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:

Autocrine activation of the MET receptor tyrosine kinase in acute myeloid leukemia

Abstract

Although the treatment of acute myeloid leukemia (AML) has improved substantially in the past three decades, more than half of all patients develop disease that is refractory to intensive chemotherapy1,2. Functional genomics approaches offer a means to discover specific molecules mediating the aberrant growth and survival of cancer cells3,4,5,6,7,8. Thus, using a loss-of-function RNA interference genomic screen, we identified the aberrant expression of hepatocyte growth factor (HGF) as a crucial element in AML pathogenesis. We found HGF expression leading to autocrine activation of its receptor tyrosine kinase, MET, in nearly half of the AML cell lines and clinical samples we studied. Genetic depletion of HGF or MET potently inhibited the growth and survival of HGF-expressing AML cells. However, leukemic cells treated with the specific MET kinase inhibitor crizotinib developed resistance resulting from compensatory upregulation of HGF expression, leading to the restoration of MET signaling. In cases of AML where MET is coactivated with other tyrosine kinases, such as fibroblast growth factor receptor 1 (FGFR1)9, concomitant inhibition of FGFR1 and MET blocked this compensatory HGF upregulation, resulting in sustained logarithmic cell killing both in vitro and in xenograft models in vivo. Our results show a widespread dependence of AML cells on autocrine activation of MET, as well as the key role of compensatory upregulation of HGF expression in maintaining leukemogenic signaling by this receptor. We anticipate that these findings will lead to the design of additional strategies to block adaptive cellular responses that drive compensatory ligand expression as an essential component of the targeted inhibition of oncogenic receptors in human cancers.

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: Aberrant HGF expression by AML cells is associated with MET activation and is necessary for cell growth and survival.
Figure 2: HGF and MET are coexpressed in the leukemic blasts of patients with AML and are induced by leukemogenic transcription factors in primary mouse hematopoietic progenitor cells, conferring susceptibility to MET kinase inhibition.
Figure 3: Restoration of leukemic cell growth after chronic MET kinase inhibitor treatment is a result of the compensatory upregulation of HGF and MET reactivation, which can be overcome by inhibiting the compensatory upregulation of HGF.
Figure 4: Combined inhibition of MET and FGFR1 blocks the compensatory upregulation of HGF, leading to sustained inhibition of MET in KG-1 cells and near-complete regression of AML in vivo.

Similar content being viewed by others

References

  1. Grimwade, D. et al. The predictive value of hierarchical cytogenetic classification in older adults with acute myeloid leukemia (AML): analysis of 1065 patients entered into the United Kingdom Medical Research Council AML11 trial. Blood 98, 1312–1320 (2001).

    Article  CAS  Google Scholar 

  2. Burnett, A., Wetzler, M. & Lowenberg, B. Therapeutic advances in acute myeloid leukemia. J. Clin. Oncol. 29, 487–494 (2011).

    Article  Google Scholar 

  3. Westbrook, T.F., Stegmeier, F. & Elledge, S.J. Dissecting cancer pathways and vulnerabilities with RNAi. Cold Spring Harb. Symp. Quant. Biol. 70, 435–444 (2005).

    Article  CAS  Google Scholar 

  4. Bernards, R., Brummelkamp, T.R. & Beijersbergen, R.L. shRNA libraries and their use in cancer genetics. Nat. Methods 3, 701–706 (2006).

    Article  CAS  Google Scholar 

  5. Ngo, V.N. et al. A loss-of-function RNA interference screen for molecular targets in cancer. Nature 441, 106–110 (2006).

    Article  CAS  Google Scholar 

  6. Whitehurst, A.W. et al. Synthetic lethal screen identification of chemosensitizer loci in cancer cells. Nature 446, 815–819 (2007).

    Article  CAS  Google Scholar 

  7. Turner, N.C. et al. A synthetic lethal siRNA screen identifying genes mediating sensitivity to a PARP inhibitor. EMBO J. 27, 1368–1377 (2008).

    Article  CAS  Google Scholar 

  8. Scholl, C. et al. Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells. Cell 137, 821–834 (2009).

    Article  CAS  Google Scholar 

  9. Gu, T.L. et al. Phosphotyrosine profiling identifies the KG-1 cell line as a model for the study of FGFR1 fusions in acute myeloid leukemia. Blood 108, 4202–4204 (2006).

    Article  CAS  Google Scholar 

  10. Wang, C., Curtis, J.E., Minden, M.D. & McCulloch, E.A. Expression of a retinoic acid receptor gene in myeloid leukemia cells. Leukemia 3, 264–269 (1989).

    CAS  Google Scholar 

  11. Birchmeier, C., Birchmeier, W., Gherardi, E. & Vande Woude, G.F. MET, metastasis, motility, and more. Nat. Rev. Mol. Cell Biol. 4, 915–925 (2003).

