9,208
Views
4
CrossRef citations to date
0
Altmetric
Review

EZH2 Inhibition: A Promising Strategy to Prevent Cancer Immune Editing

ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 1457-1476 | Received 06 May 2020, Accepted 28 Aug 2020, Published online: 17 Sep 2020

References

  • Vaddepally RK , KharelP , PandeyRet al. Review of indications of FDA-approved immune checkpoint inhibitors per NCCN Guidelines with the level of evidence. Cancers (Basel)12(3), 738–756 (2020).
  • O’Donnell JS , TengMWL , SmythMJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol.16(3), 151–167 (2019).
  • Sharma P , Hu-LieskovanS , WargoJAet al. Primary, adaptive and acquired resistance to cancer immunotherapy. Cell168(4), 707–723 (2017).
  • Dunn GP , OldLJ , SchreiberRD. The three Es of cancer immunoediting. Annu. Rev. Immunol.22, 329–360 (2004).
  • Schreiber RD , OldLJ , SmythMJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science331(6024), 1565–1570 (2011).
  • Deans C , MaggertKA. What do you mean, “epigenetic”. Genetics199(4), 887–896 (2015).
  • Burr ML , SparbierCE , ChanKLet al. An evolutionarily conserved function of polycomb silences the MHC class I antigen presentation pathway and enables immune evasion in cancer. Cancer Cell36(4), 385–401.e8 (2019).
  • Toyokawa G , TakadaK , TagawaTet al. A positive correlation between the EZH2 and PD-L1 expression in resected lung adenocarcinomas. Ann. Thorac. Surg.107(2), 393–400 (2019).
  • Wang D , QuirosJ , MahuronKet al. Targeting EZH2 reprograms intratumoral regulatory T cells to enhance cancer immunity. Cell Rep.23(11), 3262–3274 (2018).
  • Xia L , ZhuX , ZhangL , XuY , ChenG , LuoJ. EZH2 enhances expression of CCL5 to promote recruitment of macrophages and invasion in lung cancer. Biotechnol. Appl. Biochem.https://doi.org/10.1002/bab.1875(2019).
  • Burnet FM . The concept of immunological surveillance. Prog. Exp. Tumor Res.13, 1–27 (1970).
  • Thomas L . On immunosurveillance in human cancer. Yale J. Biol. Med.55(3–4), 329–333 (1982).
  • Appay V , DouekDC , PriceDA. CD8+ T cell efficacy in vaccination and disease. Nat. Med.14(6), 623–628 (2008).
  • Malmberg KJ , CarlstenM , BjörklundAet al. Natural killer cell-mediated immunosurveillance of human cancer. Semin. Immunol.31, 20–29 (2017).
  • Iannello A , ThompsonTW , ArdolinoMet al. Immunosurveillance and immunotherapy of tumors by innate immune cells. Curr. Opin. Immunol.38, 52–58 (2016).
  • Hayday AC . γδ T cell update: adaptate orchestrators of immune surveillance. J. Immunol.203(2), 311–320 (2019).
  • Melssen M , SlingluffCL. Vaccines targeting helper T cells for cancer immunotherapy. Curr. Opin. Immunol.47, 85–92 (2017).
  • Pardoll DM , TopalianSL. The role of CD4+ T cell responses in antitumor immunity. Curr. Opin. Immunol.10(5), 588–594 (1998).
  • Pawelec G . Tumour escape: antitumour effectors too much of a good thing. Cancer Immunol. Immunother.53(3), 262–274 (2004).
  • Vinay DS , RyanEP , PawelecGet al. Immune evasion in cancer: mechanistic basis and therapeutic strategies. Semin. Cancer Biol.35(Suppl.), S185–S198 (2015).
  • McGranahan N , SwantonC. Cancer evolution constrained by the immune microenvironment. Cell170(5), 825–827 (2017).
  • Haq R . Trapping cancers as they adapt to survive. Cancer Discov.7(11), 1216–1217 (2017).
  • Rosenthal R , CadieuxEL , SalgadoRet al. Neoantigen-directed immune escape in lung cancer evolution. Nature567(7749), 479–485 (2019).
  • Palucka AK , CoussensLM. The basis of oncoimmunology. Cell164(6), 1233–1247 (2016).
  • Choi SY , GoutPW , CollinsCCet al. Epithelial immune cell-like transition (EIT): a proposed transdifferentiation process underlying immune-suppressive activity of epithelial cancers. Differentiation83(5), 293–298 (2012).
  • Dunn GP , BruceAT , IkedaHet al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat. Immunol.3(11), 991–998 (2002).
  • Beatty GL , GladneyWL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res.21(4), 687–692 (2015).
  • Talukdar S , BhoopathiP , EmdadLet al. Dormancy and cancer stem cells: an enigma for cancer therapeutic targeting. Adv. Cancer Res.141, 43–84 (2019).
  • Garrido F , AptsiauriN , DoorduijnEMet al. The urgent need to recover MHC class I in cancers for effective immunotherapy. Curr. Opin. Immunol.39, 44–51 (2016).
  • Han LY , FletcherMS , UrbauerDLet al. HLA class I antigen processing machinery component expression and intratumoral T-Cell infiltrate as independent prognostic markers in ovarian carcinoma. Clin. Cancer Res.14(11), 3372–3379 (2008).
  • Bukur J , JasinskiS , SeligerB. The role of classical and non-classical HLA class I antigens in human tumors. Semin. Cancer Biol.22(4), 350–358 (2012).
  • Adrián Cabestré F , MoreauP , RiteauBet al. HLA-G expression in human melanoma cells: protection from NK cytolysis. J. Reprod. Immunol.43(2), 183–193 (1999).
  • Seliger B , Jasinski-BergnerS , QuandtDet al. HLA-E expression and its clinical relevance in human renal cell carcinoma. Oncotarget7(41), 67360–67372 (2016).
  • Pardoll DM . The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer12(4), 252–264 (2012).
  • Schlößer HA , TheurichS , Shimabukuro-VornhagenAet al. Overcoming tumor-mediated immunosuppression. Immunotherapy6(9), 973–988 (2014).
  • Anderson AC , JollerN , KuchrooVK. Lag-3, Tim-3 and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity44(5), 989–1004 (2016).
  • Marin-Acevedo JA , DholariaB , SoyanoAEet al. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J. Hematol. Oncol.11(1), 39 (2018).
  • Schnell A , BodL , MadiAet al. The yin and yang of co-inhibitory receptors: toward anti-tumor immunity without autoimmunity. Cell Res.30, 285–299 (2020).
  • Burkholder B , HuangRY , BurgessRet al. Tumor-induced perturbations of cytokines and immune cell networks. Biochim. Biophys. Acta1845(2), 182–201 (2014).
  • Prendergast GC , MalachowskiWP , DuHadawayJBet al. Discovery of IDO1 inhibitors: from bench to bedside. Cancer Res.77(24), 6795–6811 (2017).
  • Choi SY , CollinsCC , GoutPWet al. Cancer-generated lactic acid: a regulatory, immunosuppressive metabolite. J. Pathol.230(4), 350–355 (2013).
  • Morrot A , da FonsecaLM , SalustianoEJet al. Metabolic symbiosis and immunomodulation: how tumor cell-derived lactate may disturb innate and adaptive immune responses. Front Oncol8, 81 (2018).
  • Binnewies M , RobertsEW , KerstenKet al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med.24(5), 541–550 (2018).
  • Guo C , ManjiliMH , SubjeckJRet al. Therapeutic cancer vaccines: past, present and future. Adv. Cancer Res.119, 421–475 (2013).
  • Hinrichs CS , RosenbergSA. Exploiting the curative potential of adoptive T-cell therapy for cancer. Immunol. Rev.257(1), 56–71 (2014).
  • Scott AM , WolchokJD , OldLJ. Antibody therapy of cancer. Nat. Rev. Cancer12(4), 278–287 (2012).
  • Gallotta M , AssiH , DegagnéÉet al. Inhaled TLR9 agonist renders lung tumors permissive to PD-1 blockade by promoting optimal CD4. Cancer Res.78(17), 4943–4956 (2018).
  • Kauffman EC , LiuH , SchwartzMJet al. Toll-like receptor 7 agonist therapy with imidazoquinoline enhances cancer cell death and increases lymphocytic infiltration and proinflammatory cytokine production in established tumors of a renal cell carcinoma mouse model. J. Oncol.2012, 103298 (2012).
  • Wu X , LiJ , ConnollyEMet al. Combined anti-VEGF and Anti-CTLA-4 therapy elicits humoral immunity to galectin-1 which is associated with favorable clinical outcomes. Cancer Immunol. Res.5(6), 446–454 (2017).
  • Kujawski M , ZhangC , HerrmannAet al. Targeting STAT3 in adoptively transferred T cells promotes their in vivo expansion and antitumor effects. Cancer Res.70(23), 9599–9610 (2010).
  • Lu C , TalukderA , SavageNMet al. JAK-STAT-mediated chronic inflammation impairs cytotoxic T lymphocyte activation to decrease anti-PD-1 immunotherapy efficacy in pancreatic cancer. Oncoimmunology6(3), e1291106 (2017).
