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:

Genomic subtyping and therapeutic targeting of acute erythroleukemia

Abstract

Acute erythroid leukemia (AEL) is a high-risk leukemia of poorly understood genetic basis, with controversy regarding diagnosis in the spectrum of myelodysplasia and myeloid leukemia. We compared genomic features of 159 childhood and adult AEL cases with non-AEL myeloid disorders and defined five age-related subgroups with distinct transcriptional profiles: adult, TP53 mutated; NPM1 mutated; KMT2A mutated/rearranged; adult, DDX41 mutated; and pediatric, NUP98 rearranged. Genomic features influenced outcome, with NPM1 mutations and HOXB9 overexpression being associated with a favorable prognosis and TP53, FLT3 or RB1 alterations associated with poor survival. Targetable signaling mutations were present in 45% of cases and included recurrent mutations of ALK and NTRK1, the latter of which drives erythroid leukemogenesis sensitive to TRK inhibition. This genomic landscape of AEL provides the framework for accurate diagnosis and risk stratification of this disease, and the rationale for testing targeted therapies in this high-risk leukemia.

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

Fig. 1: Demographic, clinical and genomic patient characteristics.
Fig. 2: Mutation rates in patients with AEL (WHO 2008), non-erythroid AML and MDS.
Fig. 3: Genomic classification of AEL.
Fig. 4: Association with clinical outcome.
Fig. 5: NTRK1 mutations in AEL.

Similar content being viewed by others

Data availability

Genomic data including both sequencing and copy number data have been deposited in the European Genome-phenome Archive (EGA), accession EGAS00001002537. This includes the following datasets: whole-exome sequencing data (EGAD00001003413), RNA-sequencing data (EGAD00001003412) and SNP6 Affymetrix copy number data (EGAD00010001443). Moreover, genomic data including nonsilent SNV, indels, ITD sequence mutations, in-frame gene fusions, structural variations, copy number aberrations and gene expression data can be explored interactively at the St. Jude PeCan Data Portal88 (https://pecan.stjude.cloud/proteinpaint/study/ael).

References

  1. Cancer Genome Atlas Research Network Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368, 2059–2074 (2013).

    Google Scholar 

  2. Papaemmanuil, E. et al. Genomic classification and prognosis in acute myeloid leukemia. N. Engl. J. Med. 374, 2209–2221 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Cervera, N. et al. Molecular characterization of acute erythroid leukemia (M6-AML) using targeted next-generation sequencing. Leukemia 30, 966–970 (2016).

    CAS  PubMed  Google Scholar 

  4. Grossmann, V. et al. Acute erythroid leukemia (AEL) can be separated into distinct prognostic subsets based on cytogenetic and molecular genetic characteristics. Leukemia 27, 1940–1943 (2013).

    CAS  PubMed  Google Scholar 

  5. Ping, N. et al. Exome sequencing identifies highly recurrent somatic GATA2 and CEBPA mutations in acute erythroid leukemia. Leukemia 31, 195–202 (2017).

    CAS  PubMed  Google Scholar 

  6. Liu, W. et al. Pure erythroid leukemia: a reassessment of the entity using the 2008 World Health Organization classification. Mod. Pathol. 24, 375–383 (2011).

    CAS  PubMed  Google Scholar 

  7. Almeida, A. M. et al. Clinical outcomes of 217 patients with acute erythroleukemia according to treatment type and line: a retrospective multinational study. Int. J. Mol. Sci. 18, E837 (2017).

    PubMed  Google Scholar 

  8. Dameshek, W. & Baldini, M. The Di Guglielmo syndrome. Blood 13, 192–194 (1958).

    CAS  PubMed  Google Scholar 

  9. Bennett, J. M. et al. Proposed revised criteria for the classification of acute myeloid leukemia: a report of the French-American-British Cooperative Group. Ann. Intern. Med. 103, 620–625 (1985).

    CAS  PubMed  Google Scholar 

  10. Vardiman, J. W. et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood 114, 937–951 (2009).

    CAS  PubMed  Google Scholar 

  11. Arber, D. A. et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 127, 2391–2405 (2016).

    CAS  PubMed  Google Scholar 

  12. Kowal-Vern, A. et al. Diagnosis and characterization of acute erythroleukemia subsets by determining the percentages of myeloblasts and proerythroblasts in 69 cases. Am. J. Hematol. 65, 5–13 (2000).

    CAS  PubMed  Google Scholar 

  13. Lichtman, M. A. The disappearance of acute erythroid leukemia: an act of legerdemain at the world health organization. Blood Cells Mol. Dis. 61, 54–57 (2016).

    PubMed  Google Scholar 

  14. Arber, D. A. Revisiting erythroleukemia. Curr. Opin. Hematol. 24, 146–151 (2017).

    PubMed  Google Scholar 

  15. Wang, S. A. et al. Acute erythroid leukemia with <20% bone marrow blasts is clinically and biologically similar to myelodysplastic syndrome with excess blasts. Mod. Pathol. 29, 1221–1231 (2016).

    CAS  PubMed  Google Scholar 

  16. Hasserjian, R. P. et al. Acute erythroid leukemia: a reassessment using criteria refined in the 2008 WHO classification. Blood 115, 1985–1992 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Greenberg, P. L. et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood 120, 2454–2465 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Pounds, S. et al. A genomic random interval model for statistical analysis of genomic lesion data. Bioinformatics 29, 2088–2095 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Forbes, S. A. et al. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res. 45, D777–D783 (2017).

    CAS  PubMed  Google Scholar 

  20. Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371, 2488–2498 (2014).

    PubMed  PubMed Central  Google Scholar 

  21. Xie, M. et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 20, 1472–1478 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 371, 2477–2487 (2014).

    PubMed  PubMed Central  Google Scholar 

  23. Stephens, P. J. et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Rausch, T. et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell 148, 59–71 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Abaigar, M. et al. Chromothripsis is a recurrent genomic abnormality in high-risk myelodysplastic syndromes. PLoS One 11, e0164370 (2016).

    PubMed  PubMed Central  Google Scholar 

  26. Yang, C. T. et al. Activation of KLF1 enhances the differentiation and maturation of red blood cells from human pluripotent stem cells. Stem Cells 35, 886–897 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Holmfeldt, P. et al. Nfix is a novel regulator of murine hematopoietic stem and progenitor cell survival. Blood 122, 2987–2996 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Panagopoulos, I. et al. Fusion of ZMYND8 and RELA genes in acute erythroid leukemia. PLoS One 8, e63663 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Quelen, C. et al. Identification of a transforming MYB-GATA1 fusion gene in acute basophilic leukemia: a new entity in male infants. Blood 117, 5719–5722 (2011).

    CAS  PubMed  Google Scholar 

  30. Wang, G. G. et al. Haematopoietic malignancies caused by dysregulation of a chromatin-binding PHD finger. Nature 459, 847–851 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Majeti, R. et al. Dysregulated gene expression networks in human acute myelogenous leukemia stem cells. Proc. Natl Acad. Sci. USA 106, 3396–3401 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Andersson, A., Eden, P., Olofsson, T. & Fioretos, T. Gene expression signatures in childhood acute leukemias are largely unique and distinct from those of normal tissues and other malignancies. BMC Med. Genomics 3, 6 (2010).

    PubMed  PubMed Central  Google Scholar 

  33. Gatalica, Z., Xiu, J., Swensen, J. & Vranic, S. Molecular characterization of cancers with NTRK gene fusions. Mod. Pathol. 32, 147–153 (2018).

    PubMed  Google Scholar 

  34. Taylor, J. et al. Oncogenic TRK fusions are amenable to inhibition in hematologic malignancies. J. Clin. Invest. 128, 3819–3825 (2018).

    PubMed  PubMed Central  Google Scholar 

  35. Roberts, K. G. et al. Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. N. Engl. J. Med. 371, 1005–1015 (2014).

    PubMed  PubMed Central  Google Scholar 

  36. Roberts, K. G. et al. ETV6-NTRK3 induces aggressive acute lymphoblastic leukemia highly sensitive to selective TRK inhibition. Blood 132, 861–865 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Lang, G. A. et al. Gain of function of a p53 hot spot mutation in a mouse model of Li-Fraumeni syndrome. Cell 119, 861–872 (2004).

    CAS  PubMed  Google Scholar 

  38. Grunewald, T. G. et al. Chimeric EWSR1-FLI1 regulates the Ewing sarcoma susceptibility gene EGR2 via a GGAA microsatellite. Nat. Genet. 47, 1073–1078 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Doebele, R. C. et al. An oncogenic NTRK fusion in a patient with soft-tissue sarcoma with response to the tropomyosin-related kinase inhibitor LOXO-101. Cancer Discov. 5, 1049–1057 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Khotskaya, Y. B. et al. Targeting TRK family proteins in cancer. Pharmacol Ther. 173, 58–66 (2017).

    CAS  PubMed  Google Scholar 

  41. Dohner, H. et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 129, 424–447 (2017).

    PubMed  PubMed Central  Google Scholar 

  42. Welch, J. S. et al. TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes. N. Engl. J. Med. 375, 2023–2036 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Bertrand, T. et al. The crystal structures of TrkA and TrkB suggest key regions for achieving selective inhibition. J. Mol. Biol. 423, 439–453 (2012).

    CAS  PubMed  Google Scholar 

  44. Bertrand, T. Crystal structures of neurotrophin receptors kinase domain. Vitam. Horm. 104, 1–18 (2017).

    PubMed  Google Scholar 

  45. Pession, A. et al. Results of the AIEOP AML 2002/01 multicenter prospective trial for the treatment of children with acute myeloid leukemia. Blood 122, 170–178 (2013).

    CAS  PubMed  Google Scholar 

  46. Tomizawa, D. et al. Excess treatment reduction including anthracyclines results in higher incidence of relapse in core binding factor acute myeloid leukemia in children. Leukemia 27, 2413–2416 (2013).

    CAS  PubMed  Google Scholar 

  47. Schlenk, R. F. et al. All-trans retinoic acid as adjunct to intensive treatment in younger adult patients with acute myeloid leukemia: results of the randomized AMLSG 07-04 study. Ann. Hematol. 95, 1931–1942 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Lange, B. J. et al. Outcomes in CCG-2961, a children’s oncology group phase 3 trial for untreated pediatric acute myeloid leukemia: a report from the children’s oncology group. Blood 111, 1044–1053 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Buchner, T. et al. Acute Myeloid Leukemia (AML): different treatment strategies versus a common standard arm–combined prospective analysis by the German AML Intergroup. J. Clin. Oncol. 30, 3604–3610 (2012).

    PubMed  Google Scholar 

  50. Farrar, J. E. et al. Genomic profiling of pediatric acute myeloid leukemia reveals a changing mutational landscape from disease diagnosis to relapse. Cancer Res 76, 2197–2205 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Cancer Genome Atlas Research Network et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368, 2059–2074 (2013).

    Google Scholar 

  52. Bolouri, H. et al. The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions. Nat. Med. 24, 103–112 (2018).

    CAS  PubMed  Google Scholar 

  53. Lindsley, R. C. et al. Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation. N. Engl. J. Med. 376, 536–547 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Kitamura, T. et al. Establishment and characterization of a unique human cell line that proliferates dependently on GM-CSF, IL-3, or erythropoietin. J. Cell Physiol. 140, 323–334 (1989).

    CAS  PubMed  Google Scholar 

  55. Martin, P. & Papayannopoulou, T. HEL cells: a new human erythroleukemia cell line with spontaneous and induced globin expression. Science 216, 1233–1235 (1982).

    CAS  PubMed  Google Scholar 

  56. Drmanac, R. et al. Human genome sequencing using unchained base reads on self-assembling DNA nanoarrays. Science 327, 78–81 (2010).

    CAS  PubMed  Google Scholar 

  57. Liu, Y. et al. The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia. Nat. Genet. 49, 1211–1218 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhang, J. et al. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature 481, 157–163 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Zhang, J. et al. SNPdetector: a software tool for sensitive and accurate SNP detection. PLoS Comput. Biol. 1, e53 (2005).

    PubMed  PubMed Central  Google Scholar 

  60. Edmonson, M. N. et al. Bambino: a variant detector and alignment viewer for next-generation sequencing data in the SAM/BAM format. Bioinformatics 27, 865–866 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Downing, J. R. et al. The pediatric cancer genome project. Nat. Genet. 44, 619–622 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Roberts, K. G. et al. High frequency and poor outcome of Philadelphia chromosome-like acute lymphoblastic leukemia in adults. J. Clin. Oncol. 35, 394–401 (2017).

    PubMed  Google Scholar 

  63. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  64. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. 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).

    CAS  PubMed  Google Scholar 

  66. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    PubMed  PubMed Central  Google Scholar 

  67. Pawlikowska, I. et al. Dunn Index Bootstrap (DIBS): A procedure to empirically select a cluster analysis method that identifies biologically and clinically relevant molecular disease subgroups. BMC Bioinformatics 16, P2 (2015).

    Google Scholar 

  68. Rousseeuw, P. J. Least median of squares regression. J. Am. Stat. Assoc. 79, 871–880 (1984).

    Google Scholar 

  69. He, J. et al. Integrated genomic DNA/RNA profiling of hematologic malignancies in the clinical setting. Blood 127, 3004–3014 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Kohlmann, A. et al. The Interlaboratory RObustness of Next-generation sequencing (IRON) study: a deep sequencing investigation of TET2, CBL and KRAS mutations by an international consortium involving 10 laboratories. Leukemia 25, 1840–1848 (2011).

    CAS  PubMed  Google Scholar 

  71. Delic, S. et al. Application of an NGS-based 28-gene panel in myeloproliferative neoplasms reveals distinct mutation patterns in essential thrombocythaemia, primary myelofibrosis and polycythaemia vera. Br. J. Haematol. 175, 419–426 (2016).

    CAS  PubMed  Google Scholar 

  72. Weisser, M. et al. Risk assessment by monitoring expression levels of partial tandem duplications in the MLL gene in acute myeloid leukemia during therapy. Haematologica 90, 881–889 (2005).

    CAS  PubMed  Google Scholar 

  73. Schnittger, S. et al. Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease. Blood 100, 59–66 (2002).

    CAS  PubMed  Google Scholar 

  74. Mullighan, C. G. et al. Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature 446, 758–764 (2007).

    CAS  PubMed  Google Scholar 

  75. Lin, M. et al. dChipSNP: significance curve and clustering of SNP-array-based loss-of-heterozygosity data. Bioinformatics 20, 1233–1240 (2004).

    CAS  PubMed  Google Scholar 

  76. Venkatraman, E. S. & Olshen, A. B. A faster circular binary segmentation algorithm for the analysis of array CGH data. Bioinformatics 23, 657–663 (2007).

    CAS  PubMed  Google Scholar 

  77. Wilcoxon, F. Individual comparisons by ranking methods. Biometrics 1, 80–83 (1945).

    Google Scholar 

  78. Storey, J. D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl Acad. Sci. USA 100, 9440–9445 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal 6, pl1 (2013).

    PubMed  PubMed Central  Google Scholar 

  80. Huntley, R. P. et al. The GOA database: gene Ontology annotation updates for 2015. Nucleic Acids Res. 43, D1057–D1063 (2015).

    CAS  PubMed  Google Scholar 

  81. Huether, R. et al. The landscape of somatic mutations in epigenetic regulators across 1,000 paediatric cancer genomes. Nat. Commun. 5, 3630 (2014).

    PubMed  Google Scholar 

  82. The PyMOL Molecular Graphics System v.1.3r1 (Schrodinger LLC, 2010).

  83. Iacobucci, I. et al. Truncating erythropoietin receptor rearrangements in acute lymphoblastic leukemia. Cancer Cell 29, 186–200 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Mullighan, C. G. et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N. Engl. J. Med. 360, 470–480 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Mantel, N. Evaluation of survival data and two new rank order statistics arising in its consideration. Cancer Chemother. Rep. 50, 163–170 (1966).

    CAS  PubMed  Google Scholar 

  86. Fine, J. P. & Gray, R. J. A proportional hazards model for the subdistribution of a competing risk. J. Am. Stat. Assoc. 94, 496–509 (1999).

    Google Scholar 

  87. R Development Core Team. R: a Language and Environment for Statistical Computing (R Foundation for Statistical Computing, Vienna, Austria, 2009).

  88. Zhou, X. et al. Exploring genomic alteration in pediatric cancer using ProteinPaint. Nat. Genet. 48, 4–6 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported in part by the American Lebanese Syrian Associated Charities of St. Jude Children’s Research Hospital; a Stand Up to Cancer Innovative Research Grant and a St. Baldrick’s Foundation Robert J. Arceci Innovation Award (to C.G.M.); a Leukemia and Lymphoma Society Specialized Center of Research grant (to C.G.M.); the Henry Schueler 41&9 Foundation (to C.G.M.); a Lady Tata Memorial Trust Award (to I.I.); St. Jude Children’s Research Hospital Hematological Malignancies Program Garwood Fellowship (to I.I.); Italian Scientists and Scholars in North America Foundation (ISSNAF) to I.I.; a Leukemia and Lymphoma Society Translational Research Program (to C.G.M.); a National Cancer Institute Outstanding Investigator Award R35 CA197695 (to C.G.M.); the R25CA23944 from the National Cancer Institute (to St. Jude Pediatric Oncology Education program); a St. Jude Summer Plus Fellowship, Rhodes College (to S.M.M.); NIH Cancer Center Support Grant P30 CA21765 (to C.G.M.); Fondazione Cariparo (to G.B.) and AIRC (to G.B.); AIRC 5 × 1.000 ‘Immunity in Cancer Spreading and Metastasis’ (to F.L.); National Medical Research Council, Singapore (NMRC/CSA/0053/2013) (to A.E.J.Y.); and Cancer Science Institute of Singapore (to A.E.J.Y.). This work was also supported by a Project Grant (516726 to B.T.K.) and Program Grants (1016647 and 1113577 to B.T.K.), a Fellowship (1063008 to B.T.K.), and an Independent Research Institutes Infrastructure Support Scheme Grant (361646) from the Australian National Health and Medical Research Council (to B.T.K.); the Leukaemia Foundation of Australia (to C.L.C. and B.T.K.); the Sylvia & Charles Viertel Foundation (to B.T.K.); the Australian Cancer Research Fund (to B.T.K.); Cancer Council of South Australia Beat Cancer Project (1145385, to A.L.B., H.S.S. and C.N.H.) and a Victorian State Government Operational Infrastructure Support Grant (to B.T.K.). We thank the ALLG Tissue Bank at the Princess Alexandra Hospital, Brisbane (now the Cancer Collaborative Biobank), for providing samples. The ALLG Tissue Bank received funding support from the Leukaemia Foundation (to P.M.), the National Health and Medical Research Council (to P.M.), and Queensland Health and Pathology Queensland for the ALLG Tissue Bank (to P.M.). The South Australian Cancer Research Biobank was supported by the Cancer Council SA Beat Cancer Project, University of Adelaide, University of South Australia, South Australian Health and Medical Research Institute, SA Health, Health Service Gifts and Charitable Board of the Central Adelaide Local Health Network, Medvet Laboratories Pty Ltd and the Government of South Australia (to L.B.T., R.D.A., H.S. and I.L.). We thank the staff of the Biorepository, the Hartwell Centre for Bioinformatics and Biotechnology, the Flow Cytometry and Cell Sorting Core Facility, the Cell and Tissue Imaging Facility, the Animal Resources Center and the Small Animal Imaging Center, the Compound Management Center and the Department of Chemical Biology & Therapeutics of St. Jude Children’s Research Hospital. We thank Loxo Oncology, Inc. for providing larotrectinib and support in dosing.

Author information

Authors and Affiliations

Authors

Contributions

I.I. led the collaboration, coordination and processing of patient samples, analyzed genomic data, performed experiments and wrote the manuscript. J.W. analyzed sequencing data. J.K.C. performed central review of immunophenotypic data. L.J.J. performed histopathology analyses. K.E.M., S.M.M., T.B.A., D.P.-T., M.V. and V.V. performed experiments. G.S. analyzed copy number data. E.J.E. performed structure modeling. M.M, C.L.C., D.T., N.K., G.B., F.L., S.K.Y.K., A.E.J.Y., R.E.R., E.S., A.H.W., L.B.T., I.D.L., R.J.D.A., B.T.K., A.L.B., H.S.S., C.N.H., P.M., S.M., M.L.L. and T.H. provided patient samples and clinical data. R.C.L. and B.L.E. shared data for the MDS comparison cohort. Y.L., C.Q., X.M., X.Z., E.S., S.P.H. and M.R. performed genomic sequencing, analysis and support. L.S., S.B.P., D.P. and C.C. performed statistical analyses. C.G.M. designed and oversaw the study, analyzed genomic data and wrote the manuscript. All the authors read and approved the final version of the manuscript.

Corresponding author

Correspondence to Charles G. Mullighan.

Ethics declarations

Competing interests

M.M., employed by MLL Munich Leukemia Laboratory; R.C.L., research funding from MedImmune and Jazz, and consulting fees from Takeda; B.L.E., research funding from Celgene and Deerfield, and consulting fees from GRAIL; T.H., equity ownership of MLL Munich Leukemia Laboratory; C.G.M., research funding from Loxo Oncology for TRK inhibitors in acute lymphoblastic leukemia, and Abbvie and Pfizer for studies unrelated to those described herein. C.G.M. has received honoraria and travel support from Pfizer and Amgen.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Mutation distribution.

(a) Genetic variants including non-silent SNVs, indels or ITD sequence mutations, in-frame gene fusions, structural variations and focal copy number aberrations in AEL patients (n = 159 biologically independent samples). (b) Variant burden according to age (P value is from one-way ANOVA), IPSS-R cytogenetic risk (adjusted P values are from one-way ANOVA with post-hoc Tukey Honestly Significant Difference Test Calculator) and WHO 2008 (two-sided P value is from t test) and revised WHO 2016 diagnosis criteria (P value is from one-way ANOVA) (n = 159 biologically independent samples). Mean and S.D. are shown. (c) Proportion of nucleotide transition and transversion mutations according to age (n = 142 biologically independent samples with WES). Abbreviations: MAF, Mutant allele frequency.

Supplementary Figure 2 Mutations in DNA methylation genes and tumor suppressor genes.

(a) Heat map showing the distribution of mutations in DNA methylation genes and (b) tumor suppressor genes across age, IPSS-R cytogenetic risk groups and leukemia subtypes (WHO 2008 and WHO 2016). (c) Frequency of TP53 mutations according to the indicated age groups (n = 159 biologically independent samples), IPSS-R cytogenetic risk (n = 158 biologically independent samples), WHO 2008 (n = 150 biologically independent samples) and WHO 2016 (n = 149 biologically independent samples) criteria. Two-side P values are from Chi-square test (see Supplementary Table 9 for numbers for each group). Mean and S.D. are shown. Differences were identified according to age and cytogenetic risk. (d) Distribution of TP53 variant allele frequency in adult (n = 7) and older adult patients (n = 41). No significant differences (t test, two-sided P value = 0.7763) were identified. Mean and S.D. are shown. (e) Pie chart showing the co-occurrence of two TP53 mutations (MUT/MUT), a mutation and a loss of the other allele (MUT/LOSS) or a mutation and copy neutral loss of heterozygosis (LOH) (MUT/LOH). The percentages are calculated on the total mutated TP53 AEL cases (n = 51). (f) ProteinPaint visualization of TP53 (see URLs). (g) FISH with ZBTB4 5´ and ZBTB4 3´ showing one allele missing and the other allele broken apart (left). Addition of TP53 5´ shows pairing with ZBTB4 3´ (right). (h) Sanger sequencing electropherograms showing the rearrangement involving TP53 intron 1 and ZBTB4 intron 1 in SJAEL011863 (upper) and TP53 intron 1 and an intragenic region on chromosome 17 in SJAEL015002 (lower). This latter alteration was identifiable only by whole genome sequencing. (i) Log2 counts per million reads (CPM) for ZBTB4 from RNA-seq in the case with the TP53-ZBTB4 rearrangement (SJAEL011863) in red and the remaining AEL cases (total cases, n = 139). (j) Log2CPM values for TP53 from RNA-seq data (n = 139 biologically independent samples). Interestingly, the samples with rearrangements in intron 1 of TP53 had the lowest expression values for TP53 among the entire AEL cohort.

Supplementary Figure 3 Mutations in signaling genes.

(a) Pie charts showing the distribution of the different signaling pathway mutations in the whole AEL cohort (n = 159 biologically independent samples). (b) Matrix showing the distribution of mutations in genes involved in signaling pathways across age, IPSS-R cytogenetic risk groups and leukemia subtypes (WHO 2008 and WHO 2016). (c) Heatmap showing the variant allele frequency (VAF) for genes in the different signaling pathways. Each column is a patient. (d) Variant allele frequency boxplots comparing the VAF ranges in samples with one Ras pathway mutation (n = 16) and in those with at least 2 Ras pathway mutations (n = 13). Two-sided P value is from t test.

Supplementary Figure 4 Copy number alteration events detected by using SNP6 microarrays.

(a) Patients (n = 137 independent biological samples) are grouped by cytogenetic risk according to IPSS-R. (b) Correlation of copy number alterations with IPSS-R cytogenetic risk (Very good/Good: n = 59; Intermediate: n = 21; Poor: n = 6 and Very poor: n = 51; adjusted P values are from one-way ANOVA with post-hoc Tukey Honestly Significant Difference Test Calculator), TP53 mutations (TP53 wild-type, WT: n = 87; TP53 mutated, MUT: n = 50; P value is from t test), age (Pediatric: n = 20; Young adult: n = 6; Adult: n = 29; Older Adult: n = 82; adjusted P values are from one-way ANOVA with post-hoc Tukey Honestly Significant Difference Test Calculator) and M6 phenotype (M6a: n = 126; M6b: n = 11; two-sided P value is from t test). Mean and S.D. are shown. Correlation of loss of chromosomes 5 (c) and 7 (d) with age (left panel) and IPSS-R cytogenetic risk (right panel). Two-sided P values are from Chi-square test. Independent biological samples for each group are shown in the figure. (e) Number of CNAs in each chromosome. (f) Chromosomes affected by chromothripsis.

Supplementary Figure 5 Chromothripsis.

(a) Views by UCSC Genome Browser of chromosome 19 zoomed on the common deleted region. The Y axis shows the log2 copy number ratio. (b) Pie charts show the distribution of the recurrently mutated pathways in patients with chromothripsis.

Supplementary Figure 6 Comparison of mutation rates in pediatric and adult AEL cases according to WHO 2008 and reclassified as MDS according to WHO 2016 criteria (referred as AEL/MDS) and MDS.

(a) Comparison of mutation rates in pediatric (0-20 years) AEL/MDS (n = 11 biologically independent samples) and MDS (n= 104 biologically independent samples). (b) Comparison of mutation rates in adult AEL/MDS (n = 111 biologically independent samples) and MDS (n = 1410 biologically independent samples). Only cases for which sequencing data were available for both cohorts are reported. Data are from non-synonymous SNVs and indels. P values are from Fisher’s test (see Supplementary Table 19 for numbers for each group).

Supplementary Figure 7 Chimeric in-frame gene fusions in pediatric and adult AEL.

ProteinPaint schematic representation of chimeric gene fusions (right: a, MYB-GATA1; b, ASNS-PTPN1; c, ZMYND8-RELA; d, APLP2-EPOR; e, PFN1-SCHIP; f, NPM1-MLF1) and Sanger sequencing electropherograms showing the gene fusions (left). Abbreviations. LMSTEN, Leucine- Methionine- Serine- Threonine- Glutamic Acid- Asparagine motif in Myb proteins; Asn, Asparagine synthetase; PTPc, protein tyrosine phosphatase catalytic domain; PTTP, Pro-Trp-Trp-Pro motif; MYND finger, myeloid, Nervy, and DEAF-1 domain; RHD-n_RelA, Rel homology domain of RelA; IPT_NFkappaB, Immunoglobulin-like fold, Plexins, Transcription factors domain of NFkappaB; BPTI/Kunitz domain, bovine pancreatic trypsin inhibitor/Kunitz domain; APP, amyloid precursor protein; IL6Ra-bindInterleukin-6 receptor alpha chain, binding; FN3Fibronectin type 3 domain; SCHIP-1, Schwannomin-interacting protein 1; Mlf1IP, Myelodysplasia-myeloid leukemia factor 1-interacting protein. (g) Fluorescence in situ hybridization (FISH) for MYB-GATA1. MYB 5′ and GATA1 3′ (left) followed by addition of GATA1 5′ (right) showing insertion of GATA1 3′ into MYB 5′ in SJAEL011884. One hundred cells were analyzed and 96 (96%) were positive for MYB-GATA1 fusion. (h) FISH for ZMYND8-RELA fusion. ZMYND8 5′ and ZMYND8 3′ (left) followed by addition of RELA 3′ (right) demonstrating the fusions of ZMYND8 to RELA in SJAEL011882. One hundred cells were analyzed and 75 (75%) had the ZMYND8-RELA rearrangement.

Supplementary Figure 8 Pairwise associations between gene mutations.

Top rows indicate grouping of genes according to pathway from 159 patients. Left/upper triangular half of the figure displays the number of cases having alterations of both genes. Right/lower half of the figure displays the odds ratio (represented by color of square) and P value from the Fisher exact test (represented by size of black dots) testing the association between two genes. Odds ratio (OR) represents the odds of genomic event in one gene, compared to odds of genomic event in the other gene. OR>1 indicates a positive association between two genes. Conversely, OR<1 indicates a negative association. When there is at least one cell with zero in the 2*2 contingency table, the corresponding cell color in the co-occurrence matrix is set to white (which is not indicated in the figure legend) since the calculated odd ratio is either zero or infinite.

Supplementary Figure 9 Overall survival according to genomic features.

Kaplan–Meier survival curves with overall survival distributions according to gene expression clusters (n = 119), chromothripsis (n = 125 biologically independent samples), number of driver gene mutations in the same sample (1-2, 3-4, 5-6, 7-8 or > 8) (n = 147), functional pathway (cell cycle regulation, cohesin, DNA methylation, DNA repair, epigenomic, NPM1, signaling, RNA/processing/splicing and transcription regulation) (n = 147 biologically independent samples) and mutated genes (TP53, FLT3 and RB1) (n = 147 biologically independent samples). Survival curves were estimated for each group, considered separately, using the Kaplan–Meier method and compared statistically using the log rank test. Two-side P values were reported, multiple comparisons were not adjusted. See also Supplementary Table 25. Y: mutated; N: wild-type.

Supplementary Figure 10 NUP98 fusions in AEL.

(a) Schematic visualization of NUP98 fusions. Abbreviations: JmjN, Jumonji N-terminal domain; JmjC, Jumonji C-terminal domain. (b) Supervised hierarchical clustering analysis (using the top 500 differentially expressed coding and non-coding RNAs in pediatric cases (0-20 years) with/without NUP98 fusions (n = 20 biologically independent samples) (see also Supplementary Table 29 for the complete list of genes). (c) Volcano plot showing the differentially expressed genes between NUP98-fusion positive AEL and NUP98-fusion negative AEL cases (n = 20 biologically independent samples). Two-sided P values were identified by Limma Voom method. (d) Colony forming unit-granulocyte, macrophage (CFU-GM) and burst forming unit-erythroid (BFU-E) of lineage-negative mouse fetal liver cells expressing a fusion gene or transduced with empty vector (MIG). Columns show means of three replicates ± S.D. The experiment was performed independently twice with similar results. (e) Bioluminescent images from sublethally C57BL/6 mice transplanted with fetal liver lin-negative HSPCs expressing NUP98-KDM5A fusion or empty vector and captured after 12 weeks following transplantation from each group. (f) Splenic cells harvested from moribund primary recipients (median latency of 119 days) were GFP-sorted and injected into sublethally irradiated secondary recipient mice (shorter latency with a median of 90 days, P = 0.018) and Kaplan–Meier survival curves are shown for each group (Primary: n = 5; Secondary: n = 12). The Kaplan–Meier survival curve from primary mice (n = 5) transplanted with lin-negative HSPCs transduced with empty vector is shown as comparison. Outcome associations were analyzed with the log-rank test. Spleen weight in primary and secondary recipient mice expressing NUP98-KDM5A is reported. The mean expression is shown by the horizontal line in the scatter dot plot and the error bars represent the S.D. (g)(left) Representative histology showing multisystemic infiltration of NUP98-KDM5A induced leukemia in a primary mouse recipient. Scale bars represent 100 μm (bone marrow and uterus), 250 μm (meninges, skin-subcutis, kidney and liver) and 500 μm (pancreatic lymph node and spinal cord). Necropsy revealed moderate to marked leukemia infiltrates in multiple organs assessed including bone marrow, adrenal gland, brain, kidneys, liver, lymph nodes, salivary gland liver, skin, stomach and uterus confirming the systemic nature of the disease. (g)(right) Hematoxylin and eosin staining and MPO, B220, PAX5, CD41, GATA1, RUNX1 and GlyA labeling of lymph node from a representative mouse with NUP98-KDM5A induced leukemia. Scale bars are 50 μm. An erythroid phenotype (left) was excluded and the myeloid phenotype supported by the positive expression of RUNX1 and the negative expression of GATA1 and Glycophorin A; a megakaryocytic phenotype was ruled out by the negative expression of CD41. This analysis was performed in additional three independent primary mouse tumors and in two secondary recipient mice, obtaining similar results. (h) Flow cytometry of bone marrow cells harvested from a representative moribund primary (left) and secondary (right) recipient mouse with NUP98-KDM5A leukemia is shown. Cells were gated based on GFP expression. This analysis was performed on all mice in study that developed leukemia obtaining similar results.

