Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2015

Open Access 01-12-2015 | Research

High expression of myocyte enhancer factor 2C (MEF2C) is associated with adverse-risk features and poor outcome in pediatric acute myeloid leukemia: a report from the Children’s Oncology Group

Authors: George S. Laszlo, Todd A. Alonzo, Chelsea J. Gudgeon, Kimberly H. Harrington, Alex Kentsis, Robert B. Gerbing, Yi-Cheng Wang, Rhonda E. Ries, Susana C. Raimondi, Betsy A. Hirsch, Alan S. Gamis, Soheil Meshinchi, Roland B. Walter

Published in: Journal of Hematology & Oncology | Issue 1/2015

Login to get access

Abstract

Background

Recent studies have identified myocyte enhancer factor 2C (MEF2C) as cooperating oncogene in acute myeloid leukemia (AML) and suggested a contribution to the aggressive nature of at least some subtypes of AML, raising the possibility that MEF2C could serve as marker of poor-risk AML and, therefore, have prognostic significance.

Methods

To test this hypothesis, we retrospectively quantified MEF2C expression in pretreatment bone marrow specimens in participants of the AAML0531 trial by reverse-transcriptase polymerase chain reaction and correlated expression levels with disease characteristics and clinical outcome.

Results

In all 751 available patient specimens, MEF2C messenger RNA (mRNA) was detectable and varied >3000-fold relative to β-glucuronidase. Patients with the highest relative MEF2C expression (4th quartile) less likely achieved a complete remission after one course of chemotherapy than the other patients (67 vs. 78 %, P = 0.005). They also had an inferior overall survival (P = 0.014; at 5 years 55 ± 8 vs. 67 ± 4 %), inferior event-free survival (P < 0.001; at 5 years 38 ± 7 vs. 54 ± 4 %), and higher relapse risk than patients within the lower 3 quartiles of MEF2C expression (P < 0.001; at 5 years 53 ± 9 vs. 35 ± 5 %). These differences were accounted for by lower prevalence of cytogenetically/molecularly defined low-risk disease (16 vs. 46 %, P < 0.001) and higher prevalence of standard-risk disease (68 vs. 42 %, P < 0.001) in patients with high MEF2C expression, suggesting that MEF2C cooperates with additional pathogenic abnormalities.

Conclusions

High MEF2C expression identifies a subset of AML patients with adverse-risk disease features and poor outcome. With confirmation that high MEF2C mRNA expression leads to overexpression of MEF2C protein, these findings provide the rationale for therapeutic targeting of MEF2C transcriptional activation in AML.
Literature
1.
go back to reference Black BL, Olson EN. Transcriptional control of muscle development by myocyte enhancer factor-2 (MEF2) proteins. Annu Rev Cell Dev Biol. 1998;14:167–96.CrossRefPubMed Black BL, Olson EN. Transcriptional control of muscle development by myocyte enhancer factor-2 (MEF2) proteins. Annu Rev Cell Dev Biol. 1998;14:167–96.CrossRefPubMed
2.
go back to reference Lin Q, Schwarz J, Bucana C, Olson EN. Control of mouse cardiac morphogenesis and myogenesis by transcription factor MEF2C. Science. 1997;276(5317):1404–7.CrossRefPubMedPubMedCentral Lin Q, Schwarz J, Bucana C, Olson EN. Control of mouse cardiac morphogenesis and myogenesis by transcription factor MEF2C. Science. 1997;276(5317):1404–7.CrossRefPubMedPubMedCentral
3.
go back to reference Canté-Barrett K, Pieters R, Meijerink JP. Myocyte enhancer factor 2C in hematopoiesis and leukemia. Oncogene. 2014;33(4):403–10.CrossRefPubMed Canté-Barrett K, Pieters R, Meijerink JP. Myocyte enhancer factor 2C in hematopoiesis and leukemia. Oncogene. 2014;33(4):403–10.CrossRefPubMed
4.
go back to reference Schüler A, Schwieger M, Engelmann A, Weber K, Horn S, Müller U, et al. The MADS transcription factor Mef2c is a pivotal modulator of myeloid cell fate. Blood. 2008;111(9):4532–41.CrossRefPubMed Schüler A, Schwieger M, Engelmann A, Weber K, Horn S, Müller U, et al. The MADS transcription factor Mef2c is a pivotal modulator of myeloid cell fate. Blood. 2008;111(9):4532–41.CrossRefPubMed
5.
go back to reference Stehling-Sun S, Dade J, Nutt SL, DeKoter RP, Camargo FD. Regulation of lymphoid versus myeloid fate ‘choice’ by the transcription factor Mef2c. Nat Immunol. 2009;10(3):289–96.CrossRefPubMed Stehling-Sun S, Dade J, Nutt SL, DeKoter RP, Camargo FD. Regulation of lymphoid versus myeloid fate ‘choice’ by the transcription factor Mef2c. Nat Immunol. 2009;10(3):289–96.CrossRefPubMed
6.
go back to reference Zheng R, Wang X, Studzinski GP. 1,25-Dihydroxyvitamin D3 induces monocytic differentiation of human myeloid leukemia cells by regulating C/EBPbeta expression through MEF2C. J Steroid Biochem Mol Biol. 2015;148:132–7.CrossRefPubMed Zheng R, Wang X, Studzinski GP. 1,25-Dihydroxyvitamin D3 induces monocytic differentiation of human myeloid leukemia cells by regulating C/EBPbeta expression through MEF2C. J Steroid Biochem Mol Biol. 2015;148:132–7.CrossRefPubMed
7.
go back to reference Nagel S, Meyer C, Quentmeier H, Kaufmann M, Drexler HG, MacLeod RA. MEF2C is activated by multiple mechanisms in a subset of T-acute lymphoblastic leukemia cell lines. Leukemia. 2008;22(3):600–7.CrossRefPubMed Nagel S, Meyer C, Quentmeier H, Kaufmann M, Drexler HG, MacLeod RA. MEF2C is activated by multiple mechanisms in a subset of T-acute lymphoblastic leukemia cell lines. Leukemia. 2008;22(3):600–7.CrossRefPubMed
8.
go back to reference Nagel S, Venturini L, Meyer C, Kaufmann M, Scherr M, Drexler HG, et al. Transcriptional deregulation of oncogenic myocyte enhancer factor 2C in T-cell acute lymphoblastic leukemia. Leuk Lymphoma. 2011;52(2):290–7.CrossRefPubMed Nagel S, Venturini L, Meyer C, Kaufmann M, Scherr M, Drexler HG, et al. Transcriptional deregulation of oncogenic myocyte enhancer factor 2C in T-cell acute lymphoblastic leukemia. Leuk Lymphoma. 2011;52(2):290–7.CrossRefPubMed
9.
go back to reference Homminga I, Pieters R, Langerak AW, de Rooi JJ, Stubbs A, Verstegen M, et al. Integrated transcript and genome analyses reveal NKX2-1 and MEF2C as potential oncogenes in T cell acute lymphoblastic leukemia. Cancer Cell. 2011;19(4):484–97.CrossRefPubMed Homminga I, Pieters R, Langerak AW, de Rooi JJ, Stubbs A, Verstegen M, et al. Integrated transcript and genome analyses reveal NKX2-1 and MEF2C as potential oncogenes in T cell acute lymphoblastic leukemia. Cancer Cell. 2011;19(4):484–97.CrossRefPubMed
10.
go back to reference Zuurbier L, Gutierrez A, Mullighan CG, Cante-Barrett K, Gevaert AO, de Rooi J, et al. Immature MEF2C-dysregulated T-cell leukemia patients have an early T-cell precursor acute lymphoblastic leukemia gene signature and typically have non-rearranged T-cell receptors. Haematologica. 2014;99(1):94–102.CrossRefPubMedPubMedCentral Zuurbier L, Gutierrez A, Mullighan CG, Cante-Barrett K, Gevaert AO, de Rooi J, et al. Immature MEF2C-dysregulated T-cell leukemia patients have an early T-cell precursor acute lymphoblastic leukemia gene signature and typically have non-rearranged T-cell receptors. Haematologica. 2014;99(1):94–102.CrossRefPubMedPubMedCentral
11.
go back to reference Schwieger M, Schüler A, Forster M, Engelmann A, Arnold MA, Delwel R, et al. Homing and invasiveness of MLL/ENL leukemic cells is regulated by MEF2C. Blood. 2009;114(12):2476–88.CrossRefPubMed Schwieger M, Schüler A, Forster M, Engelmann A, Arnold MA, Delwel R, et al. Homing and invasiveness of MLL/ENL leukemic cells is regulated by MEF2C. Blood. 2009;114(12):2476–88.CrossRefPubMed
12.
go back to reference Somervaille TCP, Cleary ML. Grist for the MLL: how do MLL oncogenic fusion proteins generate leukemia stem cells? Int J Hematol. 2010;91(5):735–41.CrossRefPubMed Somervaille TCP, Cleary ML. Grist for the MLL: how do MLL oncogenic fusion proteins generate leukemia stem cells? Int J Hematol. 2010;91(5):735–41.CrossRefPubMed
13.
go back to reference de Boer J, Walf-Vorderwulbecke V, Williams O. In focus: MLL-rearranged leukemia. Leukemia. 2013;27(6):1224–8.CrossRefPubMed de Boer J, Walf-Vorderwulbecke V, Williams O. In focus: MLL-rearranged leukemia. Leukemia. 2013;27(6):1224–8.CrossRefPubMed
14.
go back to reference Gole B, Wiesmüller L. Leukemogenic rearrangements at the mixed lineage leukemia gene (MLL)-multiple rather than a single mechanism. Front Cell Dev Biol. 2015;3:41.CrossRefPubMedPubMedCentral Gole B, Wiesmüller L. Leukemogenic rearrangements at the mixed lineage leukemia gene (MLL)-multiple rather than a single mechanism. Front Cell Dev Biol. 2015;3:41.CrossRefPubMedPubMedCentral
15.
16.
go back to reference Jin G, Yamazaki Y, Takuwa M, Takahara T, Kaneko K, Kuwata T, et al. Trib1 and Evi1 cooperate with Hoxa and Meis1 in myeloid leukemogenesis. Blood. 2007;109(9):3998–4005.CrossRefPubMed Jin G, Yamazaki Y, Takuwa M, Takahara T, Kaneko K, Kuwata T, et al. Trib1 and Evi1 cooperate with Hoxa and Meis1 in myeloid leukemogenesis. Blood. 2007;109(9):3998–4005.CrossRefPubMed
17.
go back to reference Yoshimi A, Goyama S, Watanabe-Okochi N, Yoshiki Y, Nannya Y, Nitta E, et al. Evi1 represses PTEN expression and activates PI3K/AKT/mTOR via interactions with polycomb proteins. Blood. 2011;117(13):3617–28.CrossRefPubMed Yoshimi A, Goyama S, Watanabe-Okochi N, Yoshiki Y, Nannya Y, Nitta E, et al. Evi1 represses PTEN expression and activates PI3K/AKT/mTOR via interactions with polycomb proteins. Blood. 2011;117(13):3617–28.CrossRefPubMed
18.
go back to reference Glass C, Wilson M, Gonzalez R, Zhang Y, Perkins AS. The role of EVI1 in myeloid malignancies. Blood Cells Mol Dis. 2014;53(1-2):67–76.CrossRefPubMed Glass C, Wilson M, Gonzalez R, Zhang Y, Perkins AS. The role of EVI1 in myeloid malignancies. Blood Cells Mol Dis. 2014;53(1-2):67–76.CrossRefPubMed
19.
go back to reference Bindels EM, Havermans M, Lugthart S, Erpelinck C, Wocjtowicz E, Krivtsov AV, et al. EVI1 is critical for the pathogenesis of a subset of MLL-AF9-rearranged AMLs. Blood. 2012;119(24):5838–49.CrossRefPubMedPubMedCentral Bindels EM, Havermans M, Lugthart S, Erpelinck C, Wocjtowicz E, Krivtsov AV, et al. EVI1 is critical for the pathogenesis of a subset of MLL-AF9-rearranged AMLs. Blood. 2012;119(24):5838–49.CrossRefPubMedPubMedCentral
20.
go back to reference Rommer A, Steinmetz B, Herbst F, Hackl H, Heffeter P, Heilos D, et al. EVI1 inhibits apoptosis induced by antileukemic drugs via upregulation of CDKN1A/p21/WAF in human myeloid cells. PLoS One. 2013;8(2):e56308.CrossRefPubMedPubMedCentral Rommer A, Steinmetz B, Herbst F, Hackl H, Heffeter P, Heilos D, et al. EVI1 inhibits apoptosis induced by antileukemic drugs via upregulation of CDKN1A/p21/WAF in human myeloid cells. PLoS One. 2013;8(2):e56308.CrossRefPubMedPubMedCentral
21.
go back to reference Heller G, Rommer A, Steinleitner K, Etzler J, Hackl H, Heffeter P, et al. EVI1 promotes tumor growth via transcriptional repression of MS4A3. J Hematol Oncol. 2015;8:28.CrossRefPubMedPubMedCentral Heller G, Rommer A, Steinleitner K, Etzler J, Hackl H, Heffeter P, et al. EVI1 promotes tumor growth via transcriptional repression of MS4A3. J Hematol Oncol. 2015;8:28.CrossRefPubMedPubMedCentral
22.
go back to reference Du Y, Spence SE, Jenkins NA, Copeland NG. Cooperating cancer-gene identification through oncogenic-retrovirus-induced insertional mutagenesis. Blood. 2005;106(7):2498–505.CrossRefPubMedPubMedCentral Du Y, Spence SE, Jenkins NA, Copeland NG. Cooperating cancer-gene identification through oncogenic-retrovirus-induced insertional mutagenesis. Blood. 2005;106(7):2498–505.CrossRefPubMedPubMedCentral
23.
go back to reference Krivtsov AV, Twomey D, Feng Z, Stubbs MC, Wang Y, Faber J, et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature. 2006;442(7104):818–22.CrossRefPubMed Krivtsov AV, Twomey D, Feng Z, Stubbs MC, Wang Y, Faber J, et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature. 2006;442(7104):818–22.CrossRefPubMed
24.
go back to reference Gamis AS, Alonzo TA, Meshinchi S, Sung L, Gerbing RB, Raimondi SC, et al. Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: results from the randomized phase III Children’s Oncology Group trial AAML0531. J Clin Oncol. 2014;32(27):3021–32.CrossRefPubMedPubMedCentral Gamis AS, Alonzo TA, Meshinchi S, Sung L, Gerbing RB, Raimondi SC, et al. Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: results from the randomized phase III Children’s Oncology Group trial AAML0531. J Clin Oncol. 2014;32(27):3021–32.CrossRefPubMedPubMedCentral
25.
go back to reference Vitali C, Bassani C, Chiodoni C, Fellini E, Guarnotta C, Miotti S, et al. SOCS2 controls proliferation and stemness of hematopoietic cells under stress conditions and its deregulation marks unfavorable acute leukemias. Cancer Res. 2015;75(11):2387–99.CrossRefPubMed Vitali C, Bassani C, Chiodoni C, Fellini E, Guarnotta C, Miotti S, et al. SOCS2 controls proliferation and stemness of hematopoietic cells under stress conditions and its deregulation marks unfavorable acute leukemias. Cancer Res. 2015;75(11):2387–99.CrossRefPubMed
26.
go back to reference Laszlo GS, Ries RE, Gudgeon CJ, Harrington KH, Alonzo TA, Gerbing RB, et al. High expression of suppressor of cytokine signaling-2 predicts poor outcome in pediatric acute myeloid leukemia: a report from the Children’s Oncology Group. Leuk Lymphoma. 2014;55(12):2817–21.CrossRefPubMedPubMedCentral Laszlo GS, Ries RE, Gudgeon CJ, Harrington KH, Alonzo TA, Gerbing RB, et al. High expression of suppressor of cytokine signaling-2 predicts poor outcome in pediatric acute myeloid leukemia: a report from the Children’s Oncology Group. Leuk Lymphoma. 2014;55(12):2817–21.CrossRefPubMedPubMedCentral
27.
go back to reference Badodi S, Baruffaldi F, Ganassi M, Battini R, Molinari S. Phosphorylation-dependent degradation of MEF2C contributes to regulate G2/M transition. Cell Cycle. 2015;14(10):1517–28.CrossRefPubMedPubMedCentral Badodi S, Baruffaldi F, Ganassi M, Battini R, Molinari S. Phosphorylation-dependent degradation of MEF2C contributes to regulate G2/M transition. Cell Cycle. 2015;14(10):1517–28.CrossRefPubMedPubMedCentral
28.
go back to reference Seita J, Sahoo D, Rossi DJ, Bhattacharya D, Serwold T, Inlay MA, et al. Gene Expression Commons: an open platform for absolute gene expression profiling. PLoS One. 2012;7(7):e40321.CrossRefPubMedPubMedCentral Seita J, Sahoo D, Rossi DJ, Bhattacharya D, Serwold T, Inlay MA, et al. Gene Expression Commons: an open platform for absolute gene expression profiling. PLoS One. 2012;7(7):e40321.CrossRefPubMedPubMedCentral
29.
go back to reference Gentles AJ, Plevritis SK, Majeti R, Alizadeh AA. Association of a leukemic stem cell gene expression signature with clinical outcomes in acute myeloid leukemia. JAMA. 2010;304(24):2706–15.CrossRefPubMedPubMedCentral Gentles AJ, Plevritis SK, Majeti R, Alizadeh AA. Association of a leukemic stem cell gene expression signature with clinical outcomes in acute myeloid leukemia. JAMA. 2010;304(24):2706–15.CrossRefPubMedPubMedCentral
31.
go back to reference Ho PA, Alonzo TA, Gerbing RB, Pollard J, Stirewalt DL, Hurwitz C, et al. Prevalence and prognostic implications of CEBPA mutations in pediatric acute myeloid leukemia (AML): a report from the Children’s Oncology Group. Blood. 2009;113(26):6558–66.CrossRefPubMedPubMedCentral Ho PA, Alonzo TA, Gerbing RB, Pollard J, Stirewalt DL, Hurwitz C, et al. Prevalence and prognostic implications of CEBPA mutations in pediatric acute myeloid leukemia (AML): a report from the Children’s Oncology Group. Blood. 2009;113(26):6558–66.CrossRefPubMedPubMedCentral
32.
go back to reference Loken MR, Alonzo TA, Pardo L, Gerbing RB, Raimondi SC, Hirsch BA, et al. Residual disease detected by multidimensional flow cytometry signifies high relapse risk in patients with de novo acute myeloid leukemia: a report from Children’s Oncology Group. Blood. 2012;120(8):1581–8.CrossRefPubMedPubMedCentral Loken MR, Alonzo TA, Pardo L, Gerbing RB, Raimondi SC, Hirsch BA, et al. Residual disease detected by multidimensional flow cytometry signifies high relapse risk in patients with de novo acute myeloid leukemia: a report from Children’s Oncology Group. Blood. 2012;120(8):1581–8.CrossRefPubMedPubMedCentral
33.
go back to reference Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–8.CrossRefPubMed Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–8.CrossRefPubMed
34.
go back to reference Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3(6):1101–8.CrossRefPubMed Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3(6):1101–8.CrossRefPubMed
35.
go back to reference Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc. 1958;53(282):457–81.CrossRef Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc. 1958;53(282):457–81.CrossRef
36.
go back to reference Kalbfleisch JD, Prentice RL. The statistical analysis of failure time data. Hoboken, NJ: John Wiley & Sons, Inc.; 2002.CrossRef Kalbfleisch JD, Prentice RL. The statistical analysis of failure time data. Hoboken, NJ: John Wiley & Sons, Inc.; 2002.CrossRef
37.
go back to reference Cox DR. Regression models and life tables (with discussion). J R Statist Soc. 1972;34(B):187–220. Cox DR. Regression models and life tables (with discussion). J R Statist Soc. 1972;34(B):187–220.
Metadata
Title
High expression of myocyte enhancer factor 2C (MEF2C) is associated with adverse-risk features and poor outcome in pediatric acute myeloid leukemia: a report from the Children’s Oncology Group
Authors
George S. Laszlo
Todd A. Alonzo
Chelsea J. Gudgeon
Kimberly H. Harrington
Alex Kentsis
Robert B. Gerbing
Yi-Cheng Wang
Rhonda E. Ries
Susana C. Raimondi
Betsy A. Hirsch
Alan S. Gamis
Soheil Meshinchi
Roland B. Walter
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2015
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-015-0215-4

Other articles of this Issue 1/2015

Journal of Hematology & Oncology 1/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine