Purpose: There is currently no reliable biomarker to predict who would benefit from anti-PD-1/PD-L1 inhibitors. We comprehensively analyzed the immunogenomic properties in The Cancer Genome Atlas (TCGA) according to the classification of tumor into four groups based on PD-L1 status and tumor-infiltrating lymphocyte recruitment (TIL), a combination that has been suggested to be a theoretically reliable biomarker of anti-PD-1/PD-L1 inhibitors.

Experimental Design: The RNA expression levels of PD-L1 and CD8A in the samples in the pan-cancer database of TCGA (N = 9,677) were analyzed. Based on their median values, the samples were classified into four tumor microenvironment immune types (TMIT). The mutational profiles, PD-L1 amplification, and viral association of the samples were compared according to the four TMITs.

Results: The proportions of TMIT I, defined by high PD-L1 and CD8A expression, were high in lung adenocarcinoma (67.1%) and kidney clear cell carcinoma (64.8%) among solid cancers. The number of somatic mutations and the proportion of microsatellite instable-high tumor in TMIT I were significantly higher than those in other TMITs, respectively (P < 0.001). PD-L1 amplification and oncogenic virus infection were significantly associated with TMIT I, respectively (P < 0.001). A multivariate analysis confirmed that the number of somatic mutations, PD-L1 amplification, and Epstein–Barr virus/human papillomavirus infection were independently associated with TMIT I.

Conclusions: TMIT I is associated with a high mutational burden, PD-L1 amplification, and oncogenic viral infection. This integrative analysis highlights the importance of the assessment of both PD-L1 expression and TIL recruitment to predict responders to immune checkpoint inhibitors. Clin Cancer Res; 22(9); 2261–70. ©2016 AACR.

See related commentary by Schalper et al., p. 2102

This article is featured in Highlights of This Issue, p. 2097

Translational Relevance

This study classified tumors and their microenvironments into four groups based on PD-L1 expression and tumor-infiltrating lymphocyte (TIL) recruitment, as assessed by CD8A expression, in the pan-cancer RNA-sequencing database of The Cancer Genome Atlas (TCGA, N = 9,677). This immunogenomic perspective of four tumor microenvironment immune types (TMIT) would be an appropriate approach for cancer immunotherapy. Across all cancer types in TCGA, TMIT I, defined by high PD-L1 and CD8A expression, was significantly associated with a high number of somatic mutations and neoantigen, PD-L1 amplification, and infection with an oncogenic virus, such as Epstein–Barr virus or human papillomavirus. This integrative analysis highlights the importance of the assessment of both PD-L1 expression and TIL recruitment to predict responders to anti-PD-1/PD-L1 therapies.

A recent strategy targeting immune checkpoints such as CTLA-4 and PD-1/PD-L1 shows promising clinical benefits and introduces a paradigm shift in cancer treatment. Immune checkpoint-blocking agents have shown a remarkable clinical efficacy with a long response duration in immunogenic tumors, such as melanoma, renal cell carcinoma, bladder cancer, non–small cell lung carcinoma, and Hodgkin's lymphoma (1–5).

Although the expression of PD-L1 on the surface of tumor cells, as measured by immunohistochemistry, may potentially serve as a predictive factor to identify patients who would benefit from anti-PD-1/PD-L1 therapy, not all PD-L1-positive patients respond well (4–6). Therefore, PD-L1 expression on tumor cells may not be a simple predictive factor. Interestingly, the degree of tumor-infiltrating lymphocyte (TIL) infiltration and PD-L1 expression in the tumor microenvironment (TME) are also correlated with the clinical outcomes of anti-PD-1/PD-L1 therapies (6 7). Moreover, recent advances in immuno-genomics have shown that tumors with a high mutational burden, abundant neoantigen, and microsatellite (MSI)-high status are associated with a good response to anti-PD-1/PD-L1 therapy (8–14). Moreover, oncogenic viruses such as Epstein–Barr virus (EBV) or human papillomavirus (HPV) are associated with an inflamed TME, and a favorable clinical outcome in response to anti-PD-1/PD-L1 therapy would also be expected (10, 15). Because of the complex nature of tumor immunity, a comprehensive immuno-genomic analysis that include analyses of the interaction between the tumor and TIL in the TME and investigations of the underlying reasons for the promotion of tumor immunogenicity is needed.

Although TIL assessment in the TME is challenging (15), Rooney and colleagues fascinatingly showed immune cytolytic activity by measuring the mRNA expression levels of granzyme A (GZMA) and perforin 1 (PRF1) from large-scale RNA-sequencing (RNAseq) data from The Cancer Genome Atlas (TCGA) (10). These researchers showed that RNAseq data constitute an appropriate model that can be used to assess the TME because the contamination of stromal cells surrounding the tumor would proportionally influence the TME gene expression profiles in an unbiased manner. However, the cytolytic activity assessed by measuring the granzyme A and perforin 1 expression levels would be influenced by the infiltration of not only CD8+ cytolytic T cells (CTL) but also other immune cells, such as natural killer T (NK-T) cells (10, 16). Because CD8+ CTL recruitment by the adaptive immune response rather than the activation of NK-T cells by innate immunity plays a crucial role in the antitumor activity of immune checkpoint inhibitors (17, 18), we aimed to investigate whether CD8A expression, instead of cytolytic activity, can be used to assess CD8+ CTLs in the TME.

The classification of tumors into four different types based on the presence or absence of CD8+ CTLs and PD-L1 expression was recently suggested (15, 19). Tumors with high PD-L1 expression and the presence of CD8+ CTLs in the microenvironment are classified as TME immune type I, which would only benefit from anti-PD-L1/PD-1 therapies (15, 19). However, this concept has not been investigated with a large-scale genomics database because a standardized methodology for assessing PD-L1 and CD8+ CTLs has not been clarified.

In this study, we classified a large set of TCGA pan-cancer samples into four TME immune types (TMIT) by measuring the mRNA expression levels of PD-L1 and CD8A. The aims of this TCGA pan-cancer analysis were to determine the associations between TMIT and (i) cancer type and clinicopathologic features, (ii) mutational burden, and (iii) PD-L1 amplification, which would provide strategic information for the use of immune checkpoint-blocking therapy.

Processing of genomic data from TCGA project

We used publicly available, level 3 data of TCGA in this study. Clinical information, gene-level somatic mutation data, copy number variation (CNV) data, and mRNA expression data obtained by RNAseq of the TCGA samples were downloaded from the UCSC Cancer Browser (https://genome-cancer.ucsc.edu) on June 3, 2015. To count the number of total somatic mutations, multiple somatic mutations including nonsynonymous mutations, insertion–deletion mutations, and silent mutations were each counted and summated, and germline mutations without somatic mutations were excluded. The amplification and deletion statuses in the CNV thresholded data, which was calculated using Gistic 2.0 (20), were documented as “2” and "−2", respectively, and mRNA expression data, which were generated using the Illumina HiSeq V2 platform, are presented as reads per kilobase per million (RPKM) and transformed into log 2 values for analysis. The patients' clinical data and microsatellite instability (MSI) status of the tumors were extracted from the TCGA database. The MSI status was available for 1,164 samples, which included COAD (N = 426) and STAD samples (N = 414). The detection of oncogenic viruses, such as EBV, HPV, and hepatitis B virus (HBV), in each tumor sample (N = 6,511) and the neoantigen number (N = 3,726) were also referenced in a previous report by Rooney and colleagues (10). Altogether, samples of 32 cancer types (N = 10,354) were included in the analysis (Supplementary Table S1).

Statistical analyses of genomic data

According to previous reports regarding the four types of immune evasion of cancer (15), after merging log 2-transformed values of the RPKM of PD-L1 and CD8A, we divided all of the TCGA samples into four groups as follows: type I, PD-L1 expression higher than the median and CD8A expression higher than the median; type II, PD-L1 expression lower than the median and CD8A expression lower than the median; type III, PD-L1 expression higher than the median and CD8A expression lower than the median; and type IV, PD-L1 expression lower than the median and CD8A expression higher than the median. The cytolytic activity of each sample was calculated using the log 2-transformed value of the geometric mean of GZMA and PRF1, as previously reported (10). The prognostic significance of the four TMIT was estimated using Kaplan–Meier plots (log-rank test) and Cox proportional hazards regression analysis. The statistical significance of two continuous variables, such as the total number of somatic mutations and the number of neoantigens, and the CD8A expression level, cytolytic activity, and PD-1 expression was calculated by linear regression analysis. The significance of the differences between continuous values, such as the number of mutations, interferon-gamma (IFNG) expression, and cytolytic activity, and categorical variables, such as TMIT, MSI status, and POLE mutation, was calculated by the Wilcoxon rank sum test or analysis of variance with Tukey post hoc test (for comparisons of more than three groups). Comparing the proportion of each TMIT according to categorical variables was performed by Fisher exact test. The number of somatic mutations and CNV, such as the amplification and deletion of a gene in a previously suggested set of 373 genes that are frequently altered in cancer (10, 21) with PD-L1 (CD274), were analyzed by logistic regression to determine whether these variables can significantly predict a specific TMIT. Univariate and multivariate logistic regression analyses were performed to determine whether there is a significant association between clinicopathologic characteristics and the ability to predict TMIT I. P values less than 0.05 were considered statistically significant. False discovery rates of less than 5% were applied to control type I errors in the analysis to determine which gene alterations would significantly predict a specific TMIT. All statistical analyses and data presentations were performed in R language 3.1.3 (http://www.r-project.org), with the exception of the survival analysis, which was performed in STATA version 12 (StataCorp LP).

Distribution and clinical implication of TMIT across cancer types

We analyzed 9,677 tumor samples from 32 cancer types included in the TCGA dataset (Supplementary Table S1). The median of the log 2-transformed RPKM values, which indicate the mRNA expression level, of PD-L1 and CD8A were 4.72 and 6.97, respectively. The log transformation has no effect on the division at the median of the distributions, but only improves the analysis of the correlation. The expression levels of PD-L1 and CD8A were generally positively correlated even though a considerable proportion of tumor samples were found to be PD-L1-high and CD8A-low or PD-L1-low and CD8A-high tumors (P < 0.001, R2 = 0.250; Fig. 1a and Supplementary Fig. S1). CD8A expression was significantly correlated with cytolytic activity (10) and PD-1 expression (P < 0.001, R2 = 0.718 and 0.712, respectively; Supplementary Fig. S2).

Figure 1.

Distribution of tumor microenvironment immune type (TMIT) and mutational burden across cancer type. A scatter plot of log 2-transformed values of RPKM of PD-L1 and CD8A is shown (A). The portion of TMIT [TMIT, I, red (34.6%); II, blue (34.6%); III, green (15.4%); IV; yellow (15.4%)] (B) and the log 2-transformed value of the total number of somatic mutations according to cancer types of TCGA (C) are graphed. The red bar indicates the median value of each column, and the blue dashed line indicates the median value of the total samples. NA, not applicable.

Figure 1.

Distribution of tumor microenvironment immune type (TMIT) and mutational burden across cancer type. A scatter plot of log 2-transformed values of RPKM of PD-L1 and CD8A is shown (A). The portion of TMIT [TMIT, I, red (34.6%); II, blue (34.6%); III, green (15.4%); IV; yellow (15.4%)] (B) and the log 2-transformed value of the total number of somatic mutations according to cancer types of TCGA (C) are graphed. The red bar indicates the median value of each column, and the blue dashed line indicates the median value of the total samples. NA, not applicable.

