Purpose:

There is an urgent need for biomarkers of radiation response in organ-sparing therapies. Bladder preservation with trimodality therapy (TMT), consisting of transurethral tumor resection followed by chemoradiation, is an alternative to radical cystectomy for muscle-invasive bladder cancer (MIBC), but molecular determinants of response are poorly understood.

Experimental Design:

We characterized genomic and transcriptomic features correlated with long-term response in a single institution cohort of patients with MIBC homogeneously treated with TMT. Pretreatment tumors from 76 patients with MIBC underwent whole-exome sequencing; 67 underwent matched transcriptomic profiling. Molecular features were correlated with clinical outcomes including modified bladder-intact event-free survival (mBI-EFS), a composite endpoint that reflects long-term cancer control with bladder preservation.

Results:

With a median follow-up of 74.6 months in alive patients, 37 patients had favorable long-term response to TMT while 39 had unfavorable long-term response. Tumor mutational burden was not associated with outcomes after TMT. DNA damage response gene alterations were associated with improved locoregional control and mBI-EFS. Of these alterations, somatic ERCC2 mutations stood out as significantly associated with favorable long-term outcomes; patients with ERCC2 mutations had significantly improved mBI-EFS [HR, 0.15; 95% confidence interval (CI), 0.06–0.37; P = 0.030] and improved BI-EFS, an endpoint that includes all-cause mortality (HR, 0.33; 95% CI, 0.15–0.68; P = 0.044). ERCC2 mutant bladder cancer cell lines were significantly more sensitive to concurrent cisplatin and radiation treatment in vitro than isogenic ERCC2 wild-type cells.

Conclusions:

Our data identify ERCC2 mutation as a candidate biomarker associated with sensitivity and long-term response to chemoradiation in MIBC. These findings warrant validation in independent cohorts.

Translational Relevance

Although trimodality therapy (TMT) can be an effective organ-sparing alternative to radical cystectomy for muscle-invasive bladder cancer (MIBC), not all patients have favorable long-term outcomes after TMT. A better understanding of molecular determinants of response to chemoradiation can provide biomarkers for the optimal selection of appropriate patients for TMT. We performed whole-exome sequencing and transcriptome profiling of pretreatment tumors in a well-curated cohort of patients with MIBC homogeneously treated with TMT with long clinical follow-up. This integrated analysis yielded insights into molecular features associated with favorable long-term response to TMT, including alterations in DNA damage response genes. Of these, mutations in ERCC2, a DNA helicase in the nucleotide excision repair pathway, were significantly associated with improved long-term outcomes after TMT, particularly in patients who received concurrent cisplatin-based chemotherapy. These findings warrant further investigation but identify potential biomarkers of chemoradiation response that can guide the precision radiation-based management of patients with MIBC.

Radiation therapy with or without chemotherapy remains a pillar of curative-intent treatment in many solid tumors (1), often allowing avoidance of radical surgery and facilitating organ sparing, such as in patients with breast cancer or muscle-invasive bladder cancer (MIBC; refs. 2, 3). Predictive biomarkers for the rational selection of systemic therapies for individual patients have transformed precision oncology (4), but few biomarkers are available clinically to aid therapeutic decisions involving radiotherapy despite the genomic heterogeneity of cancers (5, 6). There exists an urgent need to identify candidate biomarkers for predicting successful outcomes after radiotherapy to help improve selection of patients and potentially allow for de-escalation of therapy in specific clinical circumstances (7).

Promising predictive biomarkers of radiotherapy response include genomic alterations in DNA damage response (DDR) genes (8, 9). Radiation-induced DNA damage is sensed and repaired via a highly coordinated cellular DDR, and tumors that harbor alterations in DDR pathways may have increased sensitivity to radiation. Therefore, there is interest in evaluating DDR genes to determine whether specific DDR alterations are correlated with clinical radiosensitivity of tumors (9, 10). Other studies have identified potential non-DDR tumor biomarkers as predictors for radiotherapeutic efficacy or resistance (11–14), but despite interest (5), this area of precision oncology remains understudied.

Trimodality therapy (TMT), which consists of maximal transurethral resection of bladder tumor (TURBT) followed by chemoradiation, is an effective bladder-preserving treatment for well-selected patients with MIBC (3, 15). Although no randomized trials have been completed comparing TMT with the treatment of radical cystectomy with or without neoadjuvant chemotherapy (2), multiple long-term series have demonstrated comparable outcomes (16–19). MIBC has been previously molecularly characterized, with recurrent mutations identified in known cancer genes such as TP53, KMT2D, and KDM6A. Prior analyses have evaluated gene alterations with clinical outcomes following neoadjuvant chemotherapy prior to cystectomy and identified DDR genes associated with response (20, 21). MIBC molecular subtypes defined by transcriptional profiles have been found to be associated with outcomes in patients with MIBC undergoing radical cystectomy with or without neoadjuvant chemotherapy (22–24). Gene expression profiling of 136 primary tumors from patients with MIBC treated with TMT identified an association of T-cell activation and INFγ signaling expression signatures with improved disease-specific survival in the TMT cohort (25). However, there remains a need for a deeper understanding of the molecular characteristics of MIBC to aid with treatment selection and identify more accurate prognostic and predictive biomarkers of chemoradiation outcomes (26, 27). To this end, we leveraged a well-curated clinical cohort of patients with MIBC homogenously treated with TMT that included patients previously evaluated for gene expression profiling (25), and performed whole-exome sequencing (WES) in combination with gene expression profiling to identify potential molecular biomarkers associated with clinical outcomes of chemoradiation therapy.

Patients and samples

Patients with cT2-T4aN0M0 MIBC treated with definitive-intent chemoradiation as part of TMT were retrospectively identified from a single institution between 1986–2015. This study was approved by the Mass General Brigham Institutional Review Board (IRB; Protocol #2008P001128), and studies were conducted in accordance with the Declaration of Helsinki and in accordance with an assurance filed with and approved by the U.S. Department of Health and Human Services. Analyzed tumor tissues were excess discarded clinical specimens previously collected from human subjects as part of usual clinical care under an IRB-approved waiver of consent. Tumors from this cohort had previously been analyzed for whole transcriptome analysis (25), and those with sufficient remaining formalin-fixed, paraffin-embedded (FFPE) tissue available for whole-exome sequencing were included in the study.

Study endpoints

All molecular characteristics were correlated with clinical outcomes following TMT. The main clinical outcome used in this analysis was modified bladder-intact event-free survival (mBI-EFS), with an event defined as muscle-invasive recurrence, locoregional nodal progression, distant metastasis, death from bladder cancer, and radical cystectomy for either cancer recurrence or toxicity. For the purposes of this analysis, patients who had a cumulative incidence of an event were defined as having an “unfavorable” long-term mBI-EFS outcome, while those without a cumulative incidence of an event were defined as having a “favorable” long-term mBI-EFS outcome. This long-term outcome classification is a secondary endpoint of the INTACT SWOG/NRG 1806 cooperative group trial of TMT with or without atezolizumab (NCT03775265), and is closely related to bladder-intact event-free survival (BI-EFS), the primary endpoint of the trial, with the only difference being that mBI-EFS does not include all-cause mortality. These endpoints were developed by investigators from ECOG, RTOG/NRG, ALLIANCE, SWOG, and CCTG, in discussion with the NCI GU Steering Committee and the U.S. FDA, as the composite endpoint most clinically relevant for this patient population. Other clinical endpoints examined included cumulative incidence of locoregional failure, cumulative incidence of distant failure, locoregional control, mBI-EFS, BI-EFS, and overall survival (OS).

DNA extraction and whole-exome sequencing

DNA extraction, whole-exome library preparation, and sequencing were performed at the Broad Institute using methods previously described (12, 28). FFPE TURBT specimens were reviewed by a board-certified genitourinary pathologist to select high-density tumor blocks. Normal (germline) DNA samples were not available. Genomic DNA was purified from FFPE cores using a commercial kit (Qiagen QIAamp DNA FFPE Tissue Kit Quantitation Reagent). Whole-exome capture libraries were constructed from 100 ng of DNA after sample shearing, end repair, and phosphorylation and ligation to barcoded sequencing adaptors. Ligated DNA was size selected for lengths between 200–350bp and subjected to exonic hybrid capture using The Broad Institute Genomics Platform Custom Illumina bait (12). Briefly, the Illumina exome specifically targets approximately 37.7 Mb of primarily exonic territory consisting of all targets from the Agilent exome design (Agilent SureSelect All Exon), all coding regions of Gencode V11 genes, and all coding regions of RefSeq gene and KnownGene tracks from the UCSC genome browser (http://genome.ucsc.edu). The sample was multiplexed and sequenced using Illumina HiSeq/NextSeq technology. Data were analyzed using the Broad Picard Pipeline, which includes demultiplexing and data aggregation.