    Article  CAS  Google Scholar 

  12. Valk, P.J. et al. Prognostically useful gene-expression profiles in acute myeloid leukemia. N. Engl. J. Med. 350, 1617–1628 (2004).

    Article  CAS  Google Scholar 

  13. Zou, H.Y. et al. An orally available small-molecule inhibitor of c-Met, PF-2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res. 67, 4408–4417 (2007).

    Article  CAS  Google Scholar 

  14. Mohammadi, M. et al. Crystal structure of an angiogenesis inhibitor bound to the FGF receptor tyrosine kinase domain. EMBO J. 17, 5896–5904 (1998).

    Article  CAS  Google Scholar 

  15. Sporn, M.B. & Todaro, G.J. Autocrine secretion and malignant transformation of cells. N. Engl. J. Med. 303, 878–880 (1980).

    Article  CAS  Google Scholar 

  16. Loriaux, M.M. et al. High-throughput sequence analysis of the tyrosine kinome in acute myeloid leukemia. Blood 111, 4788–4796 (2008).

    Article  CAS  Google Scholar 

  17. Haq, R. et al. Constitutive p38HOG mitogen-activated protein kinase activation induces permanent cell cycle arrest and senescence. Cancer Res. 62, 5076–5082 (2002).

    CAS  Google Scholar 

  18. Majeti, R. et al. Dysregulated gene expression networks in human acute myelogenous leukemia stem cells. Proc. Natl. Acad. Sci. USA 106, 3396–3401 (2009).

    Article  CAS  Google Scholar 

  19. Robinson, D.R., Wu, Y.M. & Lin, S.F. The protein tyrosine kinase family of the human genome. Oncogene 19, 5548–5557 (2000).

    Article  CAS  Google Scholar 

  20. Zheng, R., Klang, K., Gorin, N.C. & Small, D. Lack of KIT or FMS internal tandem duplications but co-expression with ligands in AML. Leuk. Res. 28, 121–126 (2004).

    Article  CAS  Google Scholar 

  21. Zhou, J. et al. Enhanced activation of STAT pathways and overexpression of survivin confer resistance to FLT3 inhibitors and could be therapeutic targets in AML. Blood 113, 4052–4062 (2009).

    Article  CAS  Google Scholar 

  22. Sato, T. et al. FLT3 ligand impedes the efficacy of FLT3 inhibitors in vitro and in vivo. Blood 117, 3286–3293 (2011).

    Article  CAS  Google Scholar 

  23. Ngo, V.N. et al. A loss-of-function RNA interference screen for molecular targets in cancer. Nature 441, 106–110 (2006).

    Article  CAS  Google Scholar 

  24. Heinrichs, S. et al. Accurate detection of uniparental disomy and microdeletions by SNP array analysis in myelodysplastic syndromes with normal cytogenetics. Leukemia 23, 1605–1613 (2009).

    Article  CAS  Google Scholar 

  25. Mathew, P. et al. Detection of MYCN gene amplification in neuroblastoma by fluorescence in situ hybridization: a pediatric oncology group study. Neoplasia 3, 105–109 (2001).

    Article  CAS  Google Scholar 

  26. Armstrong, S.A. et al. Inhibition of FLT3 in MLL. Validation of a therapeutic target identified by gene expression based classification. Cancer Cell 3, 173–183 (2003).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Gutierrez, M. Mansour and E. Gjini for critical discussions and J. Gilbert for editorial advice. This research was supported by the US National Institutes of Health grant K08CA160660 (A.K.), the William Lawrence and Blanche Hughes Foundation (T.S.), the Samuel Waxman Cancer Research Foundation (J.D.L.), the V Foundation (A.T.L.) and the Intramural Research Program of the National Cancer Institute, Center for Cancer Research (L.M.S.).

Author information

Authors and Affiliations

Authors

Contributions

A.K., C.R., K.L.R., T.S., A.C., E.T., V.N. and L.A.M. performed experiments. A.K., S.J.R., P.J.M.V., R.D., J.L.K., S.E.D., R.J.B., J.G.C., G.V.W., J.D.L., A.L.K., L.M.S. and A.T.L. analyzed data. A.K. and A.T.L. wrote the manuscript.

Corresponding author

Correspondence to A Thomas Look.

Ethics declarations

Competing interests

J.G.C. is an employee of Pfizer.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–19, Supplementary Tables 1–3 and Supplementary Methods (PDF 2513 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kentsis, A., Reed, C., Rice, K. et al. Autocrine activation of the MET receptor tyrosine kinase in acute myeloid leukemia. Nat Med 18, 1118–1122 (2012). https://doi.org/10.1038/nm.2819

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2819

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing