Skip to main content
Top

08-12-2015 | Treatment | Article

Improving cancer immunotherapy with DNA methyltransferase inhibitors

Journal: Cancer Immunology, Immunotherapy

Authors: Mohammad H. Saleh, Lei Wang, Michael S. Goldberg

Publisher: Springer Berlin Heidelberg

Abstract

Immunotherapy confers durable clinical benefit to melanoma, lung, and kidney cancer patients. Challengingly, most other solid tumors, including ovarian carcinoma, are not particularly responsive to immunotherapy, so combination with a complementary therapy may be beneficial. Recent findings suggest that epigenetic modifying drugs can prime antitumor immunity by increasing expression of tumor-associated antigens, chemokines, and activating ligands by cancer cells as well as cytokines by immune cells. This review, drawing from both preclinical and clinical data, describes some of the mechanisms of action that enable DNA methyltransferase inhibitors to facilitate the establishment of antitumor immunity.

Introduction

The accumulation of genomic mutations in oncogenes and tumor suppressor genes has long been regarded as the core driver of tumor progression [1], but research over the past decade has highlighted the growing importance of epigenetic mechanisms in neoplastic disease [2]. The aberrant orchestration of epigenetic machinery can give rise to cancer, rendering the understanding and, as required, inhibition of these mechanisms very important.
Global hypomethylation of genomic DNA increases during carcinogenesis, contributing to chromosomal instability, reactivation of transposable elements, and loss of imprinting [3, 4]. In contrast, the focal hypermethylation of promoter CpG islands and accumulation of histone modifications silences many tumor suppressor genes [3].
This emerging role for epigenetics in cancer has inspired investigation of the therapeutic potential of drugs that selectively target and reverse abnormalities that arise from epigenetic modifications [4]. To date, there are at least four known types of DNA modifications and 16 kinds of histone modifications, including methylation, acetylation, phosphorylation, and ubiquitylation [5]. Of the five epigenetic modifiers currently approved by the US Food and Drug Administration, two inhibit DNA methyltransferases (DNMTs: azacitidine and decitabine) and three inhibit histone deacetylases (HDACs: vorinostat, romidepsin, and belinostat). Although there is considerable evidence for the anticancer effects of HDAC inhibitors [6, 7], recent studies suggest that the majority of HDAC substrates are non-histone, non-epigenetic proteins [8, 9]. This review will thus focus on the epigenetic effects of DNMT inhibition by azacitidine (AZA) or decitabine (DAC).
AZA and DAC are prodrug cytosine analogs—a ribonucleoside and a deoxyribonucleoside, respectively—that are incorporated into nucleic acids as azacytosine-guanine pairs following their uptake and phosphorylation [10]. DNMTs, which naturally bind CG pairs, become trapped in a covalent bond to DNA when they attempt to methylate the azacytosine. Such adducts result either in cell death at high analog doses or in a DNA damage response at low analog doses that degrades DNMTs and subsequently reactivates genes that were previously silenced by hypermethylation [10, 11].
While much interest has focused on the reactivation of tumor suppressor genes by DNMT inhibitors (DNMTi), recent evidence suggests that the inhibition of DNMTs produces cancer cell-extrinsic immunomodulatory effects. This has prompted several groups to study the effects of DNMTi in combination with cancer immunotherapy. Herein, we summarize the mechanisms of DNMTi on antitumor immunity and its synergy with immunotherapy in preclinical and clinical settings.

DNMTi boost tumor immunogenicity

Immune evasion is an emerging hallmark of tumor formation and progression [1]. One of the most effective evasion tactics taken by cancer cells is the impairment of antigen presentation. Specifically, levels of the class I major histocompatibility complex (MHC I) are downregulated by irreversible gene-disabling mutations or by reversible inactivating methylation patterns [12]. Addressing the latter using DNMTi has yielded promising results in a variety of cancer types. In addition to upregulating MHC I, DNMTi can increase tumor immunogenicity by increasing levels of antigens displayed in MHC I, particularly cancer-testis antigens (CTAs) [13, 14]. CTAs are a family of tumor-associated antigens expressed on tumors, but not normal tissues, aside from the testes and placenta. CTA expression is regulated primarily by DNA methylation, so DNMTi treatment increases CTA expression on tumor cells, enabling host cytotoxic T lymphocytes (CTLs) to discriminate them from healthy cells [15].
Both DAC and AZA are currently approved for the treatment of patients with myelodysplastic syndromes (MDS). The impact of these DNMTi may extend beyond their cytotoxicity, as they influence antigen presentation in hematologic cells as well. DAC treatment was shown to increase expression of MHC I and II molecules on a chronic lymphocytic leukemia (CLL) cell line and, to a lesser extent, on a primary CLL culture [16]. It also induced the de novo gene expression of multiple CTAs, including one of the most immunogenic CTAs, NY-ESO-1, and upregulated baseline levels of others, such as multiple members of the melanoma-associated antigen (MAGE) family [16]. Similar results on CTA expression were observed following in vitro treatment of acute myeloid leukemia (AML) cells with DAC, most notably inducing de novo expression of the CTA SSX2 in a majority of cell lines tested [17]. These data are supported by translational studies, as clinical administration of AZA induced SSX2 in all eight AML patients examined [17]. Similarly, the addition of AZA to an HDAC inhibitor for treatment of Hodgkin lymphoma patients resulted in a broader antitumor T cell repertoire, suggesting an increase in antigen presentation [18].
Importantly, there is ample evidence for increased immunogenicity by DMNTi therapy beyond MDS, most notably in melanoma and ovarian cancer, but also in several other cancer types (Table 1). In vitro treatment of a melanoma cell line with DAC upregulated the surface expression of MHC I, increased IFN-γ release by tumor-specific CTLs, and enhanced target cell lysis [19]. Similar results were observed with ovarian cancer cell lines and xenograft melanoma models [20, 21]. Immunizing mice with DAC-treated melanoma cells induced a significant anti-NY-ESO-1 humoral response over multiple challenges, indicating an antitumor memory response [22]. Increased CTA expression was also increased on melanoma patient samples following ex vivo treatment with DAC, notably showing a reduction in intratumor CTA heterogeneity post-treatment [23].
Table 1
Summary of cancer/testis antigens induced or upregulated by DNMT inhibition in different cancer types
Cancer/testis antigen
In vitro
In vivo
Ex vivo
Clinical
BAGE
Sar [33]
   
CT-7
Sar [33]
   
CT-10
Sar [33]
   
FATE-1
CLL [16]
 
CLL [16]
 
GAGE1-6
MM [35]; RCC [30]; Sar [33]
Mel [22]
Mel [23]; MM [35]
 
GAGE-2
CLL [16]
 
CLL [16]
 
GAGE-4
CLL [16]
 
CLL [16]
 
GAGE-7
CLL [16]
 
CLL [16]
 
LAGE-1
CLL [16]; Gli [29]; Sar [33]
   
MAGE-A1
AML [17]; CLL [16]; Sar [28, 30, 49]; MM [35]; NB [34]; Ov [21]; Thy [32]; CC, MC, LC, RCC [30]
Mel [22]
Gli [29]; Mel [23]; Sar [28]
 
MAGE-A2
MM [35]; RCC [30]
Mel [22]
Mel [23]; MM [35];
 
MAGE-A3
CLL [16]; Gli [29]; MM [35]; NB [34]; Ov [21]; Sar [28, 30, 33]; RCC, CC, MC, LC [30]
Mel [22]
Gli [29]; Mel [23] MM [35]; Sar [28]
LC, MM [36]
MAGE-A4
CLL [16]; MM [35]; Ov [21]; RCC [30]; Sar [33]; Thy [32]
Mel [22]
CLL [16]; Gli [29]; Mel [23]; MM [35]
 
MAGE-A6
Ov [21]
 
Gli [29]; Mel [23]
 
MAGE-A8
  
CLL [16]
 
MAGE-A10
Ov [21]; Sar [33]
Mel [22]
  
MAGE-A12
Ov [21]
   
MAGE-B2
CLL [16]
   
MAGE-C1
AML [17]
   
MAGE-C2
AML [17]
   
NY-ESO-1
AML [17]; CLL [16]; Gli [29]; MM [35]; NB [34]; Ov [21]; Sar [28, 30, 33]; Thy [25]; CC, MC, LC, Mel, RCC [30]
Gli [29]; Mel [22]; Thy [25]
Gli [29]; Mel [23]; MM [35]; Sar [28]
LC, MM [36]
NY-SAR-35
CLL [16]
   
NXF2
AML [17]; CLL [16]
 
CLL [16]
 
P1A*
CC, Hep, LC, Lym, MC, Mel, NB, Sar [31]
CC, LLC, MC [26]
  
PAGE-1
  
CLL [16]
 
PRAME
  
Mel [23]
AML [17]
RAGE-1
RCC, Sar [30]
   
SPANX-B
AML [17]
   
SSX1-5
  
Mel [23]
 
SSX1
CLL [16]; Gli [29]
 
CLL [16]; Gli [29]
 
SSX2
AML [17]; CLL [16]; MM [35]
 
CLL [16]; Gli [29]; MM [35]
AML [17]
SSX4
AML [17]; CLL [16]; Gli [29]
 
Gli [29]
 
TAG-1
Ov [21]
   
TAG-2a
Ov [21]
   
TAG-2b
Ov [21]
   
TAG-2C
Ov [21]
   
TPX-1
CLL [16]
   
XAGE-1
CLL [16]
 
CLL [16]
 
Columns indicate treatment conditions, with ex vivo referring to the treatment of primary culture samples
AML acute myeloid leukemia, CC colon carcinoma, CLL chronic lymphoid leukemia, Gli glioma, Hep hepatoma, LC lung carcinoma, LLC lewis lung carcinoma, Lym lymphoma, MC mammary carcinoma, Mel melanoma, MM mesothelioma, NB neuroblastoma, Ov ovarian carcinoma, RCC renal cell carcinoma, Sar sarcoma, Thy thyroid cancer
P1A is the murine analog of MAGE/GAGE/BAGE genes
Given the preclinical data illustrating the effects of DAC on NY-ESO-1 levels, a phase I dose-escalation trial was initiated to evaluate the ability of DAC to augment vaccination against NY-ESO-1. Sixty percent of evaluable ovarian cancer patients who received the combination of DAC, NY-ESO-1 vaccine, and doxorubicin achieved disease stabilization or partial clinical response [24]. The therapy was associated with manageable toxicities, increased NY-ESO-1 serum antibodies, and enhanced antigen-specific T cell responses.
MHC I upregulation by DNMTi has been demonstrated in mammary, lung, colon, and thyroid histotypes [25, 26] as well as human papilloma virus (HPV)-associated carcinomas [27], sarcomas [28], and gliomas [29]. DNMTi-mediated increases of levels of various CTAs have also been observed in a majority of these cancers [25, 26, 2833] as well as in neuroblastomas [31, 34], mesotheliomas [35], and renal, esophageal, pleural [36], and liver [31] cancers (summarized in Table 1). Clearly, downregulation of MHC I and tumor-associated antigens is a fundamental mechanism of tumor immune evasion, and DNMTi can uncloak such tumors.

DNMTi stimulate NK cell- and CD8 T cell-mediated cytotoxicity

In addition to rendering cancer cells more recognizable to T cells in antigen-specific manner, epigenetic modifiers can also enhance cytotoxic natural killer (NK) and T cell function. Specifically, epigenetic modifiers can induce expression of chemokines as well as activating ligands on the surface of tumors cells [4, 37]. DAC and valproic acid, an HDAC inhibitor, upregulate MICA, a ligand for natural-killer group 2, member D (NKG2D) expressed by NK cells as well as activated CD8 T cells [4, 38]. DAC similarly upregulates the related ligand MICB through promoter DNA demethylation and DNA damage [39]. MICA and MICB are stress response ligands and are commonly expressed within tumors.
DAC also intrinsically enhances NK cell antitumor cytotoxicity through various mechanisms. DNA methylation regulates the expression of killer immunoglobulin-like receptors (KIRs), receptors that bind MHC I to allow NK cells to recognize abnormal cells that have downregulated MHC I [40]. The epigenetic modulation of KIR is a known mechanism of immune escape in cancer. Restoring proper expression of KIR with immunosensitizing drugs is an attractive approach to improving cancer immunotherapy. Surprisingly, while DAC enhances NK cell responsiveness to activating stimuli by permitting transcription of genes involved in NK cell reactivity, AZA impairs NK cell-mediated cytotoxicity and IFN-γ production [41].
DNMTi-mediated demethylation promotes transcription of genes involved in CTL reactivity, most notably antitumor cytokines. For example, the transcription of IL-2 in non-proliferating T lymphocytes is correlated with the demethylation of a promoter-enhancer region of the IL-2 gene upon activation [42]. This demethylation pattern of the IL-2 locus (as well as that of IFN-γ) was observed early after CD8 T cell antigen exposure and is maintained throughout CD8 memory development in the presence of CD4 help [43]. This suggests that IL-2 levels could be enhanced and maintained via DNMTi-mediated hypomethylation. Indeed, comparing IL-2 production by recent thymic emigrant peripheral murine T cells to their more mature but still naïve counterparts revealed a reduced difference in IL-2 levels upon culturing in the presence of AZA [44].
Similarly, culturing primary mouse CD8 T cells in the presence of AZA increased their expression of IFN-γ up to 25-fold. Their expression of IL-3, a T cell growth and differentiation cytokine as well as a myeloid proliferation enhancer, also increased by up to 14-fold in a dose-dependent manner [45]. The IFN-γ locus is highly methylated at three specific CpG sites in naïve CD8 T cells, while it is unmethylated in effector T cells, permitting the production of large amounts of IFN-γ. In memory T cells, these sites are partially methylated but rapidly demethylate upon stimulation [46], suggesting an activation-dependent demethylation spectrum to control production levels. Indeed, AZA treatment of naïve CD8 T cells increases the number of IFN-γ-producing cells to a quantity that is comparable to the number of IFN-γ-producing memory cells [46].
Consistent with these results, following DAC treatment, we observed a significant increase in the recruitment of and antitumor cytokine production by NK cells and CD8 T cells in the ascites of mice bearing orthotopic synergenic ovarian tumors [20]. Specifically, the percentage of these cells that produced IFN-γ and TNF-α increased markedly [20]. Interestingly, NK cells produced both IFN-γ and TNF-α while, T cells produced either IFN-γ or TNF-α. The enhanced infiltration of NK cells and CD8 T cells into subcutaneous tumors following DAC treatment was also confirmed. The mechanism underlying the propensity of these cytolytic lymphocytes to migrate to the tumor site was evaluated. The ovarian cancer cells were treated with DAC ex vivo, and microarray analysis was performed. Interestingly, the most highly enriched Gene Ontology was that of “immune system process,” and top gene candidates included chemokines and mediators of innate immunity (Fig. 1) [20]. All candidates that were analyzed by qPCR were successfully validated, and Irf7 (innate immune signaling), Ccl5 and Cxcl10 (chemokines that attract NK and T cells), and GitrL (a ligand that activates these leukocytes) were among the top hits.
DNMTi can influence the behavior of helper cells as well. Culturing human CD4 T cells in the presence of AZA revealed that transcription of IL17A, a proinflammatory cytokine, is directly induced by the demethylation of its promoter region [47]. In vitro AZA treatment of CD4 T cells isolated from 68 MDS patients led to increased IL-17 production by these cells [48]. Moreover, the peripheral blood of AML and MDS patients treated with AZA contained significantly increased levels of IL-17A-secreting CD4 T cells [49]. Similarly, DAC treatment of helper T cells demethylated Th1-specific promoters, induced naïve cells toward Th1 polarization, and depolarized Th2 cells, thereby promoting production of IFN-γ [50].
Taken together, these studies point to the potential use of DNMTi for the enhancement of cytotoxic NK and CD8 T cells as well as helper CD4 cells, particularly through the induction of critical immunostimulatory cytokines.

DNMTi decrease suppression by regulatory adaptive and innate immune cells

Clinical data suggest that the reduction of immunosuppressive regulatory immune cells may be as important as stimulating effector cell-mediated antitumor immunity [51, 52]. Notably, in addition to promoting tumor immunogenicity and effector cell function, epigenetic modifiers can decrease natural and tumor-induced immunosuppression. A number of regulatory T cells (Tregs) present in peripheral blood samples from a pool of 68 MDS patients were reduced following treatment with AZA. This result is consistent with the observed reduction in proliferative capacity and suppressive function of Tregs upon AZA treatment of patient CD4 T cells in vitro [48].
Levels of myeloid-derived suppressor cells (MDSCs), another major immunosuppressive population often found in the tumor microenvironment, can also be modulated with DNMTi. In the 4T1 mouse model, a combination regimen of DAC, entinostat (an HDAC inhibitor), anti-CTLA-4, and anti-PD-1 dramatically decreased circulating granulocytic MDSCs and brought their level down to those observed in non-tumor-bearing mice. This treatment also significantly decreased intratumoral MDSCs and Tregs [53]. However, combination with HDACi is not essential for achieving similar results in other tumor models. In a syngeneic murine ovarian cancer model, DAC treatment reduced the number of tumor-associated MDSCs in the peritoneal cavity [20]. Consistently, the frequency of tumor-infiltrating MDSCs was also reduced in the B16 murine melanoma model in response to DAC treatment [54].
Similarly, mice bearing subcutaneous TRAMP-C2 (derived from prostatic adenocarcinoma) or TC-1/A9 (derived from primary lung epithelia) tumors showed a decreased percentage of MDSCs in their tumor microenvironments and spleens upon AZA treatment. A higher dose of AZA was also shown to counter the accumulation of MDSCs induced by cyclophosphamide treatment. To elucidate the mechanism underlying these effects, splenic MDSCs cultured in vitro in the presence of AZA showed a reduction in the percentage of CD11b+/Gr-1+ MDSCs and an increase in the percentages of CD11c+ and CD86+/MHCII+ cells. This suggests that DNMTi target MDSCs, at least partially, by inducing their differentiation toward DCs [55]. This explanation is supported by earlier research showing that the addition of DAC to a GM-CSF-supplemented culture of tumor-infiltrated CD11b+ myeloid cells promoted the generation of pure CD11b+ CD11c+ mature antigen presenting cells that produced lower levels of immunosuppressive and proinflammatory cytokines [56]. Taken together, these studies suggest that the success of DNMTi-based treatments can be partially attributed to their ability to counter lymphocyte- and myeloid-driven immunosuppression within the tumor milieu.

Rationale for combination of DNMTi with immunotherapy

There is also emerging evidence supporting the use of DNMTi to specifically activate a broad variety of other immunomodulatory genes in multiple cancers. Both DAC and AZA induce sustained upregulation of genes related to antigen presentation, inflammation, and immune responses in leukemic and breast cancer cells in vitro [57]. Similarly, the treatment of non-small cell lung cancer (NSCLC) lines with AZA enriches gene expression related to antigen presentation, type I and type II interferon signaling, apoptosis, viral defense, and numerous immune-related transcription factors [58]. Moreover, a gene expression and methylation analysis of 63 cancer cell lines of breast, colorectal, and ovarian origins revealed similar enrichment for immunostimulatory pathways, such as cytokine signaling and inflammation [59]. This ability to widely prime the immune system makes DNMTi a great candidate for combinations with other immunotherapies.
Immune checkpoint blockade is of particularly relevance as a combination to DNMTi. Administering DNMTi alone or in combination with an HDACi to patients previously untreated for MDS or AML resulted in upregulated expression of CTLA-4, PD-1, PD-L1, and/or PD-L2 in a majority of patients, suggesting that antibodies against these targets may compensate for any negative impact that DNMTi may have on antitumor immunity. Of note, responding patients exhibited far lower levels of PD-L1 and PD-L2 expression than those patients who were resistant to therapy, and lower baseline PD-L1 expression correlated significantly with improved overall response rate [60]. The CTLA-4 and PD-1 axes are central in orchestrating multiple delicate inhibitory pathways, and their upregulation can dampen T cell activation and enhance tumor immune evasion if not countered by immune checkpoint blockade therapy. PD-1 and CTLA-4 are also implicated in the induction and expansion of Tregs, with inhibition of CTLA-4 resulting in a reduced MDSC frequency [61]. Additionally, CTLA-4 [62] and PD-1 [63] blockade elevate IFN-γ levels at murine tumor sites, further supporting the value of combining immune checkpoint blockade with DNMTi.
As mentioned above, survival of mice bearing 4T1 tumors was significantly enhanced when they were treated with the combination of epigenetic modification and checkpoint blockade relative to epigenetic therapy alone [53]. The addition of anti-PD-1 and anti-CTLA-4 also leads to a reduction in levels of circulating granulocytic MDSC [53]. These results are supported by our recent study on the synergistic antitumor effect of DAC with anti-CTLA-4 [20]. While neither therapy produced durable responses as a monotherapy in the orthotopic syngeneic murine model of ovarian carcinoma that we evaluated, the efficacy of anti-CTLA-4 was potentiated by combination with DAC, drastically extending the survival of mice (Fig. 2). Mechanistically, this synergy was explained by the ability of DAC to increase the expression of chemokines that recruit NK cells and CD8 T cells and to promote the production of IFN-γ and TNF-α by these cells. Furthermore, it was observed that this combination promotes differentiation of naive T cells into effector T cells and prolongs cytotoxic T lymphocyte responses. These data suggest that epigenetic modifiers can induce not only cancer cell-intrinsic effects but also cancer cell-extrinsic effects that alter the localization and function of leukocytes in the tumor microenvironment.

Recent and current clinical trials

The aforementioned evidence has led to multiple clinical trials employing DNMTi in combination with a variety of immune-based therapies. One study (NCT01799083) used low-dose DAC followed by an infusion of cytokine-induced killer cells to treat solid tumors and observed an 80 % objective response rate with minimal adverse effects, compared to a 50 and 72 % rate for DAC alone or DAC combined with chemotherapy, respectively [64]. Another recently completed phase I study (NCT01241162) combined DAC with a vaccine cocktail of CTA peptides (NY-ESO-1, MAGE-A1, and MAGE-A3) to treat pediatric neuroblastomas and sarcomas. This combination was well tolerated, with 60 % (6/10) of evaluable patients developing a response to the CTA vaccines and 20 % (2/10) remaining disease-free at least 2 years after their last vaccination [65]. This regimen will be repeated in an ongoing trial (NCT02332889, currently recruiting) to treat gliomas, medulloblastomas, and central nervous system primitive neuroectodermal tumors. Another currently recruiting study (NCT01690507) aims to treat elderly MDS and AML patients using DAC combined with a modified CAG regimen (a cocktail of cytarabine, aclacinomycin, and the G-CSF cytokine) followed by an infusion of HLA haploidentical T cells.
Of particular interest is a study (NCT01928576) seeking to combine DNMTi and checkpoint blockade to treat NSCLC patients. Given the low PD-L1 expression in primary NSCLC samples and the fact that AZA upregulates the transcription and protein expression of PD-L1 in vitro [58], patients are to be treated by AZA (with or without entinostat) followed by the anti-PD-1 drug nivolumab. Given the encouraging preclinical results reported by us and others, the outlook for more clinical trials combining immune checkpoint blockade and epigenetic modification is promising.

Conclusion

DNMTi have been in clinical development for years, but they have conferred limited utility as cytotoxic agents, particularly against solid tumors. Recent studies suggest that these molecules may be utilized optimally at lower doses as priming agents to enhance the efficacy of immunotherapy. Preclinical data indicate that DNMTi are useful across diverse tumor types, and we hope that translational data will confirm that these compounds can be applied broadly in the clinical setting to break immune tolerance and promote antitumor immunity, particularly in the context of combination with cancer immunotherapy.

Acknowledgments

We thank the Ovarian Cancer Research Fund (Liz Tilberis Scholar award) and the Susan F. Smith Center for Women’s Cancer for supporting this work.

Compliance with ethical standards

Conflict of interest

The authors declare no conflict of interest.
AML
Acute myeloid leukemia
AZA
Azacitidine
CLL
Chronic lymphocytic leukemia
CTA
Cancer testis antigen
CTL
Cytotoxic T lymphocyte
DAC
Decitabine
DNMT
DNA methyltransferases
DNMTi
DNA methyltransferase inhibitor
HDAC
Histone deacetylase
MAGE
Melanoma-associated antigen
MDS
Myelodysplastic syndrome
MDSC
Myeloid-derived suppressor cell
MHC I
Major histocompatibility complex class I
NK cells
Natural killer cells
NKG2D
Natural-killer group 2, member D
NSCLC
Non-small cell lung cancer
KIR
Killer immunoglobulin-like receptor
Tregs
Regulatory T cells
Literature
1.
Hanahan D, Weinberg R (2011) Hallmarks of cancer: the next generation. Cell 144:646–674. doi:10.​1016/​j.​cell.​2011.​02.​013 CrossRefPubMed
2.
Baylin S, Jones P (2011) A decade of exploring the cancer epigenome: biological and translational implications. Nat Rev Cancer 11:726–734. doi:10.​1038/​nrc3130 CrossRefPubMedPubMedCentral
3.
Esteller M (2008) Epigenetics in cancer. N Engl J Med 358:1148–1159. doi:10.​1056/​nejmra072067 CrossRefPubMed
4.
Rodríguez-Paredes M, Esteller M (2011) Cancer epigenetics reaches mainstream oncology. Nat Med. doi:10.​1038/​nm.​2305 PubMed
5.
Dawson M, Kouzarides T (2012) Cancer epigenetics: from mechanism to therapy. Cell 150:12–27. doi:10.​1016/​j.​cell.​2012.​06.​013 CrossRefPubMed
6.
Minucci S, Pelicci P (2006) Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 6:38–51. doi:10.​1038/​nrc1779 CrossRefPubMed
7.
West A, Johnstone R (2014) New and emerging HDAC inhibitors for cancer treatment. J Clin Invest 124:30–39. doi:10.​1172/​jci69738 CrossRefPubMedPubMedCentral
8.
Xu W, Parmigiani R, Marks P (2007) Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene 26:5541–5552. doi:10.​1038/​sj.​onc.​1210620 CrossRefPubMed
9.
Choudhary C, Kumar C, Gnad F, Nielsen M, Rehman M, Walther T, Olsen J, Mann M (2009) Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science 325:834–840. doi:10.​1126/​science.​1175371 CrossRefPubMed
10.
Stresemann C, Lyko F (2008) Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer 123:8–13. doi:10.​1002/​ijc.​23607 CrossRefPubMed
11.
Derissen E, Beijnen J, Schellens J (2013) Concise drug review: azacitidine and decitabine. Oncologist 18:619–624. doi:10.​1634/​theoncologist.​2012-0465 CrossRefPubMedPubMedCentral
12.
Bubeník J (2004) MHC class I down-regulation: tumour escape from immune surveillance? (Review). Int J Oncol 25:487–491. doi:10.​3892/​ijo.​25.​2.​487 PubMed
13.
Tomasi T, Magner W, Khan A (2006) Epigenetic regulation of immune escape genes in cancer. Cancer Immunol Immun 55:1159–1184. doi:10.​1007/​s00262-006-0164-4 CrossRef
14.
Maio M, Coral S, Fratta E, Altomonte M, Sigalotti L (2003) Epigenetic targets for immune intervention in human malignancies. Oncogene 22:6484–6488. doi:10.​1038/​sj.​onc.​1206956 CrossRefPubMed
15.
Fratta E, Coral S, Covre A, Parisi G, Colizzi F, Danielli R, Marie Nicolay H, Sigalotti L, Maio M (2011) The biology of cancer testis antigens: putative function, regulation and therapeutic potential. Mol Oncol 5:164–182. doi:10.​1016/​j.​molonc.​2011.​02.​001 CrossRefPubMed
16.
Dubovsky J, McNeel D, Powers J, Gordon J, Sotomayor E, Pinilla-Ibarz J (2009) Treatment of chronic lymphocytic leukemia with a hypomethylating agent induces expression of NXF2, an immunogenic cancer testis antigen. Clin Cancer Res 15:3406–3415. doi:10.​1158/​1078-0432.​ccr-08-2099 CrossRefPubMed
17.
Atanackovic D, Luetkens T, Kloth B, Fuchs G, Cao Y, Hildebrandt Y et al (2011) Cancer-testis antigen expression and its epigenetic modulation in acute myeloid leukemia. Am J Hematol 86:918–922. doi:10.​1002/​ajh.​22141 CrossRefPubMed
18.
Cruz C, Gerdemann U, Leen A, Shafer J, Ku S, Tzou B, Horton T et al (2011) Improving T-cell therapy for relapsed EBV-negative Hodgkin lymphoma by targeting upregulated MAGE-A4. Clin Cancer Res 17:7058–7066. doi:10.​1158/​1078-0432.​ccr-11-1873 CrossRefPubMedPubMedCentral
19.
Fonsatti E, Nicolay H, Sigalotti L, Calabro L, Pezzani L, Colizzi F et al (2007) Functional up-regulation of human leukocyte antigen class I antigens expression by 5-aza-2′-deoxycytidine in cutaneous melanoma: immunotherapeutic implications. Clin Cancer Res 13:3333–3338. doi:10.​1158/​1078-0432.​ccr-06-3091 CrossRefPubMed
20.
Wang L, Amoozgar Z, Huang J, Saleh M, Xing D, Orsulic S, Goldberg M (2015) Decitabine enhances lymphocyte migration and function and synergizes with CTLA-4 blockade in a murine ovarian cancer model. Cancer Immunol Res 3:1030–1041. doi:10.​1158/​2326-6066.​cir-15-0073 CrossRefPubMed
21.
Adair S, Hogan K (2008) Treatment of ovarian cancer cell lines with 5-aza-2′-deoxycytidine upregulates the expression of cancer-testis antigens and class I major histocompatibility complex-encoded molecules. Cancer Immunol Immun 58:589–601. doi:10.​1007/​s00262-008-0582-6 CrossRef
22.
Coral S, Sigalotti L, Colizzi F, Spessotto A, Nardi G, Cortini E et al (2006) Phenotypic and functional changes of human melanoma xenografts induced by DNA hypomethylation: immunotherapeutic implications. J Cell Physiol 207:58–66. doi:10.​1002/​jcp.​20540 CrossRefPubMed
23.
Sigalotti L, Fratta E, Coral S, Tanzarella S, Danielli R, Colizzi F et al (2004) Intratumor heterogeneity of cancer/testis antigens expression in human cutaneous melanoma is methylation-regulated and functionally reverted by 5-Aza-2′-deoxycytidine. Cancer Res 64:9167–9171. doi:10.​1158/​0008-5472.​can-04-1442 CrossRefPubMed
24.
Odunsi K, Matsuzaki J, James S, Mhawech-Fauceglia P, Tsuji T, Miller A et al (2014) Epigenetic potentiation of NY-ESO-1 vaccine therapy in human ovarian cancer. Cancer Immunol Res 2:37–49. doi:10.​1158/​2326-6066.​cir-13-0126 CrossRefPubMedPubMedCentral
25.
Gunda V, Frederick D, Bernasconi M, Wargo J, Parangi S (2014) A potential role for immunotherapy in thyroid cancer by enhancing NY-ESO-1 cancer antigen expression. Thyroid 24:1241–1250. doi:10.​1089/​thy.​2013.​0680 CrossRefPubMedPubMedCentral
26.
Wachowska M, Gabrysiak M, Muchowicz A, Bednarek W, Barankiewicz J, Rygiel T et al (2014) 5-Aza-2′-deoxycytidine potentiates antitumour immune response induced by photodynamic therapy. Eur J Cancer 50:1370–1381. doi:10.​1016/​j.​ejca.​2014.​01.​017 CrossRefPubMedPubMedCentral
27.
Šímová J, Polláková V, Indrová M, Mikyšková R, Bieblová J, Štěpánek I et al (2011) Immunotherapy augments the effect of 5-azacytidine on HPV16-associated tumours with different MHC class I-expression status. Br J Cancer 105:1533–1541. doi:10.​1038/​bjc.​2011.​428 CrossRefPubMedPubMedCentral
28.
Krishnadas D, Bao L, Bai F, Chencheri S, Lucas K (2014) Decitabine facilitates immune recognition of sarcoma cells by upregulating CT antigens, MHC molecules, and ICAM-1. Tumor Biol 35:5753–5762. doi:10.​1007/​s13277-014-1764-9 CrossRef
29.
Natsume A, Wakabayashi T, Tsujimura K, Shimato S, Ito M, Kuzushima K et al (2008) The DNA demethylating agent 5-aza-2′-deoxycytidine activates NY-ESO-1 antigenicity in orthotopic human glioma. Int J Cancer 122:2542–2553. doi:10.​1002/​ijc.​23407 CrossRefPubMed
30.
Coral S, Sigalotti L, Altomonte M, Engelsberg A, Cattarossi I, Maraskovsky E et al (2002) 5-aza-2′-Deoxycytidine-induced expression of functional cancer testis antigens in human renal cell carcinoma immunotherapeutic implications. Clin Cancer Res 8:2690PubMed
31.
Guo Z (2006) De novo induction of a cancer/testis antigen by 5-aza-2′-deoxycytidine augments adoptive immunotherapy in a murine tumor model. Cancer Res 66:1105–1113. doi:10.​1158/​0008-5472.​can-05-3020 CrossRefPubMedPubMedCentral
32.
Gunda V, Cogdill A, Bernasconi M, Wargo J, Parangi S (2013) Potential role of 5-Aza-2′-deoxycytidine induced MAGE-A4 expression in immunotherapy for anaplastic thyroid cancer. Surgery 154:1456–1462. doi:10.​1016/​j.​surg.​2013.​07.​009 CrossRefPubMed
33.
Ayyoub M, Taub R, Keohan M, Hesdorffer M, Metthez G, Memeo L et al (2004) The frequent expression of cancer/testis antigens provides opportunities for immunotherapeutic targeting of sarcoma. Cancer Immun 4:7PubMed
34.
Bao L, Dunham K, Lucas K (2011) MAGE-A1, MAGE-A3, and NY-ESO-1 can be upregulated on neuroblastoma cells to facilitate cytotoxic T lymphocyte-mediated tumor cell killing. Cancer Immunol Immun 60:1299–1307. doi:10.​1007/​s00262-011-1037-z CrossRef
35.
Sigalotti L, Coral S, Altomonte M, Natali L, Gaudino G, Cacciotti P et al (2002) Cancer testis antigens expression in mesothelioma: role of DNA methylation and bioimmunotherapeutic implications. Br J Cancer 86:979–982. doi:10.​1038/​sj.​bjc.​6600174 CrossRefPubMedPubMedCentral
36.
Schrump D, Fischette M, Nguyen D, Zhao M, Li X, Kunst T et al (2006) Phase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleura. Clin Cancer Res 12:5777–5785. doi:10.​1158/​1078-0432.​ccr-06-0669 CrossRefPubMed
37.
Wu X, Tao Y, Hou J, Meng X, Shi J (2012) Valproic acid upregulates NKG2D ligand expression through an ERK-dependent mechanism and potentially enhances NK cell-mediated lysis of myeloma. Neoplasia 14:1178–1189CrossRefPubMedPubMedCentral
38.
Marbach D, Costello J, Küffner R, Vega N, Prill R, Camacho D et al (2012) Wisdom of crowds for robust gene network inference. Nat Methods 9:796–804. doi:10.​1038/​nmeth.​2016 CrossRefPubMedPubMedCentral
39.
Tang K, He C, Zeng G, Wu J, Song G, Shi Y et al (2008) Induction of MHC class I-related chain B (MICB) by 5-aza-2′-deoxycytidine. Biochem Biophys Res Commun 370:578–583. doi:10.​1016/​j.​bbrc.​2008.​03.​131 CrossRefPubMed
40.
Chan H, Kurago Z, Stewart C, Wilson M, Martin M, Mace B et al (2003) DNA methylation maintains allele-specific KIR gene expression in human natural killer cells. J Exp Med 197:245–255. doi:10.​1084/​jem.​20021127 CrossRefPubMedPubMedCentral
41.
Schmiedel B, Arélin V, Gruenebach F, Krusch M, Schmidt S, Salih H (2010) Azacytidine impairs NK cell reactivity while decitabine augments NK cell responsiveness toward stimulation. Int J Cancer 128:2911–2922. doi:10.​1002/​ijc.​25635 CrossRefPubMed
42.
Bruniquel D, Schwartz R (2003) Selective, stable demethylation of the interleukin-2 gene enhances transcription by an active process. Nat Immunol 4:235–240. doi:10.​1038/​ni887 CrossRefPubMed
43.
Northrop J, Thomas R, Wells A, Shen H (2006) Epigenetic remodeling of the IL-2 and IFN-loci in memory CD8 T cells is influenced by CD4 T cells. J Immunol 177:1062–1069. doi:10.​4049/​jimmunol.​177.​2.​1062 CrossRefPubMed
44.
Berkley A, Hendricks D, Simmons K, Fink P (2013) Recent thymic emigrants and mature naive T cells exhibit differential DNA methylation at key cytokine loci. J Immunol 190:6180–6186. doi:10.​4049/​jimmunol.​1300181 CrossRefPubMedPubMedCentral
45.
Fitzpatrick D, Shirley K, McDonald L, Bielefeldt-Ohmann H, Kay G, Kelso A (1998) Distinct methylation of the interferon (IFN-) and interleukin 3 (IL-3) genes in newly activated primary CD8+ T lymphocytes: regional IFN- promoter demethylation and mRNA expression are heritable in CD44highCD8+ T cells. J Exp Med 188:103–117. doi:10.​1084/​jem.​188.​1.​103 CrossRefPubMedPubMedCentral
46.
Kersh E, Fitzpatrick D, Murali-Krishna K, Shires J, Speck S, Boss J, Ahmed R (2006) Rapid demethylation of the IFN-gamma gene occurs in memory but not naive CD8 T cells. J Immunol 176:4083–4093. doi:10.​4049/​jimmunol.​176.​7.​4083 CrossRefPubMed
47.
Janson P, Linton L, Ahlen Bergman E, Marits P, Eberhardson M, Piehl F et al (2010) Profiling of CD4+ T cells with epigenetic immune lineage analysis. J Immunol 186:92–102. doi:10.​4049/​jimmunol.​1000960 CrossRefPubMed
48.
Costantini B, Kordasti S, Kulasekararaj A, Jiang J, Seidl T, Abellan P et al (2012) The effects of 5-azacytidine on the function and number of regulatory T cells and T-effectors in myelodysplastic syndrome. Haematologica 98:1196–1205. doi:10.​3324/​haematol.​2012.​074823 CrossRefPubMed
49.
Frikeche J, Clavert A, Delaunay J, Brissot E, Grégoire M, Gaugler B, Mohty M (2011) Impact of the hypomethylating agent 5-azacytidine on dendritic cells function. Exp Hematol 39:1056–1063. doi:10.​1016/​j.​exphem.​2011.​08.​004 CrossRefPubMed
50.
Dubovsky J, Powers J, Gao Y, Mariusso L, Sotomayor E, Pinilla-Ibarz J (2011) Epigenetic repolarization of T lymphocytes from chronic lymphocytic leukemia patients using 5-aza-2′-deoxycytidine. Leukemia Res 35:1193–1199. doi:10.​1016/​j.​leukres.​2011.​02.​007 CrossRef
51.
Sinha P, Clements V, Bunt S, Albelda S, Ostrand-Rosenberg S (2007) Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a Type 2 response. J Immunol 179:977–983. doi:10.​4049/​jimmunol.​179.​2.​977 CrossRefPubMed
52.
Guislain A, Gadiot J, Kaiser A, Jordanova E, Broeks A, Sanders J et al (2015) Sunitinib pretreatment improves tumor-infiltrating lymphocyte expansion by reduction in intratumoral content of myeloid-derived suppressor cells in human renal cell carcinoma. Cancer Immunol Immun 64:1241–1250. doi:10.​1007/​s00262-015-1735-z CrossRef
53.
Kim K, Skora A, Li Z, Liu Q, Tam A, Blosser R et al (2014) Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci USA 111:11774–11779. doi:10.​1073/​pnas.​1410626111 CrossRefPubMedPubMedCentral
54.
Triozzi P, Aldrich W, Achberger S, Ponnazhagan S, Alcazar O, Saunthararajah Y (2012) Differential effects of low-dose decitabine on immune effector and suppressor responses in melanoma-bearing mice. Cancer Immunol Immun 61:1441–1450. doi:10.​1007/​s00262-012-1204-x CrossRef
55.
Mikysková R, Indrova M, Vlkova V, Bieblova J, Šimova J, Paračková Z et al (2014) DNA demethylating agent 5-azacytidine inhibits myeloid-derived suppressor cells induced by tumor growth and cyclophosphamide treatment. J Leukoc Biol 95:743–753. doi:10.​1189/​jlb.​0813435 CrossRef
56.
Daurkin I, Eruslanov E, Vieweg J, Kusmartsev S (2009) Generation of antigen-presenting cells from tumor-infiltrated CD11b myeloid cells with DNA demethylating agent 5-aza-2′-deoxycytidine. Cancer Immunol Immun 59:697–706. doi:10.​1007/​s00262-009-0786-4 CrossRef
57.
Tsai H, Li H, Van Neste L, Cai Y, Robert C, Rassool F et al (2012) Transient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cells. Cancer Cell 21:430–446. doi:10.​1016/​j.​ccr.​2011.​12.​029 CrossRefPubMedPubMedCentral
58.
Wrangle J, Wang W, Koch A, Easwaran H, Mohammad H, Vendetti F et al (2013) Alterations of immune response of non-small cell lung cancer with azacytidine. Oncotarget 4:2067–2079CrossRefPubMedPubMedCentral
59.
Li H, Chiappinelli K, Guzzetta A, Easwaran H, Yen R, Vatapalli R et al (2014) Immune regulation by low doses of the DNA methyltransferase inhibitor 5-azacitidine in common human epithelial cancers. Oncotarget 5:587–598CrossRefPubMedPubMedCentral
60.
Yang H, Bueso-Ramos C, DiNardo C, Estecio M, Davanlou M, Geng Q et al (2013) Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia 28:1280–1288. doi:10.​1038/​leu.​2013.​355 CrossRefPubMedPubMedCentral
61.
Pico de Coaña Y, Choudhury A, Kiessling R (2015) Checkpoint blockade for cancer therapy: revitalizing a suppressed immune system. Trends Mol Med 21:482–491. doi:10.​1016/​j.​molmed.​2015.​05.​005 CrossRefPubMed
62.
Paradis T, Floyd E, Burkwit J, Cole S, Brunson B, Elliott E, Gilman S, Gladue R (2001) The anti-tumor activity of anti-CTLA-4 is mediated through its induction of IFN gamma. Cancer Immunol Immunother 50:125–133CrossRefPubMed
63.
Peng W, Liu C, Xu C, Lou Y, Chen J, Yang Y et al (2012) PD-1 blockade enhances T-cell migration to tumors by elevating IFN- inducible chemokines. Cancer Res 72:5209–5218. doi:10.​1158/​0008-5472.​can-12-1187 CrossRefPubMedPubMedCentral
64.
Fan H, Lu X, Wang X, Liu Y, Guo B, Zhang Y et al (2014) Low-dose decitabine-based chemoimmunotherapy for patients with refractory advanced solid tumors: a phase I/II report. J Immunol Res 2014:371087. doi:10.​1155/​2014/​371087 PubMedPubMedCentral
65.
Krishnadas D, Shusterman S, Bai F, Diller L, Sullivan J, Cheerva A et al (2015) A phase I trial combining decitabine/dendritic cell vaccine targeting MAGE-A1, MAGE-A3 and NY-ESO-1 for children with relapsed or therapy-refractory neuroblastoma and sarcoma. Cancer Immunol Immun 64:1251–1260. doi:10.​1007/​s00262-015-1731-3 CrossRef