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:

Targeting mitochondrial oxidative phosphorylation eradicates therapy-resistant chronic myeloid leukemia stem cells

Abstract

Treatment of chronic myeloid leukemia (CML) with imatinib mesylate and other second- and/or third-generation c-Abl-specific tyrosine kinase inhibitors (TKIs) has substantially extended patient survival1. However, TKIs primarily target differentiated cells and do not eliminate leukemic stem cells (LSCs)2,3,4. Therefore, targeting minimal residual disease to prevent acquired resistance and/or disease relapse requires identification of new LSC-selective target(s) that can be exploited therapeutically5,6. Considering that malignant transformation involves cellular metabolic changes, which may in turn render the transformed cells susceptible to specific assaults in a selective manner7, we searched for such vulnerabilities in CML LSCs. We performed metabolic analyses on both stem cell–enriched (CD34+ and CD34+CD38) and differentiated (CD34) cells derived from individuals with CML, and we compared the signature of these cells with that of their normal counterparts. Through combination of stable isotope–assisted metabolomics with functional assays, we demonstrate that primitive CML cells rely on upregulated oxidative metabolism for their survival. We also show that combination treatment with imatinib and tigecycline, an antibiotic that inhibits mitochondrial protein translation, selectively eradicates CML LSCs both in vitro and in a xenotransplantation model of human CML. Our findings provide a strong rationale for investigation of the use of TKIs in combination with tigecycline to treat patients with CML with minimal residual disease.

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: Primitive CML cells show an increase in oxidative metabolism as compared to differentiated counterparts.
Figure 2: Mitochondrial metabolic activity is enhanced in primitive CML cells in comparison to normal undifferentiated hematopoietic cells.
Figure 3: Inhibition of aberrant oxidative metabolism targets CML progenitors and LSCs.
Figure 4: Inhibition of oxidative metabolism eliminates xenotransplanted human CML LSCs.

Similar content being viewed by others

References

  1. Druker, B.J. et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N. Engl. J. Med. 355, 2408–2417 (2006).

    Article  CAS  Google Scholar 

  2. 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  Google Scholar 

  3. 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  Google Scholar 

  4. 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  Google Scholar 

  5. Holyoake, T.L. & Helgason, G.V. Do we need more drugs for chronic myeloid leukemia? Immunol. Rev. 263, 106–123 (2015).

    Article  CAS  Google Scholar 

  6. Abraham, S.A. et al. Dual targeting of p53 and c-MYC selectively eliminates leukaemic stem cells. Nature 534, 341–346 (2016).

    Article  Google Scholar 

  7. Tennant, D.A., Durán, R.V. & Gottlieb, E. Targeting metabolic transformation for cancer therapy. Nat. Rev. Cancer 10, 267–277 (2010).

    Article  CAS  Google Scholar 

  8. Rowley, J.D. A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature 243, 290–293 (1973).

    Article  CAS  Google Scholar 

  9. Groffen, J. et al. Philadelphia chromosomal breakpoints are clustered within a limited region, bcr, on chromosome 22. Cell 36, 93–99 (1984).

    Article  CAS  Google Scholar 

  10. Konopka, J.B., Watanabe, S.M. & Witte, O.N. An alteration of the human c-abl protein in K562 leukemia cells unmasks associated tyrosine kinase activity. Cell 37, 1035–1042 (1984).

    Article  CAS  Google Scholar 

  11. Rousselot, P. et al. Imatinib mesylate discontinuation in patients with chronic myelogenous leukemia in complete molecular remission for more than 2 years. Blood 109, 58–60 (2007).

    Article  CAS  Google Scholar 

  12. 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  Google Scholar 

  13. Ross, D.M. et al. Safety and efficacy of imatinib cessation for CML patients with stable undetectable minimal residual disease: results from the TWISTER study. Blood 122, 515–522 (2013).

    Article  CAS  Google Scholar 

  14. Simsek, T. et al. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell 7, 380–390 (2010).

    Article  CAS  Google Scholar 

  15. Takubo, K. et al. Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell 12, 49–61 (2013).

    Article  CAS  Google Scholar 

  16. Yu, W.M. et al. Metabolic regulation by the mitochondrial phosphatase PTPMT1 is required for hematopoietic stem cell differentiation. Cell Stem Cell 12, 62–74 (2013).

    Article  CAS  Google Scholar 

  17. Ito, K. et al. A PML–PPAR-δ pathway for fatty acid oxidation regulates hematopoietic stem cell maintenance. Nat. Med. 18, 1350–1358 (2012).

