We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

The complex role of EZH2 in the tumor microenvironment: opportunities and challenges for immunotherapy combinations

    Jing Qiu

    Oncology R&D, Pfizer Worldwide Research & Development, San Diego, CA 92121, USA

    ,
    Shikhar Sharma

    Oncology R&D, Pfizer Worldwide Research & Development, San Diego, CA 92121, USA

    ,
    Robert A Rollins

    Oncology R&D, Pfizer Worldwide Research & Development, Pearl River, NY 10965, USA

    &
    Thomas A Paul

    *Author for correspondence:

    E-mail Address: Thomas.A.Paul@pfizer.com

    Oncology R&D, Pfizer Worldwide Research & Development, San Diego, CA 92121, USA

    Published Online:https://doi.org/10.4155/fmc-2020-0072

    Abstract

    Immune dysfunction in the tumor microenvironment occurs through epigenetic changes in both tumor cells and immune cells that alter transcriptional programs driving cell fate and cell function. Oncogenic activation of the histone methyltransferase EZH2 mediates gene expression changes, governing tumor immunogenicity as well as differentiation, survival and activation states of immune lineages. Emerging preclinical studies have highlighted the potential for EZH2 inhibitors to reverse epigenetic immune suppression in tumors and combine with immune checkpoint therapies. However, EZH2 activity is essential for the development of lymphoid cells, performing critical immune effector functions within tumors. In this review, we highlight the complexity of EZH2 function in immune regulation which may impact the implementation of combination with immunotherapy agents in clinic.

    Tumor initiation and evolution requires a series of genetic and epigenetic changes which enable uncontrolled cell proliferation while limiting surveillance from specialized components of the immune system capable of eliciting antitumor responses. Epigenetic enzymes including DNA methyltransferases, histone deacetylases, histone acetyltransferases, histone methyltransferases, histone demethylases and chromatin remodeling enzymes have been implicated as critical regulators of the cellular transcriptional states that establish the equilibrium between tumor cells and their immune microenvironment [1–3]. Due to the fundamental role of epigenetic regulation in controlling cellular plasticity in both tumor cells and immune cells, increasing evidence has emerged around the role of epigenetic regulators in mediating multiple pathways of immune evasion and resistance toward immune checkpoint therapies [4,5]. Based on these observations. ongoing clinical trials are currently exploring therapeutic targeting of these classes of enzymes as opportunities to reverse epigenetic resistance mechanisms to immune checkpoint therapies including anti-PD-1, anti-PD-L1 and anti-CTLA-4 [6].

    Approaches targeting epigenetic regulators for immune therapy combinations require consideration of the diverse functional role of these enzymes and protein complexes in the heterogeneous cell lineages that comprise the innate and adaptive immune system. Ideally, epigenetic drug combinations with immune checkpoint inhibitors should enhance the immunogenicity of tumors, sustain effector functions of T cells and reprogram suppressive immune cell types in the tumor microenvironment. Paradoxically, many epigenetic targets being considered for immune therapy combinations have critical roles in immune cell differentiation, maturation, and function required for antitumor activity. Therefore, it is important to define the broader impact of inhibiting these targets in multiple cell types within the tumor microenvironment.

    In this review, we highlight emerging data supporting utilization of inhibitors of the histone methyltransferase EZH2 in clinical immunotherapy applications. We will discuss increasing evidence for the role of dysregulation of EZH2 function as a key mediator of tumor-intrinsic programs driving immune-suppression. Additionally, we describe the role of EZH2 in immune cell populations interfacing with the tumor microenvironment and how epigenetic changes within these cell types may also contribute to suppressive phenotypes observed in tumors. Finally, we will discuss the complexity of effects anticipated from inhibiting EZH2 in both tumor and immune cells and how these may both productively and negatively impact the clinical application of EZH2 inhibitors in cancer therapy.

    Role of EZH2 in epigenetic regulation

    Epigenetic regulation refers to a physiological process of stable alternations in gene expression patterns without changing underlying DNA sequence. DNA methylation, histone post-translational modifications including phosphorylation, acetylation, ubiquitination, methylation at enhancers and promoters and the activity of chromatin remodeling enzymes are believed to play a key role in the control of gene expression in response to developmental and environmental changes [7–9]. The histone methyltransferase EZH2 is a key regulator of cellular epigenetic states. As the core enzymatic subunit of PRC2 consisting of EZH2, EED, SUZ12, and RbAp46/48, EZH2 mediates H3K27me3 to influence chromatin compaction that promotes transcriptional silencing at targeted genes [10,11]. A paralog of EZH2, EZH1, has also been isolated in PRC2 complexes and appears to show a distinct expression pattern compared with EZH2. While EZH2 is highly expressed in proliferating cells, EZH1 shows abundant expression primarily in nondividing cells. As PRC2 complexes with EZH1 possess low methyltransferase activity compared with the PRC2 complex containing EZH2, nonenzymatic functions have also been ascribed to EZH1-containing PRC2 complexes [12,13].

    EZH2 and PRC2 members were initially identified in Drosophila where they play a critical role during embryonic development, regulating accurate gene expression to facilitate various biological functions, including cell proliferation, lineage commitment, X-chromosome inactivation and stem cell self-renewal. EZH2 mediated H3K27me3 to represses transcription of numerous groups of developmental genes like the HOX clusters and genes encoding the homeodomain-containing transcriptional factors. In adult tissues, EZH2 functions in maintaining homeostasis of various tissues through control of gene expression programs in tissue-specific stem cells. Through regulation of developmental gene programs, EZH2 function is also critical in regulating early and late stage of hematopoiesis.

    Dysregulation of EZH2 in cancer

    Epigenetic dysregulation of tumors, evidenced by changes in epigenetic modifications at gene promoters and enhancers, promotes transcriptional changes that shape oncogenic gene expression programs required for tumor cell growth and survival. Changes in epigenetic regulation in tumors have been directly linked to molecular alterations of chromatin regulating enzymes themselves due to mutation, overexpression, translocation or misrecruitment.

    EZH2 dysfunction has been identified as an important oncogenic mechanism in both hematological and solid tumors [14,15]. The best studied example of EZH2 alterations occur in B-cell lymphomas, follicular lymphoma (FL) and diffuse large B-cell lymphoma (DLBCL), in which recurrent heterozygous gain-of-function somatic mutations in EZH2 catalytic SET domain have been identified in 30% of germinal center (GC) B-cell-like subtype DLBCLs and 27% of FLs. These mutations enhance the efficiency of EZH2 activity toward H3K27 trimethylation resulting in more pronounced repression of EZH2 target genes. Mechanistically, these mutations behave as direct drivers of oncogenesis. Mice engineered to express mutant EZH2Y641 in GC B cells develop GC hyperplasia and accumulate high levels of H3K27me3. EZH2 gain-of-function mutations confer a gene expression signature featuring repression of genes involved in B-cell terminal differentiation and proliferation checkpoints. This observation is consistent with the critical role of EZH2 function during normal B-cell GC maturation. Within B-cell lineages, EZH2 was shown to be highly expressed in progenitor cells however decreased in expression in differentiated and mature B cells [16]. Upon activation, B cells were found to massively upregulate EZH2 expression during GC formation and immunoglobulin affinity maturation [17]. EZH2 prevents terminal differentiation of GC B cells and sustains GC B-cell survival [18–20]. Thus, EZH2 gain-of-function mutations likely serve to maintain expression of B-cell GC maturation programs which sustain cell proliferation and survival.

