Purpose: Determine the localized expression pattern and clinical significance of VISTA/PD-1H in human non–small cell lung cancer (NSCLC).

Experimental Design: Using multiplex quantitative immunofluorescence (QIF), we performed localized measurements of VISTA, PD-1, and PD-L1 protein in 758 stage I–IV NSCLCs from 3 independent cohorts represented in tissue microarray format. The targets were selectively measured in cytokeratin+ tumor epithelial cells, CD3+ T cells, CD4+ T-helper cells, CD8+ cytotoxic T cells, CD20+ B lymphocytes and CD68+ tumor-associated macrophages. We determined the association between the targets, clinicopathological/molecular variables and survival. Genomic analyses of lung cancer cases from TCGA were also performed.

Results: VISTA protein was detected in 99% of NSCLCs with a predominant membranous/cytoplasmic staining pattern. Expression in tumor and stromal cells was seen in 21% and 98% of cases, respectively. The levels of VISTA were positively associated with PD-L1, PD-1, CD8+ T cells and CD68+ macrophages. VISTA expression was higher in T-lymphocytes than in macrophages; and in cytotoxic T cells than in T-helper cells. Elevated VISTA was associated with absence of EGFR mutations and lower mutational burden in lung adenocarcinomas. Presence of VISTA in tumor compartment predicted longer 5-year survival.

Conclusions: VISTA is frequently expressed in human NSCLC and shows association with increased tumor-infiltrating lymphocytes, PD-1 axis markers, specific genomic alterations and outcome. These results support the immunomodulatory role of VISTA in human NSCLC and suggests its potential as therapeutic target. Clin Cancer Res; 24(7); 1562–73. ©2017 AACR.

Translational Relevance

VISTA/PD-1H participate in immune regulation and clinical studies are under way to assess the antitumor effect of VISTA blockade. However, the biological impact of VISTA expression in human malignancies remains largely unexplored. Our results demonstrate that VISTA is frequently expressed in human non–small cell lung cancer (NSCLC) and shows differential distribution in tumor and immune cells. Elevated VISTA in NSCLC is associated with increased PD-1 axis markers, effector T cells and CD68+ macrophages, supporting its modulation by local proinflammatory responses. VISTA protein levels are associated with specific genomic alterations in lung adenocarcinomas and its expression in tumor cells predicts longer survival in NSCLC patients. Taken together, our findings support the immunomodulatory role of VISTA in human NSCLC and suggests its potential as therapeutic target.

The antitumor immune response is essential to control tumor growth and progression. Blockade of immune coregulatory signals in the tumor immune microenvironment such as the PD-1/PD-L1 axis has revolutionized the treatment of diverse tumor types (1–3). Notably, expression of PD-L1 protein or metrics of tumor immune infiltration in pretreatment samples can be used to predict sensitivity/resistance to treatment (1–6). Therefore, careful evaluation of the tumor immune landscape could provide essential information to understand the role of the immune system during tumor progression and to define the optimal treatment modalities using both immune and nonimmune therapies. To date, most studies characterizing multiple cell/targets in the tumor immune microenvironment have used nonhuman model systems and/or methods requiring tissue grinding such as flow cytometry/CyTOF and mRNA profiling. This has limited the understanding of the spatial context and evaluation of key cell/molecular interactions in human malignancies. For instance, multiple potentially actionable immune-modulatory proteins, such as CTLA-4, PD-1, PD-L1, LAG-3, TIM-3, and other can be coexpressed on immune cells and functionally interact with each other to modulate the response against tumors (7–9).

Due to its clinical relevance, the PD-L1/PD-1 pathway has been extensively studied in human malignancies. Engagement of PD-1 receptor by its ligand PD-L1 (and also PD-L2) mediates inhibitory signals in T cells and antiapoptotic signals in tumor cells (10–13). In human neoplasms, PD-L1 is predominantly expressed in malignant tumor cells and macrophages in response to IFNγ stimulation or as a result of constitutive oncogenic signaling. Elevated levels of PD-L1 are found with variable frequency in human malignancies and blockade of this pathway produces prominent and lasting antitumor responses in a proportion of patients with cancer. This supports a fundamental role of the PD-1 pathway in the antitumor immune evasion (2, 3, 10–12). PD-L1 (also known as B7-H1 or CD274) belongs to the B7 family of immune-regulatory molecules. Since its initial description, additional members of this family have been reported containing variable degree of sequence homology including PD-L2/B7-DC, B7-H2/ICOS-L, B7-H3, B7-H4, B7-H5, VISTA/PD-1H, and B7-H6 (13). To date, little is known about the expression, biological role, and therapeutic potential of these targets in human neoplasms.

Human VISTA is a 311–amino acid long Ig domain-containing type I transmembrane protein able to suppress T-cell activation in vitro and in vivo. Notably, this protein shares sequence homology with both PD-1 and PD-L1 (14, 15) and can act as a ligand on antigen presenting cells and as receptor in T lymphocytes (14–19).

Previous studies have shown high VISTA expression in CD11b+ myeloid cells and lower levels on CD4+ and CD8+ T cells and CD4+/Foxp3+ T-regulatory lymphocytes (Treg; refs. 14–15, 18–19). To date, there is no clear evidence of VISTA expression in tumor cells (16, 18–20). Chronic inflammation and spontaneous activation of T cells have been reported in PD-1 or VISTA single knockout animals, and this phenotype is enhanced in double knockout mice (21). In addition, the magnitude of T-cell responses after a challenge with foreign antigens is synergistically increased in the double knockout mice compared with the single knockout animals, supporting nonredundant immune suppressive effects of VISTA and PD-1 pathway (21). Despite these functional associations, the expression pattern and possible link between PD-1, PD-L1, and VISTA expression in human tumors remains unexplored. Recent studies in melanoma mouse models show that blockade of VISTA using monoclonal antibodies increases the proportion of circulating tumor-specific T-cell, promotes tumor immune infiltration and reduces tumor growth (15). Furthermore, an additive antitumor effect with differential contribution of VISTA, CTLA-4, and PD-1 was reported in a squamous cell carcinoma model (22).

Here, we performed quantitative and spatially resolved measurements of VISTA protein and additional immune markers in 3 large cohorts of human non–small cell lung cancers (NSCLC). To further support our findings, we also performed analyses of The Cancer Genome Atlas (TCGA) lung cancer dataset. Our data demonstrate frequent expression of VISTA in human lung carcinomas and differential expression in tumor and immune cell subsets. We also show association of VISTA with local antitumor immune responses, expression of PD-1 axis markers, specific tumor genomic alterations, and survival in human lung cancer.

Patient cohorts and tissue microarrays

We included retrospectively collected formalin-fixed, paraffin-embedded (FFPE) tumor samples from 765 stage I–IV NSCLCs in three cohorts represented in tissue microarray (TMA) format. The first two cohorts included NSCLC cases with detailed clinicopathological annotation and have been previously reported (23–25). The TMAs were constructed by selecting areas containing viable tumor cells and stromal elements on hematoxylin and eosin–stained preparations (as assessed by a pathologist) and without enriching for specific tumor regions, tissue structures or immune-related features. The first collection is from Greek hospitals between 1991 and 2001 and included 329 specimens. The second cohort was from Yale and included 297 samples collected between 1988 and 2003. The clinicopathological characteristics of these cohorts are described in Supplementary Table S2. A third cohort from Yale (n = 139, Supplementary Table S3) including advanced lung adenocarcinomas clinically tested for EGFR and KRAS mutations was also studied. The final number of cases used for each analysis is different from the total number of cases in the cohorts (758 cases included) due to inevitable loss of histospots during the staining procedure or exclusion of cases after quality control. For the QIF measurements, we used two different TMA blocks from the cohorts, each containing two independent 0.6-mm tumor cores. Serial sections from each TMA were stained twice. Therefore, the results presented included integrated data from 2 to 4 independent tumor cores stained at least twice. To determine the levels of VISTA in nontumor lung tissue relative to paired NSCLC from the same cases, we also included a cohort of 46 primary resected NSCLCs (Supplementary Fig. S5). All tissues were used after approval from the Yale Human Investigation Committee protocols #9505008219 and #1608018220, which approved the patient consent forms or in some cases waiver of consent.

Quantitative immunofluorescence (QIF)

Using previously validated/standardized multiplexed QIF panels and serial TMA sections, we measured the levels of cytokeratin (clone AE1/AE3, eBioscience), VISTA (clone D1L2G, CST), PD-L1 (clone 405.9A11, CST), PD-1 (clone EH33, CST), CD3 (polyclonal, DAKO), CD4 (clone SP35, Spring Biosc.), CD8 (clone C8/144B, DAKO), CD20 (clone L26, DAKO), and CD68 (clone KP1, DAKO). The specific QIF panels used and experimental conditions are outlined in Supplementary Table S1 and were stained using a previously reported protocol (25–27). Briefly, freshly cut TMA serial sections were deparaffinized and antigen retrieval was carried on with 1 mmol/L EDTA pH8 (Sigma Aldrich) and boiled for 20 minutes at 97°C (PT module, Lab Vision, Thermo Sci.). Inactivation of endogenous peroxidase activity was carried on using a solution of 0.3% hydrogen peroxide in methanol for 20 minutes and then slides were incubated with a blocking solution containing 0.3% BSA in 0.05% Tween-20 and Tris-buffered solution for 30 minutes. Primary antibody dilution and incubation were carried on as described in Supplementary Table S1. Isotype-specific HRP-conjugated antibodies and tyramide-based amplification systems (Perkin Elmer) were used for signal detection. Residual HRP activity between sequential detection protocol was eliminated incubating the slides twice with a solution containing 100 mmol/L benzoic hydrazide and 50 mmol/L hydrogen peroxide in phosphate-buffered solution.

Fluorescence signal quantification and cases stratification

Quantitative measurement of the fluorescence signal was performed using the AQUA method of QIF, as previously reported (25–27). Each slide was visually examined to exclude samples with tissue/staining artifacts and those with less than 5% tumor content. Cases were considered to display detectable levels of each target when the QIF score was above the signal to noise threshold determined measuring negative control preparations and by visual examination of the sample. For VISTA, PD-1 and PD-L1 measurement, we considered the signal detected in the tumor compartment (cytokeratin-positive cells), stromal (cytokeratin-negative cells) compartment, and the total tissue area signal detected in the whole tumor sample (tumor and stroma). In addition, VISTA was selectively measured in immune cell subpopulations defined by its colocalization with the immune cell phenotype markers CD3, CD4, CD8, CD20, or CD68. For stratification purposes and statistical analysis, VISTA, PD-1 and PD-L1 detection were classified as high and low using the median score as cutoff point.

Cell culture and antibody validation

The VISTA antibody validation was carried out using antibody preabsorption with the recombinant protein and by quantitative immunodetection in FFPE cell line preparations containing parental cells and cells with siRNA-induced VISTA silencing (Supplementary Fig. S1). VISTA monoclonal antibody was preabsorbed with 2.5 and 5.0 μg of human recombinant VISTA protein (R&D Systems). Preabsorption was carried on overnight at 4°C in a solution of BSA 0.3% and Tris-buffered solution (0.05% Tween-20) containing a ratio 1:2 and 1:1 of antigen and antibody. Absorbed antibody was diluted to 94 ng/mL and used to detect the target by QIF in positive and negative control preparations. HPB-all, Ramos and Karpas cell lines were cultured in RPMI-1640 culture medium containing 10% FBS, 1 mmol/L glutamine and 10 IU streptomycin/ampicillin. VISTA knockdown in these was achieved using two single siRNAs (ID 261239 and ID 258598 from Ambion) and a mixed pool (SC-90756 from Santa Cruz Biotechnology). A scrambled siRNA was used as negative control (#12935-100, Invitrogen). Cells were transfected using Lipofectamin and 10 nmol of each siRNA for 24 hours, and then cultured with low FBS OPTI-MEM medium for additional 24 hours. Control and knockeddown cells were used fresh for protein extraction and immunoblotting; or fixed in 10% neutral buffered formalin for 8 to 12 hours and embedded in paraffin for VISTA quantification by QIF. The antibodies against additional targets used in this study have been previously validated by our group using comparable strategies (25–30). Cell lines used in this study were purchased in the ATCC, and authentication was performed every 3 to 6 months using the GenePrint 10 System in the Yale University DNA Analysis Facility.

TCGA data analysis for mRNA expression and genomic alterations

We analyzed the NSCLC samples from the TCGA (http://cancergenome.nih.gov/). Briefly, we downloaded the RNA-seq and DNA whole exome sequencing data from 370 NSCLC cases, including 250 adenocarcinomas and 120 squamous cell carcinomas. Then, we aligned the transcripts and performed variant calling using defaults TCGA pipelines. Then, we conducted single Scatterplot analysis between log2 transformed mRNA FPKM scores of VISTA, PD-L1, PD-1, and CD8A. The number of nonsynonymous mutations detected in the whole exome sequencing data was used as the mutational burden. Association was defined based on linear regression factor (R2) and the statistical significance using the linear correlation by GraphPad 5.0 software.

Statistical analysis

All statistical and survival studies were conducted using JMP 10.0 software for Windows. Graphs and images were prepared using GraphPad 5.0 and Photoshop 9.0 for Windows. A standard two-tailed Student t test was used for all statistical analyses. All samples sizes were appropriate for assumption of normal distribution, and variance was similar between compared groups. The statistical values of P < 0.05 was considered statistically significant. Adjustment for multiple comparisons was performed using the Bonferroni method and statistical significance was met with P = 0.002. Values of mean determinants are presented as mean ± SEM.

Validation of an anti-VISTA assay and multiplexed immunodetection

Stringent validation and optimization of a VISTA monoclonal antibody for use in formalin-fixed paraffin-embedded (FFPE) tissue samples was performed. To determine the target specificity, we measured VISTA protein levels using QIF in Ramos cell preparations with known (endogenous) VISTA expression using the antibody in control conditions or after preabsorption with recombinant human VISTA protein. As shown in Supplementary Fig. S1A, overnight incubation of the primary antibody with 2.5 μg of VISTA protein significantly reduced the target detection and incubation with 5 μg completely abolished VISTA signal. Preincubation of the antibody did not affect PD-L1 levels in Karpas cells (known to express PD-L1) ruling out a nonspecific effect of the recombinant peptide and cross reactivity of the primary antibody with PD-L1 protein (Supplementary Fig. S1B and S1E). PD-L1 was not detected in Ramos cells using both control and preabsorbed antibodies. To further assess the assay specificity, we performed VISTA measurements in Ramos cells with and without siRNA-induced VISTA silencing. As shown in Supplementary Fig. S1C, lower VISTA levels were detected in cells exposed for 48 hours to 2 different VISTA-specific siRNAs but no change was seen after transfection with a nonspecific/scrambled siRNA. As expected, the levels of PD-L1 protein were not affected by incubation with VISTA-specific or scrambled siRNA (Supplementary Fig. S1D). After the specificity assessment, the VISTA assay was titrated and optimized for use in multiplexing QIF panels with previously validated antibodies for PD-1, PD-L1, and immune cell markers. A detailed description of the antibodies, experimental conditions, fluorescence channels and panels used is shown in Supplementary Table S1. Human tonsil and placenta samples were used as positive human tissue controls and for standardization of the multiplexed QIF experiments. As shown in Supplementary Fig. S2A, VISTA was detected preferentially in the interfollicular tonsil areas and in trophoblastic cells of the placenta. Prominent PD-L1 signal was seen in germinal centers (Supplementary Fig. S2A, top) and trophoblastic cells of the chorionic villi, but not in the mesenchymal areas of the placenta (Supplementary Fig. S2A, bottom). Representative captions from the 4 different multiplexing panels described in Supplementary Table S1 stained in the NSCLC cases are shown in Supplementary Fig. S2B.

VISTA expression in NSCLC

To evaluate the expression of VISTA and its biological significance in human NSCLC, 636 cases represented in TMAs from two independent cohorts were studied (Supplementary Table S2). As shown in Fig. 1A–C, VISTA protein was detected with variable levels across NSCLCs. It was expressed predominantly in stromal cells and showed a cytoplasmic/membranous staining pattern. A fraction of cases showed a mixed pattern with simultaneous expression of VISTA in stromal cells and in cytokeratin-positive tumor epithelial cells (Fig. 1D). Some cases also displayed VISTA expression in immune cells infiltrating or abutting cytokeratin-positive tumor regions (Supplementary Fig. S3, bottom). In the quantitative analysis, VISTA protein levels displayed a continuous distribution and the scores were comparable in both studied cohorts (Fig. 1E and F). Overall, VISTA protein was detected in 99% of NSCLC cases with 19.4% and 22.8% of cases showing a mixed tumor/stromal staining pattern in cohorts #1 and #2, respectively. Notably, there was a positive association between the levels of VISTA in tumor and stromal cells [linear regression coefficient (R2) = 0.32 in cohort #1 and 0.33 in cohort #2, P < 0.001, Fig. 1G and H]. As shown in Supplementary Fig. S4, the levels of stromal VISTA were significantly higher in the tumor than in morphologically normal lung tissue form the same cases (P = 0.035). In the nontumor lung tissue, the majority of VISTA signal was located in nonepithelial cells underlying cytokeratin-positive pneumocytes with morphological features consistent with macrophages.

Figure 1.

VISTA is highly expressed in NSCLC. QIF for VISTA protein was performed. The protein expression, immunodetection pattern, and QIF scores were analyzed based on distribution and correlation between different compartments in two different retrospective NSCLC cohorts (cohort #1, N = 324 and cohort #2, N = 295). A–D, Representative immunolocalization of VISTA in NSCLC cases with high and low stromal detection are shown (A and B, respectively). Overall 80% of total NSCLC cases showed a stromal VISTA detection (C) and 20% of cases expressed VISTA on cytokeratin positive cells (D). For both studied cohorts, total number of cases were represented in a distribution curve showing the lowest and highest measured total QIF score using DAPI as mask (E and F). Correlation between stromal and tumor VISTA detection was analyzed and linear regression factor (R2) is indicated; QIF scores were measured using the respective compartment (cytokeratin negative and positive, respectively). Images are representative of 619 cases.

Figure 1.

VISTA is highly expressed in NSCLC. QIF for VISTA protein was performed. The protein expression, immunodetection pattern, and QIF scores were analyzed based on distribution and correlation between different compartments in two different retrospective NSCLC cohorts (cohort #1, N = 324 and cohort #2, N = 295). A–D, Representative immunolocalization of VISTA in NSCLC cases with high and low stromal detection are shown (A and B, respectively). Overall 80% of total NSCLC cases showed a stromal VISTA detection (C) and 20% of cases expressed VISTA on cytokeratin positive cells (D). For both studied cohorts, total number of cases were represented in a distribution curve showing the lowest and highest measured total QIF score using DAPI as mask (E and F). Correlation between stromal and tumor VISTA detection was analyzed and linear regression factor (R2) is indicated; QIF scores were measured using the respective compartment (cytokeratin negative and positive, respectively). Images are representative of 619 cases.

Close modal

To explore the possible role of VISTA in the immune microenvironment of NSCLC, we measured its levels in CD3+ tumor infiltrating T cells, CD4+ T-helper cells, CD8+ cytotoxic T lymphocytes, CD20+ B cells and CD68+ tumor-associated macrophages. As shown in Fig. 2A–C and in Supplementary Fig. S5, detectable levels of VISTA were recognized in all studied immune cell subsets but were significantly higher in CD3+ T cells than in CD68-positive macrophages or CD20+ B lymphocytes (Fig. 2A). In addition, higher levels of VISTA were found in CD8+ cytotoxic cells than in CD4+ helper T lymphocytes (Fig. 2B).

Figure 2.

VISTA is selectively expressed in immune cells subset. Simultaneous QIF for VISTA and most relevant immune cell subtype markers were performed in a single unrelated NSCLC cohort (N = 340). QIF scores for VISTA expression in a cellular subset were measured using the specific immune marker as mask. In A, VISTA is detected predominantly in T lymphocytes (CD3+ cells) and macrophages (CD68+ cells) than B lymphocytes (CD20+ cells) population. In B, CD8+ cytotoxic cells expressed more VISTA than CD4+ T regulatory lymphocytes. C, Representative images of VISTA, immune marker, and tumor cells (cytokeratin, CK+ cells) codetection are shown. ***, P = 0.001; ****, P < 0.0001.

Figure 2.

VISTA is selectively expressed in immune cells subset. Simultaneous QIF for VISTA and most relevant immune cell subtype markers were performed in a single unrelated NSCLC cohort (N = 340). QIF scores for VISTA expression in a cellular subset were measured using the specific immune marker as mask. In A, VISTA is detected predominantly in T lymphocytes (CD3+ cells) and macrophages (CD68+ cells) than B lymphocytes (CD20+ cells) population. In B, CD8+ cytotoxic cells expressed more VISTA than CD4+ T regulatory lymphocytes. C, Representative images of VISTA, immune marker, and tumor cells (cytokeratin, CK+ cells) codetection are shown. ***, P = 0.001; ****, P < 0.0001.

Close modal

To understand the relationship between VISTA expression and the immune composition of NSCLCs, we studied the association between cell-specific VISTA levels and tumor infiltration by different immune cell populations. As shown in Table 1, elevated VISTA protein measured in all cells of the sample (e.g., total score) or exclusively in CD3+ tumor-infiltrating lymphocytes (TIL) was significantly associated with prominent CD3+/CD8+ TILs, CD20+ B lymphocytes and CD68+ macrophages, but with low levels of CD4+ TILs. A similar trend was seen with VISTA in CD4, CD8, CD20, and CD68+ cells but without reaching statistical significance for all cell type categories, suggesting a weaker association. Altogether, these results indicate an association between higher levels of VISTA expression and increased tumor immune infiltration in human lung cancer.

Table 1.

VISTA is expressed in immune cells and correlated with TILs

CD8+ cells infiltrationCD4+ cells infiltrationCD3+ cells infiltrationCD20+ cells infiltrationCD68+ cells infiltration
462LowHigh462LowHigh675LowHigh503LowHigh297LowHigh
N231231PN232230PN326349PN252251PN149148P
total VISTA Low 241 142/31.7% 99/21.4% <0.0001 241 109/23.6% 132/28.6% 0.025 339 229/33.9% 97/14.4% <0.0001 242 138/27.4% 104/20.7% 0.0027 111 75 25.2% 36 12.1% <0.0001 
 High 221 89/19.3% 132/28.6%  221 123/26.6% 98/21.2%  336 110/16.3% 239/35.4%  261 114/22.7% 147/29.2%  186 74/24.9% 112/37.7%  
  462 Low High  462 Low High  414 Low High  411 Low High  95 Low High  
  231 231 232 230 212 202 207 204 52 43 
VISTA in CD8+ cells Low 231 130/28.1% 101/21.9% 0.0069 231 115/24.9% 116/25.1% 0.8524 208 122/29.5% 86/20.8% 0.002 205 110/26.8% 95/23.1% 0.1826 46 24/25.3% 22/23.2% 0.6267 
 High 231 101/21.9% 130/28.1%  231 117/25.3% 114/24.7%  206 90/21.7% 116/28.0%  206 97/23.6% 109/26.5%  49 28/29.5% 21/22.1%  
  462 Low High  462 Low High  414 Low High  411 Low High  95 Low High  
  231 231 232 230 212 202 207 204 52 43 
VISTA in CD4+ cells Low 231 137/29.6% 94/20.4% <0.0001 231 99/21.4% 132/28.6% 0.0015 205 109/26.3% 96/23.2% 0.4287 203 109/26.5% 94/22.9% 0.1821 45 25/26.3% 20/21.0% 0.8791 
 High 231 94/20.4% 137/29.6%  231 133/28.8% 98/21.2%  209 103/24.9% 106/25.6%  208 98/23.8% 110/26.8%  50 27/28.4% 23/24.2%  
  414 Low High  414 Low High  675 Low High  503 Low High  297 Low High  
  204 210 216 198 339 336 252 251 149 148 
VISTA in CD3+ cells Low 206 117/28.3% 89/21.5% 0.002 206 95/22.9% 111/26.8% 0.014 339 226/33.5% 113/16.7% <0.0001 255 147/29.2% 108/21.5% 0.0006 114 71/23.9% 43/14.5% 0.0009 
 High 208 87/21.0% 121/29.2%  208 121/29.2% 87/21.0%  336 113/16.7% 223/33.0%  248 105/20.9% 143/28.4%  183 78/26.3% 105/35.3%  
  411 Low High  411 Low High  503 Low High  503 Low High  125 Low High  
  203 208 215 196 255 248 252 251 63 62 
VISTA in CD20+ cells Low 205 117/28.5% 88/21.4% 0.0018 205 101/24.6% 104/25.3% 0.2177 252 136/27.0% 116/23.1% 0.1412 252 143/28.4% 109/21.7% 0.002 65 37/29.6% 28/22.4% 0.1284 
 High 206 86/20.9% 120/29.2%  206 114/27.7% 92/22.4%  251 119/23.7% 132/26.2%  251 109/21.7% 142/28.2%  60 26/20.8% 34/27.2%  
  95 Low High  95 Low High  297 Low High  125 Low High  297 Low High  
  50 45 46 49 110 187 70 55 149 148 
VISTA in CD68+ cells Low 46 30/31.6% 16/16.8% 0.0167 55 28/29.5% 18/18.9% 0.5692 149 62/20.9% 87/29.3% 0.1011 62 32/25.6% 30/24% 0.3267 149 83/27.9% 66/22.2% 0.0553 
 High 49 20/21.1% 29/30.5%  40 27/28.4% 22/23.2%  148 48/16.2% 100/33.7%  63 38/30.4% 25/20%  148 66/22.2% 82/27.6%  
CD8+ cells infiltrationCD4+ cells infiltrationCD3+ cells infiltrationCD20+ cells infiltrationCD68+ cells infiltration
462LowHigh462LowHigh675LowHigh503LowHigh297LowHigh
N231231PN232230PN326349PN252251PN149148P
total VISTA Low 241 142/31.7% 99/21.4% <0.0001 241 109/23.6% 132/28.6% 0.025 339 229/33.9% 97/14.4% <0.0001 242 138/27.4% 104/20.7% 0.0027 111 75 25.2% 36 12.1% <0.0001 
 High 221 89/19.3% 132/28.6%  221 123/26.6% 98/21.2%  336 110/16.3% 239/35.4%  261 114/22.7% 147/29.2%  186 74/24.9% 112/37.7%  
  462 Low High  462 Low High  414 Low High  411 Low High  95 Low High  
  231 231 232 230 212 202 207 204 52 43 
VISTA in CD8+ cells Low 231 130/28.1% 101/21.9% 0.0069 231 115/24.9% 116/25.1% 0.8524 208 122/29.5% 86/20.8% 0.002 205 110/26.8% 95/23.1% 0.1826 46 24/25.3% 22/23.2% 0.6267 
 High 231 101/21.9% 130/28.1%  231 117/25.3% 114/24.7%  206 90/21.7% 116/28.0%  206 97/23.6% 109/26.5%  49 28/29.5% 21/22.1%  
  462 Low High  462 Low High  414 Low High  411 Low High  95 Low High  
  231 231 232 230 212 202 207 204 52 43 
VISTA in CD4+ cells Low 231 137/29.6% 94/20.4% <0.0001 231 99/21.4% 132/28.6% 0.0015 205 109/26.3% 96/23.2% 0.4287 203 109/26.5% 94/22.9% 0.1821 45 25/26.3% 20/21.0% 0.8791 
 High 231 94/20.4% 137/29.6%  231 133/28.8% 98/21.2%  209 103/24.9% 106/25.6%  208 98/23.8% 110/26.8%  50 27/28.4% 23/24.2%  
  414 Low High  414 Low High  675 Low High  503 Low High  297 Low High  
  204 210 216 198 339 336 252 251 149 148 
VISTA in CD3+ cells Low 206 117/28.3% 89/21.5% 0.002 206 95/22.9% 111/26.8% 0.014 339 226/33.5% 113/16.7% <0.0001 255 147/29.2% 108/21.5% 0.0006 114 71/23.9% 43/14.5% 0.0009 
 High 208 87/21.0% 121/29.2%  208 121/29.2% 87/21.0%  336 113/16.7% 223/33.0%  248 105/20.9% 143/28.4%  183 78/26.3% 105/35.3%  
  411 Low High  411 Low High  503 Low High  503 Low High  125 Low High  
  203 208 215 196 255 248 252 251 63 62 
VISTA in CD20+ cells Low 205 117/28.5% 88/21.4% 0.0018 205 101/24.6% 104/25.3% 0.2177 252 136/27.0% 116/23.1% 0.1412 252 143/28.4% 109/21.7% 0.002 65 37/29.6% 28/22.4% 0.1284 
 High 206 86/20.9% 120/29.2%  206 114/27.7% 92/22.4%  251 119/23.7% 132/26.2%  251 109/21.7% 142/28.2%  60 26/20.8% 34/27.2%  
  95 Low High  95 Low High  297 Low High  125 Low High  297 Low High  
  50 45 46 49 110 187 70 55 149 148 
VISTA in CD68+ cells Low 46 30/31.6% 16/16.8% 0.0167 55 28/29.5% 18/18.9% 0.5692 149 62/20.9% 87/29.3% 0.1011 62 32/25.6% 30/24% 0.3267 149 83/27.9% 66/22.2% 0.0553 
 High 49 20/21.1% 29/30.5%  40 27/28.4% 22/23.2%  148 48/16.2% 100/33.7%  63 38/30.4% 25/20%  148 66/22.2% 82/27.6%  

NOTE: Simultaneous QIF was performed in a single unrelated NSCLC cohort. QIF scores for VISTA expression in a cellular subset were measured in the respective mask. Studied cohort was separated in cases with high and low expression using the median QIF score as cut point. Contingency analysis between immune cell infiltration and VISTA expression was performed. The number of cases per marker/mask (N) and P values for each category are indicated. Adjustment for multiple comparisons was performed using the Bonferroni method and statistical significance was met with P = 0.002. Bold P values are statistically significant after Bonferroni test.

VISTA levels and PD-1 axis in NSCLC

To evaluate the association between VISTA and major PD-1 axis components, we simultaneously measured the levels of cytokeratin, VISTA, PD-1 and PD-L1 protein in the NSCLC cohorts. As expected, PD-1 was located exclusively in immune cells of the stroma and PD-L1 was predominantly recognized in tumor epithelial cells and tumor associated macrophages (Fig. 3A and B). No cases showed simultaneous coexpression of all three markers in the same cell population. The most frequent phenotype seen was the coexpression of two of the targets in specific cell types. Notably, we found a proportion of cases containing stromal cells with simultaneous expression of VISTA/PD-1 and morphological features consistent with lymphocytes (Fig. 3A); and cells coexpressing VISTA/PD-L1 and features consistent with tumor-associated macrophages (Fig. 3B). Overall and measuring the markers in the whole tumor tissue sample, the levels of VISTA protein were significantly correlated with the levels of both PD-1 and PD-L1 (R2 = 0.53–0.61, P < 0.0001, Fig. 3C–F). In additional analysis using the median marker levels as stratification cutoff point, elevated expression of VISTA was significantly associated with high PD-1, PD-L1, and CD8+ TILs; and this association was consistent in both studied cohorts (Table 2).

Figure 3.

PD-1 axis markers are coexpressed and correlated with VISTA expression in NSCLC. Simultaneous QIF for VISTA, PD-L1 and PD-1 was performed in two different unrelated retrospective NSCLC cohorts (cohort #1, N = 324; cohort #2, N = 295). For both studied cohorts, the respective target coexpression, distribution and total QIF scores were measured. A–D, Representative coimmunolocalization of VISTA, PD-L1 and PD-1 proteins in NSCLC cases with high and low stromal detection are shown (A and B, respectively). Selected area shows an amplified view of the multiplex immunodetection. QIF scores for each studied target were analyzed by a scatterplot matrix, representing the correlation between PD-L1 and VISTA coexpression (C and D), and PD-1 and VISTA coexpression (E and F) in both NSCLC cohorts. Linear regression factor (R2) and P values are indicated. Images are representative of 619 cases.

Figure 3.

PD-1 axis markers are coexpressed and correlated with VISTA expression in NSCLC. Simultaneous QIF for VISTA, PD-L1 and PD-1 was performed in two different unrelated retrospective NSCLC cohorts (cohort #1, N = 324; cohort #2, N = 295). For both studied cohorts, the respective target coexpression, distribution and total QIF scores were measured. A–D, Representative coimmunolocalization of VISTA, PD-L1 and PD-1 proteins in NSCLC cases with high and low stromal detection are shown (A and B, respectively). Selected area shows an amplified view of the multiplex immunodetection. QIF scores for each studied target were analyzed by a scatterplot matrix, representing the correlation between PD-L1 and VISTA coexpression (C and D), and PD-1 and VISTA coexpression (E and F) in both NSCLC cohorts. Linear regression factor (R2) and P values are indicated. Images are representative of 619 cases.

Close modal
Table 2.

VISTA and PD-1 axis molecules are associated and correlated with tumor-infiltrating CD8+ cytotoxic lymphocytes

Cohort #1Cohort #2
VISTA expressionVISTA expression
NHighLowPNHighLowP
PD-1 324 162 162  292 146 146  
 High 162 41.4% 8.6% <0.0001 147 43.0% 6.8% <0.0001 
 Low 162 8.6% 41.4%  145 6.8% 43.4%  
PD-L1 324 162 162  292 146 146  
 High 162 41.0% 9.0% <0.0001 147 41.4% 8.5% <0.0001 
 Low 162 8.9% 41.1%  145 8.5% 41.6%  
CD8 277 145 132  288 147 141  
 High 140 32.5% 18.1% <0.0001 145 33.6% 15.5% 0.001 
 Low 137 19.9% 29.5%  143 20.0% 30.9%  
Cohort #1Cohort #2
VISTA expressionVISTA expression
NHighLowPNHighLowP
PD-1 324 162 162  292 146 146  
 High 162 41.4% 8.6% <0.0001 147 43.0% 6.8% <0.0001 
 Low 162 8.6% 41.4%  145 6.8% 43.4%  
PD-L1 324 162 162  292 146 146  
 High 162 41.0% 9.0% <0.0001 147 41.4% 8.5% <0.0001 
 Low 162 8.9% 41.1%  145 8.5% 41.6%  
CD8 277 145 132  288 147 141  
 High 140 32.5% 18.1% <0.0001 145 33.6% 15.5% 0.001 
 Low 137 19.9% 29.5%  143 20.0% 30.9%  

NOTE: Simultaneous QIF was performed and QIF scores for the respective target were measured in two different unrelated retrospective NSCLC cohorts (cohort #1, N = 324 and cohort #2, N = 295). Studied cohorts were separated in cases with high and low expression using the median QIF score as cut point. Contingency analysis for each cohort was independently performed and the P values for each category are indicated.

To further support our findings, we studied the association between the levels of VISTA, PD-1, PD-L1, and CD8A mRNA transcripts in the NSCLC datasets from TCGA. As shown in Fig. 4A and B, we found a positive association between the levels of VISTA, PD-1, and CD8 mRNA in both lung adenocarcinomas (N = 250, Fig. 4A) and squamous cell carcinomas (N = 120, Fig. 4B). However, a significant association between VISTA and PD-L1 transcript was seen only in lung adenocarcinomas from TCGA, but not in squamous tumors.

Figure 4.

VISTA mRNA is correlated with PD-L1 and PD-1 molecules and CD8+ cytotoxic lymphocytes in The TCGA data set. Scatterplot analysis of 250 cases of lung adenocarcinomas and 120 cases of lung squamous cell carcinoma from the TCGA data set were analyzed. Linear regression factor (R2) and P values for the association between VISTA, PD-L1, PD-1, and CD8a are indicated.

Figure 4.

VISTA mRNA is correlated with PD-L1 and PD-1 molecules and CD8+ cytotoxic lymphocytes in The TCGA data set. Scatterplot analysis of 250 cases of lung adenocarcinomas and 120 cases of lung squamous cell carcinoma from the TCGA data set were analyzed. Linear regression factor (R2) and P values for the association between VISTA, PD-L1, PD-1, and CD8a are indicated.

Close modal

Genotype/phenotype associations of VISTA expression in NSCLC

To explore the association of VISTA levels with major molecular subtypes of lung cancer, we studied a third cohort including 139 primary lung adenocarcinomas clinically tested for activating mutations in KRAS and EGFR (Supplementary Table S3). As shown in Fig. 5A, total or tumor-specific VISTA levels were comparable across cases with and without KRAS/EGFR mutations. However, elevated VISTA protein in stromal cells was significantly lower in tumors harboring oncogenic mutations in EGFR than in neoplasms lacking KRAS and EGFR mutations (Fig. 5B).

Figure 5.

VISTA shows a minimal association with lung cancer mutational status. An unrelated and retrospective cohort with major molecular adenocarcinoma variants was analyzed (cohort #3, N = 139). A and B, Levels of VISTA protein in the whole tumor tissue (A) or in the stromal area (B) in primary lung adenocarcinomas lacking mutations in KRAS and EGFR (nonmutated) and in tumors with oncogenic KRAS or EGFR variants (mutants). Scatterplot analysis of 250 cases of lung adenocarcinomas (C) and 120 cases of lung squamous cell carcinoma (D) from the TCGA data set were analyzed. Linear regression factor (R2) and P values for the association between VISTA mRNA expression and global mutational load are indicated. *, P < 0.05.

Figure 5.

VISTA shows a minimal association with lung cancer mutational status. An unrelated and retrospective cohort with major molecular adenocarcinoma variants was analyzed (cohort #3, N = 139). A and B, Levels of VISTA protein in the whole tumor tissue (A) or in the stromal area (B) in primary lung adenocarcinomas lacking mutations in KRAS and EGFR (nonmutated) and in tumors with oncogenic KRAS or EGFR variants (mutants). Scatterplot analysis of 250 cases of lung adenocarcinomas (C) and 120 cases of lung squamous cell carcinoma (D) from the TCGA data set were analyzed. Linear regression factor (R2) and P values for the association between VISTA mRNA expression and global mutational load are indicated. *, P < 0.05.

Close modal

To explore the association between VISTA levels and the somatic genomic landscape in NSCLC, we studied the relationship between VISTA mRNA expression and the nonsynonymous mutational burden in lung cancer cases from TCGA. As shown in Fig. 5C and D, we found an inverse relationship between VISTA transcript levels and the number of nonsynonymous DNA variants in lung adenocarcinomas (Fig. 5C). However, no significant association was seen in squamous lung malignancies (Fig. 5D).

Biological implications of VISTA expression in NSCLC

To assess the biological relevance of VISTA expression in lung carcinomas, we determined the association between VISTA levels, major clinicopathological variables and survival in the NSCLC cohorts. As shown in Table 3 and using the median score as stratification cutoff point, we found no consistent association between the level of total VISTA in the samples and major clinicopathological variables in the cohorts. However, elevated expression of VISTA measured exclusively in the tumor area was significantly associated with longer 5-year overall survival, and this was consistent in both the training cohort #1 and in the validation set/cohort #2 (log-rank P = 0.027–0.038, Fig. 6). No consistent association between stromal VISTA levels or VISTA measured in the total tissue area and survival were detected using the median score as stratification cutoff point (Supplementary Fig. S6).

Table 3.

Clinical relevance of VISTA expression in NSCLC

Cohort #1Cohort #2
VISTA expressionVISTA expression
NHighLowPNHighLowP
Total patients 324 167 157  292 150 142  
Age (years) 
 <70 247 39.9% 36.5% 0.589 125 29.7% 27.7% 0.814 
 ≥70 77 11.5% 12.1%  167 21.5% 21.2%  
Sex 
 Male 287 45.7% 42.9% 0.980 139 25.0% 22.6% 0.708 
 Female 37 5.9% 5.6%  153 26.4% 26.0%  
Smoking status 
 Never-smoker 26 4.5% 4.5% 0.911 39 7.6% 5.9% 0.529 
 Smoker 262 46.5% 44.4%  249 44.1% 42.4%  
 Unknown 36       
Histology 
 Adenocarcinoma 122 23.2% 20.4% 0.516 162 37.9% 31.9% 0.315 
 Squamous cell carcinoma 158 27.9% 28.6%  70 14.2% 16.0%  
 Others 44    57    
 Unknown       
Stage 
 I–II 191 29.2% 30.7% 0.293 229 41.7% 37.2% 0.462 
 III–IV 128 21.9% 18.2%  61 10.0% 11.0%  
 Unknown       
Cohort #1Cohort #2
VISTA expressionVISTA expression
NHighLowPNHighLowP
Total patients 324 167 157  292 150 142  
Age (years) 
 <70 247 39.9% 36.5% 0.589 125 29.7% 27.7% 0.814 
 ≥70 77 11.5% 12.1%  167 21.5% 21.2%  
Sex 
 Male 287 45.7% 42.9% 0.980 139 25.0% 22.6% 0.708 
 Female 37 5.9% 5.6%  153 26.4% 26.0%  
Smoking status 
 Never-smoker 26 4.5% 4.5% 0.911 39 7.6% 5.9% 0.529 
 Smoker 262 46.5% 44.4%  249 44.1% 42.4%  
 Unknown 36       
Histology 
 Adenocarcinoma 122 23.2% 20.4% 0.516 162 37.9% 31.9% 0.315 
 Squamous cell carcinoma 158 27.9% 28.6%  70 14.2% 16.0%  
 Others 44    57    
 Unknown       
Stage 
 I–II 191 29.2% 30.7% 0.293 229 41.7% 37.2% 0.462 
 III–IV 128 21.9% 18.2%  61 10.0% 11.0%  
 Unknown       

NOTE: Contingency analysis between major clinicopathological variables and total QIF scores for VISTA expression from two unrelated retrospective NSCLC cohorts (cohort #1, N = 324 and cohort #2, N = 295) was performed. Studied cohorts were separated in cases with high and low expression using the median QIF score as cutoff point. P values for each category are indicated.

Figure 6.

Association between VISTA levels and overall survival in NSCLC. Kaplan–Meier graphical analysis of the 5-year overall survival in NSCLC cases from cohort #1 (A) and cohort #2 (B) based on the levels of VISTA protein expression in the tumor area. Patients from two independent cohorts were stratified by VISTA QIF score (low vs. high expression) using the median as a cutoff point. P values comparing risk groups were calculated with the log-rank test.

Figure 6.

Association between VISTA levels and overall survival in NSCLC. Kaplan–Meier graphical analysis of the 5-year overall survival in NSCLC cases from cohort #1 (A) and cohort #2 (B) based on the levels of VISTA protein expression in the tumor area. Patients from two independent cohorts were stratified by VISTA QIF score (low vs. high expression) using the median as a cutoff point. P values comparing risk groups were calculated with the log-rank test.

Close modal

Despite the prominent interest and possible role of VISTA/PD-1H as a novel anticancer immunotherapy target in human malignancies, little is known about its expression in cancer tissues. Here, we have carefully validated a protein detection assay and interrogated the expression, tissue distribution, and role of VISTA in a sizable number of human lung carcinomas from 4 independent populations. Our data demonstrate that VISTA is expressed in the vast majority of human NSCLCs with specific geographical/spatial patterns and positively associated with tumor immune infiltration and PD-1/PD-L1 expression. The levels of VISTA are not associated with specific NSCLC histological subtypes or patient clinical/demographical variables. However, compartment specific expression of VISTA in stromal or tumor epithelial cells is associated with oncogenic EGFR mutations and with the nonsynonymous mutational burden in lung adenocarcinomas; and with overall survival in two independent NSCLC cohorts. Taken together, these findings suggest an immune-modulatory effect of VISTA in human lung cancer.

Most studies addressing VISTA expression in tissues have used mRNA expression or flow cytometry of mechanically/enzymatically disaggregated samples, thus potentially altering the native cell conditions and precluding assessment of the spatial context. In mice, high expression of VISTA has been detected in hematopoietic cells, lung and small intestine, and very low levels of mRNA have been found in the heart, brain, muscle, kidney, testis, and placenta (14, 15). In humans, VISTA has also been reported to be mainly expressed in hematopoietic cells (14–19). In immune cells, the highest level of VISTA expression is detected in monocytes and dendritic cells, intermediate levels on neutrophils, and very low levels on natural killer (NK) cells (16). VISTA expression was also detected with variable levels in both parenchymal and stromal/immune cells from morphologically normal human brain, thyroid, stomach, spleen, liver and in human peripheral blood mononuclear cells (PBMC) (19). Notably, the levels of VISTA in PBMCs or isolated human monocytes were prominently upregulated by TLR3-, TLR5-ligand, IL10 and IFNγ (19). The latter indicates the inducible nature of VISTA under inflammatory conditions and supports the need for context-specific measurements.

To our knowledge, no studies reliably evaluating the expression and location of VISTA protein in human tumors have been reported. For instance, previous reports failed to demonstrate VISTA expression in colon and lung cancer samples using a noncommercial monoclonal antibody (clone GG8) after seeing limited sensitivity and affinity of the assay for use in immunohistochemistry studies (16, 20). In our work and using a validated commercially available assay that is suitable for FFPE tissue samples, we found expression of VISTA in >99% of lung cancer samples from over 700 cases studied. Notably, we found distinct expression patterns with most cases showing stromal-only staining and nearly 20% showing staining in stromal and epithelial tumor cells. The expression in tumor cells was predominantly focal and spatially connected with immune infiltration and PD-1/PD-L1 expression, suggesting that locally secreted factors such as interleukins or interferons could mediate VISTA upregulation. In support of this notion, VISTA expression has been shown to be directly and synergistically induced by IL10 and IFNγ (19). Additional studies will be required to determine the effect of multiple cytokines/chemokines (alone or in combination) in VISTA expression on tumor and immune cells. To date, only few studies have directly assessed the role of VISTA protein in tumor cells. One study described that VISTA overexpression in a murine fibro-sarcoma model promotes tumor growth and the effect was T-cell dependent (15). However, the biological (e.g., immune) determinants of this finding and the ligand/receptor function of VISTA in tumor cells remain unclear.

In human lung cancer, VISTA protein was found in different TIL populations and in tumor-associated macrophages. Moreover, higher levels of VISTA were seen in effector CD8+ T cells than in CD4-helper TILs and expression of VISTA in the whole sample or in immune cells was in general associated with increased tumor immune infiltration by CD8+, CD20+, and CD68+ cells, but not by CD4+ helper T lymphocytes. The latter suggest that VISTA expression in the tumor immune microenvironment could be related with antitumor immune pressure. We also observed limited association between the tumor mutational burden and VISTA levels in lung carcinomas. The determinants for this are currently under investigation.

Several preclinical mouse models have reported that VISTA upregulation in stromal cells can act as an immune evasion mechanism. For instance, VISTA expression is higher on immature dendritic cells, myeloid-derived suppressor cells (MDSC) and Tregs than in peripheral tissues (18). Due to prominent limitations in the current understanding and validation of markers to characterize specific myeloid cell subsets in human specimens, our study did not include major immune suppressive cell types such as alternatively polarized macrophages (e.g., M2-type macrophages) and MDSCs. Studies are under way to determine the optimal strategy to accurately interrogate such populations.

After major clinical success of blocking the PD-1 axis using monoclonal antibodies, the current landscape of anticancer immunotherapies includes numerous combinations of immune and nonimmune agents. Therefore, identification of independent or redundant immune evasion pathways could be used to support optimal treatment modalities and rational design of clinical trials. Previous work using VISTA and PD-1 knockout mice demonstrated that the immune-regulatory pathways for PD-1 and VISTA are functionally nonredundant in antigen-specific responses and during autoimmune inflammatory conditions (21). However, little is known about the association between VISTA and PD-1 pathways in human malignancies. In our study, we found a consistent and prominent association between the levels of VISTA protein, PD-1 and PD-L1 in NSCLC cohorts represented in TMA format and also measuring mRNA transcripts in whole tissue lung cancer specimens from TCGA. The latter indicates the robustness of the association and excludes a possible impact/bias of measuring the targets in relatively small TMA histospots. Although we noted distinct cellular coexpression patterns of VISTA, PD-1 and PD-L1 in the human tumors (Fig. 3A and B), our results demonstrate frequent regional colocalization of the markers in lung cancer and suggest a synergetic/cooperative immune evasion effect. The previously reported induction of VISTA and PD-L1 by proinflammatory cytokines such as IFNγ could mechanistically support the markers coexpression and the positive association with TILs. Therefore, simultaneous blockade of both VISTA and PD-1 pathway could represent an effective antitumor strategy in human NSCLC. The results from ongoing and future human clinical trials using anti-VISTA monotherapy or combination therapies will be required to support this observation.

Conflicting results about the antitumor effect of dual VISTA/PD-1 axis blockade have been reported using two different monoclonal VISTA antibodies in preclinical models (21, 22). A synergistic effect of the combination of a monoclonal therapy against VISTA and PD-L1 was described in a colon cancer mouse model, showing a reduction in the tumor growth and an increased overall survival in comparison to monotherapy regimens (21). However, and despite seeing increased CD8 T-cell function, no effect on tumor growth was detected after treatment with the combination of anti-VISTA/PD-1 treatment in an oral squamous cell carcinoma model (22). In support of our findings in human tumors, a recent study showed a positive association between VISTA and PD-L1 protein levels; and a cooperative effect of VISTA and CD8+ cells to predict survival in a retrospective cohort of human oral squamous carcinomas (31). Taken together, these data suggest that the VISTA/PD-1H pathway may contribute to the adaptive resistance mechanisms operating in the tumor microenvironment.

Increased VISTA expression could also participate in acquired resistance to immune checkpoint blockade. In this regard, VISTA upregulation after CTLA-4 blockade with ipilimumab was recently reported in prostate carcinomas (32). In this study, VISTA was upregulated in tumor-infiltrating immune cells, particularly in CD68+ tumor-associated macrophages. The possible role of VISTA in mediating resistance to immune checkpoint blockers in human lung carcinomas is currently under investigation.

V. Velcheti is a consultant/advisory board member for Amgen, AstraZeneca, Bristol-Myers Squibb, Celgene, Fulgent Health, Genentech, Merck, and Navigate Biopharma. L. Chen reports receiving commercial research grants from Boehringer Ingelheim, NextCure, and Pfizer, and is a consultant/advisory board member for GenomiCare, Pfizer, and VcanBio. K.A. Schalper reports receiving commercial research grants from Navigate, Onkaido, Surface Oncology, Takeda, Tesaro, and Vasculox, speakers bureau honoraria from Merck and Takeda, and is a consultant/advisory board member for Celgene and Shattuck Labs. No potential conflicts of interest were disclosed by the other authors.

Conception and design: F. Villarroel-Espindola, K. Syrigos, L. Chen, R.S. Herbst, K.A. Schalper

Development of methodology: F. Villarroel-Espindola, I. Datar, V. Velcheti, L. Chen, R.S. Herbst, K.A. Schalper

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): F. Villarroel-Espindola, M. Sanmamed, V. Velcheti, K. Syrigos, R.S. Herbst

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): F. Villarroel-Espindola, X. Yu, V. Velcheti, K. Syrigos, H. Zhao, L. Chen, R.S. Herbst, K.A. Schalper

Writing, review, and/or revision of the manuscript: F. Villarroel-Espindola, I. Datar, M. Sanmamed, V. Velcheti, K. Syrigos, M. Toki, H. Zhao, L. Chen, R.S. Herbst, K.A. Schalper

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): F. Villarroel-Espindola, N. Mani, M. Sanmamed, M. Toki

Study supervision: F. Villarroel-Espindola, L. Chen, R.S. Herbst, K.A. Schalper

Thanks to Lori Charette and Yale Pathology Tissue Services for the technical support.

This study was supported by the Lung Cancer Research Foundation (LCRF), Yale SPORE in Lung Cancer (P50CA196530), Department of Defense-Lung Cancer Research Program Career Development Award (LC150383), and Stand Up to Cancer – American Cancer Society Lung Cancer Dream Team Translational Research Grant (grant number: SU2C-AACR-DT17-15). Stand Up To Cancer is a program of the Entertainment Industry Foundation. Research grants are administered by the American Association for Cancer Research, the scientific partner of SU2C, sponsored research support by Navigate Biopharma and Yale Cancer Center Support Grant (P30CA016359).

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.
Sznol
M
,
Chen
L
. 
Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human cancer–response
.
Clin Cancer Res
2013
;
19
:
5542
.
2.
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
.
3.
Butte
MJ
,
Keir
ME
,
Phamduy
TB
,
Sharpe
AH
,
Freeman
GJ
. 
Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses
.
Immunity
2007
;
27
:
111
22
.
4.
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
.
5.
Herbst
RS
,
Baas
P
,
Kim
DW
,
Felip
E
,
Pérez-Gracia
JL
,
Han
JY
, et al
Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial
.
Lancet
2016
;
387
:
1540
50
.
6.
Larkin
J
,
Lao
CD
,
Urba
WJ
,
McDermott
DF
,
Horak
C
,
Jiang
J
,
Wolchok
JD
. 
Efficacy and safety of nivolumab in patients with BRAF V600 mutant and BRAF wild-type advanced melanoma: a pooled analysis of 4 clinical trials
.
JAMA Oncol
2015
;
1
:
433
40
.
7.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Cowey
CL
,
Lao
CD
, et al
Combined nivolumab and ipilimumab or monotherapy in untreated melanoma
.
N Engl J Med
2015
;
373
:
23
34
.
8.
Lavin
Y
,
Kobayashi
S
,
Leader
A
,
Amir
ED
,
Elefant
N
,
Bigenwald
C
, et al
Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses
.
Cell
2017
;
169
:
750
65
.
e17
.
9.
Hirsh
V
. 
New developments in the treatment of advanced squamous cell lung cancer: focus on afatinib
.
Onco Targets Ther
2017
;
10
:
2513
26
.
10.
Dong
H
,
Zhu
G
,
Tamada
K
,
Chen
L
. 
B7-H1, a third member of the B7 family, costimulates T cell proliferation and interleukin-10 secretion
.
Nat Med
1999
;
5
:
1365
9
.
11.
Freeman
GJ
,
Long
AJ
,
Iwai
Y
,
Bourque
K
,
Chernova
T
,
Nishimura
H
, et al
Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation
.
J Exp Med
2000
;
192
:
1027
34
.
12.
Azuma
T
,
Yao
S
,
Zhu
G
,
Flies
AS
,
Flies
SJ
,
Chen
L
. 
B7-H1 is a ubiquitous antiapoptotic receptor on cancer cells
.
Blood
2008
;
111
:
3635
43
.
13.
Zou
W
,
Chen
L
. 
Inhibitory B7-family molecules in the tumour microenvironment
.
Nat Rev Immunol
2008
;
8
:
467
77
.
14.
Flies
DB
,
Wang
S
,
Xu
H
,
Chen
L
. 
Cutting edge: a monoclonal antibody specific for the programmed death-1 homolog prevents graft-versus-host disease in mouse models
.
J Immunol
2011
;
187
:
1537
41
.
15.
Wang
L
,
Rubinstein
R
,
Lines
JL
,
Wasiuk
A
,
Ahonen
C
,
Guo
Y
, et al
VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses
.
J Exp Med
2011
;
208
:
577
92
.
16.
Lines
JL
,
Pantazi
E
,
Mak
J
,
Sempere
LF
,
Wang
L
,
O'Connell
S
, et al
VISTA is an immune checkpoint molecule for human T cells
.
Cancer Res
2014
;
74
:
1924
32
.
17.
Flies
DB
,
Han
X
,
Higuchi
T
,
Zheng
L
,
Sun
J
,
Ye
JJ
, et al
Coinhibitory receptor PD-1H preferentially suppresses CD4+ T cell-mediated immunity
.
J Clin Invest
2014
;
124
:
1966
75
.
18.
Le Mercier
I
,
Chen
W
,
Lines
JL
,
Day
M
,
Li
J
,
Sergent
P
, et al
VISTA regulates the development of protective antitumor immunity
.
Cancer Res
2014
;
74
:
1933
44
.
19.
Bharaj
P
,
Chahar
HS
,
Alozie
OK
,
Rodarte
L
,
Bansal
A
,
Goepfert
PA
, et al
Characterization of programmed death-1 homologue-1 (PD-1H) expression and function in normal and HIV infected individuals
.
PLoS One
2014
;
9
:
e109103
.
20.
Lines
JL
,
Sempere
LF
,
Broughton
T
,
Wang
L
,
Noelle
R
. 
VISTA is a novel broad-spectrum negative checkpoint regulator for cancer immunotherapy
.
Cancer Immunol Res
2014
;
2
:
510
7
.
21.
Liu
J
,
Yuan
Y
,
Chen
W
,
Putra
J
,
Suriawinata
AA
,
Schenk
AD
, et al
Immune-checkpoint proteins VISTA and PD-1 nonredundantly regulate murine T-cell responses
.
Proc Natl Acad Sci U S A
2015
;
112
:
6682
7
.
22.
Kondo
Y
,
Ohno
T
,
Nishii
N
,
Harada
K
,
Yagita
H
,
Azuma
M
. 
Differential contribution of three immune checkpoint (VISTA, CTLA-4, PD-1) pathways to antitumor responses against squamous cell carcinoma
.
Oral Oncol
2016
;
57
:
54
60
.
23.
Velcheti
V
,
Schalper
KA
,
Carvajal
DE
,
Anagnostou
VK
,
Syrigos
KN
,
Sznol
M
, et al
Programmed death ligand-1 expression in non-small cell lung cancer
.
Lab Invest
2014
;
94
:
107
16
.
24.
Anagnostou
VK
,
Syrigos
KN
,
Bepler
G
,
Homer
RJ
,
Rimm
DL
. 
Thyroid transcription factor 1 is an independent prognostic factor for patients with stage I lung adenocarcinoma
.
J Clin Oncol
2009
;
27
:
271
8
.
25.
Schalper
KA
,
Brown
J
,
Carvajal-Hausdorf
D
,
McLaughlin
J
,
Velcheti
V
,
Syrigos
KN
, et al
Objective measurement and clinical significance of TILs in non-small cell lung cancer
.
J Natl Cancer Inst
2015
;
107
.
pii: dju435
.
26.
Carvajal-Hausdorf
DE
,
Schalper
KA
,
Neumeister
VM
,
Rimm
DL
. 
Quantitative measurement of cancer tissue biomarkers in the lab and in the clinic
.
Lab Invest
2015
;
95
:
385
96
.
27.
Schalper
KA
,
Carvajal-Hausdorf
D
,
McLaughlin
J
,
Altan
M
,
Velcheti
V
,
Gaule
P
, et al
Differential expression and significance of PD-L1, IDO-1, and B7-H4 in human lung cancer
.
Clin Cancer Res
2017
;
23
:
370
8
.
28.
Mani
NL
,
Schalper
KA
,
Hatzis
C
,
Saglam
O
,
Tavassoli
F
,
Butler
M
, et al
Quantitative assessment of the spatial heterogeneity of tumor-infiltrating lymphocytes in breast cancer
.
Breast Cancer Res
2016
;
18
:
78
84
.
29.
Gaule
P
,
Smithy
JW
,
Toki
M
,
Rehman
J
,
Patell-Socha
F
,
Cougot
D
, et al
A quantitative comparison of antibodies to programmed cell death 1 ligand 1
.
JAMA Oncol
2017
;
3
:
256
9
.
30.
Bordeaux
J
,
Welsh
A
,
Agarwal
S
,
Killiam
E
,
Baquero
M
,
Hanna
J
, et al
Antibody validation
.
Biotechniques
2010
;
48
:
197
209
.
31.
Wu
L
,
Deng
WW
,
Huang
CF
,
Bu
LL
,
Yu
GT
,
Mao
L
, et al
Expression of VISTA correlated with immunosuppression and synergized with CD8 to predict survival in human oral squamous cell carcinoma
.
Cancer Immunol Immunother
2017
;
66
:
627
36
.
32.
Gao
J
,
Ward
JF
,
Pettaway
CA
,
Shi
LZ
,
Subudhi
SK
,
Vence
LM
, et al
VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer
.
Nat Med
2017
;
23
:
551
5
.