    Article  CAS  Google Scholar 

  18. Carracedo, A., Cantley, L.C. & Pandolfi, P.P. Cancer metabolism: fatty acid oxidation in the limelight. Nat. Rev. Cancer 13, 227–232 (2013).

    Article  CAS  Google Scholar 

  19. Sullivan, L.B. et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162, 552–563 (2015).

    Article  CAS  Google Scholar 

  20. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    Article  CAS  Google Scholar 

  21. Cardaci, S. et al. Pyruvate carboxylation enables growth of SDH-deficient cells by supporting aspartate biosynthesis. Nat. Cell Biol. 17, 1317–1326 (2015).

    Article  CAS  Google Scholar 

  22. Norberg, E. et al. Differential contribution of the mitochondrial translation pathway to the survival of diffuse large B-cell lymphoma subsets. Cell Death Differ. 24, 251–262 (2017).

    Article  CAS  Google Scholar 

  23. Skrtić, M. et al. Inhibition of mitochondrial translation as a therapeutic strategy for human acute myeloid leukemia. Cancer Cell 20, 674–688 (2011).

    Article  Google Scholar 

  24. Mackay, G.M., Zheng, L., van den Broek, N.J. & Gottlieb, E. Analysis of cell metabolism using LC–MS and isotope tracers. Methods Enzymol. 561, 171–196 (2015).

    Article  CAS  Google Scholar 

  25. Karvela, M. et al. ATG7 regulates energy metabolism, differentiation and survival of Philadelphia-chromosome-positive cells. Autophagy 12, 936–948 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to dedicate this work to the memory of Prof. Tessa Holyoake who sadly passed away recently. Tessa was a dedicated clinician and an enthusiastic scientist who combined both aspects of her work for patients' benefit. She will be gravely missed.

We thank all patients and healthy donors who contributed samples and National Health Service (NHS) Greater Glasgow and Clyde Biorepository; A. Hair for sample processing; T. Gilbey and T. Harvey for cell sorting; N. Van Den Broek and G. MacKay for technical assistance; and A. King for editorial work. This study was supported by Cancer Research UK; the Cancer ResearchUK Glasgow Centre (C596/A18076) and the Biological Services Unit facilities at the Cancer Research UK Beatson Institute (C596/A17196); Medical Research Council/AstraZeneca project grants (MR/K014854/1); the Glasgow Experimental Cancer Medicine Centre (ECMC), which is funded by Cancer Research UK and by the Chief Scientist's Office (Scotland); the Howat Foundation and Friends of Paul O'Gorman; the Bloodwise Specialist Programme (14033); the Kay Kendall Leukaemia Fund (KKLF) (KKL501 and KKL698); Lady Tata International Award; and Leuka. G.V.H. is a KKLF Intermediate Research Fellow, Leadership Fellow and John Goldman Fellow.

Author information

Authors and Affiliations

Authors

Contributions

E.M.K., G.V.H. and E.G. wrote the manuscript. E.M.K., P.B., T.L.H., G.V.H. and E.G. designed experiments and interpreted data. E.M.K. and P.B. performed experiments and analyzed data. A.M.M. and K.D. assisted with the in vivo work. S.T. provided the custom-formulated culture medium. T.L.H. and G.V.H. provided primary cells. G.V.H. and E.G. supervised the project. All authors reviewed the manuscript.

Corresponding authors

Correspondence to G Vignir Helgason or Eyal Gottlieb.

Ethics declarations

Competing interests

E.G. is a founder and shareholder of MetaboMed, Ltd. T.L.H. has previously received research support from Bristol-Myers Squibb and Novartis.

Supplementary information

Supplementary Figures

Supplementary Figures 1–8. (PDF 39660 kb)

Life Sciences Reporting Summary (PDF 207 kb)

Supplementary Table 1

Comparative steady-state metabolomics analysis of patient-matched CD34+ and CD34 CML cells measured by LC–MS. (XLSX 18 kb)

Supplementary Table 2

Comparative steady-state metabolomics analysis of CD34+ CML and normal CD34+ cells measured by LC–MS. (XLSX 16 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kuntz, E., Baquero, P., Michie, A. et al. Targeting mitochondrial oxidative phosphorylation eradicates therapy-resistant chronic myeloid leukemia stem cells. Nat Med 23, 1234–1240 (2017). https://doi.org/10.1038/nm.4399

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4399

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