Supplementary Figure 11 NTRK1 mutations in AEL.

(a) NTRK1 expression from RNA-seq and expressed as log2 counts per million, CPM, mapped reads according to NTRK1 mutational status (left panel, P value is from unpaired t test) and according to genomic subgroups (right panel, P value is from one-way ANOVA). The number of independent samples for each group is reported in each panel. Mean and S.D. are shown. (b) Immunofluorescence for NTRK1 expression in NIH 3T3 cells expressing WT or mutated NTRK1 (H498R, G617D and H766R) tagged at the C-terminus with a 6X-histidine sequence. Scale bars represent 10 μm. (c) Focus formation assay in NIH/ 3T3 cells expressing WT or mutated NTRK1 and treated with entrectinib or vehicle (CTRL). Number of foci are from 2 week culture and two biological replicates. Mean and S.D. are shown. (d) Representative histology showing infiltration of the lung and sternal bone marrow by NTRK1H498R/TP53R172H induced leukemia in a primary mouse recipient. Scale bars are 50 μm. This experiment was repeated in an independent mouse with NTRK1H498R/TP53R172H obtaining the same result. (e) Flow cytometry of bone marrow cells harvested from moribund representative primary recipient mice with NTRK1/TP53R172H, NTRK1H498R/TP53R172H, NTRK1G617D/TP53R172H or NTRK1H766R/TP53R172H induced leukemia is shown. Cells were gated based on GFP expression. Leukemia cells from NTRK/TP53 induced tumors were positive for the erythroid markers Ter119 and CD71 and negative for myeloid (Mac1, Gr1), megakaryoblastic (CD41) and lymphoid (B220, CD19, CD3) markers (data not shown). This analysis was performed on all mice in study that developed leukemia obtaining similar results. (f) Unsupervised hierarchical clustering analysis. (g) Kaplan–Meier survival curves and spleen weight from sublethally irradiated C57BL/6 primary recipients transplanted with bone marrow TP53R172H lin- HSPC expressing WT NTRK1 or mutated NTRK1 (G617D or H766R) are shown. The Kaplan–Meier survival curve from WT NTRK1 primary mice is from Fig. 5d. The number of independent mice is reported in each panel. Outcome associations were analyzed with the log-rank test. In the spleen weight graph the mean expression is shown by the horizontal line in the scatter dot plot and the error bars represent the S.D. (h) Hematoxylin and eosin staining and positive IHC labeling for GlyA, Ter119, mCD45, GATA1 and human TRKA in liver from mice with NTRK1/TP53R172H, NTRK1G617D/TP53R172H or NTRK1H766R/TP53R172H. All cells were strongly positive for human TRKA. This experiment was repeated in an independent mouse with NTRK1/TP53R172H obtaining the same result. (i) SKY karyotype of a representative mouse with NTRK1G617D /Tp53R172H mutated erythroid leukemia. The image shows the spectral (RGB) color image on the left and the classified (pseudocolor) chromosome after per-pixel classification of the spectral data on the right. The image has the following karyotype: 44~45,XX,+3[3],i(3)[1],+8[3],cf(8;14)[3],+11[3],+11[2]+15[3],+15[3][cp3]. The number of cells with each alteration (composite karyotype, cp) is shown by providing the number of cells in square brackets after each alteration. Abbreviation: cf: centric fusion. (j) Copy number alteration events detected by whole exome sequencing and showing perfect overlap with data generated from SKY karyotype. In red are copy number gains, in blues copy number losses and white is diploid copy number state. Data are from 11 independent mice. (k) Kaplan–Meier survival curves in mice with NTRK1G617D/TP53R172H induced leukemia treated with larotrectinib (n = 5) or vehicle (n = 5). Outcome associations were analyzed with the log-rank test. In the drug-treated group larotrectinib was stopped after 31 days.

Supplementary Figure 12 Pipelines for sequencing analysis.

(a) Analysis of whole exome sequencing (WES) for diagnosis/relapse samples with paired germline samples. (b) Analysis of WES for diagnosis/relapse samples without paired germline samples. (c) Pipeline for RNA-seq analysis. Abbreviations: SNV, single nucleotide variation; PCGP, Pediatric Cancer Genome Project; NHLBI ESP, NHLBI GO Exome Sequencing Project.

Supplementary information

Supplementary Information

Supplementary Figures 1–12 and Supplementary Note

Reporting Summary

Supplementary Tables

Supplementary Tables 1–39

Supplementary Data

Supplementary Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Iacobucci, I., Wen, J., Meggendorfer, M. et al. Genomic subtyping and therapeutic targeting of acute erythroleukemia. Nat Genet 51, 694–704 (2019). https://doi.org/10.1038/s41588-019-0375-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-019-0375-1

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing