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.

  • Letter
  • Published:

Myelodysplastic syndrome progression to acute myeloid leukemia at the stem cell level

A Publisher Correction to this article was published on 19 December 2018

This article has been updated

Abstract

Myelodysplastic syndromes (MDS) frequently progress to acute myeloid leukemia (AML); however, the cells leading to malignant transformation have not been directly elucidated. As progression of MDS to AML in humans provides a biological system to determine the cellular origins and mechanisms of neoplastic transformation, we studied highly fractionated stem cell populations in longitudinal samples of patients with MDS who progressed to AML. Targeted deep sequencing combined with single-cell sequencing of sorted cell populations revealed that stem cells at the MDS stage, including immunophenotypically and functionally defined pre-MDS stem cells (pre-MDS-SC), had a significantly higher subclonal complexity compared to blast cells and contained a large number of aging-related variants. Single-cell targeted resequencing of highly fractionated stem cells revealed a pattern of nonlinear, parallel clonal evolution, with distinct subclones within pre-MDS-SC and MDS-SC contributing to generation of MDS blasts or progression to AML, respectively. Furthermore, phenotypically aberrant stem cell clones expanded during transformation and stem cell subclones that were not detectable in MDS blasts became dominant upon AML progression. These results reveal a crucial role of diverse stem cell compartments during MDS progression to AML and have implications for current bulk cell–focused precision oncology approaches, both in MDS and possibly other cancers that evolve from premalignant conditions, that may miss pre-existing rare aberrant stem cells that drive disease progression and leukemic transformation.

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: Higher subclonal diversity at the stem cell level than in blasts in patients with MDS and sAML.
Fig. 2: Schematic models of subclonal evolution of stem cell and blast populations during progression from MDS to sAML.
Fig. 3: Spatiotemporal subclonal evolution during the progression from MDS to sAML determined by single-cell sequencing of sorted stem and blast cells.
Fig. 4: Proposed model of subclonal evolution of stem cells during the progression of MDS to sAML.

Similar content being viewed by others

Data availability

The high-throughput DNA sequencing data have been deposited in the database of Genotypes and Phenotypes (dbGaP) .

Change history

  • 19 December 2018

    In the version of this article originally published, Ulrich Steidl’s name was listed as “and Ulrich Steidl.” His name has been updated to “Ulrich Steidl.” The error has been fixed in the print, PDF and HTML versions of this article.

References

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

    Article  CAS  Google Scholar 

  2. Ades, L., Itzykson, R. & Fenaux, P. Myelodysplastic syndromes. Lancet 383, 2239–2252 (2014).

    Article  Google Scholar 

  3. Fialkow, P. J. et al. Clonal development, stem-cell differentiation, and clinical remissions in acute nonlymphocytic leukemia. N. Engl. J.Med. 317, 468–473 (1987).

    Article  CAS  Google Scholar 

  4. Nilsson, L. et al. Involvement and functional impairment of the CD34+CD38-Thy-1+ hematopoietic stem cell pool in myelodysplastic syndromes with trisomy 8. Blood 100, 259–267 (2002).

    CAS  PubMed  Google Scholar 

  5. Steidl, U. et al. Essential role of Jun family transcription factors in PU.1 knockdown-induced leukemic stem cells. Nat. Genet. 38, 1269–1277 (2006).

    Article  CAS  Google Scholar 

  6. Will, B. et al. Stem and progenitor cells in myelodysplastic syndromes show aberrant stage-specific expansion and harbor genetic and epigenetic alterations. Blood 120, 2076–2086 (2012).

    Article  CAS  Google Scholar 

  7. Jan, M. et al. Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia. Sci. Transl. Med. 4, 149ra118 (2012).

    Article  Google Scholar 

  8. Pang, W. W. et al. Hematopoietic stem cell and progenitor cell mechanisms in myelodysplastic syndromes. Proc. Natl Acad. Sci. USA 110, 3011–3016 (2013).

    Article  CAS  Google Scholar 

  9. Corces-Zimmerman, M. R., Hong, W. J., Weissman, I. L., Medeiros, B. C. & Majeti, R. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc. Natl Acad. Sci. USA 111, 2548–2553 (2014).

    Article  CAS  Google Scholar 

  10. Shlush, L. I. et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 506, 328–333 (2014).

    Article  CAS  Google Scholar 

  11. Will, B. et al. Minimal PU.1 reduction induces a preleukemic state and promotes development of acute myeloid leukemia. Nat. Med. 21, 1172–1181 (2015).

    Article  CAS  Google Scholar 

  12. Walter, M. J. et al. Clonal architecture of secondary acute myeloid leukemia. N. Engl. J. Med. 366, 1090–1098 (2012).

    Article  CAS  Google Scholar 

  13. Walter, M. J. et al. Clonal diversity of recurrently mutated genes in myelodysplastic syndromes. Leukemia 27, 1275–1282 (2013).

    Article  CAS  Google Scholar 

  14. Makishima, H. et al. Dynamics of clonal evolution in myelodysplastic syndromes. Nat. Genet. 49, 204–212 (2017).

    Article  CAS  Google Scholar 

  15. Goardon, N. et al. Coexistence of LMPP-like and GMP-like leukemia stem cells in acute myeloid leukemia. Cancer Cell 19, 138–152 (2011).

    Article  CAS  Google Scholar 

  16. Jordan, C. et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia 14, 1777 (2000).

    Article  CAS  Google Scholar 

  17. Barreyro, L. et al. Overexpression of IL-1 receptor accessory protein in stem and progenitor cells and outcome correlation in AML and MDS. Blood 120, 1290–1298 (2012).

    Article  CAS  Google Scholar 

  18. Mitchell, K. et al. IL1RAP potentiates multiple oncogenic signaling pathways in AML. J. Exp. Med. 215, 1709–1727 (2018).

    Article  CAS  Google Scholar 

  19. Jan, M. et al. Prospective separation of normal and leukemic stem cells based on differential expression of TIM3, a human acute myeloid leukemia stem cell marker. Proc. Natl Acad. Sci. USA 108, 5009–5014 (2011).

    Article  CAS  Google Scholar 

  20. Chung, S. S. et al. CD99 is a therapeutic target on disease stem cells in myeloid malignancies. Sci.Transl. Med. 9, eaaj2025 (2017).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  22. McGranahan, N. et al. Clonal status of actionable driver events and the timing of mutational processes in cancer evolution. Sci. Transl. Med. 7, 283ra254 (2015).

    Article  Google Scholar 

  23. Blokzijl, F. et al. Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538, 260–264 (2016).

    Article  CAS  Google Scholar 

  24. Adams, P. D., Jasper, H. & Rudolph, K. L. Aging-induced stem cell mutations as drivers for disease and cancer. Cell. Stem. Cell. 16, 601–612 (2015).

    Article  CAS  Google Scholar 

  25. Rossi, D. J. et al. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447, 725–729 (2007).

    Article  CAS  Google Scholar 

  26. Mandal, P. K., Blanpain, C. & Rossi, D. J. DNA damage response in adult stem cells: pathways and consequences. Nat. Rev. Mol. Cell Biol. 12, 198–202 (2011).

    Article  CAS  Google Scholar 

  27. Mohrin, M. et al. Hematopoietic stem cell quiescence promotes error-prone DNA repair and mutagenesis. Cell. Stem. Cell. 7, 174–185 (2010).

    Article  CAS  Google Scholar 

  28. Miller, C. A. et al. SciClone: inferring clonal architecture and tracking the spatial and temporal patterns of tumor evolution. PLoS Comput. Biol. 10, e1003665 (2014).

    Article  Google Scholar 

  29. Yanagisawa, B., Ghiaur, G., Smith, B. D. & Jones, R. J. Translating leukemia stem cells into the clinical setting: harmonizing the heterogeneity. Exp. Hematol. 44, 1130–1137 (2016).

    Article  CAS  Google Scholar 

  30. Haferlach, T. et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 28, 241–247 (2014).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

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

    Article  Google Scholar 

  35. Arends, C. M. et al. Hematopoietic lineage distribution and evolutionary dynamics of clonal hematopoiesis. Leukemia 32, 1908–1919 (2018).

    Article  CAS  Google Scholar 

  36. Abelson, S. et al. Prediction of acute myeloid leukaemia risk in healthy individuals. Nature 559, 400–404 (2018).

    Article  CAS  Google Scholar 

  37. Desai, P. et al. Somatic mutations precede acute myeloid leukemia years before diagnosis. Nat. Med. 24, 1015 (2018).

    Article  CAS  Google Scholar 

  38. Herbaux, C. et al. Incidence of ATRX mutations in myelodysplastic syndromes, the value of microcytosis. Am. J. Hematol. 90, 737–738 (2015).

    Article  CAS  Google Scholar 

  39. Steensma, D. P., Higgs, D. R., Fisher, C. A. & Gibbons, R. J. Acquired somatic ATRX mutations in myelodysplastic syndrome associated with alpha thalassemia (ATMDS) convey a more severe hematologic phenotype than germline ATRX mutations. Blood 103, 2019–2026 (2004).

    Article  CAS  Google Scholar 

  40. Bacher, U., Haferlach, T., Kern, W., Haferlach, C. & Schnittger, S. A comparative study of molecular mutations in 381 patients with myelodysplastic syndrome and in 4130 patients with acute myeloid leukemia. Haematologica 92, 744–752 (2007).

    Article  CAS  Google Scholar 

  41. Shastri, A., Will, B., Steidl, U. & Verma, A. Stem and progenitor cell alterations in myelodysplastic syndromes. Blood 129, 1586–1594 (2017).

    Article  CAS  Google Scholar 

  42. Thomas, D. & Majeti, R. Biology and relevance of human acute myeloid leukemia stem cells. Blood 129, 1577–1585 (2017).

    Article  CAS  Google Scholar 

  43. McIntosh, B. E. et al. Nonirradiated NOD,B6.SCID Il2rγ-/- Kit(W41/W41) (NBSGW) mice support multilineage engraftment of human hematopoietic cells. Stem Cell Reports 4, 171–180 (2015).

    Article  CAS  Google Scholar 

  44. Woll, P. S. et al. Myelodysplastic syndromes are propagated by rare and distinct human cancer stem cells in vivo. Cancer Cell. 25, 794–808 (2014).

    Article  CAS  Google Scholar 

  45. Terwijn, M. et al. Leukemic stem cell frequency: a strong biomarker for clinical outcome in acute myeloid leukemia. PLoS ONE 9, e107587 (2014).

    Article  Google Scholar 

  46. Wang, C. et al. Non-lethal ionizing radiation promotes aging-like phenotypic changes of human hematopoietic stem and progenitor cells in humanized mice. PLoS ONE 10, e0132041 (2015).

    Article  Google Scholar 

  47. Lu, R., Czechowicz, A., Seita, J., Jiang, D. & Weissman, I. L. Clonal level lineage commitment pathways of hematopoietic stem cells in vivo. Preprint at https://doi.org/10.1101/262774 (2018).

  48. Hosono, S. et al. Unbiased whole-genome amplification directly from clinical samples. Genome Res. 13, 954–964 (2003).

    Article  CAS  Google Scholar 

  49. de Bourcy, C. F. et al. A quantitative comparison of single-cell whole genome amplification methods. PLoS ONE 9, e105585 (2014).

    Article  Google Scholar 

  50. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  Google Scholar 

  51. McKenna, A. et al. The genome analysis toolkit: a mapreduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  Google Scholar 

  52. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).

    Article  CAS  Google Scholar 

  53. Garrison, E. & Marth, G. Haplotype-based variant detection from short-read sequencing. Preprint at https://arxiv.org/abs/1207.3907 (2012).

  54. Cingolani, P. et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w(1118); iso-2; iso-3. Fly 6, 80–92 (2012).

    Article  CAS  Google Scholar 

  55. Rosenthal, R., McGranahan, N., Herrero, J., Taylor, B. S. & Swanton, C. DeconstructSigs: delineating mutational processes in single tumors distinguishes DNA repair deficiencies and patterns of carcinoma evolution. Genome. Biol. 17, 31 (2016).

    Article  Google Scholar 

  56. Shen, R. & Seshan, V. E. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 44, e131 (2016).

    Article  Google Scholar 

  57. Popic, V. et al. Fast and scalable inference of multi-sample cancer lineages. Genome. Biol. 16, 91 (2015).

    Article  Google Scholar 

  58. Smith, M.A. et al. E-scape: interactive visualization of single-cell phylogenetics and cancer evolution. Nat. Methods 14, 549–550 (2017).

    Article  CAS  Google Scholar 

  59. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. Preprint at https://arxiv.org/abs/1303.3997 (2013).

  60. Yu, Y., Ceredig, R. & Seoighe, C. LymAnalyzer: a tool for comprehensive analysis of next generation sequencing data of T cell receptors and immunoglobulins. Nucleic Acids Res. 44, e31 (2016).

    Article  Google Scholar 

Download references

Acknowledgements

We thank P. Schultes from the Department of Cell Biology for expert technical assistance. We thank A. Fiallo from the Einstein Genomics Core Facility for technical assistance in single-cell targeted sequencing, and S. Maqbool and S. Mi from Einstein Epigenomics Core Facility for assistance in targeted sequencing with the HiSeq platform. We thank V. Thiruthuvanathan from the Department of Cell Biology for assistance in processing the patient samples. We also thank W. Li for advice regarding whole-genome amplification, and F. C. Chan, C. Steidl, and H. Steidl for helpful discussion. This work was supported by NIH grants no. R01CA166429, no. R01CA217092 (to U.S.), no. R01HL139487, no. R01DK103961 (to A.V.), and no. K01DK105134 (to B.W.); Translational Research Program grants from the Leukemia & Lymphoma Society (to U.S. and A.V., respectively); a research grant from the Taub Foundation for MDS Research (to U.S.); and a research grant from the Evans Foundation (to A.V.). J.C. was supported by The Einstein Training Program in Stem Cell Research from the Empire State Stem Cell Fund through New York State Department of Health Contract (no. C30292GG). U.S. is a Research Scholar of the Leukemia and Lymphoma Society and the Diane and Arthur B. Belfer Faculty Scholar in Cancer Research of the Albert Einstein College of Medicine. This work was supported through the Albert Einstein Cancer Center core support grant (no. P30CA013330).

Author information

Authors and Affiliations

Authors

Contributions

J.C., U.S., and A.V. designed the study and analyzed and interpreted data. J.C., Y.K., and T.I.T. collected and analyzed clinical samples. J.C., Y.K., D.S., S.N., and B.W. performed the FACS experiments. J.C. and S.N. performed the xenotransplantation assays. J.C. performed the methylcellulose assay and TCR sequencing. J.C. and D.R. performed single-cell targeted sequencing. C.M., A.V., and U.S. designed the targeted capture panel. J.C. analyzed the sequencing data. J.C., A.V., and U.S. wrote the manuscript. All authors reviewed and approved the final version of the manuscript.

Corresponding authors

Correspondence to Amit Verma or Ulrich Steidl.

Ethics declarations

Competing interests

U.S. has received research funding from GlaxoSmithKline, Bayer Healthcare, Aileron Therapeutics, and Novartis; has received compensation for consultancy services and for serving on scientific advisory boards from GlaxoSmithKline, Bayer Healthcare, Celgene, Aileron Therapeutics, Stelexis Therapeutics, and Pieris Pharmaceuticals; and has equity ownership in and is serving on the board of directors of Stelexis Therapeutics. A.V. has received research funding from GlaxoSmithKline, Incyte, MedPacto, Novartis, and Eli Lilly and Company, has received compensation as a scientific advisor to Novartis, Stelexis Therapeutics, Acceleron Pharma, and Celgene, and has equity ownership in Stelexis Therapeutics. B.W. has received research support from Novartis Pharmaceuticals.

Additional information

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

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15

Reporting Summary

Supplementary Table 1

Patient characteristics

Supplementary Table 2

Genes for targeted capture sequencing

Supplementary Table 3

Somatic mutations detected by targeted capture sequencing in each patient

Supplementary Table 4

Antibodies for FACS experiments

Supplementary Table 5

Primers for single-cell targeted sequencing with Fluidigm platform

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, J., Kao, YR., Sun, D. et al. Myelodysplastic syndrome progression to acute myeloid leukemia at the stem cell level. Nat Med 25, 103–110 (2019). https://doi.org/10.1038/s41591-018-0267-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0267-4

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