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.

Dual targeting of p53 and c-MYC selectively eliminates leukaemic stem cells

Abstract

Chronic myeloid leukaemia (CML) arises after transformation of a haemopoietic stem cell (HSC) by the protein-tyrosine kinase BCR–ABL. Direct inhibition of BCR–ABL kinase has revolutionized disease management, but fails to eradicate leukaemic stem cells (LSCs), which maintain CML. LSCs are independent of BCR–ABL for survival, providing a rationale for identifying and targeting kinase-independent pathways. Here we show—using proteomics, transcriptomics and network analyses—that in human LSCs, aberrantly expressed proteins, in both imatinib-responder and non-responder patients, are modulated in concert with p53 (also known as TP53) and c-MYC regulation. Perturbation of both p53 and c-MYC, and not BCR–ABL itself, leads to synergistic cell kill, differentiation, and near elimination of transplantable human LSCs in mice, while sparing normal HSCs. This unbiased systems approach targeting connected nodes exemplifies a novel precision medicine strategy providing evidence that LSCs can be eradicated.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: p53 and c-MYC network in CML regulation.
Figure 2: Validation of proteomic network.
Figure 3: Modulation of p53 and c-MYC demonstrates CML sensitivity.
Figure 4: p53 and c-MYC abrogation in normal and primitive CML cells.
Figure 5: Mechanism and clinical relevance of treatment.
Figure 6: Targeting p53 and c-MYC in CML elicits synergistic kill in BCR–ABL+ LSCs.

Similar content being viewed by others

Accession codes

Primary accessions

European Nucleotide Archive

Gene Expression Omnibus

Data deposits

The CML and normal CD34+ mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifiers PXD001502, PXD001503, PXD001504; SCOPE3/SCOPE4 data are also available using PXD001505 and PXD002782 respectively. Transcriptomic data are publicly available via the accession codes E-MTAB-2581, E-MTAB-2508, E-MIMR-17 at ArrayExpress (https://www.ebi.ac.uk/arrayexpress/) and GSE47927, GSE5550, GSE24739, GSE14671 at GEO (http://www.ncbi.nlm.nih.gov/geo/). RNA-seq data (fastq) have been deposited in the European Nucleotide Archive under accession number PRJEB9942.

References

  1. O’Hare, T., Zabriskie, M. S., Eiring, A. M. & Deininger, M. W. Pushing the limits of targeted therapy in chronic myeloid leukaemia. Nature Rev. Cancer 12, 513–526 (2012)

    Article  CAS  Google Scholar 

  2. Druker, B. J. et al. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N. Engl. J. Med. 344, 1038–1042 (2001)

    Article  CAS  PubMed  Google Scholar 

  3. Graham, S. M. et al. Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro. Blood 99, 319–325 (2002)

    Article  CAS  PubMed  Google Scholar 

  4. Mahon, F. X. et al. Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete molecular remission for at least 2 years: the prospective, multicentre Stop Imatinib (STIM) trial. Lancet Oncol. 11, 1029–1035 (2010)

    Article  CAS  PubMed  Google Scholar 

  5. Corbin, A. S. et al. Human chronic myeloid leukemia stem cells are insensitive to imatinib despite inhibition of BCR-ABL activity. J. Clin. Invest. 121, 396–409 (2011)

    Article  CAS  PubMed  Google Scholar 

  6. Neviani, P. et al. PP2A-activating drugs selectively eradicate TKI-resistant chronic myeloid leukemic stem cells. J. Clin. Invest. 123, 4144–4157 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Dijkstra, E. W. A note on two problems in connexion with graphs. Numer. Math. 1, 269–271 (1959)

    Article  MathSciNet  MATH  Google Scholar 

  8. Van Dang, C. & McMahon, S. B. Emerging concepts in the analysis of transcriptional targets of the MYC oncoprotein: are the targets targetable? Genes Cancer 1, 560–567 (2010)

    Article  PubMed  CAS  Google Scholar 

  9. Boichenko, A. P. et al. A panel of regulated proteins in serum from patients with cervical intraepithelial neoplasia and cervical cancer. J. Proteome Res. 13, 4995–5007 (2014)

    Article  CAS  PubMed  Google Scholar 

  10. Liu, Y. et al. Glycoproteomic analysis of prostate cancer tissues by SWATH mass spectrometry discovers N-acylethanolamine acid amidase and protein tyrosine kinase 7 as signatures for tumor aggressiveness. Mol. Cell. Proteomics 13, 1753–1768 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sutton, C. W. et al. Quantitative proteomic profiling of matched normal and tumor breast tissues. J. Proteome Res. 9, 3891–3902 (2010)