Quality control and variant calling

Data processing and analysis of exome sequence data were performed using Broad Institute pipelines as described previously (12, 28). We used the Getz Lab CGA WES Characterization pipeline developed at the Broad Institute to call, filter, and annotate somatic mutations and copy number variations. The pipeline employs the following tools: MuTect (29), ContEst (30), Strelka (31), Orientation Bias Filter (32), DeTiN (33), AllelicCapSeg (34), MAFPoNFilter (35), RealignmentFilter, ABSOLUTE (36), GATK (37), PicardTools (38), Variant Effect Predictor (39), and Oncotator (40). ContEst (30) was used to evaluate cross-individual contaminations and samples with <4% estimated contamination were included. The MuTect algorithm (29) was applied to identify single-nucleotide variants in targeted exons, and Strelka (31) was used to identify small deletions or insertions. All alterations were annotated with Oncotator (40). Due to the absence of germline DNA, a common variant filtering strategy was implemented to remove likely germline artifacts, using the ExAC database, now a part of gnomAD (41). This tool is also used by AACR Genie (42). Common variants were annotated and soft filtered if the variant appeared in at least 10 alleles across any ancestral subpopulation in ExAC. Mutations were examined for distribution and type and confirmed using Integrative Genomics Viewer (43, 44). Genes comprising known oncogenic signaling pathways (as described in Robertson and colleagues; ref. 45) as well as the following frequently mutated genes in MIBC were analyzed:

TP53/Cell Cycle pathway: ATM, TP53, MDM2, CDKN2A, RB1, CCND1, CDKN1A, PTEN, CCNE1, FBXW7, CDKN1B, CCND1/2/3, CDK4/6

RTK/RAS/PIK3K pathway: PIK3CA, FGFR1/3, ERBB2/3, RAF1, PTEN, TSC1/2, EGFR, AKT1/2, NF1, RAC1, H/N/KRAS, JAK1/2, BRAF

Histone modification pathway: EP300, CREBBP, KMT2C/D, KDM6A, BAP1, ASXL1/2, SETD2

SWI/SNF pathway: ARID1A, ARID1B, ARID2

DDR pathway: ERCC2, BRIP1, ATM, BRCA1/2, RAD21/50, CHEK1

Cohesin complex pathway: STAGE1/2, RAD21, SMC1A/3

Oxidative stress pathway: NFE2L2, KEAP1, CUL3, TXNIP

Alternative splicing pathway: RMB10, SF3B1, U2AF1, CDK12

Other frequently mutated MIBC genes: KMT2A, FAT1, ELF3, SPTAN1

In addition, we examined alterations in specific genes that are being evaluated in on-going clinical trials that incorporated a risk-adapted chemotherapy-based approach to the treatment of MIBC: RETAIN (NCT02710734) and Alliance A031701 (NCT03609216). In RETAIN BLADDER, a mutation-positive patient is defined as any alteration present in ATM, RB1, FANCC, or ERCC2. In the Alliance trial, pretreatment TURBT specimens are evaluated for deleterious DDR gene alternations (truncating mutations including frameshift, nonsense, splice site) in 8 genes (ATM, ATR, FANCC, BRCA1, BRCA2, RECQL4, RAD51C, ERCC5) or any mutation in ERCC2. Loss-of-function mutations in these genes were identified using OncoKB (46).

Mutational signatures (COSMIC Signatures v3.2) were derived from tumor samples using deconstructSigs (https://github.com/raerose01/deconstructSigs) to determine the weights of each mutational signature contributing to the mutational spectrum of a tumor sample (47).

Transcriptome analysis

RNA extraction from FFPE cores and expression profile generation was previously performed (25). Briefly, transcriptome-wide gene expression profiles were generated using Human Exon Array 1.0 ST oligonucleotide microarrays in a clinic-grade laboratory (Veracyte).

The Genomic subtyping classifier (GSC) subtypes were generated by first identifying neuroendocrine-like (NE-like cases; ref. 48), with the remaining tumors classified using the Seiler 2017 model (23). A T-cell–inflamed immune content signature (25, 49), MIBC consensus class (50), as well as The Cancer Genomic Atlas (TCGA 2017) subtyping models were also used to classify our tumors into various subtypes. Transcriptomic data was available for 67 of the 76 patients (88%) in this cohort.

Cell culture and clonogenic assay

The bladder cancer cell lines used here have been previously described (51, 52). KU19–19 (RRID: CVCL_1344) and SW1710 (RRID: CVCL_1721) cell lines were originally purchased from DSMZ and were validated by STR profiling. KE1 is a derivative of the KU19–19 bladder cancer cell line and SC14 is a derivative of the SW1710 bladder cancer cell line. Both KE1 and SC14 harbor ERCC2 mutations introduced by CRISPR/Cas9 techniques and have been shown to be nucleotide excision repair (NER) deficient. The KU19–19 and KE1 cell lines were cultured in RPMI media supplemented with 10% FBS. SW1710 and SC14 cell lines were cultured in DMEM supplemented with 10% FBS. Early-passage cells (≤6 passages) were used for all experiments and cell lines were tested monthly to confirm absence of Mycoplasma. For cisplatin and radiation sensitivity assays, cells were plated in 6 well plates (KU19 and KE: 5,000 cells/well; SW1710 and SC14: 8,000 cells/well) and the following day were treated with cisplatin and/or radiation. Cells were then incubated for 10 days, fixed with 4% paraformaldehyde for 20 minutes, stained with 1% crystal violet for 1 hour, and then washed with tap water, allowed to dry, and photographed. For quantification, 1 mL of 1% SDS solution was added to each well to dissolve the crystal violet and the sample solutions were then scanned on a microplate reader at a wavelength of 570 nm. Survival for each condition was calculated as the signal divided by the signal in untreated cells. Survival curves were plotted using Graphpad Prism and the survival fraction was calculated using a linear-quadratic (LQ) model. Combenefit software was used to perform synergy analysis (53).

Statistical analysis

Differences in single-nucleotide variations and tumor mutational burden (TMB) between those with favorable versus unfavorable clinical outcomes were calculated using χ2 analyses and unpaired t tests, respectively. A Benjamini–Hochberg FDR of 0.1 was used to account for multiple hypothesis testing. COSMIC mutational signatures were evaluated using Kruskal–Wallis H and Wilcoxon rank sum tests. Differences in expression profile subtypes between favorable and unfavorable long-term mBI-EFS outcomes were calculated using χ2 test. Associations of transcriptomic signature scores and gene mutations versus wild-type (WT) were analyzed using Mann–Whitney U tests. Association of select gene mutations with mBI-EFS, BI-EFS, locoregional control, and OS was assessed using the Kaplan–Meier method and log rank test with P < 0.05 considered to be statistically significant.

Data availability

The WES data generated in this study are publicly available in dbGAP at phs003402.v1.p1. Expression profile data analyzed in this study were obtained from Gene Expression Omnibus at GSE128701.

Integrated analysis of genomic and transcriptomic data from patients with MIBC undergoing TMT

There were 92 patients who met initial criteria for inclusion in the study. Seventy-six cases passed quality control metrics for WES and comprise our final cohort (Supplementary Fig. S1). Clinical characteristics of the cohort are summarized in Table 1. The median age of the cohort was 72.3 years, with 24% female patients. All patients had MIBC (86% T2, 14% T3/T4) and the majority were treated with cisplatin-based chemotherapy (66%) concurrent with radiation (median dose 64.3 Gy, interquartile range, 60.7–64.3).

Table 1.

Clinical characteristics of bladder TMT cohort.

CharacteristicsNumber%
Age at Diagnosis 72.3 yr (IQR, 63.8–77.8) 
Sex 
 Male 58 76 
 Female 18 24 
Clinical T stage 
 T2 65 86 
 T3 11 
 T4 
Concurrent chemotherapy 
 Cisplatin/5-FU 33 43 
 Cisplatin/taxol 11 14 
 Mitomycin/5-FU 11 14 
 Cisplatin 
 Paclitaxel 
 5-FU 
 Gemcitabine 
 NA 
Grade 
 Moderate 
 Poor 73 96 
Hydronephrosis 
 Present 11 14 
 Absent 65 86 
Tumor-associated CIS 
 Present 12 16 
 Absent 64 84 
TURBT 
 Visibly complete 59 78 
 Visibly incomplete 17 22 
Pathologic response to CRT 
 Complete 59 78 
 Incomplete 17 22 
Radiation dose (median)  64.3 Gy (IQR 60.7–64.3) 
Histology Pure urothelial 55 72 
 Squamous differentiation 11 
 Glandular differentiation 
 Micropapillary 
 Neuroendocrine differentiation 
 Sarcomatoid differentiation 
 Spindle cell 
 NA 
CharacteristicsNumber%
Age at Diagnosis 72.3 yr (IQR, 63.8–77.8) 
Sex 
 Male 58 76 
 Female 18 24 
Clinical T stage 
 T2 65 86 
 T3 11 
 T4 
Concurrent chemotherapy 
 Cisplatin/5-FU 33 43 
 Cisplatin/taxol 11 14 
 Mitomycin/5-FU 11 14 
 Cisplatin 
 Paclitaxel 
 5-FU 
 Gemcitabine 
 NA 
Grade 
 Moderate 
 Poor 73 96 
Hydronephrosis 
 Present 11 14 
 Absent 65 86 
Tumor-associated CIS 
 Present 12 16 
 Absent 64 84 
TURBT 
 Visibly complete 59 78 
 Visibly incomplete 17 22 
Pathologic response to CRT 
 Complete 59 78 
 Incomplete 17 22 
Radiation dose (median)  64.3 Gy (IQR 60.7–64.3) 
Histology Pure urothelial 55 72 
 Squamous differentiation 11 
 Glandular differentiation 
 Micropapillary 
 Neuroendocrine differentiation 
 Sarcomatoid differentiation 
 Spindle cell 
 NA 

Abbreviations: CIS, carcinoma in situ; CRT, chemoradiation therapy; 5-FU, 5-fluorouracil; Gy, Gray; IQR, interquartile range; NA, not available; yr, years.

