Purpose: The survival of patients with clear cell metastatic renal cell carcinoma (cc-mRCC) has improved substantially since the introduction of tyrosine kinase inhibitors (TKI). With the fact that TKIs interact with immune responses, we investigated whether polymorphisms of genes involved in immune checkpoints are related to the clinical outcome of cc-mRCC patients treated with sunitinib as first TKI.

Experimental Design: Twenty-seven single-nucleotide polymorphisms (SNP) in CD274 (PD-L1), PDCD1 (PD-1), and CTLA-4 were tested for a possible association with progression-free survival (PFS) and overall survival (OS) in a discovery cohort of 550 sunitinib-treated cc-mRCC patients. SNPs with a significant association (P < 0.05) were tested in an independent validation cohort of 138 sunitinib-treated cc-mRCC patients. Finally, data of the discovery and validation cohort were pooled for meta-analysis.

Results:CTLA-4 rs231775 and CD274 rs7866740 showed significant associations with OS in the discovery cohort after correction for age, gender, and Heng prognostic risk group [HR, 0.84; 95% confidence interval (CI), 0.72–0.98; P = 0.028, and HR, 0.73; 95% CI, 0.54–0.99; P = 0.047, respectively]. In the validation cohort, the associations of both SNPs with OS did not meet the significance threshold of P < 0.05. After meta-analysis, CTLA-4 rs231775 showed a significant association with OS (HR, 0.83; 95% CI, 0.72–0.95; P = 0.008). Patients with the GG genotype had longer OS (35.1 months) compared with patients with an AG (30.3 months) or AA genotype (24.3 months). No significant associations with PFS were found.

Conclusions: The G-allele of rs231775 in the CTLA-4 gene is associated with an improved OS in sunitinib-treated cc-mRCC patients and could potentially be used as a prognostic biomarker. Clin Cancer Res; 24(10); 2350–6. ©2018 AACR.

Patients with metastatic renal cell carcinoma (mRCC) have several treatment options, including therapies targeting the angiogenesis pathway and immune checkpoint pathway, but there is no biomarker to predict clinical outcome. It has been reported that there is an interaction between both pathways. The immune checkpoint, such as PD-L1 expression, has been associated with treatment outcome of tyrosine kinase inhibitors (TKI). We thus hypothesized that genetic variants in the immune checkpoint pathway might be used to predict the outcome of the TKI-sunitinib treatment. In a discovery cohort and subsequent combined cohort of sunitinib-treated patients with clear cell mRCC, we observed that patients with the GG genotype of CTLA-4 rs231775 had longer OS compared with patients with an AG or AA genotype. Yet, no significant associations with PFS were found. This study suggests that CTLA-4 rs231775 is a prognostic biomarker.

Renal cell carcinoma (RCC) is the most common type of kidney cancer in adults. Almost 70% of RCC is of clear cell histology, and 20% to 25% of patients have metastatic spread by the time they are diagnosed with RCC (1, 2). Even though new cases of RCC remain relatively few compared with other more frequent malignancies, the global incidence and mortality are steadily increasing at a rate of 2% to 3% per decade (2).

For decades, the outcome for metastatic RCC (mRCC) treatment with IL2 and IFNα was disappointing. With the discovery of von Hippel–Lindau (VHL) gene mutations in the majority of clear cell RCC, therapies targeting angiogenesis were introduced in early 2003 including tyrosine kinase inhibitors (TKI), mTOR inhibitors, and the anti-VEGF mAb bevacizumab. With an improved understanding of how tumor cells evade antitumor response and how T cells can be redirected through activating or inhibiting receptors, the immune checkpoint pathway broadens the knowledge of the biological behavior of RCC. Checkpoint inhibitors, such as programmed death-1 (PD-1) inhibitors (pembrolizumab and nivolumab), PD-1 ligand (PD-L1) inhibitor (atezolizumab), and cytotoxic T-lymphocyte antigen 4 (CTLA-4) inhibitor (ipilimumab), have emerged as promising RCC treatments (3). Given the fact that angiogenesis pathway inhibitors interact with immune responses (4, 5), the potential synergy between checkpoint inhibitors and angiogenesis inhibitors has led to several combination trials in mRCC, such as sunitinib or pazopanib combined with nivolumab (6).

Upon activation, some T-cell receptors (such as CD28) positively regulate T cells, leading to tumor destruction, whereas others (such as PD-1 and CTLA-4) negatively regulate T cells, resulting in prolonged tumor survival. When PD-1 binds to PD-L1, the activation of T cells is inhibited, resulting in the suppression of T-cell attack and tumor immune escape (7). After binding to B7.1 (CD80) and B7.2 (CD86), CTLA-4 reduces the activated T-cell response to tumor cells, thereby failing to halt tumor progression (8). Generally, there is a balance among the receptors which makes the immune response of T cells maintain a proper intensity in order to protect normal cells from collateral damage (7). However, overexpression of PD-L1 and CTLA-4 was observed in patients with many tumor types, including clear cell mRCC (cc-mRCC; refs. 8–11).

Recently, the prognostic and/or predictive value of aforementioned immune checkpoints has been evaluated in several tumor types. It was reported in RCC patients that higher PD-L1 expression was associated with a larger tumor size and a higher risk of death (10, 12, 13). Subsequently, the association of higher PD-L1 tumor expression with shorter progression-free survival (PFS) and/or overall survival (OS) in mRCC patients treated by TKIs was reported by Choueiri and colleagues (14) and Fukuda and colleagues (15). The possible association between CTLA-4 expression and OS has been investigated in different tumor types with controversial results. A meta-analysis was conducted to pool all data, but consistent results were lacking possibly due to heterogeneity of the tumor types, differences in experimental methods (immunohistochemistry, ELISA, or PCR), and varying sample sources (blood or tumor) used in the studies (16).

Thus far, there are no studies investigating the role of single-nucleotide polymorphisms (SNP) in genes related to the immune checkpoints in cc-mRCC patients. In this study, we investigated whether genetic variants within CD274 (PD-L1), PDCD1 (PD-1), and CTLA-4 could be useful as predictive biomarkers for sunitinib treatment outcome in cc-mRCC patients.

Discovery cohort

The discovery cohort was composed of patients who participated in the EuroTARGET project (17). In brief, the EuroTARGET is a multicenter observational study aiming to identify biomarkers for prediction of response to TKI treatment in mRCC, within which a hypothesis-free genome-wide association study was conducted on sunitinib efficacy in cc-mRCC patients. Thus, clinical information and germline DNA variant chip data (Illumina Human OmniExpress BeadChip) were available (17). After quality control checks, 679,324 SNPs met the quality criteria applied (Supplementary Document S1). A total of 6,540,327 SNPs were available after imputation by impute2 using the 1000 Genomes (phase III integrated data set of 2,504 individuals) as reference panel. In the present study, patients with cc-mRCC who were treated by sunitinib as first TKI were included in the discovery cohort.

SNP selection

In order to capture the genetic variation in the target genes, genomic sequences of CD274, PDCD1, and CTLA-4 genes were retrieved from the 1000 Genomes Project (GRCh37.p13). For our study, we selected tagging SNPs using the SNP Tagger approach of the Haploview software package (version 4.2). Pairwise tagging with r2 threshold 0.8 was used. Based on the 1000 Genomes Project, only SNPs with minor allele frequency (MAF) of 0.05 or higher were included. This approach allows to cover the common genetic variations in candidate genes (18), while reducing the number of tested SNPs.

Genotyping results of the selected tagging SNPs were extracted from the EuroTARGET project. To assess the quality of imputation, an estimated imputation “info” score was used, which was computed by impute 2 and took values between 0 and 1. An “info” score near 1 indicates that an SNP has been imputed with high certainty. SNPs with an “info” score lower than 0.9 were excluded from statistical analysis.

Validation cohort and genotyping

Positive hits were tested in an independent validation cohort consisting of cc-mRCC patients treated with sunitinib as first TKI. Details have been described previously (19). In brief, patients were enrolled between 2004 and 2010 in five medical centers in the Netherlands and in the Cleveland Clinic Foundation Taussig Cancer Institute in the United States. Germline DNA was isolated from whole blood, serum, plasma, or peripheral blood mononuclear cell samples and genotyped. A flowchart of the study design is shown in Fig. 1. Genotyping methods are described in Supplementary Document S2.

Figure 1.

Flowchart of the study design.

Figure 1.

Flowchart of the study design.

Close modal

Statistical analysis

Study endpoints consisted of PFS and OS. PFS was defined as the time in months between the first day of sunitinib treatment and the date of progressive disease (according to RECIST version 1.1) or death due to cancer. If no progression was observed during sunitinib treatment, PFS was censored at the time of the last follow-up or death due to other reasons (whichever occurred first). OS was defined as the time in months between the first day of sunitinib treatment and the date of death or the date at which patients were last known to be alive. In the discovery cohort, associations between tagging SNPs and endpoints were univariately tested using an additive genetic model. Individual SNPs with P values lower than 0.1 were tested in a multivariable Cox-regression model together with well-established covariates age, gender, and Heng prognostic risk group (20), which have been repeatedly associated with PFS and OS. Subsequently, individual SNPs with a P value < 0.05 in the multivariable analysis were tested in the validation cohort using the same Cox-regression model and endpoint definitions. Finally, data from the discovery and validation cohort were pooled using a fixed effect meta-analysis. Patient characteristics between discovery and validation cohorts were compared by t test, χ2 test, Mann–Whitney U test, or Kaplan–Meier survival analysis with the log-rank test depending on the type of data. All tests were two-sided and carried out using SPSS Statistical Package for Windows (version 23.0, IBM Corp). R (version 2.3.2) and package MICE (Multivariate Imputation by Chained Equations; ref. 21) were used to impute missing values for variables included in Heng prognostic risk group (World Health Organization performance status, hemoglobin, neutrophil count, thrombocytes, calcium, and time from diagnosis until start of sunitinib).

Bioinformatic analysis

We used several bioinformatic tools to assess the possible functional relevance for genetic variants showing significant associations with endpoints in the final pooled analysis. The Genotype-Tissue Expression (GTEx, http://www.gtexportal.org/home/) portal was employed to identify potential associations between genetic variants and gene expression levels (eQTL) in all available tissues (22). The OncoLnc (http://www.oncolnc.org/; ref. 23) was used to link mRNA expression levels to the survival data from The Cancer Genome Atlas Kidney Renal Clear Cell Carcinoma (TCGA-KIRC) collection (24), which included 537 patients with different stages of cc-RCC.

Patient characteristics

A total of 550 and 138 sunitinib-treated cc-mRCC patients were included in the discovery and validation cohort, respectively. Patient characteristics for the discovery and validation cohorts are presented in Table 1. In brief, patients included in the discovery cohort were older (median age 63) than those in the validation cohort (median age 59). Nearly 93% of patients in the discovery cohort were classified in the intermediate or poor Heng prognostic risk group, whereas in the validation cohort, only 72% belonged to these prognostic groups. Consequently, patients in the validation cohort had longer PFS and OS compared with those in the discovery cohort. Only 86 (16%) patients in the discovery cohort and 14 (10%) patients in the validation cohort had one or more values missing as variables required for inclusion in a Heng prognostic risk group.

Table 1.

Patient characteristics

Discovery cohortValidation cohortPooled cohort
Characteristics(n = 550)(n = 138)(n = 688)P valuea
Median age at sunitinib start in years (range) 63 (33–87) 59 (28–80) 62 (28–87) <0.05 
Male 405 (74%) 97 (70%) 502 (73%) 0.453 
Heng prognostic risk groupb    <0.05 
 Good (0 risk factor) 38 (6.9%) 38 (27.5%) 76 (11.0%)  
 Intermediate (1–2 risk factors) 291 (52.9%) 68 (49.3%) 359 (52.2%)  
 Poor (3–6 risk factors) 221 (40.2%) 32 (23.2%) 235 (36.8%)  
Sunitinib starting dose    0.011 
 50 mg 482 (87.6%) 134 (97.1%) 616 (89.5%)  
 37.5 mg 46 (8.4%) 4 (2.9%) 50 (7.3%)  
 25 mg 20 (3.6%) 20 (2.9%)  
 12.5 mg 2 (0.4%) 2 (0.3%)  
Mean number of metastatic sites 2.1 2.3 2.1 0.024 
PFS     
 Median in months 12.6 20.1 13.4 0.099 
 Number of events 341 (62.0%) 95 (68.8%) 436 (63.4%) 0.140 
OS     
 Median in months 28.8 33.4 29.1 0.025 
 Number of deaths 341 (62.0%) 88 (63.8%) 429 (62.4%) 0.768 
Discovery cohortValidation cohortPooled cohort
Characteristics(n = 550)(n = 138)(n = 688)P valuea
Median age at sunitinib start in years (range) 63 (33–87) 59 (28–80) 62 (28–87) <0.05 
Male 405 (74%) 97 (70%) 502 (73%) 0.453 
Heng prognostic risk groupb    <0.05 
 Good (0 risk factor) 38 (6.9%) 38 (27.5%) 76 (11.0%)  
 Intermediate (1–2 risk factors) 291 (52.9%) 68 (49.3%) 359 (52.2%)  
 Poor (3–6 risk factors) 221 (40.2%) 32 (23.2%) 235 (36.8%)  
Sunitinib starting dose    0.011 
 50 mg 482 (87.6%) 134 (97.1%) 616 (89.5%)  
 37.5 mg 46 (8.4%) 4 (2.9%) 50 (7.3%)  
 25 mg 20 (3.6%) 20 (2.9%)  
 12.5 mg 2 (0.4%) 2 (0.3%)  
Mean number of metastatic sites 2.1 2.3 2.1 0.024 
PFS     
 Median in months 12.6 20.1 13.4 0.099 
 Number of events 341 (62.0%) 95 (68.8%) 436 (63.4%) 0.140 
OS     
 Median in months 28.8 33.4 29.1 0.025 
 Number of deaths 341 (62.0%) 88 (63.8%) 429 (62.4%) 0.768 

aP value shows the comparison between discovery and validation cohorts using the t test, χ2 test, Mann–Whitney U test, or Kaplan–Meier survival analysis with the log-rank test, depending on the type of data.

bThe Heng prognostic risk group is based on six risk factors: World Health Organization performance status (≥1), low hemoglobin [<lower limit of normal (LLN); for males, LLN = 8.1 mmol/L or 13 g/dL; for females, LLN = 7.1 mmol/L or 11.5 g/dL], high calcium (>2.5 mmol/L), time from initial diagnosis to treatment with sunitinib (<1 year), neutrophil count [>upper limit of normal (ULN)], and thrombocytes (>ULN; ref. 20). R (version 2.3.2) and package MICE (21) were used to impute missing values for variables included in the Heng prognostic risk group.

Discovery study on the associations of the selected SNPs with PFS and OS

In the discovery analysis, we selected a total of 34 tagging SNPs in CD274, PDCD1, and CTLA-4 according to our predefined tagging SNP approach. Genotype results for 27 of 34 tagging SNPs were imputed based on 1000 genomes project. Seven SNPs with “info” score lower than 0.9 were excluded from statistical analysis (Supplementary Table S1).

In the univariate analysis, CTLA-4 rs231775 and CD274 rs7866740 were associated with PFS and OS with P values lower than 0.1 (Supplementary Table S1). After correction for age, gender, and Heng prognostic risk group, CTLA-4 rs231775 and CD274 rs7866740 remained significantly associated with OS [HR, 0.84; 95% confidence interval (CI), 0.72–0.98; P = 0.028 and HR, 0.73; 95% CI, 0.54–0.99; P = 0.047, respectively). The minor allele carriers of both SNPs had a better OS compared with that of major allele carriers. The association of CTLA-4 rs231775 and CD274 rs7866740 with PFS did not reach the significance threshold in the multivariate analysis (shown in Table 2).

Table 2.

Results of the multivariable Cox regression model for the association of genetic variants with PFS and OS in cc-mRCC patients treated with sunitinib as first TKIa

PFSOS
ProteinGeners numberHR (95% CI)bP valueHR (95% CI)bP value
Discovery cohort (n = 550) 
 PD-L1 CD274 rs7866740 0.77 (0.56–1.05) 0.093 0.73 (0.54–0.99) 0.047 
 CTLA-4 CTLA-4 rs231775 0.86 (0.74–1.01) 0.059 0.84 (0.72–0.98) 0.028 
Validation cohort (n = 138) 
 PD-L1 CD274 rs7866740 1.03 (0.64–1.65) 0.911 1.35 (0.85–2.14) 0.197 
 CTLA-4 CTLA-4 rs231775 1.02 (0.78–1.35) 0.867 0.74 (0.55–1.01) 0.057 
Pooled cohort (n = 688) 
 PD-L1 CD274 rs7866740 0.83 (0.65–1.07) 0.160 0.89 (0.69–1.14) 0.358 
 CTLA-4 CTLA-4 rs231775 0.88 (0.77–1.01) 0.073 0.83 (0.72–0.95) 0.008 
PFSOS
ProteinGeners numberHR (95% CI)bP valueHR (95% CI)bP value
Discovery cohort (n = 550) 
 PD-L1 CD274 rs7866740 0.77 (0.56–1.05) 0.093 0.73 (0.54–0.99) 0.047 
 CTLA-4 CTLA-4 rs231775 0.86 (0.74–1.01) 0.059 0.84 (0.72–0.98) 0.028 
Validation cohort (n = 138) 
 PD-L1 CD274 rs7866740 1.03 (0.64–1.65) 0.911 1.35 (0.85–2.14) 0.197 
 CTLA-4 CTLA-4 rs231775 1.02 (0.78–1.35) 0.867 0.74 (0.55–1.01) 0.057 
Pooled cohort (n = 688) 
 PD-L1 CD274 rs7866740 0.83 (0.65–1.07) 0.160 0.89 (0.69–1.14) 0.358 
 CTLA-4 CTLA-4 rs231775 0.88 (0.77–1.01) 0.073 0.83 (0.72–0.95) 0.008 

aMultivariable analysis was adjusted by age, gender, and Heng prognostic risk group.

bMajor allele was the reference.

Validation study on the associations of polymorphisms with PFS and OS

In the validation cohort, the genotype call rate for CTLA-4 rs231775 and CD274 rs7866740 was 99.3% and 98.6%, respectively. Both SNPs were in Hardy–Weinberg equilibrium (P > 0.05). The MAF of both SNPs was similar in both the discovery cohort and the National Center for Biotechnology Information (NCBI) dbSNP database (CEU population, which represents Utah residents with Northern and Western European ancestry). Only 4 patients with the CD274 rs7866740 GG genotype were detected. As a consequence, a dominant model was used for the genetic association analysis.

In the validation study, none of the SNPs was significantly associated with PFS or OS either in the univariate analysis or after correction for age, gender, and Heng prognostic risk group. Nevertheless, CTLA-4 rs231775 showed the same direction of effect and comparable effect size for OS (HR, 0.74; 95% CI, 0.55–1.01; P = 0.057; Table 2) than that calculated in the discovery cohort (HR, 0.84; 95% CI, 0.72–0.98; P = 0.028).

Pooled analysis of the genetic associations with PFS and OS

In the pooled cohort, CTLA-4 rs231775 showed a significant association with OS (HR, 0.83; 95% CI, 0.72–0.95; P = 0.008). Patients with the GG genotype had longer OS (35.1 months) compared with the patients with an AG or AA genotype (30.3 and 24.3 months, respectively). The Kaplan–Meier plot for OS is presented in Supplementary Fig. S1. No significant associations were found for PFS (shown in Table 2).

Functional effect

We utilized data from GTEx and OncoLnc to provide possible explanations for our interesting finding that CTLA-4 rs231775 was associated with OS. In the vast majority of tissues in GTEx (Release V6p), GG genotype of CTLA-4 rs231775 was associated with a decreased mRNA expression of the CTLA-4 gene compared with the AA and AG genotypes (Supplementary Fig. S2A). Due to the strict significance threshold, the above association between CTLA-4 rs231775 and mRNA expression was only observed in testis tissue (P = 1.0 × 10−7; Supplementary Fig. S2B). By OncoLnc, OS was compared between cc-RCC patients with lower and higher CTLA-4 mRNA expression. It was revealed that patients with lower CTLA-4 mRNA expression (lower 50 percentile) had a longer OS (P = 0.00255, Supplementary Fig. S3).

In the present study, we explored genetic variants in the checkpoint-related genes CD274 (PD-L1), PDCD1 (PD-1), and CTLA-4 for a possible association with PFS and OS in cc-mRCC patients that received sunitinib as first TKI. The most important finding of our study is the identification of CTLA-4 rs231775 as a potential prognostic biomarker for OS. Patients with the GG genotype showed an increased OS compared with those with the GA or AA genotype. To our knowledge, this is the first time that an association of the CTLA-4 rs231775 genetic polymorphism with OS in this specific patient population is reported.

CTLA-4 rs231775 is located at position +49 in exon 1 of the CTLA-4 gene and is a common nonsynonymous CTLA-4 polymorphism. The +49 G allele encodes a Thr to Ala substitution at codon 17 in the signal peptide of the CTLA-4 protein (25). Anjos and colleagues have found in a cell-free model of in vitro reconstitution of translation and endoplasmic reticulum processing that the G-allele of CTLA-4 rs231775 was associated with inefficient glycosylation, which leads to a decrease of CTLA-4 expression in cell surface (26). In addition, Sun and colleagues have demonstrated that CTLA-4-Ala (coded by the G-allele) had a lower capability to bind the CTLA-4 ligand and a weaker inhibitory effect on T-cell activation compared with the functional effects of CTLA-4-Thr (coded by the A-allele; ref. 27). In a large Chinese population, they also found that subjects carrying the AA genotype were more susceptible for developing cancer than those with the GG genotype (27). Either by the decrease of protein expression or through lower binding capability, both studies reach to the conclusion that the G-allele is associated with reduced inhibition of activated T cells (27). In addition, Ligers and colleagues have reported in patients with myasthenia gravis and multiple sclerosis that G-allele carriers showed a significant decrease of CTLA-4 mRNA and protein expression (28). Therefore, it seems likely that the G-allele of CTLA-4 rs231775 is associated with a decrease of CTLA-4 expression.

We sought evidence that the decreased CTLA-4 expression contributes to an improved OS. CTLA-4 is thought to play a negative regulatory role after binding with its ligands. Hence, it could be hypothesized that low CTLA-4 expression leads to low ligand binding, because of which T cells can only be weakly inhibited facilitating an enhanced autoimmune response of possible benefit for the patient with cancer. We, therefore, investigated the relationship of CTLA-4 mRNA expression levels with OS by using the TCGA-KIRC dataset (23, 24), which revealed that cc-RCC patients (regardless of tumor stage) with lower CTLA-4 mRNA expression had longer OS. Combining all findings, the G-allele of CTLA-4 rs231775 is indeed associated with lower CTLA-4 mRNA expression, and lower mRNA expression links to longer OS, which is consistent with our results.

Our finding provides insight into RCC prognosis and corroborates the current strategies of new drug development for RCC. In this respect, ipilimumab, which is designed to inhibit the CTLA-4 protein, has demonstrated clinical efficacy and manageable toxicity as monotherapy or combined with nivolumab in mRCC (29). To date, there are no studies investigating the role of CTLA-4 polymorphisms or CTLA-4 protein expression on ipilimumab outcome in mRCC patients. However, in patients with melanoma, Breunis and colleagues have assessed seven CTLA-4 polymorphisms with ipilimumab response. In responding patients, there were proportionally more rs231775 A-allele carriers than G-allele carriers, whereas there was no difference in nonresponding patients (OR, 0.39; 95% CI, 0.18–0.82; P = 0.009; ref. 30). If confirmed in mRCC patients on the basis of the results presented here, rs231775 might be important for the optimization of ipilimumab treatment of mRCC.

In the present study in cc-mRCC patients that received sunitinib as first TKI, we were able to demonstrate an association of CTLA-4 rs231775 with OS, but not with PFS. Interestingly, Song and colleagues have also identified CTLA-4 rs231775 is a prognostic biomarker in patients with advanced non–small cell lung cancer (31). Patients with a GG or GA genotype experienced a significantly longer OS than those with an AA genotype after correction for many covariates among which was treatment (31). These findings strongly suggest that the CTLA-4 genetic variant is more of influence on the biological behavior of the disease than with the effects of treatment, implying the prognostic role of this polymorphism. Introduction of checkpoint inhibitors in the treatment of cancer patients may, however, change the relevance of CTLA-4 rs231775 for PFS.

Polymorphisms in PDCD1 (PD-1) and CD274 (PD-L1) are thought to be promising genetic candidates to explain differences in immunosuppressive function. It has been reported that PDCD1 rs10204525 was associated with OS in 439 patients with locoregional gastric cancer (32) as well as in 668 patients with resectable colorectal cancer (33), suggesting that it may serve as a prognostic factor. In our patient population of metastatic disease, however, we did not observe any significant association of PDCD1 rs10204525 (which was captured by PDCD1 rs41386349) with survival outcome. CD274 rs4143815 has been reported to be significantly associated with worse OS in a total of 354 patients with non–small cell lung cancer who underwent curative resection (34). We observed an association of CD274 rs7866740 with OS in the discovery cohort, but failed confirmation in the final analysis. Owing to the fact that genotype of CD274 rs7866740 was imputed and the MAF was relatively small, the significant association with CD274 rs7866740 in the discovery cohort might be detected by chance. Future studies should focus on the relevance of these SNPs in patients with no/microscopic disease after surgery versus those with advanced disease.

Whereas previous studies have focused on SNPs in genes of the VEGF signaling pathway, we are the first to explore the potential association of the genetic variability in three genes encoding immune checkpoints with the outcome in sunitinib-treated patients. Although we did not formally confirm the association in the validation cohort, the meta-analysis showed similar results to those of the discovery cohort and with stronger statistical evidence. In addition, a biological and mechanistic rationale was provided using expression and survival data from GTEx (22) and TCGA (23). However, we should interpret results from GTEx and TCGA carefully. It is because the significant association of genotype with CTLA-4 expression is found in testis not in kidney tissue (only 39 samples available in GTEx) or T cells in microenvironment. Moreover, the TCGA-KIRC datasets include patients with different stages, and follow-up period is not long enough. As a result, the plot in Supplementary Fig. S3 might be changed when tumor stage is taken into account and after enough follow-up period. The possible reasons of the failed validation could be the relatively small sample size as well as differences in patient characteristics between the two cohorts. Due to the need for imputation, adjustment of PFS and OS might slightly be distorted. To reduce this chance, a multiple imputation procedure instead of a single imputation was used. Moreover, imputation was implemented in 16% and 10% patients in the discovery and validation cohort, respectively, which could be considered negligible. The significance threshold in the discovery study was not adjusted by the strict Bonferroni correction in this exploratory study, because Bonferroni correction will increase the type II error (the chances of false-negative results; ref. 35). In our opinion, an inflated type I error due to multiple comparisons is better solved by a validation study than by a Bonferroni correction.

In conclusion, data from the present study show that the G-allele of CTLA-4 rs231775 is associated with an improved OS in cc-mRCC patients receiving sunitinib treatment, suggesting this polymorphism may serve as a prognostic marker.

K. Junker reports receiving commercial research grants and speakers bureau honoraria from, and is a consultant/advisory board member for Novartis. T. Eisen is an employee of AstraZeneca, reports receiving commercial research grants from AstraZeneca and Pfizer, holds ownership interest (including patents) in AstraZeneca, and is a consultant/advisory board member for EUSA. B.I. Rini reports receiving commercial research grants from and is a consultant/advisory board member for Pfizer. No potential conflicts of interest were disclosed by the other authors.

Conception and design: X. Liu, J.J. Swen, H. Gelderblom, E. Oosterwijk, T. Eisen, L.A.L.M. Kiemeney, B.I. Rini, H.-J. Guchelaar

Development of methodology: X. Liu, J.J. Swen, M.H.M. Diekstra, A. Sverrisdottir, H.-J. Guchelaar

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J.J. Swen, M.H.M. Diekstra, E. Boven, D. Castellano, H. Gelderblom, R.H.J. Mathijssen, E. Oosterwijk, K. Junker, M. Roessler, K. Alexiusdottir, A. Sverrisdottir, M.T. Radu, V. Ambert, T. Eisen, A. Warren, C. Rodríguez-Antona, J. García-Donas, L.A.L.M. Kiemeney

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): X. Liu, J.J. Swen, E. Boven, D. Castellano, E. Oosterwijk, A. Sverrisdottir, C. Rodríguez-Antona, S. Böhringer, K.K.M. Koudijs, H.-J. Guchelaar

Writing, review, and/or revision of the manuscript: X. Liu, J.J. Swen, M.H.M. Diekstra, E. Boven, H. Gelderblom, R.H.J. Mathijssen, S.H. Vermeulen, E. Oosterwijk, A. Sverrisdottir, M.T. Radu, T. Eisen, C. Rodríguez-Antona, J. García-Donas, S. Böhringer, L.A.L.M. Kiemeney, B.I. Rini, H.-J. Guchelaar

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): X. Liu, M.H.M. Diekstra, S.H. Vermeulen, E. Oosterwijk, L.A.L.M. Kiemeney

Study supervision: J.J. Swen, D. Castellano, T. Eisen, L.A.L.M. Kiemeney, H.-J. Guchelaar

Other (review of histology): A. Warren

The authors thank Renée Baak-Pablo and Danielle Klootwijk for their assistance with genotyping.

The research leading to these results has received funding from the European Union's Seventh Framework Programme (FP7/2007-2013) under grant agreement no. 259939. X. Liu is supported by the Chinese Scholarship Council (No. 201406220186).

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.
Prasad
SR
,
Humphrey
PA
,
Catena
JR
,
Narra
VR
,
Srigley
JR
,
Cortez
AD
, et al
Common and uncommon histologic subtypes of renal cell carcinoma: imaging spectrum with pathologic correlation
.
Radiographics
2006
;
26
:
1795
806
.
2.
Znaor
A
,
Lortet-Tieulent
J
,
Laversanne
M
,
Jemal
A
,
Bray
F
. 
International variations and trends in renal cell carcinoma incidence and mortality
.
Eur Urol
2015
;
67
:
519
30
.
3.
Ghatalia
P
,
Zibelman
M
,
Geynisman
DM
,
Plimack
ER
. 
Checkpoint inhibitors for the treatment of renal cell carcinoma
.
Curr Treat Options Oncol
2017
;
18
:
7
.
4.
Kusmartsev
S
,
Eruslanov
E
,
Kubler
H
,
Tseng
T
,
Sakai
Y
,
Su
Z
, et al
Oxidative stress regulates expression of VEGFR1 in myeloid cells: link to tumor-induced immune suppression in renal cell carcinoma
.
J Immunol
2008
;
181
:
346
53
.
5.
Manning
EA
,
Ullman
JG
,
Leatherman
JM
,
Asquith
JM
,
Hansen
TR
,
Armstrong
TD
, et al
A vascular endothelial growth factor receptor-2 inhibitor enhances antitumor immunity through an immune-based mechanism
.
Clin Cancer Res
2007
;
13
:
3951
9
.
6.
Amin
A
,
Plimack
ER
,
Infante
JR
,
Ernstoff
M
,
Rini
BI
,
McDermott
DF
, et al
1052pdnivolumab (N) (Anti-Pd-1; Bms-936558, Ono-4538) in combination with sunitinib (S) or pazopanib (P) in patients (Pts) with metastatic renal cell carcinoma (Mrcc)
.
Ann Oncol
2014
;
25
(
suppl_4
):
iv362
iv3
.
7.
He
J
,
Hu
Y
,
Hu
M
,
Li
B
. 
Development of PD-1/PD-L1 pathway in tumor immune microenvironment and treatment for non-small cell lung cancer
.
Sci Rep
2015
;
5
:
13110
.
8.
Antczak
A
,
Pastuszak-Lewandoska
D
,
Gorski
P
,
Domanska
D
,
Migdalska-Sek
M
,
Czarnecka
K
, et al
Ctla-4 expression and polymorphisms in lung tissue of patients with diagnosed non-small-cell lung cancer
.
BioMed Res Int
2013
;
2013
:
576486
.
9.
Iacovelli
R
,
Nole
F
,
Verri
E
,
Renne
G
,
Paglino
C
,
Santoni
M
, et al
Prognostic role of PD-L1 expression in renal cell carcinoma. A systematic review and meta-analysis
.
Target Oncol
2016
;
11
:
143
8
.
10.
Thompson
RH
,
Kuntz
SM
,
Leibovich
BC
,
Dong
H
,
Lohse
CM
,
Webster
WS
, et al
Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up
.
Cancer Res
2006
;
66
:
3381
5
.
11.
Mao
H
,
Zhang
L
,
Yang
Y
,
Zuo
W
,
Bi
Y
,
Gao
W
, et al
New insights of CTLA-4 into its biological function in breast cancer
.
Curr Cancer Drug Targets
2010
;
10
:
728
36
.
12.
Frigola
X
,
Inman
BA
,
Lohse
CM
,
Krco
CJ
,
Cheville
JC
,
Thompson
RH
, et al
Identification of a soluble form of B7-H1 that retains immunosuppressive activity and is associated with aggressive renal cell carcinoma
.
Clin Cancer Res
2011
;
17
:
1915
23
.
13.
Leite
KR
,
Reis
ST
,
Junior
JP
,
Zerati
M
,
Gomes Dde
O
,
Camara-Lopes
LH
, et al
PD-L1 expression in renal cell carcinoma clear cell type is related to unfavorable prognosis
.
Diagn Pathol
2015
;
10
:
189
.
14.
Choueiri
TK
,
Figueroa
DJ
,
Fay
AP
,
Signoretti
S
,
Liu
Y
,
Gagnon
R
, et al
Correlation of PD-L1 tumor expression and treatment outcomes in patients with renal cell carcinoma receiving sunitinib or pazopanib: results from COMPARZ, a randomized controlled trial
.
Clin Cancer Res
2015
;
21
:
1071
7
.
15.
Fukuda
T
,
Kamai
T
,
Masuda
A
,
Nukui
A
,
Abe
H
,
Arai
K
, et al
Higher preoperative serum levels of PD-L1 and B7-H4 are associated with invasive and metastatic potential and predictable for poor response to VEGF-targeted therapy and unfavorable prognosis of renal cell carcinoma
.
Cancer Med
2016
;
5
:
1810
20
.
16.
Hu
P
,
Liu
Q
,
Deng
G
,
Zhang
J
,
Liang
N
,
Xie
J
, et al
The prognostic value of cytotoxic T-lymphocyte antigen 4 in cancers: a systematic review and meta-analysis
.
Sci Rep
2017
;
7
:
42913
.
17.
van der Zanden
LF
,
Vermeulen
SH
,
Oskarsdottir
A
,
Maurits
JS
,
Diekstra
MH
,
Ambert
V
, et al
Description of the EuroTARGET cohort: A European collaborative project on TArgeted therapy in renal cell cancer-GEnetic- and tumor-related biomarkers for response and toxicity
.
Urol Oncol
2017
;
35
:
529.e9
529.e16
.
18.
de Bakker
PI
,
Yelensky
R
,
Pe'er
I
,
Gabriel
SB
,
Daly
MJ
,
Altshuler
D
. 
Efficiency and power in genetic association studies
.
Nat Genet
2005
;
37
:
1217
23
.
19.
Liu
X
,
Swen
JJ
,
Boven
E
,
Castellano
D
,
Gelderblom
H
,
Mathijssen
RH
, et al
Meta-analysis on the association of VEGFR1 genetic variants with sunitinib outcome in metastatic renal cell carcinoma patients
.
Oncotarget
2017
;
8
:
1204
12
.
20.
Heng
DY
,
Xie
W
,
Regan
MM
,
Warren
MA
,
Golshayan
AR
,
Sahi
C
, et al
Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study
.
J Clin Oncol
2009
;
27
:
5794
9
.
21.
van Buuren
S
,
Groothuis-Oudshoorn
K
. 
mice: Multivariate Imputation by Chained Equations in R
.
J Stat Software
2011
;
45
:
67
.
22.
GTEx Consortium
The genotype-tissue expression (GTEx) project
.
Nat Genet
2013
;
45
:
580
5
.
23.
Anaya
J
. 
OncoLnc: linking TCGA survival data to mRNAs, miRNAs, and lncRNAs
.
PeerJ Comput Sci
2016
;
2
:
e67
.
24.
Cancer Genome Atlas Research Network
. 
Comprehensive molecular characterization of clear cell renal cell carcinoma
.
Nature
2013
;
499
:
43
9
.
25.
Nistico
L
,
Buzzetti
R
,
Pritchard
LE
,
Van der Auwera
B
,
Giovannini
C
,
Bosi
E
, et al
The CTLA-4 gene region of chromosome 2q33 is linked to, and associated with, type 1 diabetes. Belgian Diabetes Registry
.
Hum Mol Genet
1996
;
5
:
1075
80
.
26.
Anjos
S
,
Nguyen
A
,
Ounissi-Benkalha
H
,
Tessier
MC
,
Polychronakos
C
. 
A common autoimmunity predisposing signal peptide variant of the cytotoxic T-lymphocyte antigen 4 results in inefficient glycosylation of the susceptibility allele
.
J Biol Chem
2002
;
277
:
46478
86
.
27.
Sun
T
,
Zhou
Y
,
Yang
M
,
Hu
Z
,
Tan
W
,
Han
X
, et al
Functional genetic variations in cytotoxic T-lymphocyte antigen 4 and susceptibility to multiple types of cancer
.
Cancer Res
2008
;
68
:
7025
34
.
28.
Ligers
A
,
Teleshova
N
,
Masterman
T
,
Huang
WX
,
Hillert
J
. 
CTLA-4 gene expression is influenced by promoter and exon 1 polymorphisms
.
Genes Immun
2001
;
2
:
145
52
.
29.
Hammers
HJ
,
Plimack
ER
,
Infante
JR
,
Rini
BI
,
McDermott
DF
,
Lewis
LD
, et al
Safety and efficacy of nivolumab in combination with ipilimumab in metastatic renal cell carcinoma: The CheckMate 016 Study
.
J Clin Oncol
2017
;
35
:
3851
8
.
30.
Breunis
WB
,
Tarazona-Santos
E
,
Chen
R
,
Kiley
M
,
Rosenberg
SA
,
Chanock
SJ
. 
Influence of cytotoxic T lymphocyte-associated antigen 4 (CTLA4) common polymorphisms on outcome in treatment of melanoma patients with CTLA-4 blockade
.
J Immunother
2008
;
31
:
586
90
.
31.
Song
B
,
Liu
Y
,
Liu
J
,
Song
X
,
Wang
Z
,
Wang
M
, et al
CTLA-4 +49A>G polymorphism is associated with advanced non-small cell lung cancer prognosis
.
Respiration
2011
;
82
:
439
44
.
32.
Sunakawa
Y
,
Cao
S
,
Volz
NB
,
Berger
MD
,
Yang
D
,
Parekh
A
, et al
Genetic variations in immunomodulatory pathways to predict survival in patients with locoregional gastric cancer
.
Pharmacogenomics J
2017
;
17
:
528
34
.
33.
Yoon
S
,
Kang
BW
,
Park
SY
,
Kim
HJ
,
Park
JS
,
Choi
GS
, et al
Prognostic relevance of genetic variants involved in immune checkpoints in patients with colorectal cancer
.
J Cancer Res Clin Oncol
2016
;
142
:
1775
80
.
34.
Lee
SY
,
Jung
DK
,
Choi
JE
,
Jin
CC
,
Hong
MJ
,
Do
SK
, et al
Functional polymorphisms in PD-L1 gene are associated with the prognosis of patients with early stage non-small cell lung cancer
.
Gene
2017
;
599
:
28
35
.
35.
Rothman
KJ
. 
No adjustments are needed for multiple comparisons
.
Epidemiology
1990
;
1
:
43
6
.

Supplementary data