Close modal

All tumor samples were divided into four groups of TMIT according to the median values of PD-L1 and CD8A expression. Among all of the evaluated samples, 34.6% of the samples were classified as TMIT I, defined by high PD-L1 expression in the tumor and surrounding TME and high CD8A expression, which indicates a high proportion of CD8+ CTLs. The proportions of TMIT II (low PD-L1/low CD8A), III (high PD-L1/low CD8A), and IV (low PD-L1/high CD8A) were 34.6%, 15.4%, and 15.4%, respectively. The proportion of TMIT I, II, III, and IV samples of skin cutaneous melanoma (SKCM) were 43.4%, 33.2%, 5.9%, and 18.4%, respectively, and these values are comparable to those included in previous reports (38%, 41%, 1%, and 20%, respectively; refs. 15, 19).

The proportion of TMIT samples was analyzed according to cancer type (Fig. 1b). As expected, cancers derived from lymphoproliferative tissues, such as thymoma (THYM) and diffuse large B cell lymphoma (DLBC), had the highest proportion of TMIT I tumors among all cancer types (84.0% and 70.8%, respectively), further supporting the hypothesis that the signature reliably reflects the TMIT in cancer tissues. Among the solid cancers, lung adenocarcinoma (LUAD, 67.1%), kidney clear cell carcinoma (KIRC, 64.8%), lung squamous cell carcinoma (LUSC, 63.5%), and head and neck squamous cell carcinoma (HNSC, 54.1%) had the highest proportion of TMIT I samples. The clinicopathological features of the TCGA patients are summarized according to TMIT in Supplementary Table S2. Older patients (>60 years) had a significantly higher proportion of immune type I tumors compared with younger patients, although the actual difference between two groups was small (37.4% vs. 32.4%, P < 0.001). A survival analysis according to immune type showed that the overall survival of patients with immune type I tumors was significantly more favorable compared with that of patients with immune type III [hazards ratio (HR) 1.20; 95% confidence interval (CI), 1.07–1.35; P = 0.002; Supplementary Fig. S3a). The poor prognostic impact of immune type III compared with that of immune type I was most significant in bladder urothelial carcinoma (BLCA), in which PD-L1 expression and TIL are regarded as a predictive factor for the response to anti-PD-1/PD-L1 therapy (ref. 22; HR 1.98; 95% CI, 1.16–3.39; P = 0.012; Supplementary Fig. S3b). Although the overall prognosis of TMIT II and IV across all cancer types was not significant, a subgroup analysis showed that SKCM patients with immune type II and IV tumors had a significantly poorer prognosis compared with those with immune type I tumors (immune type II vs. type I; HR 2.04; 95% CI, 1.48–2.82; P < 0.001, immune type IV vs. type I; HR 1.58, 95% CI, 1.05–2.38; P = 0.028; Supplementary Fig. S4).

High mutational burden is associated with TMIT I

Because previous reports have shown that the degree of mutational burden and the presence of neoantigen are correlated with the immunogenic features of the tumor and reliably predict a good response to an anti-PD-1/PD-L1 treatment strategy (10,12–14), we compared the proportion of different TMIT groups according to the mutational burden. According to cancer type, we observed a tendency toward a correlation between the total number of somatic mutations (hereby, number of mutations) and the number of neoantigens and the proportion of TMIT I (Fig. 1c; Supplementary Fig. S5). Interestingly, TMIT I tumors had a significantly higher number of mutations and a higher number of neoantigens compared with the other TMIT groups (P < 0.001, Fig. 2a). Tumor samples with a higher number of mutations than the median value (46) had a significantly higher proportion of TMIT I compared with those with fewer mutations (40.3% vs. 30.4%; P < 0.001; Fig. 2b).

Figure 2.

High-mutational burden is associated with tumor microenvironment immune type (TMIT) I. Box plot of log 2-transformed values of the number of total somatic mutations (first, third, fifth, and seventh columns, the color scheme is as follows: I, red; II, blue; III, green; IV, yellow) and neoantigens (second, fourth, sixth, and eighth columns) according to TMIT are plotted (A). Proportion of TMIT according to a number of total mutations higher or lower than the median is compared (B). Box plot of log 2-transformed value of the number of mutations according to MSI status is plotted (C). The proportion of TMIT according to MSI status is compared (D). Total MT, total number of somatic mutations; NeoAg, number of neoantigens; MSI-H, microsatellite instability-high; MSI-L, microsatellite instability-low.

Figure 2.

High-mutational burden is associated with tumor microenvironment immune type (TMIT) I. Box plot of log 2-transformed values of the number of total somatic mutations (first, third, fifth, and seventh columns, the color scheme is as follows: I, red; II, blue; III, green; IV, yellow) and neoantigens (second, fourth, sixth, and eighth columns) according to TMIT are plotted (A). Proportion of TMIT according to a number of total mutations higher or lower than the median is compared (B). Box plot of log 2-transformed value of the number of mutations according to MSI status is plotted (C). The proportion of TMIT according to MSI status is compared (D). Total MT, total number of somatic mutations; NeoAg, number of neoantigens; MSI-H, microsatellite instability-high; MSI-L, microsatellite instability-low.

Close modal

The MSI has also been reported as a type of high mutational burden with immunogenic features (14, 23). As expected, MSI-high tumors had a significantly higher number of mutations compared with MSI-low tumors and microsatellite stable (MSS) tumors (Fig. 2c). Moreover, MSI-high tumors had a high proportion of TMIT I compared MSI-low tumors and MSS tumors (52.2% versus 25.0% and 29.9%, respectively, P < 0.001; Fig. 2d). This association was still clearly observed in the subgroup of COAD and STAD (Supplementary Fig. S6). Because recent studies showed that the POLE mutation is associated with defects in DNA proofreading and an enhanced immune response (24), we also determined whether the POLE mutation is associated with TMIT I. Among samples with different mutational profiles (N = 5,830), the POLE mutation was detected in 112 (1.9%) samples, which had a significantly increased number of mutations compared with the number of mutations found in the samples without the POLE mutation (Supplementary Fig. S7). Interestingly, the proportion of TMIT I samples was higher in the tumor samples with the POLE mutation than in the wild-type samples (46.4% vs. 34.8%, P = 0.021; Supplementary Fig. S7).

Because the mutational burden is significantly associated with TMIT I, we then assessed the association of TMIT I with somatic mutations in 373 genes that are frequently mutated in many cancers (21) and the PD-L1 (CD274) gene (Table 1 and Supplementary Table S3a and Supplementary Fig. S8). Other than the POLE mutation, we found that VHL, PBRM1, CASP8, and ATM mutations are significantly associated with TMIT I. In contrast, EGFR and BRAF mutations were found to be correlated with TMIT III.

Table 1.

Specific genomic alterations of cancer driver genes to predict tumor microenvironment immune type (TMIT)

GeneGenomic alterationOdds ratioP valueFDR
VHL Mutation 3.37 2.21 × 10−20 4.15 × 10−18 
CASP8 Mutation 3.13 2.56 × 10−8 8.81 × 10−7 
PBRM1 Mutation 2.40 6.59 × 10−12 8.23 × 10−10 
NCOR1 Mutation 2.37 2.58 × 10−8 8.81 × 10−7 
MXRA5 Mutation 2.13 5.93 × 10−10 4.44 × 10−8 
FAT1 Mutation 1.99 1.44 × 10−8 6.00 × 10−7 
ANK3 Mutation 1.95 1.20 × 10−8 5.64 × 10−7 
MUC17 Mutation 1.94 1.90 × 10−11 1.78 × 10−9 
BRAF Mutation 1.70 5.50 × 10−9 3.44 × 10−7 
FLG Mutation 1.69 7.05 × 10−9 3.78 × 10−7 
CD274 Amplification 3.46 2.05 × 10−14 7.43 × 10−12 
APC Amplification 3.33 6.10 × 10−9 3.68 × 10−7 
DIAPH1 Amplification 3.09 1.16 × 10−9 1.40 × 10−7 
CDX1 Amplification 2.73 1.69 × 10−8 8.75 × 10−7 
NPM1 Amplification 2.54 2.27 × 10−9 1.65 × 10−7 
PIGZ Amplification 1.67 4.94 × 10−10 8.94 × 10−8 
MUC4 Amplification 1.64 2.03 × 10−9 1.65 × 10−7 
ACVR2B Deletion 4.50 1.54 × 10−11 5.57 × 10−9 
MYD88 Deletion 4.32 3.56 × 10−11 5.58 × 10−9 
TGFBR2 Deletion 3.42 4.64 × 10−11 5.58 × 10−9 
RPSA Deletion 4.26 1.52 × 10−10 1.31 × 10−8 
SLC22A14 Deletion 3.92 1.81 × 10−10 1.31 × 10−8 
EAF1 Deletion 4.10 6.79 × 10−10 3.50 × 10−8 
MRPS25 Deletion 4.19 6.45 × 10−10 3.50 × 10−8 
CTNNB1 Deletion 3.50 7.86 × 10−10 3.55 × 10−8 
ZNF620 Deletion 3.78 1.55 × 10−9 6.21 × 10−8 
GeneGenomic alterationOdds ratioP valueFDR
VHL Mutation 3.37 2.21 × 10−20 4.15 × 10−18 
CASP8 Mutation 3.13 2.56 × 10−8 8.81 × 10−7 
PBRM1 Mutation 2.40 6.59 × 10−12 8.23 × 10−10 
NCOR1 Mutation 2.37 2.58 × 10−8 8.81 × 10−7 
MXRA5 Mutation 2.13 5.93 × 10−10 4.44 × 10−8 
FAT1 Mutation 1.99 1.44 × 10−8 6.00 × 10−7 
ANK3 Mutation 1.95 1.20 × 10−8 5.64 × 10−7 
MUC17 Mutation 1.94 1.90 × 10−11 1.78 × 10−9 
BRAF Mutation 1.70 5.50 × 10−9 3.44 × 10−7 
FLG Mutation 1.69 7.05 × 10−9 3.78 × 10−7 
CD274 Amplification 3.46 2.05 × 10−14 7.43 × 10−12 
APC Amplification 3.33 6.10 × 10−9 3.68 × 10−7 
DIAPH1 Amplification 3.09 1.16 × 10−9 1.40 × 10−7 
CDX1 Amplification 2.73 1.69 × 10−8 8.75 × 10−7 
NPM1 Amplification 2.54 2.27 × 10−9 1.65 × 10−7 
PIGZ Amplification 1.67 4.94 × 10−10 8.94 × 10−8 
MUC4 Amplification 1.64 2.03 × 10−9 1.65 × 10−7 
ACVR2B Deletion 4.50 1.54 × 10−11 5.57 × 10−9 
MYD88 Deletion 4.32 3.56 × 10−11 5.58 × 10−9 
TGFBR2 Deletion 3.42 4.64 × 10−11 5.58 × 10−9 
RPSA Deletion 4.26 1.52 × 10−10 1.31 × 10−8 
SLC22A14 Deletion 3.92 1.81 × 10−10 1.31 × 10−8 
EAF1 Deletion 4.10 6.79 × 10−10 3.50 × 10−8 
MRPS25 Deletion 4.19 6.45 × 10−10 3.50 × 10−8 
CTNNB1 Deletion 3.50 7.86 × 10−10 3.55 × 10−8 
ZNF620 Deletion 3.78 1.55 × 10−9 6.21 × 10−8 

Please see Supplementary Table S3 for raw data of all significance genomic alterations of genes.

Odds ratio was calculated that the proportion of TMIT I in each genomic alteration-positive divided by those in the genomic alteration-negative, by logistic regression.

Filtering condition of this table is odds ratio > 1.5 and FDR < 1.00 × 10−7.

FDR, false discovery rate.

PD-L1 amplification is associated with TMIT I

The association of PD-L1 amplification and TMIT was analyzed because PD-L1 amplification has been reported to serve as a good predictive marker of the response to anti-PD-1/PD-L1 therapy in Hodgkin's lymphoma (4, 25) and has been found to be associated with high immune cytolytic activity (10). The frequency of PD-L1 amplification in all cancers was 1.6% (149 of 9,364 samples), and HNSC (4.8%), sarcoma (4.7%), ovarian serous cystadenocarcinoma (4.3%), and DLBC (4.2%) had a relatively high frequency of PD-L1 amplification (Fig. 3a). Compared with the wild-type tumors, a high proportion of tumors with PD-L1 amplification were TMIT I (64.4% vs. 34.4%, P < 0.001; Fig. 3b and c).

Figure 3.

PD-L1 amplification is associated with tumor microenvironment immune type (TMIT) I. The frequency of PD-L1 amplification according to cancer type is shown (A). A scatter plot of log 2-transformed values of RPKM of PD-L1 and CD8A according to PD-L1 amplification is shown (B). The proportion of TMIT according to PD-L1 amplification is compared (C).

Figure 3.

PD-L1 amplification is associated with tumor microenvironment immune type (TMIT) I. The frequency of PD-L1 amplification according to cancer type is shown (A). A scatter plot of log 2-transformed values of RPKM of PD-L1 and CD8A according to PD-L1 amplification is shown (B). The proportion of TMIT according to PD-L1 amplification is compared (C).

Close modal

In addition to PD-L1 amplification, the analysis of 373 oncogenic genes in cancer revealed that APC, NPM1, and CDX1 amplifications as well as CTNNB1 deletion are associated with TMIT I. Moreover, EGFR amplification and CDKN2A deletion were found to be associated with TMIT III (Table 1 and Supplementary Table S3b and S3c and Supplementary Fig. S9).

Oncogenic virus infection is associated with TMIT I

We then aimed to determine whether there is association between infection with an oncogenic virus and TMIT. Considerable proportions of EBV in STAD, HPV in CESC and HNSC, and HBV in liver hepatocellular carcinoma were detected (LIHC; Supplementary Fig. S10), which is consistent with previous reports (10, 26). A high proportion of tumors resulting from EBV infection and HPV infection were classified as TMIT I compared with those resulting from HBV infection or those with no virus (84% and 56.4% vs. 18.5% and 35.9%, respectively, P < 0.001; Fig. 4a and b). This trend was also significant in EBV-positive and EBV-negative STAD (Supplementary Fig. S11a) and HPV-positive and HPV-negative HNSC (Supplementary Fig. S11b), although HBV positivity did not influence the proportion of TMIT tumors in LIHC (Supplementary Fig. S11c).

Figure 4.

Oncogenic viral infection with increased cytolytic activity is associated with tumor microenvironment immune type (TMIT) I. A scatter plot of log 2-transformed values of RPKM of PD-L1 and CD8A according to virus detection and PD-L1 amplification is shown (A). The proportion of TMIT according to PD-L1 amplification is compared (B). Box plots of log 2-transformed values of RPKM of IFNG (C) and cytolytic activity (D) according to TMITs are shown. A box plot of cytolytic activity according to virus detection and TMIT is shown (E).

Figure 4.

Oncogenic viral infection with increased cytolytic activity is associated with tumor microenvironment immune type (TMIT) I. A scatter plot of log 2-transformed values of RPKM of PD-L1 and CD8A according to virus detection and PD-L1 amplification is shown (A). The proportion of TMIT according to PD-L1 amplification is compared (B). Box plots of log 2-transformed values of RPKM of IFNG (C) and cytolytic activity (D) according to TMITs are shown. A box plot of cytolytic activity according to virus detection and TMIT is shown (E).

Close modal

IFNG expression and cytolytic activity were significantly higher in TMIT I tumors (Supplementary Figs. S5c and S5d), and cytolytic activity was significantly higher in EBV-infected and HPV-infected tumors compared with HBV-infected and uninfected samples (Supplementary Fig. S5e).

Summary of clinicopathologic features correlated with TMIT I

Taken together, the multivariate analysis results show that a high number of mutations, EBV infection, HPV infection, and PD-L1 amplification are independently associated with TMIT I (Table 2).

Table 2.

Logistic regression analysis for predicting tumor microenvironment immune type I according to clinicopathological characteristics

UnivariateMultivariate
OR (95% CI)P valueOR (95% CI)P value
Age ≥Median 1.24 (1.14–1.35) 5.78 × 10−7 1.11 (0.96–1.28) 0.147 
Gender Men (vs. women) 0.97 (0.89–1.06) 0.493 Not entered Not entered 
Number of mutations ≥Median 1.55 (1.40–1.72) <2.00 × 10−16 1.44 (1.25–1.67) 6.21 × 10−7 
Presence of virus EBV 8.14 (3.07–28.0) 1.32 × 10−4 6.29 (2.34–21.8) 9.01 × 10−4 
 HPV 2.31 (1.75–3.07) 5.05 × 10−9 1.98 (1.47–2.67) 8.31 × 10−6 
 HBV 0.19 (0.07–0.42) 1.54 × 10−4 0.18 (0.07–0.40) 1.08 × 10−4 
PD-L1 amplification Yes 3.46 (2.50–4.87) 6.47 × 10−13 3.45 (1.96–6.33) 3.10 × 10−5 
UnivariateMultivariate
OR (95% CI)P valueOR (95% CI)P value
Age ≥Median 1.24 (1.14–1.35) 5.78 × 10−7 1.11 (0.96–1.28) 0.147 
Gender Men (vs. women) 0.97 (0.89–1.06) 0.493 Not entered Not entered 
Number of mutations ≥Median 1.55 (1.40–1.72) <2.00 × 10−16 1.44 (1.25–1.67) 6.21 × 10−7 
Presence of virus EBV 8.14 (3.07–28.0) 1.32 × 10−4 6.29 (2.34–21.8) 9.01 × 10−4 
 HPV 2.31 (1.75–3.07) 5.05 × 10−9 1.98 (1.47–2.67) 8.31 × 10−6 
 HBV 0.19 (0.07–0.42) 1.54 × 10−4 0.18 (0.07–0.40) 1.08 × 10−4 
PD-L1 amplification Yes 3.46 (2.50–4.87) 6.47 × 10−13 3.45 (1.96–6.33) 3.10 × 10−5 

OR, odds ratio.

Using a large-scale TCGA pan-cancer dataset, we classified all types of cancer into one of four TMITs based on their PD-L1 and CD8A mRNA expression levels assessed by RNAseq. TMIT varies across cancer type, and this result is likely accompanied by the prevalence of somatic mutations across the cancer types. TMIT I, defined by high PD-L1 and high CD8A expression, is associated with a high mutational burden/neoantigen, high MSI, PD-L1 amplification, and infection with an oncogenic virus such as EBV and HPV. Our results suggest that PD-L1 positivity should be comprehensively interpreted with TME and that classification into four TMITs may be an appropriate model through which to tailor cancer immunotherapeutic modules (Supplementary Fig. S12).

The clinical effects of anti-PD-1/PD-L1 therapies have been modest except in a subset of cancer types, including malignant melanoma, lung cancer, and kidney cancer (2, 3, 27). The underlying biology of such limitations has not been clearly understood until recent studies, which showed that the mutational burden correlates with immunogenic features and favorable clinical outcomes of anti-PD-1/PD-L1 therapies (8, 12–14). Consistent with previous observations, the proportion of TMIT I tumors, which reflects the adaptive immune response (15), was higher in lung cancer and melanoma, as well as head and neck cancer, stomach cancer, and bladder cancer, which are expected to have a favorable clinical outcome in response to anti-PD-1/PD-L1 therapies (5, 28, 29).

The assessment of the tumor mutational burden has indicated that the mutational burden varies between studies, and the prediction of neoantigens by calculating the interaction between a specific mutation and HLA genotype would be theoretically appropriate (10, 12, 30). However, the number of nonsynonymous mutations is also clearly correlated with the mutational burden and the clinical outcome of anti-PD-1 antibody treatment (8, 13). In this study, the number of predicted neoantigens, as referenced by Rooney and colleagues (10), was found to be significantly correlated with the total number of somatic mutations assessed by whole-exome sequencing. Considering that the prediction of neoantigens is only performed in silico (31, 32), calculating the number of somatic mutations would be a more convenient method for assessing the mutational burden because the total number of mutations is also significantly correlated with the number of nonsynonymous mutations (data not shown). The number of mutations is also correlated with the MSI status as well as POLE mutation, which was found to be correlated with TMIT I in this study. This finding is consistent with previous results, which showed that anti-PD-1 antibody exhibits a good clinical outcome in MSI-high tumors (14) and TIL recruitment in POLE-mutated tumors (24).

Consistent with a previous report that showed that oncogenic virus infection increases the cytolytic activity of a tumor (10), a high proportion of tumors associated with EBV and HPV are TMIT I. Viral infection generates various viral antigens inside the tumor, and this phenomenon increases the immunogenicity of the tumor by activating the IFNG pathway (33), as supported by recent findings of the association of PD-L1 with EBV-associated malignancies (34, 35). Moreover, PD-L1 amplification is also clearly correlated with TMIT I. Previous results have clearly shown that Hodgkin's lymphoma presents variable copy number gains of chromosome 9p24.1, a genomic region that includes PD-L1, PDCD1LG2 (encoding PD-L2, another ligand of PD-1), and JAK2, which activates the JAK/STAT/IFNG pathway (25, 36). This finding is consistent with our analysis, which showed a frequent co-amplification of PD-L1 and JAK2 in the TCGA pan-cancer data (data not shown). These findings are also clearly supported by a recent finding of anti-PD-1 antibody in Hodgkin's lymphoma, which is known to be associated with both EBV and PD-L1 amplification (4).

Along with the investigation of PD-L1 expression as a predictive marker of the response to anti-PD-1/PD-L1 therapies (5, 6), the prognostic impact of PD-L1 has also been actively studied (37, 38). In addition to the fact that the optimal cutoff of PD-L1 has not been defined, the prognostic significance of PD-L1 has been reported to differ according to cancer type. The results from this study show that even in patients with high PD-L1, the overall prognosis differed according to CD8A expression (e.g., TMIT I vs. TMIT III), which would account for the previously published findings of an inconsistent prognostic significance of PD-L1. Therefore, the prognostic impact of PD-L1 expression, along with CD8+ TIL and the underlying etiology of high PD-L1 expression, such as high mutational burden and viral infection that could recruit immunogenicity, should be assessed.

In this study, the proportion of TMIT I tumors was found to be correlated with the number of mutations across cancer type and viral infection status, but considerable discrepancies also exist. KIRC is known as a highly immunogenic tumor type (39), and the results from this study show a high proportion of TMIT I. However, this tumor has a relatively low mutational burden. Interestingly, VHL and PBRM1 mutations, which are frequently shown in KIRC (40), were clearly observed to be correlated with TMIT I. Moreover, hepatocellular carcinoma has a low proportion of TMIT I tumors, although it has a relatively high mutational burden and an association with HBV. These findings are likely the result of the unique anatomical characteristics of immune privilege, such as in glioblastoma (41, 42).

In this study, we used RNAseq data from a mixture of cancer cells and surrounding tissues, including TILs, although this contamination of surrounding stromal tissues was not intentional. Interestingly, as interpreted by Rooney and colleagues (10), this limitation would actually be advantageous to assessing the infiltration of surrounding tissues, such as the TIL of CD8+ CTLs. In this regard, the assessment of CD8A gene expression in a mixture of cancer and stromal cells would be more practical in the clinical setting than the immunohistochemical assessment of CD8+ CTL recruitment or activity, which would be difficult to judge uniformly across different tumor types. Nevertheless, the clinical validation of this approach is definitely warranted.

Although this study has some limitations, including that the cutoff values of PD-L1 and CD8A need clinical validation, it is nevertheless valuable because we systematically assessed the TMIT of most cancer types by using data from TCGA project. Our findings are clearly consistent with previous findings. Future investigations and clinical validations regarding the use of this approach for the assessment of immuno-genomic features across cancer types are warranted.

In conclusion, from the pan-cancer immuno-genomic perspective, the classification of tumors into four TMITs based on PD-L1 status and CD8A+ TIL is an appropriate approach for cancer immunotherapy. TMIT I (PD-L1+/CD8A+) was found to be associated with high PD-L1 expression, high mutational burden/neoantigen, high MSI, PD-L1 amplification, and the presence of an oncogenic virus. These factors are likely good predictive factors for the response to anti-PD-1/PD-L1 therapy. Our findings will open up opportunities for finding new anti-PD-1/PD-L1 therapy strategies that can identify a subset of patients who may receive a greater benefit from anticancer immunotherapy.

No potential conflicts of interest were disclosed.

Conception and design: C.-Y. Ock, B. Keam, D.H. Chung, D.S. Heo

Development of methodology: C.-Y. Ock, B. Keam

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): C.-Y. Ock, B. Keam, S. Kim, D.-W. Kim

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): C.-Y. Ock, B. Keam, J.-S. Lee, M. Kim, T.M. Kim, Y.K. Jeon, D.S. Heo

Writing, review, and/or revision of the manuscript: C.-Y. Ock, B. Keam, J.-S. Lee, T.M. Kim, Y.K. Jeon, D.H. Chung, D.S. Heo

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C.-Y. Ock, B. Keam, D.H. Chung

Study supervision: C.-Y. Ock, B. Keam, D.H. Chung, D.S. Heo

The authors thank the patients and their families, who generously donated their tissues to TCGA and the members of TCGA who collected and disclosed valuable data. The authors also thank Rooney and colleagues, who generously generated the valuable data regarding neoantigen and virus detection in TCGA samples.

This study was supported by the SNUH Research Fund (Grant No. 03-2015-0380). This study was supported by grants from the Innovative Research Institute for Cell Therapy, Republic of Korea (A062260).