The genomic tumor landscape of the cohort is summarized in Fig. 1 and Supplementary Fig. S2. Figure 1 displays all genes comprising known oncogenic signaling pathways and known frequently mutated MIBC genes (Methods) with at least 5% mutation frequency. Mean target coverage was 109X for somatic tumor DNA (Supplementary Table S1). Mean nonsynonymous TMB for the entire cohort was 12.4 mutations/Mb (range, 3.4–31.5). Overall, TP53 was the most frequently mutated gene with 52.6% of samples having an alteration, followed by KMT2D at 40.8%. Mutational signature analysis identified four active signatures: Signatures 1, 2, 5, and 13. Signature 1, present in 62/76 (81.6%) samples, is associated with age-associated spontaneous or enzymatic deamination of 5-methylcystosine to thymine (54). Signature 5, which is the signature previously associated with ERCC2 mutations in urothelial tumors and with smoking history, was present in 68/76 (89.5%) samples (55). Signatures 2 and 13, present in 44/76 (57.9%) and 39/76 (51.3%) cases, respectively, are associated with APOBEC-mediated mutations (54), and have been described previously in MIBC (45). There were significant differences in the relative contribution of mutations across mutational signatures (Supplementary Fig. S3; Kruskal–Wallis H, P < 0.001), similar to previous observations in MIBC cohorts (56). Signature 5 had a significantly higher contribution than other signatures (Wilcoxon rank sum, P < 0.001), but patients with ERCC2-mutant tumors did not have higher contribution from signature 5 compared with patients with ERCC2 WT tumors (P = 0.813).

Figure 1.

Integrative molecular characterization of the MIBC cohort. Mutational signatures, TMB, gene expression molecular subtypes (GSC, TCGA, and MIBC consensus), transcriptomic T-cell–inflamed immune signature score, select clinical characteristics, and somatic alternations in select genes in pathways (Methods) for each tumor in the cohort (n = 76). TURBT complete refers to whether the initial TURBT was complete versus incomplete. Complete response defined as no visible tumor on cystoscopy and a negative tumor site rebiopsy following TMT after a period of induction therapy (16).

Figure 1.

Integrative molecular characterization of the MIBC cohort. Mutational signatures, TMB, gene expression molecular subtypes (GSC, TCGA, and MIBC consensus), transcriptomic T-cell–inflamed immune signature score, select clinical characteristics, and somatic alternations in select genes in pathways (Methods) for each tumor in the cohort (n = 76). TURBT complete refers to whether the initial TURBT was complete versus incomplete. Complete response defined as no visible tumor on cystoscopy and a negative tumor site rebiopsy following TMT after a period of induction therapy (16).

Close modal

Sixty-seven samples had transcriptomic data available (Fig. 1). Among the GSC subtypes (23, 48), Luminal was most common (21/67, 31.3%), while the Luminal papillary subtype was most common among TCGA subtypes (21/67, 31.3%), and the Basal squamous subtype was most common among the MIBC consensus class (19/67, 28.4%). The distribution of TCGA subtypes in our cohort was not significantly different from the distribution in the TCGA MIBC cohort (Supplementary Fig. S4; ref. 45). Overall, the mean T-cell–inflamed signature score was 0.23 (range, −0.04 to 0.98).

To verify that the genomic data correlates with the transcriptomic data, we examined how a few genomic alterations associated with corresponding transcriptomic signature scores. FGFR3 mutations are common in non–MIBC as well as in tumors of the upper tract, but are also present at lower frequencies in MIBC (57). In this cohort, 6 of 76 tumors (7.9%) harbored FGFR3 mutations (Supplementary Fig. S5A). A lncRNA-based genomic classifier (58) revealed significant enrichment for FGFR3+ (“FGFR3 active”) cases in FGFR3 mutant tumors (4/6 cases; 66.7%) compared with FGFR3 WT tumors (4/61 cases; 6.6%, P < 0.0001). Similarly, tumors with FGFR3 mutations also had higher FGFR3 transcriptional signature scores (59) compared with FGFR3 WT tumors (median 0.528 versus 0.191; P < 0.001; Supplementary Fig. S5B).

Mutations in TP53 were most common in the cohort (40/76 samples mutated). Gene expression profiling revealed higher hallmark p53 pathway scores (60) among those with TP53 WT tumors compared with mutant (median 0.3593 versus 0.1171; P = 0.0001; Supplementary Fig. S6A). Separately, upon evaluation of a previously described novel TP53 mutation signature in prostate cancers (61), tumors with TP53 mutations had higher signature scores compared with WT tumors (median 0.9945 versus 0.9786; P = 0.020; Supplementary Fig. S6B), consistent with prior associations (61).

Genomic predictors of favorable mBI-EFS outcomes

Median follow-up for the cohort was 74.6 (range, 20.0–266.4) months in alive patients. Thirty-seven of 76 patients (49%) were characterized as having favorable long-term outcomes for mBI-EFS, and 39 (51%) as having unfavorable long-term outcomes for mBI-EFS (see study endpoints in Methods). Five-year mBI-EFS for the entire cohort was 45.4% (Supplementary Fig. S7A). Five-year BI-EFS for the entire cohort was 37.9% (Supplementary Fig. S7B), and 5-year OS for the entire cohort was 48.6% (Supplementary Fig. S7C). There were 19 locoregional failures and 22 distant failures.

TMB has been shown to be associated with response to immune checkpoint inhibitors (62, 63), and TMB has also been suggested to be associated with radiation response in other cancers (64). We therefore evaluated whether there were differences in TMB in patients with MIBC with favorable compared with unfavorable clinical outcomes after TMT in our cohort. Interestingly, there was no significant difference in TMB between those with favorable (13.3 mutations/Mb, range 3.4–31.5) and unfavorable mBI-EFS outcomes (11.5 mutations/Mb, range 6.4–26.8), P = 0.1584 (Supplementary Fig. S8).

We then performed an exploratory analysis of DDR genes as defined in the Alliance A031701 (NCT03609216) and RETAIN (NCT02710734) clinical trials of risk-adapted MIBC treatment to determine whether mutations in any of these 10 DDR genes correlated with clinical outcomes after TMT (Fig. 2A). The following mutations were identified in our cohort: ATM (n = 13), RB1 (n = 17), FANCC (n = 4), ERCC2 (n = 8), ATR (n = 9), BRCA1 (n = 7), BRCA2 (n = 11), RECQL4 (n = 0), RAD51C (n = 0), and ERCC5 (n = 2; Supplementary Table S2). TMB was higher among DDR mutant tumors (13.4 mutations/Mb, range 3.4–31.5) compared with DDR WT tumors (10.8 mutations/Mb, range 5.5–24.7, P = 0.04). In the TCGA cohort which included patients with MIBC who underwent surgical resection (45), the presence of these DDR mutations was not significantly associated with improved survival [median months survival in patients with DDR mutations: 41.72 (95% confidence interval (CI), 26.91–92.9) versus 33.11 (95% CI, 22.86–54.86); P = 0.442; Supplementary Fig. S9]. However, in our TMT cohort, having any DDR gene mutation present was associated with improved mBI-EFS (HR, 0.51; 95% CI, 0.27–0.97; P = 0.040; Fig. 2B), as well as improved locoregional control (HR, 0.31; 95% CI, 0.12–0.80; log-rank P = 0.009; Fig. 2C). This was also consistent when evaluating cumulative incidence of locoregional failures (n = 19); patients without locoregional failure were enriched for the presence of any DDR gene mutation present [39/57 (68.4%) with mutations] compared with patients with locoregional failure [7/19 (37%) with mutations; χ2P = 0.014]. There were no associations between DDR gene alterations and distant failures (n = 22 total distant failures, 11 among patients with DDR gene-altered tumors). In addition, there was no association between patients with TP53 mutations and favorable or unfavorable mBI-EFS.

Figure 2.

Association of select DDR genes and long-term clinical outcomes following TMT. A, TMB, concurrent cisplatin with radiation, locoregional failure (LRF), mBI-EFS outcome (F, favorable; UF, unfavorable), along with select DDR gene alterations. Samples are ordered by favorable mBI-EFS outcome and unfavorable mBI-EFS outcome. Patients without locoregional failure were enriched for presence of any DDR mutation compared to patients with locoregional failure (χ2 0.014). More patients with favorable mBI-EFS outcomes had mutations in ERCC2 compared with those with unfavorable mBI-EFS outcomes (χ2P = 0.020). B, Having any DDR mutation present associated with improved locoregional control (HR, 0.31; 95% CI, 0.12–0.80; log-rank P = 0.0088). C, Having any DDR mutation present associated with improved mBI-EFS (HR, 0.51; 95% CI, 0.27–0.97; P = 0.004).

Figure 2.

Association of select DDR genes and long-term clinical outcomes following TMT. A, TMB, concurrent cisplatin with radiation, locoregional failure (LRF), mBI-EFS outcome (F, favorable; UF, unfavorable), along with select DDR gene alterations. Samples are ordered by favorable mBI-EFS outcome and unfavorable mBI-EFS outcome. Patients without locoregional failure were enriched for presence of any DDR mutation compared to patients with locoregional failure (χ2 0.014). More patients with favorable mBI-EFS outcomes had mutations in ERCC2 compared with those with unfavorable mBI-EFS outcomes (χ2P = 0.020). B, Having any DDR mutation present associated with improved locoregional control (HR, 0.31; 95% CI, 0.12–0.80; log-rank P = 0.0088). C, Having any DDR mutation present associated with improved mBI-EFS (HR, 0.51; 95% CI, 0.27–0.97; P = 0.004).

Close modal

Next, because missense mutations in DDR genes are often non-functional passenger events, we further evaluated whether the presence of known loss of function (LOF) mutations in DDR genes (as annotated in OncoKB; ref. 46) impacts outcomes after TMT (Supplementary Tables S3 and S4). We found that patients without locoregional failure continued to be enriched for the presence of any DDR (LOF) mutation present [26/57 (66.7%) with mutations] compared with patients with locoregional failure [2/19 (11%) with mutations; χ2P = 0.006]. Patients with favorable mBI-EFS outcomes were enriched for the presence of an ERCC2 LOF mutation [4/37 (11%) with mutations] compared with patients with unfavorable mBI-EFS outcomes [1/39 (3%) with mutations; χ2P < 0.001].

We then evaluated the proportion of patients with favorable mBI-EFS outcomes after TMT who would have met DDR gene mutation-specific inclusion criteria for either of the risk-adapted bladder-sparing clinical trials that are testing whether chemotherapy alone is sufficient to omit upfront local therapy (cystectomy or radiation) in patients with MIBC with DDR gene mutations (Supplementary Fig. S10A and S10B). Upon applying these inclusion criteria [Alliance A031701 included any truncating mutations including frameshift, nonsense, splice site in 8 genes (ATM, ATR, FANCC, BRCA1/2, RECQL4, RAD51C, ERCC5) or any mutation in ERCC2; RETAIN included any alteration present in ATM, RB1, FANCC, or ERCC2], it was found that 20/37 (54%) patients with favorable mBI-EFS had DDR gene mutations necessary for eligibility for one or both trials (9 RETAIN only, 2 Alliance only, 9 both trials), while 17/37 (46%) patients did not meet any of the mutation eligibility criteria for these trials (Supplementary Fig. S10A; Supplementary Table S4). Thus, nearly half of patients who had favorable mBI-EFS outcomes after bladder-sparing TMT would not have been eligible for participation in chemotherapy-only–based risk-adapted bladder preservation clinical trials.

Of the DDR gene alterations evaluated, ERCC2 mutations stood out as being significantly associated with favorable mBI-EFS outcomes (n = 7) as compared with unfavorable outcomes (n = 1, locoregional failure; Fisher exact P = 0.027; Fig. 2A). ERCC2 is a DNA helicase that plays an important role in the NER pathway (65). Upon examination of the distribution of 8 ERCC2 mutations, 7 were located within one of the two conserved helicase domains (Fig. 3A; Supplementary Table S5). The distribution of ERCC2 mutations in this cohort reflects the overall mutation distribution present in the TCGA bladder cohort (Supplementary Fig. S11; ref. 45). Six of the 8 patients with ERCC2 mutations received cisplatin-based chemotherapy concurrently with radiation (all with favorable mBI-EFS outcomes); of the 2 that did not, 1 received mitomycin C, which also creates DNA lesions that require the NER pathway for repair of DNA damage, and was also associated with favorable mBI-EFS outcome. The 1 patient with an unfavorable mBI-EFS outcome received concurrent 5-FU. Therefore, among patients who received cisplatin-based chemoradiotherapy, all 6 patients with an ERCC2 mutation had favorable mBI-EFS outcomes. Patients with ERCC2 mutations had significantly improved mBI-EFS (HR, 0.15; 95% CI, 0.06–0.37; P = 0.030; Fig. 3B), and improved BI-EFS, an endpoint that includes all-cause mortality (HR, 0.33; 95% CI, 0.15–0.68; P = 0.044; Fig. 3C). A trend was also noted toward improved OS in patients with ERCC2 mutations (HR, 0.36; 95% CI, 0.17–0.80; P = 0.075; Fig. 3D). There was no significant difference in TMB among ERCC2 mutation tumors (14.0 mutations/Mb, range 4.2–24.3) versus ERCC2 WT tumors (12.2 mutations/Mb, range 3.4–31.5; P = 0.38).

Figure 3.

ERCC2 mutations associate with favorable mBI-EFS outcomes following TMT. A, Lollipop figure of ERCC2 gene depicting the location of missense mutations in the 6 patients with a favorable mBI-EFS outcome (black), as well as the location of the missense mutation in the patient with an unfavorable mBI-EFS outcome (red). One patient with a favorable outcome had I17T and Y24C compound mutations (denoted by bracket). One patient with a favorable outcome had a splice site mutation (not shown). B, Patients with ERCC2 mutations had improved mBI-EFS (HR, 0.15; 95% CI, 0.06–0.37; P = 0.030). C, Patients with ERCC2 mutations had improved BI-EFS, which includes all-cause mortality (HR, 0.33; 95% CI, 0.15–0.68; P = 0.044). D, Patients with ERCC2 mutations had a trend toward improved OS (HR, 0.36; 95% CI, 0.17–0.80; P = 0.075).

Figure 3.

ERCC2 mutations associate with favorable mBI-EFS outcomes following TMT. A, Lollipop figure of ERCC2 gene depicting the location of missense mutations in the 6 patients with a favorable mBI-EFS outcome (black), as well as the location of the missense mutation in the patient with an unfavorable mBI-EFS outcome (red). One patient with a favorable outcome had I17T and Y24C compound mutations (denoted by bracket). One patient with a favorable outcome had a splice site mutation (not shown). B, Patients with ERCC2 mutations had improved mBI-EFS (HR, 0.15; 95% CI, 0.06–0.37; P = 0.030). C, Patients with ERCC2 mutations had improved BI-EFS, which includes all-cause mortality (HR, 0.33; 95% CI, 0.15–0.68; P = 0.044). D, Patients with ERCC2 mutations had a trend toward improved OS (HR, 0.36; 95% CI, 0.17–0.80; P = 0.075).

Close modal

The mBI-EFS endpoint includes cystectomy due to treatment toxicity as an event, and in our cohort, 2 patients had their bladders removed due to toxicity (Supplementary Table S4). To evaluate whether excluding these events would affect our analysis, we evaluated a new endpoint that does not include cystectomy for toxicity (called “re-modified BI-EFS”). We found that having any DDR mutation was significantly associated with improved re-modified BI-EFS (HR, 0.51; 95% CI, 0.26–0.99; P = 0.040; Supplementary Fig. S12) and patients with ERCC2 mutations had significantly improved re-modified BI-EFS (HR, 0.15; 95% CI, 0.06–0.37; P = 0.037; Supplementary Fig. S13).

ERCC2 mutations were associated with a Luminal papillary subtype

We then evaluated the transcriptional landscape of ERCC2 mutant tumors compared with ERCC2 WT tumors. Patient samples with ERCC2 mutations (n = 6 with transcriptomic data available) had differential enrichment of several transcriptomic signature scores and molecular subtypes compared with ERCC2 WT tumors (n = 61; Fig. 4A). Samples with ERCC2 mutations had higher hallmark DNA repair scores (ref. 60; median 0.110 versus −0.318; P = 0.020), consistent with dysregulation of DDR (Supplementary Fig. S14). In addition, ERCC2 mutant tumors were enriched for TCGA Luminal papillary subtype compared with ERCC2 WT (5/6, 83% versus 26% Luminal papillary; P = 0.004; Fig. 4B). These same patients were also enriched for GSC Luminal subtype compared with ERCC2 WT tumors (4/6, 67% versus 28% Luminal; P = 0.050, Supplementary Fig. S15). ERCC2 mutant tumors were not associated with higher or lower T-cell–inflamed signature scores compared with ERCC2 WT tumors.

Figure 4.

Gene expression subtypes and signature scores associate with ERCC2 mutations. A, TCGA and GSC expression subtypes in the TMT cohort with transcriptomic data (n = 67), hallmark DNA repair signature score, and ERCC2 alterations in a TMT cohort. B,ERCC2 mutation tumors were enriched for TCGA Luminal papillary subtype compared with ERCC2 WT (83% versus 26% Luminal papillary, P = 0.004).

Figure 4.

Gene expression subtypes and signature scores associate with ERCC2 mutations. A, TCGA and GSC expression subtypes in the TMT cohort with transcriptomic data (n = 67), hallmark DNA repair signature score, and ERCC2 alterations in a TMT cohort. B,ERCC2 mutation tumors were enriched for TCGA Luminal papillary subtype compared with ERCC2 WT (83% versus 26% Luminal papillary, P = 0.004).

Close modal

Functional characterization of ERCC2 mutations in response to cisplatin and radiation

We next tested the functional impact of ERCC2 mutations on cisplatin and radiation sensitivity in bladder cancer cell lines. KU19–19 and SW1710 are bladder cancer cell lines with WT ERCC2, while KE1 and SC14 are derivatives of KU19–19 and SW1710, respectively, with ERCC2 mutations introduced by CRISPR-mediated gene editing (52). We have previously shown that the ERCC2-mutant KE1 and SC14 lines are NER-deficient and have increased cisplatin sensitivity compared with their respective WT ERCC2 parental lines (51, 52). To model the impact of ERCC2 functional loss on cisplatin-based chemoradiotherapy response, we compared the sensitivity of the KU19–19/KE1 and SW1710/SC14 cell pairs to varying doses of combined cisplatin and radiation treatment. As expected, the ERCC2-mutant lines were significantly more sensitive to cisplatin monotherapy than the WT ERCC2 parental lines (Fig. 5AD). The addition of sublethal doses of cisplatin to radiation did not significantly impact sensitivity in the parental KU19–19 and SW1710 cell lines; however, there was a significant increase in sensitivity with dose enhancement factors of 1.55 and 1.40 when cisplatin was added to radiation in the ERCC2-mutant SC14 and KE1 cell lines, respectively (Fig. 5EJ). The ERCC2 mutant clones demonstrated mild increase in radiation sensitivity compared with WT, but this was much less pronounced than the increase in cisplatin sensitivity (Fig. 5E and F). Taken together, these data demonstrate that the presence of a deleterious ERCC2 mutation is sufficient to drive sensitivity to combined radiation and cisplatin treatment in bladder cancer preclinical models.

Figure 5.

ERCC2 mutations confer sensitivity to cisplatin-based chemoradiotherapy in bladder cancer cell lines. A, Representative images of clonogenic cell survival following treatment with cisplatin and ionizing radiation (IR) over a range of concentrations and doses in SW1710 (ERCC2 WT) and SC14 (ERCC2 mutant, mut) bladder cancer cell lines. B, Representative images of clonogenic cell survival following treatment with cisplatin and IR for KU19–19 (ERCC2 WT) and KE1 (ERCC2 mut) bladder cancer cell lines. C and D, Clonogenic survival fractions for SW1710 versus SC14 and KU19–19 vs. KE1 cell lines following cisplatin treatment at the concentrations shown. E, Clonogenic survival fractions for ERCC2 WT cell lines following treatment with IR with versus without cisplatin at ∼IC70 (SW1710) or ∼IC50 (KU19–19) concentrations. Data points were fitted with the LQ formula with the IR dose range determined by the sensitivity of the ERCC2 mut lines to combined IR/cisplatin. F, Analogous to Panel E, clonogenic survival fractions of ERCC2 mut cell lines using cisplatin at ∼IC70 (SC14) and ∼IC50 (KE1) concentrations. Dose enhancement factors (DEF) for cisplatin were estimated at 0.5 survival fraction. GJ, Synergy analysis was performed over the full range of treatment doses and concentrations (Combenefit). All data points represent three independent repeats. Comparison of data points by t test, two-sided, and comparison of fitted survival curves by F-test, two-sided (GraphPad Prism 6.1). NS, not significant.

Figure 5.

ERCC2 mutations confer sensitivity to cisplatin-based chemoradiotherapy in bladder cancer cell lines. A, Representative images of clonogenic cell survival following treatment with cisplatin and ionizing radiation (IR) over a range of concentrations and doses in SW1710 (ERCC2 WT) and SC14 (ERCC2 mutant, mut) bladder cancer cell lines. B, Representative images of clonogenic cell survival following treatment with cisplatin and IR for KU19–19 (ERCC2 WT) and KE1 (ERCC2 mut) bladder cancer cell lines. C and D, Clonogenic survival fractions for SW1710 versus SC14 and KU19–19 vs. KE1 cell lines following cisplatin treatment at the concentrations shown. E, Clonogenic survival fractions for ERCC2 WT cell lines following treatment with IR with versus without cisplatin at ∼IC70 (SW1710) or ∼IC50 (KU19–19) concentrations. Data points were fitted with the LQ formula with the IR dose range determined by the sensitivity of the ERCC2 mut lines to combined IR/cisplatin. F, Analogous to Panel E, clonogenic survival fractions of ERCC2 mut cell lines using cisplatin at ∼IC70 (SC14) and ∼IC50 (KE1) concentrations. Dose enhancement factors (DEF) for cisplatin were estimated at 0.5 survival fraction. GJ, Synergy analysis was performed over the full range of treatment doses and concentrations (Combenefit). All data points represent three independent repeats. Comparison of data points by t test, two-sided, and comparison of fitted survival curves by F-test, two-sided (GraphPad Prism 6.1). NS, not significant.

Close modal

Biomarkers of treatment outcomes to guide precision oncology are needed to select patients for radiation or chemoradiation-based organ-sparing treatment approaches. In MIBC, TMT can be an effective alternative to radical cystectomy for bladder preservation, and there is an urgent unmet need for biomarkers to guide treatment selection. In this study, we performed integrated genomic and transcriptomic analyses to evaluate genomic determinants of clinical outcomes following TMT in the largest MIBC cohort with such data available. When evaluating molecular associations with clinical outcomes, including BI-EFS and mBI-EFS which are clinically relevant endpoints for this patient population (66), it was found that having any DDR gene alteration as per genes defined in the Alliance A031701 (NCT03609216) and RETAIN BLADDER (NCT02710734) trials was associated with improved locoregional control as well as mBI-EFS. Of these, ERCC2 mutations stood out as being significantly associated with improved mBI-EFS and BI-EFS, with a trend towards improved OS. This is the first study to associate molecular biomarkers with BI-EFS and mBI-EFS, clinical trials endpoints previously defined by researchers and stakeholders invested in relevant outcomes for patients with MIBC.

Genomic alterations in DDR genes are prevalent in bladder cancer, and these alterations have been previously studied and associated with improved responses to chemotherapy and immune checkpoint inhibitors (20, 21, 67, 68). DDR gene mutations have been previously found to be associated with higher TMB (69, 70), and our cohort similarly demonstrated higher TMB among DDR mutated tumors versus DDR WT tumors. Although TMB was not associated with outcomes after TMT, DDR gene alterations were associated with favorable long-term outcomes in our TMT cohort. Interestingly, DDR gene mutations are not associated with improved survival in the TCGA cohort, which is comprised of patients treated with radical cystectomy, indicating that DDR gene mutations are not inherently prognostic. ERCC2 mutations have been associated with sensitivity to cisplatin-based chemotherapy, and functional profiling of clinically observed ERCC2 missense mutations has demonstrated that nearly all mutations arising within the conserved helicase domains are unable to support normal cellular NER, including mutations at Y24, N238, G606, and G665 as observed in this cohort (52, 65). While ERCC2-dependent NER has no known role in the repair of radiation-induced DNA damage (71), our functional data demonstrate that the presence of an ERCC2 mutation is sufficient to sensitize bladder cancer cells to combination treatment with cisplatin and radiation. Consistent with this notion, 75% (6/8) of the patients with ERCC2 mutations received concurrent cisplatin-based chemotherapy in our cohort. The one patient with unfavorable mBI-EFS in the cohort with an ERCC2 mutation did not receive cisplatin-based chemotherapy. Although we cannot parse out the relative contributions of chemotherapy versus radiation to the favorable mBI-EFS outcomes observed in patients with DDR or ERCC2 mutations, contemporary TMT includes both concurrent radiation and low-dose radiosensitizing cisplatin (or other DNA damaging) chemotherapy. Future investigations could evaluate relative contribution of these mutations in neoadjuvant full-dose cisplatin cohorts compared with TMT cohorts receiving concurrent low-dose radiosensitizing cisplatin, or by comparing with outcomes in chemotherapy-only bladder-sparing trials. Nevertheless, our data and data from others (72) suggest that ERCC2 could serve as a potential biomarker for TMT treatment selection in cisplatin-eligible patients.

Our analysis also evaluated patients who had transcriptomic signature/subtype data available, and we observed higher hallmark DNA repair scores (60) among samples with ERCC2 mutations. These tumors were also enriched for GSC Luminal subtype and TCGA Luminal papillary subtype, compared with ERCC2 WT tumors. Prior data indicate that the Luminal/Luminal papillary subtypes are associated with heterogeneous responses to neoadjuvant chemotherapy (73), while a separate analysis did not find any association between molecular subtype and rate of complete response following TMT (25). In the TCGA cohort, ERCC2 mutant tumors were not enriched for Luminal papillary subtype (74). Therefore, our findings must be validated in larger cohorts.

Due to the prevalence of DDR mutations in MIBC, trials are currently underway which use a “risk-adapted” bladder preservation approach based on DDR gene mutational status to guide treatment strategies. The RETAIN BLADDER phase II trial, the Alliance A031701 trial, and the HCRN GU 16–257 (ref. 75; NCT03558087) are several examples exploring such an approach, but these bladder-sparing trials omit upfront local therapy (cystectomy or radiation) and employ chemotherapy-only regimens. In our cohort, we found that tumors with any DDR alteration were associated with improved locoregional control and mBI-EFS following standard-of-care TMT. In addition, 46% of patients with favorable mBI-EFS outcomes after TMT did not meet mutation criteria for inclusion in either of these chemotherapy-only trials.

To our knowledge, this is the largest study to date of integrated molecular profiling of samples from a cohort of TMT patients, and one of few studies leveraging both genomic and transcriptomic data, along with functional characterization of our findings. There are several limitations to this study, including its retrospective nature and small numbers. In addition, the lack of matched normal tissue could lead to overcalling alterations, although we used several approaches to account for this (41, 42). The mean TMB in the TCGA bladder cohort is 7.8 mutations/Mb (range 0.03–118.2; ref. 45), and although the mean TMB in our cohort [12.4 mutations/Mb (range, 3.4–31.5)] is significantly different (P < 0.001), it falls well within the range reported by TCGA and is not near the upper end of the range. Therefore, our computational approaches helped control for the lack of matched normal sample, and the molecular characteristics of our cohort are consistent with other large MIBC cohorts. We also lack a validation cohort to confirm our findings; however, a cohort of patients with MIBC who underwent TMT at multiple institutions from the NRG Oncology/Radiation Therapy Oncology Group trials will be analyzed as a separate study. Finally, tumor spatial heterogeneity cannot be inferred from this study as we do not have data from multiple areas of each tissue sample. Our study is strengthened by the use of a homogenously treated TMT cohort that is well-representative of the general MIBC patient population; the frequency of gene alterations in our TMT cohort were consistent with other large MIBC cohorts, including alterations in TP53 (n = 40, 52.6%), KMT2D (n = 31, 40.8%), and KMT2C (n = 16, 21.1%). The frequency of FGFR3 alterations was relatively low in this cohort (n = 6, 7.9%), consistent with a lower frequency in MIBC compared with non-invasive bladder cancer (76). We found mutation signatures previously associated with MIBC including Signatures 1, 2, 5, and 13 (77). The distribution of ERCC2 mutations in this cohort reflects the overall mutation distribution in cancers of the urogenital tract per TCGA bladder, supportive of the representative nature of our MIBC cohort. Therefore, our findings can be broadly applicable to patients with MIBC.

In summary, our study leverages integrated molecular profiling of a large cohort of patients with MIBC homogenously treated with TMT to evaluate for biomarkers of response. Alterations in a DDR gene, ERCC2, associated with improved mBI-EFS following standard-of-care TMT, as well as a trend toward improved OS. These data require validation but demonstrate early promise in elucidating novel biomarkers to guide precision oncology for radiation-based management in patients with MIBC.

C.-L. Wu reports other support from GoPath Diagnostics, as well as personal fees from OrigiMed Co. Ltd. outside the submitted work. A.S. Feldman reports research funding from Convergent Genomics. R.J. Lee reports personal fees from Exelixis, Bayer, Dendreon, and Blue Earth and grants and personal fees from Janssen outside the submitted work. E. Van Allen reports personal fees from Serinus Bio, Tango Therapeutics, Genome Medical, Genomic Life, Monte Rosa Therapeutics, Manifold Bio, Illumina, Enara Bio, Foaley & Hoag, and Riva Therapeutics; grants and personal fees from Novartis and Janssen; and grants from BMS and Sanofi outside the submitted work. In addition, E. Van Allen has a patent for Institutional patents filed on chromatin mutations and immunotherapy response, and methods for clinical interpretation pending and issued. T.S. Hong reports consulting for Synthetic Biologics, Novocure, Boston Scientific, Neogenomics, Merck, GSK, and NextCure; scientific advisory board member of PanTher Therapeutics (equity) and Lustgarten; and research funding (clinical trials) from Taiho, AstraZeneca, BMS, GSK, IntraOp, and Ipsen. Y. Liu reports personal fees from Veracyte during the conduct of the study, as well as personal fees from Veracyte outside the submitted work. E. Davicioni reports other support from Veracyte during the conduct of the study, as well as other support from Veracyte outside the submitted work; in addition, E. Davicioni has a patent for US201762448921 pending. E.A. Gibb reports other support from Veracyte during the conduct of the study, as well as other support from Veracyte outside the submitted work. K.W. Mouw reports personal fees from Riva Therapeutics, as well as personal fees from UroGen outside the submitted work. J.A. Efstathiou reports personal fees from Blue Earth Diagnostics, Boston Scientific, AstraZeneca, Merck, Roivant Pharma, Myovant Sciences, Janssen, Bayer Healthcare, Progenics Pharmaceuticals, Pfizer, Genentech, Gilead, Lantheus Medical Imaging, Angiodynamics, IBA, and Estellas outside the submitted work. D.T. Miyamoto reports grants from NCI, Radiation Oncology Institute, and George E. Safiol Foundation during the conduct of the study, as well as other support from Cardiff Oncology outside the submitted work. No disclosures were reported by the other authors.

S.C. Kamran: Data curation, formal analysis, investigation, visualization, methodology, writing–original draft, writing–review and editing. Y. Zhou: Formal analysis, investigation, methodology. K. Otani: Investigation. M. Drumm: Data curation, investigation. Y. Otani: Investigation. S. Wu: Investigation, writing–review and editing. C.-L. Wu: Investigation, writing–review and editing. A.S. Feldman: Investigation, writing–review and editing. M. Wszolek: Investigation, writing–review and editing. R.J. Lee: Investigation, writing–review and editing. P.J. Saylor: Investigation, writing–review and editing. J. Lennerz: Investigation, writing–review and editing. E. Van Allen: Investigation, writing–review and editing. H. Willers: Investigation, project administration, writing–review and editing. T.S. Hong: Funding acquisition, investigation, project administration, writing–review and editing. Y. Liu: Data curation, formal analysis, investigation, visualization. E. Davicioni: Data curation, software, supervision, investigation. E.A. Gibb: Data curation, software, investigation, visualization, writing–review and editing. W.U. Shipley: Funding acquisition, investigation, project administration. K.W. Mouw: Conceptualization, resources, formal analysis, supervision, investigation, project administration, writing–review and editing. J.A. Efstathiou: Conceptualization, resources, data curation, supervision, funding acquisition, investigation, writing–original draft, project administration, writing–review and editing. D.T. Miyamoto: Conceptualization, resources, supervision, funding acquisition, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing.

We gratefully acknowledge Drs. David B. Solit and Sizhi P. Gao for providing the ERCC2-mutant KE1 and SC14 cell lines. This work was supported by grants from the NIH (C06 CA059267 to J.A. Efstathiou; U01CA220714 to H. Willers; R0125737 to K.W. Mouw; R01CA259007 to D.T. Miyamoto), the Radiation Oncology Institute (ROI2021–9151 to D.T. Miyamoto), and the George E. Safiol Foundation (to D.T. Miyamoto).

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Baumann
M
,
Krause
M
,
Overgaard
J
,
Debus
J
,
Bentzen
SM
,
Daartz
J
, et al
.
Radiation oncology in the era of precision medicine
.
Nat Rev Cancer
2016
;
16
:
234
49
.
2.
Huddart
RA
,
Hall
E
,
Lewis
R
,
Birtle
A
.
Life and death of spare (selective bladder preservation against radical excision): reflections on why the spare trial closed
.
BJU Int
2010
;
106
:
753
5
.
3.
James
ND
,
Hussain
SA
,
Hall
E
,
Jenkins
P
,
Tremlett
J
,
Rawlings
C
, et al
.
Radiotherapy with or without chemotherapy in muscle-invasive bladder cancer
.
N Engl J Med
2012
;
366
:
1477
88
.
4.
Rosen
E
,
Drilon
A
,
Chakravarty
D
.
Precision oncology: 2022 in review
.
Cancer Discov
2022
;
12
:
2747
53
.
5.
Hall
WA
,
Bergom
C
,
Thompson
RF
,
Baschnagel
AM
,
Vijayakumar
S
,
Willers
H
, et al
.
Precision oncology and genomically guided radiation therapy: a report from the American Society for Radiation Oncology/American Association of Physicists in Medicine/National Cancer Institute precision medicine conference
.
Int J Radiat Oncol Biol Phys
2018
;
101
:
274
84
.
6.
Kirsch
DG
,
Diehn
M
,
Kesarwala
AH
,
Maity
A
,
Morgan
MA
,
Schwarz
JK
, et al
.
The future of radiobiology
.
J Natl Cancer Inst
2018
;
110
:
329
40
.
7.
Riaz
N
,
Sherman
E
,
Pei
X
,
Schöder
H
,
Grkovski
M
,
Paudyal
R
, et al
.
Precision radiotherapy: reduction in radiation for oropharyngeal cancer in the 30 ROC trial
.
J Natl Cancer Inst
2021
;
113
:
742
51
.
8.
Knijnenburg
TA
,
Wang
L
,
Zimmermann
MT
,
Chambwe
N
,
Gao
GF
,
Cherniack
AD
, et al
.
Genomic and molecular landscape of DNA damage repair deficiency across The Cancer Genome Atlas
.
Cell Rep
2018
;
23
:
239
54
.
9.
Pitter
KL
,
Casey
DL
,
Lu
YC
,
Hannum
M
,
Zhang
Z
,
Song
X
, et al
.
Pathogenic ATM mutations in cancer and a genetic basis for radiotherapeutic efficacy
.
J Natl Cancer Inst
2021
;
113
:
266
73
.
10.
Groelly
FJ
,
Fawkes
M
,
Dagg
RA
,
Blackford
AN
,
Tarsounas
M
.
Targeting DNA damage response pathways in cancer
.
Nat Rev Cancer
2023
;
23
:
78
94
.
11.
Binkley
MS
,
Jeon
YJ
,
Nesselbush
M
,
Moding
EJ
,
Nabet
BY
,
Almanza
D
, et al
.
KEAP1/NFE2L2 mutations predict lung cancer radiation resistance that can be targeted by glutaminase inhibition
.
Cancer Discov
2020
;
10
:
1826
41
.
12.
Kamran
SC
,
Lennerz
JK
,
Margolis
CA
,
Liu
D
,
Reardon
B
,
Wankowicz
SA
, et al
.
Integrative molecular characterization of resistance to neoadjuvant chemoradiation in rectal cancer
.
Clin Cancer Res
2019
;
25
:
5561
71
.
13.
Hong
TS
,
Wo
JY
,
Borger
DR
,
Yeap
BY
,
McDonnell
EI
,
Willers
H
, et al
.
Phase II study of proton-based stereotactic body radiation therapy for liver metastases: importance of tumor genotype
.
J Natl Cancer Inst
2017
;
109
.
14.
Sitthideatphaiboon
P
,
Galan-Cobo
A
,
Negrao
MV
,
Qu
X
,
Poteete
A
,
Zhang
F
, et al
.
STK11/LKB1 mutations in NSCLC are associated with KEAP1/NRF2-dependent radiotherapy resistance targetable by glutaminase inhibition
.
Clin Cancer Res
2021
;
27
:
1720
33
.
15.
National Comprehensive Cancer Network
.
Bladder cancer - version 6.2021
.
2021
. Available from
: https://www.nccn.org/professionals/physician_gls/pdf/bladder.pdf.
16.
Giacalone
NJ
,
Shipley
WU
,
Clayman
RH
,
Niemierko
A
,
Drumm
M
,
Heney
NM
, et al
.
Long-term outcomes after bladder-preserving tri-modality therapy for patients with muscle-invasive bladder cancer: an updated analysis of the Massachusetts General Hospital experience
.
Eur Urol
2017
;
71
:
952
60
.
17.
Kulkarni
GS
,
Hermanns
T
,
Wei
Y
,
Bhindi
B
,
Satkunasivam
R
,
Athanasopoulos
P
, et al
.
Propensity score analysis of radical cystectomy versus bladder-sparing trimodal therapy in the setting of a multidisciplinary bladder cancer clinic
.
J Clin Oncol
2017
;
35
:
2299
305
.
18.
Mak
RH
,
Hunt
D
,
Shipley
WU
,
Efstathiou
JA
,
Tester
WJ
,
Hagan
MP
, et al
.
Long-term outcomes in patients with muscle-invasive bladder cancer after selective bladder-preserving combined-modality therapy: a pooled analysis of radiation therapy oncology group protocols 8802, 8903, 9506, 9706, 9906, and 0233
.
J Clin Oncol
2014
;
32
:
3801
9
.
19.
Zlotta
AR
,
Ballas
LK
,
Niemierko
A
,
Lajkosz
K
,
Kuk
C
,
Miranda
G
, et al
.
Radical cystectomy versus trimodality therapy for muscle-invasive bladder cancer: a multi-institutional propensity score matched and weighted analysis
.
Lancet Oncol
2023
;
24
:
669
81
.
20.
Teo
MY
,
Bambury
RM
,
Zabor
EC
,
Jordan
E
,
Al-Ahmadie
H
,
Boyd
ME
, 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
.
21.
Plimack
ER
,
Dunbrack
RL
,
Brennan
TA
,
Andrake
MD
,
Zhou
Y
,
Serebriiskii
IG
, et al
.
Defects in DNA repair genes predict response to neoadjuvant cisplatin-based chemotherapy in muscle-invasive bladder cancer
.
Eur Urol
2015
;
68
:
959
67
.
22.
Choi
W
,
Porten
S
,
Kim
S
,
Willis
D
,
Plimack
ER
,
Hoffman-Censits
J
, et al
.
Identification of distinct basal and
luminal
subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy
.
Cancer Cell
2014
;
25
:
152
65
.
23.
Seiler
R
,
Ashab
HAD
,
Erho
N
,
van Rhijn
BWG
,
Winters
B
,
Douglas
J
, et al
.
Impact of molecular subtypes in muscle-invasive bladder cancer on predicting response and survival after neoadjuvant chemotherapy
.
Eur Urol
2017
;
72
:
544
54
.
24.
Sjödahl
G
,
Abrahamsson
J
,
Holmsten
K
,
Bernardo
C
,
Chebil
G
,
Eriksson
P
, et al
.
Different responses to neoadjuvant chemotherapy in urothelial carcinoma molecular subtypes
.
Eur Urol
2022
;
81
:
523
32
.
25.
Efstathiou
JA
,
Mouw
KW
,
Gibb
EA
,
Liu
Y
,
Wu
CL
,
Drumm
MR
, et al
.
Impact of immune and stromal infiltration on outcomes following bladder-sparing trimodality therapy for muscle-invasive bladder cancer
.
Eur Urol
2019
;
76
:
59
68
.
26.
Miyamoto
DT
,
Mouw
KW
,
Feng
FY
,
Shipley
WU
,
Efstathiou
JA
.
Molecular biomarkers in bladder preservation therapy for muscle-invasive bladder cancer
.
Lancet Oncol
2018
;
19
:
e683
e95
.
27.
Magliocco
AM
,
Moughan
J
,
Miyamoto
DT
,
Simko
J
,
Shipley
WU
,
Gray
PJ
, et al
.
Analysis of MRE11 and mortality among adults with muscle-invasive bladder cancer managed with trimodality therapy
.
JAMA Netw Open
2022
;
5
:
e2242378
.
28.
Liu
D
,
Abbosh
P
,
Keliher
D
,
Reardon
B
,
Miao
D
,
Mouw
K
, et al
.
Mutational patterns in chemotherapy resistant muscle-invasive bladder cancer
.
Nat Commun
2017
;
8
:
2193
.
29.
Cibulskis
K
,
Lawrence
MS
,
Carter
SL
,
Sivachenko
A
,
Jaffe
D
,
Sougnez
C
, et al
.
Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples
.
Nat Biotechnol
2013
;
31
:
213
9
.
30.
Cibulskis
K
,
McKenna
A
,
Fennell
T
,
Banks
E
,
DePristo
M
,
Getz
G
.
ContEst:estimating cross-contamination of human samples in next-generation sequencing data
.
Bioinformatics
2011
;
27
:
2601
2
.
31.
Saunders
CT
,
Wong
WS
,
Swamy
S
,
Becq
J
,
Murray
LJ
,
Cheetham
RK
.
Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs
.
Bioinformatics
2012
;
28
:
1811
7
.
32.
Costello
M
,
Pugh
TJ
,
Fennell
TJ
,
Stewart
C
,
Lichtenstein
L
,
Meldrim
JC
, et al
.
Discovery and characterization of artifactual mutations in deep coverage targeted capture sequencing data due to oxidative DNA damage during sample preparation
.
Nucleic Acids Res
2013
;
41
:
e67
.
33.
Taylor-Weiner
A
,
Stewart
C
,
Giordano
T
,
Miller
M
,
Rosenberg
M
,
Macbeth
A
, et al
.
DeTiN: overcoming tumor-in-normal contamination
.
Nat Methods
2018
;
15
:
531
4
.
34.
Landau
DA
,
Carter
SL
,
Stojanov
P
,
McKenna
A
,
Stevenson
K
,
Lawrence
MS
, et al
.
Evolution and impact of subclonal mutations in chronic lymphocytic leukemia
.
Cell
2013
;
152
:
714
26
.
35.
Lawrence
MS
,
Stojanov
P
,
Mermel
CH
,
Robinson
JT
,
Garraway
LA
,
Golub
TR
, et al
.
Discovery and saturation analysis of cancer genes across 21 tumor types
.
Nature
2014
;
505
:
495
501
.
36.
Carter
SL
,
Cibulskis
K
,
Helman
E
,
McKenna
A
,
Shen
H
,
Zack
T
, et al
.
Absolute quantification of somatic DNA alterations in human cancer
.
Nat Biotechnol
2012
;
30
:
413
21
.
37.
McKenna
A
,
Hanna
M
,
Banks
E
,
Sivachenko
A
,
Cibulskis
K
,
Kernytsky
A
, et al
.
The genome analysis toolkit: a mapreduce framework for analyzing next-generation DNA sequencing data
