|Table of Contents|

Advances of epigenetic therapy in acute myeloid leukemia

Journal Of Modern Oncology[ISSN:1672-4992/CN:61-1415/R]

Issue:
2023 23
Page:
4467-4471
Research Field:
Publishing date:

Info

Title:
Advances of epigenetic therapy in acute myeloid leukemia
Author(s):
LI QiLI Yinghua
Department of Hematology,First Affiliated Hospital of Harbin Medical University,Heilongjiang Harbin 150000,China.
Keywords:
epigenetic drugsacute myeloid leukemiatargeted therapyDNMTHDAC
PACS:
R733.71
DOI:
10.3969/j.issn.1672-4992.2023.23.033
Abstract:
With the development of second-generation sequencing and other technologies,more and more epigenetic regulation-related genes have been found mutations or deletions in acute myeloid leukemia(AML).These factors are often associated with treatment resistance and poor prognosis.Epigenetic regulation is mainly regulated through DNA methylation,histone modification,non-coding RNA and other mechanisms,which play an important role in the occurrence and development of AML.In clinical practice,compared with traditional chemotherapy,epigenetic drugs combined with chemotherapy have achieved significant efficacy in the treatment of AML,and improved the remission rate of patients.This article reviews the research progress of epigenetic therapy for AML.

References:

[1] DHNER H,WEISDORF DJ,BLOOMFIELD CD.Acute myeloid leukemia[J].N Engl J Med,2015,373(12):1136-1152.
[2] DHNER H,ESTEY E,GRIMWADE D,et al.Diagnosis and management of AML in adults:2017 ELN recommendations from an international expert panel[J].Blood,2017,129(4):424-447.
[3] ZAPPASODI R,MERGHOUB T,WOLCHOK JD.Emerging concepts for immune checkpoint blockade-based combination therapies[J].Cancer Cell,2018,33(4):581-598.
[4] BOSE P,VACHHANI P,CORTES JE.Treatment of relapsed/refractory acute myeloid leukemia[J].Curr Treat Options Oncol,2017,18(3):17.
[5] NEFF T,ARMSTRONG SA.Recent progress toward epigenetic therapies:the example of mixed lineage leukemia[J].Blood,2013,121(24):4847-4853.
[6] ROY DM,WALSH LA,CHAN TA.Driver mutations of cancer epigenomes[J].Protein Cell,2014,5(4):265-296.
[7] ALLIS CD,JENUWEIN T.The molecular hallmarks of epigenetic control[J].Nat Rev Genet,2016,17(8):487-500.
[8] DAWSON MA,KOUZARIDES T.Cancer epigenetics:from mechanism to therapy[J].Cell,2012,150(1):12-27.
[9] GREER EL,SHI Y.Histone methylation:a dynamic mark in health,disease and inheritance[J].Nat Rev Genet,2012,13(5):343-357.
[10] VU LP,PICKERING BF,CHENG Y,et al.The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells[J].Nat Med,2017,23(11):1369-1376.
[11] CAO R,WANG L,WANG H,et al.Role of histone H3 lysine 27 methylation in Polycomb-group silencing[J].Science,2002,298(5595):1039-1043.
[12] DHALL A,ZEE BM,YAN F,et al.Intersection of epigenetic and metabolic regulation of histone modifications in acute myeloid leukemia[J].Front Oncol,2019,9:432.
[13] KHAN SN,JANKOWSKA AM,MAHFOUZ R,et al.Multiple mechanisms deregulate EZH2 and histone H3 lysine 27 epigenetic changes in myeloid malignancies[J].Leukemia,2013,27(6):1301-1309.
[14] TANAKA S,MIYAGI S,SASHIDA G,et al.Ezh2 augments leukemogenicity by reinforcing differentiation blockage in acute myeloid leukemia[J].Blood,2012,120(5):1107-1117.
[15] SINGH AA,PETRAGLIA F,NEBBIOSO A,et al.Multi-omics profiling reveals a distinctive epigenome signature for high-risk acute promyelocytic leukemia[J].Oncotarget,2018,9(39):25647-25660.
[16] NG HH,FENG Q,WANG H,et al.Lysine methylation within the globular domain of histone H3 by Dot1 is important for telomeric silencing and Sir protein association[J].Genes Dev,2002,16(12):1518-1527.
[17] FENG Q,WANG H,NG HH,et al.Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain[J].Curr Biol,2002,12(12):1052-1058.
[18] DAIGLE SR,OLHAVA EJ,THERKELSEN CA,et al.Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor[J].Cancer Cell,2011,20(1):53-65.
[19] CUSAN M,CAI SF,MOHAMMAD HP,et al.LSD1 inhibition exerts its antileukemic effect by recommissioning PU.1- and C/EBPα-dependent enhancers in AML[J].Blood,2018,131(15):1730-1742.
[20] STEIN EM,GARCIA-MANERO G,RIZZIERI DA,et al.The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia[J].Blood,2018,131(24):2661-2669.
[21] SAN JOS-ENRIZ E,GIMENEZ-CAMINO N,AGIRRE X,et al.HDAC inhibitors in acute myeloid leukemia[J].Cancers (Basel),2019,11(11):1794.
[22] LI Y,WANG Y,ZHOU Y,et al.Cooperative effect of chidamide and chemotherapeutic drugs induce apoptosis by DNA damage accumulation and repair defects in acute myeloid leukemia stem and progenitor cells[J].Clin Epigenetics,2017,9:83.
[23] XU WS,PARMIGIANI RB,MARKS PA.Histone deacetylase inhibitors:molecular mechanisms of action[J].Oncogene,2007,26(37):5541-5552.
[24] BOTS M,VERBRUGGE I,MARTIN BP,et al.Differentiation therapy for the treatment of t(8;21) acute myeloid leukemia using histone deacetylase inhibitors[J].Blood,2014,123(9):1341-1352.
[25] MIMS AS,MISHRA A,ORWICK S,et al.A novel regimen for relapsed/refractory adult acute myeloid leukemia using a KMT2A partial tandem duplication targeted therapy:results of phase 1 study NCI 8485[J].Haematologica,2018,103(6):982-987.
[26] 李捷瑶,徐宛婷,卓然,等.西达本胺抗肿瘤作用机制的研究进展[J].肿瘤药学,2019,9(01):6-9,25. LI JY,XU WT,ZHUO R,et al.The research progress of chidamide antitumor mechanism[J].Anti-Tumor Pharmacy,2019,9(01):6-9,25.
[27] 郭丹,李英花.表观遗传学调控及治疗在急性髓系白血病中的研究进展[J].临床与病理杂志,2021,41(6):1411-1419. GUO D,LI YH.Research progress of epigenetic regulation and therapy in acute myeloid leukemia[J].Journal of Clinical and Pathological Research,2021,41(6):1411-1419.
[28] MOORE LD,LE T,FAN G.DNA methylation and its basic function[J].Neuropsychopharmacology,2013,38(1):23-38.
[29] CUI D,XU X.DNA methyltransferases,DNA methylation,and age-associated cognitive function[J].Int J Mol Sci,2018,19(5):1315-1319.
[30] OHGAMI RS,ARBER DA.The diagnostic and clinical impact of genetics and epigenetics in acute myeloid leukemia[J].Int J Lab Hematol,2015,37 Suppl 1:122-132.
[31] MOMPARLER RL.Pharmacology of 5-Aza-2'-deoxycytidine (decitabine)[J].Semin Hematol,2005,42(3 Suppl 2):9-16.
[32] NIETO M,DEMOLIS P,BHANZIN E,et al.The European Medicines Agency review of decitabine (Dacogen) for the treatment of adult patients with acute myeloid leukemia:summary of the scientific assessment of the committee for medicinal products for human use[J].Oncologist,2016,21(6):692-700.
[33] ZHENG L,HUANG L,HUI Y,et al.Clinical efficacy of decitabine-containing induction chemotherapy in de novo non-elderly acute myeloid leukemia[J].Int J Oncol,2020,56(6):1521-1528.
[34] SCHAEFER M,HAGEMANN S,HANNA K,et al.Azacytidine inhibits RNA methylation at DNMT2 target sites in human cancer cell lines[J].Cancer Res,2009,69(20):8127-8132.
[35] ZEIDAN AM,WANG R,WANG X,et al.Clinical outcomes of older patients with AML receiving hypomethylating agents:a large population-based study in the United States[J].Blood Adv,2020,4(10):2192-2201.
[36] CHEN X,ZHOU W,SONG RH,et al.Tumor suppressor CEBPA interacts with and inhibits DNMT3A activity[J].Sci Adv,2022,8(4):eabl5220.
[37] YE J,ZHA J,SHI Y,et al.Co-inhibition of HDAC and MLL-menin interaction targets MLL-rearranged acute myeloid leukemia cells via disruption of DNA damage checkpoint and DNA repair[J].Clin Epigenetics,2019,11(1):137.
[38] ABDEL-MAGID AF.Potential treatment of acute leukemia with inhibitors of Menin/MLL interaction[J].ACS Med Chem Lett,2018,9(9):868-869.
[39] KRIVTSOV AV,EVANS K,GADREY JY,et al.A Menin-MLL inhibitor induces specific chromatin changes and eradicates disease in models of MLL-rearranged leukemia[J].Cancer Cell,2019,36(6):660-673.
[40] GREMBECKA J,HE S,SHI A,et al.Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia[J].Nat Chem Biol,2012,8(3):277-284.
[41] BORKIN D,HE S,MIAO H,et al.Pharmacologic inhibition of the Menin-MLL interaction blocks progression of MLL leukemia in vivo[J].Cancer Cell,2015,27(4):589-602.
[42] DZAMA MM,STEINER M,RAUSCH J,et al.Synergistic targeting of FLT3 mutations in AML via combined Menin-MLL and FLT3 inhibition[J].Blood,2020,136(21):2442-2456.
[43] PREZ-SALVIA M,ESTELLER M.Bromodomain inhibitors and cancer therapy:From structures to applications[J].Epigenetics,2017,12(5):323-339.
[44] ZUBER J,SHI J,WANG E,et al.RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia[J].Nature,2011,478(7370):524-528.
[45] DELMORE JE,ISSA GC,LEMIEUX ME,et al.BET bromodomain inhibition as a therapeutic strategy to target c-Myc[J].Cell,2011,146(6):904-917.
[46] HERRMANN H,BLATT K,SHI J,et al.Small-molecule inhibition of BRD4 as a new potent approach to eliminate leukemic stem- and progenitor cells in acute myeloid leukemia AML[J].Oncotarget,2012,3(12):1588-1599.
[47] BERENGUER-DAIZ C,ASTORGUES-XERRI L,ODORE E,et al.OTX015 (MK-8628),a novel BET inhibitor,displays in vitro and in vivo antitumor effects alone and in combination with conventional therapies in glioblastoma models[J].Int J Cancer,2016,139(9):2047-2055.
[48] JIN Q,MARTINEZ CA,ARCIPOWSKI KM,et al.USP7 cooperates with NOTCH1 to drive the oncogenic transcriptional program in T-cell leukemia[J].Clin Cancer Res,2019,25(1):222-239.
[49] HOHMANN AF,MARTIN LJ,MINDER JL,et al.Sensitivity and engineered resistance of myeloid leukemia cells to BRD9 inhibition[J].Nat Chem Biol,2016,12(9):672-679.
[50] JIANG Y,HU T,WANG T,et al.AMP-activated protein kinase links acetyl-CoA homeostasis to BRD4 recruitment in acute myeloid leukemia[J].Blood,2019,134(24):2183-2194.
[51] DINARDO CD,RAVANDI F,AGRESTA S,et al.Characteristics,clinical outcome,and prognostic significance of IDH mutations in AML[J].Am J Hematol,2015,90(8):732-736.
[52] ZEIDNER JF.Differentiating the differentiation syndrome associated with IDH inhibitors in AML[J].Clin Cancer Res,2020,26(16):4174-4176.
[53] SULKOWSKI PL,CORSO CD,ROBINSON ND,et al.2-Hydroxyglutarate produced by neomorphic IDH mutations suppresses homologous recombination and induces PARP inhibitor sensitivity[J].Sci Transl Med,2017,9(375):eaal2463.
[54] POPOVICI-MULLER J,LEMIEUX RM,ARTIN E,et al.Discovery of AG-120 (ivosidenib):a first-in-class mutant IDH1 inhibitor for the treatment of IDH1 mutant cancers[J].ACS Med Chem Lett,2018,9(4):300-305.
[55] STEIN EM,DINARDO CD,POLLYEA DA,et al.Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia[J].Blood,2017,130(6):722-731.
[56] DINARDO CD,STEIN AS,STEIN EM,et al.Mutant isocitrate dehydrogenase 1 inhibitor ivosidenib in combination with azacitidine for newly diagnosed acute myeloid leukemia[J].J Clin Oncol,2021,39(1):57-65.
[57] STEIN EM,DINARDO CD,FATHI AT,et al.Ivosidenib or enasidenib combined with intensive chemotherapy in patients with newly diagnosed AML:a phase 1 study[J].Blood,2021,137(13):1792-1803.
[58] KIM D,KIM KI,BAEK SH.Roles of lysine-specific demethylase 1 (LSD1) in homeostasis and diseases[J].J Biomed Sci,2021,28(1):41.
[59] SCHULTE JH,LIM S,SCHRAMM A,et al.Lysine-specific demethylase 1 is strongly expressed in poorly differentiated neuroblastoma:implications for therapy[J].Cancer Res,2009,69(5):2065-2071.
[60] HARRIS WJ,HUANG X,LYNCH JT,et al.The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells[J].Cancer Cell,2012,21(4):473-487.
[61] JOHNSTON G,RAMSEY HE,LIU Q,et al.Nascent transcript and single-cell RNA-seq analysis defines the mechanism of action of the LSD1 inhibitor INCB059872 in myeloid leukemia[J].Gene,2020,752:144758.
[62] SCHENK T,CHEN WC,GLLNER S,et al.Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia[J].Nat Med,2012,18(4):605-611.
[63] ISHIKAWA Y,NAKAYAMA K,MORIMOTO M,et al.Synergistic anti-AML effects of the LSD1 inhibitor T-3775440 and the NEDD8-activating enzyme inhibitor pevonedistat via transdifferentiation and DNA rereplication[J].Oncogenesis,2017,6(9):e377.

Memo

Memo:
-
Last Update: 2023-10-31