. Moreover, cocultivation with FGFR3-inhibited bladder cancer cells significantly reduced the proportion of activated CD8+ T cells indicated by Ki67, TNFα, granzyme B, and perforin positive ratios (Fig. 3DG).

Figure 3.

FGFR3 inhibition or silence in bladder cancer cells suppresses T-cell responses. A, Illustration shows the in vitro procedure of human and mouse CD8+ T cells isolation and cocultivation with cancer cells. B, Results from the proliferation assay of SW780 (left) and MB49 (right) cells, which were grown in the presence of indicated drugs with/without T cells. DMSO was used as vehicle control and the homogenized absorbance is shown in each panel. C, Results from the proliferation assay of SW780 (left) and MB49 (right) cells with FGFR3 knocked down in the presence of T cells. Nonsilencing shRNA was used as negative control. D, Flow cytometry plots and graphs show Ki67 (left) and TNFα (right) expression of human T cells that were cocultured with SW780 cells in the presence of erdafitinib and infigratinib treatment (mean ± SD, n = 3). E, Flow cytometry plots and graphs show granzyme B (left) and perforin (right) expression of human T cells that were cocultured with SW780 cells in the presence of erdafitinib and infigratinib treatment (mean ± SD, n = 3). F, Flow cytometry plots and graphs show Ki67 (left) and TNFα (right) expression of human T cells that were cocultured with SW780 cells treated with FGFR3 shRNA (mean ± SD, n = 3). G, Flow cytometry plots and graphs show granzyme B (left) and perforin (right) expression of human T cells that cocultured with SW780 cells treated with FGFR3 shRNAs. Nonsilencing shRNA was used as negative control (mean ± SD, n = 3). n.s., not significant; ***, P < 0.001; ****, P < 0.0001; one-way ANOVA. PBMC, peripheral blood mononuclear cells. NS, nonsilencing shRNA.

Figure 3.

FGFR3 inhibition or silence in bladder cancer cells suppresses T-cell responses. A, Illustration shows the in vitro procedure of human and mouse CD8+ T cells isolation and cocultivation with cancer cells. B, Results from the proliferation assay of SW780 (left) and MB49 (right) cells, which were grown in the presence of indicated drugs with/without T cells. DMSO was used as vehicle control and the homogenized absorbance is shown in each panel. C, Results from the proliferation assay of SW780 (left) and MB49 (right) cells with FGFR3 knocked down in the presence of T cells. Nonsilencing shRNA was used as negative control. D, Flow cytometry plots and graphs show Ki67 (left) and TNFα (right) expression of human T cells that were cocultured with SW780 cells in the presence of erdafitinib and infigratinib treatment (mean ± SD, n = 3). E, Flow cytometry plots and graphs show granzyme B (left) and perforin (right) expression of human T cells that were cocultured with SW780 cells in the presence of erdafitinib and infigratinib treatment (mean ± SD, n = 3). F, Flow cytometry plots and graphs show Ki67 (left) and TNFα (right) expression of human T cells that were cocultured with SW780 cells treated with FGFR3 shRNA (mean ± SD, n = 3). G, Flow cytometry plots and graphs show granzyme B (left) and perforin (right) expression of human T cells that cocultured with SW780 cells treated with FGFR3 shRNAs. Nonsilencing shRNA was used as negative control (mean ± SD, n = 3). n.s., not significant; ***, P < 0.001; ****, P < 0.0001; one-way ANOVA. PBMC, peripheral blood mononuclear cells. NS, nonsilencing shRNA.

Close modal

FGFR3 regulates PD-L1 ubiquitination via the ubiquitin E3 ligase NEDD4

To investigate the mechanism under FGFR3 regulating PD-L1 levels, we first assessed the relationship between FGFR3 inhibition and PD-L1 mRNA levels by RT-PCR. The results showed that the levels of PD-L1 mRNA with FGFR3 inhibition were not substantially upregulated in accordance with the protein levels (Fig. 4A). Clinical patient data from the TCGA/GTEx (n = 404) and Metabric (n = 1,904) databases revealed that FGFR3 expression has no obvious correlation with PD-L1 expression in cancer at mRNA level (Supplementary Fig. S4I). On the basis of this observation, we sought to determine whether the posttranslational pathway was the most relevant to PD-L1 upregulation in bladder cancer cells by screening common protein degradation pathways using a proteasome inhibitor (MG132) and several autophagy inhibitors. Our results revealed that only MG132 significantly increased PD-L1 in SW780 and MB49 cells (Fig. 4B and C). Broadly speaking, these results confirmed that FGFR3 might participate in PD-L1 degradation mediated by the ubiquitin–proteasome pathway (UPP). PD-L1 in SW780 cells was then pulled down by immunoprecipitation and subjected to ubiquitination analysis. Although PD-L1 had notable basal ubiquitination, FGFR3 interference abolished the ubiquitination of PD-L1 in SW780 cells (Fig. 4D, left). In concert with this, erdafitinib and infigratinib treatment also significantly inhibited PD-L1 ubiquitination (Fig. 4D, right). These results supported the notion that FGFR3 activation induced PD-L1 ubiquitination in bladder cancer cells. To identify the regulatory factor that serves as a bridge between FGFR3 and PD-L1, we first analyzed the protein–protein interaction (PPI) between FGFR3 and common E3 ubiquitin ligases in tumors through the STRING interaction network (Supplementary Table S3; refs. 34, 35). Among the 70 E3 ubiquitin ligases, only five were related to FGFR3 (Fig. 4E). To better identify which E3 ubiquitin ligase participates in the process of PD-L1 ubiquitination, we used the UbiBrowser database, a website that can predict human ubiquitin ligase (E3) substrate interactions, and displayed the top 20 E3 ubiquitin ligases that might participate in PD-L1 ubiquitination (Fig. 4F; ref. 36). To pinpoint which E3 ubiquitin ligase controls PD-L1 ubiquitination in bladder cancer cells, we used siRNA to reduce the relevant E3 ubiquitin ligase levels based on the five E3 ubiquitin ligases predicted to interact with FGFR3. To this end, we found that only NEDD4 interference could upregulate PD-L1 levels, similar to FGFR3 inhibition in human and mouse bladder cancer cells. (Fig. 4G and H; Supplementary Figs. S4J and S4K). This result was consistent with the intersection of the protein interaction predictions and E3 ubiquitin ligase predictions.

Figure 4.

FGFR3 inhibition upregulates PD-L1 level through suppressing ubiquitination mediated by NEDD4. A, The PD-L1 mRNA levels were measured by RT-PCR in SW780, RT4, and MB49 cell lines treated with erdafitinib and infigratinib (mean ± SD, n = 3). B, Representative flow cytometric images (left) and statistical diagrams (right) show the PD-L1 levels expressed in SW780 and MB49 cells with MG-132, chloroquine, 3-methyladenine, and bafilomycin A1 treatment (mean ± SD, n = 3). C, Immunoblot analysis for PD-L1 levels in SW780 and MB49 cells with different treatment. Actin was used as a control. D, Coimmunoprecipitation analysis of the ubiquitination of endogenous PD-L1 in SW780 cells treated with FGFR3 shRNAs (left) or erdafitinib and infigratinib (right). E, Protein–protein interaction network analysis showed five E3 ubiquitin ligases that interact directly with FGFR3. F, The PD-L1 targeting E3 ubiquitin ligases were predicted via the Ubibrowser database (ubibrowser.ncpsb.org/) and the top 20 E3 are displayed. G, Representative flow cytometric images and statistical diagrams show the PD-L1 levels expressed in SW780 cells after knockdown of NEDD4, CBL, MDM2, ITCH, and WWP1 by siRNAs as compared with nonsilencing and nontargeted control (mean ± SD, n = 3). H, Western blot representative images show the PD-L1 and five E3 expression in SW780 cells that were treated with five E3 siRNAs. n.s., not significant; *, P < 0.05; ****, P < 0.0001; one-way ANOVA.

Figure 4.

FGFR3 inhibition upregulates PD-L1 level through suppressing ubiquitination mediated by NEDD4. A, The PD-L1 mRNA levels were measured by RT-PCR in SW780, RT4, and MB49 cell lines treated with erdafitinib and infigratinib (mean ± SD, n = 3). B, Representative flow cytometric images (left) and statistical diagrams (right) show the PD-L1 levels expressed in SW780 and MB49 cells with MG-132, chloroquine, 3-methyladenine, and bafilomycin A1 treatment (mean ± SD, n = 3). C, Immunoblot analysis for PD-L1 levels in SW780 and MB49 cells with different treatment. Actin was used as a control. D, Coimmunoprecipitation analysis of the ubiquitination of endogenous PD-L1 in SW780 cells treated with FGFR3 shRNAs (left) or erdafitinib and infigratinib (right). E, Protein–protein interaction network analysis showed five E3 ubiquitin ligases that interact directly with FGFR3. F, The PD-L1 targeting E3 ubiquitin ligases were predicted via the Ubibrowser database (ubibrowser.ncpsb.org/) and the top 20 E3 are displayed. G, Representative flow cytometric images and statistical diagrams show the PD-L1 levels expressed in SW780 cells after knockdown of NEDD4, CBL, MDM2, ITCH, and WWP1 by siRNAs as compared with nonsilencing and nontargeted control (mean ± SD, n = 3). H, Western blot representative images show the PD-L1 and five E3 expression in SW780 cells that were treated with five E3 siRNAs. n.s., not significant; *, P < 0.05; ****, P < 0.0001; one-way ANOVA.

Close modal

FGFR3 binds to and phosphorylates NEDD4 to enhance its E3 activity

To understand how NEDD4 influenced bladder cancer cell PD-L1 levels, we first examined the relationship between upstream FGFR3 and NEDD4. Confocal immunofluorescence analysis revealed that p-FGFR3 colocalized with NEDD4 at the cell surface of bladder cancer cells (Fig. 5A and B). To assess whether FGFR3 activation was necessary for NEDD4 binding, we examined the effect of FGF1, the FGFR3-specific ligand, in 293T cells transfected with plasmids expressing Myc-tagged FGFR3 and Flag-tagged NEDD4. The immunoprecipitation and immunofluorescence results only showed a detectable FGFR3–NEDD4 combination and NEDD4 phosphorylation after FGF1 exposure (Fig. 5C; Supplementary Fig. S5A), whereas FGFR3 activity inhibited by erdafitinib prevented FGF1-induced FGFR3 and NEDD4 binding, consistent with no NEDD4 phosphorylation (Fig. 5D). It has been reported that NEDD4 can be phosphorylated to greatly improve its ubiquitination capacity (19). In the bladder cancer cell line SW780 with FGFR3-activating mutations, both FGFR3 and NEDD4 were phosphorylated at high levels, which were substantially reduced when FGFR3 was inhibited by FGFR inhibitors or shRNAs (Fig. 5E and F). These results revealed that the activation of FGFR3 was necessary for NEDD4 binding and phosphorylation. To further verify whether NEDD4 was critical for PD-L1 degradation in bladder cancer cells, we knocked out NEDD4 using a specific CRISPR/Cas9-sgRNA in SW780 and MB49 cells. As expected, depletion of NEDD4 led to a remarkable elevation of PD-L1 levels in both human and mouse bladder cancer cells (Fig. 5G and H). These results revealed that NEDD4 was a critical regulator for PD-L1 levels in bladder cancer with FGFR3 activation.

Figure 5.

FGFR3 binds to and phosphorylates NEDD4 to enhance its ubiquitination activity. A and B, Immunofluorescence images for NEDD4 (red), p-FGFR3 (green), and DAPI (blue) of SW780 and MB49 cells. Scale bars, 20 μm. Intensity profiles of p-FGFR3 (green lines) and NEDD4 (red lines) colocalization signal are shown in plotted lines at three random sites. C, Immunoprecipitation analysis of the interaction of FGFR3 with NEDD4 and phosphorylation of NEDD4, using Myc-FGFR3– and Flag-NEDD4–transfected 293T cells with/without FGF1 treatment. D, Immunoprecipitation analysis of the interaction of FGFR3 with NEDD4 and phosphorylation of NEDD4, using Myc-FGFR3– and Flag-NEDD4–transfected 293T cells with/without FGF1 and erdafitinib treatment. E, The immunoprecipitation analysis of the phosphorylation of NEDD4 in the SW780 cells treated with erdafitinib and infigratinib. DMSO was used as negative control. F, The immunoprecipitation analysis of the phosphorylation of NEDD4 in the SW780 cells treated with FGFR3 shRNAs. Nonsilencing shRNA was used as negative control. G, Western blot representative images show the expression of PD-L1 and NEDD4 in SW780 (left) and MB49 (right) cells after NEDD4 knockout. Mock transfection was used as a control. H, Representative flow cytometric images (left) and statistical diagrams (right) show the PD-L1 levels expressed in SW780 and MB49 cells with NEDD4-knockout. (mean ± SD, n = 3). n.s., not significant; ****, P < 0.0001; one-way ANOVA.

Figure 5.

FGFR3 binds to and phosphorylates NEDD4 to enhance its ubiquitination activity. A and B, Immunofluorescence images for NEDD4 (red), p-FGFR3 (green), and DAPI (blue) of SW780 and MB49 cells. Scale bars, 20 μm. Intensity profiles of p-FGFR3 (green lines) and NEDD4 (red lines) colocalization signal are shown in plotted lines at three random sites. C, Immunoprecipitation analysis of the interaction of FGFR3 with NEDD4 and phosphorylation of NEDD4, using Myc-FGFR3– and Flag-NEDD4–transfected 293T cells with/without FGF1 treatment. D, Immunoprecipitation analysis of the interaction of FGFR3 with NEDD4 and phosphorylation of NEDD4, using Myc-FGFR3– and Flag-NEDD4–transfected 293T cells with/without FGF1 and erdafitinib treatment. E, The immunoprecipitation analysis of the phosphorylation of NEDD4 in the SW780 cells treated with erdafitinib and infigratinib. DMSO was used as negative control. F, The immunoprecipitation analysis of the phosphorylation of NEDD4 in the SW780 cells treated with FGFR3 shRNAs. Nonsilencing shRNA was used as negative control. G, Western blot representative images show the expression of PD-L1 and NEDD4 in SW780 (left) and MB49 (right) cells after NEDD4 knockout. Mock transfection was used as a control. H, Representative flow cytometric images (left) and statistical diagrams (right) show the PD-L1 levels expressed in SW780 and MB49 cells with NEDD4-knockout. (mean ± SD, n = 3). n.s., not significant; ****, P < 0.0001; one-way ANOVA.

Close modal

NEDD4 targets and catalyzes K48-linked polyubiquitination of PD-L1

A hypothesis arising from the above results is that NEDD4 and PD-L1 can interact molecularly. To confirm this hypothesis, we performed fluorescence confocal localization of NEDD4 and PD-L1. As expected, NEDD4 colocalized with PD-L1 in bladder cancer cells (Fig. 6A and B). We knocked out PD-L1 from SW780 and MB49 cells and found that the NEDD4 was still distributed in the cell, but PD-L1 antibody only remains little nonspecific staining (Supplementary Figs. S5B and S5C). To further determine whether NEDD4 interacts with PD-L1, we expressed Myc-tagged FGFR3, Flag-tagged NEDD4, and V5-tagged PD-L1 in 293T cells. The levels of NEDD4 coimmunoprecipitated with PD-L1 significantly increased in cells in which FGFR3 was activated by FGF1 and NEDD4 was phosphorylated (Fig. 6C). These results indicated that the interaction between NEDD4 and PD-L1 was obviously enhanced in cells after NEDD4 was phosphorylated by FGFR3 activation. In addition, we also found that NEDD4 significantly coimmunoprecipitated with PD-L1 in SW780 and MB49 cells (Supplementary Fig. S5D). To further verify whether NEDD4 regulated PD-L1 protein levels through its E3 ligase activity, we assessed the NEDD4-mediated ubiquitination process of PD-L1 in bladder cancer cells. NEDD4-mediated polyubiquitination of PD-L1 was clearly detectable in SW780 cells transfected with plasmids expressing V5-tagged PD-L1 and hemagglutinin (HA)-tagged ubiquitin in the presence of a plasmid expressing Flag-tagged NEDD4 (Fig. 6D). To investigate the type of NEDD4-mediated polyubiquitination of PD-L1, we used vectors expressing HA-tagged mutant ubiquitin (K48) or ubiquitin (K63), which contain substitution of arginine for all lysine residues except the lysine at position 48 or 63, respectively. NEDD4 catalyzed the polyubiquitination of PD-L1 in the presence of HA-tagged wild-type ubiquitin [HA–ubiquitin (WT)] and HA–ubiquitin (K48) but not in the presence of HA–ubiquitin (K63) in SW780 cells (Fig. 6D). These data suggested that NEDD4 catalyzed the K48-linked polyubiquitination of PD-L1. In addition to the above findings, we identified that NEDD4 was necessary for PD-L1 ubiquitination by using SW780 cells with NEDD4 knockout. The ubiquitination of PD-L1 was significantly inhibited after NEDD4 knockout or treatment with erdafitinib (Fig. 6E). Pulse-chase analysis using cycloheximide revealed that NEDD4 deletion significantly prolonged the half-life of the PD-L1 protein in SW780 cells, whereas NEDD4 overexpression significantly shortened the PD-L1 protein half-life (Fig. 6F and G). The cocultivation results revealed that NEDD4 deletion in bladder cancer cells protected them from being killed by T cells to a great extent (Fig. 6H). Moreover, cocultivation with NEDD4-knockout bladder cancer cells significantly diminished the proportion of Ki67, TNFα, granzyme B, and perforin positive CD8+ T cells (Fig. 6I and J). To further determine the effects of NEDD4 on bladder cancer control in vivo, we used NEDD4 WT/KO MB49 cells to establish bladder cancer models in immunocompetent C57BL/6 mice (Supplementary Fig. S6A). The tumor growth speed in mice bearing NEDD4-KO MB49 tumors was significantly increased compared with that in mice bearing NEDD4-WT MB49 tumors (Supplementary Figs. S6B–S6F). The gap in tumor growth speed between the two groups was largely narrowed after anti-PD-1 antibody treatment (Supplementary Figs. S6B–S6F). Notably, tumors with NEDD4 deletion had markedly increased PD-L1 levels and decreased proportions of CD8+ T cells and granzyme B expression, which were reversed by treatment with an anti-PD-1 antibody (Supplementary Fig. S6G). These findings revealed that NEDD4 could downregulate the PD-L1 in MB49 cells and that NEDD4 deletion could help tumor cells subvert immune surveillance.

Figure 6.

NEDD4 binds to and catalyzes K48-linked polyubiquitination of PD-L1. A and B, Immunofluorescence images for NEDD4 (red), PD-L1 (green), and DAPI (blue) of SW780 (left) and MB49 (right) cells. Scale bars, 20 μm. Intensity profiles of PD-L1 (green lines) and NEDD4 (red lines) colocalization signal are shown in plotted lines at three random sites. C, Immunoprecipitation analysis of the interaction of NEDD4 with PD-L1 and phosphorylation of NEDD4 in 293T cells transfected with Myc-FGFR3, V5-PD-L1, and Flag-NEDD4 with/without FGF1 treatment. D, Immunoprecipitation analysis of the ubiquitination of PD-L1 in SW780 cells transfected with plasmids encoding V5-PD-L1, Flag-NEDD4, HA–ubiquitin (WT), HA–ubiquitin (K48), or HA–ubiquitin (K63). E, Immunoprecipitation analysis of the ubiquitination of PD-L1 in WT and NEDD4-KO SW780 cells with/without erdafitinib treatment. F, Western blot representative images show the expression of PD-L1 and NEDD4 in SW780 cells with NEDD4 knockout and overexpression in the presence of cycloheximide (CHX) for indicated time period. Actin was used as a control. G, The quantification of PD-L1 degradation kinetics in indicated groups after treatment with cycloheximide (n = 3). H, Results from the proliferation assay of T-cell cocultivation with SW780 (top) and MB49 (bottom) cells. Mock transfection was used as negative control and the homogenized absorbance is shown in each panel. I, Flow cytometry plots and graphs show the Ki67 and TNFα expression of human T cells that were cocultured with NEDD4-knockout SW780 cells (mean ± SD, n = 3). J, Flow cytometry plots and graphs show granzyme B (left) and perforin (right) expression of human T cells that were cocultured with NEDD4-KO SW780 cells. n.s., not significant; ***, P < 0.001; ****, P < 0.0001; one-way ANOVA.

Figure 6.

NEDD4 binds to and catalyzes K48-linked polyubiquitination of PD-L1. A and B, Immunofluorescence images for NEDD4 (red), PD-L1 (green), and DAPI (blue) of SW780 (left) and MB49 (right) cells. Scale bars, 20 μm. Intensity profiles of PD-L1 (green lines) and NEDD4 (red lines) colocalization signal are shown in plotted lines at three random sites. C, Immunoprecipitation analysis of the interaction of NEDD4 with PD-L1 and phosphorylation of NEDD4 in 293T cells transfected with Myc-FGFR3, V5-PD-L1, and Flag-NEDD4 with/without FGF1 treatment. D, Immunoprecipitation analysis of the ubiquitination of PD-L1 in SW780 cells transfected with plasmids encoding V5-PD-L1, Flag-NEDD4, HA–ubiquitin (WT), HA–ubiquitin (K48), or HA–ubiquitin (K63). E, Immunoprecipitation analysis of the ubiquitination of PD-L1 in WT and NEDD4-KO SW780 cells with/without erdafitinib treatment. F, Western blot representative images show the expression of PD-L1 and NEDD4 in SW780 cells with NEDD4 knockout and overexpression in the presence of cycloheximide (CHX) for indicated time period. Actin was used as a control. G, The quantification of PD-L1 degradation kinetics in indicated groups after treatment with cycloheximide (n = 3). H, Results from the proliferation assay of T-cell cocultivation with SW780 (top) and MB49 (bottom) cells. Mock transfection was used as negative control and the homogenized absorbance is shown in each panel. I, Flow cytometry plots and graphs show the Ki67 and TNFα expression of human T cells that were cocultured with NEDD4-knockout SW780 cells (mean ± SD, n = 3). J, Flow cytometry plots and graphs show granzyme B (left) and perforin (right) expression of human T cells that were cocultured with NEDD4-KO SW780 cells. n.s., not significant; ***, P < 0.001; ****, P < 0.0001; one-way ANOVA.

Close modal

FGFR3 blockade combined with anti-PD-1 therapy improves the antitumor effect

Because FGFR3 inhibition could both inhibit tumor growth and regulate PD-L1 stability, we sought to determine whether FGFR3 inhibition combined with ICB would enhance antitumor immunity accompanied by tumor growth inhibition. We established three mouse models including MB49 in a C57BL/6 mouse model, SW780 in a humanized immune reconstruction mouse model, and PDX mouse models bearing tumors with FGFR3 activating mutations from clinical patients. C57BL/6 mice were randomly grouped on the basis of tumor fluorescence intensity and treated with each formulation from 3 days post-inoculation with Luci+ MB49 cells (Supplementary Fig. S7A). In the initial 11 days after drug administration, remarkable tumor regression was observed in the combination therapy group compared with that in the monotherapy groups (Supplementary Figs. S7B–S7E). Combination therapy also significantly prolonged the survival of tumor-bearing mice. (Supplementary Fig. S7F). Ultrasound imaging and tumor tissue section staining further confirmed the excellent tumor regression effect and Ki67 inhibition of combination administration (Supplementary Fig. S7G). Interestingly, erdafitinib combined with anti-PD-1 antibody therapy increased the total proportion and activated CD8+ T cells compared with monotherapy administration, which indicated that combination therapy could fully activate the tumor-killing effect of the mouse immune system (Supplementary Fig. S7H). The safety and tolerability evaluation of the combination therapy showed that no significant relevant changes existed (Supplementary Figs. S8A–S8C).

To verify the therapeutic effect of combination therapy on human bladder cancer, we established a humanized immune system in NOG mice using human CD34+ HSC transplantation after radiation to achieve human bladder cancer cell implantation (Fig. 7A). Hu-HSC-NOG mice were randomly grouped and treated with each formulation from 5 days post-inoculation with Luci+ SW780 cells (Fig. 7B). Combination therapy achieved the best effect on suppressing tumor growth and prolonging survival time among all the treated groups (Fig. 7CG). The results from ultrasound imaging and flow cytometric analysis showed that SW780 tumor-bearing mice treated with combination therapy could suppress tumor growth by inhibiting cell proliferation and stimulating antitumor immune responses centered around CD8+ T cells (Fig. 7HK).

Figure 7.

Combination of FGFR3 blockade and anti-PD-1 therapy displays excellent antitumor activity in hu-HSC-NOG mice. A, Schematic of hu-HSC-NOG mice establishment. B, Schematic of experimental design to SW780 tumor-bearing mice model. C, Bioluminescence images of hu-HSC-NOG mice bearing Luci+ SW780 in different groups. D, Quantification of bioluminescence intensity in tumor-bearing mice (mean ± SD, n = 5). E, Macroscopic image of excised bladder tumors. Scale bars, 1 cm. F, Tumor weight after erdafitinib and anti-PD-1 treatment (mean ± SD, n = 5). G, Kaplan–Meier survival analysis of tumor-bearing mice in different groups [**, P < 0.01; log-rank (Mantel–Cox) test]. H, Ultrasound images, hematoxylin and eosin (H&E)–stained sections of tumors, and immunofluorescence images for Ki67 (red), cytokine 7 (green), and DAPI (blue) in SW780 tumor–bearing mice model. Scale bars in ultrasound images, 2 mm, 500 μm for hematoxylin and eosin staining, and 10 μm for immunofluorescence images. I, Flow cytometry analysis and statistical diagram showing the total proportion of CD3+CD8+ T cells in tumor (mean ± SD, n = 3). J, Flow cytometry analysis and statistical diagram showing the granzyme B-positive ratio of CD3+CD8+ T cells and granzyme B+CD8+ T cells in tumor (mean ± SD, n = 3). K, Flow cytometry analysis and statistical diagram showing the TNFα-positive ratio in CD8+ T cells and TNFα+ CD8+ T cells in tumor (mean ± SD, n = 3). L, Experimental design to evaluate the in vivo effect of combination treatment in PDX mice model. M, Ultrasound images of orthotopic bladder tumors in PDX mice model. Scale bars, 2 mm. N, Macroscopic image of excised bladder tumors. Scale bars, 1 cm. O, Tumor weight after erdafitinib and anti-PD-1 treatment (mean ± SD, n = 5). P, Kaplan–Meier survival analysis of PDX mice model treated with erdafitinib and anti-PD-1 [**, P < 0.01, log-rank (Mantel–Cox) test]. Q, PD-L1 t-SNE plots from PDX tumor tissue in different groups. The statistical diagram showing the proportion of PD-L1–positive cells in PDX tumor tissue (mean ± SD, n = 3). R, Flow cytometry analysis and statistical plots showing the PD-1, Tim-3, and LAG-3 expression level of CD8+ T cells in PDX tumor tissues (mean ± SD, n = 3). S, Schematic showing FGFR3–NEDD4–PD-L1 axis in FGFR3-activated bladder cancer. n.s., not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; two tailed Student t test, one-way ANOVA.

Figure 7.

Combination of FGFR3 blockade and anti-PD-1 therapy displays excellent antitumor activity in hu-HSC-NOG mice. A, Schematic of hu-HSC-NOG mice establishment. B, Schematic of experimental design to SW780 tumor-bearing mice model. C, Bioluminescence images of hu-HSC-NOG mice bearing Luci+ SW780 in different groups. D, Quantification of bioluminescence intensity in tumor-bearing mice (mean ± SD, n = 5). E, Macroscopic image of excised bladder tumors. Scale bars, 1 cm. F, Tumor weight after erdafitinib and anti-PD-1 treatment (mean ± SD, n = 5). G, Kaplan–Meier survival analysis of tumor-bearing mice in different groups [**, P < 0.01; log-rank (Mantel–Cox) test]. H, Ultrasound images, hematoxylin and eosin (H&E)–stained sections of tumors, and immunofluorescence images for Ki67 (red), cytokine 7 (green), and DAPI (blue) in SW780 tumor–bearing mice model. Scale bars in ultrasound images, 2 mm, 500 μm for hematoxylin and eosin staining, and 10 μm for immunofluorescence images. I, Flow cytometry analysis and statistical diagram showing the total proportion of CD3+CD8+ T cells in tumor (mean ± SD, n = 3). J, Flow cytometry analysis and statistical diagram showing the granzyme B-positive ratio of CD3+CD8+ T cells and granzyme B+CD8+ T cells in tumor (mean ± SD, n = 3). K, Flow cytometry analysis and statistical diagram showing the TNFα-positive ratio in CD8+ T cells and TNFα+ CD8+ T cells in tumor (mean ± SD, n = 3). L, Experimental design to evaluate the in vivo effect of combination treatment in PDX mice model. M, Ultrasound images of orthotopic bladder tumors in PDX mice model. Scale bars, 2 mm. N, Macroscopic image of excised bladder tumors. Scale bars, 1 cm. O, Tumor weight after erdafitinib and anti-PD-1 treatment (mean ± SD, n = 5). P, Kaplan–Meier survival analysis of PDX mice model treated with erdafitinib and anti-PD-1 [**, P < 0.01, log-rank (Mantel–Cox) test]. Q, PD-L1 t-SNE plots from PDX tumor tissue in different groups. The statistical diagram showing the proportion of PD-L1–positive cells in PDX tumor tissue (mean ± SD, n = 3). R, Flow cytometry analysis and statistical plots showing the PD-1, Tim-3, and LAG-3 expression level of CD8+ T cells in PDX tumor tissues (mean ± SD, n = 3). S, Schematic showing FGFR3–NEDD4–PD-L1 axis in FGFR3-activated bladder cancer. n.s., not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; two tailed Student t test, one-way ANOVA.

Close modal

To be closer to the clinical application, we established PDX mouse models on hu-HSC-NOG mice using a UC of the bladder specimen from an elderly male patient with high-grade UC of the bladder that had FGFR3–TACC3 gene fusion detected by NGS. Bladder cancer PDX mice with a humanized immune system were randomly grouped on the basis of ultrasound imaging assessment and treated with each formulation from 5 days post-inoculation (Fig. 7L). The combination treatment inhibited tumor growth and prolonged median overall survival more effectively than that in control mice or mice treated with monotherapy (Fig. 7MP). We examined the PD-L1 expression levels in the PDX tumors, and the results indicated that PD-L1-positive cells ratio conspicuously increased in the tumors treated with erdafitinib (Fig. 7Q). Besides, exhaustion markers including Tim-3, LAG-3, and PD-1 on CD8+ T cell showed no obvious difference among different treated group (Fig. 7R). Strikingly, these results demonstrated that our research had important potential value that could be translated into clinical application.

Previous studies on molecular subtypes and ICB therapy have demonstrated that there is a relatively low immune signature and decreased expression of PD-L1 in UC of the bladder with FGFR3 mutation or high expression (6, 9, 13, 37), but the regulatory mechanism is obscure. Our study reveals that the activation of FGFR3 pathways leads to the downregulation of PD-L1 and that the PD-L1 level is elevated after administration of FGFR inhibitors in UC of the bladder with FGFR3 activation. More importantly, we uncovered the underlying mechanism about the significant number of patients with FGFR3-activated UC of the bladder could not benefit from FGFR inhibition and proposed the combination therapy to address this challenge. Most studies have revealed that the therapeutic effect of PD-1/PD-L1 inhibitors is closely related to PD-L1 expression levels in various cancers (38, 39). However, the specific mechanism underlying which PD-L1 is regulated in UC of the bladder remains unclear. Herein, our study illustrates the intrinsic pathway of PD-L1 regulation using a series of in vivo and in vitro experiments in which PD-L1 can be destabilized via NEDD4 phosphorylation caused by FGFR3 activation.

The abnormal regulation of NEDD4 plays a critical role in oncogenesis and tumor progression (40, 41). NEDD4, which is frequently overexpressed in most cancers, is associated with chemotherapy and targeted therapy resistance (40), whereas there are no studies concerning the influence of NEDD4 on immunotherapy. Our findings reveal potentially critical roles of NEDD4 in the regulation of PD-L1 expression. The expression level and activating status of NEDD4 may play roles in the response to PD-1/PD-L1 inhibitors. In our study, we found that although NEDD4 knockout led to rapid tumor growth, the response to anti-PD-1 antibody was more sensitive than that in NEDD4 WT tumors. Hence, the NEDD4 E3 ligase may be a promising predictive biomarker and therapeutic target in advanced UC of the bladder with immunotherapy or combined therapy.

Mutations and high expression of FGFR3 have different effects on bladder cancer prognosis. Patients enrolled in clinical trials with different FGFR inhibitors had different inclusion criteria for mutations or high expression, and the clinical trial results were also different (42). Our study included both bladder cancer with high expression and mutations of FGFR3, and we conclude that the activation of FGFR3 is the key issue rather than expression or mutation. An aberrant FGFR signaling axis was found to function in oncogenesis, tumor progression, and resistance to anticancer therapy across different tumor types (43). FGFR3 mutation or fusion was also found in breast cancer, glioblastoma, intrahepatic cholangiocarcinoma, cervical cancer, and so on, but the mutation frequency was lower than that of UC of the bladder (44). Numerous FGFR inhibitors are currently being assessed in preclinical, phase 1, phase 2, and phase 3 clinical trials in different cancer types (37). It has been reported that erdafitinib inhibits PD-L1 expression in human lung tumor and FGFR2 promoted expression of PD-L1 in colorectal cancer (45, 46). In this study, we showed that erdafitinib and infigratinib induce PD-L1 expression in bladder cancer cells. Furthermore, we verified this phenomenon in another two FGFR3 activating mutations cancer cell lines and found that FGFR3 inhibition also induced PD-L1 expression in these cells. On the basis of this, we speculated the phenomenon that erdafitinib and infigratinib inducing PD-L1 expression in bladder cancer cells may be cancer selective. Future studies should be conducted to extend this study to other cancer types and generalize our hypothesis.

Recent studies have demonstrated that transcriptional and posttranscriptional modifications of PD-L1 through several signaling pathways including JAK/STAT3, GSK3β, SPOP, and STT3 have emerged as important regulatory mechanisms that modulate immunosuppression in cancer (15, 16). Previous studies have shown that chemotherapy treatment can also induce PD-L1 expression in lung cancer (47, 48). However, chemotherapy and targeted therapy differ greatly in their mechanisms of action and therapeutic indication. The particularity of PD-L1 regulation in bladder cancer, especially in certain subtypes of bladder cancer, has hardly been well elucidated in previous studies. Here, we identified FGFR3 as a new regulator of PD-L1 protein levels through NEDD4 in FGFR3-activated bladder cancer. On the basis of the available data, we conclude that the activation of FGFR3 in bladder cancer recruits NEDD4 and phosphorylates it, whereas NEDD4 phosphorylation can activate its ability to ubiquitinate PD-L1 protein (Fig. 7S). Furthermore, the observations that FGFR3 activating mutation in clinical data is inversely correlated with PD-L1 levels suggest a model in which FGFR3 is correlated with bladder cancer immunology by influencing PD-L1. In line with this, it can be speculated that FGFR3 and NEDD4 in bladder cancer may fulfill an immunoregulatory role by influencing the ubiquitination of the immune checkpoint molecule PD-L1. Finally, multiple mouse models, including PDX mice bearing an FGFR3–TACC3 gene fusion bladder tumor from clinical excision, showed that erdafitinib and anti-PD-1 antibody combination treatment significantly enhanced the effectiveness of the current ICB and targeted therapy. These results may change our understanding of FGFR3 targeted therapy in UC of the bladder.

No disclosure was reported by the authors.

W. Jing: Conceptualization, data curation, software, formal analysis, visualization, methodology, writing–original draft. G. Wang: Software, formal analysis, visualization, methodology, writing–original draft. Z. Cui: Formal analysis, methodology. G. Xiong: Methodology. X. Jiang: Formal analysis, investigation. Y. Li: Methodology. W. Li: Resources. B. Han: Formal analysis. S. Chen: Resources, supervision, writing–review and editing. B. Shi: Resources, supervision, funding acquisition, project administration, writing–review and editing.

This research was supported by the National Natural Science Foundation of China (grants 81670687 and 81970661 to B. Shi; grant 81800672 to S. Chen), the Tai Shan Scholar Foundation (ts201511092 to B. Shi); Primary Research & Development Plan of Shandong Province (2019GSF108123 to S. Chen) and CSCO Clinical Oncology Research Foundation (Y-2019AZQN-0557 to S. Chen). All schematic figures were created with BioRender.com.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Sung
H
,
Ferlay
J
,
Siegel
RL
,
Laversanne
M
,
Soerjomataram
I
,
Jemal
A
, et al
Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 Countries
.
CA Cancer J Clin
2021
;
71
:
209
49
.
2.
Antoni
S
,
Ferlay
J
,
Soerjomataram
I
,
Znaor
A
,
Jemal
A
,
Bray
F
. 
Bladder cancer incidence and mortality: a global overview and recent trends
.
Eur Urol
2017
;
71
:
96
108
.
3.
Lenis
AT
,
Lec
PM
,
Chamie
K
,
Mshs
MD
. 
Bladder cancer: a review
.
JAMA
2020
;
324
:
1980
91
.
4.
Bellmunt
J
,
Théodore
C
,
Demkov
T
,
Komyakov
B
,
Sengelov
L
,
Daugaard
G
, et al
Phase III trial of vinflunine plus best supportive care compared with best supportive care alone after a platinum-containing regimen in patients with advanced transitional cell carcinoma of the urothelial tract
.
J Clin Oncol
2009
;
27
:
4454
61
.
5.
Galsky
MD
,
JÁA
A
,
Bamias
A
,
Davis
ID
,
De Santis
M
,
Kikuchi
E
, 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
.
6.
Rosenberg
JE
,
Hoffman-Censits
J
,
Powles
T
,
van der Heijden
MS
,
Balar
AV
,
Necchi
A
, 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
.
7.
van Rhijn
BWG
,
Mertens
LS
,
Mayr
R
,
Bostrom
PJ
,
Real
FX
,
Zwarthoff
EC
, et al
FGFR3 mutation status and FGFR3 expression in a large bladder cancer cohort treated by radical cystectomy: implications for anti-FGFR3 treatment?
Eur Urol
2020
;
78
:
682
7
.
8.
Blinova
E
,
Buzdin
A
,
Enikeev
D
,
Roshchin
D
,
Suntsova
M
,
Samyshina
E
, et al
Prognostic role of FGFR3 expression status and tumor-related MicroRNAs level in association with PD-L1 expression in primary luminal non-muscular invasive bladder carcinoma
.
Life (Basel)
2020
;
10
:
305
.
9.
Rosenberg
JE
,
Gajate
P
,
Morales-Barrera
R
,
Lee
JL
,
Necchi
A
,
Penel
N
, et al
Safety and preliminary efficacy of rogaratinib in combination with atezolizumab in a phase Ib/II study (FORT-2) of first-line treatment in cisplatin-ineligible patients (pts) with locally advanced or metastatic urothelial cancer (UC) and FGFR mRNA overexpression
.
J Clin Oncol
38
:
15s
, 
2020
(
suppl. abstr. 5014
).
10.
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
.
11.
Patel
VG
,
Oh
WK
,
Galsky
MD
. 
Treatment of muscle-invasive and advanced bladder cancer in 2020
.
CA Cancer J Clin
2020
;
70
:
404
23
.
12.
Qin
Q
,
Patel
V
,
Galsky
MD
. 
Urothelial carcinoma: the development of FGFR inhibitors in combination with immune checkpoint inhibitors
.
Expert Rev Anticancer Ther
2020
;
20
:
503
12
.
13.
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
56
.
14.
Wang
L
,
Gong
Y
,
Saci
A
,
Szabo
PM
,
Martini
A
,
Necchi
A
, et al
Fibroblast growth factor receptor 3 alterations and response to PD-1/PD-L1 blockade in patients with metastatic urothelial cancer
.
Eur Urol
2019
;
76
:
599
603
.
15.
Hsu
JM
,
Li
CW
,
Lai
YJ
,
Hung
MC
. 
Posttranslational modifications of PD-L1 and their applications in cancer therapy
.
Cancer Res
2018
;
78
:
6349
53
.
16.
Zhang
J
,
Bu
X
,
Wang
H
,
Zhu
Y
,
Geng
Y
,
Nihira
NT
, et al
Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance
.
Nature
2018
;
553
:
91
95
.
17.
Rotin
D
,
Kumar
S
. 
Physiological functions of the HECT family of ubiquitin ligases
.
Nat Rev Mol Cell Biol
2009
;
10
:
398
409
.
18.
Xu
C
,
Fan
CD
,
Wang
X
. 
Regulation of Mdm2 protein stability and the p53 response by NEDD4–1 E3 ligase
.
Oncogene
2015
;
34
:
281
9
.
19.
Persaud
A
,
Alberts
P
,
Mari
S
,
Tong
J
,
Murchie
R
,
Maspero
E
, et al
Tyrosine phosphorylation of NEDD4 activates its ubiquitin ligase activity
.
Sci Signal
2014
;
7
:
ra95
.
20.
Chalmers
ZR
,
Connelly
CF
,
Fabrizio
D
,
Gay
L
,
Ali
SM
,
Ennis
R
, et al
Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden
.
Genome Med
2017
;
9
:
34
.
21.
Vichai
V
,
Kirtikara
K
. 
Sulforhodamine B colorimetric assay for cytotoxicity screening
.
Nat Protoc
2006
;
1
:
1112
6
.
22.
Feoktistova
M
,
Geserick
P
,
Leverkus
M
. 
Crystal violet assay for determining viability of cultured cells
.
Cold Spring Harb Protoc
2016
;
2016
:
pdb.prot087379
.
23.
Kang
S
,
Dong
S
,
Gu
TL
,
Guo
A
,
Cohen
MS
,
Lonial
S
, et al
FGFR3 activates RSK2 to mediate hematopoietic transformation through tyrosine phosphorylation of RSK2 and activation of the MEK/ERK pathway
.
Cancer Cell
2007
;
12
:
201
14
.
24.
Du
X
,
Wang
QR
,
Chan
E
,
Merchant
M
,
Liu
J
,
French
D
, et al
FGFR3 stimulates stearoyl CoA desaturase 1 activity to promote bladder tumor growth
.
Cancer Res
2012
;
72
:
5843
55
.
25.
LaFleur
MW
,
Nguyen
TH
,
Coxe
MA
,
Miller
BC
,
Yates
KB
,
Gillis
JE
, et al
PTPN2 regulates the generation of exhausted CD8+ T cell subpopulations and restrains tumor immunity
.
Nat Immunol
2019
;
20
:
1335
47
.
26.
Wang
W
,
Green
M
,
Choi
JE
,
Gijón
M
,
Kennedy
PD
,
Johnson
JK
, et al
CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy
.
Nature
2019
;
569
:
270
4
.
27.
Wu
YM
,
Su
F
,
Kalyana-Sundaram
S
,
Khazanov
N
,
Ateeq
B
,
Cao
X
, et al
Identification of targetable FGFR gene fusions in diverse cancers
.
Cancer Discov
2013
;
3
:
636
47
.
28.
Acquaviva
J
,
He
S
,
Zhang
C
,
Jimenez
JP
,
Nagai
M
,
Sang
J
, et al
FGFR3 translocations in bladder cancer: differential sensitivity to HSP90 inhibition based on drug metabolism
.
Mol Cancer Res
2014
;
12
:
1042
54
.
29.
Langle
YV
,
Belgorosky
D
,
McCormick
BP
,
Sahores
A
,
Góngora
A
,
Baldi
A
, et al
FGFR3 down-regulation is involved in bacillus Calmette-Guérin induced bladder tumor growth inhibition
.
J Urol
2016
;
195
:
188
97
.
30.
Boulay
G
,
Sandoval
GJ
,
Riggi
N
,
Iyer
S
,
Buisson
R
,
Naigles
B
, et al
Cancer-specific retargeting of BAF complexes by a prion-like domain
.
Cell
2017
;
171
:
163
78
.
e19
.
31.
Ronchetti
D
,
Greco
A
,
Compasso
S
,
Colombo
G
,
Dell'Era
P
,
Otsuki
T
, et al
Deregulated FGFR3 mutants in multiple myeloma cell lines with t(4;14): comparative analysis of Y373C, K650E and the novel G384D mutations
.
Oncogene
2001
;
20
:
3553
62
.
32.
Sanli
O
,
Dobruch
J
,
Knowles
MA
,
Burger
M
,
Alemozaffar
M
,
Nielsen
ME
, et al
Bladder cancer
.
Nat Rev Dis Primers
2017
;
3
:
17022
.
33.
Fridman
WH
,
Pagès
F
,
Sautès-Fridman
C
,
Galon
J
. 
The immune contexture in human tumours: impact on clinical outcome
.
Nat Rev Cancer
2012
;
12
:
298
306
.
34.
Senft
D
,
Qi
J
,
Ronai
ZA
. 
Ubiquitin ligases in oncogenic transformation and cancer therapy
.
Nat Rev Cancer
2018
;
18
:
69
88
.
35.
Szklarczyk
D
,
Gable
AL
,
Lyon
D
,
Junge
A
,
Wyder
S
,
Huerta-Cepas
J
, et al
STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets
.
Nucleic Acids Res
2019
;
47
:
D607
13
.
36.
Li
Y
,
Xie
P
,
Lu
L
,
Wang
J
,
Diao
L
,
Liu
Z
, et al
An integrated bioinformatics platform for investigating the human E3 ubiquitin ligase-substrate interaction network
.
Nat Commun
2017
;
8
:
347
.
37.
Roghmann
F
,
Wirtz
R
,
Jarczyk
J
,
Kriegmair
MC
,
Worst
TS
,
Sikic
D
, et al
Prognostic role of FGFR alterations and FGFR mRNA expression in metastatic urothelial cancer treated with anti-PD(L1) inhibitors in first and second line setting
.
Eur Urol Open Sci
2020
;
19
:
e830
.
38.
Gibney
GT
,
Weiner
LM
,
Atkins
MB
. 
Predictive biomarkers for checkpoint inhibitor-based immunotherapy
.
Lancet Oncol
2016
;
17
:
e542
51
.
39.
Yi
M
,
Jiao
D
,
Xu
H
,
Liu
Q
,
Zhao
W
,
Han
X
, et al
Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors
.
Mol Cancer
2018
;
17
:
129
.
40.
Wang
ZW
,
Hu
X
,
Ye
M
,
Lin
M
,
Chu
M
,
Shen
X
. 
NEDD4 E3 ligase: functions and mechanism in human cancer
.
Semin Cancer Biol
2020
;
67
:
92
101
.
41.
Bernassola
F
,
Karin
M
,
Ciechanover
A
,
Melino
G
. 
The HECT family of E3 ubiquitin ligases: multiple players in cancer development
.
Cancer Cell
2008
;
14
:
10
21
.
42.
Flippot
R
,
Loriot
Y
. 
The FGFR3 story in bladder cancer: another piece of the puzzle?
Eur Urol
2020
;
78
:
688
9
.
43.
Babina
IS
,
Turner
NC
. 
Advances and challenges in targeting FGFR signalling in cancer
.
Nat Rev Cancer
2017
;
17
:
318
32
.
44.
Krook
MA
,
Reeser
JW
,
Ernst
G
,
Barker
H
,
Wilberding
M
,
Li
G
, et al
Fibroblast growth factor receptors in cancer: genetic alterations, diagnostics, therapeutic targets and mechanisms of resistance
.
Br J Cancer
2021
;
124
:
880
92
.
45.
Palakurthi
S
,
Kuraguchi
M
,
Zacharek
SJ
,
Zudaire
E
,
Huang
W
,
Bonal
DM
, et al
The combined effect of FGFR inhibition and PD-1 blockade promotes tumor-intrinsic induction of antitumor immunity
.
Cancer Immunol Res
2019
;
7
:
1457
71
.
46.
Li
P
,
Huang
T
,
Zou
Q
,
Liu
D
,
Wang
Y
,
Tan
X
, et al
FGFR2 promotes expression of PD-L1 in colorectal cancer via the JAK/STAT3 signaling pathway
.
J Immunol
2019
;
202
:
3065
75
.
47.
Shin
J
,
Chung
JH
,
Kim
SH
,
Lee
KS
,
Suh
KJ
,
Lee
JY
, et al
Effect of platinum-based chemotherapy on PD-L1 expression on tumor cells in non-small cell lung cancer
.
Cancer Res Treat
2019
;
51
:
1086
97
.
48.
Guo
L
,
Song
P
,
Xue
X
,
Guo
C
,
Han
L
,
Fang
Q
, et al
Variation of programmed death ligand 1 expression after platinum-based neoadjuvant chemotherapy in lung cancer
.
J Immunother
2019
;
42
:
215
20
.