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.
Hodi
FS
,
O'Day
SJ
,
McDermott
DF
,
Weber
RW
,
Sosman
JA
,
Haanen
JB
, et al
Improved survival with ipilimumab in patients with metastatic melanoma
.
N Engl J Med
2010
;
363
:
711
23
.
2.
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
.
3.
Hamid
O
,
Robert
C
,
Daud
A
,
Hodi
FS
,
Hwu
WJ
,
Kefford
R
, et al
Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma
.
N Engl J Med
2013
;
369
:
134
44
.
4.
Ansell
SM
,
Lesokhin
AM
,
Borrello
I
,
Halwani
A
,
Scott
EC
,
Gutierrez
M
, et al
PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma
.
N Engl J Med
2015
;
372
:
311
9
.
5.
Garon
EB
,
Rizvi
NA
,
Hui
R
,
Leighl
N
,
Balmanoukian
AS
,
Eder
JP
, et al
Pembrolizumab for the treatment of non-small-cell lung cancer
.
N Engl J Med
2015
;
372
:
2018
28
.
6.
Herbst
RS
,
Soria
JC
,
Kowanetz
M
,
Fine
GD
,
Hamid
O
,
Gordon
MS
, et al
Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients
.
Nature
2014
;
515
:
563
7
.
7.
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
.
8.
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
.
9.
Rutledge
WC
,
Kong
J
,
Gao
J
,
Gutman
DA
,
Cooper
LA
,
Appin
C
, et al
Tumor-infiltrating lymphocytes in glioblastoma are associated with specific genomic alterations and related to transcriptional class
.
Clin Cancer Res
2013
;
19
:
4951
60
.
10.
Rooney
MS
,
Shukla
SA
,
Wu
CJ
,
Getz
G
,
Hacohen
N
. 
Molecular and genetic properties of tumors associated with local immune cytolytic activity
.
Cell
2015
;
160
:
48
61
.
11.
Brown
SD
,
Warren
RL
,
Gibb
EA
,
Martin
SD
,
Spinelli
JJ
,
Nelson
BH
, et al
Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival
.
Genome Res
2014
;
24
:
743
50
.
12.
Schumacher
TN
,
Schreiber
RD
. 
Neoantigens in cancer immunotherapy
.
Science
2015
;
348
:
69
74
.
13.
Rizvi
NA
,
Hellmann
MD
,
Snyder
A
,
Kvistborg
P
,
Makarov
V
,
Havel
JJ
, et al
Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer
.
Science
2015
;
348
:
124
8
.
14.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
15.
Teng
MW
,
Ngiow
SF
,
Ribas
A
,
Smyth
MJ
. 
Classifying cancers based on T-cell infiltration and PD-L1
.
Cancer Res
2015
;
75
:
2139
45
.
16.
Lee
JC
,
Lee
KM
,
Kim
DW
,
Heo
DS
. 
Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients
.
J Immunol
2004
;
172
:
7335
40
.
17.
Tumeh
PC
,
Harview
CL
,
Yearley
JH
,
Shintaku
IP
,
Taylor
EJ
,
Robert
L
, et al
PD-1 blockade induces responses by inhibiting adaptive immune resistance
.
Nature
2014
;
515
:
568
71
.
18.
Gajewski
TF
,
Schreiber
H
,
Fu
YX
. 
Innate and adaptive immune cells in the tumor microenvironment
.
Nat Immunol
2013
;
14
:
1014
22
.
19.
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
.
20.
Mermel
CH
,
Schumacher
SE
,
Hill
B
,
Meyerson
ML
,
Beroukhim
R
,
Getz
G
. 
GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers
.
Genome Biol
2011
;
12
:
R41
.
21.
Lawrence
MS
,
Stojanov
P
,
Polak
P
,
Kryukov
GV
,
Cibulskis
K
,
Sivachenko
A
, et al
Mutational heterogeneity in cancer and the search for new cancer-associated genes
.
Nature
2013
;
499
:
214
8
.
22.
Powles
T
,
Eder
JP
,
Fine
GD
,
Braiteh
FS
,
Loriot
Y
,
Cruz
C
, et al
MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer
.
Nature
2014
;
515
:
558
62
.
23.
Kloor
M
,
Michel
S
,
von Knebel Doeberitz
M
. 
Immune evasion of microsatellite unstable colorectal cancers
.
Int J Cancer
2010
;
127
:
1001
10
.
24.
van Gool
IC
,
Eggink
FA
,
Freeman-Mills
L
,
Stelloo
E
,
Marchi
E
,
de Bruyn
M
, et al
POLE Proofreading mutations elicit an antitumor immune response in endometrial cancer
.
Clin Cancer Res
2015
;
21
:
3347
55
.
25.
Green
MR
,
Monti
S
,
Rodig
SJ
,
Juszczynski
P
,
Currie
T
,
O'Donnell
E
, et al
Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma
.
Blood
2010
;
116
:
3268
77
.
26.
Tang
KW
,
Alaei-Mahabadi
B
,
Samuelsson
T
,
Lindh
M
,
Larsson
E
. 
The landscape of viral expression and host gene fusion and adaptation in human cancer
.
Nature Commun
2013
;
4
:
2513
.
27.
Rosenberg
SA
. 
Progress in human tumour immunology and immunotherapy
.
Nature
2001
;
411
:
380
4
.
28.
Robert
C
,
Schachter
J
,
Long
GV
,
Arance
A
,
Grob
JJ
,
Mortier
L
, et al
Pembrolizumab versus Ipilimumab in advanced melanoma
.
N Engl J Med
2015
;
372
:
2521
32
.
29.
Brahmer
J
,
Reckamp
KL
,
Baas
P
,
Crino
L
,
Eberhardt
WE
,
Poddubskaya
E
, et al
Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer
.
N Engl J Med
2015
;
373
:
123
35
.
30.
Gubin
MM
,
Zhang
X
,
Schuster
H
,
Caron
E
,
Ward
JP
,
Noguchi
T
, et al
Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens
.
Nature
2014
;
515
:
577
81
.
31.
Nielsen
M
,
Lundegaard
C
,
Blicher
T
,
Lamberth
K
,
Harndahl
M
,
Justesen
S
, et al
NetMHCpan, a method for quantitative predictions of peptide binding to any HLA-A and -B locus protein of known sequence
.
PloS One
2007
;
2
:
e796
.
32.
Rajasagi
M
,
Shukla
SA
,
Fritsch
EF
,
Keskin
DB
,
DeLuca
D
,
Carmona
E
, et al
Systematic identification of personal tumor-specific neoantigens in chronic lymphocytic leukemia
.
Blood
2014
;
124
:
453
62
.
33.
Dong
H
,
Chen
X
. 
Immunoregulatory role of B7-H1 in chronicity of inflammatory responses
.
Cell Mol Immunol
2006
;
3
:
179
87
.
34.
Chen
BJ
,
Chapuy
B
,
Ouyang
J
,
Sun
HH
,
Roemer
MG
,
Xu
ML
, et al
PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies
.
Clin Cancer Res
2013
;
19
:
3462
73
.
35.
Fang
W
,
Zhang
J
,
Hong
S
,
Zhan
J
,
Chen
N
,
Qin
T
, et al
EBV-driven LMP1 and IFN-gamma up-regulate PD-L1 in nasopharyngeal carcinoma: implications for oncotargeted therapy
.
Oncotarget
2014
;
5
:
12189
202
.
36.
Marzec
M
,
Zhang
Q
,
Goradia
A
,
Raghunath
PN
,
Liu
X
,
Paessler
M
, et al
Oncogenic kinase NPM/ALK induces through STAT3 expression of immunosuppressive protein CD274 (PD-L1, B7-H1)
.
Proc Natl Acad Sci U S A
2008
;
105
:
20852
7
.
37.
Zhang
Y
,
Kang
S
,
Shen
J
,
He
J
,
Jiang
L
,
Wang
W
, et al
Prognostic significance of programmed cell death 1 (PD-1) or PD-1 ligand 1 (PD-L1) expression in epithelial-originated cancer: a meta-analysis
.
Medicine
2015
;
94
:
e515
.
38.
Wang
A
,
Wang
HY
,
Liu
Y
,
Zhao
MC
,
Zhang
HJ
,
Lu
ZY
, et al
The prognostic value of PD-L1 expression for non-small cell lung cancer patients: a meta-analysis
.
Eur J Surg Oncol
2015
;
41
:
450
6
.
39.
Klapper
JA
,
Downey
SG
,
Smith
FO
,
Yang
JC
,
Hughes
MS
,
Kammula
US
, et al
High-dose interleukin-2 for the treatment of metastatic renal cell carcinoma: a retrospective analysis of response and survival in patients treated in the surgery branch at the National Cancer Institute between 1986 and 2006
.
Cancer
2008
;
113
:
293
301
.
40.
Cancer Genome Atlas Research
N
. 
Comprehensive molecular characterization of clear cell renal cell carcinoma
.
Nature
2013
;
499
:
43
9
.
41.
Perng
P
,
Lim
M
. 
Immunosuppressive mechanisms of malignant gliomas: parallels at non-CNS sites
.
Front Oncol
2015
;
5
:
153
.
42.
Sprinzl
MF
,
Galle
PR
. 
Immune control in hepatocellular carcinoma development and progression: role of stromal cells
.
Sem Liver Dis
2014
;
34
:
376
88
.

Supplementary data