Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The creatine kinase pathway is a metabolic vulnerability in EVI1-positive acute myeloid leukemia

Abstract

Expression of the MECOM (also known as EVI1) proto-oncogene is deregulated by chromosomal translocations in some cases of acute myeloid leukemia (AML) and is associated with poor clinical outcome. Here, through transcriptomic and metabolomic profiling of hematopoietic cells, we reveal that EVI1 overexpression alters cellular metabolism. A screen using pooled short hairpin RNAs (shRNAs) identified the ATP-buffering, mitochondrial creatine kinase CKMT1 as necessary for survival of EVI1-expressing cells in subjects with EVI1-positive AML. EVI1 promotes CKMT1 expression by repressing the myeloid differentiation regulator RUNX1. Suppression of arginine–creatine metabolism by CKMT1-directed shRNAs or by the small molecule cyclocreatine selectively decreased the viability, promoted the cell cycle arrest and apoptosis of human EVI1-positive cell lines, and prolonged survival in both orthotopic xenograft models and mouse models of primary AML. CKMT1 inhibition altered mitochondrial respiration and ATP production, an effect that was abrogated by phosphocreatine-mediated reactivation of the arginine–creatine pathway. Targeting CKMT1 is thus a promising therapeutic strategy for this EVI1-driven AML subtype that is highly resistant to current treatment regimens.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: EVI1 overexpression imparts new metabolic dependencies on AML cells.
Figure 2: EVI1-positive cells have high expression of CKMT1 and are dependent on CKMT1 for survival.
Figure 3: EVI1-mediated downregulation of RUNX1 expression promotes CKMT1 expression.
Figure 4: Blockade of the arginine–creatine pathway after CKMT1 inhibition impairs both mitochondrial respiration and ATP production in cells from patients with EVI1-positive AML.
Figure 5: Inhibition of the creatine kinase pathway alters the viability of AML cells derived from individuals with EVI-1-positive AML via cell cycle blockade and apoptosis induction.
Figure 6: CKMT1 knockdown preferentially impairs development of EVI1-positive human and mouse myeloid leukemias without affecting the viability of healthy progenitor cells.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

  1. Baysal, B.E. et al. Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma. Science 287, 848–851 (2000).

    Article  CAS  PubMed  Google Scholar 

  2. Janeway, K.A. et al. Defects in succinate dehydrogenase in gastrointestinal stromal tumors lacking KIT and PDGFRA mutations. Proc. Natl. Acad. Sci. USA 108, 314–318 (2011).

    Article  CAS  PubMed  Google Scholar 

  3. Ricketts, C. et al. Germline SDHB mutations and familial renal cell carcinoma. J. Natl. Cancer Inst. 100, 1260–1262 (2008).

    Article  CAS  PubMed  Google Scholar 

  4. Kim, S., Kim, D.H., Jung, W.H. & Koo, J.S. Succinate dehydrogenase expression in breast cancer. Springerplus 2, 299 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Cairns, R.A. & Mak, T.W. Oncogenic isocitrate dehydrogenase mutations: mechanisms, models and clinical opportunities. Cancer Discov. 3, 730–741 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Figueroa, M.E. et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function and impair hematopoietic differentiation. Cancer Cell 18, 553–567 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lu, C. et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 483, 474–478 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ward, P.S. et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting α-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 17, 225–234 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cantor, J.R. & Sabatini, D.M. Cancer cell metabolism: one hallmark, many faces. Cancer Discov. 2, 881–898 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Burnett, A., Wetzler, M. & Löwenberg, B. Therapeutic advances in acute myeloid leukemia. J. Clin. Oncol. 29, 487–494 (2011).

    Article  PubMed  Google Scholar 

  11. Patel, J.P. et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N. Engl. J. Med. 366, 1079–1089 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Glass, C., Wilson, M., Gonzalez, R., Zhang, Y. & Perkins, A.S. The role of EVI1 in myeloid malignancies. Blood Cells Mol. Dis. 53, 67–76 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Goyama, S. & Kurokawa, M. Evi1 as a critical regulator of leukemic cells. Int. J. Hematol. 91, 753–757 (2010).

    Article  CAS  PubMed  Google Scholar 

  14. 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. 28, 2101–2107 (2010).

    Article  PubMed  CAS  Google Scholar 

  15. Lugthart, S. et al. Clinical, molecular and prognostic significance of WHO type inv(3)(q21q26.2)/t(3;3)(q21;q26.2) and various other 3q abnormalities in acute myeloid leukemia. J. Clin. Oncol. 28, 3890–3898 (2010).

    Article  PubMed  Google Scholar 

  16. Kustikova, O.S. et al. Activation of Evi1 inhibits cell cycle progression and differentiation of hematopoietic progenitor cells. Leukemia 27, 1127–1138 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Goyama, S. et al. Evi1 is a critical regulator for hematopoietic stem cells and transformed leukemic cells. Cell Stem Cell 3, 207–220 (2008).

    Article  CAS  PubMed  Google Scholar 

  18. Kataoka, K. et al. Evi1 is essential for hematopoietic stem cell self-renewal, and its expression marks hematopoietic cells with long-term multilineage repopulating activity. J. Exp. Med. 208, 2403–2416 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bard-Chapeau, E.A. et al. Ecotopic viral integration site 1 (EVI1) regulates multiple cellular processes important for cancer and is a synergistic partner for FOS protein in invasive tumors. Proc. Natl. Acad. Sci. USA 109, 2168–2173 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Glass, C. et al. Global identification of EVI1 target genes in acute myeloid leukemia. PLoS One 8, e67134 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Furter, R., Furter-Graves, E.M. & Wallimann, T. Creatine kinase: the reactive cysteine is required for synergism but is non-essential for catalysis. Biochemistry 32, 7022–7029 (1993).

    Article  CAS  PubMed  Google Scholar 

  22. Burgess, M.R. et al. Preclinical efficacy of MEK inhibition in Nras-mutant AML. Blood 124, 3947–3955 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Li, Q. et al. Hematopoiesis and leukemogenesis in mice expressing oncogenic NrasG12D from the endogenous locus. Blood 117, 2022–2032 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Herranz, D. et al. Metabolic reprogramming induces resistance to anti-NOTCH1 therapies in T cell acute lymphoblastic leukemia. Nat. Med. 21, 1182–1189 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wang, Y.H. et al. Cell-state-specific metabolic dependency in hematopoiesis and leukemogenesis. Cell 158, 1309–1323 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mussai, F. et al. Arginine dependence of acute myeloid leukemia blast proliferation: a novel therapeutic target. Blood 125, 2386–2396 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Plunkett, W. Arginine addiction in AML. Blood 125, 3971–3972 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Miraki-Moud, F. et al. Arginine deprivation using pegylated arginine deiminase has activity against primary acute myeloid leukemia cells in vivo. Blood 125, 4060–4068 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Wallimann, T., Wyss, M., Brdiczka, D., Nicolay, K. & Eppenberger, H.M. Intracellular compartmentation, structure and function of creatine kinase isoenzymes in tissues with high and fluctuating energy demands: the 'phosphocreatine circuit' for cellular energy homeostasis. Biochem. J. 281, 21–40 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Roberts, D.J. & Miyamoto, S. Hexokinase II integrates energy metabolism and cellular protection: Akting on mitochondria and TORCing to autophagy. Cell Death Differ. 22, 248–257 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Arora, K.K. & Pedersen, P.L. Functional significance of mitochondrial-bound hexokinase in tumor cell metabolism. Evidence for preferential phosphorylation of glucose by intramitochondrially generated ATP. J. Biol. Chem. 263, 17422–17428 (1988).

    CAS  PubMed  Google Scholar 

  32. Mathupala, S.P., Ko, Y.H. & Pedersen, P.L. Hexokinase II: cancer's double-edged sword acting as both facilitator and gatekeeper of malignancy when bound to mitochondria. Oncogene 25, 4777–4786 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wyss, M. & Kaddurah-Daouk, R. Creatine and creatinine metabolism. Physiol. Rev. 80, 1107–1213 (2000).

    Article  CAS  PubMed  Google Scholar 

  34. Cimino, D. et al. Identification of new genes associated with breast cancer progression by gene expression analysis of predefined sets of neoplastic tissues. Int. J. Cancer 123, 1327–1338 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Gaidzik, V.I. et al. RUNX1 mutations in acute myeloid leukemia: results from a comprehensive genetic and clinical analysis from the AML study group. J. Clin. Oncol. 29, 1364–1372 (2011).

    Article  PubMed  Google Scholar 

  36. Zuber, J. et al. Toolkit for evaluating genes required for proliferation and survival using tetracycline-regulated RNAi. Nat. Biotechnol. 29, 79–83 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Banerji, V. et al. The intersection of genetic and chemical genomic screens identifies GSK-3α as a target in human acute myeloid leukemia. J. Clin. Invest. 122, 935–947 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Robinson, M.D., McCarthy, D.J. & Smyth, G.K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  39. Luo, B. et al. Highly parallel identification of essential genes in cancer cells. Proc. Natl. Acad. Sci. USA 105, 20380–20385 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ben-Sahra, I., Howell, J.J., Asara, J.M. & Manning, B.D. Stimulation of de novo pyrimidine synthesis by growth signaling through mTOR and S6K1. Science 339, 1323–1328 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Jones, S.J. Prediction of genomic functional elements. Annu. Rev. Genomics Hum. Genet. 7, 315–338 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Thorvaldsdóttir, H., Robinson, J.T. & Mesirov, J.P. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013).

    Article  PubMed  CAS  Google Scholar 

  43. Bernt, K.M. et al. MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell 20, 66–78 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Puissant, A. et al. SYK is a critical regulator of FLT3 in acute myeloid leukemia. Cancer Cell 25, 226–242 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Reich, M. et al. GenePattern 2.0. Nat. Genet. 38, 500–501 (2006).

    Article  CAS  PubMed  Google Scholar 

  46. Mootha, V.K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  47. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Merico, D., Isserlin, R. & Bader, G.D. Visualizing gene-set-enrichment results using the Cytoscape plug-in enrichment map. Methods Mol. Biol. 781, 257–277 (2011).

    Article  CAS  PubMed  Google Scholar 

  49. Merico, D., Isserlin, R., Stueker, O., Emili, A. & Bader, G.D. Enrichment map: a network-based method for gene-set-enrichment visualization and interpretation. PLoS One 5, e13984 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Li, B. & Dewey, C.N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Leng, N. et al. EBSeq: an empirical Bayes hierarchical model for inference in RNA-seq experiments. Bioinformatics 29, 1035–1043 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Novershtern, N. et al. Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell 144, 296–309 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Stegmaier, K. et al. Gene-expression-based high-throughput screening (GE–HTS) and application to leukemia differentiation. Nat. Genet. 36, 257–263 (2004).

    Article  CAS  PubMed  Google Scholar 

  55. Zuber, J. et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukemia. Nature 478, 524–528 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Daigle, S.R. et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20, 53–65 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wouters, B.J. et al. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood 113, 3088–3091 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Tomasson, M.H. et al. Somatic mutations and germline sequence variants in the expressed tyrosine kinase genes of patients with de novo acute myeloid leukemia. Blood 111, 4797–4808 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. Sato and M. Kurokawa (University of Tokyo) for providing plasmid constructs and detailed procedures for Evi1 overexpression in mouse hematopoietic cells. We also thank J.F. Clark (University of Cincinnati) for advice on the use of cyclocreatine in vivo. This research was supported with grants from the US National Cancer Institute (NCI) (NIH 1R35 CA210030-01 (K. Stegmaier) and R37 CA72614 (K. Shannon)), the Stand-up-to-Cancer Program (K. Stegmaier); the Bridge Project, a collaboration between the Koch Institute for Integrative Cancer Research at MIT and the Dana-Farber–Harvard Cancer Center (DF–HCC) (K. Stegmaier and M.T.H.) and the Koch Institute Cancer Center Support (NCI grant P30-CA14051; M.T.H.), and with support from the Cubans Curing Children's Cancers (4C's Fund) (K. Stegmaier). A.P. is a recipient of support from the ATIP–AVENIR and LNCC French research programs, the EHA research grant for a Non-Clinical Advanced fellow, and is supported by the St. Louis Association for leukemia research. K. Stegmaier is an LLS Scholar. A.P., N.F. and I.B.-S. were awarded the 'Prix Jeune Chercheur' from the Bettencourt Foundation and the Franco-American Exchange Prize from Philippe Foundation Inc.

Author information

Authors and Affiliations

Authors

Contributions

N.F. and C.F.B. contributed equally to the manuscript as joint first authors. N.F. and C.F.B. developed the study, established conditions for in vivo and in vitro experiments, acquired and analyzed the data, and wrote the manuscript; I.B.-S. designed and performed the metabolism-related experiments; L.B., A.R., Y.P., A.S.C. and F.L. designed, performed, and analyzed the in vivo experiments; Q.L., M.R.B. and K. Shannon revised the manuscript and provided NrasG12D and NrasG12D + Evi1 mouse models and methodology for in vivo functional analyses; G.A. revised the manuscript and performed statistical analysis, biostatistics, and computational analysis of the RNA sequencing, the publicly available patient sample cohorts, and the shRNA screening experiments; A.S.P. and Y.Z. provided reagents and ChIP-seq data for the ChIP–qPCR experiments performed on endogenous mouse Evi1; I.G., D.J.D. and R.M.S. provided patient samples and revised the manuscript; P.A. revised the manuscript; M.T.H., A.P. and K Stegmaier contributed equally to this work as joint senior authors; M.T.H., A.P. and K. Stegmaier supervised the study, wrote and revised the manuscript, designed the in vitro and in vivo experiments, analyzed the data and provided funding for the study.

Corresponding author

Correspondence to Kimberly Stegmaier.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7, Supplementary Tables 1–8 and Supplementary Note (PDF 19995 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fenouille, N., Bassil, C., Ben-Sahra, I. et al. The creatine kinase pathway is a metabolic vulnerability in EVI1-positive acute myeloid leukemia. Nat Med 23, 301–313 (2017). https://doi.org/10.1038/nm.4283

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4283

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer