Immune checkpoint inhibitors have revolutionized the treatment of patients with advanced-stage metastatic melanoma, as well as patients with many other solid cancers, yielding long-lasting responses and improved survival. However, a subset of patients who initially respond to immunotherapy, later relapse and develop therapy resistance (termed “acquired resistance”), whereas others do not respond at all (termed “primary resistance”). Primary and acquired resistance are key clinical barriers to further improving outcomes of patients with metastatic melanoma, and the known mechanisms underlying each involves various components of the cancer immune cycle, and interactions between multiple signaling molecules and pathways. Due to this complexity, current knowledge on resistance mechanisms is still incomplete. Overcoming therapy resistance requires a thorough understanding of the mechanisms underlying immune evasion by tumors. In this review, we explore the mechanisms of primary and acquired resistance to immunotherapy in melanoma and detail potential therapeutic strategies to prevent and overcome them. Clin Cancer Res; 24(6); 1260–70. ©2017 AACR.

Immune checkpoint inhibitors have revolutionized the treatment of advanced melanoma (1–5) and have significant clinical activity across an increasing range of many other solid malignancies, including non–small cell lung cancer (6, 7), renal cell carcinoma (8), head and neck cancer (9), Merkel cell carcinoma (10), and bladder cancer (11, 12). Understanding the biology behind response and resistance to immune checkpoint blockade is critical to further improving outcomes of patients with metastatic melanoma.

The first immune checkpoint to be clinically targeted, the cytotoxic T-lymphocyte antigen 4 (CTLA-4), is expressed on the surface of activated T cells and binds to its ligands, B7-1 and B7-2, on antigen-presenting cells (APC), resulting in the transmission of inhibitory signals to T cells. In patients with metastatic melanoma, phase III clinical trials of ipilimumab, a fully human IgG1 monoclonal antibody inhibiting CTLA-4, demonstrated a significant improvement in progression-free survival (PFS) and overall survival (OS) when compared with a gp100 vaccine (13) or dacarbazine chemotherapy (4).

Drugs targeting the programmed cell death receptor 1 (PD-1, PDCD1) showed a further increase in response rates, PFS (2), and OS (14–16) compared with anti–CTLA-4 blockade. PD-1 is also expressed on the surface of activated T cells and binds to the programmed cell death ligand 1 (PD-L1, CD274) to negatively regulate T-cell activation and differentiation. PD-L1 is constitutively expressed by T cells, macrophages, and dendritic cells (DC), as well as by some tumor cells including melanoma (17). Follow-up data from phase I clinical trials of the fully human IgG4 monoclonal antibody, nivolumab, showed a median OS of 17.3 months, with a 5 year OS rate of 34% (18). In a phase III study of nivolumab versus dacarbazine in patients with BRAF wild-type metastatic melanoma, the median OS was not reached for nivolumab at the most recent analysis, versus 11.2 months for dacarbazine [hazard ratio (HR), 0.43, P < 0.001], and the 1- and 2-year OS rates were 73% and 58%, respectively, for nivolumab (1, 14). Pembrolizumab, a humanized IgG4 monoclonal antibody against PD-1, also showed 1-, 2-, and 3-year OS rates of 67%, 50%, and 40%, respectively, in a phase I trial of ipilimumab-treated and ipilimumab-naïve patients with advanced melanoma (3). Furthermore, in a phase III trial of pembrolizumab versus ipilimumab, the 2-year OS rates were 55% versus 43%, respectively (5, 15).

More recently, combined anti–CTLA-4 and anti–PD-1 immunotherapies have shown improved response rates and clinical outcomes in comparison to ipilimumab monotherapy (the study was not powered to compare the two nivolumab treating arms: nivolumab plus ipilimumab and nivolumab alone). A phase III study showed an increase in the median PFS of patients treated with nivolumab and ipilimumab (11.5 months; HR, 0.42, P < 0.001) and nivolumab alone (6.9 months; HR, 0.57, P < 0.001) compared with ipilimumab alone (2.9 months; ref. 2). At a minimum follow-up of 28 months, the median OS had not been reached in the combination or nivolumab-alone groups and was 20 months for ipilimumab alone [HR: combination vs. ipilimumab, 0.55 (P < 0.0001); nivolumab vs. ipilimumab, 0.63 (P < 0.0001); ref. 16]. The two-year OS rates were 64%, 59%, and 45% in the combination, nivolumab, and ipilimumab groups, respectively (16).

The results of these clinical trials highlight the significant impact immunotherapies have had on the clinical management of patients with advanced-stage metastatic melanoma. However, although approximately 35% to 60% of patients have a RECIST response (10%–12% a complete response) to anti–PD-1-based immunotherapy (2, 14, 15), 40% to 65% have shown minimal or no RECIST response at the outset, and 43% of responders develop acquired resistance by 3 years (3). The underlying mechanisms driving these variations in response are not yet well understood. For an immunotherapy to elicit an efficient antitumor immune response, the cancer immune cycle must be initiated and the subsequent steps successfully completed. This involves efficient (i) antigen presentation and T-cell activation, (ii) T-cell trafficking and tumor infiltration, and (iii) T-cell killing activity within the tumor microenvironment (Fig. 1). Studies examining possible predictive biomarkers of response to immunotherapy have reported a higher density of preexisting cytotoxic T lymphocytes in tumor biopsies of patients who displayed a greater response to anti–PD-1/PD-L1 immunotherapy (19–21), and more significantly, an increased influx of T cells and PD-L1+ macrophages early during treatment (22). In this review, we discuss the different forms of immunotherapy resistance, the mechanisms by which tumors evade the immune system, and strategies to overcome or prevent resistance in the future.

Figure 1.

The cancer immune cycle. The induction of an effective antitumor immune response requires successful (i) antigen presentation and T-cell activation, (ii) T-cell trafficking and tumor infiltration, and (iii) T-cell killing activity within the tumor microenvironment. Various immune escape mechanisms present at each of these stages can result in primary or acquired resistance to immunotherapy. Potential therapeutic strategies can be used at each stage to overcome immunotherapy resistance. A2AR, A2A receptor; B2M, beta-2-microglobulin; HDAC, histone deacetylase; JAK1/JAK2, janus kinases 1 and 2; IDO, indoleamine 2,3-dioxygenase; IPRES, innate anti–PD-1 resistance signature; LAG-3, lymphocyte activation gene 3; TIM-3, T-cell immunoglobulin and mucin domain 3; Tregs, regulatory T cells; VEGF, vascular endothelial growth factor.

Figure 1.

The cancer immune cycle. The induction of an effective antitumor immune response requires successful (i) antigen presentation and T-cell activation, (ii) T-cell trafficking and tumor infiltration, and (iii) T-cell killing activity within the tumor microenvironment. Various immune escape mechanisms present at each of these stages can result in primary or acquired resistance to immunotherapy. Potential therapeutic strategies can be used at each stage to overcome immunotherapy resistance. A2AR, A2A receptor; B2M, beta-2-microglobulin; HDAC, histone deacetylase; JAK1/JAK2, janus kinases 1 and 2; IDO, indoleamine 2,3-dioxygenase; IPRES, innate anti–PD-1 resistance signature; LAG-3, lymphocyte activation gene 3; TIM-3, T-cell immunoglobulin and mucin domain 3; Tregs, regulatory T cells; VEGF, vascular endothelial growth factor.

Close modal

Primary resistance to immune checkpoint blockade occurs in approximately 40% to 65% of patients with melanoma treated with anti–PD-1 based therapy (Fig. 2), depending on whether anti–PD-1 is given upfront or after progression on other therapies (2, 14, 15), and >70% of those treated with anti–CTLA-4 therapy (4, 13). This key unsolved clinical problem occurs when there is failure to induce an effective antitumor immune response at any of the three stages of the cancer immune cycle (Fig. 1). To date, the clinicopathologic factors that have been associated with primary resistance are elevated levels of baseline serum LDH (23), increased baseline tumor burden (24), lack of PD-L1 expression in baseline melanoma tissue samples (Fig. 3; ref. 25), lack of T-cell infiltration (Fig. 3; ref. 21), the absence of PD-1 T cells and PD-L1 macrophages in melanoma biopsies taken early during treatment (22), insufficient neoantigens and low mutational burden (26), the presence of an innate anti–PD-1 resistance signature (IPRES) transcriptional signature (27), or absence of an interferon signature (28). It is currently unknown whether these measures are surrogates for resistance or have a direct mechanistic role in preventing response. Here, we discuss the immune escape mechanisms that can occur at each stage of the cancer immune cycle (Fig. 1), thereby promoting both the growth and metastasis of tumors and resistance to immune checkpoint inhibitor therapies.

Figure 2.

Primary resistance in metastatic melanoma. CT scans of a 55-year-old patient with metastatic melanoma treated with combined anti–CTLA-4 and anti–PD-1 immunotherapy at baseline (A) and after 12 weeks of therapy (B), demonstrating widespread metastatic disease, including significant liver metastases, who did not respond, indicating primary resistance to therapy. The patient did not respond to subsequent chemotherapy and died 156 days after commencing combined immunotherapy.

Figure 2.

Primary resistance in metastatic melanoma. CT scans of a 55-year-old patient with metastatic melanoma treated with combined anti–CTLA-4 and anti–PD-1 immunotherapy at baseline (A) and after 12 weeks of therapy (B), demonstrating widespread metastatic disease, including significant liver metastases, who did not respond, indicating primary resistance to therapy. The patient did not respond to subsequent chemotherapy and died 156 days after commencing combined immunotherapy.

Close modal
Figure 3.

Variable expression of immune markers in patients with metastatic melanoma treated with anti–PD-1 immunotherapy. Multiplex immunofluorescent images illustrating baseline PD-L1 expression and tumor-infiltrating lymphocytes (TIL) in a 59-year-old patient who responded to anti–PD-1 monotherapy (A and C), and lack thereof in a 55-year-old patient who did not respond to immunotherapy (B and D).

Figure 3.

Variable expression of immune markers in patients with metastatic melanoma treated with anti–PD-1 immunotherapy. Multiplex immunofluorescent images illustrating baseline PD-L1 expression and tumor-infiltrating lymphocytes (TIL) in a 59-year-old patient who responded to anti–PD-1 monotherapy (A and C), and lack thereof in a 55-year-old patient who did not respond to immunotherapy (B and D).

Close modal

Antigen presentation and T-cell activation

Upon encountering and engulfing foreign antigens, such as that of cancer cells, DCs migrate from the tumor to regional lymph nodes where they present the antigens on major histocompatibility complex (MHC) class I molecules to CD8+ T cells, resulting in activation of the latter. Barriers at this stage of the cancer immune cycle prevent optimal T-cell priming and activation, hence resulting in evasion of the immune system by the tumor (Table 1).

Table 1.

Mechanisms of resistance to immune checkpoint blockade

Mechanisms of resistanceContributing factorsReferences
Insufficient antigen presentation and recognition Low mutational burden (26) 
 Lack of neoantigen recognition (26, 35) 
 Loss of B2M (99, 100) 
 Loss of MHC class I (99) 
Insufficient T-cell activation Lack of mature DCs (44, 45) 
 STAT3 expression (46, 49) 
Absence of T cells from tumor microenvironment Lack of chemokines (50, 51, 54) 
 VEGF overexpression (63, 64) 
Upregulation of immunosuppressive markers PD-L1 (69, 70) 
 IDO (80, 81) 
 Tregs (82–85) 
Decreased sensitivity to IFN-γ signaling Mutations in the JAK/STAT pathway (93, 101) 
Immune checkpoint markers TIM-3 (105) 
 LAG-3 (106) 
Mechanisms of resistanceContributing factorsReferences
Insufficient antigen presentation and recognition Low mutational burden (26) 
 Lack of neoantigen recognition (26, 35) 
 Loss of B2M (99, 100) 
 Loss of MHC class I (99) 
Insufficient T-cell activation Lack of mature DCs (44, 45) 
 STAT3 expression (46, 49) 
Absence of T cells from tumor microenvironment Lack of chemokines (50, 51, 54) 
 VEGF overexpression (63, 64) 
Upregulation of immunosuppressive markers PD-L1 (69, 70) 
 IDO (80, 81) 
 Tregs (82–85) 
Decreased sensitivity to IFN-γ signaling Mutations in the JAK/STAT pathway (93, 101) 
Immune checkpoint markers TIM-3 (105) 
 LAG-3 (106) 

Abbreviations: B2M, beta-2-microglobulin; IDO, indoleamine 2,3-dioxygenase; IFN-γ, interferon gamma; STAT3, signal transducer and activator of transcription 3; TIM-3, T-cell immunoglobulin and mucin domain 3; Tregs, regulatory T cells; VEGF, vascular endothelial growth factor.

Poor immunogenicity.

Some tumors lack sufficient antigen presentation by the immune system (29, 30) or do not present antigens that can be recognized as foreign (31, 32). The process of distinguishing tumor cells from normal cells depends on T-cell recognition of tumor-specific or tumor-associated antigens (TAA; ref. 33). Tumor immune evasion by TAA-negative cells was reported in patients with melanoma who relapsed after responding to peptide vaccinations (34). Recognition of tumor neoantigens by T cells has been associated with increased and durable response to immunotherapies and increased tumor regression, indicating a significant role for neoantigens in improving the outcome of patients with metastatic melanoma (26, 35). Circulating CD8+PD-1+ lymphocytes in peripheral blood of patients with melanoma can target patient-specific neoantigens, and neoantigen-specific T cells can in turn recognize autologous tumors (36). The immunogenicity of neoantigens can be predicted by combining exome sequencing and mass spectrometry data, thereby facilitating the identification of antigens that can be used to generate active T-cell responses (37).

Analysis of The Cancer Genome Atlas (TCGA) data from melanoma cases revealed that cutaneous melanoma displays a high mutational burden and UV signature (38). In addition to neoantigen recognition, a high mutational load was also found to correlate with clinical benefit to immune checkpoint blockade (26). Similarly, a positive correlation was observed between a higher mutational load and increased CD8+ T-cell infiltration (39). Furthermore, an increased mutational burden is associated with elevated PD-L1 expression in advanced melanoma (40). In a pooled analysis of 832 patients with melanoma, an increased PFS was observed in PD-L1–positive patients treated with nivolumab and ipilimumab combined immunotherapy, as well as in patients treated with nivolumab alone, compared with PD-L1–negative patients (2, 41). Similarly, PD-L1–positive patients treated with pembrolizumab had increased PFS, OS, and overall response rate (ORR), highlighting PD-L1 as a potential biomarker of response (42). As PD-L1 positivity is associated with improved response in patients with melanoma, a lack of PD-L1 correlates with primary resistance (Fig. 3). Nevertheless, some patients with PD-L1–positive tumors do not respond to PD-1 blockade, and conversely, some patients with PD-L1–negative tumors respond. For these reasons, intratumoral PD-L1 expression is a suboptimal predictive biomarker (2, 41). Together, the aforementioned data indicate that PD-L1 expression is a possible surrogate for lack of immunogenicity, as well as other failures further down the immune cycle.

Impaired DC maturation.

In order to efficiently activate T cells, DCs must undergo a process called maturation, where they increase their capacity to stimulate T cells by displaying increased expression of various costimulatory molecules required for T-cell activation, such as MHC class I/II, CD80, CD86, and CD40 (43). The density of DCs strongly correlates with activated T cells in melanoma (44). The function of DCs can be impaired via multiple pathways. Interleukin (IL) 37b, a protein with a critical role in the inhibition of the innate immune response, suppresses DC maturation and function by decreasing CD80 and CD86 expression via the ERK/S6K/NF-κB signaling pathways (45). Furthermore, DC maturation and tumor infiltration increased significantly in melanoma following the inhibition of signal transducer and activator of transcription 3 (STAT3), a transcription factor that is required for tumor growth and metastasis (46). STAT3 is also involved in the cross-talk between melanoma cells and immune cells, resulting in the induction of other immunosuppressive factors such as the vascular endothelial growth factor (VEGF), IL10, regulatory T cells (Treg), and transforming growth factor β (TGF-β), all of which have inhibitory effects on DC maturation (47–49).

T-cell trafficking and tumor infiltration

Tumors can use a number of immune evasive mechanisms to prevent T-cell trafficking and infiltration into tumors. Assuming that the T cells were successfully activated in the previous steps, disruption during this stage is a likely cause for lack of response to immunotherapy.

Downregulation of chemokines required for T-cell recruitment.

The differential expression of chemokine receptors is required for effective T-cell homing and recruitment in cancer. In particular, CXCR3 has been identified as an important chemokine receptor critical for T-cell infiltration. In mouse melanoma models, CXCR3 was highly expressed on a number of T-cell subsets, and transfection with its ligand, CXCL9, resulted in a significant increase in both CD4+ and CD8+ T-cell infiltration (50). Similarly, human melanoma samples with high CD8+ T-cell expression were associated with increased levels of CXCL9 and CXCL10 (51). Interferon gamma (IFN-γ, IFNG) has previously been shown to mediate trafficking of Tregs, T helper cells, and cytotoxic T cells (52, 53). STAT3 inhibits CXCL10 production by tumor-associated myeloid cells and T-cell recruitment into tumors by downregulating IFN-γ production by CD8+ T cells (54). Conversely, Stat3 ablation increases CXCR3 expression on CD8+ T cells, allowing T-cell tumor infiltration (54).

Epigenetic alterations including DNA methylation and histone modifications have also been identified as important mechanisms of chemokine repression and tumor progression. Epigenetic modifications are heritable modifications to DNA that result in changes to the gene expression profiles of tumor cells, thereby allowing them to evade the immune system (55). Epigenetic silencing resulted in the suppression of CXCL9 and CXCL10 in ovarian cancer, and treatment with epigenetic modulators increased chemokine expression and T-cell infiltration into tumors (56). Increased expression of chemokines, T-cell recruitment, and tumor regression was also observed in lung cancer cell lines and mice treated with the histone deacetylase (HDAC) inhibitor romidepsin (57).

Upregulation of the endothelin B receptor.

T-cell trafficking through the tumor and lymph nodes is controlled by a number of endothelial signals, regulating T-cell homing, adhesion, and migration (58). The interaction between endothelin 1 (ET-1, EDN1) and the endothelin A receptor (ETAR, EDNRA) promotes tumorigenesis through various pathways including cell proliferation, invasion, angiogenesis, bone remodeling, and inhibition of apoptosis (59). The endothelin B receptor (ETBR, EDNRB) counterregulates ET-1/ETAR activity via the increased production of nitric oxide, activation of apoptotic pathways, and clearance of ET-1 (60). The endothelin system has been implicated in the pathogenesis of a number of cancers, including ovarian cancer, prostate cancer, and colon cancer. Interestingly, ETBR is upregulated in melanoma and has also been proposed as a marker of melanoma progression, suggesting a role for ETBR in melanoma tumorigenesis (61). ETBR inhibition in 10 human melanoma cell lines using the ETBR antagonist BQ788 resulted in an increase in apoptosis and cell death, as well as an increase in angiogenesis in the tumors (62). In human ovarian cancers, ETBR was found to correlate with an absence of tumor-infiltrating lymphocytes (TIL) as well as decreased patient survival time. Administration of BQ788 increased T-cell homing to tumors and improved the efficacy of immunotherapy (58), highlighting ETBR as a potential immune escape mechanism and future target in patients who fail to respond to immunotherapy.

Overexpression of VEGF.

Increased levels of the proangiogenic factor VEGF in plasma and tissue samples have also been associated with the growth and progression of melanoma (63, 64). VEGF-A downregulates T-cell adhesion to the endothelium via the suppression of intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) on endothelial cells (65). Increased expression of VEGF in tumor was associated with an absence of intratumoral TILs and a shorter OS time in patients with ovarian carcinoma (66). Inhibition of VEGF resulted in an increase in T-cell infiltration into B16 melanoma tumors via the upregulation of CXCL10 and CXCL11 chemokines (67). Additionally, VEGF-A, along with prostaglandin E2 (PGE2) and IL10, upregulates the Fas ligand, resulting in CD8+ T-cell death, and subsequent inhibition of VEGF and PGE2 increased CD8+ T-cell infiltration (68). Corresponding with these data, in melanoma tumor biopsies, increased levels of VEGFA were observed in nonresponders to anti–CTLA-4 and anti–PD-1 immunotherapies in comparison to responders (19).

T-cell killing activity within the tumor microenvironment

Primary resistance to immunotherapy also occurs when T cells become successfully activated and infiltrate the tumor; however, their function is hindered by the presence of immunosuppressive molecules within the tumor microenvironment (31).

Upregulation of PD-L1.

Primary resistance can be driven by the constitutive expression of PD-L1 through oncogenic signaling (69, 70). The increased expression of PD-L1 by cells in the tumor microenvironment results in decreased function of cytotoxic T cells and apoptosis, hence providing an immune escape mechanism for tumor cells.

Several studies have revealed a correlation between loss of the phosphatase and tensin homolog (PTEN) in cancer and the upregulation of PD-L1, implicating PD-L1 in tumor immune evasion. PTEN is a tumor suppressor that negatively regulates the P13K/AKT pathway. This pathway is responsible for the regulation of cellular processes such as proliferation and survival. The loss of PTEN and activation of the P13K/AKT pathway in human glioma cell lines resulted in an increase in posttranscriptional CD274 expression (71). PD-L1 expression was also upregulated in lung squamous cell carcinoma following the simultaneous depletion of Pten and Lkb1 [also known as Stk11 (serine–threonine kinase 11); ref. 72]. In melanoma, loss of PTEN led to a decrease in T-cell trafficking, infiltration, and T-cell activity (73). However, silencing PTEN did not significantly alter the expression of PD-L1 in melanoma cell lines in vitro or in xenograft models in vivo, indicating that PD-L1 regulation may not be the principal mechanism of immune resistance resulting from a loss of PTEN (73).

Other mechanisms that have been shown to have a role in the constitutive upregulation of PD-L1 include the transcription factor interferon regulatory factor 1 (IRF-1) and mutations in the epidermal growth factor receptor (EGFR). IRF-1 is responsible for the regulation of cell proliferation, apoptosis, and immunity (74). The knockdown of IRF-1 using siRNA resulted in the decrease in transcription and translation of PD-L1 in a lung carcinoma cell line (75). Similarly, activation of the EGFR pathway resulted in the increased expression of PD-L1 in lung cancer cell lines (76, 77) and tissue (76). Increased expression of markers of T-cell exhaustion, such as PD-1 and FOXP3, was also observed in the tumor microenvironment (76). PD-1 blockade increased cytotoxic T-cell numbers as well as effector T-cell function (76), highlighting the role of the PD-1/PD-L1 axis in immune evasion and its manipulation as a therapeutic strategy.

Induction of IDO.

Another molecule proposed to play a critical role in the negative regulation of T-cell function is indoleamine 2,3-dioxygenase (IDO, IDO1). IDO is expressed in a wide range of human cancers and is the rate-limiting enzyme responsible for the degradation of tryptophan into kynurenine (78, 79). T lymphocytes undergo arrest in response to this tryptophan depletion, resulting in the suppression of T-cell proliferation and activity (80). To understand the mechanism of immunosuppression induced by IDO, Holmgaard and colleagues (81) developed a B16 melanoma tumor model overexpressing IDO and revealed a correlation between IDO expression and increased tumor-infiltrating myeloid-derived suppressor cells (MDSC), as well as CD4+FOXP3+ Tregs. This association demonstrated that IDO suppresses T-cell activity through the recruitment and activation of MDSCs in a Treg-dependent fashion (81). Systemic inhibition of IDO in mice using a tryptophan analogue, 1-methyl-L-tryptophan (1MT), reduced tumor progression in a T-cell–dependent manner (79). Similarly, administration of 1MT in combination with anti–CTLA-4 immunotherapy in B16F10 mouse melanoma models resulted in a significant delay in tumor growth and an increase in OS (78). These findings provide a strong rationale for targeting IDO to improve the efficacy of immunotherapies in patients with melanoma.

Upregulation of Tregs.

The upregulation of FOXP3-expressing Tregs has been observed in a number of melanoma studies, revealing a possible role for Tregs in melanoma tumorigenesis (82–84). Tregs promote tumor growth by inhibiting the activity of T-cell subsets, either through direct cell-to-cell contact or indirectly through the secretion of anti-inflammatory cytokines, such as IL10 and TGF-β (85, 86). The presence of CD4+CD25+ FOXP3 Tregs was observed amongst TILs in metastatic melanoma (87) and the transfer of CD25 (IL2RA)-depleted splenic T cells into B16 mouse melanoma models resulted in the suppression of tumor growth in vivo (82). Populations of tumor-infiltrating Tregs also significantly correlated with increased tumor growth in B16BL6 mice (88). Furthermore, a decrease in FOXP3+ Tregs significantly correlated with increased tumor control and survival in patients with melanoma treated with ipilimumab (89), highlighting the immunosuppressive function of Tregs in melanoma.

Upregulation of the CD73/adenosine pathway.

Elevated levels of extracellular adenosine and CD73 (NT5E) have also been implicated in immune suppression and tumor progression. Adenosine is produced via the conversion of extracellular ATP by the ectoenzymes CD39 (ENTPD1) and CD73 and binds to the adenosine A2A receptor (A2AR, ADORA2A) to inhibit effector T-cell function (90). Increased CD73 expression correlated with advanced-stage disease in melanoma (91), and an upregulation in CD73 was observed in patients who progressed following treatment with anti–PD-1 immunotherapy (92). A2AR inhibition increased CD8+ T-cell infiltration and significantly reduced tumor growth in mouse melanoma models (91), suggesting a role for the CD73/adenosine axis in promoting immune escape.

Expression of IPRES signature.

The expression of IPRES has recently been identified as a mechanism of primary resistance to immunotherapy. Transcriptomal analyses of responding and nonresponding pretreatment melanoma biopsies from patients treated with anti–PD-1 immunotherapy revealed the coenrichment of genes associated with mesenchymal transition, wound healing, and angiogenesis in nonresponding patient samples (27). This was observed not only in metastatic melanoma but also in other major cancer types such as pancreatic cancer (27).

Loss-of-function mutations.

Mutations in the janus kinases 1 and 2 (JAK1/2) have also been shown to be involved in primary resistance to anti–PD-1 immunotherapy. JAK1/2 loss-of-function mutations identified in one of 23 melanoma tumor biopsies, and two of 48 human melanoma cell lines via whole-exome sequencing, resulted in a lack of PD-L1 expression due to an inability to respond to IFN-γ signaling (93). Furthermore, the recent development of a two cell type-CRISPR (2CT-CRISPR) screening assay revealed an important role for apelin receptor (APLNR) loss-of-function mutations in disturbing effector T-cell function (94). Retroviral overexpression of APLNR correlated with an increase in JAK1 as well as tumor sensitivity to effector T-cell function (94). Conversely, APLNR-knockout cells demonstrated decreased activation of the JAK/STAT pathway following IFN-γ treatment, and Aplnr knockout in mouse melanoma in vivo resulted in a decrease in the efficacy of anti–CTLA-4 immunotherapy (94). These findings provide a strong rationale for further investigating APLNR as a potential target to prevent immune evasion by tumors.

Mechanisms under investigation

The composition of the gut microbiome.

Recent studies have highlighted a possible role for the gut microbiome in patient response to immunotherapy. Dysbiosis (an imbalance of the microbiota) involves decreases in the diversity and stability of the microbiome, thereby promoting tumorigenesis (95). Sequencing of the oral and gut microbiome of patients with metastatic melanoma revealed a correlation between higher gut microbiome diversity and response to anti–PD-1 monotherapy (96). Responders also had a significantly different gut microbiome composition in comparison with nonresponders, and this correlated with differences in PFS (96). The increased abundance of specific bacteria in the gut microbiome also correlated with a higher CD8+ T-cell density in responders to anti–PD-1 immunotherapy (96). Similarly, the composition of the baseline gut microbiome in patients with metastatic melanoma was associated with response to ipilimumab, and improved PFS and OS were associated with specific groups of bacteria such as Faecalibacterium and other Firmicutes (97). Additionally, a significant decrease in TILs and lack of response to CTLA-4 blockade was observed in tumors of mice housed in germ-free conditions (98). The anticancer therapeutic effects of the anti–CTLA-4 antibody were restored upon oral feeding of the germ-free mice with Bacteroides fragilis (B. fragilis), a Bacteroides isolate, as well as with the adoptive transfer of B. fragilis–specific memory T cells (98). The mechanisms through which the gut microbiome influences the immune response are currently being investigated.

Acquired resistance occurs in patients who relapse after exhibiting an initial response to immunotherapy (Fig. 4). Currently, little is understood about the mechanisms that give rise to acquired resistance, and many are likely to be similar to those underlying primary resistance (Table 1).

Figure 4.

Acquired resistance in metastatic melanoma. Images of a 74-year-old patient with metastatic melanoma treated with anti–PD-1 monotherapy, showing complete resolution of melanoma in right-neck lymph nodes from baseline (A), week 4 (B), and week 12 (C). However, acquired resistance developed in the right preauricular region at 12 months after the commencement of anti–PD-1 monotherapy (D). The patient subsequently received radiotherapy and ipilimumab in addition to continuing anti–PD-1, with an excellent ongoing response 12 months after acquired resistance.

Figure 4.

Acquired resistance in metastatic melanoma. Images of a 74-year-old patient with metastatic melanoma treated with anti–PD-1 monotherapy, showing complete resolution of melanoma in right-neck lymph nodes from baseline (A), week 4 (B), and week 12 (C). However, acquired resistance developed in the right preauricular region at 12 months after the commencement of anti–PD-1 monotherapy (D). The patient subsequently received radiotherapy and ipilimumab in addition to continuing anti–PD-1, with an excellent ongoing response 12 months after acquired resistance.

Close modal

Acquired resistance to immunotherapy can develop when there is Darwinian selection of subpopulations of tumor cells with genetic and epigenetic traits that allow them to evade the immune system (32). An example of one such trait is beta-2-microglobulin (B2M), a component of MHC class I molecules that is necessary for their functional expression. The loss of B2M expression was reported in melanoma cell lines from five patients who had been treated with immunotherapy and cytokine–gene therapy (99). This resulted in a loss of MHC class I expression and, therefore, a subsequent decrease in recognition by CD8+ T cells. Archival tissues taken prior to immunotherapy from three of these patients were B2M positive, suggesting loss of B2M expression as a mechanism of acquired resistance (99). Similarly, the loss of B2M has been observed in sequential lesions obtained from a patient with metastatic melanoma following immunotherapy treatment with DCs transfected with autologous tumor mRNA (100).

JAK1/2 mutations have also recently been identified as genetic markers of acquired resistance to immunotherapy in melanoma. These mutations in tumor cells lead to decreased sensitivity to IFN-γ, ultimately preventing IFN-γ–induced cell growth arrest (101). Upon exposure to IFN-γ (produced by activated T cells), JAK1/2 become activated and subsequently phosphorylate a tyrosine residue present on STATs (102). This JAK/STAT signaling pathway is responsible for cell proliferation, differentiation, cell migration, and apoptosis (102). However, IFN-γ also results in the upregulation of PD-L1 on tumor cells, thus inactivating antitumor T cells (70). Loss-of-function mutations in the genes encoding JAK1 or JAK2 were found in relapsed tumors in two of four patients following whole-exome sequencing of baseline and progression biopsies; all patients had an objective response to treatment with pembrolizumab and then progressed after a median of 1.8 years (101). The anti–PD-1-resistant cells harboring JAK mutations were derived from cells clonally selected from the baseline tumor. These findings demonstrate the role of the JAK/STAT pathway in promoting acquired resistance to immunotherapy.

In addition, acquired resistance can also occur on the level of the individual cells, whereby tumor cells alter their gene expression in response to immune molecules within the tumor microenvironment (32). For example, PD-L1 can be upregulated by tumor cells in response to immune cytokines, such as IFN-γ released by T cells, hence limiting T-cell function (70), and can occur in both primary and acquired resistance (32, 103).

Other immune checkpoint markers such as lymphocyte activation gene 3 (LAG-3) and T-cell immunoglobulin and mucin domain 3 (TIM-3, HAVCR2) have also been revealed to interfere with the activity of T cells (70, 104), resulting in acquired resistance to immunotherapy. In a recent study, TIM-3 upregulation was observed in patients who developed adaptive immune resistance to anti–PD-1 immunotherapy (105). Furthermore, TIM-3 blockade in mice resulted in a significant increase in survival time, as well as increased production of IFN-γ and proliferation of CD8+ T cells (105). LAG-3 is also overexpressed in PD-L1–positive melanoma, suggesting LAG-3 upregulation as a potential immune evasion mechanism (106).

From the above, it is clear that there exist multiple immune evasive mechanisms that can be utilized by tumors at each of the different stages of the cancer immune cycle that may induce either primary or acquired resistance to immunotherapy. Determining the specific mechanisms underlying resistance to immunotherapy in these patients is a crucial step toward effective treatment and ultimately producing durable responses for them.

Combinatorial therapies

The immune escape mechanisms discussed above do not act in isolation. Together, the overlap between various signaling pathways and the interactions between several of the immunosuppressive molecules leads to resistance. It is likely that combinations of therapy will be more effective than single-agent therapies for a given patient. However, the challenge remains to determine which of these combinations are most effective and in which patient given interpatient heterogeneity. Currently, multiple clinical trials are underway examining the activity and toxicity of combined immunotherapies, particularly using an anti–PD-1 drug in combination with an agent targeting a complimentary part of the immune system (Supplementary Table S1).

Dual blockade of PD-1 and LAG-3 in mouse cancer models resulted in the regression of tumors in most mice, as well as an increased survival rate (107). In a recent study, all mice that were treated with a triple combination of LAG-3 blockade, PD-1 blockade, and poxvirus-based immunotherapy demonstrated complete tumor regression (108). Phase I clinical trials involving LAG-3 blockade with and without PD-1 blockade in solid tumors are currently underway, and in patients who had progressed on anti–PD-1 monotherapy, a response rate of 16% (any tumor reduction in 45%) was observed in those patients whose tumors expressed LAG-3 (109). Similarly, in mouse models, combined TIM-3 and PD-L1 blockade significantly reduced tumor growth in comparison with single-agent immunotherapy (110). Furthermore, dual blockade of TIM-3 and PD-L1 increased the ability of CD8+ T cells to produce IFN-γ, thereby restoring their function (110). Consistent with this, anti–TIM-3 and anti–PD-1/anti–CTLA-4 combined immunotherapy resulted in a significant decrease in the tumor sizes of multiple cancers (111).

Phase I/II clinical trials testing the combination of an IDO inhibitor with PD-1/PD-L1 inhibitors in metastatic melanoma are also currently underway. These include the administration of nivolumab and a PD-L1/IDO peptide vaccine (NCT03047928), and indoximod (IDO inhibitor) in combination with CTLA-4 or PD-1 inhibitors (NCT02073123). Furthermore, the combination of epacadostat (an IDO1 inhibitor) plus pembrolizumab was generally well tolerated and correlated with improved response in various cancers (112–114), leading to the initiation of phase III studies such as that in patients with treatment-naïve advanced melanoma (NCT02752074).

Many studies have also demonstrated the increased efficacy of combined radiotherapy and immune checkpoint blockade. Radiotherapy activates the inflammatory pathways and induces DNA damage, resulting in the release of antigens from irradiated cells and an increase in tumor sensitization to T-cell immune responses (115). Radiotherapy in patients with advanced melanoma who had progressed following treatment with ipilimumab led to an abscopal effect in some patients, whereby irradiation of the primary tumor induced regression in other nonirradiated metastases, which correlated with longer OS (116). Significant tumor regression was also observed in patients with metastatic melanoma who received combined radiotherapy and anti–CTLA-4 immunotherapy, and experiments in mice showed that a combination of anti–CTLA-4 immunotherapy, anti–PD-L1 immunotherapy, and radiotherapy was required to achieve optimum response (117).

The combination of epigenetic modulators and immune checkpoint blockade has recently been shown to be more effective than single-agent immunotherapy in patients with melanoma. The inhibition of HDAC using LBH589 in combination with PD-1 blockade in B16F10 mouse melanoma models resulted in delayed tumor growth and increased survival compared with the control group and PD-1 blockade alone (118). Similarly, a reduction in tumor size and Tregs was observed in B16F10 mouse melanoma models treated with a combination of anti–CTLA-4 immunotherapy and epigenetic modulation of trimethylation of lysine 27 on histone H3 (H3K27me3) compared with CTLA-4 blockade alone (119). The efficacy of combined epigenetic and immune checkpoint therapies is currently being tested in various human cancers (Supplementary Table S1).

These findings provide a strong rationale for the development of combinations of different treatment strategies in clinical trials as an effective means of melanoma treatment.

Checkpoint inhibitor immunotherapy has revolutionized the treatment of multiple cancer types. However, responses in patients with metastatic melanoma are diverse, with many patients displaying primary or acquired resistance. An important ongoing, major unmet clinical need remains to identify predictors and causes of this resistance and strategies to overcome them. A key element of effective immunotherapy is identifying the various mechanisms by which the tumors evade the immune system on an individual basis. Improving our understanding of these mechanisms and determining which immune markers to target in each patient will then allow for the administration of the most appropriate form of therapy to achieve optimal response and improve the overall outcomes of patients with metastatic melanoma.

G.V. Long reports receiving speakers bureau honoraria from Bristol-Myers Squibb, Incyte, Merck, Novartis, and Roche, and is a consultant/advisory board member for Amgen, Array, Bristol-Myers Squibb, Incyte, Merck, Novartis, Pierre Fabre, and Roche. No potential conflicts of interest were disclosed by the other authors.

This work was supported by Melanoma Institute Australia, the New South Wales Ministry of Health, NSW Health Pathology, National Health and Medical Research Council of Australia (NHMRC), and Cancer Institute NSW. J.S. Wilmott, R.A. Scolyer, and G.V. Long are supported by NHMRC Fellowships. J.S. Wilmott is also supported by a CINSW Fellowship. G.V. Long is also supported by the University of Sydney Medical Foundation. T.N. Gide is supported by The University of Sydney and Melanoma Institute Australia Scholarships.

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.
Atkinson
V
,
Ascierto
PA
,
Long
GV
,
Brady
B
,
Dutriaux
C
,
Maio
M
, et al
Two-year survival and safety update in patients (pts) with treatment-naïve advanced melanoma (MEL) receiving nivolumab (NIVO) or dacarbazine (DTIC) in CheckMate 066
. 
2015 Nov 18–21
;
San Francisco, CA, USA
.
2.
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
.
3.
Robert
C
,
Ribas
A
,
Hamid
O
,
Daud
A
,
Wolchok
JD
,
Joshua
AM
, et al
Three-year overall survival for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001
.
J Clin Oncol
34
, 
2016
(
suppl; abstr 9503
).
4.
Robert
C
,
Thomas
L
,
Bondarenko
I
,
O'Day
S
,
Weber
J
,
Garbe
C
, et al
Ipilimumab plus dacarbazine for previously untreated metastatic melanoma
.
N Engl J Med
2011
;
364
:
2517
26
.
5.
Schachter
J
,
Ribas
A
,
Long
GV
,
Arance
A
,
Grob
JJ
,
Mortier
L
, et al
Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival analysis of KEYNOTE-006
.
J Clin Oncol
34
, 
2016
(
suppl; abstr 9504
).
6.
Brahmer
J
,
Reckamp
KL
,
Baas
P
,
Crino
L
,
Eberhardt
WE
,
Poddubskaya
E
, et al
Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer
.
N Engl J Med
2015
;
373
:
123
35
.
7.
Herbst
RS
,
Baas
P
,
Kim
DW
,
Felip
E
,
Perez-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
.
8.
Motzer
RJ
,
Escudier
B
,
McDermott
DF
,
George
S
,
Hammers
HJ
,
Srinivas
S
, et al
Nivolumab versus everolimus in advanced renal-cell carcinoma
.
N Engl J Med
2015
;
373
:
1803
13
.
9.
Ferris
RL
,
Blumenschein
G
 Jr
,
Fayette
J
,
Guigay
J
,
Colevas
AD
,
Licitra
L
, et al
Nivolumab for recurrent squamous-cell carcinoma of the head and neck
.
N Engl J Med
2016
;
375
:
1856
67
.
10.
Kaufman
HL
,
Russell
J
,
Hamid
O
,
Bhatia
S
,
Terheyden
P
,
D'Angelo
SP
, et al
Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial
.
Lancet Oncol
2016
;
17
:
1374
85
.
11.
Balar
AV
,
Galsky
MD
,
Rosenberg
JE
,
Powles
T
,
Petrylak
DP
,
Bellmunt
J
, et al
Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial
.
Lancet
2017
;
389
:
67
76
.
12.
Sharma
P
,
Retz
M
,
Siefker-Radtke
A
,
Baron
A
,
Necchi
A
,
Bedke
J
, et al
Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial
.
Lancet Oncol
2017
;
18
:
312
22
.
13.
Hodi
FS
,
O'Day
SJ
,
McDermott
DF
,
Weber
RW
,
Sosman
JA
,
Haanen
JB
, et al
Improved survival with ipilimumab in patients with metastatic melanoma
.
N Engl J Med
2010
;
363
:
711
23
.
14.
Robert
C
,
Long
GV
,
Brady
B
,
Dutriaux
C
,
Maio
M
,
Mortier
L
, et al
Nivolumab in previously untreated melanoma without BRAF mutation
.
N Engl J Med
2015
;
372
:
320
30
.
15.
Robert
C
,
Schachter
J
,
Long
GV
,
Arance
A
,
Grob
JJ
,
Mortier
L
, et al
Pembrolizumab versus ipilimumab in advanced melanoma
.
N Engl J Med
2015
;
372
:
2521
32
.
16.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Rutkowski
P
,
Grob
J-J
,
Cowey
CL
, et al
Overall survival (OS) results from a phase III trial of nivolumab (NIVO) combined with ipilimumab (IPI) in treatment-naïve patients with advanced melanoma (CheckMate 067)
[abstract]. In
:
Proceedings of the American Association for Cancer Research Annual Meeting 2017
; 
2017 Apr 1–5
;
Washington, DC. Philadelphia (PA)
:
AACR
;
Cancer Res 2017;77(13 Suppl): Abstract nr CT075
.
17.
Herbst
RS
,
Soria
JC
,
Kowanetz
M
,
Fine
GD
,
Hamid
O
,
Gordon
MS
, et al
Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients
.
Nature
2014
;
515
:
563
7
.
18.
Hodi
FS
,
Kluger
H
,
Sznol
M
,
Carvajal
R
,
Lawrence
D
,
Atkins
M
, et al
Durable, long-term survival in previously treated patients with advanced melanoma (MEL) who received nivolumab (NIVO) monotherapy in a phase I trial [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16–20; New Orleans, LA. Philadelphia (PA): AACR;
Cancer Res
2016
;
76
(
14 Suppl
):
Abstract nr CT001
.
19.
Chen
PL
,
Roh
W
,
Reuben
A
,
Cooper
ZA
,
Spencer
CN
,
Prieto
PA
, et al
Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade
.
Cancer Discov
2016
;
6
:
827
37
.
20.
Powderly
JD
,
Koeppen
H
,
Hodi
FS
,
Sosman
JA
,
Gettinger
SN
,
Desai
R
, et al
Biomarkers and associations with the clinical activity of PD-L1 blockade in a MPDL3280A study
.
J Clin Oncol
31
, 
2013
(suppl; abstr 3001).
21.
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
.
22.
Vilain
RE
,
Menzies
AM
,
Wilmott
JS
,
Kakavand
H
,
Madore
J
,
Guminski
A
, et al
Dynamic changes in PD-L1 expression and immune infiltrates early during treatment predict response to PD-1 blockade in melanoma
.
Clin Cancer Res
2017
;
23
:
5024
33
.
23.
Diem
S
,
Kasenda
B
,
Spain
L
,
Martin-Liberal
J
,
Marconcini
R
,
Gore
M
, et al
Serum lactate dehydrogenase as an early marker for outcome in patients treated with anti-PD-1 therapy in metastatic melanoma
.
Br J Cancer
2016
;
114
:
256
61
.
24.
Nishino
M
,
Giobbie-Hurder
A
,
Manos
MP
,
Bailey
N
,
Buchbinder
EI
,
Ott
PA
, et al
Immune-related tumor response dynamics in melanoma patients treated with pembrolizumab: identifying markers for clinical outcome and treatment decisions
.
Clin Cancer Res
2017
;
23
:
4671
9
.
25.
Taube
JM
,
Klein
A
,
Brahmer
JR
,
Xu
H
,
Pan
X
,
Kim
JH
, et al
Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy
.
Clin Cancer Res
2014
;
20
:
5064
74
.
26.
Snyder
A
,
Makarov
V
,
Merghoub
T
,
Yuan
J
,
Zaretsky
JM
,
Desrichard
A
, et al
Genetic basis for clinical response to CTLA-4 blockade in melanoma
.
N Engl J Med
2014
;
371
:
2189
99
.
27.
Hugo
W
,
Zaretsky
JM
,
Sun
L
,
Song
C
,
Moreno
BH
,
Hu-Lieskovan
S
, et al
Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma
.
Cell
2016
;
165
:
35
44
.
28.
Ayers
M
,
Lunceford
J
,
Nebozhyn
M
,
Murphy
E
,
Loboda
A
,
Kaufman
DR
, et al
IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade
.
J Clin Invest
2017
;
127
:
2930
40
.
29.
Korkolopoulou
P
,
Kaklamanis
L
,
Pezzella
F
,
Harris
AL
,
Gatter
KC
. 
Loss of antigen-presenting molecules (MHC class I and TAP-1) in lung cancer
.
Br J Cancer
1996
;
73
:
148
53
.
30.
Restifo
NP
,
Kawakami
Y
,
Marincola
F
,
Shamamian
P
,
Taggarse
A
,
Esquivel
F
, et al
Molecular mechanisms used by tumors to escape immune recognition: immunogenetherapy and the cell biology of major histocompatibility complex class I
.
J Immunother Emphasis Tumor Immunol
1993
;
14
:
182
90
.
31.
Kelderman
S
,
Schumacher
TN
,
Haanen
JB
. 
Acquired and intrinsic resistance in cancer immunotherapy
.
Mol Oncol
2014
;
8
:
1132
9
.
32.
Restifo
NP
,
Smyth
MJ
,
Snyder
A
. 
Acquired resistance to immunotherapy and future challenges
.
Nat Rev Cancer
2016
;
16
:
121
6
.
33.
Drake
CG
,
Jaffee
E
,
Pardoll
DM
. 
Mechanisms of immune evasion by tumors
.
Adv Immunol
2006
;
90
:
51
81
.
34.
Jager
E
,
Ringhoffer
M
,
Altmannsberger
M
,
Arand
M
,
Karbach
J
,
Jager
D
, et al
Immunoselection in vivo: independent loss of MHC class I and melanocyte differentiation antigen expression in metastatic melanoma
.
Int J Cancer
1997
;
71
:
142
7
.
35.
McGranahan
N
,
Furness
AJS
,
Rosenthal
R
,
Ramskov
S
,
Lyngaa
R
,
Saini
SK
, et al
Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade
.
Science
2016
;
351
:
1463
9
.
36.
Gros
A
,
Parkhurst
MR
,
Tran
E
,
Pasetto
A
,
Robbins
PF
,
Ilyas
S
, et al
Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients
.
Nat Med
2016
;
22
:
433
8
.
37.
Yadav
M
,
Jhunjhunwala
S
,
Phung
QT
,
Lupardus
P
,
Tanguay
J
,
Bumbaca
S
, et al
Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing
.
Nature
2014
;
515
:
572
6
.
38.
Zhang
T
,
Dutton-Regester
K
,
Brown
KM
,
Hayward
NK
. 
The genomic landscape of cutaneous melanoma
.
Pigment Cell Melanoma Res
2016
;
29
:
266
83
.
39.
Reuben
A
,
Spencer
CN
,
Prieto
PA
,
Gopalakrishnan
V
,
Reddy
SM
,
Miller
JP
, et al
Genomic and immune heterogeneity are associated with differential responses to therapy in melanoma
.
NPJ Genom Med
2017
;
2
:
10
.
40.
Madore
J
,
Strbenac
D
,
Vilain
R
,
Menzies
AM
,
Yang
JY
,
Thompson
JF
, et al
PD-L1 negative status is associated with lower mutation burden, differential expression of immune-related genes, and worse survival in stage III melanoma
.
Clin Cancer Res
2016
;
22
:
3915
23
.
41.
Long
GV
,
Larkin
J
,
Ascierto
PA
,
Hodi
FS
,
Rutkowski
P
,
Sileni
V
, et al
PD-L1 expression as a biomarker for nivolumab (NIVO) plus ipilimumab (IPI) and NIVO alone in advanced melanoma (MEL): a pooled analysis
.
Ann Oncol
2016
;
27
(
6_suppl
):
1112PD
PD
.
42.
Carlino
M
,
Ribas
A
,
Gonzalez
R
,
Hoeller
C
,
Bar-Sela
G
,
Barrow
C
, et al
KEYNOTE-006: PD-L1 expression and efficacy in patients (Pts) treated with pembrolizumab (pembro) vs. ipilimumab (IPI) for advanced melanoma [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2017 Apr 16–20; New Orleans, LA. Philadelphia (PA): AACR;
Cancer Res
2016
;
76
(
14 Suppl
):
Abstract nr CT004
.
43.
Ni
K
,
O'Neill
HC
. 
The role of dendritic cells in T cell activation
.
Immunol Cell Biol
1997
;
75
:
223
30
.
44.
Ladanyi
A
,
Kiss
J
,
Somlai
B
,
Gilde
K
,
Fejos
Z
,
Mohos
A
, et al
Density of DC-LAMP(+) mature dendritic cells in combination with activated T lymphocytes infiltrating primary cutaneous melanoma is a strong independent prognostic factor
.
Cancer Immunol Immunother
2007
;
56
:
1459
69
.
45.
Wu
W
,
Wang
W
,
Wang
Y
,
Li
W
,
Yu
G
,
Li
Z
, et al
IL-37b suppresses T cell priming by modulating dendritic cell maturation and cytokine production via dampening ERK/NF-kappaB/S6K signalings
.
Acta Biochim Biophys Sin (Shanghai)
2015
;
47
:
597
603
.
46.
Emeagi
PU
,
Maenhout
S
,
Dang
N
,
Heirman
C
,
Thielemans
K
,
Breckpot
K
. 
Downregulation of Stat3 in melanoma: reprogramming the immune microenvironment as an anticancer therapeutic strategy
.
Gene Ther
2013
;
20
:
1085
92
.
47.
Chattopadhyay
G
,
Shevach
EM
. 
Antigen-specific induced T regulatory cells impair dendritic cell function via an IL-10/MARCH1-dependent mechanism
.
J Immunol
2013
;
191
:
5875
84
.
48.
Hargadon
KM
,
Bishop
JD
,
Brandt
JP
,
Hand
ZC
,
Ararso
YT
,
Forrest
OA
. 
Melanoma-derived factors alter the maturation and activation of differentiated tissue-resident dendritic cells
.
Immunol Cell Biol
2016
;
94
:
24
38
.
49.
Lindenberg
JJ
,
van de Ven
R
,
Lougheed
SM
,
Zomer
A
,
Santegoets
SJAM
,
Griffioen
AW
, et al
Functional characterization of a STAT3-dependent dendritic cell-derived CD14+ cell population arising upon IL-10-driven maturation
.
OncoImmunology
2013
;
2
:
e23837
.
50.
Hong
M
,
Puaux
AL
,
Huang
C
,
Loumagne
L
,
Tow
C
,
Mackay
C
, et al
Chemotherapy induces intratumoral expression of chemokines in cutaneous melanoma, favoring T-cell infiltration and tumor control
.
Cancer Res
2011
;
71
:
6997
7009
.
51.
Harlin
H
,
Meng
Y
,
Peterson
AC
,
Zha
Y
,
Tretiakova
M
,
Slingluff
C
, et al
Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment
.
Cancer Res
2009
;
69
:
3077
85
.
52.
Fu
H
,
Kishore
M
,
Gittens
B
,
Wang
G
,
Coe
D
,
Komarowska
I
, et al
Self-recognition of the endothelium enables regulatory T-cell trafficking and defines the kinetics of immune regulation
.
Nat Commun
2014
;
5
:
3436
.
53.
Kryczek
I
,
Bruce
AT
,
Gudjonsson
JE
,
Johnston
A
,
Aphale
A
,
Vatan
L
, et al
Induction of IL-17(+) T cell trafficking and development by IFN-γ: mechanism and pathological relevance in psoriasis
.
J Immunol
2008
;
181
:
4733
41
.
54.
Yue
C
,
Shen
S
,
Deng
J
,
Priceman
SJ
,
Li
W
,
Huang
A
, et al
STAT3 in CD8+ T cells inhibits their tumor accumulation by downregulating CXCR3/CXCL10 axis
.
Cancer Immunol Res
2015
;
3
:
864
70
.
55.
Maio
M
,
Covre
A
,
Fratta
E
,
Di Giacomo
AM
,
Taverna
P
,
Natali
PG
, et al
Molecular pathways: at the crossroads of cancer epigenetics and immunotherapy
.
Clin Cancer Res
2015
;
21
:
4040
7
.
56.
Peng
D
,
Kryczek
I
,
Nagarsheth
N
,
Zhao
L
,
Wei
S
,
Wang
W
, et al
Epigenetic silencing of Th1 type chemokines shapes tumor immunity and immunotherapy
.
Nature
2015
;
527
:
249
53
.
57.
Zheng
H
,
Zhao
W
,
Yan
C
,
Watson
CC
,
Massengill
M
,
Xie
M
, et al
HDAC inhibitors enhance T-cell chemokine expression and augment response to PD-1 immunotherapy in lung adenocarcinoma
.
Clin Cancer Res
2016
;
22
:
4119
32
.
58.
Buckanovich
RJ
,
Facciabene
A
,
Kim
S
,
Benencia
F
,
Sasaroli
D
,
Balint
K
, et al
Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy
.
Nat Med
2008
;
14
:
28
36
.
59.
Nelson
J
,
Bagnato
A
,
Battistini
B
,
Nisen
P
. 
The endothelin axis: emerging role in cancer
.
Nat Rev Cancer
2003
;
3
:
110
6
.
60.
Lalich
M
,
McNeel
DG
,
Wilding
G
,
Liu
G
. 
Endothelin receptor antagonists in cancer therapy
.
Cancer Invest
2007
;
25
:
785
94
.
61.
Demunter
A
,
De Wolf-Peeters
C
,
Degreef
H
,
Stas
M
,
van den Oord
JJ
. 
Expression of the endothelin-B receptor in pigment cell lesions of the skin. Evidence for its role as tumor progression marker in malignant melanoma
.
Virchows Arch
2001
;
438
:
485
91
.
62.
Lahav
R
,
Suvà
M-L
,
Rimoldi
D
,
Patterson
PH
,
Stamenkovic
I
. 
Endothelin receptor B inhibition triggers apoptosis and enhances angiogenesis in melanomas
.
Cancer Res
2004
;
64
:
8945
53
.
63.
Redondo
P
,
Bandres
E
,
Solano
T
,
Okroujnov
I
,
Garcia-Foncillas
J
. 
Vascular endothelial growth factor (VEGF) and melanoma. N-acetylcysteine downregulates VEGF production in vitro
.
Cytokine
2000
;
12
:
374
8
.
64.
Rajabi
P
,
Neshat
A
,
Mokhtari
M
,
Rajabi
MA
,
Eftekhari
M
,
Tavakoli
P
. 
The role of VEGF in melanoma progression
.
J Res Med Sci
2012
;
17
:
534
9
.
65.
Bouzin
C
,
Brouet
A
,
De Vriese
J
,
DeWever
J
,
Feron
O
. 
Effects of vascular endothelial growth factor on the lymphocyte-endothelium interactions: identification of caveolin-1 and nitric oxide as control points of endothelial cell anergy
.
J Immunol
2007
;
178
:
1505
11
.
66.
Zhang
L
,
Conejo-Garcia
JR
,
Katsaros
D
,
Gimotty
PA
,
Massobrio
M
,
Regnani
G
, et al
Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer
.
N Engl J Med
2003
;
348
:
203
13
.
67.
Huang
H
,
Langenkamp
E
,
Georganaki
M
,
Loskog
A
,
Fuchs
PF
,
Dieterich
LC
, et al
VEGF suppresses T-lymphocyte infiltration in the tumor microenvironment through inhibition of NF-kappaB-induced endothelial activation
.
FASEB J
2015
;
29
:
227
38
.
68.
Motz
GT
,
Santoro
SP
,
Wang
LP
,
Garrabrant
T
,
Lastra
RR
,
Hagemann
IS
, et al
Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors
.
Nat Med
2014
;
20
:
607
15
.
69.
Dong
ZY
,
Wu
SP
,
Liao
RQ
,
Huang
SM
,
Wu
YL
. 
Potential biomarker for checkpoint blockade immunotherapy and treatment strategy
.
Tumour Biol
2016
;
37
:
4251
61
.
70.
Pardoll
DM
. 
The blockade of immune checkpoints in cancer immunotherapy
.
Nat Rev Cancer
2012
;
12
:
252
64
.
71.
Parsa
AT
,
Waldron
JS
,
Panner
A
,
Crane
CA
,
Parney
IF
,
Barry
JJ
, et al
Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma
.
Nat Med
2007
;
13
:
84
8
.
72.
Xu
C
,
Fillmore
CM
,
Koyama
S
,
Wu
H
,
Zhao
Y
,
Chen
Z
, et al
Loss of Lkb1 and Pten leads to lung squamous cell carcinoma with elevated PD-L1 expression
.
Cancer Cell
2014
;
25
:
590
604
.
73.
Peng
W
,
Chen
JQ
,
Liu
C
,
Malu
S
,
Creasy
C
,
Tetzlaff
MT
, et al
Loss of PTEN promotes resistance to T cell-mediated immunotherapy
.
Cancer Discov
2016
;
6
:
202
16
.
74.
Kröger
A
,
Köster
M
,
Schroeder
K
,
Hauser
H
,
Mueller
PP
. 
Activities of IRF-1
.
J Interferon Cytokine Res
2002
;
22
:
5
14
.
75.
Lee
SJ
,
Jang
BC
,
Lee
SW
,
Yang
YI
,
Suh
SI
,
Park
YM
, et al
Interferon regulatory factor-1 is prerequisite to the constitutive expression and IFN-gamma-induced upregulation of B7-H1 (CD274)
.
FEBS Lett
2006
;
580
:
755
62
.
76.
Akbay
EA
,
Koyama
S
,
Carretero
J
,
Altabef
A
,
Tchaicha
JH
,
Christensen
CL
, et al
Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors
.
Cancer Discov
2013
;
3
:
1355
63
.
77.
Chen
N
,
Fang
W
,
Zhan
J
,
Hong
S
,
Tang
Y
,
Kang
S
, et al
Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: implication for optional immune targeted therapy for NSCLC patients with EGFR mutation
.
J Thorac Oncol
2015
;
10
:
910
23
.
78.
Holmgaard
RB
,
Zamarin
D
,
Munn
DH
,
Wolchok
JD
,
Allison
JP
. 
Indoleamine 2,3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4
.
J Exp Med
2013
;
210
:
1389
402
.
79.
Uyttenhove
C
,
Pilotte
L
,
Theate
I
,
Stroobant
V
,
Colau
D
,
Parmentier
N
, et al
Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase
.
Nat Med
2003
;
9
:
1269
74
.
80.
Munn
DH
,
Shafizadeh
E
,
Attwood
JT
,
Bondarev
I
,
Pashine
A
,
Mellor
AL
. 
Inhibition of T cell proliferation by macrophage tryptophan catabolism
.
J Exp Med
1999
;
189
:
1363
72
.
81.
Holmgaard
RB
,
Zamarin
D
,
Li
Y
,
Gasmi
B
,
Munn
DH
,
Allison
JP
, et al
Tumor-expressed IDO recruits and activates MDSCs in a Treg-dependent manner
.
Cell Rep
2015
;
13
:
412
24
.
82.
Gajewski
TF
. 
Identifying and overcoming immune resistance mechanisms in the melanoma tumor microenvironment
.
Clin Cancer Res
2006
;
12
:
2326s
30s
.
83.
Jandus
C
,
Bioley
G
,
Speiser
DE
,
Romero
P
. 
Selective accumulation of differentiated FOXP3(+) CD4 (+) T cells in metastatic tumor lesions from melanoma patients compared to peripheral blood
.
Cancer Immunol Immunother
2008
;
57
:
1795
805
.
84.
Viguier
M
,
Lemaitre
F
,
Verola
O
,
Cho
MS
,
Gorochov
G
,
Dubertret
L
, et al
Foxp3 expressing CD4+CD25high regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells
.
J Immunol
2004
;
173
:
1444
53
.
85.
Strauss
L
,
Bergmann
C
,
Szczepanski
M
,
Gooding
W
,
Johnson
JT
,
Whiteside
TL
. 
A unique subset of CD4+CD25highFoxp3+ T cells secreting interleukin-10 and transforming growth factor-beta1 mediates suppression in the tumor microenvironment
.
Clin Cancer Res
2007
;
13
:
4345
54
.
86.
Wang
HY
,
Lee
DA
,
Peng
G
,
Guo
Z
,
Li
Y
,
Kiniwa
Y
, et al
Tumor-specific human CD4+ regulatory T cells and their ligands
.
Immunity
2004
;
20
:
107
18
.
87.
Harlin
H
,
Kuna
TV
,
Peterson
AC
,
Meng
Y
,
Gajewski
TF
. 
Tumor progression despite massive influx of activated CD8+ T cells in a patient with malignant melanoma ascites
.
Cancer Immunol Immunother
2006
;
55
:
1185
97
.
88.
Wei
SC
,
Levine
JH
,
Cogdill
AP
,
Zhao
Y
,
Anang
NAS
,
Andrews
MC
, et al
Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade
.
Cell
2017
;
170
:
1120
33
.
e17
.
89.
Simeone
E
,
Gentilcore
G
,
Giannarelli
D
,
Grimaldi
AM
,
Caracò
C
,
Curvietto
M
, et al
Immunological and biological changes during ipilimumab treatment and their potential correlation with clinical response and survival in patients with advanced melanoma
.
Cancer Immunol Immunother
2014
;
63
:
675
83
.
90.
Umansky
V
,
Shevchenko
I
,
Bazhin
AV
,
Utikal
J
. 
Extracellular adenosine metabolism in immune cells in melanoma
.
Cancer Immunol Immunother
2014
;
63
:
1073
80
.
91.
Young
A
,
Ngiow
SF
,
Madore
J
,
Reinhardt
J
,
Landsberg
J
,
Chitsazan
A
, et al
Targeting adenosine in BRAF-mutant melanoma reduces tumor growth and metastasis
.
Cancer Res
2017
;
77
:
4684
96
.
92.
Reinhardt
J
,
Landsberg
J
,
Schmid-Burgk
JL
,
Ramis
BB
,
Bald
T
,
Glodde
N
, et al
MAPK signaling and inflammation link melanoma phenotype switching to induction of CD73 during immunotherapy
.
Cancer Res
2017
;
77
:
4697
709
.
93.
Shin
DS
,
Zaretsky
JM
,
Escuin-Ordinas
H
,
Garcia-Diaz
A
,
Hu-Lieskovan
S
,
Kalbasi
A
, et al
Primary resistance to PD-1 blockade mediated by JAK1/2 mutations
.
Cancer Discov
2017
;
7
:
188
201
.
94.
Patel
SJ
,
Sanjana
NE
,
Kishton
RJ
,
Eidizadeh
A
,
Vodnala
SK
,
Cam
M
, et al
Identification of essential genes for cancer immunotherapy
.
Nature
2017
;
548
:
537
42
.
95.
Bhatt
AP
,
Redinbo
MR
,
Bultman
SJ
. 
The role of the microbiome in cancer development and therapy
.
CA Cancer J Clin
2017
;
67
:
326
44
.
96.
Wargo
JA
,
Gopalakrishnan
V
,
Spencer
C
,
Karpinets
T
,
Reuben
A
,
Andrews
MC
, et al
Association of the diversity and composition of the gut microbiome with responses and survival (PFS) in metastatic melanoma (MM) patients (pts) on anti-PD-1 therapy
.
J Clin Oncol
35
, 
2017
(
suppl; abstr 3008
).
97.
Chaput
N
,
Lepage
P
,
Coutzac
C
,
Soularue
E
,
Le Roux
K
,
Monot
C
, et al
Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab
.
Ann Oncol
2017
;
28
:
1368
79
.
98.
Vetizou
M
,
Pitt
JM
,
Daillere
R
,
Lepage
P
,
Waldschmitt
N
,
Flament
C
, et al
Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota
.
Science
2015
;
350
:
1079
84
.
99.
Restifo
NP
,
Marincola
FM
,
Kawakami
Y
,
Taubenberger
J
,
Yannelli
JR
,
Rosenberg
SA
. 
Loss of functional beta 2-microglobulin in metastatic melanomas from five patients receiving immunotherapy
.
J Natl Cancer Inst
1996
;
88
:
100
8
.
100.
del Campo
AB
,
Kyte
JA
,
Carretero
J
,
Zinchencko
S
,
Mendez
R
,
Gonzalez-Aseguinolaza
G
, et al
Immune escape of cancer cells with beta2-microglobulin loss over the course of metastatic melanoma
.
Int J Cancer
2014
;
134
:
102
13
.
101.
Zaretsky
JM
,
Garcia-Diaz
A
,
Shin
DS
,
Escuin-Ordinas
H
,
Hugo
W
,
Hu-Lieskovan
S
, et al
Mutations associated with acquired resistance to PD-1 blockade in melanoma
.
N Engl J Med
2016
;
375
:
819
29
.
102.
Dutta
P
,
Li
WX
. 
Role of the JAK-STAT signalling pathway in cancer
.
eLS2013
.
DOI: 10.1002/9780470015902.a0025214
.
103.
Sharma
P
,
Hu-Lieskovan
S
,
Wargo
JA
,
Ribas
A
. 
Primary, adaptive, and acquired resistance to cancer immunotherapy
.
Cell
2017
;
168
:
707
23
.
104.
Mellman
I
,
Coukos
G
,
Dranoff
G
. 
Cancer immunotherapy comes of age
.
Nature
2011
;
480
:
480
9
.
105.
Koyama
S
,
Akbay
EA
,
Li
YY
,
Herter-Sprie
GS
,
Buczkowski
KA
,
Richards
WG
, et al
Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints
.
Nat Commun
2016
;
7
:
10501
.
106.
Taube
JM
,
Young
GD
,
McMiller
TL
,
Chen
S
,
Salas
JT
,
Pritchard
TS
, et al
Differential expression of immune-regulatory genes associated with PD-L1 display in melanoma: implications for PD-1 pathway blockade
.
Clin Cancer Res
2015
;
21
:
3969
76
.
107.
Woo
SR
,
Turnis
ME
,
Goldberg
MV
,
Bankoti
J
,
Selby
M
,
Nirschl
CJ
, et al
Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape
.
Cancer Res
2012
;
72
:
917
27
.
108.
Foy
SP
,
Sennino
B
,
dela Cruz
T
,
Cote
JJ
,
Gordon
EJ
,
Kemp
F
, et al
Poxvirus-based active immunotherapy with PD-1 and LAG-3 dual immune checkpoint inhibition overcomes compensatory immune regulation, yielding complete tumor regression in mice
.
PLoS One
2016
;
11
:
e0150084
.
109.
Ascierto
PA
,
Melero
I
,
Bhatia
S
,
Bono
P
,
Sanborn
RE
,
Lipson
EJ
, et al
Initial efficacy of anti-lymphocyte activation gene-3 (anti–LAG-3; BMS-986016) in combination with nivolumab (nivo) in pts with melanoma (MEL) previously treated with anti–PD-1/PD-L1 therapy
.
J Clin Oncol
35
, 
2017
(
suppl; abstr 9520
).
110.
Sakuishi
K
,
Apetoh
L
,
Sullivan
JM
,
Blazar
BR
,
Kuchroo
VK
,
Anderson
AC
. 
Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity
.
J Exp Med
2010
;
207
:
2187
94
.
111.
Ngiow
SF
,
von Scheidt
B
,
Akiba
H
,
Yagita
H
,
Teng
MW
,
Smyth
MJ
. 
Anti-TIM3 antibody promotes T cell IFN-gamma-mediated antitumor immunity and suppresses established tumors
.
Cancer Res
2011
;
71
:
3540
51
.
112.
Gangadhar
TC
,
Hamid
O
,
Smith
DC
,
Bauer
TM
,
Wasser
JS
,
Olszanski
AJ
, et al
Epacadostat plus pembrolizumab in patients with advanced melanoma and select solid tumors: updated phase 1 results from ECHO-202/KEYNOTE-037
.
Ann Oncol
2016
;
27
(
6_suppl
):
1110PD
PD
.
113.
Gangadhar
TC
,
Schneider
BJ
,
Bauer
TM
,
Wasser
JS
,
Spira
AI
,
Patel
SP
, et al
Efficacy and safety of epacadostat plus pembrolizumab treatment of NSCLC: preliminary phase I/II results of ECHO-202/KEYNOTE-037
.
J Clin Oncol
35
, 
2017
(
suppl; abstr 9014
).
114.
Smith
DC
,
Gajewski
T
,
Hamid
O
,
Wasser
JS
,
Olszanski
AJ
,
Patel
SP
, et al
Epacadostat plus pembrolizumab in patients with advanced urothelial carcinoma: preliminary phase I/II results of ECHO-202/KEYNOTE-037
.
J Clin Oncol
35
, 
2017
(
suppl; abstr 4503
).
115.
Spiotto
M
,
Fu
YX
,
Weichselbaum
RR
. 
The intersection of radiotherapy and immunotherapy: mechanisms and clinical implications
.
Sci Immunol
2016
;
1
:
eaag1266
.
116.
Grimaldi
AM
,
Simeone
E
,
Giannarelli
D
,
Muto
P
,
Falivene
S
,
Borzillo
V
, et al
Abscopal effects of radiotherapy on advanced melanoma patients who progressed after ipilimumab immunotherapy
.
Oncoimmunology
2014
;
3
:
e28780
.
117.
Twyman-Saint Victor
C
,
Rech
AJ
,
Maity
A
,
Rengan
R
,
Pauken
KE
,
Stelekati
E
, et al
Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer
.
Nature
2015
;
520
:
373
7
.
118.
Woods
DM
,
Sodre
AL
,
Villagra
A
,
Sarnaik
A
,
Sotomayor
EM
,
Weber
J
. 
HDAC inhibition upregulates PD-1 ligands in melanoma and augments immunotherapy with PD-1 blockade
.
Cancer Immunol Res
2015
;
3
:
1375
85
.
119.
Goswami
S
,
Zhao
H
,
Zhang
X
,
Sharma
P
. 
Epigenetic changes in T cells in response to immune checkpoint blockade
.
J Clin Oncol
34
, 
2016
(
suppl; abstr 11549
).