    In addition to EZH2 activating mutations, overexpression of EZH2 has been discovered in over 80% B-cell lymphomas including Burkitt lymphomas, FLs [21,22]. Similarly, overexpression of EZH2 has also been identified in mantle cell lymphoma in which the transcription factor c-MYC contributes to EZH2 upregulation via repression of the EZH2 targeting miR-26a microRNA [23,24]. A correlation between elevated EZH2 expression and various myeloid malignancies has been described in patients. For instance, EZH2 is commonly overexpressed in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) along with PRC1 members RING1 and BMI1, driving abnormal epigenetic repression of the tumor suppressor p15 (INK4B) [25]. In another study, c-MYC dysfunction has been linked to the enhancement of EZH2 expression in AML [26]. Moreover, the inhibition of EZH2 in a murine AML model prevented cancer progression by driving myeloid cell differentiation [27]. In contrast to the oncogenic function of EZH2 in cancers described above, potential tumor suppressor roles of EZH2 have also been observed in certain hematological cancers highlighting its highly lineage-specific function. Inactivation of EZH2 due to genomic aberrations including allelic deletion and frameshift mutations were found in a subset of patients with MDS, myeloproliferative neoplasms and MDS/myeloproliferative neoplasm overlap disorder [28,29]. Loss-of-function mutations or deletion of EZH2 and other subunits (e.g., SUZ12 and EED) of PRC2 have also been identified in human T-cell acute lymphoblastic leukemia [30,31]. Altogether, EZH2 exhibits dual roles as an oncogene and tumor suppressor in a context and lineage-specific manner leading to diverse hematological malignancies.

    EZH2 overexpression has been observed in several solid tumors including prostate cancer, endometrial cancer, small-cell-lung cancer (SCLC), breast cancer and melanoma. In these tumors EZH2 overexpression is associated with a more aggressive and advanced form of each disease [10,32–37]. In prostate cancer, EZH2 has been shown to regulate the proper chromatin binding of androgen receptor in castration-resistant disease. Notably, EZH2 can also act independently of PRC2 and/or its histone methyltransferase activities in breast and prostate cancer [25,38–41].

    Dysregulation of EZH2 function can occur through mutations in epigenetic enzyme complexes that antagonize the effects of H3K27 methylation. Recurrent cancer mutations in the H3K27 histone demethylase UTX (ubiquitously transcribed tetratricopeptide repeat gene on X chromosome) and the chromatin remodeling complexes switch/sucrose non-fermentable (SWI/SNF) are associated with altered EZH2 functions. This is best exemplified in malignant rhabdoid tumors where loss of SNF5 (INI1) results in the altered genomic occupancy of the repressive chromatin mark deposited by PRC2 at the H3K27me3 residues, leading to the repression of lineage-specific targets [42–45].

    Epigenetic regulation in tumor immunity

    In addition to the direct impact on tumor cell growth and survival, epigenetic changes in cancer cells can elicit a variety of mechanisms mediating escape from recognition and elimination by the immune system [2,46–49]. These mechanism include loss of expression of tumor-associated antigens or neoantigens, impairment of cell surface antigen presentation, changes in expression of immunosuppressive molecules and proinflammatory cytokines and aberrant expression of checkpoint pathway proteins such as PD-L1 [50–53].

    Epigenetic changes have also been observed in the many cell types that comprise the tumor microenvironment including tumor-infiltrating immune cells. Chronic antigen stimulation of tumor-associated CD8+ T cells results in an ‘exhausted’ phenotype characterized by rearrangement in chromatin accessibility conferring altered transcriptional programs [47,48,54,55]. Changes in the epigenetic landscape have also been described in additional tumor-infiltrating effector immune cell populations including dendritic cells and natural killer (NK) cells where transcriptional changes confer gradual loss of their mature antigen-presenting function and cytotoxicity toward cancer cells [56–58]. Epigenetic regulators also functionally impact transcriptional programs driving increased differentiation and proliferation/survival of immunosuppressive cells such as myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages and Tregs in the tumor microenvironment [59–64]. Within the tumor microenvironment, signaling molecules tied to chronic inflammation, hypoxia and altered metabolism have been associated with functional changes in the activity of epigenetic regulators influencing the production of cytokines, chemokines, growth factors and adhesion molecules [65,66].

    EZH2 functions as an essential mediator of tumor immunity

    Epigenetic changes conferred by EZH2 dysregulation have been identified as a major mechanism of transcriptional repression of genes that confer immunogenicity to tumor cells. This is best exemplified by the role of EZH2 in transcriptional repression of genes involved in antigen presenting pathways in cancer [67,68]. In a recent functional genomics screen conducted in K562 leukemia cells, inhibition of EZH2 and other PRC2 genes were identified as modulators of the IFN-γ response leading to MHC-I upregulation [69]. These results were further substantiated by Burr et al. who demonstrated a pivotal role for EZH2-mediated H3K27me3 repressive marks in maintenance of MHC-I silencing in MHC-I-deficient cancers. Interestingly, in neuroendocrine tumors including SCLC and neuroblastoma, EZH2 function appears to hijack a conserved developmental program in neural progenitor cells to confer immune privilege via MHC-I down-regulation. Genetic inhibition of EZH2 in a mouse genetically-engineered model of SCLC led to MHC-I up-regulation and immune-mediated tumor regression [70].

    In uveal melanoma, a malignant tumor of the eye, EZH2-mediated repression of the MHC class II master control transcription factor, CIITA, prevents expression of MHC-II molecules after IFN-γ induction. These observations were extended into additional tumor types in a recent study, by Ennishi and colleagues, who noted that EZH2 activating mutations were enriched in 30% of DLBCL patients which lack MHC-II cell surface expression [64]. Moreover, 77% of EZH2 mutated cases lost either MHC-I and/or MHC-II expression in DLBCL, suggesting epigenetic repression occurs in both class I and II presentation pathways. Experimentally, introduction of either EZH2Y641N or EZH2Y641F mutations into B-cell lymphoma increases H3K27me3 at the promoters of NLRC5 and CIITA which could be reversed by EZH2 inhibitor treatment. These results indicate EZH2 inhibition could be a therapeutically useful approach for restoration of MHC expression.

    EZH2 function has also been identified as a mechanism of transcriptional repression of chemokines and cytokines in various cancers. In a mouse MYC-driven prostate cancer model, IFNGR1 was identified as a direct target of EZH2 transcriptional repression. In this model, EZH2 overexpression was found to be regulated in a MYC-dependent manner consistent with observations in metastatic prostate cancer patients. Therapeutic treatment of mouse MYC-driven prostate cancer tumors with EZH2 inhibitors enhanced antitumor effects in response to IFN-γ treatment [71]. Overexpression of EZH2 in tumors has also been linked to the downregulation of Th1 chemokines CXCL9 and CXCL10, which are essential for mediating T cell migration to tumor sites for effective tumor killing [50,72]. In the ID8 ovarian cancer syngeneic tumor model, EZH2 inhibition in combination with DNA methyltransferase inhibitors (5-AZA dC) promoted re-expression of Th1 chemokines resulting in increased tumor-infiltrating T cells and tumor regression when combined with immune checkpoint inhibition. In a separate study, Zingg et al. demonstrated that EZH2 overexpression may serve as an immune escape mechanism during immunotherapy. In the B16-F10 mouse melanoma model treated with CTLA-4 or IL-2 blockade, increased TNF-α production from tumor-infiltrating T cells resulted in increased EZH2 expression causing loss of dominant tumor antigens and silencing of the antigen-processing and -presenting machinery [73]. Genetic or pharmacological inactivation of EZH2 in this model produced a synergistic effect with anti-CTLA-4 and IL-2, suppressing tumor growth. EZH2-mediated transcriptional repression has also been observed at the promoters of many IFN-γ stimulated genes, where EZH2 function antagonizes the chromatin remodeling activity of the SWI/SNF complex [74]. Thus, EZH2 regulation may broadly impact IFN-γ signaling pathways within tumor cells.

    Immune recognition of transformed/mutated self-peptides is critical for host protection against cancer [75]. Tumor-associated antigens expressed on their cell surface can be presented by MHC molecules, which when recognized by T cells, mediate tumor rejection. Many recent studies have illustrated the importance of epigenetic regulation of tumor antigen expression and presentation. For example, X-linked tumor-associated antigens (e.g., MAGE-A1 and NY-ESO-1) showed downregulated expression via epigenetic repression, including EZH2 function, in several tumors such as ovary, lung, pancreas cancer, as well as melanoma and myeloma [76–78]. EZH2 was also identified in the epigenetic silencing of a specific type of IFN-γ-inducible antisense 3′-UTR endogenous retroviral elements called SPARCS [79]. Derepression of SPARCS following EZH2 inhibition results in double-stranded RNA generation following IFN-γ exposure and engagement of MAVS and STING innate antitumor immunity pathways.

