1,466
Views
0
CrossRef citations to date
0
Altmetric
Articles

Clinical values of gene alterations as marker of minimal residual disease in non-M3 acute myeloid leukemia

, &

References

  • Schuurhuis GJ, et al. Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD working party. Blood. 2018;131(12):1275–1291.
  • Gomez-Arteaga A, Guzman ML. Minimal residual disease in acute myeloid leukemia. Adv Exp Med Biol. 2018;1100:111–125.
  • Moors I, et al. Clinical implications of measurable residual disease in AML: review of current evidence. Crit Rev Oncol Hematol. 2019;133:142–148.
  • Ossenkoppele G, et al. Can we incorporate MRD assessment into clinical practice in AML? Best Pract Res Clin Haematol. 2019;32(2):186–191.
  • Feller N, et al. Defining consensus leukemia-associated immunophenotypes for detection of minimal residual disease in acute myeloid leukemia in a multicenter setting. Blood Cancer J. 2013;3(8):e129–e129.
  • Rossi G, et al. Leukemia-associated immunophenotypes subdivided in “categories of specificity” improve the sensitivity of minimal residual disease in predicting relapse in acute myeloid leukemia. Cytometry B Clin Cytom. 2020;98(3):216–225.
  • Cruz NM, et al. Minimal residual disease in acute myelogenous leukemia. Int J Lab Hematol. 2017;39(Suppl 1):53–60.
  • Ravandi F, Walter RB, Freeman SD. Evaluating measurable residual disease in acute myeloid leukemia. Blood Adv. 2018;2(11):1356–1366.
  • Luskin MR, Stone RM. Can minimal residual disease determination in acute myeloid leukemia Be used in clinical practice? J Oncol Pract. 2017;13(8):471–480.
  • Cilloni D, et al. Digital PCR in myeloid malignancies: ready to replace quantitative PCR? Int J Mol Sci. 2019;20(9):2249. DOI:https://doi.org/10.3390/ijms20092249
  • Waterhouse M, et al. Droplet digital PCR for the simultaneous analysis of minimal residual disease and hematopoietic chimerism after allogeneic cell transplantation. Clin Chem Lab Med. 2019;57(5):641–647.
  • Levine RL, Valk PJM. Next-generation sequencing in the diagnosis and minimal residual disease assessment of acute myeloid leukemia. Haematologica. 2019;104(5):868–871.
  • Behjati S, Tarpey PS. What is next generation sequencing? Arch Dis Child Educ Pract Ed. 2013;98(6):236–238.
  • Voso MT, et al. MRD in AML: the role of new techniques. Front Oncol. 2019;9:655. DOI:https://doi.org/10.3389/fonc.2019.00655
  • Flach J, et al. Clinical potential of introducing next-generation sequencing in patients at relapse of acute myeloid leukemia. Hematol Oncol. 2020;38(4):425–431.
  • Yang F, Anekpuritanang T, Press RD. Clinical utility of next-generation sequencing in acute myeloid leukemia. Mol Diagn Ther. 2020;24(1):1–13.
  • Valk PJM. Molecular measurable residual disease detection in AML. HemaSphere. 2020;4:S2: 115-117. Educational Updates in Hematology Book, doi:https://doi.org/10.1097/HS9.0000000000000444.
  • Pourrajab F, et al. Genetic characterization and risk stratification of acute myeloid leukemia. Cancer Manag Res. 2020;12:2231–2253.
  • Chiaretti S, et al. Genomic characterization of acute leukemias. Med Princ Pract. 2014;23(6):487–506.
  • Abdelhamid E, et al. Minimal residual disease monitoring based on FLT3 internal tandem duplication in adult acute myeloid leukemia. Leuk Res. 2012;36(3):316–323.
  • Perl AE, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N Engl J Med. 2019;381(18):1728–1740.
  • Chou WC, et al. Sensitive measurement of quantity dynamics of FLT3 internal tandem duplication at early time points provides prognostic information. Ann Oncol. 2011;22(3):696–704.
  • Levis, M., FLT3 mutations in acute myeloid leukemia: what is the best approach in 2013? Hematol Am Soc Hematol Educ Program, 2013. 2013: p. 220–226.
  • Gaballa S, et al. Relapse risk and survival in patients with FLT3 mutated acute myeloid leukemia undergoing stem cell transplantation. Am J Hematol. 2017;92(4):331–337.
  • Candoni A, et al. Predictive value of pretransplantation molecular minimal residual disease assessment by WT1 gene expression in FLT3-positive acute myeloid leukemia. Exp Hematol. 2017;49:25–33.
  • Grunwald MR, et al. Improved FLT3 internal tandem duplication PCR assay predicts outcome after allogeneic transplant for acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20(12):1989–1995.
  • Palmisano M, et al. NPM1 mutations are more stable than FLT3 mutations during the course of disease in patients with acute myeloid leukemia. Haematologica. 2007;92(9):1268–1269.
  • Levis M, et al. FLT3 inhibitors added to induction therapy induce deeper remissions. Blood. 2020;135(1):75–78.
  • Ambinder AJ, Levis M. Potential targeting of FLT3 acute myeloid leukemia. Haematologica. 2021;106(3):671–681.
  • Levis MJ, et al. A next-generation sequencing-based assay for minimal residual disease assessment in AML patients with FLT3-ITD mutations. Blood Adv. 2018;2(8):825–831.
  • Gebru MT, et al. Glucocorticoids enhance the antileukemic activity of FLT3 inhibitors in FLT3-mutant acute myeloid leukemia. Blood. 2020;136(9):1067–1079.
  • Alnagar AA, et al. Outcome of core binding factor acute myeloid leukemia by receptor tyrosine kinase mutation. Clin Lymphoma Myeloma Leuk. 2020;20(7):459–467.
  • Burchert A, et al. Sorafenib maintenance after allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia with FLT3-internal tandem duplication mutation (SORMAIN). J Clin Oncol. 2020;38(26):2993–3002.
  • Schlenk RF, et al. Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood. 2014;124(23):3441–3449.
  • Helbig G, et al. Pre-transplant FLT3/ITD status predicts outcome in FLT3-mutated acute myeloid leukemia following allogeneic stem cell transplantation. Ann Hematol. 2020;99(8):1845–1853.
  • Balagopal V, et al. Measurable residual disease monitoring for patients with acute myeloid leukemia following hematopoietic cell transplantation using error corrected hybrid capture next generation sequencing. PLoS One. 2019;14(10):e0224097. DOI:https://doi.org/10.1371/journal.pone.0224097
  • Rücker FG, et al. Measurable residual disease monitoring in acute myeloid leukemia with t(8;21)(q22;q22.1): results from the AML study group. Blood. 2019;134(19):1608–1618.
  • Ishikawa Y, et al. Prospective evaluation of prognostic impact of KIT mutations on acute myeloid leukemia with RUNX1-RUNX1T1 and CBFB-MYH11. Blood Adv. 2020;4(1):66–75.
  • Marková J, et al. Monitoring of minimal residual disease in patients with core binding factor acute myeloid leukemia and the impact of C-KIT, FLT3, and JAK2 mutations on clinical outcome. Leuk Lymphoma. 2009;50(9):1448–1460.
  • Liu X, et al. RAS mutations in acute myeloid leukaemia patients: a review and meta-analysis. Clin Chim Acta. 2019;489:254–260.
  • Casey G, et al. N-ras mutation in acute myeloid leukemia: incidence, prognostic significance and value as a marker of minimal residual disease. Pathology. 1993;25(1):57–62.
  • Ishida H, et al. Panel-based next-generation sequencing facilitates the characterization of childhood acute myeloid leukemia in clinical settings. Biomed Rep. 2020;13(5):1–1.
  • Thol F, et al. Measurable residual disease monitoring by NGS before allogeneic hematopoietic cell transplantation in AML. Blood. 2018;132(16):1703–1713.
  • Puckrin R, et al. Measurable residual disease monitoring provides insufficient lead-time to prevent morphologic relapse in the majority of patients with core-binding factor acute myeloid leukemia. Haematologica. 2020;106(1):56–63.
  • Duployez N, et al. Prognosis and monitoring of core-binding factor acute myeloid leukemia: current and emerging factors. Expert Rev Hematol. 2015;8(1):43–56.
  • Perea G, et al. Prognostic value of minimal residual disease (MRD) in acute myeloid leukemia (AML) with favorable cytogenetics [t(8;21) and inv(16)]. Leukemia. 2006;20(1):87–94.
  • Wang L, et al. High prognostic value of minimal residual disease detected by flow-cytometry-enhanced fluorescence in situ hybridization in core-binding factor acute myeloid leukemia (CBF-AML). Ann Hematol. 2014;93(10):1685–1694.
