|Table of Contents|

Research progress on therapeutic effect and prognosis of different gene mutations in patients with acute myeloid leukemia

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

Issue:
2023 22
Page:
4284-4289
Research Field:
Publishing date:

Info

Title:
Research progress on therapeutic effect and prognosis of different gene mutations in patients with acute myeloid leukemia
Author(s):
LIU YangLIU Shuchuan
Department of Hematology,the First Affiliated Hospital of Harbin Medical University,Heilongjiang Harbin 150001,China.
Keywords:
acute myeloid leukemiaFMS-like tyrosine kinase 3 genenuclear phosphoprotein generunt-related transcription factor geneWilms tumor 1 gene
PACS:
R733.71
DOI:
10.3969/j.issn.1672-4992.2023.22.036
Abstract:
Acute myeloid leukemia (AML) is a group of hematological malignancies originated from hematopoietic stem cells.In recent years,cytogenetic changes have been considered as an important independent prognostic factor in patients with AML.FLT3,NPM1,RUNX1 and WT1 gene mutations have become important risk stratification indicators.Among them,FLT3-ITD mutation can lead to uncontrolled proliferation of leukemic cells,which is an independent poor prognostic factor.NPM1 mutation indicates a better prognosis,so we can consider the combination of all-trans retinoic acid and arsenic,anti-CD33 monoclonal antibody,actinomycin and so on.RUNX1 gene mutation is a hot spot mutation found in myeloid tumors in recent years,which occurs frequently in AML with normal karyotype,which is related to short overall survival time and immature cell morphology,and is considered to be a necessary tumor suppressor for the survival of AML.The expression of WT1 gene,a tumor suppressor gene regulating hematopoiesis and apoptosis,may be related to clinical remission and recurrence,and can be used as a potential prognostic factor and a new target for specific immunotherapy.Quantitative PCR is more advantageous than WT1 in detecting NPM1 mutation in patients with relapse.The above molecular markers are reviewed in this paper.The purpose of this study is to find potential gene targets and to provide reference for the development of new gene targeting drugs for accurate treatment of AML.

References:

[1] 宋慧慧,李明春.急性髓系白血病基因水平发病机制的研究进展[J].中国现代应用药学,2020,37(3):371-377. SONG HH,LI MC.Research progress on gene level pathogenesis of acute myeloid leukemia[J].Chinese Modern Applied Pharmacy,2020,37(3):371-377.
[2] MAKKAR H,MAJHI RK,GOEL H,et al.Acute myeloid leukemia:novel mutations and their clinical implications[J].Am J Blood Res,2023,13(1):12-27.
[3] TSAPOGAS P,MOONEY CJ,BROWN G,et al.The cytokine Flt3-ligand in normal and malignant hematopoiesis[J].Int J Mol Sci,2017,18(6):1115.
[4] POUBEL CP,MANSUR MB,BORONI M,et al.FLT3 overexpression in acute leukaemias:New insights into the search for molecular mechanisms[J].BBA -Reviews on Cancer,2019,1872(1):80-88.
[5] CHENG J,QU L,WANG J,et al.High expression of FLT3 is a risk factor in leukemia[J].Mol Med Rep,2018,17(2):2885-2892.
[6] TARLOCK K,ALONZO TA,LOKEN MR,et al.Disease characteristics and prognostic implications of cell-surface FLT3 receptor (CD135) expression in pediatric acute myeloid leukemia:a report from the Children's Oncology Group[J].Clin Cancer Res,2017,23(14):3649-3656.
[7] DOHNER 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.
[8]刘彦,克晓燕,王晶,等.伴FLT3-ITD突变的急性髓系白血病的临床特征和预后[J].中国实验血液学杂志,2018,26(2):354-358. LIU Y,KE XY,WANG J,et al.Clinical characteristics and prognosis of acute myeloid leukemia with FLT3-ITD mutation[J].Chinese Journal of Experimental Hematology,2018,26(2):354-358.
[9] DAVER N,SCHLENK RF,RUSSELL NH,et al.Targeting FLT3 mutations in AML:review of current knowledge and evidence[J].Leukemia,2019,33(2):299-312.
[10] 李沛颖,刘晓燕,宗志峰,等.FLT3、NPM1、DNMT3A及IDH基因突变对非M3型急性髓系白血病预后影响的研究进展.临床医药文献电子杂志,2020,7(21):49. LI PY,LIU XY,ZONG ZF,et al.Research progress of FLT3,NPM1,DNMT3A and IDH gene mutations on the prognosis of non-M3 acute myeloid leukemia[J].Electronic Journal of Clinical Medical Literature,2020,7(21):49.
[11]CHEN F,SUN J,YIN C,et al.Impact of FLT3-ITD allele ratio and ITD length on therapeutic outcome in cytogenetically normal AML patients without NPM1 mutation[J].Bone Marrow Transplant,2020,55(4):740-748.
[12] DAVER N,SCHLENK RF,RUSSELL NH,et al.Targeting FLT3 mutations in AML:review of current knowledge and evidence[J].Leukemia,2019,33(10):299-312.
[13]KUROSAWA S,YAMAGUCHI H,YAMAGUCHI T,et al.The prognostic impact of FLT3-ITD,NPM1 and CEBPa in cytogenetically intermediate-risk AML after first relapse[J].Int J Hematol,2020,112(2):200-209.
[14] ZHAO JC,AGARWAL S,AHMAD H,et al.A review of FLT3 inhibitors in acute myeloid leukemia[J].Blood Rev,2022,52:100905.
[15]BOX JK,PAQUET N,ADAMS MN,et al.Nucleophosmin:from structure and function to disease development[J].BMC Mol Biol,2016,17(1):19.
[16]HEATH M,CHAN SM,MINDEN MD,et al.Biological and clinical consequences of NPM1-mutations in AML[J].Leukemia,2017,31(20):798-807.
[17] BRODSKA B,SASINKOVA M,KUZELOVA K,et al.Nucleophosmin in leukemia:consequences of anchor loss[J].Int J Biochem Cell Biol,2019,111:52-62.
[18] YANG K,YANG J,YI J.Nucleolar stress:hallmarks,sensing mechanism and diseases[J].Cell Stress J,2018,2(6):125-140.
[19] POLETTO M,MALFATTI MC,DORJSUREN D,et al.Tell,inhibitors of the apurinic/apyrimidinic endonuclease 1(APE1)/nucleophosmin (NPM1) interaction that display anti-tumor properties[J].Mol Carcinog,2016,55(5):688-704.
[20] IVEY A,HILLS RK,SIMPSON MA,et al.Assessment of minimal residual disease in standard-risk AML[J].N Engl J Med,2016,374(5):422-433.
[21]LINCH DC,HILLS RK,BURNETT AK,et al.Analysis of the clinical impact of NPM1 mutant allele burden in a large cohort of younger adult patients with acute myeloid leukemia[J].Br J Haematol,2020,188(6):852-859.
[22] HEATH EM,CHAN SM,MINDEN MD,et al.Biological and clinical conseq-uences of NPM1 mutations in AML[J].Leukemia,2017,31(4):798-807.
[23]DOHNER H,WEI AH,APPELBAUM FR,et al.Diagnosis and management of AML in adults:2022 recommendations from an international expert panel on behalf of the ELN[J].Blood,2022,140(12):1345-1377.
