V-domain Ig suppressor of T-cell activation (VISTA) is a novel negative checkpoint ligand that is homologous to PD-L1 and suppresses T-cell activation. This study demonstrates the multiple mechanisms whereby VISTA relieves negative regulation by hematopoietic cells and enhances protective antitumor immunity. VISTA is highly expressed on myeloid cells and Foxp3+CD4+ regulatory cells, but not on tumor cells within the tumor microenvironment (TME). VISTA monoclonal antibody (mAb) treatment increased the number of tumor-specific T cells in the periphery and enhanced the infiltration, proliferation, and effector function of tumor-reactive T cells within the TME. VISTA blockade altered the suppressive feature of the TME by decreasing the presence of monocytic myeloid-derived suppressor cells and increasing the presence of activated dendritic cells within the tumor microenvironment. In addition, VISTA blockade impaired the suppressive function and reduced the emergence of tumor-specific Foxp3+CD4+ regulatory T cells. Consequently, VISTA mAb administration as a monotherapy significantly suppressed the growth of both transplantable and inducible melanoma. Initial studies explored a combinatorial regimen using VISTA blockade and a peptide-based cancer vaccine with TLR agonists as adjuvants. VISTA blockade synergized with the vaccine to effectively impair the growth of established tumors. Our study therefore establishes a foundation for designing VISTA-targeted approaches either as a monotherapy or in combination with additional immune-targeted strategies for cancer immunotherapy. Cancer Res; 74(7); 1933–44. ©2014 AACR.

Immune responses against cancer are negatively regulated by multiple checkpoints, including CTLA-4, PD-L1/PD-1, and B7-H4 pathways. Targeting of these negative immune regulators has proved to be a clinically effective strategy to enhance tumor-specific immune responses (1, 2).

The critical role of T CTLA-4 in suppressing tumor-specific immunity was demonstrated when antibody-mediated CTLA-4 targeting in combination with a cellular vaccine induced regression of established, poorly immunogenic B16 melanoma (3). Ipilimumab, an anti-human CTLA-4 monoclonal antibody (mAb), as a monotherapy has proved to exert clinical benefit in late-stage melanoma in patients and has been approved for treating advanced melanoma, as well as undergoing early-phase trials for other cancers (4–6).

Programmed death-1 (PD-1) and its ligands PD-L1/PD-L2 represent another immune negative checkpoint axis (7, 8). The PD-1 pathway downregulates tumor-specific immunity by impairing T-cell responses and promoting the induction of Foxp3+ Tregs in the periphery (1, 9). Blocking the PD-L1/PD-1 pathway, in conjunction with other immune therapies inhibits tumor progression (10–15). MDX-1106/BMS-936558, the human αPD-1 mAb, as well as a human αPD-L1 mAb, have entered clinical trials, and early studies have shown resounding clinical results (16–18).

Given the success of immune-checkpoint regulator blockade in improving both endogenous and vaccine-elicited antitumor immune responses, identification of additional negative checkpoint regulator pathways would likely have important therapeutic implications. We have recently discovered a novel immunoglobulin (Ig) superfamily ligand, designated V-domain Ig suppressor of T-cell activation (VISTA; Genbank: JN602184; ref. 19). VISTA bears homology to PD-L1 but displays a distinct expression pattern. Within the hematopoietic compartment, VISTA is constitutively and highly expressed on CD11bhigh myeloid cells, and expressed at lower levels on CD4+ and CD8+ T cells and Foxp3+ Tregs. The human and murine homologs share 90% homology, have indistinguishable functional properties, and are similar in their lineage restricted expression (20). VISTA expressed on APCs directly suppresses CD4+ and CD8+ T-cell proliferation and cytokine production (19).

We hypothesize that VISTA is a novel negative checkpoint regulator and a promising new target for cancer immunotherapy. This study has utilized a VISTA-specific blocking mAb to examine the role of VISTA in regulating antitumor immunity. Our data show that VISTA mAb treatment impaired the suppressive character of the tumor microenvironment (TME) and enhanced protective antitumor immunity. Furthermore, initial studies exploring a combination regimen of VISTA blockade together with a peptide vaccine show synergistic efficacy. As such, our study establishes a foundation for designing optimal therapeutic approaches that incorporate VISTA blockade either as a monotherapy or in combination with additional immune-targeted therapies.

Mice

C57BL/6 mice were from NCI. TRP1 and OTII CD4 transgenic mice were from the Jackson Laboratory. FoxP3-GFP reporter mice were as described (21) and were generously provided by Dr. Alexander Rudensky (University of Washington School of Medicine, Seattle, WA). The triple transgenic mice strain B6.Cg-BrafCA/+ Ptenlox5/lox5 Tg (Tyr::Cre/ERT2) was obtained from Dr. Bosenberg (Yale School of Medicine, New Haven, CT). All animals were maintained in a pathogen-free facility at Dartmouth Medical School. All animal protocols were Institutional Animal Care and Use Committee approved at the Dartmouth College.

Tumor models, tumor vaccine, and treatment

MB49 (300,000), B16OVA (120,000), and B16BL6 (18,000) tumor cells were inoculated on the right flank of female mice. For the PTEN/BRAF melanoma model, tumors were induced by intradermal injection of 10 μL tamoxifen (10 μL dissolved in dimethyl sulfoxide) on the lower back. Tumor vaccine consisted of CD40 agonistic antibody FGK (100 μg), LPS (30 μg), polyI:C (100 μg), CpG (ODN1826, 30 μg), Gardiquimod (30 μg), tumor antigen peptide TRP1 (106–130; 100 μg), and a mutated TRP2 peptide DeltaV-TRP2 (180–188; 100 μg; refs. 22–24). Vaccine mixture was applied with split dose subcutaneously on indicated days. For prophylactic anti-VISTA mAb treatment, mice were treated every 2 days with 300 μg antibody subcutaneously, starting on day 0 for the entire duration of the experiment. For therapeutic treatment, mice were treated with subcutaneous injection of 300 μg mAb every day for 10 days on day+2 or day+7 after tumor inoculation, followed by continuous antibody injection every 2 days for the entire duration of the experiment.

Flow cytometry and analysis

Flow cytometry analysis was performed either on FACSCAN using CellQuest software (BD Biosciences) or on MACSquant 7 color analyzer (Miltenyi). Data analysis was performed using FlowJo software (Treestar).

Graphs and statistical analysis

All graphs and statistical analysis were generated using Prism 4 (GraphPad Software, Inc.). A Student t test (two-tailed) was used for the data analyses. ***, P < 0.005; **, P < 0.025; *, P < 0.05.

Materials and Methods are described in detail in the Supplementary Data.

VISTA mAb treatment impairs tumor growth in immunogenic transplantable tumor models

We have previously generated a hamster monoclonal antibody (clone 13F3) that neutralizes the suppressive activity of VISTA (19). We hypothesized that VISTA mAb-mediated blockade would enhance antitumor immune responses. This hypothesis was tested in murine tumor models. We first examined the immunogenicity and in vivo clearance of 13F3 (Supplementary Fig. S1). Our data show that mice developed strong immune response against 13F3 and accumulated high levels neutralizing antibodies, which presumably leads to fast clearance of 13F3. In fact, after a week of continuous treatment, we can no longer detect any 13F3 in the serum when blood was analyzed 24 hours after each injection. Our data indicates that the most effective window of 13F3-mediated VISTA blockade in vivo might be within the first week of treatment.

Next, we examined the impact of VISTA mAb treatment in mice bearing melanoma B16OVA, which expresses the chicken ovalbumin as a neo tumor antigen. Despite the apparent immunogenicity and short half-life of 13F3 in vivo, 13F3 treatment significantly suppressed tumor growth in the B16OVA model (Fig. 1A). Increased number of IFN-γ–producing cells in the tumor-draining lymph node was detected by ELISPOT assay in response to irradiated tumor cells, indicating that VISTA mAb treatment enhanced tumor-specific T-cell responses. Even though we cannot detect any expression of VISTA on nonhematopoietic cells, we examined in vitro cultured tumor cells to exclude the possibility that VISTA mAb directly affected tumor cell proliferation and apoptosis (Supplementary Fig. S2).

Figure 1.

VISTA mAb treatment suppressed tumor growth and altered the tumor microenvironment in B16OVA melanoma model. A, B16OVA tumor-bearing mice (n = 8 for control group; n = 10 for 13F3 group) were treated with anti-VISTA mab (13F3) or control-Ig starting from day 0, every 2 days throughout the duration of the experiment. Tumor size was measured with a caliper. IFN-γ ELISPOT was performed using tumor-draining lymph node cells on day 14. Cells were restimulated in vitro with irradiated tumor cells for 20 hours. IFN-γ–producing cells were visualized and counted. B and C, cells were harvested from peripheral lymph node, tumor-draining lymph node, and tumor tissues around day 20 when tumors reached approximately 8–10 mm diameter. VISTA expression on myeloid-derived suppressor cells (MDSC, CD11bhi CD11c Gr1+) and myeloid DCs (CD11b+ CD11c+), and a comparison of VISTA and PD-L1 expression on tumor cells and tumor-infiltrating leukocytes (TIL) was shown by flow cytometry. D, VISTA expression within B16 melanoma tumor tissue was examined by immunofluorescence. 4′, 6-diamidino-2-phenylindole (DAPI), blue; Cd11b, green; VISTA or control-IgG, red. E–H, VISTA mAb treatment altered the TME and improved the effector function of tumor-infiltrating CD8+ T cells. B16OVA-bearing mice were treated with 13F3 or control Ig from day 0 until tumors in the control group reached 9–10 mm diameter (∼day+19). Tumors were harvested and CD45+ TILs were analyzed E. Various TIL populations, including CD4+ and CD8+ T cells, and MDSC were identified by flow cytometry. Proliferation of tumor-infiltrating CD8+ T cells was analyzed on the basis of Ki67 expression (F). G and H, TIL CD8+ cells were stimulated with irradiated tumor cells for 20 hours. Cytokine production (IFN-γ), surface mobilization of CD107ab, and granzyme B expression were quantified. Data are representative of 2–3 independent experiments.

Figure 1.

VISTA mAb treatment suppressed tumor growth and altered the tumor microenvironment in B16OVA melanoma model. A, B16OVA tumor-bearing mice (n = 8 for control group; n = 10 for 13F3 group) were treated with anti-VISTA mab (13F3) or control-Ig starting from day 0, every 2 days throughout the duration of the experiment. Tumor size was measured with a caliper. IFN-γ ELISPOT was performed using tumor-draining lymph node cells on day 14. Cells were restimulated in vitro with irradiated tumor cells for 20 hours. IFN-γ–producing cells were visualized and counted. B and C, cells were harvested from peripheral lymph node, tumor-draining lymph node, and tumor tissues around day 20 when tumors reached approximately 8–10 mm diameter. VISTA expression on myeloid-derived suppressor cells (MDSC, CD11bhi CD11c Gr1+) and myeloid DCs (CD11b+ CD11c+), and a comparison of VISTA and PD-L1 expression on tumor cells and tumor-infiltrating leukocytes (TIL) was shown by flow cytometry. D, VISTA expression within B16 melanoma tumor tissue was examined by immunofluorescence. 4′, 6-diamidino-2-phenylindole (DAPI), blue; Cd11b, green; VISTA or control-IgG, red. E–H, VISTA mAb treatment altered the TME and improved the effector function of tumor-infiltrating CD8+ T cells. B16OVA-bearing mice were treated with 13F3 or control Ig from day 0 until tumors in the control group reached 9–10 mm diameter (∼day+19). Tumors were harvested and CD45+ TILs were analyzed E. Various TIL populations, including CD4+ and CD8+ T cells, and MDSC were identified by flow cytometry. Proliferation of tumor-infiltrating CD8+ T cells was analyzed on the basis of Ki67 expression (F). G and H, TIL CD8+ cells were stimulated with irradiated tumor cells for 20 hours. Cytokine production (IFN-γ), surface mobilization of CD107ab, and granzyme B expression were quantified. Data are representative of 2–3 independent experiments.

Close modal

VISTA mAb treatment alters the cellular composition of the tumor immune microenvironment

The TME plays a crucial role in suppressing tumor-specific T-cell responses (25, 26). VISTA is found to be highly expressed on tumor-infiltrating myeloid cells, including myeloid DCs (CD11b+ CD11c+Gr1) and myeloid-derived suppressor cells (CD11b+Gr1+CD11C), but is not expressed on B16 tumor cells (Fig. 1B–D). This expression pattern is in contrast to PD-L1, which is expressed on both tumor cells and tumor-infiltrating leukocytes (TIL; Fig. 1C).

VISTA mAb treatment altered the TIL composition in the B16OVA model. αVISTA administration increased total CD45+ leukocytes tumor infiltration from 25.77% ± 5.95% to 57.78% ± 3.97% (Fig. 1E). Within the TIL CD45+ population, a decreased percentage of CD11b+Gr1+CD11C MDSCs (from 37.74% ± 2.44% to 25.64% ± 0.93%), and increased percentages of tumor-specific CD4+ T cells (from 6.38% ± 0.27% to 11.74% ± 0.97%) and CD8+ T cells (from 9.25% ± 0.95% to 17.74% ± 0.41%) were seen with αVISTA treatment. Tumor-infiltrating CD8+ T cells expressed phenotypes of CD44hiCD62Llow (data not shown), produced effector molecules (i.e., IFN-γ and granzyme B), and mobilized CD107ab upon restimulation with irradiated tumor cells ex vivo, indicating that these tumor-infiltrating T cells contained tumor-specific effector T-cell populations (Fig. 1F–H). VISTA mAb administration enhanced the proliferation and the effector function of tumor-infiltrating CD8+ T cells, as evidenced by increased number of Ki67+ cells, enhanced effector molecule production (i.e., IFN-γ and granzyme B), and CD107 mobilization (Fig. 1F–H and Supplementary Fig. S1). There are no significant alterations of myeloid cells and T-cell lineages in the spleen or tumor-draining lymph nodes upon VISTA mAb treatment (data not shown), indicating that VISTA mAb did not induce antibody-mediated deletion of VISTA+ cells. VISTA blockade therefore appears to act predominantly on altering the TME by enhancing the frequency of tumor-infiltrating effector T cells as well as their effector functions, resulting in enhanced control of tumor growth.

Similar analysis was performed in an immunogenic bladder tumor model MB49 (27). Consistent with that observed in the melanoma model, VISTA mAb treatment significantly suppressed MB49 tumor growth (Fig. 2A). Similar to the B16OVA model, VISTA is highly expressed on tumor-infiltrating myeloid cell populations (Fig. 2B). Unlike the B16OVA tumors, VISTA mAb treatment in MB49 tumors did not reduce the relative percentage of MDSCs within the CD45+ TILs. Further analysis indicated that more than 90% of the MDSC populations infiltrating MB49 tumors were of the granulocytic phenotype (Cd11b+Gr1hiLy6G+Ly6C-int), which is in contrast to the predominant monocytic MDSC populations (Cd11b+Gr1intLy6GLy6Chi) infiltrating B16 melanoma (Supplementary Fig. S3). Despite the unaltered presence of MDSCs, VISTA mAb enhanced the activation status of tumor-associated CD11c+ DCs, which showed higher expression of MCHII and CD80, and higher production of interleukin (IL)-12 and TNF-α (Fig. 2D). In αVISTA–treated mice, tumor-specific T-cell responses were significantly enhanced, both in the tumor-draining lymph node and within the tumor tissue (Fig. 2).

Figure 2.

Effects of VISTA mAb treatment on the growth and the tumor microenvironment of MB49 bladder tumors. MB49 tumor-bearing mice (n = 14 per group) were treated with control-Ig or 13F3 every 2 days starting from day 0. Tumor size was measured with a caliper. A, IFN-γ ELISPOT was performed using tumor-draining lymph node cells on day 14 as described for the B16OVA tumor model in Fig. 1. B, when MB49 tumors reached 9–10 mm diameter (∼day 20), cells were harvested from peripheral lymph node, tumor-draining lymph node, and tumor tissues. VISTA expression on myeloid-derived suppressor cells (MDSC, CD11bhi CD11c Gr1+) and myeloid DCs (CD11b+ CD11c+) was analyzed by flow cytometry. C–E, VISTA mAb treatment altered the TME and improved the effector function of tumor-infiltrating T cells. MB49-bearing mice were treated with 13F3 or control Ig from day 0 until tumors in the control group reached 9–10 mm diameter. Tumors were harvested and total CD45+ TILs were quantified. Tumor-infiltrating DCs were analyzed by flow cytometry for their surface expression of MHCII and CD80 and cytokine production (i.e., IL12p40 and TNF-α). Tumor-infiltrating CD4+ and CD8+ T cells were stimulated with irradiated tumor cells for 20 hours. Cytokine production (i.e., IFN-γ and TNF-α) was analyzed by flow cytometry and quantified. Data are representative of 2–3 independent experiments.

Figure 2.

Effects of VISTA mAb treatment on the growth and the tumor microenvironment of MB49 bladder tumors. MB49 tumor-bearing mice (n = 14 per group) were treated with control-Ig or 13F3 every 2 days starting from day 0. Tumor size was measured with a caliper. A, IFN-γ ELISPOT was performed using tumor-draining lymph node cells on day 14 as described for the B16OVA tumor model in Fig. 1. B, when MB49 tumors reached 9–10 mm diameter (∼day 20), cells were harvested from peripheral lymph node, tumor-draining lymph node, and tumor tissues. VISTA expression on myeloid-derived suppressor cells (MDSC, CD11bhi CD11c Gr1+) and myeloid DCs (CD11b+ CD11c+) was analyzed by flow cytometry. C–E, VISTA mAb treatment altered the TME and improved the effector function of tumor-infiltrating T cells. MB49-bearing mice were treated with 13F3 or control Ig from day 0 until tumors in the control group reached 9–10 mm diameter. Tumors were harvested and total CD45+ TILs were quantified. Tumor-infiltrating DCs were analyzed by flow cytometry for their surface expression of MHCII and CD80 and cytokine production (i.e., IL12p40 and TNF-α). Tumor-infiltrating CD4+ and CD8+ T cells were stimulated with irradiated tumor cells for 20 hours. Cytokine production (i.e., IFN-γ and TNF-α) was analyzed by flow cytometry and quantified. Data are representative of 2–3 independent experiments.

Close modal

Because both B16OVA and MB49 tumors express neoantigens that elicit strong immune responses, we sought to evaluate whether VISTA blockade might impact on the growth of poorly immunogenic tumors, such as the B16BL6 melanoma. VISTA mAb in this model treatment significantly delayed tumor growth (Fig. 3A). Similar to the B16OVA model, reduction of tumor-infiltrating monocytic MDSCs and increase of tumor-infiltrating CD4+ and CD8+ T cells were observed upon VISTA mAb treatment (Fig. 3B). To determine whether VISTA mAb could directly promote T-cell infiltration to tumor tissues, we tracked the tumor-infiltration of TRP1 TCR transgenic CD4+ T cells that are specific for the melanoma antigen tyrosinase-related protein-1 (TRP1), in B16 tumor-bearing mice. Our data show that VISTA mAb treatment significantly enhanced tumor infiltration of TRP1 transgenic CD4+ T cells (Fig. 3C). Phenotypic analysis of tumor-infiltrating polyclonal populations of CD4+ and CD8+ T cells demonstrated heightened activation status upon VISTA blockade, which was evidenced by enhanced Ki67+ cells and heightened frequency of cells bearing a CD44hiCD62Llow surface phenotype (Fig. 3D). When restimulated with BMDCs pulsed with tumor antigens in vitro, TIL CD4+ and CD8+ T cells produced enhanced levels of effector molecules (IFN-γ and/or granzyme B; Fig. 3E). Consistent with the MB49 model, tumor-infiltrating CD11c+ DCs expressed higher level of MHCII and CD80, as well as inflammatory cytokines IL-12 and TNF-α (Fig. 3F). Taken together, these results demonstrated the ability of VISTA mAb to alter the suppressive signature of the TME and promote tumor-specific effector T-cell function, which likely contributed to reduced tumor growth.

Figure 3.

Effects of VISTA mAb treatment on the growth and the tumor microenvironment of B16-BL6 melanoma. A, mice (n = 12 per group) were inoculated with B16BL6 tumor cells (18,000) on the right flank. Mice were treated with anti-VISTA mAb every 2 days, starting from day +2 for the entire duration of the experiment. Tumor growth was measured by a caliper every 2 days. B, tumors were harvested when control tumors reached ∼9–10 mm diameter, which typically corresponded to day19–20 posttumor inoculation. TIL populations, including CD4+ and CD8+ T cells, and MDSCs (CD11b+ CD11C Gr1+) were analyzed by flow cytometry. C, purified TRP1 transgenic CD4+ T cells (25,000) were adoptively transferred into congenic B16-tumor bearing mice that were treated with anti-VISTA mAb or control Ig. The number of tumor-infiltrating TRP1 cells was analyzed day+10 post transfer. D, the proliferative marker (Ki67 expression) and activation status (CD44 and CD62L expression) of tumor-infiltrating polyclonal CD4+ and CD8+ T cells were analyzed. E, the expression level of effector molecules (i.e., IFN-γ and granzyme B) by TIL CD8+ T cells was analyzed after overnight stimulation by antigen-loaded BMDCs. F, expression levels of surface markers (i.e., MHCII and CD80) and cytokine production (i.e., IL12p40 and TNF-α) of tumor-infiltrating CD11C+ DCs were analyzed. Data are representative of 2–3 independent experiments.

Figure 3.

Effects of VISTA mAb treatment on the growth and the tumor microenvironment of B16-BL6 melanoma. A, mice (n = 12 per group) were inoculated with B16BL6 tumor cells (18,000) on the right flank. Mice were treated with anti-VISTA mAb every 2 days, starting from day +2 for the entire duration of the experiment. Tumor growth was measured by a caliper every 2 days. B, tumors were harvested when control tumors reached ∼9–10 mm diameter, which typically corresponded to day19–20 posttumor inoculation. TIL populations, including CD4+ and CD8+ T cells, and MDSCs (CD11b+ CD11C Gr1+) were analyzed by flow cytometry. C, purified TRP1 transgenic CD4+ T cells (25,000) were adoptively transferred into congenic B16-tumor bearing mice that were treated with anti-VISTA mAb or control Ig. The number of tumor-infiltrating TRP1 cells was analyzed day+10 post transfer. D, the proliferative marker (Ki67 expression) and activation status (CD44 and CD62L expression) of tumor-infiltrating polyclonal CD4+ and CD8+ T cells were analyzed. E, the expression level of effector molecules (i.e., IFN-γ and granzyme B) by TIL CD8+ T cells was analyzed after overnight stimulation by antigen-loaded BMDCs. F, expression levels of surface markers (i.e., MHCII and CD80) and cytokine production (i.e., IL12p40 and TNF-α) of tumor-infiltrating CD11C+ DCs were analyzed. Data are representative of 2–3 independent experiments.

Close modal

VISTA regulates the induction of Foxp3+ iTregs from naïve CD4+ T cells as well as the suppressive activity of nTregs

Our previous study showed that VISTA-Ig fusion protein directly suppressed T-cell activation by inhibiting T-cell proliferative responses and cytokine production (19). Our current data show that VISTA-Ig promoted the induction of Foxp3+CD4+ regulatory T cells (iTregs) in the presence of TGF-β in vitro (Fig. 4C). This effect is also seen on human CD4+ T cells treated with human-VISTA-Ig fusion protein in vitro (manuscript submitted). Tregs induced by VISTA-Ig obtained similar suppressive activity in vitro when compared with control iTregs (Fig. 4C). To validate the role of VISTA on promoting the differentiation of iTregs, we examined the effect of VISTA mAb treatment on the induction of tumor-specific iTregs, as previously described (9). Naïve OVA-specific OTII CD4+ T cells (purified from Foxp3GFP reporter mice as CD25CD62Lhi Foxp3GFP) were adoptively transferred into sublethally irradiated host bearing the B16OVA tumor. Mice were treated with either control-Ig or VISTA mAb. The induction of Foxp3GFP+ OTII iTreg cells in the tumor-draining lymph node and within the tumor tissue was examined when tumors reached >8 mm diameter (approximately day 20). As shown in Fig. 4D, VISTA blockade significantly diminished the percentage of Foxp3GFP+ iTregs within the tumor-infiltrating OTII transgenic CD4+ T-cell population (from 61.51% ± 5.71% to 35.75% ± 7.09%). Similar reduction was seen in the tumor-draining lymph node (from 3.51% ± 0.49% to 1.75% ± 0.25%).

Figure 4.

VISTA regulates the suppressive function of Foxp3+CD4+ nTregs, as well as the de novo induction of iTregs. A, VISTA expression on nTreg subsets. nTreg subsets from spleens of 7–8-week-old naïve mice were distinguished based on markers CD62L and ICOS. B, higher levels of VISTA expression on Tregs within the TME, when compared with Tregs from the peripheral LN. C, VISTA-Ig promotes the de novo induction of Foxp3+CD4+ iTregs. Naïve murine CD4+ T cells were stimulated with plate-bound αCD3 (2.5 μg/mL) together with either control-Ig or VISTA-Ig (5 μg/mL). Exogenous TGF-β (2.5 μg/mL) was added as indicated. Cells were examined for the induction of Foxp3 after 72 hours. Foxp3GFP+ iTregs were sorted on day+5 and their suppressive activity was examined as described in Materials and Methods. D, VISTA mAb treatment diminished tumor-mediated induction of iTregs. Congenic naïve OTII CD4+ T cells were adoptively transferred into B16OVA tumor-bearing mice (n = 14 for control, n = 17 for 13F3 group), which were treated with control-Ig or VISTA mab 13F3 every 2 days starting from day 0. Mice were analyzed around day 20 when tumor size in control group reached 9–10 mm diameter. When necessary, small size tumors (diameter less than 5 mm) were pooled within the group to obtain sufficient cell number for analysis. Conversion of naïve OTII to Foxp3+ iTregs in tumor-draining lymph node and within the tumor tissue was quantified and shown as the percentages of Foxp3+ cells among gated OTII cells. E, the suppressive activity of Treg subsets is partially impaired by VISTA mAb. Foxp3+CD4+ nTreg subsets were sorted from spleen based on expression level of ICOS and CD62L. These nTregs were cocultured with naïve CD4+ T cells in indicated ratios, in the presence of VISTA mAb or control IgG. The proliferation of T cells was measured by tritium incorporation. Data are representative of 2–3 independent experiments.

Figure 4.

VISTA regulates the suppressive function of Foxp3+CD4+ nTregs, as well as the de novo induction of iTregs. A, VISTA expression on nTreg subsets. nTreg subsets from spleens of 7–8-week-old naïve mice were distinguished based on markers CD62L and ICOS. B, higher levels of VISTA expression on Tregs within the TME, when compared with Tregs from the peripheral LN. C, VISTA-Ig promotes the de novo induction of Foxp3+CD4+ iTregs. Naïve murine CD4+ T cells were stimulated with plate-bound αCD3 (2.5 μg/mL) together with either control-Ig or VISTA-Ig (5 μg/mL). Exogenous TGF-β (2.5 μg/mL) was added as indicated. Cells were examined for the induction of Foxp3 after 72 hours. Foxp3GFP+ iTregs were sorted on day+5 and their suppressive activity was examined as described in Materials and Methods. D, VISTA mAb treatment diminished tumor-mediated induction of iTregs. Congenic naïve OTII CD4+ T cells were adoptively transferred into B16OVA tumor-bearing mice (n = 14 for control, n = 17 for 13F3 group), which were treated with control-Ig or VISTA mab 13F3 every 2 days starting from day 0. Mice were analyzed around day 20 when tumor size in control group reached 9–10 mm diameter. When necessary, small size tumors (diameter less than 5 mm) were pooled within the group to obtain sufficient cell number for analysis. Conversion of naïve OTII to Foxp3+ iTregs in tumor-draining lymph node and within the tumor tissue was quantified and shown as the percentages of Foxp3+ cells among gated OTII cells. E, the suppressive activity of Treg subsets is partially impaired by VISTA mAb. Foxp3+CD4+ nTreg subsets were sorted from spleen based on expression level of ICOS and CD62L. These nTregs were cocultured with naïve CD4+ T cells in indicated ratios, in the presence of VISTA mAb or control IgG. The proliferation of T cells was measured by tritium incorporation. Data are representative of 2–3 independent experiments.

Close modal

VISTA expression level on tumor-infiltrating Tregs is higher than Tregs from peripheral lymph nodes (Fig. 4B), indicating that VISTA expressed on Tregs within the TME might play a role in suppressing tumor-specific immunity. Studies were developed to functionally assess whether VISTA mAb-mediated blockade impaired the suppressive function of Tregs. Thymus-derived natural Tregs (nTregs) contain different subsets that are distinguished by surface markers. Our analysis show that VISTA is more highly expressed on CD62L and ICOS Treg subsets, when compared with CD62L+ and ICOS+ Treg subsets (Fig. 4A; refs. 28, 29). No difference of VISTA expression was observed on other Treg subsets based on surface markers such as CD25, GITR, CD73, Folate receptor 4, and IL-7 receptor (data not shown). Treg subsets were sorted from naïve mice based on markers ICOS and CD62L and tested for their suppressive activity in vitro. VISTA mAb enhanced naive target T-cell proliferation in the presence of all the Treg subsets tested. This data indicates that VISTA blockade might directly impair the suppressive activity of Tregs. Notably, VISTA mAb also enhanced T-cell proliferation in the absence of Tregs, indicating an alternative possibility that VISTA blockade might enhance the resistance of naïve T cells to Treg-mediated suppression. Either mechanism might ultimately contribute to the therapeutic efficacy of VISTA mAb-mediated blockade in tumor models.

VISTA mAb treatment suppresses tumor progression in a genetic model of melanoma

Encouraged by results seen in the transplantable tumor models, we tested the therapeutic impact of VISTA mAb administration in an inducible melanoma model. This model relies on the interbreeding of three transgenic mouse lines (30): (i) BrafCA, which carries a conditional BrafV600E allele. (ii) Ptenlox5, which carries a conditional allele of Pten, permitting Cre-mediated deletion. (iii) Tyr::Cre/ERT2, which carries a 4-hydroxytamoxifen (4-OHT)-inducible CreERT2 allele, allowing melanocyte-specific inducible expression of Cre. The triple mutant mice, B6.Cg-BrafCA/+ Ptenlox5/lox5 Tg (Tyr::Cre/ERT2), develop pigmented skin lesions upon topical treatment of 4-OHT within 21 days, which quickly progress to malignant melanoma (Fig. 5A). Analysis of tumor-infiltrating leukocytes reveals high levels of VISTA expression on tumor-associated myeloid cells (Fig. 5B). No VISTA expression was observed on tumor cells, whereas some level of PD-L1 expression was detected (Fig. 5C). Importantly, VISTA mAb treatment significantly delayed tumor progression (Fig. 5A). Further analysis of TIL populations demonstrated similar alterations of TME as shown in the transplantable B16 tumor models, including increased tumor infiltration of T cells, decreased tumor-infiltration of myeloid cells, and enhanced IFN-γ production of tumor-infiltrating CD8+ T lymphocytes (Fig. 5D–F).

Figure 5.

VISTA monoclonal antibody treatment suppressed tumor growth and altered the tumor microenvironment in the PTEN/BRAF inducible melanoma model. Tumors were induced by application of tamoxifen on the back skin. Mice were treated with VISTA mAb 13F3 (n = 10) or control hamster Ig (n = 8) every 2 days throughout the duration of the experiment. A, representative pictures of melanomas grown on mice, as well as dissected tumors are shown. Because these inducible tumors grew with irregular and diverse height, which is in contrast to the relatively uniform shape and height of transplantable B16 tumors, tumor size was measured with a caliper and is shown as mm3 (length × width × height). B, high levels of VISTA expression on tumor-infiltrating myeloid cells. C, expression of VISTA and PD-L1 on CD45+ TILs and CD45 tumor cells is shown. D–F, analysis of tumor-infiltrating CD8+ T cells (D), MDSCs (CD11bhi CD11c Gr1+; E), and cytokine (IFN-γ) production of tumor-infiltrating CD8+ T cells upon in vitro stimulation with anti-CD3 (F) is shown. Data are representative of 2–3 independent experiments.

Figure 5.

VISTA monoclonal antibody treatment suppressed tumor growth and altered the tumor microenvironment in the PTEN/BRAF inducible melanoma model. Tumors were induced by application of tamoxifen on the back skin. Mice were treated with VISTA mAb 13F3 (n = 10) or control hamster Ig (n = 8) every 2 days throughout the duration of the experiment. A, representative pictures of melanomas grown on mice, as well as dissected tumors are shown. Because these inducible tumors grew with irregular and diverse height, which is in contrast to the relatively uniform shape and height of transplantable B16 tumors, tumor size was measured with a caliper and is shown as mm3 (length × width × height). B, high levels of VISTA expression on tumor-infiltrating myeloid cells. C, expression of VISTA and PD-L1 on CD45+ TILs and CD45 tumor cells is shown. D–F, analysis of tumor-infiltrating CD8+ T cells (D), MDSCs (CD11bhi CD11c Gr1+; E), and cytokine (IFN-γ) production of tumor-infiltrating CD8+ T cells upon in vitro stimulation with anti-CD3 (F) is shown. Data are representative of 2–3 independent experiments.

Close modal

VISTA mAb synergizes with a tumor vaccine to achieve optimal therapeutic outcome

Although VISTA mAb as a single-agent therapy delayed tumor progression, it was not curative under the conditions tested. In an attempt to increase the magnitude of the tumor-specific immune response, a peptide-based cancer vaccine was used in combination with VISTA blockade. Based on previous melanoma vaccine studies from our lab (R.J. Noelle) and others (31–34), we applied a modified vaccine platform containing the agonistic CD40 antibody FGK, TLR agonists, and tumor antigen peptides. Both a MHCI-restricted mutant TRP2 peptide and a MHCII-restricted TRP1 peptide were incorporated (22–24). Early therapeutic intervention on 2-day tumors using a combinatorial therapy of αVISTA mAb and a single dose of peptide vaccine effectively eradicated tumors in a majority of mice, whereas either reagent alone only transiently impaired tumor growth without significant survival benefit (Fig. 6A). Combinatorial treatment of 7-day established tumors (with average tumor diameter ∼3mm) showed significant suppression of tumor growth and led to approximately 30% long-term survival, whereas monotherapy had little effect (Fig. 6B). A prime-boost vaccine regimen was applied on day+7 and +14 for treating 7-day established tumors, and showed better long-term survival (from 10% to 30%) than a single vaccine dose in this setting (Fig. S6). Tumor-infiltrating T cells showed synergistically enhanced responses against immunizing peptide, indicating that the development of T-cell–mediated immune responses might be critical for the efficacy of the combination therapy (Fig. 6C). Additional evidence supporting this hypothesis is provided by the loss of therapeutic efficacy upon predepletion of CD8+ T cells, or when treating immune-deficient tumor-bearing Rag−/− hosts (Supplementary Fig. S7). This data is consistent with the mechanisms of action associated with CD40/TLR-based vaccines (31–34). A second tumor challenge of survivors at day +60 post-treatment showed delayed tumor growth (8/12) or no tumor growth (4/12), indicating that certain levels of T-cell memory was developed during the combination therapy (Fig. 6D). The protection rate for the rechallenge did not improve regardless whether a second boost vaccine was introduced during the primary tumor challenge (data not shown). These studies set the stage for extensive efforts in the future to define successful combination therapies with αVISTA mAb as a platform.

Figure 6.

VISTA mAb synergized with tumor vaccine to impair the growth of established B16-BL6 tumors. Naïve mice (n = 13 per group for A and n = 8–10 per group for B) were inoculated with B16-BL6 tumor cells (18,000) on the right flank. On day+2 (A) or day+7 (B) postinoculations, mice were treated with peptide vaccine complex (1 dose) or anti-VISTA mAb 13F3 (continuous treatment for the entire duration of the experiment) or the combination as indicated. C, to determine whether the combination therapy induced optimal T-cell responses when compared with single treatment, tumor-bearing mice were treated on day+7 with vaccine or 13F3 or the combination as indicated. Tumor-infiltrating lymphocytes were harvested ∼day+21, restimulated with the immunizing peptides TRP1/deltaV-TRP2, and analyzed for their cytokine production (i.e., IFN-γ) by flow cytometry as described in Materials and Methods. D, mice that survived the first B16BL6 tumor challenge (n = 12) after combination treatment were rechallenged with B16BL6 cells (18,000) on the same flank on day+60 post primary tumor rejection. Naïve mice were used as the control group. Tumor size was measured every 2–3 days with a caliper. Data are representative of 2–3 independent experiments.

Figure 6.

VISTA mAb synergized with tumor vaccine to impair the growth of established B16-BL6 tumors. Naïve mice (n = 13 per group for A and n = 8–10 per group for B) were inoculated with B16-BL6 tumor cells (18,000) on the right flank. On day+2 (A) or day+7 (B) postinoculations, mice were treated with peptide vaccine complex (1 dose) or anti-VISTA mAb 13F3 (continuous treatment for the entire duration of the experiment) or the combination as indicated. C, to determine whether the combination therapy induced optimal T-cell responses when compared with single treatment, tumor-bearing mice were treated on day+7 with vaccine or 13F3 or the combination as indicated. Tumor-infiltrating lymphocytes were harvested ∼day+21, restimulated with the immunizing peptides TRP1/deltaV-TRP2, and analyzed for their cytokine production (i.e., IFN-γ) by flow cytometry as described in Materials and Methods. D, mice that survived the first B16BL6 tumor challenge (n = 12) after combination treatment were rechallenged with B16BL6 cells (18,000) on the same flank on day+60 post primary tumor rejection. Naïve mice were used as the control group. Tumor size was measured every 2–3 days with a caliper. Data are representative of 2–3 independent experiments.

Close modal

Our study introduces a new negative immune-checkpoint regulator, VISTA, whose expression within the TME plays a critical role in regulating protective immunity to cancer. Our data show that αVISTA monotherapy impairs tumor growth in multiple tumor models. We have dissected the multiple mechanisms whereby VISTA mAb-mediated blockade enhances antitumor immune responses.

First, VISTA mAb enhanced tumor-specific T-cell response, both in the periphery and within the TME. VISTA is constitutively highly expressed on myeloid cells, with even higher densities observed within the TME. This data suggests that VISTA may be abundantly present within the TME of any solid tumor types that are infiltrated with myeloid cells and T cells, indicating a broad clinical applicability for VISTA-blockade therapy. The high expression of VISTA on myeloid cells within the TME suggests that VISTA might directly suppress tumor-infiltrating effector T cells. This hypothesis is supported by enhanced proliferation, activation, and effector function of tumor-infiltrating T cells in VISTA mAb-treated mice (Figs. 1–3). We have not detected VISTA expression on tumor cells investigated. As such, we predict that VISTA targeted immunotherapy will be effective independent of its expression on tumors. Similarly restricted hematopoietic expression of human VISTA has been observed in human melanoma, colorectal cancer, and lung cancer (data not shown). In contrast, PD-L1 is known to be expressed at high levels on nonhematopoietic tumor cells (Fig. 1; refs. 2, 35). It is therefore clinically relevant that VISTA mAb demonstrated efficacy despite the high levels of PD-L1 within the TME. Our future studies will compare the efficacy of VISTA-blockade with PD1 blockade and define synergistic treatment regimens for the remission of established tumors.

Second, VISTA blockade impaired the suppressive effect of natural Tregs and the differentiation of tumor-specific iTregs (Fig. 4). It remains to be determined whether VISTA expression on Tregs directly contributes to their suppressive function, even though VISTA neutralization in vitro failed to show any direct effect on Treg proliferation, apoptosis, and Foxp3 stability on in vitro cultured Tregs (Supplementary Fig. S4). Alternatively, it is possible that VISTA neutralization might enhance naïve T-cell proliferation, making them more resistant to Treg-mediated suppression. Future studies will utilize VISTAKO mice to tease out the underlying mechanisms whereby VISTA regulates the suppressive function of Tregs. In this context, both CTLA-4 and PD-1 impact on either the suppressive function and/or the peripheral induction of Foxp3+ Tregs (9, 36–38).

Third, in the B16 melanoma model, VISTA blockade altered the suppressive cellular signature of the TME, by reducing the tumor-infiltrating monocytic MDSCs while increasing the frequency of infiltrating effector T cells. Because there is no apparent deletion of both myeloid lineages and T-cell lineages in the periphery upon VISTA mAb treatment (data not shown), such alterations within the tumor tissue likely reflect either the impaired migration of monocytic MDSCs into tumor tissues, or impaired infiltration of immature myeloid progenitor cells, which might differentiate into MDSCs at tumor site (39, 40). In contrast to melanoma, VISTA blockade did not significantly alter the infiltration of the granuolytic MDSC population within the MB49 bladder tumors. This intriguing result might indicate different mechanisms whereby VISTA regulates monocytic and granuolytic myelopoiesis during tumor development.

In addition to affecting MDSCs, VISTA blockade directly enhanced the migration of tumor-specific effector T cells (i.e., TRP1 Tg CD4+ T cells) into tumor tissue (Fig. 3). Furthermore, VISTA blockade enhanced the immune-stimulatory phenotype of TIL DCs, which showed higher expression levels of MHCII and CD80, and cytokine production (i.e., IL-12 and TNF-α; Figs. 2 and 3). Collectively, these multiple effects of VISTA mAb treatment facilitate the establishment of an immune-stimulatory TME, which lead to enhanced antitumor immunity.

It is clear that combination immunotherapy for cancer using multiple biologics will be critical for improving the therapeutic outcome. In addition to the monotherapeutic targeting of negative checkpoint regulators, combining multiple checkpoint blockades, or combining checkpoint blockade with chemotherapy or cancer vaccines is moving into clinical trials. Some examples of such combinatorial approaches include CTLA4 blockade combined with GVax or Flt3vax (3, 41), CTLA4 blockade combined with PD-L1/PD1 blockade (42, 43), PD1 blockade combined with vaccine (44, 45). Our initial studies explore a combination regimen using VISTA blockade together with a peptide vaccine (22–24, 33). Our data show promising synergistic effects for treating both 2-day and 7-day established tumors. Future studies will focus on elucidating the cellular and molecular mechanisms of such synergy, as well as determining the limitations of this combination platform and strategies for improvement. For example, the use of defined peptides might lead to loss-of-antigen escape variants (46). The delivery of vaccine component might be optimized through the use of nanoparticles or other particulate formulations or TLR-peptide conjugates (47–49). Additional immune-suppressive pathways (i.e., other immune checkpoint pathways such as PD-1, and immune suppressive cytokines) might be blocked to achieve better long-term tumor-specific memory T-cell responses. Furthermore, the immunogenicity and the FcR binding activity of the VISTA mAb might be critical limiting factors for achieving optimal target neutralization and therapeutic efficacy, thus warrant future studies (50, 51).

Recent breakthroughs using CTLA-4 and PD-1 checkpoint blockade have reinvigorated the field of cancer immunotherapy (2, 35). Multiple checkpoints control the development of antitumor immunity, each with their own unique signature and mechanisms. Our studies provide compelling evidence supporting the role of VISTA as a negative immune checkpoint regulator that controls immunity against cancer. Taken together with the findings that VISTA expression and function on human leukocytes and within the human tumors have recapitulated the murine data (20), we present VISTA as a promising new target for cancer immunotherapy, either as a single target or in combination with other immunotherapeutic strategies.

L. Wang is a consultant/advisory board member of Immunext. J.L. Lines is a consultant/advisory board member of Immunext. M. Day is a research associate and has ownership interest (including patents) in ImmuNext. R.J. Noelle is CSO, has commercial research grant, other commercial research support, ownership interest (including patents), and is a consultant/advisory board member of ImmuNext. No potential conflicts of interest were disclosed by the other authors.

Conception and design: L. Wang, R.J. Noelle

Development of methodology: L. Wang, W. Chen

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): L. Wang, I. LeMercier, J.L. Lines, P. Sergent

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): L. Wang, I. LeMercier, W. Chen, J. Li, R.J. Noelle

Writing, review, and/or revision of the manuscript: L. Wang, P. Sergent, R.J. Noelle

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): L. Wang, M. Day, J. Li, P. Sergent

The authors thank Dr. Alexander Rudensky (University of Washington School of Medicine, Seattle, WA) for providing the Foxp3-GFP reporter mice, Dr. Marcus Bosenberg (Yale School of Medicine) for providing the B6.Cg-BrafCA/+ Ptenlox5/lox5 Tg (Tyr::Cre/ERT2)-inducible melanoma model, Dr. Mary Jo Turk (Geisel School of Medicine) for maintaining the inducible melanoma model and providing insightful discussions, and Dartlab Flow Cytometry Core Facility for technical support.

This work is supported by NIH grant CA164225 (L. Wang), Melanoma Research Foundation Career Development Award (L. Wang), Hitchcock Foundation Research grant (L. Wang), AI048667 (to R.J. Noelle), Wellcome Trust (R.J. Noelle), and the Medical Research Council Centre for Transplantation and Biomedical Research Center at King's College London (R.J. Noelle).

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.
Zou
W
,
Chen
L
. 
Inhibitory B7-family molecules in the tumour microenvironment
.
Nat Rev Immunol
2008
;
8
:
467
77
.
2.
Pardoll
DM
. 
The blockade of immune checkpoints in cancer immunotherapy
.
Nat Rev Cancer
2012
;
12
:
252
64
.
3.
van Elsas
A
,
Hurwitz
AA
,
Allison
JP
. 
Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation
.
J Exp Med
1999
;
190
:
355
66
.
4.
Hoos
A
,
Ibrahim
R
,
Korman
A
,
Abdallah
K
,
Berman
D
,
Shahabi
V
, et al
Development of ipilimumab: contribution to a new paradigm for cancer immunotherapy
.
Semin Oncol
2010
;
37
:
533
46
.
5.
Calabro
L
,
Danielli
R
,
Sigalotti
L
,
Maio
M
. 
Clinical studies with anti-CTLA-4 antibodies in non-melanoma indications
.
Semin Oncol
2010
;
37
:
460
7
.
6.
Peggs
KS
,
Quezada
SA
. 
Ipilimumab: attenuation of an inhibitory immune checkpoint improves survival in metastatic melanoma
.
Expert Rev Anticancer Ther
2010
;
10
:
1697
701
.
7.
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
.
8.
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
.
9.
Wang
L
,
Pino-Lagos
K
,
de Vries
VC
,
Guleria
I
,
Sayegh
MH
,
Noelle
RJ
. 
Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+CD4+ regulatory T cells
.
Proc Natl Acad Sci U S A
2008
;
105
:
9331
6
.
10.
Blank
C
,
Brown
I
,
Peterson
AC
,
Spiotto
M
,
Iwai
Y
,
Honjo
T
, et al
PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells
.
Cancer Res
2004
;
64
:
1140
5
.
11.
Blank
C
,
Gajewski
TF
,
Mackensen
A
. 
Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: implications for tumor immunotherapy
.
Cancer Immunol Immunother
2005
;
54
:
307
14
.
12.
Iwai
Y
,
Ishida
M
,
Tanaka
Y
,
Okazaki
T
,
Honjo
T
,
Minato
N
. 
Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade
.
Proc Natl Acad Sci U S A
2002
;
99
:
12293
7
.
13.
Geng
H
,
Zhang
GM
,
Xiao
H
,
Yuan
Y
,
Li
D
,
Zhang
H
, et al
HSP70 vaccine in combination with gene therapy with plasmid DNA encoding sPD-1 overcomes immune resistance and suppresses the progression of pulmonary metastatic melanoma
.
Int J Cancer
2006
;
118
:
2657
64
.
14.
Hirano
F
,
Kaneko
K
,
Tamura
H
,
Dong
H
,
Wang
S
,
Ichikawa
M
, et al
Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity
.
Cancer Res
2005
;
65
:
1089
96
.
15.
Curiel
TJ
,
Wei
S
,
Dong
H
,
Alvarez
X
,
Cheng
P
,
Mottram
P
, et al
Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity
.
Nat Med
2003
;
9
:
562
7
.
16.
Brahmer
JR
,
Drake
CG
,
Wollner
I
,
Powderly
JD
,
Picus
J
,
Sharfman
WH
, et al
Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates
.
J Clin Oncol
2010
;
28
:
3167
75
.
17.
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
.
18.
Brahmer
JR
,
Tykodi
SS
,
Chow
LQ
,
Hwu
WJ
,
Topalian
SL
,
Hwu
P
, et al
Safety and activity of anti-PD-L1 antibody in patients with advanced cancer
.
N Engl J Med 2012
;
366
:
2455
65
.
19.
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
.
20.
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
.
21.
Fontenot
JD
,
Rasmussen
JP
,
Williams
LM
,
Dooley
JL
,
Farr
AG
,
Rudensky
AY
. 
Regulatory T cell lineage specification by the forkhead transcription factor foxp3
.
Immunity
2005
;
22
:
329
41
.
22.
McWilliams
JA
,
McGurran
SM
,
Dow
SW
,
Slansky
JE
,
Kedl
RM
. 
A modified tyrosinase-related protein 2 epitope generates high-affinity tumor-specific T cells but does not mediate therapeutic efficacy in an intradermal tumor model
.
J Immunol
2006
;
177
:
155
61
.
23.
Ahonen
CL
,
Wasiuk
A
,
Fuse
S
,
Turk
MJ
,
Ernstoff
MS
,
Suriawinata
AA
, et al
Enhanced efficacy and reduced toxicity of multifactorial adjuvants compared with unitary adjuvants as cancer vaccines
.
Blood
2008
;
111
:
3116
25
.
24.
Muranski
P
,
Boni
A
,
Antony
PA
,
Cassard
L
,
Irvine
KR
,
Kaiser
A
, et al
Tumor-specific Th17-polarized cells eradicate large established melanoma
.
Blood
2008
;
112
:
362
73
.
25.
Mantovani
A
,
Allavena
P
,
Sica
A
,
Balkwill
F
. 
Cancer-related inflammation
.
Nature
2008
;
454
:
436
44
.
26.
Topalian
SL
,
Weiner
GJ
,
Pardoll
DM
. 
Cancer immunotherapy comes of age
.
J Clin Oncol
2011
;
29
:
4828
36
.
27.
Wasiuk
A
,
Dalton
DK
,
Schpero
WL
,
Stan
RV
,
Conejo-Garcia
JR
,
Noelle
RJ
. 
Mast cells impair the development of protective anti-tumor immunity
.
Cancer Immunol Immunother
2012
;
61
:
2273
82
.
28.
Fu
S
,
Yopp
AC
,
Mao
X
,
Chen
D
,
Zhang
N
,
Chen
D
, et al
CD4+ CD25+ CD62+ T-regulatory cell subset has optimal suppressive and proliferative potential
.
Am J Transplant
2004
;
4
:
65
78
.
29.
Ito
T
,
Hanabuchi
S
,
Wang
YH
,
Park
WR
,
Arima
K
,
Bover
L
, et al
Two functional subsets of FOXP3+ regulatory T cells in human thymus and periphery
.
Immunity
2008
;
28
:
870
80
.
30.
Dankort
D
,
Curley
DP
,
Cartlidge
RA
,
Nelson
B
,
Karnezis
AN
,
Damsky
WE
 Jr
, et al
Braf(V600E) cooperates with Pten loss to induce metastatic melanoma
.
Nat Genet
2009
;
41
:
544
52
.
31.
Davila
E
,
Kennedy
R
,
Celis
E
. 
Generation of antitumor immunity by cytotoxic T lymphocyte epitope peptide vaccination, CpG-oligodeoxynucleotide adjuvant, and CTLA-4 blockade
.
Cancer Res
2003
;
63
:
3281
8
.
32.
Ahonen
CL
,
Doxsee
CL
,
McGurran
SM
,
Riter
TR
,
Wade
WF
,
Barth
RJ
, et al
Combined TLR and CD40 triggering induces potent CD8+ T cell expansion with variable dependence on type I IFN
.
J Exp Med
2004
;
199
:
775
84
.
33.
Cho
HI
,
Celis
E
. 
Optimized peptide vaccines eliciting extensive CD8 T-cell responses with therapeutic antitumor effects
.
Cancer Res
2009
;
69
:
9012
9
.
34.
Cho
HI
,
Lee
YR
,
Celis
E
. 
Interferon gamma limits the effectiveness of melanoma peptide vaccines
.
Blood
2011
;
117
:
135
44
.
35.
Pardoll
D
,
Drake
C
. 
Immunotherapy earns its spot in the ranks of cancer therapy
.
J Exp Med
2012
;
209
:
201
9
.
36.
Greenwald
RJ
,
Freeman
GJ
,
Sharpe
AH
. 
The B7 family revisited
.
Annu Rev Immunol
2005
;
23
:
515
48
.
37.
Teft
WA
,
Kirchhof
MG
,
Madrenas
J
. 
A molecular perspective of CTLA-4 function
.
Annu Rev Immunol
2006
;
24
:
65
97
.
38.
Francisco
LM
,
Salinas
VH
,
Brown
KE
,
Vanguri
VK
,
Freeman
GJ
,
Kuchroo
VK
, et al
PD-L1 regulates the development, maintenance, and function of induced regulatory T cells
.
J Exp Med
2009
;
206
:
3015
29
.
39.
Gabrilovich
DI
,
Nagaraj
S
. 
Myeloid-derived suppressor cells as regulators of the immune system
.
Nat Rev Immunol
2009
;
9
:
162
74
.
40.
Gabrilovich
DI
,
Ostrand-Rosenberg
S
,
Bronte
V
. 
Coordinated regulation of myeloid cells by tumours
.
Nat Rev Immunol
2012
;
12
:
253
68
.
41.
Curran
MA
,
Allison
JP
. 
Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors
.
Cancer Res
2009
;
69
:
7747
55
.
42.
Curran
MA
,
Montalvo
W
,
Yagita
H
,
Allison
JP
. 
PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors
.
Proc Natl Acad Sci U S A
2010
;
107
:
4275
80
.
43.
Wolchok
JD
,
Kluger
H
,
Callahan
MK
,
Postow
MA
,
Rizvi
NA
,
Lesokhin
AM
, et al
Nivolumab plus ipilimumab in advanced melanoma
.
N Engl J Med
2013
;
369
:
122
33
.
44.
Li
B
,
VanRoey
M
,
Wang
C
,
Chen
TH
,
Korman
A
,
Jooss
K
. 
Anti-programmed death-1 synergizes with granulocyte macrophage colony-stimulating factor–secreting tumor cell immunotherapy providing therapeutic benefit to mice with established tumors
.
Clin Cancer Res
2009
;
15
:
1623
34
.
45.
Pilon-Thomas
S
,
Mackay
A
,
Vohra
N
,
Mule
JJ
. 
Blockade of programmed death ligand 1 enhances the therapeutic efficacy of combination immunotherapy against melanoma
.
J Immunol
2010
;
184
:
3442
9
.
46.
Sanchez-Perez
L
,
Kottke
T
,
Diaz
RM
,
Ahmed
A
,
Thompson
J
,
Chong
H
, et al
Potent selection of antigen loss variants of B16 melanoma following inflammatory killing of melanocytes in vivo
.
Cancer Res
2005
;
65
:
2009
17
.
47.
Hafner
AM
,
Corthesy
B
,
Merkle
HP
. 
Particulate formulations for the delivery of poly(I:C) as vaccine adjuvant
.
Adv Drug Deliv Rev
2013
;
65
:
1386
99
.
48.
Zom
GG
,
Khan
S
,
Filippov
DV
,
Ossendorp
F
. 
TLR ligand-peptide conjugate vaccines: toward clinical application
.
Adv Immunol
2012
;
114
:
177
201
.
49.
Silva
JM
,
Videira
M
,
Gaspar
R
,
Preat
V
,
Florindo
HF
. 
Immune system targeting by biodegradable nanoparticles for cancer vaccines
.
J Control Release
2013
;
168
:
179
99
.
50.
Bulliard
Y
,
Jolicoeur
R
,
Windman
M
,
Rue
SM
,
Ettenberg
S
,
Knee
DA
, et al
Activating Fc gamma receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies
.
J Exp Med
2013
;
210
:
1685
93
.
51.
Simpson
TR
,
Li
F
,
Montalvo-Ortiz
W
,
Sepulveda
MA
,
Bergerhoff
K
,
Arce
F
, et al
Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma
.
J Exp Med
2013
;
210
:
1695
710
.