    Taken together these observations highlight tumor immune suppression as an important consequence of EZH2 function in the cancer. It is anticipated that EZH2 inhibition should elicit gene expression changes in tumors by increasing the recognition and recruitment of the immune system to the tumor microenvironment. However, certain tumors with genetic lesions including loss-of-function mutations in JAK1 or JAK2, deletion of the wild-type allele of JAK1 or JAK2 and truncation in the antigen-presenting protein B2M may be ‘hardwired’ for defects in antigen presentation that may not be overcome by EZH2 inhibition [80]. Moreover, transcriptional repression of genes may feature additional types of epigenetic repression such as DNA methylation or H3K9me3 that will not be overcome by EZH2 inhibition alone. Along these lines, the combination of EZH2 inhibitors with DNA methyltransferase inhibitors was shown to enhance reactivation of CXCL9 and CXCL10 in ovarian tumor models [50].

    Role of EZH2 in regulation of effector T-cell function

    Immune cell development is tightly regulated by epigenetic regulators that maintain gene expression programs governing self-renewal and cell differentiation in stem and progenitor cell lineages. The important role of EZH2 function as an essential regulator in hematopoiesis has been highlighted by multiple mouse genetic knockout models, demonstrating that EZH2 controls the balance between self-renewal and multipotency of hematopoietic stem cells through H3K27me3 suppression of developmental gene programs [81–83].

    Consistent with the function of EZH2 in hematopoietic cell development, EZH2 also appears to be an important regulator of the lymphoid and myeloid cell types that comprise the tumor microenvironment. During the T-cell maturation process, EZH2-mediated transcriptional regulation plays a key role in the establishment of epigenetic programs necessary for lineage specification and divergent T-cell function (Figure 1). Tarakhovsky and colleagues first demonstrated the importance of EZH2 function in early T-cell development through adoptive transfer experiments utilizing conditional EZH2 knockout (KO) mice. In these experiments bone marrow derived from EZH2 knockout mice failed to support normal thymocyte development through arrest at the early CD4, CD8 double negative CD44intCD25hi stage [3]. Interestingly, this observed defect in T-cell maturation was found to be dependent on a unique cytosolic function of EZH2 as opposed to the canonical role as a histone methyltransferase.

    Figure 1. The function of enhancer of zeste homolog 2 in T-cell differentiation.

    In naive CD4+ T cells (left), EZH2 represses the expression of transcriptional factors T-bet and GATA3; and Th1 and Th2 cytokines such as IFN-γ, IL-4 and IL-5, which inhibit Th1 and Th2 polarization. EZH2 is required to stabilize FOXP3 expression in regulatory T cells, which is important to maintain their suppressive function upon activation. In naive CD8+ T cells (right), EZH2 silences both pro-effector and pro-memory genes to maintain developmental plasticity. In differentiated CD8+ effector T cells, EZH2 selectively inhibits memory precursor signature genes such as Id3 to restrict memory fate while EZH2 mediated suppression is not required in memory precursor cells to maintain their pluripotency.

    Treg: Regulatory T cell.

    The chromatin regulatory function of EZH2 also appears critical for the specification, functional regulation and survival of both CD4+ and CD8+ T cells (Figure 2). In mouse tumor models, tumor-infiltrating CD8+ T cells expressing EZH2 exhibited a survival advantage due to up-regulation of BCL-2, which is consistent with the anti-apoptotic role of BCL-2 in human effector T cells [84]. Consistent with this result, EZH2-deficient CD8+ T cells were also found to display enhanced T cell apoptosis during immune response [85,86]. Mechanistically, EZH2 repression of the Notch pathway suppressors NUMB and FBXW27 activates the Notch pathway in CD8+ T cells, stimulating T-cell cytokine expression and survival via BCL-2 signaling.

    Figure 2. Summary of enhancer of zeste homolog 2-mediated immune suppression/response in the tumor microenvironment.

    In the tumor microenvironment (left), EZH2 expression in tumor cells downregulates the expression of MHC class I and class II molecules as well as TAAs to escape the immune recognition. EZH2 suppresses the expression of CXCL9 and CXCL10 chemokines to inhibit T-cell infiltration to the tumor and also inhibits IFN-γ production in Th1 CD4+ T cells. EZH2 is also required on Treg to stable FOXP3 to maintain their inhibitory function. Upon EZH2 inhibition (right), many EZH2-mediated mechanisms of immune suppression on tumor cells are re-activated including the re-expression of TAAs and antigen presentation machinery. The blockage of EZH2 can promote antitumor immune response through increased infiltration of functional CD8+ T cells by the disruption of Treg function as well as the elevated expression of CXCL9 and CXCL10. Th1 CD4+ T cells and NK cells also exhibit enhanced cytotoxic activity following EZH2 inhibition. However, the inhibition of EZH2 can lead to the accumulation of MDSCs in the tumor sites which results in decreased IFN-γ production by infiltration T cells.

    DC: Dendritic cell; MDSC: Myeloid-derived suppressor cell; NK: Natural killer; TAA: Tumor-associated antigen; Treg: Regulatory T cell.

    The expression level of EZH2 in tumor-associated CD8+ T cells can be impacted by glucose restriction in the tumor microenvironment. Low glucose conditions maintain high expression of microRNAs miR-101 and miR-26a, which constrain expression of EZH2 [87]. This finding was consistent with a previous study conducted by Long et al., which identified upregulation of miR-26a in CD8+ T cells to be associated with decreased EZH2 expression and impaired effector T-cell survival and function [88].

    Using a mouse lymphocytic choriomeningitis virus (LCMV) infection model, Kakaradov et al. identified an important role of EZH2 in CD8+ T cell fate specification. EZH2, along with genes encoding other PRC2 components EED and SUZ12, is highly expressed in effector T cells relative to memory T cells upon LCMV challenge, suggesting a role of EZH2 in regulating terminal effector cell differentiation. High expression of EZH2 in terminally differentiated effector cells was associated with gains in H3K27me3 at promoters including memory associated transcription factors TCF7 and EOMES, suggesting that epigenetic silencing may be more crucial for the differentiation of terminal effector cells compared with that of memory cells [89]. Functionally, CD8+ T cells adoptively transferred from EZH2-/-.mice showed reduced expansion compared with control T cells when challenged with LCMV and exhibited an impaired capacity to secrete inflammatory cytokines. It is of interest that EZH2-deficient CD8+ T cells were not impaired in their ability to undergo activation or proliferation, however, demonstrated increased propensity to undergo apoptosis after antigen stimulation. In an independent study using a different EZH2 knockout transgenic mouse model featuring a tamoxifen Gzmb promoter-driven Cre, EZH2 was similarly found to be required for the expansion and terminal differentiation of effector CD8+ T cells when challenged with LCMV. Compared with the previous study EZH2-/- CD8+ T cells did not appear impacted in their survival, however, appeared skewed in their differentiation, acquiring memory-like states at the expense of effector states [90].

    Contrasting results for the role of EZH2 in T cell maturation were observed in a different mouse model employing melanoma-associated antigen gp100-specific CD8+ T-cell receptor-transgenic Pmel-1 cells. In this study EZH2-/- naive CD8+ T cells were found to have restrained memory differentiation and increased terminal effector differentiation compared with controls following adoptive transfer into B16 melanoma tumor bearing mouse [91]. Similarly, Goswami et al. did not observe any deleterious effects on effector T cells when studying the in vitro and in vivo effects of the EZH2 inhibitor CPI-1205 in human CD8+ T cells. Enhanced cytotoxicity and increased IFN-γ and TNF-α levels were noted in the supernatant of activated human T cells following CPI-1205 treatment, which suggested that these cells have enhanced effector function [92]. Moreover, the authors found in vivo EZH2 inhibitor treatment led to T cell-mediated tumor growth inhibition in mouse syngeneic tumor models MB49 bladder and B16-F10 melanoma either as monotherapy or in combination with anti-CTLA4. These results suggest survival of effector T-cells populations may not be detrimentally impacted by EZH2 inhibitors at least for shorter durations of treatment.

    Altogether, these findings highlight the importance of EZH2-mediated epigenetic regulation during CD8+ T-cell maturation however they also indicate that the function of EZH2 might have opposite roles during different phases of an immune response. It is clear from these studies that under certain circumstances inhibiting EZH2 may have a negative impact on effector T-cell development, function and survival and thus should be taken into consideration when exploring combination of EZH2 inhibitors with immuno-oncology agents.

    In addition to CD8+ T cells, the chromatin regulatory function of EZH2 appears critical for specification of naive CD4+ helper cells into effector lineages including Th1, Th2 and Th17 and their subsequent proliferation and survival following differentiation (Figure 1). Using a CD4-specific knockout of EZH2, Tumes et al. demonstrated that loss of EZH2 in naive CD4+ T cells was associated with differentiation skewing and lineage plasticity in both Th1 and Th2 cells [93]. Loss of EZH2-mediated H3K27me3 at the IFNγ, EOMES and TBX21 loci resulted in upregulation of IFN-γ production. EZH2 deficient CD4+ Th cells enhanced IL4, IL5 and IL13 cytokine production through upregulation of GATA3 expression under Th2 polarizing condition both in vitro and in vivo. Similarly, a separate study by Yang et al. identified EZH2-deficient CD4+ T cells as producing higher amounts of IFN-γ, IL-13 and IL-17 in Th1, Th2 and Th17 cells, respectively [94]. Although these data indicate that EZH2 loss promotes increased production and activation of CD4+ effector cells, Zhang et al. found loss of EZH2 also negatively impacted survival of Th1, Th2 and Th17 cells upon differentiation through reactivation of genes in apoptosis pathways. Consistent with this result, CD4+ T-cell-specific knockout of EZH2 in a mouse model of graft-versus-host disease protected mice from disease due to the inability of allogeneic effector CD4+ T-cell expansion [95,96]. Overall, these studies highlight the central role of EZH2 in instructing CD4+ T cell differentiation into Th1 and Th2 functional states as well as impacting their overall activation and survival. Recent data has highlighted the importance of both antigen-specific CD8+ and CD4+ T cells as well as MHC class II-restricted antigens for successful antitumor immune responses. As indicated in these genetic studies, inhibition of EZH2 may transiently increase CD4+ T-cell effector function, although longer duration of treatment may detrimentally impact the survival of effector CD4+ T-cell populations within the tumor microenvironment.

    EZH2 function in NK cells

    Although the activation program and recognition mechanism of NK cells are distinct from cytotoxic T cells, they share similar features of lytic granule exocytosis in the elimination of cancer cells [97]. Reduced numbers or impaired function of NK cells have displayed a profound impact on overall immune response, which is often associated with cancer progression [98]. Several studies have demonstrated the contribution of EZH2 epigenetic regulation on NK cell lineage commitment and functional maturation (Figure 2). An early study by Nagel et al. described the role of PRC2 in NK cell differentiation through regulation of HOXA9 and HOXA10 [99]. A subsequent study showed that the loss of EZH2 function enhances NK cell lineage commitment partially by promoting the survival of NK cell precursors. Further mechanistic studies revealed that genetic or pharmacological perturbation of EZH2 increases proliferation, activation and cytotoxicity of NK cells concomitant with upregulation of the IL-15 receptor CD12 and NK cell activating receptor NKG2D [100].

    More recent papers highlighted the immunosuppressive role of EZH2 in NK cell-mediated eradication of carcinoma cells. Bugide et al. discovered that in a mouse model of hepatocellular carcinoma, EZH2 serves as a transcriptional repressor of NKG2D ligands which enables evasion of tumor cells from immune recognition by NK cells thereby conferring resistance to NK cell-mediated cytotoxicity [101]. Inhibition of EZH2 in hepatocellular carcinoma cells lead to re-expression of NKG2D ligands such as ULBP1, MICA and MICB, enhancing the ability of NK cells to mediate tumor clearance. Similarly, EZH2 inhibition in a muscle invasive bladder cancer model not only limited proliferation of tumor cells in the context of KDM6A and SWI/SNF mutations, but also promoted NK-cell activity [102]. Interestingly, EZH2 inhibitor treatment in this model leads to upregulation of genes associated with activated NK signaling including MIP-1α, ICAM1, ICAM2 and CD86 and increased expression of IFN-γ. Although the exact mechanism of epigenetic regulation of NK cell maturation and cytotoxic activity in the tumor microenvironment is not understood, EZH2 has been presented as an exciting therapeutic target to enhance NK cell-mediated antitumor immunity.

    Potential for EZH2 inhibitors in reprogramming tumor immunosuppressive cells

    Tregs are a unique subset of CD4+ T cells defined by expression of the transcription FOXP3 with inhibitory function on effector T cells during immune response. The control of EZH2 in Treg cell function was first postulated by observations of significant EZH2 upregulation in activated Treg cells as well as co-immunoprecipitation experiments which demonstrated that EZH2 forms complexes with FOXP3 in activated mouse Tregs (Figure 1). Furthermore, EZH2 knockout prevented inducible Treg differentiation in vitro through loss of EZH2-mediated H3K27me3 at FOXP3-regulated target genes [103]. Deletion of EZH2 in CD4+ T cells in a mouse model of autoimmune colitis reduced FOXP3+ Treg numbers in the spleen and mesenteric lymph nodes and also impaired Treg cell function through reduced expression of FOXP3 [94]. Interestingly, induction of antibodies against IFN-γ and IL-4 can rescue FOXP3 expression due to EZH2 deficiency, suggesting that downregulation of FOXP3 upon loss of EZH2 in CD4+ T cells is possibly mediated by aberrant cytokine production [86,94]. In addition, EZH2-deficient T cells showed defects in maintenance of the Treg population due to instability of lineage-specific transcriptional program upon CD28 activation [104].

    More recently, studies by Wang et al. highlighted the critical role of EZH2 activity in mediating Treg function in anticancer immunity (Figure 2). Selective upregulation of EZH2 was observed in tumor-infiltrating Tregs but not in effector T cells and Treg cells in peripheral blood. Genetic deletion or pharmacological inhibition of EZH2 in vitro or in vivo destabilized FOXP3 expression thus promoting immune-mediated rejection of tumors in mouse syngeneic models [105]. These results were further confirmed by Goswami et al., who recently demonstrated that genetic depletion of EZH2 in Tregs or EZH2 catalytic inhibition using CPI-1205 elicited phenotypic and functional alterations of the Tregs leading to robust antitumor immunity. Collectively, EZH2-mediated epigenetic programs are essential for the lineage commitment as well as functional suppressive activity of Treg cells, providing rationale for exploring combination therapies of EZH2 inhibitors with immune checkpoint inhibitors [92,105]. In patients receiving ipilimumab (anti-CTLA) treatment, increased expression of EZH2 was observed in multiple immune cell types including CD4+ effector T cells, FOXP3+ Tregs and CD8+ T cells. Consistent with this result, T cells derived from CTLA-4-/- KO mice were found to overexpress EZH2 in a CD28-dependent manner [104,105]. Given the importance of FOXP3+ Tregs in tumor immunosuppression and the important role of EZH2 in regulation of FOXP3+ Tregs cells, EZH2 overexpression was postulated as a mechanism of resistance to anti-CTLA-4 therapy. To this extent, EZH2 inhibition combined with anti-CTLA-4 resulted in fewer FOXP3-expressing Treg cells and augmented antitumor responses compared with anti-CTLA-4 alone [73,92,105].

    MDSCs are a heterogeneous population consisting of granulocytes, macrophages or dendritic cells derived from immature myeloid cells with the capacity to suppress T cell functions [106]. Epigenetic changes influenced by the tumor microenvironment result in abnormal differentiation and function of myeloid cells leading to MDSCs. Huang et al. demonstrated that inhibition of EZH2 promotes the expansion of MDSCs during tumor development and attenuates antitumor immune responses (Figure 2). Treatment of mouse syngeneic tumor models with the EZH2 inhibitor GSK126 resulted in accumulation of CD11b+Gr-1+MDSCs in the tumor tissue as well as decreased IFN-γ producing CD8+ and CD4+ T cells [107]. Interestingly when MDSCs were depleted from the tumor microenvironment using chemotherapy agents, GSK126 promoted antitumor immune responses through increased infiltration of functional T cells into the tumor microenvironment. These data highlight the divergent roles of EZH2 function in various immune cell subtypes contributing to antitumor immune responses. Although EZH2 has been linked to the development of MDSCs that might tip the balance against antitumor immunity, the mechanistic understanding of how EZH2 regulates differentiation and migration in this population through DNA/histone methylation needs to be further elucidated.

    Targeting EZH2 in cancer therapy

    Unlike genetic mutations, epigenetic aberrations are reversible so targeting the relevant epigenetic factors by small molecules is potentially an efficient approach to ‘fix’ dysregulated gene/chromosome-regulatory systems caused by epigenetic changes in cancer. Along these lines, small molecule inhibitors of EZH2 and related PRC2 complexes, CPI-1205, CPI-0209, DS-3201, EPZ-6438 (tazemetostat), GSK-126 (GSK2816126), MAK683 and PF-06821497, have entered clinical trials targeting EZH2-driven epigenetic alterations in the regulation of cancer cell processes (Figures 3 & 4). These small molecules mainly inhibit EZH2 enzyme function, leading to reactivation of EZH2-target genes. DS-3201 and CPI-0209 are dual inhibitors of EHZ1 and EZH2 reported to have enhanced activity over EZH2-selective inhibitors via their targeting of potential H3K27me3 compensation driven by EZH1 paralog function [108]. Additionally, MAK683, an allosteric PRC2 inhibitor developed by Novartis, which targets the H3K27me3 binding pocket of EED, has also entered clinical studies [109].

    Figure 3. Clinical landscape around EZH2 inhibitor development.

    FDA approval January 2020.

    ALL: Acute lymphoblastic leukemia; AML: Acute myeloid leukemia; DLBCL: Diffuse large B-cell lymphoma; FL: Follicular lymphoma; mCRPC: Metastatic castration-resistant prostate cancer; mut.: Mutation; NHL: Non-Hodgkin lymphoma; R/R: Relapsed/refractory; SCLC: Small-cell lung cancer.

    Figure 4. Small molecule inhibitors of EZH2 and related polycomb repressive complex 2 members.

    Early clinical exploration around EZH2 inhibitors have focused on molecularly targeted patient populations in DLBCL and FL where activating mutations in EZH2 have been linked as drivers of disease. In FL, response rates to tazemetostat have been reported at 71% in patients harboring EZH2 activating mutations [110]. Interestingly, a 33% response rate observed in patients with wild-type EZH2 suggests broader function of EZH2 in the cell-of-origin of this disease. Additional clinical exploration for EZH2 inhibitors has focused largely on solid tumors with dysregulation of wild-type EZH2 function through overexpression (e.g., SCLC and prostate cancer) or mutations in other epigenetic remodeling complexes that antagonize EZH2 function such as BAP1 mutation (e.g., mesothelioma), SNF5/INI1 mutation (e.g., epithelioid sarcoma) [14,111]. In January 2020, tazemetostat was approved by the US FDA for patients with metastatic or locally advanced epithelioid sarcoma, marking the first clinical approval of an EZH2 inhibitor [112].

    While clinical data assessing the efficacy of EZH2 inhibitors in additional oncology indications is still emerging, the favorable safety profile of this class of drugs raises the prospects around the utility of EZH2 inhibitors in drug combinations with chemotherapies and other targeted therapies [113]. Indeed, numerous drug combinations are currently being assessed with EZH2 inhibitors to enhance responses in certain cancers where single agent responses have been limited (Figure 3). In a recent Phase II clinical trial in relapsed or refractory follicular lymphoma, 18% of patients dosed with tazemetostat reported grade ≥3 treatment-related adverse advents, most commonly thrombocytopenia and anemia. The lack of broader hematopoietic toxicity observations in early clinical trials with EZH2 inhibitors also highlights the potential for combinations with immune checkpoint inhibitors. Interestingly, a recent report by Gounder et al. highlighted an exceptional patient response to EZH2 inhibitors due to the activation of antitumor immunity, suggesting the potential for tumor extrinsic effects for this class of drugs [114]. In this patient, tazemetostat resulted in a significant increase in tumor-infiltrating CD8+ T cells and immune cells expressing checkpoint regulators PD-1 and LAG-3, suggesting EZH2 inhibition can promote a sustained antitumor response. It is of consideration that genetically engineered mouse models have demonstrated that dual inhibition of EZH1 and EZH2 present with additional hematopoietic defects, thus EZH1/EZH2 dual inhibitors, as well as EED inhibitors, may exhibit unique toxicities not previously observed with EZH2-selective inhibitors [115].

    Future perspective

    Based off the emerging clinical and preclinical data highlighting the combination potential of EZH2 inhibitors with immune checkpoint inhibitors described in this review, clinical trials have been initiated assessing the clinical combination of CPI-1205 with ipilimumab (NCT03525795) and tazemetostat with pembrolizumab (NCT03854474) or atezolizumab (NCT02220842). In these studies, it will be critical to assess whether the immune promoting effects of EZH2 inhibition will overcome the potential for immune suppressing effects anticipated in certain immune cell populations such as effector T cells. Like other epigenetic drugs previously pursued in the clinic, one possibility would be to explore dosing schedules that ‘prime’ the immune system to checkpoint inhibitors but remove drug prior to checkpoint therapy. Further assessment of the immunomodulatory effects of EZH2 inhibitors will help inform the best utilization of this promising class of drugs in the clinic.

    Executive summary

    Role of EZH2 in epigenetic regulation

    • EZH2 is a key epigenetic regulator of repressive H3K27 histone methylation controlling gene expression programs involved in cell differentiation, cell proliferation, stem cell function and immune regulation.

    Dysregulation of EZH2 in cancer

    • EZH2 dysfunction, including activating mutation and overexpression, has been discovered in a variety of hematological and solid tumors and impacts cancer gene expression programs.

    EZH2 functions as an essential mediator of tumor intrinsic & tumor extrinsic immunity

    • In certain tumor types, transcriptional regulation by EZH2 is a major mechanism of tumor immune suppression through repression of chemokine and cytokine genes, genes involved in tumor antigen expression and MHC presentation pathways.

    • Development and function of tumor-infiltrating lymphoid and myeloid cell types are dependent on the regulatory function of EZH2.

    Role of EZH2 in regulation of effector T-cell function

    • EZH2 controls gene expression programs required for cell specification in naive CD8+ T cells as they mature into effector CD8+ T cells in the context of infection and tumorigenesis. Inhibition of EZH2 may have a deleterious effect on effector CD8+ T-cell function.

    • In CD4+ T cells, EZH2 plays a central role in mediating cell differentiation programs required for Th1, Th2 and Th17 functional states through regulation of gene involved in lineage-specific cytokine production.

    EZH2 function in natural killer cells

    • EZH2 regulates genes involved in natural killer cell lineage commitment and maturation.

    • Inhibition of EZH2 has been shown to promote natural killer cell proliferation and cytotoxicity function.

    Potential for EZH2 inhibitors in reprogramming tumor immunosuppressive cells

    • Genetic and pharmacological inhibition of EZH2 activity in regulatory T cells reprograms their suppressive function through derepression of FOXP3+ driven gene programs.

    • EZH2 inhibition promotes the expansion of myeloid-derived suppressor cells thus may promote immunosuppression in the tumor microenvironment.

    Targeting EZH2 in cancer therapy

    • Small molecules inhibitors of EZH2 and related PRC2 members, EZH1 and EED, are under clinical evaluation as monotherapy and various drug combinations.

    • In preclinical models, EZH2 inhibitors have demonstrated immune-driven antitumor activity when combined with immune checkpoint inhibitors.

    • Given the lack of broader toxicity and potential activation of antitumor immunity, utilization of EZH2 inhibitors are currently being explored in combination with immune checkpoint inhibitors.

    Future perspective

    • Given the complex function of EZH2 in immune regulation, evaluation of the immune promoting and immune suppressive effects of EZH2 inhibition will be critical to assess the best clinical application of EZH2 inhibitors in cancer therapy.

    Author contributions

    J Qiu performed the initial draft and designed Figures 12. J Qiu and TA Paul wrote the manuscript. TA Paul designed Figure 3. S Sharma, RA Rollins and TA Paul revised the manuscript. All authors approved the manuscript.

    Acknowledgments

    The authors gratefully thank MA White for critical scientific insights on this review article.

    Financial & competing interests disclosure

    The authors of this manuscript are current employees of Pfizer, Inc. J Qiu is a postdoctoral fellow in the Pfizer WR&D Postdoc Program. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Dunn J, Rao S. Epigenetics and immunotherapy: the current state of play. Mol. Immunol. 87, 227–239 (2017).
    • 2. Loo Yau H, Ettayebi I, de Carvalho DD. The cancer epigenome: exploiting its vulnerabilities for immunotherapy. Trends Cell Biol. 29(1), 31–43 (2019).
    • 3. Su IH, Dobenecker MW, Dickinson E et al. Polycomb group protein EZH2 controls actin polymerization and cell signaling. Cell 121(3), 425–436 (2005).
    • 4. Chiappinelli KB, Zahnow CA, Ahuja N, Baylin SB. Combining epigenetic and immunotherapy to combat cancer. Cancer Res. 76(7), 1683–1689 (2016).
    • 5. Jones PA, Ohtani H, Chakravarthy A, De Carvalho DD. Epigenetic therapy in immune-oncology. Nat. Rev. Cancer 19(3), 151–161 (2019). •• Comprehensive review of rationale and strategies for applying epigenetic drugs in combination with immune checkpoint inhibitors.
    • 6. Mazzone R, Zwergel C, Mai A, Valente S. Epi-drugs in combination with immunotherapy: a new avenue to improve anticancer efficacy. Clin. Epigenetics 9, 59 (2017).
    • 7. Clapier CR, Cairns BR. The biology of chromatin remodeling complexes. Annu. Rev. Biochem. 78, 273–304 (2009).
    • 8. Tessarz P, Kouzarides T. Histone core modifications regulating nucleosome structure and dynamics. Nat. Rev. Mol. Cell Biol. 15(11), 703–708 (2014).
    • 9. Schubeler D. Function and information content of DNA methylation. Nature 517(7534), 321–326 (2015).
    • 10. Margueron R, Reinberg D. The polycomb complex PRC2 and its mark in life. Nature 469(7330), 343–349 (2011).
    • 11. Di Croce L, Helin K. Transcriptional regulation by Polycomb group proteins. Nat. Struct. Mol. Biol. 20(10), 1147–1155 (2013).
    • 12. Margueron R, Li G, Sarma K et al. EZH1 and EZH2 maintain repressive chromatin through different mechanisms. Mol. Cell. 32(4), 503–518 (2008).
    • 13. Dobenecker MW, Park JS, Marcello J et al. Signaling function of PRC2 is essential for TCR-driven T cell responses. J. Exp. Med. 215(4), 1101–1113 (2018).
    • 14. Kim KH, Roberts CW. Targeting EZH2 in cancer. Nat. Med. 22(2), 128–134 (2016). • Covers the biomarker-drivern startegies for pursuit of EZH2 inhibitors in select patient population harboring mutations in EZH2 or epigenetic enzymatic complex which interact with EZH2.
    • 15. Souroullas GP, Jeck WR, Parker JS et al. An oncogenic EZH2 mutation induces tumors through global redistribution of histone 3 lysine 27 trimethylation. Nat. Med. 22(6), 632–640 (2016).
    • 16. Su IH, Basavaraj A, Krutchinsky AN et al. EZH2 controls B cell development through histone H3 methylation and Igh rearrangement. Nat. Immunol. 4(2), 124–131 (2003).
    • 17. van Galen JC, Dukers DF, Giroth C et al. Distinct expression patterns of polycomb oncoproteins and their binding partners during the germinal center reaction. Eur. J. Immunol. 34(7), 1870–1881 (2004).
    • 18. Velichutina I, Shaknovich R, Geng H et al. EZH2-mediated epigenetic silencing in germinal center B cells contributes to proliferation and lymphomagenesis. Blood 116(24), 5247–5255 (2010).
    • 19. Beguelin W, Popovic R, Teater M et al. EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation. Cancer Cell 23(5), 677–692 (2013). • Experimental evidence for the oncogenic of EZH2 gain-of-function mutations and mechanistic linkage to normal function of EZH2 in B cell germinal center maturation.
    • 20. Caganova M, Carrisi C, Varano G et al. Germinal center dysregulation by histone methyltransferase EZH2 promotes lymphomagenesis. J. Clin. Invest. 123(12), 5009–5022 (2013).
    • 21. Abd Al Kader L, Oka T, Takata K et al. In aggressive variants of non-Hodgkin lymphomas, EZH2 is strongly expressed and polycomb repressive complex PRC1.4 dominates over PRC1.2. Virchows Arch. 463(5), 697–711 (2013).
    • 22. Yan J, Ng SB, Tay JL et al. EZH2 overexpression in natural killer/T-cell lymphoma confers growth advantage independently of histone methyltransferase activity. Blood 121(22), 4512–4520 (2013).
    • 23. Visser HP, Gunster MJ, Kluin-Nelemans HC et al. The polycomb group protein EZH2 is upregulated in proliferating, cultured human mantle cell lymphoma. Br. J. Haematol. 112(4), 950–958 (2001).
    • 24. Zhang X, Zhao X, Fiskus W et al. Coordinated silencing of MYC-mediated miR-29 by HDAC3 and EZH2 as a therapeutic target of histone modification in aggressive B-cell lymphomas. Cancer Cell 22(4), 506–523 (2012).
    • 25. Xu F, Li X, Wu L et al. Overexpression of the EZH2, RING1 and BMI1 genes is common in myelodysplastic syndromes: relation to adverse epigenetic alteration and poor prognostic scoring. Ann. Hematol. 90(6), 643–653 (2011).
    • 26. Salvatori B, Iosue I, Djodji Damas N et al. Critical role of c-myc in acute myeloid leukemia involving direct regulation of miR-26a and histone methyltransferase EZH2. Genes Cancer 2(5), 585–592 (2011).
    • 27. Tanaka S, Miyagi S, Sashida G et al. EZH2 augments leukemogenicity by reinforcing differentiation blockage in acute myeloid leukemia. Blood 120(5), 1107–1117 (2012).
    • 28. Ernst T, Chase AJ, Score J et al. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat. Genet. 42(8), 722–726 (2010).
    • 29. Nikoloski G, Langemeijer SM, Kuiper RP et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat. Genet. 42(8), 665–667 (2010).
    • 30. Ntziachristos P, Tsirigos A, Van Vlierberghe P et al. Genetic inactivation of the polycomb repressive complex 2 in T cell acute lymphoblastic leukemia. Nat. Med. 18(2), 298–301 (2012).
    • 31. Simon C, Chagraoui J, Krosl J et al. A key role for EZH2 and associated genes in mouse and human adult T-cell acute leukemia. Genes Dev. 26(7), 651–656 (2012).
    • 32. Varambally S, Dhanasekaran SM, Zhou M et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature 419(6907), 624–629 (2002).
    • 33. Bracken AP, Pasini D, Capra M, Prosperini E, Colli E, Helin K. EZH2 is downstream of the pRB–E2F pathway, essential for proliferation and amplified in cancer. EMBO J. 22(20), 5323–5335 (2003).
    • 34. Bachmann IM, Halvorsen OJ, Collett K et al. EZH2 expression is associated with high proliferation rate and aggressive tumor subgroups in cutaneous melanoma and cancers of the endometrium, prostate and breast. J. Clin. Oncol. 24(2), 268–273 (2006).
    • 35. Sauvageau M, Sauvageau G. Polycomb group proteins: multi-faceted regulators of somatic stem cells and cancer. Cell Stem Cell 7(3), 299–313 (2010).
    • 36. Comet I, Riising EM, Leblanc B, Helin K. Maintaining cell identity: PRC2-mediated regulation of transcription and cancer. Nat. Rev. Cancer 16(12), 803–810 (2016). •• Covers the molecular mechanisms linking EZH2-mediated H3K27me3 to gene expression changes in cancer.
    • 37. Yamagishi M, Uchimaru K. Targeting EZH2 in cancer therapy. Curr. Opin. Oncol. 29(5), 375–381 (2017).
    • 38. Jung HY, Jun S, Lee M et al. PAF and EZH2 induce Wnt/beta-catenin signaling hyperactivation. Mol. Cell 52(2), 193–205 (2013).
    • 39. Gonzalez ME, Moore HM, Li X et al. EZH2 expands breast stem cells through activation of NOTCH1 signaling. Proc. Natl Acad. Sci. USA 111(8), 3098–3103 (2014).
    • 40. Kim J, Lee Y, Lu X et al. Polycomb- and methylation-independent roles of EZH2 as a transcription activator. Cell Rep. 25(10), 2808–2820 (2018).
    • 41. Xiao L, Tien JC, Vo J et al. Epigenetic reprogramming with antisense oligonucleotides enhances the effectiveness of androgen receptor inhibition in castration-resistant prostate cancer. Cancer Res. 78(20), 5731–5740 (2018).
    • 42. van Haaften G, Dalgliesh GL, Davies H et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat. Genet. 41(5), 521–523 (2009).
    • 43. Wilson BG, Wang X, Shen X et al. Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation. Cancer Cell 18(4), 316–328 (2010).
    • 44. Jankowska AM, Makishima H, Tiu RV et al. Mutational spectrum analysis of chronic myelomonocytic leukemia includes genes associated with epigenetic regulation: UTX, EZH2 and DNMT3A. Blood 118(14), 3932–3941 (2011).
    • 45. Kadoch C, Hargreaves DC, Hodges C et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat. Genet. 45(6), 592–601 (2013).
    • 46. Scott-Browne JP, Lopez-Moyado IF, Trifari S et al. Dynamic changes in chromatin accessibility occur in CD8+ T cells responding to viral infection. Immunity 45(6), 1327–1340 (2016).
    • 47. Sen DR, Kaminski J, Barnitz RA et al. The epigenetic landscape of T cell exhaustion. Science 354(6316), 1165–1169 (2016).
    • 48. Philip M, Fairchild L, Sun L et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 545(7655), 452–456 (2017).
    • 49. Corces MR, Granja JM, Shams S et al. The chromatin accessibility landscape of primary human cancers. Science 362(6413), 2018).
    • 50. Peng D, Kryczek I, Nagarsheth N et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 527(7577), 249–253 (2015).
    • 51. Gao J, Shi LZ, Zhao H et al. Loss of IFN-gamma pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 167(2), 397–404; e399 (2016).
    • 52. Mcgranahan N, Rosenthal R, Hiley CT et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 171(6), 1259–1271.e1211 (2017).
    • 53. Sade-Feldman M, Jiao YJ, Chen JH et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat. Commun. 8(1), 1136 (2017).
    • 54. Pauken KE, Sammons MA, Odorizzi PM et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 354(6316), 1160–1165 (2016).
    • 55. Ghoneim HE, Fan Y, Moustaki A et al. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell 170(1), 142–157; e119 (2017).
    • 56. Gunawan M, Venkatesan N, Loh JT et al. The methyltransferase EZH2 controls cell adhesion and migration through direct methylation of the extranuclear regulatory protein talin. Nat. Immunol. 16(5), 505–516 (2015).
    • 57. Tian Y, Meng L, Zhang Y. Epigenetic regulation of dendritic cell development and function. Cancer J. 23(5), 302–307 (2017).
    • 58. Bugide S, Janostiak R, Wajapeyee N. Epigenetic mechanisms dictating eradication of cancer by natural killer cells. Trends Cancer 4(8), 553–566 (2018).
    • 59. Zhang C, Wang S, Liu Y, Yang C. Epigenetics in myeloid derived suppressor cells: a sheathed sword towards cancer. Oncotarget 7(35), 57452–57463 (2016).
    • 60. Guerriero JL, Sotayo A, Ponichtera HE et al. Class IIa HDAC inhibition reduces breast tumours and metastases through anti-tumour macrophages. Nature 543(7645), 428–432 (2017).
    • 61. Kwon HK, Chen HM, Mathis D, Benoist C. Different molecular complexes that mediate transcriptional induction and repression by FoxP3. Nat. Immunol. 18(11), 1238–1248 (2017).
    • 62. Rodriguez-Ubreva J, Catala-Moll F, Obermajer N et al. Prostaglandin E2 leads to the acquisition of DNMT3A-dependent tolerogenic functions in human myeloid-derived suppressor cells. Cell Rep. 21(1), 154–167 (2017).
    • 63. de Groot AE, Pienta KJ. Epigenetic control of macrophage polarization: implications for targeting tumor-associated macrophages. Oncotarget 9(29), 20908–20927 (2018).
    • 64. Ennishi D, Takata K, Beguelin W et al. Molecular and genetic characterization of MHC deficiency identifies EZH2 as therapeutic target for enhancing immune recognition. Cancer Discov. 9(4), 546–563 (2019).
    • 65. Poli V, Fagnocchi L, Zippo A. Tumorigenic cell reprogramming and cancer plasticity: interplay between signaling, microenvironment and epigenetics. Stem Cells Int. 2018, 4598195 (2018).
    • 66. Ziani L, Chouaib S, Thiery J. Alteration of the antitumor immune response by cancer-associated fibroblasts. Front. Immunol. 9, 414 (2018).
    • 67. Serrano A, Tanzarella S, Lionello I et al. Rexpression of HLA class I antigens and restoration of antigen-specific CTL response in melanoma cells following 5-aza-2′-deoxycytidine treatment. Int. J. Cancer 94(2), 243–251 (2001).
    • 68. Garcia-Lora A, Martinez M, Algarra I, Gaforio JJ, Garrido F. MHC class I-deficient metastatic tumor variants immunoselected by T lymphocytes originate from the coordinated downregulation of APM components. Int. J. Cancer 106(4), 521–527 (2003).
    • 69. Pech MF, Fong LE, Villalta JE et al. Systematic identification of cancer cell vulnerabilities to natural killer cell-mediated immune surveillance. Elife 8, e47362 (2019).
    • 70. Burr ML, Sparbier CE, Chan KL et al. An evolutionarily conserved function of polycomb silences the MHC class I antigen presentation pathway and enables immune evasion in cancer. Cancer Cell 36(4), 385–401; e388 (2019). •• Presents evidence for evolutionary selection of EZH2-mediated suppression of MHC class I expression in certain cancers and the therapuetic potential for EZH2 inhibitors to overcome this repression.
    • 71. Wee ZN, Li Z, Lee PL, Lee ST, Lim YP, Yu Q. EZH2-mediated inactivation of IFN-gamma-JAK–STAT1 signaling is an effective therapeutic target in MYC-driven prostate cancer. Cell Rep. 8(1), 204–216 (2014).
    • 72. Nagarsheth N, Peng D, Kryczek I et al. PRC2 epigenetically silences Th1-type chemokines to suppress effector T-cell trafficking in colon cancer. Cancer Res. 76(2), 275–282 (2016).
    • 73. Zingg D, Arenas-Ramirez N, Sahin D et al. The histone methyltransferase EZH2 controls mechanisms of adaptive resistance to tumor immunotherapy. Cell Rep. 20(4), 854–867 (2017).
    • 74. Abou El Hassan M, Huang K, Eswara MB et al. Cancer cells hijack PRC2 to modify multiple cytokine pathways. PLoS ONE 10(6), e0126466 (2015).
    • 75. Houghton AN, Guevara-Patino JA. Immune recognition of self in immunity against cancer. J. Clin. Invest. 114(4), 468–471 (2004).
    • 76. Scanlan MJ, Gure AO, Jungbluth AA, Old LJ, Chen YT. Cancer/testis antigens: an expanding family of targets for cancer immunotherapy. Immunol. Rev. 188, 22–32 (2002).
    • 77. James SR, Link PA, Karpf AR. Epigenetic regulation of X-linked cancer/germline antigen genes by DNMT1 and DNMT3b. Oncogene 25(52), 6975–6985 (2006).
    • 78. Heninger E, Krueger TE, Lang JM. Augmenting antitumor immune responses with epigenetic modifying agents. Front. Immunol. 6, 29 (2015).
    • 79. Canadas I, Thummalapalli R, Kim JW et al. Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses. Nat. Med. 24(8), 1143–1150 (2018).
    • 80. Shin DS, Zaretsky JM, Escuin-Ordinas H et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. 7(2), 188–201 (2017).
    • 81. Kamminga LM, Bystrykh LV, De Boer A et al. The polycomb group gene EZH2 prevents hematopoietic stem cell exhaustion. Blood 107(5), 2170–2179 (2006).
    • 82. Chou RH, Yu YL, Hung MC. The roles of EZH2 in cell lineage commitment. Am. J. Transl. Res. 3(3), 243–250 (2011).
    • 83. Mochizuki-Kashio M, Mishima Y, Miyagi S et al. Dependency on the polycomb gene EZH2 distinguishes fetal from adult hematopoietic stem cells. Blood 118(25), 6553–6561 (2011).
    • 84. Marrack P, Kappler J. Control of T cell viability. Annu. Rev. Immunol. 22, 765–787 (2004).
    • 85. He S, Wang J, Kato K et al. Inhibition of histone methylation arrests ongoing graft-versus-host disease in mice by selectively inducing apoptosis of alloreactive effector T cells. Blood 119(5), 1274–1282 (2012).
    • 86. Zhang Y, Kinkel S, Maksimovic J et al. The polycomb repressive complex 2 governs life and death of peripheral T cells. Blood 124(5), 737–749 (2014).
    • 87. Zhao E, Maj T, Kryczek I et al. Cancer mediates effector T cell dysfunction by targeting microRNAs and EZH2 via glycolysis restriction. Nat. Immunol. 17(1), 95–103 (2016).
    • 88. Long H, Xiang T, Luo J et al. The tumor microenvironment disarms CD8+ T lymphocyte function via a miR-26a-EZH2 axis. Oncoimmunology 5(12), e1245267 (2016).
    • 89. Kakaradov B, Arsenio J, Widjaja CE et al. Early transcriptional and epigenetic regulation of CD8+ T cell differentiation revealed by single-cell RNA sequencing. Nat. Immunol. 18(4), 422–432 (2017). • Provides genetic evidence for the function of EZH2 in T-cell development and function specification.
    • 90. Gray SM, Amezquita RA, Guan T, Kleinstein SH, Kaech SM. Polycomb repressive complex 2-mediated chromatin repression guides effector CD8+ T cell terminal differentiation and loss of multipotency. Immunity 46(4), 596–608 (2017).
    • 91. He S, Liu Y, Meng L et al. EZH2 phosphorylation state determines its capacity to maintain CD8+ T memory precursors for antitumor immunity. Nat. Commun. 8(1), 2125 (2017).
    • 92. Goswami S, Apostolou I, Zhang J et al. Modulation of EZH2 expression in T cells improves efficacy of anti-CTLA-4 therapy. J. Clin. Invest. 128(9), 3813–3818 (2018).
    • 93. Tumes DJ, Onodera A, Suzuki A et al. The polycomb protein EZH2 regulates differentiation and plasticity of CD4+ T helper Type 1 and Type 2 cells. Immunity 39(5), 819–832 (2013).
    • 94. Yang XP, Jiang K, Hirahara K et al. EZH2 is crucial for both differentiation of regulatory T cells and T effector cell expansion. Sci. Rep. 5, 10643 (2015).
    • 95. He S, Xie F, Liu Y et al. The histone methyltransferase EZH2 is a crucial epigenetic regulator of allogeneic T-cell responses mediating graft-versus-host disease. Blood 122(25), 4119–4128 (2013).
    • 96. Tong Q, He S, Xie F et al. EZH2 regulates transcriptional and posttranslational expression of T-bet and promotes Th1 cell responses mediating aplastic anemia in mice. J. Immunol. 192(11), 5012–5022 (2014).
    • 97. Chiang SC, Theorell J, Entesarian M et al. Comparison of primary human cytotoxic T-cell and natural killer cell responses reveal similar molecular requirements for lytic granule exocytosis but differences in cytokine production. Blood 121(8), 1345–1356 (2013).
    • 98. Vivier E, Ugolini S, Blaise D, Chabannon C, Brossay L. Targeting natural killer cells and natural killer T cells in cancer. Nat. Rev. Immunol. 12(4), 239–252 (2012). • Demonstrates the lineage-specific ablation of EZH2 in FOXP3+ Treg cells is sufficient to reprogram suppressive function.
    • 99. Nagel S, Venturini L, Marquez VE et al. Polycomb repressor complex 2 regulates HOXA9 and HOXA10, activating ID2 in NK/T-cell lines. Mol. Cancer 9, 151 (2010).
    • 100. Yin J, Leavenworth JW, Li Y et al. EZH2 regulates differentiation and function of natural killer cells through histone methyltransferase activity. Proc. Natl Acad. Sci. USA 112(52), 15988–15993 (2015).
    • 101. Bugide S, Green MR, Wajapeyee N. Inhibition of enhancer of zeste homolog 2 (EZH2) induces natural killer cell-mediated eradication of hepatocellular carcinoma cells. Proc. Natl Acad. Sci. USA 115(15), E3509–E3518 (2018).
    • 102. Ramakrishnan S, Granger V, Rak M et al. Inhibition of EZH2 induces NK cell-mediated differentiation and death in muscle-invasive bladder cancer. Cell Death Differ. 26,2100–2114 (2019)
    • 103. Arvey A, van der Veeken J, Samstein RM, Feng Y, Stamatoyannopoulos JA, Rudensky AY. Inflammation-induced repression of chromatin bound by the transcription factor Foxp3 in regulatory T cells. Nat. Immunol. 15(6), 580–587 (2014).
    • 104. Dupage M, Chopra G, Quiros J et al. The chromatin-modifying enzyme EZH2 is critical for the maintenance of regulatory T cell identity after activation. Immunity 42(2), 227–238 (2015).
    • 105. Wang D, Quiros J, Mahuron K et al. Targeting EZH2 reprograms intratumoral regulatory T Cells to enhance cancer immunity. Cell Rep. 23(11), 3262–3274 (2018).
    • 106. Umansky V, Blattner C, Gebhardt C, Utikal J. The role of myeloid-derived suppressor cells (MDSC) in cancer progression. Vaccines (Basel) 4(4), 36 (2016).
    • 107. Huang S, Wang Z, Zhou J et al. EZH2 inhibitor GSK126 suppresses antitumor immunity by driving production of myeloid-derived suppressor cells. Cancer Res. 79(8), 2009–2020 (2019).
    • 108. Yamagishi M, Hori M, Fujikawa D et al. Targeting excessive EZH1 and EZH2 activities for abnormal histone methylation and transcription network in malignant lymphomas. Cell Rep. 29(8), 2321–2337 (2019).
    • 109. Qi W, Zhao K, Gu J et al. An allosteric PRC2 inhibitor targeting the H3K27me3 binding pocket of EED. Nat. Chem. Biol. 13(4), 381–388 (2017).
    • 110. Pagel J, Morschhauser F, Herve I et al. Interim update from a Phase II multicenter study of tazemetostat, an EZH2 Inhibitor, in patients with relapsed or refractory (R/R) follicular lymphoma (FL). Clin. Lymphoma Myeloma Leuk. 18, S278–S279 (2018).
    • 111. Gulati N, Beguelin W, Giulino-Roth L. Enhancer of zeste homolog 2 (EZH2) inhibitors. Leuk. Lymphoma 59(7), 1574–1585 (2018).
    • 112. Rothbart SB, Baylin SB. Epigenetic therapy for epithelioid sarcoma. Cell 181(2), 211 (2020).
    • 113. Morschhauser F, Tilly H, Chaidos A et al. Interim update from a phase 2 multicenter study of tazemetostat, an EZH2 inhibitor, in patients with relapsed or refractory follicular lymphoma. Hematol. Oncol. 37(52), 154–156 (2019).
    • 114. Gounder MM, Zhu G, Roshal L et al. Immunologic correlates of the abscopal effect in a SMARCB1/INI1-negative poorly differentiated chordoma after EZH2 inhibition and radiotherapy. Clin. Cancer Res. 25(7), 2064–2071 (2019).
    • 115. Apostol AC, Beaudin AE. Reversing time: EZH1 deficiency hastens definitive hematopoiesis. Cell Stem Cell 22(3), 285–287 (2018).