.
Genome Res
2010
;
20
:
1297
303
.
39.
McLaren
W
,
Gil
L
,
Hunt
SE
,
Riat
HS
,
Ritchie
GR
,
Thormann
A
, et al
.
The ensembl variant effect predictor
.
Genome Biol
2016
;
17
:
122
.
40.
Ramos
AH
,
Lichtenstein
L
,
Gupta
M
,
Lawrence
MS
,
Pugh
TJ
,
Saksena
G
, et al
.
Oncotator: cancer variant annotation tool
.
Hum Mutat
2015
;
36
:
E2423
9
.
41.
Karczewski
KJ
,
Francioli
LC
,
Tiao
G
,
Cummings
BB
,
Alföldi
J
,
Wang
Q
, et al
.
The mutational constraint spectrum quantified from variation in 141,456 humans
.
Nature
2020
;
581
:
434
43
.
42.
AACR Project GENIE Consortium
.
AACR project GENIE: powering precision medicine through an international consortium
.
Cancer Discov
2017
;
7
:
818
31
.
43.
Robinson
JT
,
Thorvaldsdottir
H
,
Winckler
W
,
Guttman
M
,
Lander
ES
,
Getz
G
, et al
.
Integrative genomics viewer
.
Nat Biotechnol
2011
;
29
:
24
6
.
44.
Thorvaldsdottir
H
,
Robinson
JT
,
Mesirov
JP
.
Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration
.
Briefings Bioinf
2013
;
14
:
178
92
.
45.
Robertson
AG
,
Kim
J
,
Al-Ahmadie
H
,
Bellmunt
J
,
Guo
G
,
Cherniack
AD
, et al
.
Comprehensive molecular characterization of muscle-invasive bladder cancer
.
Cell
2018
;
174
:
1033
.
46.
Chakravarty
D
,
Gao
J
,
Phillips
SM
,
Kundra
R
,
Zhang
H
,
Wang
J
, et al
.
OncoKB: a precision oncology knowledge base
.
JCO Precis Oncol
2017
;
2017
:
PO.17.00011
.
47.
Rosenthal
R
,
McGranahan
N
,
Herrero
J
,
Taylor
BS
,
Swanton
C
.
DeconstructSigs: delineating mutational processes in single tumors distinguishes DNA repair deficiencies and patterns of carcinoma evolution
.
Genome Biol
2016
;
17
:
31
.
48.
Batista da
Costa J
,
Gibb
EA
,
Bivalacqua
TJ
,
Liu
Y
,
Oo
HZ
,
Miyamoto
DT
, et al
.
Molecular characterization of neuroendocrine-like bladder cancer
.
Clin Cancer Res
2019
;
25
:
3908
20
.
49.
Sweis
RF
,
Spranger
S
,
Bao
R
,
Paner
GP
,
Stadler
WM
,
Steinberg
G
, et al
.
Molecular drivers of the non-T cell-inflamed tumor microenvironment in urothelial bladder cancer
.
Cancer Immunol Res
2016
;
4
:
563
8
.
50.
Kamoun
A
,
de Reyniès
A
,
Allory
Y
,
Sjödahl
G
,
Robertson
AG
,
Seiler
R
, et al
.
A consensus molecular classification of muscle-invasive bladder cancer
.
Eur Urol
2020
;
77
:
420
33
.
51.
Börcsök
J
,
Sztupinszki
Z
,
Bekele
R
,
Gao
SP
,
Diossy
M
,
Samant
AS
, et al
.
Identification of a synthetic lethal relationship between nucleotide excision repair deficiency and irofulven sensitivity in urothelial cancer
.
Clin Cancer Res
2021
;
27
:
2011
22
.
52.
Li
Q
,
Damish
AW
,
Frazier
Z
,
Liu
D
,
Reznichenko
E
,
Kamburov
A
, et al
.
ERCC2 helicase domain mutations confer nucleotide excision repair deficiency and drive cisplatin sensitivity in muscle-invasive bladder cancer
.
Clin Cancer Res
2019
;
25
:
977
88
.
53.
Di Veroli
GY
,
Fornari
C
,
Wang
D
,
Mollard
S
,
Bramhall
JL
,
Richards
FM
, et al
.
Combenefit: an interactive platform for the analysis and visualization of drug combinations
.
Bioinformatics
2016
;
32
:
2866
8
.
54.
Alexandrov
LB
,
Nik-Zainal
S
,
Wedge
DC
,
Aparicio
SA
,
Behjati
S
,
Biankin
AV
, et al
.
Signatures of mutational processes in human cancer
.
Nature
2013
;
500
:
415
21
.
55.
Kim
J
,
Mouw
KW
,
Polak
P
,
Braunstein
LZ
,
Kamburov
A
,
Kwiatkowski
DJ
, et al
.
Somatic ERCC2 mutations are associated with a distinct genomic signature in urothelial tumors
.
Nat Genet
2016
;
48
:
600
6
.
56.
Mossanen
M
,
Carvalho
FLF
,
Muralidhar
V
,
Preston
MA
,
Reardon
B
,
Conway
JR
, et al
.
Genomic features of muscle-invasive bladder cancer arising after prostate radiotherapy
.
Eur Urol
2022
;
81
:
466
73
.
57.
van Rhijn
BW
,
Lurkin
I
,
Radvanyi
F
,
Kirkels
WJ
,
van der Kwast
TH
,
Zwarthoff
EC
.
The fibroblast growth factor receptor 3 (FGFR3) mutation is a strong indicator of superficial bladder cancer with low recurrence rate
.
Cancer Res
2001
;
61
:
1265
8
.
58.
de Jong
JJ
,
Liu
Y
,
Robertson
AG
,
Seiler
R
,
Groeneveld
CS
,
van der Heijden
MS
, et al
.
Long non-coding RNAs identify a subset of
luminal
muscle-invasive bladder cancer patients with favorable prognosis
.
Genome Medicine
2019
;
11
:
60
.
59.
Sjodahl
G
,
Lauss
M
,
Lovgren
K
,
Chebil
G
,
Gudjonsson
S
,
Veerla
S
, et al
.
A molecular taxonomy for urothelial carcinoma
.
Clin Cancer Res
2012
;
18
:
3377
86
.
60.
Liberzon
A
,
Birger
C
,
Thorvaldsdottir
H
,
Ghandi
M
,
Mesirov
JP
,
Tamayo
P
.
The Molecular Signatures Database (MSigDB) hallmark gene set collection
.
Cell Syst
2015
;
1
:
417
25
.
61.
Chipidza
FE
,
Alshalalfa
M
,
Mahal
BA
,
Karnes
RJ
,
Liu
Y
,
Davicioni
E
, et al
.
Development and validation of a novel TP53 mutation signature that predicts risk of metastasis in primary prostate cancer
.
Clin Genitourin Cancer
2021
;
19
:
246
54
.
62.
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, multicenter, phase II trial
.
Lancet
2016
;
387
:
1909
20
.
63.
Marcus
L
,
Fashoyin-Aje
LA
,
Donoghue
M
,
Yuan
M
,
Rodriguez
L
,
Gallagher
PS
, et al
.
FDA approval summary: pembrolizumab for the treatment of tumor mutational burden-high solid tumors
.
Clin Cancer Res
2021
;
27
:
4685
9
.
64.
Shaverdian
N
,
Shepherd
AF
,
Li
X
,
Offin
M
,
Lengel
HB
,
Gelblum
DY
, et al
.
Effects of tumor mutational burden and gene alterations associated with radiation response on outcomes of postoperative radiation therapy in non-small cell lung cancer
.
Int J Radiat Oncol Biol Phys
2022
;
113
:
335
44
.
65.
Van Allen
EM
,
Mouw
KW
,
Kim
P
,
Iyer
G
,
Wagle
N
,
Al-Ahmadie
H
, et al
.
Somatic ERCC2 mutations correlate with cisplatin sensitivity in muscle-invasive urothelial carcinoma
.
Cancer Discov
2014
;
4
:
1140
53
.
66.
Singh
P
,
Tangen
C
,
Efstathiou
JA
,
Lerner
SP
,
Jhavar
SG
,
Hahn
NM
, et al
.
INTACT: Phase III randomized trial of concurrent chemoradiotherapy with or without atezolizumab in localized muscle-invasive bladder cancer—SWOG/NRG1806
.
J Clin Oncol
2020
;
38
:
TPS586
TPS
.
67.
Font
A
,
Taron
M
,
Gago
JL
,
Costa
C
,
Sánchez
JJ
,
Carrato
C
, et al
.
BRCA1 mRNA expression and outcome to neoadjuvant cisplatin-based chemotherapy in bladder cancer
.
Ann Oncol
2011
;
22
:
139
44
.
68.
Teo
MY
,
Seier
K
,
Ostrovnaya
I
,
Regazzi
AM
,
Kania
BE
,
Moran
MM
, 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
.
69.
Dai
J
,
Jiang
M
,
He
K
,
Wang
H
,
Chen
P
,
Guo
H
, et al
.
DNA damage response and repair gene alterations increase tumor mutational burden and promote poor prognosis of advanced lung cancer
.
Front Oncol
2021
;
11
:
708294
.
70.
Song
Y
,
Huang
J
,
Liang
D
,
Hu
Y
,
Mao
B
,
Li
Q
, et al
.
DNA damage repair gene mutations are indicative of a favorable prognosis in colorectal cancer treated with immune checkpoint inhibitors
.
Front Oncol
2020
;
10
:
549777
.
71.
Scharer
OD
.
Nucleotide excision repair in eukaryotes
.
Cold Spring Harb Perspect Biol
2013
;
5
:
a012609
.
72.
Desai
NB
,
Scott
SN
,
Zabor
EC
,
Cha
EK
,
Hreiki
J
,
Sfakianos
JP
, et al
.
Genomic characterization of response to chemoradiation in urothelial bladder cancer
.
Cancer
2016
;
122
:
3715
23
.
73.
Sjödahl
G
,
Abrahamsson
J
,
Bernardo
C
,
Eriksson
P
,
Höglund
M
,
Liedberg
F
.
Molecular subtypes as a basis for stratified use of neoadjuvant chemotherapy for muscle-invasive bladder cancer-a narrative review
.
Cancers
2022
;
14
:
1692
.
74.
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
.
75.
Galsky
MD
,
Daneshmand
S
,
Chan
KG
,
Dorff
TB
,
Cetnar
JP
,
Neil
BO
, et al
.
Phase II trial of gemcitabine, cisplatin, plus nivolumab with selective bladder sparing in patients with muscle-invasive bladder cancer (MIBC): HCRN GU 16–257
.
J Clin Oncol
2021
;
39
:
4503
.
76.
Neuzillet
Y
,
van Rhijn
BW
,
Prigoda
NL
,
Bapat
B
,
Liu
L
,
Bostrom
PJ
, et al
.
FGFR3 mutations, but not FGFR3 expression and FGFR3 copy-number variations, are associated with favorable non–muscle-invasive bladder cancer
.
Virchows Arch
2014
;
465
:
207
13
.
77.
Roberts
SA
,
Lawrence
MS
,
Klimczak
LJ
,
Grimm
SA
,
Fargo
D
,
Stojanov
P
, et al
.
An APOBEC cytidine deaminase mutagenesis pattern is widespread in human cancers
.
Nat Genet
2013
;
45
:
970
6
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.

Supplementary data