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:

Axitinib effectively inhibits BCR-ABL1(T315I) with a distinct binding conformation

Abstract

The BCR-ABL1 fusion gene is a driver oncogene in chronic myeloid leukaemia and 30–50% of cases of adult acute lymphoblastic leukaemia1. Introduction of ABL1 kinase inhibitors (for example, imatinib) has markedly improved patient survival2, but acquired drug resistance remains a challenge3,4,5. Point mutations in the ABL1 kinase domain weaken inhibitor binding6 and represent the most common clinical resistance mechanism. The BCR–ABL1 kinase domain gatekeeper mutation Thr315Ile (T315I) confers resistance to all approved ABL1 inhibitors except ponatinib7,8, which has toxicity limitations. Here we combine comprehensive drug sensitivity and resistance profiling of patient cells ex vivo with structural analysis to establish the VEGFR tyrosine kinase inhibitor axitinib as a selective and effective inhibitor for T315I-mutant BCR–ABL1-driven leukaemia. Axitinib potently inhibited BCR–ABL1(T315I), at both biochemical and cellular levels, by binding to the active form of ABL1(T315I) in a mutation-selective binding mode. These findings suggest that the T315I mutation shifts the conformational equilibrium of the kinase in favour of an active (DFG-in) A-loop conformation, which has more optimal binding interactions with axitinib. Treatment of a T315I chronic myeloid leukaemia patient with axitinib resulted in a rapid reduction of T315I-positive cells from bone marrow. Taken together, our findings demonstrate an unexpected opportunity to repurpose axitinib, an anti-angiogenic drug approved for renal cancer, as an inhibitor for ABL1 gatekeeper mutant drug-resistant leukaemia patients. This study shows that wild-type proteins do not always sample the conformations available to disease-relevant mutant proteins and that comprehensive drug testing of patient-derived cells can identify unpredictable, clinically significant drug-repositioning opportunities.

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: Ex vivo drug sensitivity and resistance testing (DSRT) of primary leukaemic cells establishes axitinib as a selective BCR-ABL1(T315I) inhibitor.
Figure 2: Co-crystal structures of axitinib bound to ABL1, ABL1(T315I) and VEGFR2 demonstrating different binding conformations.
Figure 3: Axitinib suppresses BCR–ABL1(T315I) autophosphorylation and proliferation of Ba/F3 cells expressing BCR–ABL1(T315I).
Figure 4: Axitinib potently and selectively targets BCR–ABL1(T315I)-expressing patient cells.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

Data deposits

X-ray crystallographic coordinates and structure factor files for axitinib–ABL1(T315I) and axitinib–wild-type-ABL1 complexes have been deposited in the Protein Data Bank under accession numbers 4TWP and 4WA9, respectively.

References

  1. Kurzrock, R., Kantarjian, H. M., Druker, B. J. & Talpaz, M. Philadelphia chromosome-positive leukemias: from basic mechanisms to molecular therapeutics. Ann. Intern. Med. 138, 819–830 (2003)

    Article  CAS  Google Scholar 

  2. Deininger, M., Buchdunger, E. & Druker, B. J. The development of imatinib as a therapeutic agent for chronic myeloid leukemia. Blood 105, 2640–2653 (2005)

    Article  CAS  Google Scholar 

  3. 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 

  4. Hochhaus, A. et al. Dasatinib induces notable hematologic and cytogenetic responses in chronic-phase chronic myeloid leukemia after failure of imatinib therapy. Blood 109, 2303–2309 (2007)

    Article  CAS  Google Scholar 

  5. Hochhaus, A. et al. Six-year follow-up of patients receiving imatinib for the first-line treatment of chronic myeloid leukemia. Leukemia 23, 1054–1061 (2009)

    Article  CAS  Google Scholar 

  6. Branford, S. et al. High frequency of point mutations clustered within the adenosine triphosphate-binding region of BCR/ABL in patients with chronic myeloid leukemia or Ph-positive acute lymphoblastic leukemia who develop imatinib (STI571) resistance. Blood 99, 3472–3475 (2002)

    Article  CAS  Google Scholar 

  7. Bradeen, H. A. et al. Comparison of imatinib mesylate, dasatinib (BMS-354825), and nilotinib (AMN107) in an N-ethyl-N-nitrosourea (ENU)-based mutagenesis screen: high efficacy of drug combinations. Blood 108, 2332–2338 (2006)

    Article  CAS  Google Scholar 

  8. Redaelli, S. et al. Activity of bosutinib, dasatinib, and nilotinib against 18 imatinib-resistant BCR/ABL mutants. J. Clin. Oncol. 27, 469–471 (2009)

    Article  CAS  Google Scholar 

  9. Senior, M. FDA halts then allows sales of Ariad’s leukemia medication. Nature Biotechnol. 32, 9–11 (2014)

    Article  CAS  Google Scholar 

  10. Gibbons, D. L. et al. Molecular dynamics reveal BCR-ABL1 polymutants as a unique mechanism of resistance to PAN-BCR-ABL1 kinase inhibitor therapy. Proc. Natl Acad. Sci. USA 111, 3550–3555 (2014)

    Article  ADS  CAS  Google Scholar 

  11. Zabriskie, M. S. et al. BCR-ABL1 compound mutations combining key kinase domain positions confer clinical resistance to ponatinib in Ph chromosome-positive leukemia. Cancer Cell 26, 428–442 (2014)

    Article  CAS  Google Scholar 

  12. Pemovska, T. et al. Individualized systems medicine strategy to tailor treatments for patients with chemorefractory acute myeloid leukemia. Cancer Discov. 3, 1416–1429 (2013)

    Article  CAS  Google Scholar 

  13. Yadav, B. et al. Quantitative scoring of differential drug sensitivity for individually optimized anticancer therapies. Sci. Rep. 4, 5193 (2014)

    Article  CAS  Google Scholar 

  14. McTigue, M. et al. Molecular conformations, interactions, and properties associated with drug efficiency and clinical performance among VEGFR TK inhibitors. Proc. Natl Acad. Sci. USA 109, 18281–18289 (2012)

    Article  ADS  CAS  Google Scholar 

  15. Rini, B. I. et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 378, 1931–1939 (2011)

    Article  CAS  Google Scholar 

  16. Solowiej, J. et al. Characterizing the effects of the juxtamembrane domain on vascular endothelial growth factor receptor-2 enzymatic activity, autophosphorylation, and inhibition by axitinib. Biochemistry 48, 7019–7031 (2009)

    Article  CAS  Google Scholar 

  17. Azam, M., Seeliger, M. A., Gray, N. S., Kuriyan, J. & Daley, G. Q. Activation of tyrosine kinases by mutation of the gatekeeper threonine. Nature Struct. Mol. Biol. 15, 1109–1118 (2008)

    Article  CAS  Google Scholar 

  18. Dixit, A. & Verkhivker, G. M. Hierarchical modeling of activation mechanisms in the ABL and EGFR kinase domains: thermodynamic and mechanistic catalysts of kinase activation by cancer mutations. PLOS Comput. Biol. 5, e1000487 (2009)

    Article  ADS  Google Scholar 

  19. Zhou, T. et al. Structural mechanism of the pan-BCR-ABL inhibitor ponatinib (AP24534): lessons for overcoming kinase inhibitor resistance. Chem. Biol. Drug Des. 77, 1–11 (2011)

    Article  CAS  Google Scholar 

  20. O'Hare, T. et al. AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell 16, 401–412 (2009)

    Article  CAS  Google Scholar 

  21. Gontarewicz, A. et al. Simultaneous targeting of Aurora kinases and Bcr-Abl kinase by the small molecule inhibitor PHA-739358 is effective against imatinib-resistant BCR-ABL mutations including T315I. Blood 111, 4355–4364 (2008)

    Article  CAS  Google Scholar 

  22. Chan, W. W. et al. Conformational control inhibition of the BCR-ABL1 tyrosine kinase, including the gatekeeper T315I mutant, by the switch-control inhibitor DCC-2036. Cancer Cell 19, 556–568 (2011)

    Article  CAS  Google Scholar 

  23. Davis, M. I. et al. Comprehensive analysis of kinase inhibitor selectivity. Nature Biotechnol. 29, 1046–1051 (2011)

    Article  CAS  Google Scholar 

  24. Gross-Goupil, M., Francois, L., Quivy, A. & Ravaud, A. Axitinib: a review of its safety and efficacy in the treatment of adults with advanced renal cell carcinoma. Clin. Med. Insights Oncol. 7, 269–277 (2013)

    Article  Google Scholar 

  25. Bracarda, S. et al. Axitinib safety in metastatic renal cell carcinoma: suggestions for daily clinical practice based on case studies. Expert Opin. Drug Saf. 13, 497–510 (2014)

    Article  CAS  Google Scholar 

  26. Verzoni, E. et al. Targeted treatments in advanced renal cell carcinoma: focus on axitinib. Pharmgenomics. Pers. Med. 7, 107–116 (2014)

    PubMed  PubMed Central  Google Scholar 

  27. Josephs, D. H., Fisher, D. S., Spicer, J. & Flanagan, R. J. Clinical pharmacokinetics of tyrosine kinase inhibitors: implications for therapeutic drug monitoring. Ther. Drug Monit. 35, 562–587 (2013)

    Article  CAS  Google Scholar 

  28. Chen, Y. et al. Clinical pharmacology of axitinib. Clin. Pharmacokinet. 52, 713–725 (2013)

    Article  CAS  Google Scholar 

  29. 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  Google Scholar 

  30. O'Hare, T. et al. Threshold levels of ABL tyrosine kinase inhibitors retained in chronic myeloid leukemia cells determine their commitment to apoptosis. Cancer Res. 73, 3356–3370 (2013)

    Article  CAS  Google Scholar 

  31. Morrison, J. F. Kinetics of the reversible inhibition of enzyme-catalysed reactions by tight-binding inhibitors. Biochim. Biophys. Acta 185, 269–286 (1969)

    Article  CAS  Google Scholar 

  32. Kapust, R. B. et al. Tobacco etch virus protease: mechanism of autolysis and rational design of stable mutants with wild-type catalytic proficiency. Protein Eng. 14, 993–1000 (2001)

    Article  CAS  Google Scholar 

  33. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007)

    Article  CAS  Google Scholar 

  34. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501 (2010)

    Article  CAS  Google Scholar 

  35. Murshudov, G. N., Vagin, A. A. & Dodson, E. J. Refinement of macromolecular structures by the maximum-likelihood method. Acta Crystallogr. D Biol. Crystallogr. 53, 240–255 (1997)

    Article  CAS  Google Scholar 

  36. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010)

    Article  CAS  Google Scholar 

  37. le Coutre, P. et al. In vivo eradication of human BCR/ABL-positive leukemia cells with an ABL kinase inhibitor. J. Natl. Cancer Inst. 91, 163–168 (1999)

    Article  CAS  Google Scholar 

  38. Solowiej, J., Chen, J. H., Zou, H. Y., Grant, S. K. & Murray, B. W. Substrate-specific conformational regulation of the receptor tyrosine kinase VEGFR2 catalytic domain. ACS Chem. Biol. 8, 978–986 (2013)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank the patients and their families for participating in this study and donating their samples for research. We acknowledge the High Throughput Biomedicine Unit at the Institute for Molecular Medicine Finland (FIMM) for technical assistance, the laboratory of C. Gambacorti-Passerini (University of Milano-Bicocca) for engineered Ba/F3 cell mutant panel profiling data, and T. Lundán (Department of Clinical Chemistry and TYKSLAB, Turku University Central Hospital, University of Turku) for the BCR–ABL1(T315I) transcript-level quantification. This work was supported by the Jane and Aatos Erkko Foundation (to K.W.), Academy of Finland (K.W. and O.K.), Finnish Cancer Societies (O.K. and K.P.), Sigrid Juselius Foundation (O.K.), Instrumentarium Foundation (M.K.), and FinPharma Doctoral Program-Drug Discovery section (T.P.).

Author information

Authors and Affiliations

Authors

Contributions

T.P., K.P., B.W.M. and K.W. conceived the study, designed experiments and wrote the manuscript. T.P. performed the DSRT and ex vivo assays on patient samples and associated data analysis. E.J., C.C., M.M. and B.W.M. designed, performed and interpreted the crystallography experiments. M.K. and K.P. coordinated the sampling of patient material, collection of associated clinical data, and clinical translation. G.A.R. and O.K. contributed to study design and manuscript writing. J.C., P.W. and B.W.M. coordinated and performed the in vitro biochemical and cellular experiments. K.P., B.W.M. and K.W. supervised the experimental and clinical analysis. All authors discussed the results, commented and edited the manuscript.

Corresponding authors

Correspondence to Brion W. Murray or Krister Wennerberg.

Ethics declarations

Competing interests

B.W.M., P.W., C.N.C., M.M. and E.J. are all Pfizer employees.

Extended data figures and tables

Extended Data Figure 1 Axitinib adopts significantly different binding conformation in the active site of ABL1(T315I) relative to VEGFR2.

Overlay of the bound conformation of axitinib from VEGFR2 (green) (PDB ID: 4AGC) and ABL1(T315I) (purple) co-crystal structures.

Extended Data Figure 2 Axitinib exhibits selective inhibition to BCR–ABL1 gatekeeper mutants in engineered Ba/F3 cells.

a, Bar graph depicting in vitro cell proliferation data are displayed as pIC50 values. b, The corresponding IC50 values are shown in table format.

Extended Data Figure 3 Ex vivo sensitivity to BCR–ABL1 inhibitors and axitinib in primary cells derived from a CML patient harbouring the T315I mutation (FHRB.1408).

a, Waterfall plot showing the sDSS of axitinib, ponatinib, dasatinib, imatinib and nilotinib. b, Dose-response data of all approved BCR–ABL1 inhibitors and axitinib.

Extended Data Figure 4 Comparison of publicly available target specificity profiles of axitinib14,38 and ponatinib20.

The target specificity profiles were evaluated at Ki/IC50 up to tenfold ABL1(T315I) potency (Ki = 0.1 nM axitinib; IC50 = 2 nM ponatinib) illustrating that axitinib is a much less promiscuous inhibitor than ponatinib and likely to have a better safety profile in Ph+ leukaemia patients. Green labelled kinases are targeted by both inhibitors.

Extended Data Table 1 X-ray data collection and refinement statistics
Extended Data Table 2 Clinical characteristics of patients
Extended Data Table 3 Drug sensitivity scores for axitinib in a panel of BCR–ABL1(T315I)-positive, CML, AML and healthy donor control samples

Supplementary information

Supplementary Table 1

This file contains the oncology drug collection, which depicts the anti-cancer inhibitors that have been used to phenotypically profile the CML and Ph+ ALL patient samples included in the study. The names, research codes, aliases, mechanisms of action, approval status, range of tested concentrations, suppliers and suppliers references of the inhibitors are provided. (XLSX 620 kb)

Supplementary Table 2

This file contains the drug sensitivity profile of Ph+ ALL patient cells (FHRB.1278) harbouring the T315I mutation, it shows the source drug sensitivity testing results of cancer cells derived from patient FHRB.1278. The four curve fitting parameters are provided (EC50, slope, minimum and maximum inhibition) along with the % survival at each of the tested concentration of a given compound. In addition, the calculated drug sensitivity score value is shown for each of the tested inhibitors. (XLSX 147 kb)

Supplementary Table 3

This file contains drug sensitivity profiles of CML and Ph+ ALL patient samples, it shows the source drug sensitivity testing results of the remaining CML and Ph+ ALL patient samples included in the study. The four curve fitting parameters are provided (EC50, slope, minimum and maximum inhibition) along with the % survival at each of the tested concentration of a given compound. In addition, the calculated drug sensitivity score value is shown for each of the tested inhibitors. (XLSX 228 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pemovska, T., Johnson, E., Kontro, M. et al. Axitinib effectively inhibits BCR-ABL1(T315I) with a distinct binding conformation. Nature 519, 102–105 (2015). https://doi.org/10.1038/nature14119

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14119

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: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research