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:

Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia

A Corrigendum to this article was published on 16 April 2014

This article has been updated

Abstract

In acute myeloid leukaemia (AML), the cell of origin, nature and biological consequences of initiating lesions, and order of subsequent mutations remain poorly understood, as AML is typically diagnosed without observation of a pre-leukaemic phase. Here, highly purified haematopoietic stem cells (HSCs), progenitor and mature cell fractions from the blood of AML patients were found to contain recurrent DNMT3A mutations (DNMT3Amut) at high allele frequency, but without coincident NPM1 mutations (NPM1c) present in AML blasts. DNMT3Amut-bearing HSCs showed a multilineage repopulation advantage over non-mutated HSCs in xenografts, establishing their identity as pre-leukaemic HSCs. Pre-leukaemic HSCs were found in remission samples, indicating that they survive chemotherapy. Therefore DNMT3Amut arises early in AML evolution, probably in HSCs, leading to a clonally expanded pool of pre-leukaemic HSCs from which AML evolves. Our findings provide a paradigm for the detection and treatment of pre-leukaemic clones before the acquisition of additional genetic lesions engenders greater therapeutic resistance.

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: Recurrent somatic DNMT3A mutations are common in T cells from AML patients.
Figure 2: DNMT3A mutation precedes NPM1 mutation in human AML and is present in stem/progenitor cells at diagnosis and remission.
Figure 3: Pre-leukaemic HSCs bearing DNMT3Amut generate multilineage engraftment and have a competitive advantage in xenograft repopulation assays.
Figure 4: Identification of pre-leukaemic HSCs with IDH2 mutation.

Similar content being viewed by others

Change history

References

  1. 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 

  2. McCulloch, E. A., Howatson, A. F., Buick, R. N., Minden, M. D. & Izaguirre, C. A. Acute myeloblastic leukemia considered as a clonal hemopathy. Blood Cells 5, 261–282 (1979)

    CAS  PubMed  Google Scholar 

  3. Vogelstein, B., Fearon, E. R., Hamilton, S. R. & Feinberg, A. P. Use of restriction fragment length polymorphisms to determine the clonal origin of human tumors. Science 227, 642–645 (1985)

    Article  ADS  CAS  Google Scholar 

  4. Kandoth, C. et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333–339 (2013)

    Article  ADS  CAS  Google Scholar 

  5. Greaves, M. & Maley, C. C. Clonal evolution in cancer. Nature 481, 306–313 (2012)

    Article  ADS  CAS  Google Scholar 

  6. Yates, L. R. & Campbell, P. J. Evolution of the cancer genome. Nature Rev. Genet. 13, 795–806 (2012)

    Article  CAS  Google Scholar 

  7. Anderson, K. et al. Genetic variegation of clonal architecture and propagating cells in leukaemia. Nature 469, 356–361 (2011)

    Article  ADS  CAS  Google Scholar 

  8. Campbell, P. J. et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467, 1109–1113 (2010)

    Article  ADS  CAS  Google Scholar 

  9. Ding, L. et al. Genome remodelling in a basal-like breast cancer metastasis and xenograft. Nature 464, 999–1005 (2010)

    Article  ADS  CAS  Google Scholar 

  10. Ding, L. et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 481, 506–510 (2012)

    Article  ADS  CAS  Google Scholar 

  11. Notta, F. et al. Evolution of human BCRABL1 lymphoblastic leukaemia-initiating cells. Nature 469, 362–367 (2011)

    Article  ADS  CAS  Google Scholar 

  12. Shah, S. P. et al. Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution. Nature 461, 809–813 (2009)

    Article  ADS  CAS  Google Scholar 

  13. Yachida, S. et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467, 1114–1117 (2010)

    Article  ADS  CAS  Google Scholar 

  14. Mullighan, C. G. et al. Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science 322, 1377–1380 (2008)

    Article  ADS  CAS  Google Scholar 

  15. Shlush, L. I. et al. Cell lineage analysis of acute leukemia relapse uncovers the role of replication-rate heterogeneity and microsatellite instability. Blood 120, 603–612 (2012)

    Article  CAS  Google Scholar 

  16. Sgroi, D. C. Preinvasive breast cancer. Annu. Rev. Pathol. 5, 193–221 (2010)

    Article  CAS  Google Scholar 

  17. Wistuba, I. I., Mao, L. & Gazdar Smoking molecular damage in bronchial epithelium. Oncogene 21, 7298–7306 (2002)

    Article  CAS  Google Scholar 

  18. Balaban, G. B., Herlyn, M., Clark, W. H., Jr & Nowell, P. C. Karyotypic evolution in human malignant melanoma. Cancer Genet. Cytogenet. 19, 113–122 (1986)

    Article  CAS  Google Scholar 

  19. Vogelstein, B. et al. Genetic alterations during colorectal-tumor development. N. Engl. J. Med. 319, 525–532 (1988)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  21. Doulatov, S., Notta, F., Laurenti, E. & Dick, J. E. Hematopoiesis: a human perspective. Cell Stem Cell 10, 120–136 (2012)

    Article  CAS  Google Scholar 

  22. Raza, A. & Galili, N. The genetic basis of phenotypic heterogeneity in myelodysplastic syndromes. Nature Rev. Cancer 12, 849–859 (2012)

    Article  CAS  Google Scholar 

  23. Shih, A. H., Abdel-Wahab, O., Patel, J. P. & Levine, R. L. The role of mutations in epigenetic regulators in myeloid malignancies. Nature Rev. Cancer 12, 599–612 (2012)

    Article  CAS  Google Scholar 

  24. Busque, L. et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nature Genet. 44, 1179–1181 (2012)

    Article  CAS  Google Scholar 

  25. Fialkow, P. J., Gartler, S. M. & Yoshida, A. Clonal origin of chronic myelocytic leukemia in man. Proc. Natl Acad. Sci. USA 58, 1468–1471 (1967)

    Article  ADS  CAS  Google Scholar 

  26. 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 

  27. Miyamoto, T., Weissman, I. L. & Akashi, K. AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc. Natl Acad. Sci. USA 97, 7521–7526 (2000)

    Article  ADS  CAS  Google Scholar 

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

  29. Yan, X. J. et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nature Genet. 43, 309–315 (2011)

    Article  CAS  Google Scholar 

  30. Ley, T. J. et al. DNMT3A mutations in acute myeloid leukemia. N. Engl. J. Med. 363, 2424–2433 (2010)

    Article  CAS  Google Scholar 

  31. 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  Google Scholar 

  32. Krönke, J. et al. Clonal evolution in relapsed NPM1-mutated acute myeloid leukemia. Blood 122, 100–108 (2013)

    Article  Google Scholar 

  33. Doulatov, S. et al. Revised map of the human progenitor hierarchy shows the origin of macrophages and dendritic cells in early lymphoid development. Nature Immunol. 11, 585–593 (2010)

    Article  CAS  Google Scholar 

  34. Notta, F. et al. Isolation of single human hematopoietic stem cells capable of long-term multilineage engraftment. Science 333, 218–221 (2011)

    Article  ADS  CAS  Google Scholar 

  35. Laurenti, E. et al. The transcriptional architecture of early human hematopoiesis identifies multilevel control of lymphoid commitment. Nature Immunol. 14, 756–763 (2013)

    Article  CAS  Google Scholar 

  36. Kim, H. J. et al. Many multipotential gene-marked progenitor or stem cell clones contribute to hematopoiesis in nonhuman primates. Blood 96, 1–8 (2000)

    CAS  PubMed  Google Scholar 

  37. Fialkow, P. J., Janssen, J. W. & Bartram, C. R. Clonal remissions in acute nonlymphocytic leukemia: evidence for a multistep pathogenesis of the malignancy. Blood 77, 1415–1417 (1991)

    CAS  PubMed  Google Scholar 

  38. Eppert, K. et al. Stem cell gene expression programs influence clinical outcome in human leukemia. Nature Med. 17, 1086–1093 (2011)

    Article  CAS  Google Scholar 

  39. Jankowska, A. M. et al. Mutational spectrum analysis of chronic myelomonocytic leukemia includes genes associated with epigenetic regulation: UTX, EZH2, and DNMT3A. Blood 118, 3932–3941 (2011)

    Article  CAS  Google Scholar 

  40. Kikushige, Y. et al. Self-renewing hematopoietic stem cell is the primary target in pathogenesis of human chronic lymphocytic leukemia. Cancer Cell 20, 246–259 (2011)

    Article  CAS  Google Scholar 

  41. Walter, M. J. et al. Recurrent DNMT3A mutations in patients with myelodysplastic syndromes. Leukemia 25, 1153–1158 (2011)

    Article  CAS  Google Scholar 

  42. Chan, S. M. & Majeti, R. Role of DNMT3A, TET2, and IDH1/2 mutations in pre-leukemic stem cells in acute myeloid leukemia. Int. J. Hematol. 98, 648–657 (2013)

    Article  CAS  Google Scholar 

  43. Challen, G. A. et al. Dnmt3a is essential for hematopoietic stem cell differentiation. Nature Genet. 44, 23–31 (2012)

    Article  CAS  Google Scholar 

  44. Tadokoro, Y., Ema, H., Okano, M., Li, E. & Nakauchi, H. De novo DNA methyltransferase is essential for self-renewal, but not for differentiation, in hematopoietic stem cells. J. Exp. Med. 204, 715–722 (2007)

    Article  CAS  Google Scholar 

  45. Kim, S. J. et al. A DNMT3A mutation common in AML exhibits dominant-negative effects in murine ES cells. Blood 122, 4086–4089 (2013)

    Article  CAS  Google Scholar 

  46. Clappier, E. et al. Clonal selection in xenografted human T cell acute lymphoblastic leukemia recapitulates gain of malignancy at relapse. J. Exp. Med. 208, 653–661 (2011)

    Article  CAS  Google Scholar 

  47. Inaba, H., Greaves, M. & Mullighan, C. G. Acute lymphoblastic leukaemia. Lancet 381, 1943–1955 (2013)

    Article  Google Scholar 

  48. Yasuda, T. et al. Leukemic evolution of donor-derived cells harboring IDH2 and DNMT3A mutations after allogeneic stem cell transplantation. Leukemia http://dx.doi.org/10.1038/leu.2013.278 (15 October 2013)

  49. Hu, Y. & Smyth, G. K. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J. Immunol. Methods 347, 70–78 (2009)

    Article  CAS  Google Scholar 

  50. Kottaridis, P. D. et al. Studies of FLT3 mutations in paired presentation and relapse samples from patients with acute myeloid leukemia: implications for the role of FLT3 mutations in leukemogenesis, minimal residual disease detection, and possible therapy with FLT3 inhibitors. Blood 100, 2393–2398 (2002)