  • Luke JJ , BaoR , SweisRFet al. WNT/β-catenin pathway activation correlates with immune exclusion across human cancers. Clin. Cancer Res.25(10), 3074–3083 (2019).
  • Yang H , LeeWS , KongSJet al. STING activation reprograms tumor vasculatures and synergizes with VEGFR2 blockade. J. Clin. Invest.130, 4350–4364 (2019).
  • Anderson KG , StromnesIM , GreenbergPD. Obstacles posed by the tumor microenvironment to T cell activity: a case for synergistic therapies. Cancer Cell31(3), 311–325 (2017).
  • Bonifazi M , RossiM , MojaLet al. Bevacizumab in clinical practice: prescribing appropriateness relative to national indications and safety. Oncologist17(1), 117–124 (2012).
  • Anwar MA , ShahM , KimJet al. Recent clinical trends in Toll-like receptor targeting therapeutics. Med. Res. Rev.39(3), 1053–1090 (2019).
  • Zou W , WolchokJD , ChenL. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: mechanisms, response biomarkers and combinations. Sci. Transl. Med.8(328), 328rv4 (2016).
  • Seidel JA , OtsukaA , KabashimaK. Anti-PD-1 and Anti-CTLA-4 therapies in cancer: mechanisms of action, efficacy and limitations. Front. Oncol.8, 86 (2018).
  • Harjunpää H , GuillereyC. TIGIT as an emerging immune checkpoint. Clin. Exp. Immunol.200(2), 108–119 (2019).
  • Friedlaender A , AddeoA , BannaG. New emerging targets in cancer immunotherapy: the role of TIM3. ESMO Open4(Suppl. 3), e000497 (2019).
  • Long L , ZhangX , ChenFet al. The promising immune checkpoint LAG-3: from tumor microenvironment to cancer immunotherapy. Genes Cancer9(5–6), 176–189 (2018).
  • Sheng S , ZhangJ , AiJet al. Aberrant expression of IL-23/IL-23R in patients with breast cancer and its clinical significance. Mol. Med. Rep.17(3), 4639–4644 (2018).
  • Baird AM , LeonardJ , NaickerKMet al. IL-23 is pro-proliferative, epigenetically regulated and modulated by chemotherapy in non-small-cell lung cancer. Lung Cancer79(1), 83–90 (2013).
  • Soares KC , RuckiAA , KimVet al. TGF-β blockade depletes T regulatory cells from metastatic pancreatic tumors in a vaccine dependent manner. Oncotarget6(40), 43005–43015 (2015).
  • Terabe M , RobertsonFC , ClarkKet al. Blockade of only TGF-β 1 and 2 is sufficient to enhance the efficacy of vaccine and PD-1 checkpoint blockade immunotherapy. Oncoimmunology6(5), e1308616 (2017).
  • de Gramont A , FaivreS , RaymondE. Novel TGF-β inhibitors ready for prime time in onco-immunology. Oncoimmunology6(1), e1257453 (2017).
  • Morse MA , HobeikaAC , OsadaTet al. Depletion of human regulatory T cells specifically enhances antigen-specific immune responses to cancer vaccines. Blood112(3), 610–618 (2008).
  • Ohue Y , NishikawaH. Regulatory T (Treg) cells in cancer: can Treg cells be a new therapeutic target. Cancer Sci110(7), 2080–2089 (2019).
  • Miret JJ , KirschmeierP , KoyamaSet al. Suppression of myeloid cell arginase activity leads to therapeutic response in a NSCLC mouse model by activating anti-tumor immunity. J. Immunother. Cancer7(1), 32 (2019).
  • Fleming V , HuX , WeberRet al. Targeting myeloid-derived suppressor cells to bypass tumor-induced immunosuppression. Front. Immunol.9, 398 (2018).
  • Myers KV , AmendSR , PientaKJ. Targeting Tyro3, Axl and MerTK (TAM receptors): implications for macrophages in the tumor microenvironment. Mol. Cancer18(1), 94 (2019).
  • Poh AR , ErnstM. Targeting macrophages in cancer: from bench to bedside. Front. Oncol.8, 49 (2018).
  • WARBURG O . On respiratory impairment in cancer cells. Science124(3215), 269–270 (1956).
  • Feron O . Pyruvate into lactate and back: from the Warburg effect to symbiotic energy fuel exchange in cancer cells. Radiother. Oncol.92(3), 329–333 (2009).
  • Marchiq I , PouysségurJ. Hypoxia, cancer metabolism and the therapeutic benefit of targeting lactate/H(+) symporters. J. Mol. Med. (Berl)94(2), 155–171 (2016).
  • Kouidhi S , BenAyed F , ElgaaiedBenammar A. Targeting tumor metabolism: a new challenge to improve immunotherapy. Front. Immunol.9, 353 (2018).
  • Choi SYC , EttingerSL , LinDet al. Targeting MCT4 to reduce lactic acid secretion and glycolysis for treatment of neuroendocrine prostate cancer. Cancer Med.7(7), 3385–3392 (2018).
  • Todenhöfer T , SeilerR , StewartCet al. Selective inhibition of the lactate transporter MCT4 reduces growth of invasive bladder cancer. Mol. Cancer Ther.17(12), 2746–2755 (2018).
  • Polański R , HodgkinsonCL , FusiAet al. Activity of the monocarboxylate transporter 1 inhibitor AZD3965 in small cell lung cancer. Clin. Cancer Res.20(4), 926–937 (2014).
  • Romero-Garcia S , Moreno-AltamiranoMM , Prado-GarciaHet al. Lactate contribution to the tumor microenvironment: mechanisms, effects on immune cells and therapeutic relevance. Front. Immunol.7, 52 (2016).
  • Jackson CM , ChoiJ , LimM. Mechanisms of immunotherapy resistance: lessons from glioblastoma. Nat. Immunol.20(9), 1100–1109 (2019).
  • Hodi FS , O’DaySJ , McDermottDFet al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med.363(8), 711–723 (2010).
  • Wolchok JD , Chiarion-SileniV , GonzalezRet al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med.377(14), 1345–1356 (2017).
  • Luke JJ , FlahertyKT , RibasAet al. Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat. Rev. Clin. Oncol.14(8), 463–482 (2017).
  • Rizvi NA , HellmannMD , SnyderAet al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small-cell lung cancer. Science348(6230), 124–128 (2015).
  • Snyder A , MakarovV , MerghoubTet al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med.371(23), 2189–2199 (2014).
  • Hugo W , ZaretskyJM , SunLet al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell165(1), 35–44 (2016).
  • Anagnostou V , SmithKN , FordePMet al. Evolution of neoantigen landscape during immune checkpoint blockade in non-small-cell lung cancer. Cancer Discov.7(3), 264–276 (2017).
  • Gettinger S , ChoiJ , HastingsKet al. Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer. Cancer Discov.7(12), 1420–1435 (2017).
  • Peng D , KryczekI , NagarshethNet al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature527(7577), 249–253 (2015).
  • Cristescu R , MoggR , AyersMet al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science362(6411), eaar3593 (2018).
  • Koyama S , AkbayEA , LiYYet al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun.7, 10501 (2016).
  • Chapuy B , StewartC , DunfordAJet al. Genomic analyses of PMBL reveal new drivers and mechanisms of sensitivity to PD-1 blockade. Blood134(26), 2369–2382 (2019).
  • Baylin SB , OhmJE. Epigenetic gene silencing in cancer - a mechanism for early oncogenic pathway addiction. Nat. Rev. Cancer6(2), 107–116 (2006).
  • Morales V , Richard-FoyH. Role of histone N-terminal tails and their acetylation in nucleosome dynamics. Mol. Cell Biol.20(19), 7230–7237 (2000).
  • Portela A , EstellerM. Epigenetic modifications and human disease. Nat. Biotechnol.28(10), 1057–1068 (2010).
  • Kanwal R , GuptaS. Epigenetic modifications in cancer. Clin. Genet.81(4), 303–311 (2012).
  • Qamra A , XingM , PadmanabhanNet al. Epigenomic promoter alterations amplify gene isoform and immunogenic diversity in gastric adenocarcinoma. Cancer Discov.7(6), 630–651 (2017).
  • Wierzbinska JA , TothR , IshaqueNet al. Methylome-based cell-of-origin modeling (Methyl-COOM) identifies aberrant expression of immune regulatory molecules in CLL. Genome Med.12(1), 29 (2020).
  • Berglund A , MillsM , PutneyRMet al. Methylation of immune synapse genes modulates tumor immunogenicity. J. Clin. Invest.130(2), 974–980 (2020).
  • Wylie B , CheeJ , ForbesCAet al. Acquired resistance during adoptive cell therapy by transcriptional silencing of immunogenic antigens. Oncoimmunology8, 1609874 (2019).
  • Liu J , HeD , ChengLet al. p300/CBP inhibition enhances the efficacy of programmed death-ligand 1 blockade treatment in prostate cancer. Oncogene39(19), 3939–3951 (2020).
  • Floess S , FreyerJ , SiewertCet al. Epigenetic control of the foxp3 locus in regulatory T cells. PLoS Biol.5(2), e38 (2007).
  • Lu Z , ZouJ , LiSet al. Epigenetic therapy inhibits metastases by disrupting premetastatic niches. Nature579(7798), 284–290 (2020).
  • Mitra S , LaussM , CabritaRet al. Analysis of DNA methylation patterns in the tumor immune microenvironment of metastatic melanoma. Mol. Oncol.14(5), 933–950 (2020).
  • Topper MJ , VazM , MarroneKAet al. The emerging role of epigenetic therapeutics in immuno-oncology. Nat. Rev. Clin. Oncol.17(2), 75–90 (2020).
  • Chiappinelli KB , ZahnowCA , AhujaNet al. Combining epigenetic and immunotherapy to combat cancer. Cancer Res.76(7), 1683–1689 (2016).
  • Kelly AD , IssaJJ. The promise of epigenetic therapy: reprogramming the cancer epigenome. Curr. Opin. Genet. Dev.42, 68–77 (2017).
  • Símová J , PollákováV , IndrováMet al. Immunotherapy augments the effect of 5-azacytidine on HPV16-associated tumours with different MHC class I-expression status. Br. J. Cancer105(10), 1533–1541 (2011).
  • Christmas BJ , RafieCI , HopkinsACet al. Entinostat converts immune-resistant breast and pancreatic cancers into checkpoint-responsive tumors by reprogramming tumor-infiltrating MDSCs. Cancer Immunol. Res.6(12), 1561–1577 (2018).
  • Park J , ThomasS , MunsterPN. Epigenetic modulation with histone deacetylase inhibitors in combination with immunotherapy. Epigenomics7(4), 641–652 (2015).
  • Juergens RA , WrangleJ , VendettiFPet al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small-cell lung cancer. Cancer Discov.1(7), 598–607 (2011).
  • Crea F , ClermontPL , MaiAet al. Histone modifications, stem cells and prostate cancer. Curr. Pharm. Des.20(11), 1687–1697 (2014).
  • Völkel P , BaryA , RabyLet al. Ezh1 arises from Ezh2 gene duplication but its function is not required for zebrafish development. Sci. Rep.9(1), 4319 (2019).
  • Wang Q , LiuL , ZhangSet al. Long noncoding RNA NEAT1 suppresses hepatocyte proliferation in fulminant hepatic failure through increased recruitment of EZH2 to the LATS2 promoter region and promotion of H3K27me3 methylation. Exp. Mol. Med.52(3), 461–472 (2020).
  • Curry E , GreenI , Chapman-RotheNet al. Dual EZH2 and EHMT2 histone methyltransferase inhibition increases biological efficacy in breast cancer cells. Clin. Epigenetics7, 84 (2015).
  • Coward WR , Feghali-BostwickCA , JenkinsGet al. A central role for G9a and EZH2 in the epigenetic silencing of cyclooxygenase-2 in idiopathic pulmonary fibrosis. FASEB J.28(7), 3183–3196 (2014).
  • Kim E , KimM , WooDHet al. Phosphorylation of EZH2 activates STAT3 signaling via STAT3 methylation and promotes tumorigenicity of glioblastoma stem-like cells. Cancer Cell23(6), 839–852 (2013).
  • Lawrence CL , BaldwinAS. Non-canonical EZH2 transcriptionally activates RelB in triple negative breast cancer. PLoS ONE11(10), e0165005 (2016).
  • Jones A , WangH. Polycomb repressive complex 2 in embryonic stem cells: an overview. Protein Cell1(12), 1056–1062 (2010).
  • Nutt SL , KeenanC , ChopinMet al. EZH2 function in immune cell development. Biol. Chem.401(8), 933–943 (2020).
  • Deevy O , BrackenAP. PRC2 functions in development and congenital disorders. Development146(19), dev181354 (2019).
  • Cyrus S , BurkardtD , WeaverDDet al. PRC2-complex related dysfunction in overgrowth syndromes: a review of EZH2, EED and SUZ12 and their syndromic phenotypes. Am. J. Med. Genet. C Semin. Med. Genet.181(4), 519–531 (2019).
  • Matsubara T , ToyokawaG , TakadaKet al. The association and prognostic impact of enhancer of zeste homologue 2 expression and epithelial-mesenchymal transition in resected lung adenocarcinoma. PLoS ONE14(5), e0215103 (2019).
  • Wu X , ScottH , CarlssonSVet al. Increased EZH2 expression in prostate cancer is associated with metastatic recurrence following external beam radiotherapy. Prostate79(10), 1079–1089 (2019).
  • Stazi G , TaglieriL , NicolaiAet al. Dissecting the role of novel EZH2 inhibitors in primary glioblastoma cell cultures: effects on proliferation, epithelial-mesenchymal transition, migration and on the pro-inflammatory phenotype. Clin. Epigenetics11(1), 173 (2019).
  • Avan A , CreaF , PaolicchiEet al. Molecular mechanisms involved in the synergistic interaction of the EZH2 inhibitor 3-deazaneplanocin A with gemcitabine in pancreatic cancer cells. Mol. Cancer Ther.11(8), 1735–1746 (2012).
  • Huang B , HuangM , LiQ. Cancer-associated fibroblasts promote angiogenesis of hepatocellular carcinoma by VEGF-mediated EZH2/VASH1 pathway. Technol. Cancer Res. Treat18, (2019). https://doi.org/10.1177/1533033819879905
  • Zeng Z , YangY , WuH. MicroRNA-765 alleviates the malignant progression of breast cancer via interacting with EZH1. Am. J. Transl. Res.11(7), 4500–4507 (2019).
  • Nakagawa M , KitabayashiI. Oncogenic roles of enhancer of zeste homolog 1/2 in hematological malignancies. Cancer Sci.109(8), 2342–2348 (2018).
  • Quintanal-Villalonga Á , ChanJM , YuHAet al. Lineage plasticity in cancer: a shared pathway of therapeutic resistance. Nat. Rev. Clin. Oncol.17(6), 360–371 (2020).
  • Clermont PL , LinD , CreaFet al. Polycomb-mediated silencing in neuroendocrine prostate cancer. Clin. Epigenetics7, 40 (2015).
  • Zhao Y , WangXX , WuWet al. EZH2 regulates PD-L1 expression via HIF-1α in non-small-cell lung cancer cells. Biochem. Biophys. Res. Commun.517(2), 201–209 (2019).
  • Xiao G , JinLL , LiuCQet al. EZH2 negatively regulates PD-L1 expression in hepatocellular carcinoma. J. Immunother. Cancer7(1), 300 (2019).
  • Wu J , LanierLL. Natural killer cells and cancer. Adv. Cancer Res.90, 127–156 (2003).
  • Bugide S , GreenMR , WajapeyeeN. Inhibition of enhancer of zeste homolog 2 (EZH2) induces natural killer cell-mediated eradication of hepatocellular carcinoma cells. Proc. Natl Acad. Sci. USA115(15), E3509–E3518 (2018).
  • Yin J , LeavenworthJW , LiYet al. Ezh2 regulates differentiation and function of natural killer cells through histone methyltransferase activity. Proc. Natl Acad. Sci. USA112(52), 15988–15993 (2015).
  • Yin Y , QiuS , LiXet al. EZH2 suppression in glioblastoma shifts microglia toward M1 phenotype in tumor microenvironment. J. Neuroinflammation14(1), 220 (2017).
  • Pang B , ZhengXR , TianJXet al. EZH2 promotes metabolic reprogramming in glioblastomas through epigenetic repression of EAF2-HIF1α signaling. Oncotarget7(29), 45134–45143 (2016).
  • Bray M , DriscollJ , HugginsJW. Treatment of lethal Ebola virus infection in mice with a single dose of an S-adenosyl-L-homocysteine hydrolase inhibitor. Antiviral Res.45(2), 135–147 (2000).
  • Miranda TB , CortezCC , YooCBet al. DZNep is a global histone methylation inhibitor that reactivates developmental genes not silenced by DNA methylation. Mol. Cancer Ther.8(6), 1579–1588 (2009).
  • Tan J , YangX , ZhuangLet al. Pharmacologic disruption of polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev.21(9), 1050–1063 (2007).
  • Crea F , HurtEM , MathewsLAet al. Pharmacologic disruption of polycomb repressive complex 2 inhibits tumorigenicity and tumor progression in prostate cancer. Mol. Cancer10, 40 (2011).
  • Hung SW , ModyH , MarracheSet al. Pharmacological reversal of histone methylation presensitizes pancreatic cancer cells to nucleoside drugs: in vitro optimization and novel nanoparticle delivery studies. PLoS ONE8, e71196 (2013).
  • Hoy SM . Tazemetostat: first approval. Drugs80(5), 513–521 (2020).
  • Knutson SK , WarholicNM , WigleTJet al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc. Natl Acad. Sci. USA110(19), 7922–7927 (2013).
  • Knutson SK , WarholicNM , WigleTJet al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc. Natl Acad Sci. USA110(19), 7922–7927 (2013).
  • Knutson SK , KawanoS , MinoshimaYet al. Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma. Mol. Cancer Ther.13(4), 842–854 (2014).
  • Italiano A , SoriaJC , ToulmondeMet al. Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: a first-in-human, open-label, Phase I study. Lancet Oncol.19(5), 649–659 (2018).
  • Leslie M . First EZH2 inhibitor approved-for rare sarcoma. Cancer Discov.10(3), 333–334 (2020).
  • Sarkozy C , MorschhauserF , DuboisSet al. A LYSA Phase Ib study of tazemetostat (EPZ-6438) plus R-CHOP in patients with newly diagnosed diffuse large B-cell lymphoma (DLBCL) with poor prognosis features. Clin. Cancer Res.26(13), 3145–3153 (2020).
  • McCabe MT , OttHM , GanjiGet al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature492(7427), 108–112 (2012).
  • Yap TA , WinterJN , Giulino-RothLet al. Phase I study of the novel enhancer of zeste homolog 2 (EZH2) inhibitor GSK2816126 in patients with advanced hematologic and solid tumors. Clin. Cancer Res.25(24), 7331–7339 (2019).
  • Vaswani RG , GehlingVS , DakinLAet al. Identification of (R)-N-((4-Methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide (CPI-1205), a potent and selective inhibitor of histone methyltransferase EZH2, suitable for Phase I clinical trials for B-Cell lymphomas. J. Med. Chem.59(21), 9928–9941 (2016).
  • Lu B , ShenX , ZhangLet al. Discovery of EBI-2511: a highly potent and orally active EZH2 inhibitor for the treatment of non-hodgkin’s lymphoma. ACS Med. Chem. Lett.9(2), 98–102 (2018).
  • Huang Y , ZhangJ , YuZet al. Discovery of first-in-class, potent and orally bioavailable embryonic ectoderm development (EED) inhibitor with robust anticancer efficacy. J. Med. Chem.60(6), 2215–2226 (2017).
  • He Y , SelvarajuS , CurtinMLet al. The EED protein-protein interaction inhibitor A-395 inactivates the PRC2 complex. Nat. Chem. Biol.13(4), 389–395 (2017).
  • Marabondo S , KaufmanHL. High-dose interleukin-2 (IL-2) for the treatment of melanoma: safety considerations and future directions. Expert Opin. Drug Saf.16(12), 1347–1357 (2017).
  • Zingg D , Arenas-RamirezN , SahinDet al. The histone methyltransferase Ezh2 controls mechanisms of adaptive resistance to tumor immunotherapy. Cell Rep.20(4), 854–867 (2017).
  • Goswami S , ApostolouI , ZhangJet al. Modulation of EZH2 expression in T cells improves efficacy of anti-CTLA-4 therapy. J. Clin. Invest.128(9), 3813–3818 (2018).
  • Hong YK , LiY , PanditHet al. Epigenetic modulation enhances immunotherapy for hepatocellular carcinoma. Cell. Immunol.336, 66–74 (2019).
  • Zhou L , MudiantoT , MaXet al. Targeting EZH2 enhances antigen presentation, antitumor immunity and circumvents anti-PD-1 resistance in head and neck cancer. Clin. Cancer Res.26(1), 290–300 (2020).
  • Momparler RL , CôtéS , MarquezVEet al. Comparison of the antineoplastic action of 3-deazaneplanocin-A and inhibitors that target the catalytic site of EZH2 histone methyltransferase. Cancer Rep. Rev.3, 1–4 (2020).
  • Sun F , LiJ , YuQet al. Loading 3-deazaneplanocin A into pegylated unilamellar liposomes by forming transient phenylboronic acid-drug complex and its pharmacokinetic features in Sprague-Dawley rats. Eur. J. Pharm. Biopharm80(2), 323–331 (2012).
  • Chamoto K , HataeR , HonjoT. Current issues and perspectives in PD-1 blockade cancer immunotherapy. Int. J. Clin. Oncol.25(5), 790–800 (2020).
  • Kailayangiri S , AltvaterB , LeschSet al. EZH2 inhibition in ewing sarcoma upregulates G. Mol. Ther.27(5), 933–946 (2019).
  • Singh AK , McGuirkJP. CAR T cells: continuation in a revolution of immunotherapy. Lancet Oncol.21(3), e168–e178 (2020).
  • Bisserier M , WajapeyeeN. Mechanisms of resistance to EZH2 inhibitors in diffuse large B-cell lymphomas. Blood131(19), 2125–2137 (2018).
  • Crea F , FornaroL , BocciGet al. EZH2 inhibition: targeting the crossroad of tumor invasion and angiogenesis. Cancer Metastasis Rev.31(3–4), 753–761 (2012).
  • Xu Z , SunY , GuoYet al. NF-YA promotes invasion and angiogenesis by upregulating EZH2-STAT3 signaling in human melanoma cells. Oncol. Rep.35(6), 3630–3638 (2016).
  • Luo J , WangK , YehSet al. LncRNA-p21 alters the antiandrogen enzalutamide-induced prostate cancer neuroendocrine differentiation via modulating the EZH2/STAT3 signaling. Nat Commun10(1), 2571 (2019).
  • Chen Z , DuY , LiuXet al. EZH2 inhibition suppresses bladder cancer cell growth and metastasis via the JAK2/STAT3 signaling pathway. Oncol. Lett.18(1), 907–915 (2019).
  • Kim KH , RobertsCW. Targeting EZH2 in cancer. Nat. Med.22(2), 128–134 (2016).
  • Yamamoto Y , NingG , HowittBEet al. In vitro and in vivo correlates of physiological and neoplastic human Fallopian tube stem cells. J. Pathol.238(4), 519–530 (2016).
  • Xu B , AbourbihS , SircarKet al. Enhancer of zeste homolog 2 expression is associated with metastasis and adverse clinical outcome in clear cell renal cell carcinoma: a comparative study and review of the literature. Arch. Pathol. Lab. Med.137(10), 1326–1336 (2013).
  • O’Carroll D , ErhardtS , PaganiMet al. The polycomb-group gene Ezh2 is required for early mouse development. Mol. Cell. Biol.21(13), 4330–4336 (2001).
  • Wei L , WangJ , LampertEet al. Intratumoral and intertumoral genomic heterogeneity of multifocal localized prostate cancer impacts molecular classifications and genomic prognosticators. Eur. Urol.71(2), 183–192 (2017).
  • Chan TA , YarchoanM , JaffeeEet al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann. Oncol.30(1), 44–56 (2019).
  • Fornaro L , VivaldiC , OrlandiPet al. Longitudinal evaluation of angiogenesis-related circulating biomarkers during second-line treatment with paclitaxel and ramucirumab in advanced gastroesophageal cancer. Ann. Oncol.30(Suppl. 4), iv88 (2019).
  • Forschner A , BattkeF , HadaschikDet al. Tumor mutation burden and circulating tumor DNA in combined CTLA-4 and PD-1 antibody therapy in metastatic melanoma - results of a prospective biomarker study. J. Immunother. Cancer7(1), 180 (2019).
  • Holdenrieder S , StieberP. Apoptotic markers in cancer. Clin. Biochem.37(7), 605–617 (2004).
  • Holdenrieder S , NagelD , SchalhornAet al. Clinical relevance of circulating nucleosomes in cancer. Ann. NY Acad. Sci.1137, 180–189 (2008).
  • Gabler C , BlankN , HieronymusTet al. Extranuclear detection of histones and nucleosomes in activated human lymphoblasts as an early event in apoptosis. Ann. Rheum. Dis.63(9), 1135–1144 (2004).
  • Thierry AR , ElMessaoudi S , GahanPBet al. Origins, structures and functions of circulating DNA in oncology. Cancer Metastasis Rev.35(3), 347–376 (2016).
  • Holdenrieder S , StieberP , BodenmüllerHet al. Nucleosomes in serum as a marker for cell death. Clin. Chem. Lab. Med.39(7), 596–605 (2001).
  • Holdenrieder S , DharumanY , StandopJet al. Novel serum nucleosomics biomarkers for the detection of colorectal cancer. Anticancer Res.34(5), 2357–2362 (2014).
  • McAnena P , BrownJA , KerinMJ. Circulating nucleosomes and nucleosome modifications as biomarkers in cancer. Cancers (Basel)9(1), 5 (2017).
  • Bauden M , PamartD , AnsariDet al. Circulating nucleosomes as epigenetic biomarkers in pancreatic cancer. Clin. Epigenetics7, 106 (2015).
  • Rahier JF , DruezA , FaugerasLet al. Circulating nucleosomes as new blood-based biomarkers for detection of colorectal cancer. Clin. Epigenetics9, 53 (2017).
  • Guiot J , StrumanI , ChavezVet al. Altered epigenetic features in circulating nucleosomes in idiopathic pulmonary fibrosis. Clin. Epigenetics9, 84 (2017).