  • Ouyang J, et al. Comparison of multiparameter flow cytometry immunophenotypic analysis and quantitative RT-PCR for the detection of minimal residual disease of core binding factor acute myeloid leukemia. Am J Clin Pathol. 2016;145(6):769–777.
  • Yin JA, et al. Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: results of the United Kingdom MRC AML-15 trial. Blood. 2012;120(14):2826–2835.
  • Pigazzi M, et al. Minimal residual disease monitored after induction therapy by RQ-PCR can contribute to tailor treatment of patients with t(8;21) RUNX1-RUNX1T1 rearrangement. Haematologica. 2015;100(3):e99–e101.
  • Willekens C, et al. Prospective long-term minimal residual disease monitoring using RQ-PCR in RUNX1-RUNX1T1-positive acute myeloid leukemia: results of the French CBF-2006 trial. Haematologica. 2016;101(3):328–335.
  • Zhu HH, et al. MRD-directed risk stratification treatment may improve outcomes of t(8;21) AML in the first complete remission: results from the AML05 multicenter trial. Blood. 2013;121(20):4056–4062.
  • Qin YZ, et al. The dynamics of RUNX1-RUNX1T1 transcript levels after allogeneic hematopoietic stem cell transplantation predict relapse in patients with t(8;21) acute myeloid leukemia. J Hematol Oncol. 2017;10(1):44. DOI:https://doi.org/10.1186/s13045-017-0414-2
  • Tang FF, et al. Monitoring of post-transplant CBFB-MYH11 as minimal residual disease, rather than KIT mutations, can predict relapse after allogeneic haematopoietic cell transplantation in adults with inv(16) acute myeloid leukaemia. Br J Haematol. 2018;180(3):448–451.
  • Prebet T, et al. Anthracycline dose intensification improves molecular response and outcome of patients treated for core binding factor acute myeloid leukemia. Haematologica. 2014;99(10):e185–e187.
  • Ragon BK, et al. Minimal residual disease eradication with epigenetic therapy in core binding factor acute myeloid leukemia. Am J Hematol. 2017;92(9):845–850.
  • Mo XD, et al. Interferon-α Is effective for treatment of minimal residual disease in patients with t(8;21) acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation: results of a Prospective registry study. Oncologist. 2018;23(11):1349–1357.
  • Corbacioglu A, et al. Prognostic impact of minimal residual disease in CBFB-MYH11-positive acute myeloid leukemia. J Clin Oncol. 2010;28(23):3724–3729.
  • Qin YZ, et al. Allogeneic stem cell transplant may improve the outcome of adult patients with inv(16) acute myeloid leukemia in first complete remission with poor molecular responses to chemotherapy. Leuk Lymphoma. 2015;56(11):3116–3123.
  • Guièze R, et al. Prognostic value of minimal residual disease by real-time quantitative PCR in acute myeloid leukemia with CBFB-MYH11 rearrangement: the French experience. Leukemia. 2010;24(7):1386–1388.
  • Lane S, et al. A >  or=1 log rise in RQ-PCR transcript levels defines molecular relapse in core binding factor acute myeloid leukemia and predicts subsequent morphologic relapse. Leuk Lymphoma. 2008;49(3):517–523.
  • Xiaosu Z, et al. Classifying AML patients with inv(16) into high-risk and low-risk relapsed patients based on peritransplantation minimal residual disease determined by CBFβ/MYH11 gene expression. Ann Hematol. 2019;98(1):73–81.
  • Yalniz FF, et al. Significance of minimal residual disease monitoring by real-time quantitative polymerase chain reaction in core binding factor acute myeloid leukemia for transplantation outcomes. Cancer. 2020;126(10):2183–2192.
  • Gao MG, et al. The predictive value of minimal residual disease when facing the inconsistent results detected by real-time quantitative PCR and flow cytometry in NPM1-mutated acute myeloid leukemia. Ann Hematol. 2020;99(1):73–82.
  • Zhou Y, et al. Pattern associated leukemia immunophenotypes and measurable disease detection in acute myeloid leukemia or myelodysplastic syndrome with mutated NPM1. Cytometry B Clin Cytom. 2019;96(1):67–72.
  • Krönke J, et al. Monitoring of minimal residual disease in NPM1-mutated acute myeloid leukemia: a study from the German-Austrian acute myeloid leukemia study group. J Clin Oncol. 2011;29(19):2709–2716.
  • Heiblig M, et al. Prognostic value of genetic alterations in elderly patients with acute myeloid leukemia: a single institution experience. Cancers (Basel). 2019;11(4):570. DOI:https://doi.org/10.3390/cancers11040570
  • Balsat M, et al. Postinduction minimal residual disease predicts outcome and benefit from allogeneic stem cell transplantation in acute myeloid leukemia with NPM1 mutation: a study by the Acute Leukemia French Association group. J Clin Oncol. 2017;35(2):185–193.
  • Hubmann M, et al. Molecular response assessment by quantitative real-time polymerase chain reaction after induction therapy in NPM1-mutated patients identifies those at high risk of relapse. Haematologica. 2014;99(8):1317–1325.
  • Patkar N, et al. Clinical impact of measurable residual disease monitoring by ultradeep next generation sequencing in NPM1 mutated acute myeloid leukemia. Oncotarget. 2018;9(93):36613–36624.
  • Jo SY, et al. Correlation of NPM1 type A mutation burden with clinical status and outcomes in acute myeloid leukemia patients With mutated NPM1 type A. Ann Lab Med. 2016;36(5):399–404.
  • Lussana F, et al. Molecular detection of minimal residual disease before allogeneic stem cell transplantation predicts a high incidence of early relapse in adult patients with NPM1 positive acute myeloid leukemia. Cancers (Basel). 2019;11(10):1455. DOI:https://doi.org/10.3390/cancers11101455
  • Schnittger S, et al. Minimal residual disease levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML. Blood. 2009;114(11):2220–2231.
  • Shayegi N, et al. The level of residual disease based on mutant NPM1 is an independent prognostic factor for relapse and survival in AML. Blood. 2013;122(1):83–92.
  • Bill M, et al. Digital droplet PCR-based absolute quantification of pre-transplant NPM1 mutation burden predicts relapse in acute myeloid leukemia patients. Ann Hematol. 2018;97(10):1757–1765.
  • Malmberg D, et al E. Minimal residual disease assessed with deep sequencing of NPM1 mutations predicts relapse after allogeneic stem cell transplant in AML. Leuk Lymphoma. 2019;60(2):409–417.
  • Kayser S, et al. Pretransplant NPM1 MRD levels predict outcome after allogeneic hematopoietic stem cell transplantation in patients with acute myeloid leukemia. Blood Cancer J. 2016;6(7):e449–e449.
  • Linch DC, et al. Analysis of the clinical impact of NPM1 mutant allele burden in a large cohort of younger adult patients with acute myeloid leukaemia. Br J Haematol. 2020;188(6):852–859.
  • Prata PH, et al. NPM1 mutation is not associated with prolonged complete remission in acute myeloid leukemia patients treated with hypomethylating agents. Haematologica. 2018;103(10):e455–e457.
  • Lambert J, et al. MRD assessed by WT1 and NPM1 transcript levels identifies distinct outcomes in AML patients and is influenced by gemtuzumab ozogamicin. Oncotarget. 2014;5(15):6280–6288.
  • Marjanovic I, et al. Use of Wilms Tumor 1 gene expression as a reliable marker for prognosis and minimal residual disease monitoring in acute myeloid leukemia With normal karyotype patients. Clin Lymphoma Myeloma Leuk. 2017;17(5):312–319.
  • Hidaka D, et al. Wilms tumor 1 expression at diagnosis correlates with genetic abnormalities and polymorphism but is not independently prognostic in acute myelogenous leukemia: a hokkaido leukemia net study. Clin Lymphoma Myeloma Leuk. 2018;18(11):e469–e479.
  • Šálek C, et al. WT1 expression in peripheral blood at diagnosis and during the course of early consolidation treatment correlates with survival in patients with intermediate and poor-risk acute myeloid leukemia. Clin Lymphoma Myeloma Leuk. 2020;20(12):e998–e1009.
  • Cho BS, et al. WT1 measurable residual disease assay in patients with acute myeloid leukemia who underwent allogeneic hematopoietic stem cell transplantation: optimal time points, thresholds, and candidates. Biol Blood Marrow Transplant. 2019;25(10):1925–1932.
  • Xu J, et al. Clinical features and prognosis of normal karyotype acute myeloid leukemia pediatric patients with WT1 mutations: an analysis based on TCGA database. Hematology. 2020;25(1):79–84.
  • Marani C, et al. Integrating post induction WT1 quantification and flow-cytometry results improves minimal residual disease stratification in acute myeloid leukemia. Leuk Res. 2013;37(12):1606–1611.
  • Rautenberg C, et al. Wilm's tumor 1-guided preemptive treatment with hypomethylating agents for molecular relapse of AML and MDS after allogeneic transplantation. Bone Marrow Transplant. 2021;56(2):442–450.
  • Ido K, et al. The proportional relationship between pretransplant WT1 mRNA levels and risk of mortality after allogeneic hematopoietic cell transplantation in acute myeloid leukemia not in remission. Transplantation. 2019;103(10):2201–2210.
  • Andersson C, et al. Reduction in WT1 gene expression during early treatment predicts the outcome in patients with acute myeloid leukemia. Diagn Mol Pathol. 2012;21(4):225–233.
  • Mossallam GI, Abdel Hamid TM, Mahmoud HK. Prognostic significance of WT1 expression at diagnosis and end of induction in Egyptian adult acute myeloid leukemia patients. Hematology. 2013;18(2):69–73.
  • Cho BS, et al. Haploidentical vs matched unrelated donor transplantation for acute myeloid leukemia in remission: a prospective comparative study. Am J Hematol. 2021;96(1):98–109.
  • Park S, et al. Comparison of myeloablative (CyTBI, BuCy) versus reduced-Intensity (FluBu2TBI400) peripheral blood stem cell transplantation in acute myeloid leukemia patients with pretransplant low WT1 expression. Biol Blood Marrow Transplant. 2020;26(11):2018–2026.
  • Yu J, et al. Clinical implications of recurrent gene mutations in acute myeloid leukemia. Exp Hematol Oncol. 2020;9:4. DOI:https://doi.org/10.1186/s40164-020-00161-7
  • DiNardo C, Lachowiez C. Acute myeloid leukemia: from mutation profiling to treatment decisions. Curr Hematol Malig Rep. 2019;14(5):386–394.
  • Yu G, et al. Gene mutation profile and risk stratification in AML1–ETO–positive acute myeloid leukemia based on next–generation sequencing. Oncol Rep. 2019;42(6):2333–2344.
  • Yan B, et al. Low-frequency TP53 hotspot mutation contributes to chemoresistance through clonal expansion in acute myeloid leukemia. Leukemia. 2020;34(7):1816–1827.
  • Chan O, et al. Impact of TP53 gene mutation clearance and conditioning Intensity on outcome in MDS or AML patients prior to allogeneic stem cell transplantation. Blood. 2019;134(Supplement_1):149–149.
  • Wu X, et al. Prognostic significance of the EVI1 gene expression in patients with acute myeloid leukemia: a meta-analysis. Ann Hematol. 2019;98(11):2485–2496.
  • Marjanovic I, et al. Expression pattern and prognostic significance of EVI1 gene in adult acute myeloid leukemia patients with normal karyotype. Indian J Hematol Blood Transfus. 2020;36(2):292–299.
  • Paubelle E, et al. Efficacy of all-trans-retinoic acid in high-risk acute myeloid leukemia with overexpression of EVI1. Oncol Ther. 2019;7(2):121–130.
  • Ding L, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481(7382):506–510.
  • Gröschel S, et al. High EVI1 expression predicts outcome in younger adult patients with acute myeloid leukemia and is associated with distinct cytogenetic abnormalities. J Clin Oncol. 2010;28(12):2101–2107.
  • Weisser M, et al. Feasibility of using the combined MDS-EVI1/EVI1 gene expression as an alternative molecular marker in acute myeloid leukemia: a report of four cases. Cancer Genet Cytogenet. 2007;177(1):64–69.
  • Wieser R. The oncogene and developmental regulator EVI1: expression, biochemical properties, and biological functions. Gene. 2007;396(2):346–357.
  • Aytekin M, et al. Regulation of the expression of the oncogene EVI1 through the use of alternative mRNA 5'-ends. Gene. 2005;356:160–168.
  • Smol T, et al. Quantification of EVI1 transcript levels in acute myeloid leukemia by RT-qPCR analysis: a study by the ALFA group. Leuk Res. 2015;39(12):1443–1447.
  • Petrova L, et al. IDH1 and IDH2 mutations in patients with acute myeloid leukemia: suitable targets for minimal residual disease monitoring? Clin Biochem. 2018;61:34–39.
  • Grassi S, et al. Digital droplet PCR is a specific and sensitive tool for detecting IDH2 mutations in acute myeloid LeuKemia patients. Cancers (Basel). 2020;12(7):1738. DOI:https://doi.org/10.3390/cancers12071738
  • Jeziskova I, et al. Quantitative detection of IDH2 mutation for minimal residual disease monitoring in patients with acute myeloid leukemia and its comparison with mutations in NPM1 gene. Leuk Lymphoma. 2013;54(4):867–870.
  • Debarri H, et al. IDH1/2 but not DNMT3A mutations are suitable targets for minimal residual disease monitoring in acute myeloid leukemia patients: a study by the acute Leukemia French association. Oncotarget. 2015;6(39):42345–42353.
  • Petiti J, et al. Highly sensitive detection of IDH2 mutations in acute myeloid leukemia. J Clin Med. 2020;9(1):271. DOI:https://doi.org/10.3390/jcm9010271
  • Abdelhamid E, et al. Minimal residual disease assessment of IDH1/2 mutations in acute myeloid leukemia by LNA-RQ-PCR. Tunis Med. 2016;94(3):190–197.
  • Stein EM, et al. Molecular remission and response patterns in patients with mutant-IDH2 acute myeloid leukemia treated with enasidenib. Blood. 2019;133(7):676–687.
  • Roboz GJ, et al. Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia. Blood. 2020;135(7):463–471.
  • Ok CY, et al. Persistent IDH1/2 mutations in remission can predict relapse in patients with acute myeloid leukemia. Haematologica. 2019;104(2):305–311.
  • Jongen-Lavrencic M, et al. Molecular minimal residual disease in acute myeloid leukemia. N Engl J Med. 2018;378(13):1189–1199.
  • Genovese G, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014;371(26):2477–2487.
  • Getta BM, et al. Multicolor flow cytometry and multigene next-generation sequencing Are complementary and Highly predictive for relapse in acute myeloid leukemia after allogeneic transplantation. Biol Blood Marrow Transplant. 2017;23(7):1064–1071.
  • Hirsch P, et al. Genetic hierarchy and temporal variegation in the clonal history of acute myeloid leukaemia. Nat Commun. 2016;7:12475. DOI:https://doi.org/10.1038/ncomms12475
  • Brambati C, et al. Droplet digital polymerase chain reaction for DNMT3A and IDH1/2 mutations to improve early detection of acute myeloid leukemia relapse after allogeneic hematopoietic stem cell transplantation. Haematologica. 2016;101(4):e157–e161.
  • Jeziskova I, et al. Distribution of mutations in DNMT3A gene and the suitability of mutations in R882 codon for MRD monitoring in patients with AML. Int J Hematol. 2015;102(5):553–557.
  • Morita K, et al. Clearance of somatic mutations at remission and the risk of relapse in acute myeloid leukemia. J Clin Oncol. 2018;36(18):1788–1797.
  • Burillo-Sanz S, et al. MLL-rearranged acute myeloid leukemia: influence of the genetic partner in allo-HSCT response and prognostic factor of MLL 3’ region mRNA expression. Eur J Haematol. 2018;100(5):436–443.
  • Burmeister T, et al. Evidence-based RT-PCR methods for the detection of the 8 most common MLL aberrations in acute leukemias. Leuk Res. 2015;39(2):242–247.
  • Liu J, et al. Monitoring mixed lineage leukemia expression may help identify patients with mixed lineage leukemia–rearranged acute leukemia who are at high risk of relapse after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2014;20(7):929–936.
  • Huang S, et al. Prognostic significance of mixed-lineage leukemia (MLL) gene detected by real-time fluorescence quantitative PCR assay in acute myeloid leukemia. Med Sci Monit. 2016;22:3009–3017.
  • Abildgaard L, et al. A novel RT-qPCR assay for quantification of the MLL-MLLT3 fusion transcript in acute myeloid leukaemia. Eur J Haematol. 2013;91(5):394–398.
  • Huang S, et al. [Prognostic significance of detecting MLL-AF9 fusion gene expression in patients with acute myeloid leukemia by real-time fluorescence quantitative PCR]. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2013;21(6):1435–1440.
  • Ommen HB, et al. Relapse kinetics in acute myeloid leukaemias with MLL translocations or partial tandem duplications within the MLL gene. Br J Haematol. 2014;165(5):618–628.