    Article  CAS  Google Scholar 

  51. Heinrich, V. et al. The allele distribution in next-generation sequencing data sets is accurately described as the result of a stochastic branching process. Nucleic Acids Res. 40, 2426–2431 (2012)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank all members of the Dick laboratory for critical assessment of this work, A. Khandani, P. Penttilä, N. Simard, T. Velauthapillai and the SickKids-UHN flow facility for technical support, J. Claudio for management of the HALT studies that enabled the genetic analysis described herein, and J. Cui and X.-Z. Yang for curating the human AML samples used in these studies. This work was supported by a Postdoctoral Fellowship Award from the McEwen Centre for Regenerative Medicine with funding made available through the Gentle Ben Charity (L.I.S.), a Canadian Institutes for Health Research (CIHR) fellowship in partnership with the Aplastic Anemia and Myelodysplasia Association of Canada and an award from Vetenskapsradet (S.Z.), and by grants from CIHR, Canadian Cancer Society, Terry Fox Foundation, Genome Canada through the Ontario Genomics Institute, Ontario Institute for Cancer Research with funds from the province of Ontario, a Canada Research Chair, and the Ontario Ministry of Health and Long Term Care (OMOHLTC). The views expressed do not necessarily reflect those of the OMOHLTC. This work was also supported by the Cancer Stem Cell Consortium with funding from the Government of Canada through Genome Canada and the Ontario Genomics Institute (OGI-047), and through the Canadian Institutes of Health Research (CSC-105367). Contributors to the HALT Pan-Leukemia Gene Panel are listed in Supplementary Note 1.

Author information

Authors and Affiliations

Authors

Contributions

L.I.S. and S.Z. designed and performed experiments, analysed data and wrote the manuscript; A.M., W.C.C screened AML engraftment in xenotransplantation assays; J.M.B., V.G., J.A.K., A.D.S., A.C.S., K.W.Y., M.D.M. collected AML samples and assembled clinical information; J.A.K. correlated xenotransplantation engraftment data with clinical information; J.L.M., M.D. performed xenotransplantation experiments; J.J.F.M., R.M. performed ddPCR; H.J.K., K.L. performed Sanger sequencing; J.D.M., T.J.H., supervised the targeted sequencing; A.M.K.B. and F.Y. performed and analysed targeted sequencing; Q.M.T., L.D.S. performed DNMT3A data mining. M.D.M. designed the study; J.C.Y.W. supervised AML xenotransplantation screening experiments, designed the study and wrote the manuscript; J.E.D. supervised the study and wrote the manuscript.

Corresponding author

Correspondence to John E. Dick.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 FLT3-ITD is a late event in patients carrying DNMT3A mutation.

PCR analysis of FLT3-ITD50 in stem/progenitor, mature lymphoid and blast (CD45dim CD33+) cell populations from patient no. 13 (a) and no. 14 (b). FLT3-ITD was present in the blasts from both patients, and also in MLPs from patient no. 14. In contrast, DNMT3Amut without FLT3-ITD was detected in multiple non-blast cell populations (see Extended Data Fig. 2). HSC, haematopoietic stem cell; MPP, multipotent progenitor; CMP, common myeloid progenitor; MLP, multilymphoid progenitor; GMP, granulocyte monocyte progenitor; NK, natural killer cells.

Extended Data Figure 2 Frequent occurrence of DNMT3A mutation without NPM1 mutation in stem/progenitor and mature lymphoid cells in AML patients at diagnosis.

a, Summary of the allele frequency (%) of DNMT3A and NPM1 mutations in stem/progenitor, mature lymphoid, and blast (CD45dim CD33+) cell populations from 11 AML patient peripheral blood samples obtained at diagnosis, as determined by droplet digital PCR (ddPCR). Phenotypically normal cell populations were isolated by fluorescence activated cell sorting according to the strategy depicted in Fig. 2a. Mutant allele frequency 50% is consistent with a heterozygous cell population. Departures from 50% mutant allele frequency may be stochastic51, related to clonal heterogeneity, or due to the presence of copy number variations, for example loss of the wild type allele (loss of heterozygosity) or amplification of the mutant allele. NA, no cell population detected; HSC, haematopoietic stem cell; MPP, multipotent progenitor; CMP, common myeloid progenitor; MEP, megakaryocyte erythroid progenitor; MLP, multilymphoid progenitor; GMP, granulocyte monocyte progenitor; NK, natural killer cells. Blank boxes indicate no DNMT3A or NPM1 mutation detected. b, Representative plots showing ddPCR analysis of DNMT3Amut and NPM1c allele frequency in sorted cell populations from patient no. 11. The mutant allele frequency (%) is indicated on each plot.

Extended Data Figure 3 Phenotypically normal stem/progenitor and mature cell populations are present in AML patient samples at diagnosis, remission and relapse.

Flow cytometric analysis showing the gating strategy used to isolate phenotypically normal stem/progenitor and mature lymphoid cell populations from AML patient samples. Diagnosis and relapse samples are from peripheral blood; remission samples are from bone marrow.

Extended Data Figure 4 Cells bearing mutations in DNMT3A but not NPM1 are present at diagnosis in AML patients and persist at remission and relapse.

Allele frequency of DNMT3A and NPM1 mutations of patients no. 28, 35, 55, and 57 in stem/progenitor, mature and blast (CD45dim CD33+) cell populations, as determined by droplet digital PCR (ddPCR). Cells were isolated from diagnosis (blue), early remission (white), relapse (red) or late remission (yellow) samples. At remission, CD33+ myeloid cells were also analysed. HSC, haematopoietic stem cell; MPP, multipotent progenitor; MLP, multilymphoid progenitor; CMP, common myeloid progenitor; GMP, granulocyte monocyte progenitor; MEP, megakaryocyte erythroid progenitor; NK, natural killer cells.

Extended Data Figure 5 PreL-HSCs in the peripheral blood of AML patients generate multilineage human grafts in immunodeficient mice.

Summary of results of limiting dilution experiments to assess frequency of pre-leukaemic HSCs generating multilineage grafts after xenotransplantation. Cohorts of NSG mice were transplanted intrafemorally with varying numbers of peripheral blood mononuclear cells from diagnostic samples of AML patient no. 11 (a) and no. 55 (b) and analysed after 8 or 16 weeks by flow cytometry. Engraftment was defined as >0.1% human CD45+ cells in the injected right femur. Shown is the number of mice with multilineage human grafts containing both CD33+ myeloid cells and CD33CD19+ cells. The frequency of pre-leukaemic HSCs was calculated using the ELDA platform49.

Extended Data Figure 6 Frequent generation of non-leukaemic multilineage human grafts following xenotransplantation of peripheral blood cells from AML patients.

Summary of xenograft characteristics in 123 sublethally irradiated NSG mice transplanted intrafemorally with mononuclear peripheral blood cells from 20 AML patients at diagnosis and analysed after 8 weeks by flow cytometry. The proportion of myeloid (CD33+) and B-lymphoid (CD33CD19+) cells in the human (CD45+) graft is shown. Leukaemic (AML) engraftment is characterized by a dominant myeloid (CD45dimCD33+) graft, whereas non-leukaemic multilineage grafts contain both lymphoid (predominantly CD33CD19+ B cells) and myeloid (CD33+) cells. No leukaemic or multilineage graft could be detected in 65/123 mice (53%) in this cohort. Red box indicates AML grafts (27 mice, 22%); blue box indicates multilineage grafts (31 mice, 25%).

Supplementary information

Supplementary Information

This file contains the legends for Supplementary Tables 1-5 (see separate excel files) and Supplementary Notes 1-2. (PDF 163 kb)

Supplementary Table 1

This file contains Supplementary Table 1 (see Supplementary Information file for legend). (XLSX 350 kb)

Supplementary Table 2

This file contains Supplementary Table 2 (see Supplementary Information file for legend). (XLSX 12 kb)

Supplementary Table 3

This file contains Supplementary Table 3 (see Supplementary Information file for legend). (XLSX 14 kb)

Supplementary Table 4

This file contains Supplementary Table 4 (see Supplementary Information file for legend). (XLSX 26 kb)

Supplementary Table 5

This file contains Supplementary Table 5 (see Supplementary Information file for legend). (XLSX 10 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Shlush, L., Zandi, S., Mitchell, A. et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 506, 328–333 (2014). https://doi.org/10.1038/nature13038

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13038

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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