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 genomic landscape of hypodiploid acute lymphoblastic leukemia

Abstract

The genetic basis of hypodiploid acute lymphoblastic leukemia (ALL), a subtype of ALL characterized by aneuploidy and poor outcome, is unknown. Genomic profiling of 124 hypodiploid ALL cases, including whole-genome and exome sequencing of 40 cases, identified two subtypes that differ in the severity of aneuploidy, transcriptional profiles and submicroscopic genetic alterations. Near-haploid ALL with 24–31 chromosomes harbor alterations targeting receptor tyrosine kinase signaling and Ras signaling (71%) and the lymphoid transcription factor gene IKZF3 (encoding AIOLOS; 13%). In contrast, low-hypodiploid ALL with 32–39 chromosomes are characterized by alterations in TP53 (91.2%) that are commonly present in nontumor cells, IKZF2 (encoding HELIOS; 53%) and RB1 (41%). Both near-haploid and low-hypodiploid leukemic cells show activation of Ras-signaling and phosphoinositide 3-kinase (PI3K)-signaling pathways and are sensitive to PI3K inhibitors, indicating that these drugs should be explored as a new therapeutic strategy for this aggressive form of 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

Figure 1: Mutation spectrum of whole genome–sequenced hypodiploid ALL.
Figure 2: Genome-wide DNA copy-number alterations and gene expression profiling of hypodiploid ALL.
Figure 3: Recurrent alterations of Ras and RTK signaling in near-haploid ALL.
Figure 4: Frequent mutations in TP53 in low-hypodiploid ALL.
Figure 5: Recurrent deletions of the IKAROS-family genes IKZF2 (HELIOS) and IKZF3 (AIOLOS).
Figure 6: Recurring mutations in hypodiploid ALL.
Figure 7: Activation of Ras signaling.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Pui, C.H. et al. Treating childhood acute lymphoblastic leukemia without cranial irradiation. N. Engl. J. Med. 360, 2730–2741 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Kuiper, R.P. et al. High-resolution genomic profiling of childhood ALL reveals novel recurrent genetic lesions affecting pathways involved in lymphocyte differentiation and cell cycle progression. Leukemia 21, 1258–1266 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  4. Kuiper, R.P. et al. IKZF1 deletions predict relapse in uniformly treated pediatric precursor B-ALL. Leukemia 24, 1258–1264 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mullighan, C.G. et al. CREBBP mutations in relapsed acute lymphoblastic leukaemia. Nature 471, 235–239 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Harrison, C.J. et al. Three distinct subgroups of hypodiploidy in acute lymphoblastic leukaemia. Br. J. Haematol. 125, 552–559 (2004).

    Article  PubMed  Google Scholar 

  8. Heerema, N.A. et al. Hypodiploidy with less than 45 chromosomes confers adverse risk in childhood acute lymphoblastic leukemia: a report from the children's cancer group. Blood 94, 4036–4045 (1999).

    CAS  PubMed  Google Scholar 

  9. Nachman, J.B. et al. Outcome of treatment in children with hypodiploid acute lymphoblastic leukemia. Blood 110, 1112–1115 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Raimondi, S.C. et al. Reassessment of the prognostic significance of hypodiploidy in pediatric patients with acute lymphoblastic leukemia. Cancer 98, 2715–2722 (2003).

    Article  PubMed  Google Scholar 

  11. Raimondi, S.C. Cytogenetics of acute leukemias. in Childhood Leukemias (ed. Pui, C.H.) (Cambridge University Press, 2012).

  12. Carroll, A.J. et al. Masked hypodiploidy: hypodiploid acute lymphoblastic leukemia (ALL). in Children mimicking hyperdiploid ALL: a report from the Children's Oncology Group (COG) AALL03B1 study. Blood: ASH Annual Meeting Abstracts 114, 1580 (2009).

    Article  Google Scholar 

  13. Li, F.P. & Fraumeni, J.F. Jr. Soft-tissue sarcomas, breast cancer, and other neoplasms. A familial syndrome? Ann. Intern. Med. 71, 747–752 (1969).

    Article  CAS  PubMed  Google Scholar 

  14. Malkin, D. Li-Fraumeni syndrome. Genes Cancer 2, 475–484 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Xu, G.F. et al. The neurofibromatosis type 1 gene encodes a protein related to GAP. Cell 62, 599–608 (1990).

    Article  CAS  PubMed  Google Scholar 

  17. Cawthon, R.M. et al. A major segment of the neurofibromatosis type 1 gene: cDNA sequence, genomic structure, and point mutations. Cell 62, 193–201 (1990).

    Article  CAS  PubMed  Google Scholar 

  18. Viskochil, D. et al. Deletions and a translocation interrupt a cloned gene at the neurofibromatosis type 1 locus. Cell 62, 187–192 (1990).

    Article  CAS  PubMed  Google Scholar 

  19. Wallace, M.R. et al. Type 1 neurofibromatosis gene: identification of a large transcript disrupted in three NF1 patients. Science 249, 181–186 (1990).

    Article  CAS  PubMed  Google Scholar 

  20. Side, L.E. et al. Mutations of the NF1 gene in children with juvenile myelomonocytic leukemia without clinical evidence of neurofibromatosis, type 1. Blood 92, 267–272 (1998).

    Article  CAS  PubMed  Google Scholar 

  21. Balgobind, B.V. et al. Leukemia-associated NF1 inactivation in patients with pediatric T-ALL and AML lacking evidence for neurofibromatosis. Blood 111, 4322–4328 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. McLendon, R. et al. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008).

    Article  CAS  Google Scholar 

  23. Klopfenstein, K.J., Sommer, A. & Ruymann, F.B. Neurofibromatosis-Noonan syndrome and acute lymphoblastic leukemia: a report of two cases. J. Pediatr. Hematol. Oncol. 21, 158–160 (1999).

    Article  CAS  PubMed  Google Scholar 

  24. Stiller, C.A., Chessells, J.M. & Fitchett, M. Neurofibromatosis and childhood leukaemia/lymphoma: a population-based UKCCSG study. Br. J. Cancer 70, 969–972 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Messiaen, L. et al. Mosaic type-1 NF1 microdeletions as a cause of both generalized and segmental neurofibromatosis type-1 (NF1). Hum. Mutat. 32, 213–219 (2011).

    Article  CAS  PubMed  Google Scholar 

  26. Marculescu, R. et al. Recombinase, chromosomal translocations and lymphoid neoplasia: targeting mistakes and repair failures. DNA Repair (Amst.) 5, 1246–1258 (2006).

    Article  CAS  Google Scholar 

  27. Mullighan, C.G. et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature 453, 110–114 (2008).

    Article  CAS  PubMed  Google Scholar 

  28. Kohno, S., Minowada, J. & Sandberg, A.A. Chromosome evolution of near-haploid clones in an established human acute lymphoblastic leukemia cell line (NALM-16). J. Natl. Cancer Inst. 64, 485–493 (1980).

    CAS  PubMed  Google Scholar 

  29. Kratz, C.P. et al. The mutational spectrum of PTPN11 in juvenile myelomonocytic leukemia and Noonan syndrome/myeloproliferative disease. Blood 106, 2183–2185 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tartaglia, M. et al. Diversity and functional consequences of germline and somatic PTPN11 mutations in human disease. Am. J. Hum. Genet. 78, 279–290 (2006).

    Article  CAS  PubMed  Google Scholar 

  31. De Filippi, P. et al. Germ-line mutation of the NRAS gene may be responsible for the development of juvenile myelomonocytic leukaemia. Br. J. Haematol. 147, 706–709 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Matsuda, K. et al. Spontaneous improvement of hematologic abnormalities in patients having juvenile myelomonocytic leukemia with specific RAS mutations. Blood 109, 5477–5480 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Kawabuchi, M. et al. Transmembrane phosphoprotein Cbp regulates the activities of Src-family tyrosine kinases. Nature 404, 999–1003 (2000).

    Article  CAS  PubMed  Google Scholar 

  34. Brdicka, T. et al. Phosphoprotein associated with glycosphingolipid-enriched microdomains (PAG), a novel ubiquitously expressed transmembrane adaptor protein, binds the protein tyrosine kinase csk and is involved in regulation of T cell activation. J. Exp. Med. 191, 1591–1604 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Davidson, D., Bakinowski, M., Thomas, M.L., Horejsi, V. & Veillette, A. Phosphorylation-dependent regulation of T-cell activation by PAG/Cbp, a lipid raft-associated transmembrane adaptor. Mol. Cell. Biol. 23, 2017–2028 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhang, S.Q. et al. Shp2 regulates SRC family kinase activity and Ras/Erk activation by controlling Csk recruitment. Mol. Cell 13, 341–355 (2004).

    Article  PubMed  Google Scholar 

  37. Kalland, M.E., Solheim, S.A., Skanland, S.S., Tasken, K. & Berge, T. Modulation of proximal signaling in normal and transformed B cells by transmembrane adapter Cbp/PAG. Exp. Cell Res. 318, 1611–1619 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. Imamura, J., Miyoshi, I. & Koeffler, H.P. p53 in hematologic malignancies. Blood 84, 2412–2421 (1994).

    CAS  PubMed  Google Scholar 

  39. Zhang, J. et al. Key pathways are frequently mutated in high-risk childhood acute lymphoblastic leukemia: a report from the Children's Oncology Group. Blood 118, 3080–3087 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Petitjean, A. et al. Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: Lessons from recent developments in the IARC TP53 database. Hum. Mutat. 6, 622–629 (2007).

    Article  CAS  Google Scholar 

  41. Cho, Y., Gorina, S., Jeffrey, P.D. & Pavletich, N.P. Crystal structure of a p53 tumor suppressor-DNA complex: understanding tumorigenic mutations. Science 265, 346–355 (1994).

    Article  CAS  PubMed  Google Scholar 

  42. Hof, J. et al. Mutations and deletions of the TP53 gene predict nonresponse to treatment and poor outcome in first relapse of childhood acute lymphoblastic leukemia. J. Clin. Oncol. 29, 3185–93 (2011).

    Article  PubMed  Google Scholar 

  43. Wattel, E. et al. p53 mutations are associated with resistance to chemotherapy and short survival in hematologic malignancies. Blood 84, 3148–3157 (1994).

    CAS  PubMed  Google Scholar 

  44. Rovinski, B. & Benchimol, S. Immortalization of rat embryo fibroblasts by the cellular p53 oncogene. Oncogene 2, 445–452 (1988).

    CAS  PubMed  Google Scholar 

  45. el-Deiry, W.S. et al. WAF1, a potential mediator of p53 tumor suppression. Cell 75, 817–825 (1993).

    Article  CAS  PubMed  Google Scholar 

  46. Rebollo, A. & Schmitt, C. Ikaros, Aiolos and Helios: transcription regulators and lymphoid malignancies. Immunol. Cell Biol. 81, 171–175 (2003).

    Article  CAS  PubMed  Google Scholar 

  47. Georgopoulos, K. et al. The Ikaros gene is required for the development of all lymphoid lineages. Cell 79, 143–156 (1994).

    Article  CAS  PubMed  Google Scholar 

  48. Wang, J.H. et al. Selective defects in the development of the fetal and adult lymphoid system in mice with an Ikaros null mutation. Immunity 5, 537–549 (1996).

    Article  CAS  PubMed  Google Scholar 

  49. Kim, J. et al. Ikaros DNA-binding proteins direct formation of chromatin remodeling complexes in lymphocytes. Immunity 10, 345–355 (1999).

    Article  CAS  PubMed  Google Scholar 

  50. Thornton, A.M. et al. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J. Immunol. 184, 3433–3441 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Getnet, D. et al. A role for the transcription factor Helios in human CD4+CD25+ regulatory T cells. Mol. Immunol. 47, 1595–1600 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Sugimoto, N. et al. Foxp3-dependent and -independent molecules specific for CD25+CD4+ natural regulatory T cells revealed by DNA microarray analysis. Int. Immunol. 18, 1197–1209 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Hahm, K. et al. Helios, a T cell-restricted Ikaros family member that quantitatively associates with Ikaros at centromeric heterochromatin. Genes Dev. 12, 782–796 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kelley, C.M. et al. Helios, a novel dimerization partner of Ikaros expressed in the earliest hematopoietic progenitors. Curr. Bio. 8, 508–15 (1998).

    Article  CAS  Google Scholar 

  55. Goodman, R.H. & Smolik, S. CBP/p300 in cell growth, transformation, and development. Genes Dev. 14, 1553–1577 (2000).

    CAS  PubMed  Google Scholar 

  56. Kasper, L.H. et al. Two transactivation mechanisms cooperate for the bulk of HIF-1-responsive gene expression. EMBO J. 24, 3846–3858 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Margueron, R. & Reinberg, D. The Polycomb complex PRC2 and its mark in life. Nature 469, 343–349 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Beroukhim, R. et al. The landscape of somatic copy-number alteration across human cancers. Nature 463, 899–905 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hollstein, M., Sidransky, D., Vogelstein, B. & Harris, C.C. p53 mutations in human cancers. Science 253, 49–53 (1991).

    Article  CAS  PubMed  Google Scholar 

  60. Lohmann, D. Retinoblastoma. Adv. Exp. Med. Biol. 685, 220–227 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Hanel, W. & Moll, U.M. Links between mutant p53 and genomic instability. J. Cell. Biochem. 113, 433–439 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Li, M. et al. The ATM-p53 pathway suppresses aneuploidy-induced tumorigenesis. Proc. Natl. Acad. Sci. USA 107, 14188–14193 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Thompson, S.L. & Compton, D.A. Proliferation of aneuploid human cells is limited by a p53-dependent mechanism. J. Cell Biol. 188, 369–381 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Deshpande, A. & Hinds, P.W. The retinoblastoma protein in osteoblast differentiation and osteosarcoma. Curr. Mol. Med. 6, 809–817 (2006).

    CAS  PubMed  Google Scholar 

  67. Romero, F., Martinez, A.C., Camonis, J. & Rebollo, A. Aiolos transcription factor controls cell death in T cells by regulating Bcl-2 expression and its cellular localization. EMBO J. 18, 3419–3430 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Krzywinski, M. et al. Circos: an information aesthetic for comparative genomics. Genome Res. 19, 1639–1645 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Boulos, N. et al. Chemotherapeutic agents circumvent emergence of dasatinib-resistant BCR-ABL kinase mutations in a precise mouse model of Philadelphia chromosome-positive acute lymphoblastic leukemia. Blood 117, 3585–3595 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Mardis, E.R. et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N. Engl. J. Med. 361, 1058–1066 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Downing, J.R. et al. The Pediatric Cancer Genome Project. Nat. Genet. 44, 619–622 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Sherry, S.T. et al. dbSNP: the NCBI database of genetic variation. Nucleic Acids Res. 29, 308–311 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  76. Rozen, S. & Skaletsky, H.J. Bioinformatics Methods and Protocols (Methods in Molecular Biology). (Humana Press, 2000).

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Chen, K. et al. PolyScan: an automatic indel and SNP detection approach to the analysis of human resequencing data. Genome Res. 17, 659–666 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. 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 

  80. Donovan, S., See, W., Bonifas, J., Stokoe, D. & Shannon, K.M. Hyperactivation of protein kinase B and ERK have discrete effects on survival, proliferation, and cytokine expression in Nf1-deficient myeloid cells. Cancer Cell 2, 507–514 (2002).

    Article  CAS  PubMed  Google Scholar 

  81. Schubbert, S. et al. Germline KRAS mutations cause Noonan syndrome. Nat. Genet. 38, 331–336 (2006).

    Article  CAS  PubMed  Google Scholar 

  82. Peto, R. et al. Design and analysis of randomized clinical trials requiring prolonged observation of each patient. II. analysis and examples. Br. J. Cancer 35, 1–39 (1977).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. 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 

  84. 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).

    Article  Google Scholar 

  85. R Development Core Team. R: a language and environment for statistical computing (R Foundation for Statistical Computing, Vienna, 2006).

Download references

Acknowledgements

We thank J. Morris, E. Walker and A. Merriman for performing SNP and gene expression microarrays and G. Zambetti and P. Brindle for insightful discussions of TP53 and CREBBP mutational data, respectively. We also thank H. Mulder, R. Collins, M. Barbato, E. Stonerock, E. Pinto and M. Ellis for technical assistance, the Tissue Resources Core facility and the Flow Cytometry and Cell Sorting Core facility of the St. Jude Children's Research Hospital (SJCRH). This work was supported by The Henry Schueler 41&9 Foundation in conjunction with Partnership for Cures, the St. Baldrick's Foundation, US National Cancer Institute (NCI) grant RC4CA156329, US National Institutes of Health (NIH) grants CA21765 and U01 GM92666, the American Association for Cancer Research (AACR) Gertrude B. Elion Cancer Research Award and the American Lebanese and Syrian Associated Charities (ALSAC) of SJCRH. Support was also provided by NCI grants to the Children's Oncology Group, including CA98543, CA98413 and CA114766. L.H. was supported by the Swedish Research Council. S.P.H. is the Ergen Family Chair in Pediatric Cancer. M.L.L. is a Clinical Scholar in the Leukemia Lymphoma Society and supported by the Frank A. Campini Foundation. C.G.M. is a Pew Scholar in the Biomedical Sciences and a St. Baldrick's Scholar. M.L.L. and E.D.-F. were supported by the Team Connor Foundation, and S.N.P. was supported by 5R25CA023944 from NCI. This paper is dedicated to Henry 'Hank' Schueler who died from complications of hypodiploid ALL and whose Foundation is dedicated to finding a cure for hypodiploid ALL in his memory and to James B. Nachman who was instrumental in the genesis of this project and who recently passed away.

Author information

Authors and Affiliations

Authors

Contributions

L.H., C.G.M., M.L.L. and S.P.H. designed the experiments. L.H. and D.P.-T. prepared patient samples and generated xenografts. L.H., E.D.-F., M.C., K.M., S.N.P., L.A.P. and S.L.H. performed biochemical analyses. C.D. and S.B. performed ex vivo drug studies. L.H. and J.M. analyzed SNP array data. L.H. and J.E. performed transcriptome sequencing. K.B. performed exome sequencing. L.H., D.P.-T. and B.V. performed sequencing validation. L.W., J.Z., L.D., J.B., Y.T., X.C. and C.L. analyzed sequence data. S.-C.C., A.A., G.N. and L.H. analyzed expression microarray data. M.R., E.H., M.P., G.W., R.H. and G.S. provided bioinformatic support. D.P., C.C. and M.D. performed statistical analyses. R.S.F. and L.L.F. supervised whole-genome sequencing data generation. D.J.D. supervised the automated analysis pipeline. S.C.R., A.J.C. and N.A.H. performed cytogenetic analyses. M.W., C.-H.P., M.M., I.D.L., L.B.T., P.M., A.W.R., G.R., W.S., M.L.L., J.M.G.-F., R.C.R., B.W., M.J.B. and S.P.H. provided clinical samples and data. D.W.E. performed pathological analyses. R.A.D. and H.G.D. provided important reagents. L.H. and C.G.M. wrote the manuscript. L.H., L.W., J.Z., L.D., D.P.-T., M.C., A.A., S.-C.C., K.M., J.B., J.M., G.W., G.S., J.E., M.P., X.C., M.R., E.H., C.L., R.S.F., L.L.F., D.J.D., K.O., S.A.S., C.-H.P., E.R.M., R.K.W., J.R.D. and C.G.M. are part of the St. Jude Children's Research Hospital–Washington University Pediatric Cancer Genome Project.

Corresponding author

Correspondence to Charles G Mullighan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Note and Supplementary Tables 3–6, 9, 13, 16–20, 23, 24 and 27–34 and Supplementary Figures 1–22 (PDF 5157 kb)

Supplementary Table 1

Pediatric hypodiploid ALL cohort (XLSX 36 kb)

Supplementary Table 2

Adult ALL cohort (XLSX 29 kb)

Supplementary Table 7

Mutations identified by next-generation sequencing (XLSX 123 kb)

Supplementary Table 8

Structural variations identified by whole genome sequencing (XLSX 19 kb)

Supplementary Table 10

Regions of copy number alterations and copy-neutral loss-of-heterozygosity in hypodiploid ALL (XLSX 357 kb)

Supplementary Table 11

Mutations identified by Sanger sequencing in the hypodiploid ALL cohort (XLSX 26 kb)

Supplementary Table 12

Copy number alterations and mutations (XLSX 29 kb)

Supplementary Table 14

Differential expression analysis – NH versus masked NH (XLSX 5372 kb)

Supplementary Table 15

Differential expression analysis – LH versus masked LH (XLSX 5395 kb)

Supplementary Table 21

Differential expression analysis – NH versus LH (XLSX 7710 kb)

Supplementary Table 22

Gene set enrichment analysis (GSEA) – NH versus LH (XLSX 176 kb)

Supplementary Table 25

Single nucleotide variations identified by mRNA seq of NALM-16 (XLSX 17 kb)

Supplementary Table 26

Primer sequences used for targeted gene resequencing and NF1 deletion mapping (XLSX 30 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Holmfeldt, L., Wei, L., Diaz-Flores, E. et al. The genomic landscape of hypodiploid acute lymphoblastic leukemia. Nat Genet 45, 242–252 (2013). https://doi.org/10.1038/ng.2532

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2532

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