Purpose: Clear cell renal cell carcinoma (ccRCC) has shown durable responses to checkpoint blockade therapies. However, important gaps persist in the understanding of its immune microenvironment. This study aims to investigate the expression and prognostic significance of immune checkpoints in primary and metastatic ccRCC, in relation with mature dendritic cells (DC) and T-cell densities.

Experimental Design: We investigated the infiltration and the localization of CD8+ T cells and mature DC, and the expression of immune checkpoints (PD-1, LAG-3, PD-L1, and PD-L2) in relation with prognosis, in 135 primary ccRCC tumors and 51 ccRCC lung metastases. RNA expression data for 496 primary ccRCC samples were used as confirmatory cohort.

Results: We identify two groups of tumors with extensive CD8+ T-cell infiltrates. One group, characterized by high expression of immune checkpoints in the absence of fully functional mature DC, is associated with increased risk of disease progression. The second group, characterized by low expression of immune checkpoints and localization of mature DC in peritumoral immune aggregates (tertiary lymphoid structures), is associated with good prognosis.

Conclusions: The expression of the immune checkpoints and the localization of DC in the tumor microenvironment modulate the clinical impact of CD8+ T cells in ccRCC. Clin Cancer Res; 21(13); 3031–40. ©2015 AACR.

Translational Relevance

Clear cell renal cell carcinoma (ccRCC) is an enigma of the tumor microenvironment studies because, in contrast with most malignancies, high densities of CD8+ T cells correlate with poor clinical outcome. We characterize the microenvironment of ccRCC primary tumors and lung metastases associated with an extensive CD8+ T-cell infiltrate. In one group of patients, a T-cell exhausted microenvironment characterized by high expression of immune checkpoints, and the absence of fully functional mature dendritic cells (DC) was found and correlated with poor prognosis. In the second group, low expression of immune checkpoints and DC localized in peritumoral immune aggregates were found and conversely associated with good prognosis. Our data provide novel prognostic biomarkers highlighting the central role of PD-L2 and LAG-3 in the immunomodulation of ccRCC. The combination of these immune profiles could guide the selection of patients who are likely to respond to checkpoint blockade therapies.

A solid tumor is an intricate and dynamic ecosystem containing tumor and immune cells, fibroblasts, blood, and lymphatic vessels (1). The density and composition of the immune microenvironment is heterogeneous among patients and tumor types, and it is becoming a robust tool to predict postsurgical recurrence and death (1, 2). In the vast majority of cancers, tumor infiltration by CD8+ memory cytotoxic T cells and Th1 cells is associated with good clinical outcome (1). In addition, it has been reported that an organized local immune reaction, characterized by the presence of mature dendritic cells (DC) localized in tumor-associated tertiary lymphoid structures (TLS), is necessary to orchestrate this cytotoxic and Th1 immune contexture and is associated with good clinical outcome (3–8) and response to therapeutic vaccines (9, 10).

However, several recent findings challenge this concept of the unequivocal relation of effector memory CD8+ T-cell infiltration with a good clinical outcome in cancer. First, in diffuse large B-cell lymphoma (11), Hodgkin lymphoma (12), and clear cell renal cell carcinoma (ccRCC; ref. 13) high densities of tumor-infiltrating CD8+ T cells have been associated with poor prognosis in some studies. Second, a recent publication from our group showed that the infiltration of lung metastases from ccRCC by CD8+ T cells was correlated with poor overall survival (OS) whereas it was a factor of good prognosis in lung metastases from colorectal carcinoma (14). Finally, in non–small cell lung cancer (NSCLC) in which CD8+ T cells densities correlate with good prognosis, a low density of TLS-DC associated with high CD8+ T-cell infiltration identifies a group of patients with high risk of death, suggesting a functional impairment of intratumoral CD8+ T cells in this situation (4).

Immune checkpoints on infiltrating T cells are key regulators of immune escape in cancer. In primary ccRCC, several studies have shown that the expression of the inhibitory receptor PD-1 on the immune cells (15, 16) or its ligand PD-L1 on tumor cells (17–19) is associated with a poor clinical outcome. Interestingly, antibodies that block the PD-1 axis have yielded a 20% to 30% response rate in metastatic ccRCC (20, 21) that seems to be related to the expression of PD-L1 by the tumor cells (20, 22). Another inhibitory molecule that has gained recent attention is the lymphocyte activation gene-3 (LAG-3), which is coexpressed with PD-1 on CD8+ tumor-infiltrating lymphocytes in melanoma (23), and together with PD-1 synergistically regulates T-cell function (24). We expected immune checkpoints high ccRCC patients to have a worse prognosis than the immune checkpoint low.

In these newly described scenarios in which CD8+ T-cell infiltration correlates with poor prognosis, it is important to define the combination of immune-based biomarkers that will predict patients' prognosis and further guide immunotherapeutic approaches. This study aims to investigate the expression and prognostic significance of immune checkpoint receptors and paired ligands on primary and metastatic ccRCC in relation with TLS-DC and T-cell densities.

Patients

A cohort of 135 primary ccRCC human tumors and another of 51 ccRCC lung metastases were collected. The primary cases derived from specimens of radical nephrectomy operated between 1999 and 2003 at the hospital Necker-Enfants Malades (Paris, France). The ccRCC lung metastases cohort resected at the Hotel Dieu hospital (Paris, France) or Hôpital Européen George Pompidou (HEGP, Paris, France) between 1992 and 2010 was already described in ref. (14). This research was conducted according to the recommendations outlined in the Helsinki declaration and approved by the medical ethics boards of all participating institutions, and with the agreement of the Ile-de-France II ethics committee (no. 2012-0612). The demographic characteristics of the cohorts are depicted in Supplementary Table S1 (14).

In addition, expression data for 496 primary ccRCC samples with complete follow-up were downloaded from The Cancer Genome Atlas' (TCGA) KIRC study, using version 2 of the normalized RNA sequencing data. Corresponding clinical data (updated on 2013-04-06) were downloaded from (25).

Clinic and pathologic features

The original histologic diagnosis was confirmed on archival hematoxylin and eosin–stained slides, and histopathologic features such as histologic subtype (26), tumor size, regional lymph node invasion, distant metastases at surgery, Fuhrman nuclear grade (27), and sarcomatoïd features were collected. All tumors were pathologically staged according to the TNM (tumor–node–metastasis) classification (28). The duration of follow-up was calculated from the date of the surgery (nephrectomy or metastasectomy) to the date of cancer progression, last follow-up or death.

Immunohistochemical and immunofluorescence staining

Serial 5-μm formalin-fixed paraffin-embedded (FFPE) tissue sections from primary and metastatic ccRCC were stained using autostainerPlus Link 48 (Dako). Antigen retrieval and deparaffinization were carried out on a PT-Link (Dako) using the EnVision FLEX Target Retrieval Solutions (Dako). The antibodies used in this study for IHC and immunofluorescence (IF) are listed in Supplementary Table S2. IF-stained slides were scanned after secondary antibody incubation and mounting. For the IHC staining, peroxidase activity was detected using 3-amino-9-ethylcarbazole substrate or Novared and alkaline phosphatase using alkaline phosphatase substrate III (Vector Laboratories).

Tests of the specificity and sensitivity of PD-1, PD-L1, PD-L2 and LAG-3 monoclonal antibodies for IHC experiments were performed using generated FFPE cell pellets from transfected 300.19 cells (for PD-1, PD-L1, and PD-L2; ref. 29) and CHO cells (for hLAG-3), whereas parental untransfected cells served as negative controls (Supplementary Fig. S1; Costim Pharmaceuticals). Multiple organs (n = 32), human TMA (FDA999a, US Biomax), and malignant cancer tissues (n = 10) from different oncology indications were also tested using the above mentioned protocols. Normal human FFPE tonsil sections for PD-1, LAG-3, PD-L2, and normal placenta for PD-L1 were used as positive controls (Supplementary Fig. S1).

Immunohistochemical quantification

Stained slides were scanned with a Nanozoomer (Hamamatsu) and analyzed with Calopix software (Tribvn). For quantification purposes, tissues were divided into Invasive Margin (IM) and Tumor Center (TC) as previously described (30), and the density of positive cells was calculated in the whole tumor region (IM and TC). Because of the small size (or absence) of TC region in the majority of the metastases, the analysis on this cohort was done not discriminating between the two regions. The percentages of tumor cells stained positive for PD-L1 and PD-L2 were quantified by two independent reviewers (A. Lupo and N.A. Giraldo) without prior knowledge of patient outcome, and the tumors above 5% tumor cell expression were considered as positive in accordance with studies in other types of cancer (20, 31).

Statistical analysis

Comparisons among the demographic and pathologic features, immune marker densities, and PD-L1 and PD-L2 expressions were evaluated by using χ2, Fisher exact, and Wilcoxon rank-sum tests. Association of variables to prognosis was assessed using the Kaplan–Meier method, univariate and multivariate Cox regression analyses. To segregate patients in two groups based on numerical variables (cell density or gene expression), Log-rank P values of each possible cutoff were computed. The cutoff that minimized the P value of a log-rank test for DFS was retained, and the corresponding P value was corrected using the method published by Altman and colleagues (32). These cutoffs were later used to segregate patients into two groups, and their associated Kaplan–Meier curves are displayed throughout the figures. To further confirm and validate the prognosis association of the cell densities, we determined the median and third quartile cutoff and calculated the corresponding univariate Cox-regression P values (Supplementary Table S3). Only those variables univariately associated with prognosis were included in the multivariate Cox regression analysis. The duration of follow-up was calculated from the date of nephrectomy or metastasectomy to the date of death or last follow-up.

Tumor infiltration by CD8+ T lymphocytes and expression of Th1-associated genes correlate with poor prognosis in ccRCC

The density of CD8+ T cells in the IM of the primary tumors was heterogeneous in the cohort of 135 primary ccRCC. On the basis of the Optimal P value cutoff for DFS (OPv; 630 cells/mm2) we found that the CD8High (n = 41/135, 30%) group had a shorter disease-free survival (DFS, P = 0.0001, Fig. 1A) and OS (P = 0.001, Fig. 1A). The density of CD8+ T cells in the TC had no prognostic impact (Supplementary Table S4). The density of CD8+ T cells in the IM correlated with the Fuhrman grade (Supplementary Fig. S2), but not with any other pathologic variables, including TNM (data not shown).

Figure 1.

CD8+ T cells and CD8/Th1 gene signature are associated with poor prognosis in ccRCC. OS and DFS according to the presence of a high or low density of CD8+ T cells in the IM of primary ccRCC (A) and ccRCC lung metastases (B). The P values by univariate Cox regression analysis for OS on seven Th1-related genes are displayed, P = 0.05 dotted black line, HR >1.0 gray columns (C). OS according to IFNG gene expression in primary ccRCC (D).

Figure 1.

CD8+ T cells and CD8/Th1 gene signature are associated with poor prognosis in ccRCC. OS and DFS according to the presence of a high or low density of CD8+ T cells in the IM of primary ccRCC (A) and ccRCC lung metastases (B). The P values by univariate Cox regression analysis for OS on seven Th1-related genes are displayed, P = 0.05 dotted black line, HR >1.0 gray columns (C). OS according to IFNG gene expression in primary ccRCC (D).

Close modal

An independent cohort of lung metastases of ccRCC, in which the negative impact of high densities of CD8+T cells on OS had been previously described using semiquantitative counting techniques (14), was reanalyzed using a quantitative approach on 51 cases. The OPv for CD8+ T cells density was 490 cells/mm2, and the CD8High (n = 14/51, 27%) group displayed a shorter OS (P = 0.001, Fig. 1B). This result confirms our previous observations (14).

From TCGA public database of 496 primary ccRCC, we analyzed the expression of 844 immune-related genes, from which we extracted data concerning seven genes expressed in a Th1- and CD8+ T-cell–oriented response according to Galon and colleagues (30). We found that the expression of most of the genes associated with this cell signature correlated with poor prognosis: CD8A P = 0.04, TBX21 P = 0.03, IRF1 P = 0.01, GZMB P = 4.4 × 10−5, and IFNG P = 3.17 × 10−7 that displayed the lowest P value (Fig. 1C). On the basis of the OPv for IFNG, patients that had high quantities of intratumoral mRNA for this gene displayed a shorter OS (P = 0.006, Fig. 1D).

Simultaneous expression of immune checkpoints in primary and metastatic ccRCC identifies patients with poor clinical outcome

Primary tumors.

To investigate the impact of immune checkpoint molecules on the negative prognostic impact of the CD8High group, we analyzed the protein expression of PD-1, LAG-3, PD-L1, and PD-L2 molecules in this group of tumors (we could analyze 40/41 tumors) and a randomly matched group of the same size (n = 40/94) coming from the CD8Low.

On the basis of the OPv cutoff, 15 tumors of 80 were considered as PD-1High in the IM and they displayed a shorter DFS (P = 0.0005, Fig. 2A) and OS (P = 0.03, Fig. 2A). The density of PD-1+ cells in the TC was not significantly associated with prognosis (Supplementary Table S4). The Fuhrman grade was associated with the PD-1+ cell density in the IM (Supplementary Fig. S2).

Figure 2.

Expression of immune checkpoints correlates with unfavorable clinical outcome for patients with primary ccRCC. OS and DFS according to the presence of a high or low density of PD-1+ and LAG-3+ cells, and the expression of PD-L1 or PD-L2 by >5% of the tumor cells in primary ccRCC (A). OS and DFS (B) and pie chart (C) representing the percentage of patients that had progressed after 5 years of surgery according to the expression of PD-L1 and/or PD-L2 on tumor cells related with high densities of PD-1+ lymphocytes. The P values by univariate Cox regression analysis for OS on four genes are displayed, P = 0.05 dotted gray line, HR >1.0 gray columns (D).

Figure 2.

Expression of immune checkpoints correlates with unfavorable clinical outcome for patients with primary ccRCC. OS and DFS according to the presence of a high or low density of PD-1+ and LAG-3+ cells, and the expression of PD-L1 or PD-L2 by >5% of the tumor cells in primary ccRCC (A). OS and DFS (B) and pie chart (C) representing the percentage of patients that had progressed after 5 years of surgery according to the expression of PD-L1 and/or PD-L2 on tumor cells related with high densities of PD-1+ lymphocytes. The P values by univariate Cox regression analysis for OS on four genes are displayed, P = 0.05 dotted gray line, HR >1.0 gray columns (D).

Close modal

Of the 80 patients, 9 were considered as LAG-3High (subdivided by the OPv cutoff) in the IM and they displayed a shorter DFS (P = 0.02, Fig. 2A), and did not reach significance for the OS (P = 0.07, Fig. 2A). The density of LAG-3+ cells in the TC was not significantly associated with patients DFS or OS (Supplementary Table S4). Representative pictures of the IHC staining of highly and poorly PD-1– and LAG-3–infiltrated lesions are shown in Supplementary Fig. S3A and S3B, respectively.

On the basis of the 5% cutoff generally used in clinical trials with anti-checkpoint antibodies (20), 22 of 80 (27%) and 27 of 80 (34%) patients were PD-L1+ and PD-L2+, respectively (Supplementary Fig. S3C and S3D); among them, 9 tumors were double positive. Patients with PD-L1+ tumors had a shorter OS than those with less than 5% positive tumor cells (P = 0.02, Fig. 2A); a trend toward shorter DFS was also found (P = 0.06, Fig. 2A). Univariately, patients with PD-L1+ tumors were 2.9 times more likely to die in the 5 years post-nephrectomy than patients with less than 5% PD-L1+ tumor cells [OS risk ratio, 2.87; 95% confidence interval (CI), 1.2–7.1; P = 0.02, Supplementary Table S4]. Patients with PD-L2+ tumors displayed a shorter OS compared with those with less than 5% positive tumor cells (P = 0.005, Fig. 2A), but no impact on the DFS was found (P = 0.13, Fig. 2A). Univariately, patients with PD-L2+ tumors were 3.4 times more likely to die than those with negative tumor cells (risk ratio, 3.4; 95% CI, 1.4–8.5; P = 0.005, Supplementary Table S4).

Patients with tumors exhibiting both high densities of PD-1+ lymphocytes in the IM and PD-L1+ and/or PD-L2+ tumor cells (n = 11) had the worst prognosis, as assessed by DFS and OS (DFS, P = 1.3 × 10−5; OS, P = 0.005; Fig. 2B). Patients that met these criteria had 6.1 times more risk to progress after resection than patients who did not (risk ratio, 6.08; 95% CI, 2.4–15.0; P < 0.001, Supplementary Table S4). Strikingly, 91% of the patients having a PD1High infiltrate and tumor cells expressing PD-L1 and/or PD-L2 progressed in the subsequent 5 years of surgery versus 36% in the negative group (complete 5-years follow-up n = 70, Fisher exact test P = 0.001, Fig. 2C). In the TCGA public datasets of 496 primary ccRCC, the gene expression of PD-1 (PDCD1), LAG-3, and PD-L2 (PDCD1LG) was associated with shorter OS (P = 0.03, P = 0.0001, and P = 0.0003, respectively) whereas PD-L1 (CD274) was not (P = 0.67; Fig. 2D).

It has been reported that IFNγ can induce PD-L1 expression on tumor cells (27). We found a significant positive correlation between the gene expression of IFNG with PD-L1 (R = 0.13, P = 0.004) and with PD-L2 (R = 0.42, P = 2.2 × 10−16) in the TCGA cohort (Fig. 3A). In addition, a significant positive correlation was found between the densities of PD-1+ and CD8+ T cells (R = 0.31, P = 0.004), LAG-3+ and CD8+ T cells (R = 0.42, P = 0.001) and PD-1+ and LAG-3+ cells (R = 0.55, P < 0.0001) in the IM of the 80 primary ccRCC (Fig. 3B and 3C). An even stronger correlation was found at the gene-expression level between PD-1 and LAG-3 (R = 0.81, P = 0.002), PD-1 and CD8A (R = 0.95, P < 0.001), and LAG-3 and CD8A (R = 0.96, P < 0.001) in the TCGA cohort of 496 primary tumors (Fig. 3B and C). In accordance with all these observations, we also detected the presence of triple-positive CD8+/PD-1+/LAG-3+ cells in 6 of 7 cases from the CD8High group of primary tumors by IF on paraffin sections (representative pictures are displayed in Fig. 3D).

Figure 3.

Expression of immune checkpoints correlates with CD8+ T cells infiltration in primary ccRCC. Dot plot of the gene expression of PD-L1 (red dots) and PD-L2 (blue dots; y-axis) against IFNG (x-axis; A). Dot plot of the Log10 cell density and gene expression of PD-1 (red dots) and LAG-3 (blue dots; y-axis) against CD8 (x-axis; B). Dot plot of the log10 cell density and gene expression of PD-1 (y-axis) against LAG-3 (x-axis; C). Pearson r value and the number of samples for each correlation are displayed. IF staining on 1 paraffin-embedded ccRCC showing the colocalization of CD8 (green), PD-1 (red), and LAG-3 (white) proteins in lymphocytes (D); nuclei are stained with DAPI.

Figure 3.

Expression of immune checkpoints correlates with CD8+ T cells infiltration in primary ccRCC. Dot plot of the gene expression of PD-L1 (red dots) and PD-L2 (blue dots; y-axis) against IFNG (x-axis; A). Dot plot of the Log10 cell density and gene expression of PD-1 (red dots) and LAG-3 (blue dots; y-axis) against CD8 (x-axis; B). Dot plot of the log10 cell density and gene expression of PD-1 (y-axis) against LAG-3 (x-axis; C). Pearson r value and the number of samples for each correlation are displayed. IF staining on 1 paraffin-embedded ccRCC showing the colocalization of CD8 (green), PD-1 (red), and LAG-3 (white) proteins in lymphocytes (D); nuclei are stained with DAPI.

Close modal

Metastases.

Metastatic ccRCC has been one cancer in which antibodies inhibiting the PD-1 axis have induced remarkable tumor regression in some patients (20), highlighting the necessity to define prognostic and predictive immune-based biomarkers in this disease. Of the 51 patients with ccRCC lung metastasis, 13 were considered as PD-1High, and the latter displayed a shorter OS (P = 0.008, Fig. 4A). Seven of 51 were considered as LAG-3High, and again they displayed a shorter OS (P = 0.048, Fig. 4A).

Figure 4.

Expression of immune checkpoints correlates with unfavorable clinical outcome for patients with ccRCC lung metastases. OS according to the presence of a high or low density of PD-1+ and LAG-3+ cells, and the expression of PD-L1 or PD-L2 by >5% of the tumor cells in ccRCC lung metastases (A). OS (B) and pie chart (C) representing the percentage of patients that had died 5 years after the metastasectomy according to the expression of PD-L1 and/or PD-L2 on tumor cells related with high densities of PD-1+ lymphocytes. Dot plot of the Log10 density of PD-1+ (black dots) and LAG-3+ cells (gray dots; y-axis) against CD8 (x-axis; D); Pearson r value for each correlation is displayed.

Figure 4.

Expression of immune checkpoints correlates with unfavorable clinical outcome for patients with ccRCC lung metastases. OS according to the presence of a high or low density of PD-1+ and LAG-3+ cells, and the expression of PD-L1 or PD-L2 by >5% of the tumor cells in ccRCC lung metastases (A). OS (B) and pie chart (C) representing the percentage of patients that had died 5 years after the metastasectomy according to the expression of PD-L1 and/or PD-L2 on tumor cells related with high densities of PD-1+ lymphocytes. Dot plot of the Log10 density of PD-1+ (black dots) and LAG-3+ cells (gray dots; y-axis) against CD8 (x-axis; D); Pearson r value for each correlation is displayed.

Close modal

On the basis of a 5% cutoff, 5 of 51 (10%), and 15 of 51 (29%) metastases were PD-L1+ or PD-L2+, respectively; of them, 3 were double positive. Whereas patients with PD-L1+ metastases did not have a significantly worse clinical outcome (P = 0.12, Fig. 4A), the expression of PD-L2 on tumor cells was associated with a shorter OS (P = 0.03, Fig. 4A). Univariately, patients with PD-L2+ metastasis were 2.2 times more likely to die compared with patients with PD-L2- ones (risk ratio, 2.17; 95% CI, 1.03–4.35; P = 0.04, Supplementary Table S4).

Patients with high densities of PD-1+ lymphocytes and PD-L1+ and/or PD-L2+ tumor cells in their ccRCC lung metastases (n = 12) had the worst prognosis as assessed by OS (P = 0.003, Fig. 4B). The patients that met these criteria had 3.1 times more risk to die after metastasectomy than patients who did not (risk ratio, 3.1; 95% CI, 1.28–6.66; P = 0.003, Supplementary Table S4). Strikingly, 100% of the patients having a PD1High infiltrate and metastases simultaneously expressing one or both of its ligands (PD-L1 or PD-L2) died in the subsequent 5 years after surgery versus 57% in the negative group (Fisher tests, P value = 0.004, Fig. 4C).

A significant and strong correlation was found between densities of PD-1+ and CD8+ T cells (R = 0.51, P = 0.0001), LAG-3+ and CD8+ T cells (R = 0.4, P = 0.004; Fig. 4D) and PD-1+ and LAG-3+ cells (R = 0.71, P < 0.0001).

In conclusion, the combined analysis of the expression of PD-1, PD-L1, and PD-L2 identified a group of patients with a high risk of progression and death in primary and in an independent cohort of metastatic ccRCC.

Opposite correlations of TLS-DC and NTLS-DC with prognosis and their association with immune checkpoints

It has been shown that TLS-DC orchestrate intratumoral CD8+ cytotoxic T cells and Th1 responses in NSCLC (3, 4). In the primary ccRCC cohort (n = 135), DC-Lamp+ cells were detected within TLS (TLS-DC) in the IM. They coexpressed CD83 and high amounts of MHC Class II, and were localized in the vicinity of PNAd+ high endothelial venules (HEV; Fig. 5A) as described in other tumor types (3). There was a trend where high densities of TLS-DC were associated with longer DFS (OPv = 0.09), but not with OS (Supplementary Table S4). TLS-DC densities in the TC were not associated with prognosis. Interestingly, high densities of TLS-DC (based on the OPv cutoff) in the IM identified a group of patients with good prognosis among the CD8High group for both DFS (P = 0.001, Fig. 5A) and OS (P = 0.03, Fig. 5A).

Figure 5.

Characteristics of TLS-DC and NTLS-DC and their relationships with prognosis and immune checkpoints. IHC photomicrographs of DC-Lamp(Red)/CD3(Blue) illustrating the presence of mature DC in TLS (A, white arrows) or outside TLS (B, black arrows); DC-Lamp(Red)/PNAd(Blue) and DC-Lamp(Red)/CD34(Blue) illustrating TLS-DC near HEV (A) and NTLS-DC near endothelial cells (B) in primary ccRCC. IF staining showing the colocalization of CD3 (green), DC-Lamp (red) with HLA-DR or CD83 (white) expression in TLS-DC (A), but not in NTLS-DC (B); nuclei are stained with DAPI. OS and DFS according to the presence of a high- or low-density TLS-DC in the CD8High group (A, bottom) or NTLS-DC in the entire cohort (B, bottom). Dot plot of the PD-1+ against TLS-DC+ cell densities (blue dots; C); Pearson r value is displayed. OS and DFS according to the presence of a CD8High and TLS-DCLow and/or PD-1High and PD-L1 and/or L2+ immune profiles (D).

Figure 5.

Characteristics of TLS-DC and NTLS-DC and their relationships with prognosis and immune checkpoints. IHC photomicrographs of DC-Lamp(Red)/CD3(Blue) illustrating the presence of mature DC in TLS (A, white arrows) or outside TLS (B, black arrows); DC-Lamp(Red)/PNAd(Blue) and DC-Lamp(Red)/CD34(Blue) illustrating TLS-DC near HEV (A) and NTLS-DC near endothelial cells (B) in primary ccRCC. IF staining showing the colocalization of CD3 (green), DC-Lamp (red) with HLA-DR or CD83 (white) expression in TLS-DC (A), but not in NTLS-DC (B); nuclei are stained with DAPI. OS and DFS according to the presence of a high- or low-density TLS-DC in the CD8High group (A, bottom) or NTLS-DC in the entire cohort (B, bottom). Dot plot of the PD-1+ against TLS-DC+ cell densities (blue dots; C); Pearson r value is displayed. OS and DFS according to the presence of a CD8High and TLS-DCLow and/or PD-1High and PD-L1 and/or L2+ immune profiles (D).

Close modal

However, in contrast with NSCLC where most of the DC-Lamp+ cells are localized within TLS (4), in ccRCC a large percentage (79% ± 20%) of DC-Lamp+ cells with DC morphology was found isolated and outside TLS (NTLS-DC, Fig. 5B). They colocalized with CD34+ blood vessels, but not with PNAd+ HEV, expressed low amounts of MHC class II and were CD83 negative (Fig. 5B). The majority of NTLS-DC was localized in the IM (71 ± 14%) and their high densities (based on the OPv cut-off) were associated with poor clinical outcome in primary ccRCC patients (DFS P = 0.006, OS P = 5.1 × 10−5, Fig. 5B).

The opposite influences of TLS-DC and NTLS-DC on prognosis prompted us to explore their relationships with the expression of PD-1 and its ligands in the CD8High group of patients. A negative correlation was found between the densities of TLS-DC and PD-1+ cells (r = −0.23, P = 0.04, Fig. 5C). Tumors containing PD-L1+ and/or PD-L2+ tumor cells exhibited less TLS-DC (P = 0.014; Supplementary Fig. S4), but similar densities of CD8+ T cells (P = 0.96). In contrast, the density of NTLS-DC was associated with the tumor expression of PD-L1 (OR, 6.54; 95% CI, 1.23–45.45; P = 0.012) and PD-L2 (OR, 2.7; 95% CI, 1.2–18.1; P = 0.04). Most of the patients that were PD-1High and PD-L1+ and/or L2+ (8 of 11) were also CD8High and TLS-DCLo in primary ccRCC (OR, 15.02; 95% CI, 1.66–72.7; P = 0.004), and the presence of one or both patterns correlated with shorter DFS (P < 0.0001) and OS (P = 0.001, Fig. 5D).

Identification of a group of patients with worst prognosis in primary and metastatic ccRCC

To define the independent prognostic significance of the previously mentioned immune profiles (CD8High and TLS-DCLow or PD-1High and PD-L1+and/orL2+), we included them in a multivariate Cox regression analysis with the other significant prognostic clinical variables (TNM and Furhman grade). The strongest independent poor prognostic factors in primary ccRCC for DFS were to have a CD8High and TLS-DCLow (P = 0.001, Table 1) or PD-1High and PD-L1+and/orPD-L2+ (P = 0.03, Table 1) immune profile. For the metastatic cohort, we found that the strongest independent worst prognostic variable for OS was being CD8High (P = 0.004, Table 1) or having a tumor with PD-1High and PD-L1+ and/or PD-L2+ immune profile (P = 0.02, Table 1).

Table 1.

Multivariate Cox regression analysis for DFS and OS of the pathologic and immune variables in primary and metastatic ccRCC

Multivariate Cox regression analysis DFSMultivariate Cox regression analysis OS
Primary ccRCCHR (95% CI)(lower–upper)PHR (95% CI)(lower–upper)P
CD8high TLS-DClow (yes vs. no) 3.19 (1.42–3.83) 0.001 1.57 (0.45–1.82) 0.41 
PD-L1 or L2 5%–100% + PD-1high (yes vs. no) 1.88 (1.56–2.89) 0.03 2.22 (0.80–1.16) 0.24 
Fuhrman grade 1.90 (0.99–2.00) 0.06 2.68 (0.99–2.16) 0.32 
TNM stage (I–IV) 1.59 (0.18–2.33) 0.12 2.74 (1.01–3.05) 0.002 
 Multivariate Cox regression analysis OS   
Metastatic ccRCC HR (95% CI)(lower–upper) P   
CD8 group (high vs. low) 2.86 (1.21–3.11) 0.004   
PD-L1 or L2 5%–100% + PD-1high (yes vs. no) 1.41 (1.19–2.48) 0.02   
Lymph node invasion (yes vs. no) 0.72 (0.35–1.42) 0.47   
Multivariate Cox regression analysis DFSMultivariate Cox regression analysis OS
Primary ccRCCHR (95% CI)(lower–upper)PHR (95% CI)(lower–upper)P
CD8high TLS-DClow (yes vs. no) 3.19 (1.42–3.83) 0.001 1.57 (0.45–1.82) 0.41 
PD-L1 or L2 5%–100% + PD-1high (yes vs. no) 1.88 (1.56–2.89) 0.03 2.22 (0.80–1.16) 0.24 
Fuhrman grade 1.90 (0.99–2.00) 0.06 2.68 (0.99–2.16) 0.32 
TNM stage (I–IV) 1.59 (0.18–2.33) 0.12 2.74 (1.01–3.05) 0.002 
 Multivariate Cox regression analysis OS   
Metastatic ccRCC HR (95% CI)(lower–upper) P   
CD8 group (high vs. low) 2.86 (1.21–3.11) 0.004   
PD-L1 or L2 5%–100% + PD-1high (yes vs. no) 1.41 (1.19–2.48) 0.02   
Lymph node invasion (yes vs. no) 0.72 (0.35–1.42) 0.47   

Compared with other neoplasia, the immune microenvironment in ccRCC has not yet been studied in detail. However, it is of paramount importance to understand its regulation in view of the paradoxical correlation of CD8+ T-cell infiltration with poor prognosis (13, 14) and the recent advances of immunotherapy with anti-checkpoint antibodies (anti PD-1 and anti PD-L1; refs. 20, 21).

ccRCC has been described as a proinflammatory neoplasia where tumor cells produce several cytokines (such as VEGF, IL6 and TGFβ; refs. 33–35) that may lead to the recruitment and activation of polyclonal CD8+ T cells (36–39). Our data suggest that these recruited CD8+ T cells could only be locally educated when high densities of TLS-DC are present, and in only in these cases their density correlates with favorable prognosis. ccRCC is the first tumor type where a large proportion of DC-Lamp+ cells outside TLS structures have been observed (NTLS-DC). Interestingly, we found that these cells do not express activation and costimulatory markers, and they are probably recruited directly from the blood into the tumor stroma—contrary to the usual path that DC-Lamp+ cells follow in other type of tumors (5, 40). Accordingly, several studies have shown that the ccRCC microenvironment can induce a dysfunctional DC maturation, a downregulation of costimulatory molecules and tolerogenicity (34, 41, 42), whereas DC in the TLS are likely to be protected from these effects (43). Altogether, our results suggest that the particular proinflammatory milieu initiated by tumor cells induces the recruitment of CD8+ T cells that—due to the low number of fully functional mature DC present in specialized T-cell–priming sites, or the presence of DC with suppressive phenotype—are not able to mount an effective antitumor immune response, but rather express exhaustion/inhibition molecules.

This work reinforces the concept that T-cell exhaustion/inhibition (44) plays an important role in ccRCC pathogenesis. It has been described that the density of PD-1+ cells (15, 16) and the tumor expression of PD-L1 (17–19) in primary ccRCC are associated with a poor clinical outcome. In this study, we confirm these findings and extend them to ccRCC lung metastases. This information is highly relevant because metastatic ccRCC-treated patients have shown one of the highest objective durable response rates to PD-1 blockade (approximately 30%; ref. 20) and many efforts are being dedicated to define theranostic tools in this pathology. Furthermore, we describe for the first time the prognostic significance of PD-L2. This molecule seems to be expressed in a higher proportion of tumors than PD-L1, and up to 30% of them might express it solely according to our results. This might be of clinical relevance because—although in two different nonrandomized cohorts—the anti–PD-L1 treatment alone seems to have a lower response rate than the anti–PD-1 (12% and 27%, respectively; refs. 20, 21). Furthermore, there are PD-L1–negative tumors that respond to anti–PD-1 treatment (22), suggesting that indeed there are other molecules beside PD-L1 implicated in the PD-1 inhibition axis of ccRCC. Few publications have reported PD-L2 mRNA or protein expression in other tumors, including primary mediastinal large B-cell lymphoma (29), NSCLC (45), ovarian (46), and esophageal (47), where it has shown a limited impact of patients' prognosis.

To our knowledge, this is the first report on the poor prognostic impact associated with high densities of LAG-3+ cells in human tumors. Furthermore, we provide clear evidence that the expression of PD-1 and LAG-3 is highly correlated in ccRCC. Some studies on mouse models have shown a synergistic effect of the inhibition of both pathways in boosting antitumor immune response (24). Therefore, our data support the rationale of dual blockade of these molecules in ccRCC.

Although recent works have emphasized that PD-L1 is preferentially upregulated by IFNγ (31, 48), whereas PD-L2 is regulated by IL4 (49), the weak association of PD-L1 mRNA with IFNG might suggest an important role for posttranscriptional regulation of PD-L1 expression in ccRCC as for other aggressive tumors (50). Furthermore, it is highly suggestive that IFNγ can also induce PD-L2 upregulation in tumors, as in immune cells (51), and supports the rationale to use therapeutic antibodies targeting this ligand. Taken together, our results suggest that the expression of these molecules is related with a chronic inflammatory and highly suppressive process that is unselectively recruiting CD8+ and NTLS-DC cells from the circulation, and overall is associated with a poor prognosis.

Another characteristic of ccRCC is the lack of prognostic significance of the immune cells in the TC. Although CD8+, PD-1+, LAG-3+, and NTLS-DC densities in the IM of the primary tumors were associated with poor prognosis, they had no prognostic significance when present in the TC. In colorectal carcinoma, both regions are important to define the best immune score, which correlates with patient's longer survival (30). Whether this dichotomy between ccRCC and colorectal carcinoma reflects different tumor tissue organization or relates to other factors of the tumor microenvironment remains to be clarified.

Recent unsupervised gene clustering of stage IV primary ccRCC showed that tumors with high inflammatory immune infiltrate (approximately 15%) also have a high expression of PDCD1 (PD-1) and its ligands, and correlate with the worst prognosis (52). Indeed, this inflammatory/proangiogenic profile (probably originated from the ccRCC tumor cells) found in ccRCC primary tumors and in ccRCC lung metastases (14), is not found in colorectal carcinoma lung metastases (14), highly supporting that it may contribute to local immunosuppression process, the absence of fully mature DC, and high expression of immune checkpoints (53).

In summary, we identify a subset of primary and metastatic ccRCC patients characterized by (i) an extensive CD8+ T-cell infiltrate, (ii) expression of immune checkpoints, and (iii) the absence of fully functional DC, which is associated with poor clinical outcome. Our results highlight novel independent prognostic factors in ccRCC based on the concomitant quantification of densities of DC, CD8+, PD-1+, and LAG-3+ lymphocytes in addition to PD-L1/PD-L2 expression by tumor cells. These immune profiles should guide the selection of suitable patients to receive immunotherapies and need to be further validated in larger and independent cohorts. Because this choice depends on both the extent of CD8+ T-cell infiltration and the maturation and localization of DC, it invites revision of the idea that intratumoral CD8+ T cells are always associated with favorable prognosis in human tumors.

Y. Vano reports receiving other research grants from Novartis and is a consultant/advisory board member for Novartis and Pfizer. G.J. Freeman has ownership interest (including patents) in Amplimmune, Boehringer-Ingelheim, Bristol-Myers Squibb, EMD Serono, Merck, Novartis, and Roche and is a consultant/advisory board member for Novartis and Roche. S.M. Oudard reports receiving speakers bureau honoraria from and is a consultant/advisory board member for Janssen, Novartis, Pfizer, Roche, and Sanofi. W-H. Fridman is a consultant/advisory board member for Costim. No potential conflicts of interest were disclosed by the other authors.

Conception and design: N.A. Giraldo, G.J. Freeman, S. Oudard, W.H. Fridman, C. Sautès-Fridman

Development of methodology: N.A. Giraldo, G. Skliris, L. Lacroix, G.J. Freeman, M.-C. Dieu-Nosjean, S. Oudard

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): N.A. Giraldo, F. Pagès, V. Verkarre, Y. Vano, N. Saint-Aubert, L. Lacroix, I. Natario, A. Lupo, M. Alifano, D. Damotte, A. Cazes, S. Oudard

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): N.A. Giraldo, E. Becht, A. Lupo, M.-C. Dieu-Nosjean, S. Oudard, W.H. Fridman, C. Sautès-Fridman

Writing, review, and/or revision of the manuscript: N.A. Giraldo, E. Becht, F. Pagès, G. Skliris, Y. Vano, A. Mejean, F. Triebel, G.J. Freeman, M.-C. Dieu-Nosjean, S. Oudard, W.H. Fridman, C. Sautès-Fridman

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): N.A. Giraldo, F. Pagès, V. Verkarre, A. Mejean, L. Lacroix, D. Damotte, F. Triebel, S. Oudard, W.H. Fridman, C. Sautès-Fridman

Study supervision: N.A. Giraldo, W.H. Fridman, C. Sautès-Fridman

The authors thank Romain Remark for his help in the case/block selection and collection of clinical data from the metastatic cohort, Jose Balcaceres (Association pour la recherche sur les Thérapeutiques en Cancérologie) for his support in the collection of the clinical data of the primary cohort, Patricia Bonjour for cutting the paraffin-embedded blocks, Dr. Marc Riquet for the case selection of the metastatic cohort and all members of the teams 13 and 15 in the Cordeliers Research Center for their valuable discussions.

This work was supported by the “Institut National de la Santé et de la Recherche Médicale,” the University Paris-Descartes, the University Pierre et Marie Curie, the Institut National du Cancer (2011-1-PLBIO-06-INSERM 6-1 and PLBIO09-088-IDF-KROEMER), CARPEM (CAncer Research for PErsonalized Medicine), Labex Immuno-Oncology (LAXE62_9UMS872 FRIDMAN), Universidad de los Andes School of Medicine (to N.A. Giraldo), Colciencias (to N.A. Giraldo), and NIH (P50CA101942; to G.J. Freeman).

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.
Fridman
WH
,
Pagès
F
,
Sautès-Fridman
C
,
Galon
J
. 
The immune contexture in human tumours: impact on clinical outcome
.
Nat Rev Cancer
2012
;
12
:
298
306
.
2.
Giraldo
NA
,
Becht
E
,
Remark
R
,
Damotte
D
,
Sautès-Fridman
C
,
Fridman
WH
. 
The immune contexture of primary and metastatic human tumours
.
Curr Opin Immunol
2014
;
27
:
8
15
.
3.
Dieu-Nosjean
MC
,
Antoine
M
,
Danel
C
,
Heudes
D
,
Wislez
M
,
Poulot
V
, et al
Long-term survival for patients with non–small cell lung cancer with intratumoral lymphoid structures
.
J Clin Oncol
2008
;
26
:
4410
7
.
4.
Goc
J
,
Germain
C
,
Vo-Bourgais
TK
,
Lupo
A
,
Klein
C
,
Knockaert
S
, et al
Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells
.
Cancer Res
2014
;
74
:
705
15
.
5.
Dieu-Nosjean
MC
,
Goc
J
,
Giraldo
NA
,
Sautès-Fridman
C
,
Fridman
WH
. 
Tertiary lymphoid structures in cancer and beyond
.
Trends in Immunol
2014
;
35
:
571
80
.
6.
Bindea
G
,
Mlecnik
B
,
Tosolini
M
,
Kirilovsky
A
,
Waldner
M
,
Obenauf
AC
, et al
Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer
.
Immunity
2013
;
39
:
782
95
.
7.
Gu-Trantien
C
,
Loi
S
,
Garaud
S
,
Equeter
C
,
Libin
M
,
de Wind
A
. 
CD4+ follicular helper T-cell infiltration predicts breast cancer survival
.
J Clin Invest
2013
;
123
:
2873
92
.
8.
Becht
E
,
Goc
J
,
Germain
C
,
Giraldo
NA
,
Dieu-Nosjean
MC
,
Sautès-Fridman
C
, et al
Shaping of an effective immune microenvironment to and by cancer cells
.
Cancer Immunol Immunother
2014
;
63
:
991
7
.
9.
Maldonado
L
,
Teague
JE
,
Morrow
MP
,
Jotova
I
,
Wu
TC
,
Wang
C
, et al
Intramuscular therapeutic vaccination targeting HPV16 induces T-cell responses that localize in mucosal lesions
.
Sci Transl Med
2014
;
6
:
221ra13
.
10.
Lutz
ER
,
Wu
AA
,
Bigelow
E
,
Sharma
R
,
Mo
G
,
Soares
K
, et al
Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation
.
Cancer Immunol Res
2014
;
2
:
616
31
.
11.
Muris
JJ
,
Meijer
CJ
,
Cillessen
SA
,
Vos
W
,
Kummer
JA
,
Bladergroen
BA
, et al
Prognostic significance of activated cytotoxic T-lymphocytes in primary nodal diffuse large B-cell lymphomas
.
Leukemia
2004
;
18
:
589
96
.
12.
Scott
DW
,
Chan
FC
,
Hong
F
,
Rogic
S
,
Tan
KL
,
Meissner
B
, et al
Gene expression-based model using formalin-fixed paraffin-embedded biopsies predicts overall survival in advanced-stage classical Hodgkin lymphoma
.
J Clin Oncol
2013
;
31
:
692
700
.
13.
Nakano
O
,
Sato
M
,
Naito
Y
,
Suzuki
K
,
Orikasa
S
,
Aizawa
M
, et al
Proliferative activity of intratumoral CD8(+) T-lymphocytes as a prognostic factor in human renal cell carcinoma: clinicopathologic demonstration of antitumor immunity
.
Cancer Res
2001
;
61
:
5132
6
.
14.
Remark
R
,
Alifano
M
,
Cremer
I
,
Lupo
A
,
Dieu-Nosjean
MC
,
Riquet
M
, et al
Characteristics and clinical impacts of the immune environments in colorectal and renal cell carcinoma lung metastases: influence of tumor origin
.
Clin Cancer Res
2013
;
19
:
4079
91
.
15.
Thompson
RH
,
Dong
H
,
Lohse
CM
,
Leibovich
BC
,
Blute
ML
,
Cheville
JC
, et al
PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma
.
Clin Cancer Res
2007
;
13
:
1757
61
.
16.
Kang
MJ
,
Kim
KM
,
Bae
JS
,
Park
HS
,
Lee
H
,
Chung
MJ
, et al
Tumor-infiltrating PD1-positive lymphocytes and FoxP3-positive regulatory T cells predict distant metastatic relapse and survival of clear cell renal cell carcinoma
.
Transl Oncol
2013
;
6
:
282
9
.
17.
Thompson
RH
,
Gillett
MD
,
Cheville
JC
,
Lohse
CM
,
Dong
H
,
Webster
WS
, et al
Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
.
Proc Natl Acad Sci U S A
2004
;
101
:
17174
9
.
18.
Thompson
RH
,
Gillett
MD
,
Cheville
JC
,
Lohse
CM
,
Dong
H
,
Webster
WS
, et al
Costimulatory molecule B7-H1 in primary and metastatic clear cell renal cell carcinoma
.
Cancer
2005
;
104
:
2084
91
.
19.
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
.
20.
Topalian
SL
,
Hodi
FS
,
Brahmer
JR
,
Gettinger
SN
,
Smith
DC
,
McDermott
DF
, et al
Safety, activity, and immune correlates of anti–PD-1 antibody in cancer
.
N Engl J Med
2012
;
366
:
2443
54
.
21.
Brahmer
JR
,
Tykodi
SS
,
Chow
LQ
,
Hwu
WJ
,
Topalian
SL
,
Hwu
P
, et al
Safety and activity of anti-PD-L1 antibody in patients with advanced cancer
.
N Engl J Med
2012
;
366
:
2455
65
.
22.
Taube
JM
,
Klein
A
,
Brahmer
JR
,
Xu
H
,
Pan
X
,
Kim
JH
, et al
Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti–PD-1 therapy
.
Clin Cancer Res
2014
;
20
:
5064
74
.
23.
Gros
A
,
Robbins
PF
,
Yao
X
,
Li
YF
,
Turcotte
S
,
Tran
E
, et al
PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors
.
J Clin Invest
2014
;
124
:
2246
59
.
24.
Woo
SR
,
Turnis
ME
,
Goldberg
MV
,
Bankoti
J
,
Selby
M
,
Nirschl
CJ
, et al
Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape
.
Cancer Res
2012
;
72
:
917
27
.
25.
Synapse contribute to the cure [Internet]
.
Seattle
:
Sage Bionetworks
(updated on 2013–04–06). Available from:
https://www.synapse.org
using the accession number syn1446101
.
26.
Störkel
S
,
Eble
JN
,
Adlakha
K
,
Amin
M
,
Blute
ML
,
Bostwick
DG
, et al
Classification of renal cell carcinoma: workgroup No. 1. Union internationale contre le cancer (UICC) and the American Joint Committee on Cancer (AJCC)
.
Cancer
1997
;
80
:
987
9
.
27.
Fuhrman
SA
,
Lasky
LC
,
Limas
C
. 
Prognostic significance of morphologic parameters in renal cell carcinoma
.
Am J Surg Pathol
1982
;
6
:
655
63
.
28.
Edge
S
,
Byrd
DR
,
Compton
CC
,
Fritz
AG
,
Greene
FL
,
Trotti
A
. 
Kidney
. In: 
AJCC Cancer Staging Manual
.
Chicago
:
Springer-Verlag
; 
2010
. p.
479
89
.
29.
Shi
M
,
Roemer
MG
,
Chapuy
B
,
Liao
X
,
Sun
H
,
Pinkus
GS
, et al
Expression of programmed cell death 1 ligand 2 (PD-L2) Is a distinguishing feature of primary mediastinal (Thymic) large B-cell lymphoma and associated with PDCD1LG2 copy gain
.
Am J Surg Pathol
2014
;
38
:
1715
23
.
30.
Galon
J
,
Costes
A
,
Sanchez-Cabo
F
,
Kirilovsky
A
,
Mlecnik
B
,
Lagorce-Pagès
C
, et al
Type, density, and localization of immune cells within human colorectal tumors predict clinical outcome
.
Science
2006
;
313
:
1960
4
.
31.
Taube
JM
,
Anders
RA
,
Young
GD
,
Xu
H
,
Sharma
R
,
McMiller
TL
, et al
Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape
.
Sci Transl Med
2012
;
4
:
127ra37
.
32.
Altman
DG
,
Lausen
B
,
Sauerbrei
W
,
Schumacher
M
. 
Dangers of using “optimal” cutpoints in the evaluation of prognostic factors
.
J Natl Cancer Inst
1994
;
86
:
829
35
.
33.
Alberti
L
,
Thomachot
MC
,
Bachelot
T
,
Menetrier-Caux
C
,
Puisieux
I
,
Blay
JY
. 
IL-6 as an intracrine growth factor for renal carcinoma cell lines
.
Int J Cancer
2004
;
111
:
653
61
.
34.
Cabillic
F
,
Bouet-Toussaint
F
,
Toutirais
O
,
Rioux-Leclercq
N
,
Fergelot
P
,
de la Pintière
CT
, et al
Interleukin-6 and vascular endothelial growth factor release by renal cell carcinoma cells impedes lymphocyte-dendritic cell cross-talk
.
Clin Exp Immunol
2006
;
146
:
518
23
.
35.
Romero
JM
,
Aptsiauri
N
,
Vazquez
F
,
Cozar
JM
,
Canton
J
,
Cabrera
T
, et al
Analysis of the expression of HLA class I, proinflammatory cytokines and chemokines in primary tumors from patients with localized and metastatic renal cell carcinoma
.
Tissue Antigens
2006
;
68
:
303
10
.
36.
Van den Hove
LE
,
Van Gool
SW
,
Van Poppel
H
,
Baert
L
,
Coorevits
L
,
Van Damme
B
, et al
Phenotype, cytokine production and cytolytic capacity of fresh (uncultured) tumour-infiltrating T lymphocytes in human renal cell carcinoma
.
Clin Exp Immunol
1997
;
109
:
501
9
.
37.
Shabtai
M
,
Ye
H
,
Frischer
Z
,
Martin
J
,
Waltzer
WC
,
Malinowski
K
. 
Increased expression of activation markers in renal cell carcinoma infiltrating lymphocytes
.
J Urol
2002
;
168
:
2216
9
.
38.
Sittig
SP
,
Køllgaard
T
,
Grønbæk
K
,
Idorn
M
,
Hennenlotter
J
,
Stenzl
A
, et al
Clonal expansion of renal cell carcinoma-infiltrating T lymphocytes
.
Oncoimmunology
2013
;
2
:
e26014
.
39.
Gerlinger
M
,
Quezada
SA
,
Peggs
KS
,
Furness
AJ
,
Fisher
R
,
Marafioti
T
, et al
Ultra-deep T-cell receptor sequencing reveals the complexity and intratumour heterogeneity of T-cell clones in renal cell carcinomas
.
J Pathol
2013
;
231
:
424
32
.
40.
de Chaisemartin
L
,
Goc
J
,
Damotte
D
,
Validire
P
,
Magdeleinat
P
,
Alifano
M
, et al
Characterization of chemokines and adhesion molecules associated with T-cell presence in tertiary lymphoid structures in human lung cancer
.
Cancer Res
2011
;
71
:
6391
9
.
41.
Gigante
M
,
Blasi
A
,
Loverre
A
,
Mancini
V
,
Battaglia
M
,
Selvaggi
FP
, et al
Dysfunctional DC subsets in RCC patients: ex vivo correction to yield an effective anti-cancer vaccine
.
Mol Immunol
2009
;
46
:
893
901
.
42.
Teng
L
,
Chen
Y
,
Ding
D
,
Dai
H
,
Liu
G
,
Li
C
. 
Immunosuppressive effect of renal cell carcinoma on phenotype and function of dendritic cells
.
Int Urol Nephrol
2014
;
46
:
915
20
.
43.
Troy
AJ
,
Summers
KL
,
Davidson
PJ
,
Atkinson
CH
,
Hart
DN
. 
Minimal recruitment and activation of dendritic cells within renal cell carcinoma
.
Clin Cancer Res
1998
;
4
:
585
93
.
44.
Speiser
DE
,
Utzschneider
DT
,
Oberle
SG
,
Münz
C
,
Romero
P
,
Zehn
D
. 
T cell differentiation in chronic infection and cancer: functional adaptation or exhaustion
?
Nat Rev Immunol
2014
;
14
:
768
74
.
45.
Zhang
Y
,
Wang
L
,
Li
Y
,
Pan
Y
,
Wang
R
,
Hu
H
, et al
Protein expression of programmed death 1 ligand 1 and ligand 2 independently predict poor prognosis in surgically resected lung adenocarcinoma
.
Onco Targets Ther
2014
;
7
:
567
73
.
46.
Hamanishi
J
,
Mandai
M
,
Iwasaki
M
,
Okazaki
T
,
Tanaka
Y
,
Yamaguchi
K
, et al
Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer
.
Proc Natl Acad Sci U S A
2007
;
104
:
3360
5
.
47.
Ohigashi
Y
,
Sho
M
,
Yamada
Y
,
Tsurui
Y
,
Hamada
K
,
Ikeda
N
, et al
Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer
.
Clin Cancer Res
2005
;
11
:
2947
53
.
48.
Lyford-Pike
S
,
Peng
S
,
Young
GD
,
Taube
JM
,
Westra
WH
,
Akpeng
B
, et al
Evidence for a role of the PD-1:PD-L1 pathway in immune resistance of HPV-associated head and neck squamous cell carcinoma
.
Cancer Res
2013
;
73
:
1733
41
.
49.
Quandt
D
,
Jasinski-Bergner
S
,
Müller
U
,
Schulze
B
,
Seliger
B
. 
Synergistic effects of IL-4 and TNFα on the induction of B7-H1 in renal cell carcinoma cells inhibiting allogeneic T cell proliferation
.
J Transl Med
2014
;
12
:
151
.
50.
Parsa
AT
,
Waldron
JS
,
Panner
A
,
Crane
CA
,
Parney
IF
,
Barry
JJ
, et al
Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma
.
Nat Med
2007
;
13
:
84
8
.
51.
Latchman
Y
,
Wood
CR
,
Chernova
T
,
Chaudhary
D
,
Borde
M
,
Chernova
I
, et al
PD-L2 is a second ligand for PD-1 and inhibits T cell activation
.
Nat Immunol
2001
;
2
:
261
8
.
52.
Beuselinck
B
,
Job
S
,
Becht
E
,
Karadimou
A
,
Verkarre
V
,
Couchy
G
, et al
Molecular subtypes of clear cell renal cell carcinoma are associated with sunitinib response in the metastatic setting
.
Clin Cancer Res
2015
;
in press
.
53.
Mauge
L
,
Terme
M
,
Tartour
E
,
Helley
D
. 
Control of the adaptive immune response by tumor vasculature
.
Front Oncol
2014
;
4
:
61
.

Supplementary data