[24]LAMBLE AJ,BRODERSEN LE,ALONZO TA,et al.CD123 expression is associated with high-risk disease characteristics in childhood acute myeloid leukemia:a report from the children's oncology group[J].J Clin Oncol,2022,40(3):252-261.
[25]ZEIDNER JF,VINCENT BG,IVANOVA A,et al.Phase II trial of pembrolizumab after high-dose cytarabine in relapsed/refractory acute myeloid leukemia[J].Blood Cancer Discov,2021,2(6):616-629.
[26]刘艳荣,赖悦云,常艳,等.核型正常的NPM1突变阳性的急性髓系白血病的免疫表型与临床特征分析[J].中国实验血液学杂志,2013,21(6):1385-1398. LIU YR,LAI YY,CHANG Y,et al.Immunophenotypic and clinical characteristic analysis of NPM1 mutated acute myeloid leukemia with a normal karyotype[J].Chinese Journal of Experimental Hematology,2013,21(6):1385-1398.
[27] 李玲,吕晓东,米瑞华,等.急性髓系白血病患者NPM1、FLT3和C-KIT基因突变的检测及其预后分析[J].中国实验血液学杂志,2013,21(3):601-606. LI L,LYU XD,MI RH,et al.Detection of NPM1,FLT3 and C-KIT mutations in acute myeloid leukemia and their prognostic analysis[J].Chinese Journal of Experimental Hematology,2013,21(3):601-606.
[28] DE PROPRIS MS,RAPONI S,DIVERIO D,et al.High CD33 expression levels in acute myeloid leukemia cells carrying the nucleophosmin (NPM1) mutation[J].Haematologica,2011,96(10):1548-1551.
[29] ARBER DA,ORAZI A,HASSERJIAN R,et al.The 2016 revision to the World Health Organizationclassification of myeloid neoplasms and acute leukemia[J].Blood,2016,127(20):2391-2405.
[30]PATEL SS,KUO FC,GIBSON CJ,et al.High NPM1-mutant allele burden at diagnosis predicts unfavorable outcomes in de novo AML[J].Blood,2018,131(25):2816-2825.
[31] KHOURY JD,SOLARY E,ABLA O,et al.The 5th edition of the world health organization classification of haematolymphoid tumours:Myeloid and histiocytic/dendritic neoplasms[J].Leukemia,2022,36(7):1703-1719.
[32]FORGHIERI F,RIVA G,LAGRECA I,et al.Neoantigen-specific T-cell immune responses:Theparadigm of NPM1-mutated acute myeloid leukemia[J].Int J Mol Sci,2021,22(17):9159.
[33]HINDLEY A,CATHERWOOD MA,MCMULLIN MF,et al.Significance of npm1 gene mutations in aml[J].Int J Mol Sci,2021,22(18):10040.
[34] SCHLENK RF,DOHNER K,KNEBA M,et al.Gene mutations and response to treatment with all-trans retinoic acid in elderly patients with acute myeloid leukemia.Results from the AMLSG Trial AML HD98B[J].Haematologica,2009,94(1):54-60.
[35] EL HAJJ H,DASSOUKI Z,BERTHIER C,et al.Retinoic acid and arsenic trioxide trigger degradation of mutated NPM-1 resulting in apoptosis of AML cells[J].Blood,2015,125(22):3447-3455.
[36] LAMBERT J,LAMBERT J,NIBOUREL O,et al.MRD assessed by WT1 and NPM1 transcript levels identifies distinct outcomes in AML patients and is influenced by gemtuzumab ozogamicin[J].Oncotarget,2014,5(15):6280-6288.
[37] OLOMBEL G,GUERIN E,GUY J,et al.Letter to the editor:The level of blast CD33 expressin positively impacts the effect of gemtuzumab ozogamicin in patients with acute myeloid leukemia[J].Blood,2016,127(17):2155-2157.
[38]FALINI B,BRUNETTI L,MARTELLI MP.Letter to the editor:Dactinomycin in NPM1-mutated acute myeloid leukemia[J].N Engl J Med,2015,372(12):1180-1182.
[39]METZELER KH,HEROLD T,ROTHENBERG-THURLEY M,et al.Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia[J].Blood,2016,128(5):686-698.
[40]DUNLAP JB,LEONARD J,ROSENBERG M,et al.The combination of NPM1,DNMT3A,and IDH1/2mutations leads to inferior overall survival in AML[J].Am J Hematol,2019,94(8):913-920.
[41]KRONKE J,BULLINGER L,TELEANU V,et al.Clonal evolution in relapsed NPM1-mutated acute myeloid leukemia[J].Blood,2013,122(1):100-108.
[42] COCCIARDI S,DOLNIK A,KAPP-SCHWOERER S,et al.Clonal evolution patterns in acute myeloid leukemia with NPM1 mutation[J].Nature Communications,2019,10(1):1136-1152.
[43]HUANG G,SHIGESADA K,ITO K,et al.Dimerization with PEBP2beta protects RUNX1/AML1 from ubiquitin-proteasomemediated degradation[J].EMBO J,2001,20(4):723-733.
[44] DE BRUIJN M,DZIERZAK E.Runx transcription factors in the development and function of the defifinitive hematopoietic system[J].Blood,2017,129(15):2061-2069.
[45]SCHNITTGER S,DICKER F,KERN W,et al.RUNX1 mutations are frequent in de novo AML with noncomplex karyotype and confer an unfavorable prognosis[J].Blood,2011,117(8):2348-2357.
[46]MORITA K,SUZUKI K,MAEDA S,et al.Genetic regulation of the RUNX transcription factor family has antitumor effects[J].J Clin Invest,2017,127(7):2815-2828.
[47] FU L,FU H,TIAN L,et al.High expression of RUNX1 is associated with poorer outcomes in cytogenetically normal acute myeloid leukemia[J].Oncotarget,2016,7(13):15828-15839.
[48] BEHRENS K,MAUL K,TEKIN N,et al.RUNX1 cooperates with FLT3-ITD to induce leukemia[J].J Exp Med,2017,214(3):737-752.
[49]YOU E,CHO YU,JANG S,et al.Frequency and clinicopathologic features of RUNX1 mutations in patients with acute myeloid leukemia not otherwise specified[J].Am J Clin Pathol,2017,148(1):64-72.
[50] GAIDZIK VI,TELEANU V,PAPAEMMANUIL E,et al.RUNX1 mutations in acute myeloid leukemia are associated with distinct clinico-pathologic and genetic features[J].Leukemia,2016,30(11):2160-2168.
[51]SHIN SY,LEE ST,KIM HJ,et al.Mutation profiling of 19 candidate genes in acute myeloid leukemia suggests significance of DNMT3A mutations[J].Oncotarget,2016,7(34):54825-54837.
[52]YANG L,HAN Y,SUAREZ SAIZ F,et al.A tumor suppressor and oncogene:The WT1 story[J].Leukemia,2007,21(5):868-876.
[53] LI H,XING C,ZHOU B,et al.A regulatory circuitry between mir-193a/mir-600 and WT1 enhances leukemogenesis in acute myeloid leukemia[J].Exp Hematol,2018,61:59-68.
[54]TOOGEH G,RAMZI M,FARANOUSH M,et al.Prevalence and prognostic impact of Wilms' tumor 1 (WT1) gene,including SNP rs16754 in cytogenetically normal acute myeloblastic leukemia (CN-AML):An Iranian experience[J].Clin Lymphoma Myeloma Leuk,2016,16(3):e21-e26.
[55] HAO Y,CHENG Y,WU Q,et al.Combined usage of Wilms' tumor gene quantitative analysis and multipa-rameter flow cytometry for minimal residual disease monitoring of acute myeloid leukemia patients after allogeneic hematopoietic stem cells transplantation[J].Exp Ther Med,2018,15(2):1403-1409.
[56]AYATOLLAHI H,SADEGHIAN MH,NADERI M,et al.Quantitative assessment of wilms tumor 1 expression by real-time quantitative polymerase chain reaction in patients with acute myeloblastic leukemia[J].J Res Med Sci,2017,22(1):54.
[57]LIU H,WANG X,ZHANG H,et.al.Dynamic changes in the level of WT1 as an MRD marker to predict the therapeutic outcome of patients with AML with and without allogeneic stem cell transplantation[J].Mol Med Rep,2019,20(3):2426-2432.
[58]ZHU HH,JIANG H,JIANG B,et al.Cytarabine,aclarubicin and granulocyte colony-stimulating factor regimen represents an effective and safe salvage regimen for patients with acute myeloid leukemia refractory to first course of induction chemotherapy[J].Leuk Lymphoma,2013,54(11):2452-2457.
[59]REZVANI K,YONG AS,MIELKE S,et al.Leukemia-associated antigen specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies[J].Blood,2008,111(1):236-242.
[60] GRAY JX,MCMILLEN L,MOLLEE P,et al.WT1 expression as a marker of minimal residual disease predicts outcome in acute myeloid leukemia when measured post-consolidation[J].Leuk Res,2012,36(4):453-458.
[61] MASHIMA K,OH L,IKEDA T,et al.Role of sequential monitoring of WT1 gene expression in patients with acute myeloid leukemia for the early detection of leukemia relapse[J].Clin Lymphoma Myeloma Leuk,2018,18(12):e521-e527.
[62]FRAIRIA C,AYDIN S,AUDISIO E,et al.Post-remissional and pretran-splant role of minimal residual disease detected by WT1 in acute myeloid leukemia:a retro-spective cohort study[J].Leuk Res,2017,61(2017):10-17.
[63] DULERY R,NIBOUREL O,GAUTHIER J,et al.Impact of Wilms' tumor 1 expression on outcome of patients undergoing allogeneic stem cell transplantation for AML[J].Bone Marrow Transplant,2017,52(4):539-543.
[64] CANDONI A,DE MARCHI F,ZANNIER ME,et al.High prognostic value of pre-allogeneic stem cell transplantation minimal residual disease detection by WT1 gene expression in aMl transplanted in cytologic complete remission[J].Leuk Res,2017,63:22-27.

Memo

Memo:
-
Last Update: 1900-01-01