    Article  CAS  PubMed  Google Scholar 

  12. Pierce, A. et al. Eight-channel iTRAQ enables comparison of the activity of six leukemogenic tyrosine kinases. Mol. Cell. Proteomics 7, 853–863 (2008)

    Article  CAS  PubMed  Google Scholar 

  13. Issaeva, N. et al. Small molecule RITA binds to p53, blocks p53–HDM-2 interaction and activates p53 function in tumors. Nature Med. 10, 1321–1328 (2004)

    Article  CAS  PubMed  Google Scholar 

  14. Wong, C. et al. The bromodomain and extra-terminal inhibitor CPI203 enhances the antiproliferative effects of rapamycin on human neuroendocrine tumors. Cell Death Dis. 5, e1450 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mertz, J. A. et al. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc. Natl Acad. Sci. USA 108, 16669–16674 (2011)

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

  16. Hamilton, A. et al. Chronic myeloid leukemia stem cells are not dependent on Bcr-Abl kinase activity for their survival. Blood 119, 1501–1510 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Li, L. et al. Activation of p53 by SIRT1 inhibition enhances elimination of CML leukemia stem cells in combination with imatinib. Cancer Cell 21, 266–281 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Shah, N. P. et al. Transient potent BCR-ABL inhibition is sufficient to commit chronic myeloid leukemia cells irreversibly to apoptosis. Cancer Cell 14, 485–493 (2008)

    Article  CAS  PubMed  Google Scholar 

  19. Chou, T. C. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 70, 440–446 (2010)

    Article  CAS  PubMed  Google Scholar 

  20. Yong, A. S., Szydlo, R. M., Goldman, J. M., Apperley, J. F. & Melo, J. V. Molecular profiling of CD34+ cells identifies low expression of CD7, along with high expression of proteinase 3 or elastase, as predictors of longer survival in patients with CML. Blood 107, 205–212 (2006)

    Article  CAS  PubMed  Google Scholar 

  21. McWeeney, S. K. et al. A gene expression signature of CD34+ cells to predict major cytogenetic response in chronic-phase chronic myeloid leukemia patients treated with imatinib. Blood 115, 315–325 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tovar, C. et al. MDM2 small-molecule antagonist RG7112 activates p53 signaling and regresses human tumors in preclinical cancer models. Cancer Res. 73, 2587–2597 (2013)

    Article  CAS  PubMed  Google Scholar 

  23. Albrecht, B. K. et al. Identification of a benzoisoxazoloazepine inhibitor (CPI-0610) of the bromodomain and extra-terminal (BET) family as a candidate for human clinical trials. J. Med. Chem. 59, 1330–1339 (2016)

    Article  CAS  PubMed  Google Scholar 

  24. Zhang, B. et al. Effective targeting of quiescent chronic myelogenous leukemia stem cells by histone deacetylase inhibitors in combination with imatinib mesylate. Cancer Cell 17, 427–442 (2010)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Hurtz, C. et al. BCL6-mediated repression of p53 is critical for leukemia stem cell survival in chronic myeloid leukemia. J. Exp. Med. 208, 2163–2174 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lanza, F. & Bi, S. Role of p53 in leukemogenesis of chronic myeloid leukemia. Stem Cells 13, 445–452 (1995)

    Article  CAS  PubMed  Google Scholar 

  27. Gómez-Casares, M. T. et al. MYC antagonizes the differentiation induced by imatinib in chronic myeloid leukemia cells through downregulation of p27(KIP1). Oncogene 32, 2239–2246 (2013)

    Article  PubMed  CAS  Google Scholar 

  28. Winter, G. E. et al. Systems-pharmacology dissection of a drug synergy in imatinib-resistant CML. Nature Chem. Biol. 8, 905–912 (2012)

    Article  CAS  Google Scholar 

  29. Takeishi, S. et al. Ablation of Fbxw7 eliminates leukemia-initiating cells by preventing quiescence. Cancer Cell 23, 347–361 (2013)

    Article  CAS  PubMed  Google Scholar 

  30. Reavie, L. et al. Regulation of c-Myc ubiquitination controls chronic myelogenous leukemia initiation and progression. Cancer Cell 23, 362–375 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Dang, C. V. MYC, metabolism, cell growth, and tumorigenesis. Cold Spring Harb. Perspect. Med. 3, a014217 (2013)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Vousden, K. H. & Prives, C. Blinded by the light: the growing complexity of p53. Cell 137, 413–431 (2009)

    Article  CAS  PubMed  Google Scholar 

  33. Wade, M., Li, Y. C. & Wahl, G. M. MDM2, MDMX and p53 in oncogenesis and cancer therapy. Nature Rev. Cancer 13, 83–96 (2013)

    Article  CAS  Google Scholar 

  34. Zhang, H., Li, H., Ho, N., Li, D. & Li, S. Scd1 plays a tumor-suppressive role in survival of leukemia stem cells and the development of chronic myeloid leukemia. Mol. Cell. Biol. 32, 1776–1787 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hsu, T. Y. et al. The spliceosome is a therapeutic vulnerability in MYC-driven cancer. Nature 525, 384–388 (2015)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  36. Koh, C. M. et al. MYC regulates the core pre-mRNA splicing machinery as an essential step in lymphomagenesis. Nature 523, 96–100 (2015)

    Article  CAS  PubMed  ADS  Google Scholar 

  37. Fong, C. Y. et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature 525, 538–542 (2015)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  38. Prost, S. et al. Erosion of the chronic myeloid leukaemia stem cell pool by PPARγ agonists. Nature 525, 380–383 (2015)

    Article  CAS  PubMed  ADS  Google Scholar 

  39. Pizzatti, L., Sá, L. A., de Souza, J. M., Bisch, P. M. & Abdelhay, E. Altered protein profile in chronic myeloid leukemia chronic phase identified by a comparative proteomic study. Biochim. Biophys. Acta 1764, 929–942 (2006)

    Article  CAS  PubMed  Google Scholar 

  40. Cramer-Morales, K. et al. Personalized synthetic lethality induced by targeting RAD52 in leukemias identified by gene mutation and expression profile. Blood 122, 1293–1304 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Diaz-Blanco, E. et al. Molecular signature of CD34+ hematopoietic stem and progenitor cells of patients with CML in chronic phase. Leukemia 21, 494–504 (2007)

    Article  CAS  PubMed  Google Scholar 

  42. Affer, M. et al. Gene expression differences between enriched normal and chronic myelogenous leukemia quiescent stem/progenitor cells and correlations with biological abnormalities. J. Oncol. 2011, 798592 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Graham, S. M., Vass, J. K., Holyoake, T. L. & Graham, G. J. Transcriptional analysis of quiescent and proliferating CD34+ human hemopoietic cells from normal and chronic myeloid leukemia sources. Stem Cells 25, 3111–3120 (2007)

    Article  CAS  PubMed  Google Scholar 

  44. Huber, W., von Heydebreck, A., Sültmann, H., Poustka, A. & Vingron, M. Variance stabilization applied to microarray data calibration and to the quantification of differential expression. Bioinformatics 18 (suppl. 1), S96–S104 (2002)

    Article  PubMed  Google Scholar 

  45. Smyth, G. K. in Bioinformatics and Computational Biology Solutions using R and Bioconductor (eds Gentleman, R., Dudoit, S., Irizarry, R. & Huber, W. ) 397–420 (Springer, 2005)

  46. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300 (1995)

    MathSciNet  MATH  Google Scholar 

  47. Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007)

    Article  MATH  PubMed  Google Scholar 

  48. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013)

    Article  PubMed  PubMed Central  Google Scholar 

  49. Smyth, G. K. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3, e3 (2004)

    Article  MathSciNet  ADS  Google Scholar 

  50. Clark, A. G. et al. Inferring nonneutral evolution from human-chimp-mouse orthologous gene trios. Science 302, 1960–1963 (2003)

    Article  CAS  PubMed  ADS  Google Scholar 

  51. Xu, X. L. et al. Retinoblastoma has properties of a cone precursor tumor and depends upon cone-specific MDM2 signaling. Cell 137, 1018–1031 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wolfer, A. et al. MYC regulation of a “poor-prognosis” metastatic cancer cell state. Proc. Natl Acad. Sci. USA 107, 3698–3703 (2010)

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

  53. Sheth, A. et al. Inhibition of human mitochondrial peptide deformylase causes apoptosis in c-myc-overexpressing hematopoietic cancers. Cell Death Dis. 5, e1152 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Liao, Y., Smyth, G. K. & Shi, W. The Subread aligner: fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res. 41, e108 (2013)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014)

    Article  CAS  PubMed  Google Scholar 

  56. Robinson, M. D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11, R25 (2010)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Law, C. W., Chen, Y., Shi, W. & Smyth, G. K. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank all CML patients and UK haematology departments who contributed samples; A. Hair for sample processing; J. Cassels for cell sorting; A. Michie and V. Helgason for assisting with the in vivo research and providing cord blood samples; C. Wells, P. Bailey and J. Cole for discussions regarding the RNA-seq analysis. We acknowledge the Cancer Research UK (CR-UK) Glasgow Centre (C596/A18076) and the CR-UK Beatson Institute (C596/A17196) for providing animal care and housing facilities. We acknowledge Constellation Pharmaceuticals for providing CPI-203, CPI-0610 and part funding M.E.D., Roche for providing RG7112, RG7388 and part funding M.E.D. and the SPIRIT Trials Management Group for access to CML samples. This study was supported by the Glasgow and Manchester Experimental Cancer Medicine Centres (ECMC), which are funded by CR-UK and the Chief Scientist’s Office (Scotland). We acknowledge the funders who have contributed to this work: MRC stratified medicine infrastructure award (A.D.W.), CR-UK C11074/A11008 (F.P., L.E.M.H., T.L.H., A.D.W.); LLR08071 (S.A.A., E.C.); LLR11017 (M.C.); SCD/04 (M.C.); LLR13035 (S.A.A., K.D., A.D.W. and A.P.); LLR14005 (M.T.S., D.V.); KKL690 (L.E.P.); KKL698 (P.B.); LLR08004 (A.D.W., A.P. and A.J.W.); MRC CiC (M.E.D.); The Howat Foundation (fluorescence-activated cell sorting (FACS) support); Friends of Paul O’Gorman (K.D. and FACS support); ELF 67954 (S.A.A.); British Society for Haematology start-up fund (S.A.A.); MR/K014854/1 (K.D.).

Author information

Authors and Affiliations

Authors

Contributions

A.D.W. and T.L.H. supervised the entire study and research. S.A.A., L.E.M.H., A.D.W. and T.L.H. designed the research, analysed and interpreted data, and wrote the manuscript. S.A.A. conceived the hypothesis, supervised in vivo research, prepared samples for proteomic and RNA-seq, performed all in vitro work including western blotting, immunofluorescence, cloning and knockdown studies, clonogenic studies, flow cytometry and all mouse in vivo studies including tissue processing-FISH preparation and slide interpretation, engraftment determination and analysis of primitive stem cell subsets. L.E.M.H. designed and performed all in silico work including global omics handling, integration and analysis (MS, RNA-seq and microarray data); network analyses; correlation/MI calculations; functional enrichment analyses and permutation experiments for calculation of P values. E.C. performed proteomic work. A.J.K.W. performed proteomic work and generated relative proteomic quantification. M.E.D. performed virus preparation, prepared drugs for in vivo work and assisted with in vivo studies. K.D. provided maintenance and care for all mouse colonies and assisted with in vivo work. P.B. and L.E.P. provided assistance with in vivo studies and D-FISH preparation. F.P. and M.T.S. provided assistance with in vivo studies. D.V. provided analysed datasets and analysed/interpreted RNA-seq data. S.M.G. supervised and interpreted RNA-seq. C.N. performed RNA-seq experiments. K.K. and M.C. provided analysed datasets. A.P. supervised proteomic studies. All authors reviewed/edited the manuscript.

Corresponding author

Correspondence to Tessa L. Holyoake.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 BCR–ABL drives a proteomic signature mediated by p53 and c-MYC.

a, b, Equivalent to Fig. 1b, c with additional information regarding the correlations calculated from the complete list of 58 candidate proteins (rc) in addition to the correlations for the candidate network (rn) and the background (r0). Also shown is the gain in r2 obtained for the candidate network as compared to the r2 obtained for the candidate list as a whole (r2Δ). FDR calculated from 10,000 re-samplings. c, Expression changes of the network components (shown as bar plots) in the context of quiescent and primitive CML cells; data shown in each panel (left to right) are (1) CD34+ protein log2 ratios (n = 3 patient samples, n = 2 normal samples); (2) CD34+HstloPylo transcript logFC (ArrayExpress accession E-MTAB-2508); (3) CD34+HstloPylo transcript logFC (GEO accession GSE24739); (4) CD34+CD38 logFC (ArrayExpress accession E-MTAB-2581); and (5) LinCD34+CD38CD90+ logFC (GEO accession GSE47927). Down-/upregulation is indicated by turquoise/red, respectively. Where multiple probesets were found for individual genes, the probeset corresponding to the maximal log ratio was selected. d, e, Correlation of the candidate network in progenitor (CD34+) CML cells: CD34+CD38+ progenitor (top); common myeloid progenitor LinCD34+CD38+CD123+CD45RA (middle); and CD34+ cells (bottom). As in a, b, correlations for the background (r0), candidate list (58 proteins, rc) and candidate network (Fig. 1a, rn) are shown. Also shown is the gain in r2 obtained for the candidate network as compared to the r2 obtained for the candidate list as a whole (r2Δ). FDR calculated from 10,000 re-samplings; MI statistics corresponding to FDRs < 0.05 are coloured red, FDRs < 0.10 are coloured grey. f, A Venn diagram showing the overlap in protein identification of the three MS instruments: ABSciex Q-STAR Elite (Elite), Thermo LTQ Orbitrap Velos (Orbi) and ABSciex TripleTOF 5600 (5600).

Source data

Extended Data Figure 2 Validation of network candidates.

a, HDM2 and c-MYC knockdown using shRNA constructs. Western blots of c-MYC, HDM2, p53 and HSP90 in HeLa cells transduced with lentiviral constructs specific for either c-MYC (2 constructs), HDM2 (2 constructs) or scrambled control (1 construct). KD, knockdown. b, c, CML CD34+ cells were transduced with either lentiviral (GFP) shRNA constructs to HDM2 (constructs 1, 2), c-MYC (constructs 1, 2) or scramble control (1 construct). b, Transduced viable GFP+ cells (assessed as annexin-V/DAPI/GFP+ percentages multiplied by the absolute cell count) are presented as a percentage of CML CD34+ cells transduced with scramble control (n = 3 patient samples). c, Early apoptosis levels (assessed as annexin-V+/DAPI/GFP+) after transduction of CML CD34+ cells (n = 3 patient samples) as described in b. Statistical significance was calculated by a two-tailed Student’s t-test and error bars represent the s.e.m.

Source data

Extended Data Figure 3 RITA and CPI-203 synergize to eliminate CML CD34+ cells.

a, b, Western blots of CML CD34+ cells untreated or treated with 50 nM RITA; 1 μM CPI-203; or the combination of 50 nM RITA and 1 μM CPI-203 or 150 nM Das for 8 h (a) and 48 h (b). c, p53 (red, nucleus in blue) 24 h after treatment in CML CD34+ cells. d, RITA, CPI-203 and combination drug treatment eliminates CD34+ CML cells through mechanisms probably dependent on apoptosis; after 72 h of drug treatment apoptosis levels were assessed (annexin V/DAPI) using flow cytometry techniques. e, RITA or Nut cannot induce death of K562 cells that lack p53. K562 cells were treated with either 50 μM RITA or 10 μM Nut and after 72 h of drug treatment, apoptosis levels were assessed (annexin V/DAPI) using flow cytometry techniques.

Extended Data Figure 4 RITA and CPI-203 synergize to eliminate CML cells.

a, CD34+ CML cells were treated with Nut and CPI-203 for 72 h with apoptosis levels assessed (annexin V/DAPI) using flow cytometry techniques. b, Treatment of CD34+ CML cells with Nut results in the elimination of early and late progenitor cells as assessed by the functional colony-forming capacity of drug-treated CML cells. c, Sequential drug treatments (n = 3 patient samples; drug one for 24 h, then both for 48 h). d, Sequential knockdown treatments (n = 3 patient samples; knockdown one for 24 h, then both for 48 h), mean ± s.e.m. (P values: two-tailed Student’s t-test; *P < 0.05, **P < 0.01, ***P < 0.001). e, CML CD34+ primary samples were pre-treated or not with imatinib mesylate (1 μM) for 8 h followed by RITA (50 nM), CPI-203 (1 μM) or the combined treatment (RITA plus CPI-203) for 72 h (right three columns). Cell counts were obtained using trypan blue exclusion.

Source data

Extended Data Figure 5 RITA and CPI-203 selectively eliminate LSCs.

a, b, Viable cell counts (n = 3 patient samples) (a); apoptosis in normal CD34+ cells (n = 3 patient samples) in response to RITA and/or CPI-203 (b). c, Gated CML CD34+CD38 cells 72 h after treatment (n = 4 patient samples). d, Ex vivo protocol for CML/cord blood CD34+ cells in NSG mice (n = 5 mice per arm). e, f, Targeting p53 and c-MYC in CML eliminates NSG repopulating leukaemic stem cells. CML CD34+ cells were treated with RITA (70 nM) and/or CPI-203 (1 μM) or Das (150 nM) for 48 h and recovered cells were injected intravenously into 8–12-week-old, sublethally irradiated (2.5 Gy) NSG mice (2–4 mice per arm). e, Percentage of human CD45+ cell levels in peripheral blood (PB) at 8, 12 and 16 weeks. f, Percentages of human CD45+, CD34+, CD33+, CD11b+, CD19+ and CD14+ cells in the bone marrow at 16 weeks. g, CML bone marrow analyses of CD33, CD11b, CD19 and CD14 from a CML sample determined to engraft both BCR–ABL-positive and -negative cells. h, D-FISH analyses of bone marrow human engraftment studies shown in g performed twice (2 patient samples) with a minimum of n = 6 mice per arm; mean ± s.e.m. (P values: two-tailed Student’s t-test; *P < 0.05, **P < 0.01, ***P < 0.001).

Source data

Extended Data Figure 6 Mechanism of LSC elimination and clinical scope.

a, Enrichment of p53 (top); apoptosis (second from top); c-MYC (second from bottom); and differentiation MSigDB signatures (bottom) in the four treatment arms (n = 3 CML patient samples per arm) (columns named as per b). Equivalent to Fig. 5b, but with named MSigDB signatures. b, Enrichment of PANTHER pathways in the four treatment arms. Pathway enrichment calculated from the top 1,500 genes, as ranked by increasing P value (top); only those genes exhibiting an absolute FC of >0.5 in each arm (bottom left); only those genes exhibiting a P < 0.05 each arm (bottom right). c, Assessing molecular synergy of the combined RITA plus CPI-203 treatment, as compared to the individual RITA and CPI-203 arms of the RNA-seq experiments in the three in silico functional signatures: p53/apoptosis (left); c-MYC (middle); and differentiation (right). Mean expression is shown as a solid line.

Source data

Extended Data Figure 7 Mechanism of LSC elimination and clinical scope continued.

a, Gene expression patterns (logFC, n = 3 patient samples per arm) shown for the members of the three broad signatures identified in silico: p53/apoptosis (left); c-MYC (middle); and differentiation (right) (*q < 0.05); data are ordered by increasing logFC in response to combination treatment, from downregulation at the top to upregulation at the bottom. Corresponding expression data are provided in Supplementary Tables 5–7. b, Differential expression of CD34 and CD133 (markers of stemness) in the four arms of the RNA-seq experiment. c, Apoptosis levels assessed (annexin V+/DAPI) using flow cytometry on a TKI-NR CD34+ sample after 72 h treatment with RITA and CPI-203 as indicated.

Source data

Extended Data Figure 8 RG7112 and CPI-0610 as a combination decrease BCR–ABL+ cells.

a, b, DTG mice in vivo treatment (a): neutrophils normalized to control (dotted line) (b). c, Bone marrow cells stained for CD45.1/2. Drug treatment arms (minimum of n = 7 mice) mean ± s.e.m. (P values: two-tailed Student’s t-test; *P < 0.05, **P < 0.01, ***P < 0.001).

Source data

Supplementary information

Supplementary Information

This file contains the raw data for Figure 2a and Extended Data Figures 2a, 3a, 3b, Supplementary Tables 1, 3, 4, 8 and legends for Supplementary Tables 2, 5, 6 and 7 (see separate excel files). (PDF 672 kb)

Supplementary Table 2

This file contains the candidate network statistics (see Supplementary Information file for legend). (XLSX 11 kb)

Supplementary Table 5

This file shows the molecular deregulation of P53/apoptosis-related signatures and pathways (see Supplementary Information file for legend). (XLSX 35 kb)

Supplementary Table 6

This file shows the molecular deregulation of MYC-related signatures and pathways (see Supplementary Information file for legend). (XLSX 13 kb)

Supplementary Table 7

This file shows the molecular deregulation of differentiation-related signatures and pathways (see Supplementary Information file for legend). (XLSX 11 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Abraham, S., Hopcroft, L., Carrick, E. et al. Dual targeting of p53 and c-MYC selectively eliminates leukaemic stem cells. Nature 534, 341–346 (2016). https://doi.org/10.1038/nature18288

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature18288

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