Article

An engineered Axl 'decoy receptor' effectively silences the Gas6-Axl signaling axis

  • Nature Chemical Biology volume 10, pages 977983 (2014)
  • doi:10.1038/nchembio.1636
  • Download Citation
Received:
Accepted:
Published:

Abstract

Aberrant signaling through the Axl receptor tyrosine kinase has been associated with a myriad of human diseases, most notably metastatic cancer, identifying Axl and its ligand Gas6 as important therapeutic targets. Using rational and combinatorial approaches, we engineered an Axl 'decoy receptor' that binds Gas6 with high affinity and inhibits its function, offering an alternative approach from drug discovery efforts that directly target Axl. Four mutations within this high-affinity Axl variant caused structural alterations in side chains across the Gas6-Axl binding interface, stabilizing a conformational change on Gas6. When reformatted as an Fc fusion, the engineered decoy receptor bound Gas6 with femtomolar affinity, an 80-fold improvement compared to binding of the wild-type Axl receptor, allowing effective sequestration of Gas6 and specific abrogation of Axl signaling. Moreover, increased Gas6 binding affinity was critical and correlative with the ability of decoy receptors to potently inhibit metastasis and disease progression in vivo.

  • Subscribe to Nature Chemical Biology for full access:

    $59

    Subscribe

Additional access options:

Already a subscriber?  Log in  now or  Register  for online access.

Accessions

Primary accessions

Protein Data Bank

Referenced accessions

Protein Data Bank

References

  1. 1.

    & Immunobiology of the TAM receptors. Nat. Rev. Immunol. 8, 327–336 (2008).

  2. 2.

    et al. Axl as a potential therapeutic target in cancer: role of Axl in tumor growth, metastasis and angiogenesis. Oncogene 28, 3442–3455 (2009).

  3. 3.

    et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat. Genet. 44, 852–860 (2012).

  4. 4.

    , , , & ABL regulation by AXL promotes cisplatin resistance in esophageal cancer. Cancer Res. 73, 331–340 (2013).

  5. 5.

    et al. An epithelial-mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin. Cancer Res. 19, 279–290 (2013).

  6. 6.

    et al. Dominant-negative inhibition of the Axl receptor tyrosine kinase suppresses brain tumor cell growth and invasion and prolongs survival. Proc. Natl. Acad. Sci. USA 103, 5799–5804 (2006).

  7. 7.

    , , & TAM receptor tyrosine kinases: biologic functions, signaling, and potential therapeutic targeting in human cancer. Adv. Cancer Res. 100, 35–83 (2008).

  8. 8.

    et al. Differential expression of Axl and Gas6 in renal cell carcinoma reflecting tumor advancement and survival. Clin. Cancer Res. 15, 4742–4749 (2009).

  9. 9.

    , & Axl and prostasin are biomarkers for prognosis of ovarian adenocarcinoma. Ann. Diagn. Pathol. 17, 425–429 (2013).

  10. 10.

    et al. AXL is an essential factor and therapeutic target for metastatic ovarian cancer. Cancer Res. 70, 7570–7579 (2010).

  11. 11.

    et al. R428, a selective small molecule inhibitor of Axl kinase, blocks tumor spread and prolongs survival in models of metastatic breast cancer. Cancer Res. 70, 1544–1554 (2010).

  12. 12.

    et al. LY2801653 is an orally bioavailable multi-kinase inhibitor with potent activity against MET, MST1R, and other oncoproteins, and displays anti-tumor activities in mouse xenograft models. Invest. New Drugs 31, 833–844 (2013).

  13. 13.

    et al. An anti-Axl monoclonal antibody attenuates xenograft tumor growth and enhances the effect of multiple anticancer therapies. Oncogene 29, 5254–5264 (2010).

  14. 14.

    et al. Preclinical validation of AXL receptor as a target for antibody-based pancreatic cancer immunotherapy. Oncogene 10.1038/onc.2013.487 (2013).

  15. 15.

    et al. Targeting Axl with an high-affinity inhibitory aptamer. Mol. Ther. 20, 2291–2303 (2012).

  16. 16.

    et al. The E3 ligase Cbl-b and TAM receptors regulate cancer metastasis via natural killer cells. Nature 507, 508–512 (2014).

  17. 17.

    et al. A small molecule–kinase interaction map for clinical kinase inhibitors. Nat. Biotechnol. 23, 329–336 (2005).

  18. 18.

    et al. A novel site contributing to growth-arrest–specific gene 6 binding to its receptors as revealed by a human monoclonal antibody. Biochem. J. 387, 727–735 (2005).

  19. 19.

    et al. Axl receptor tyrosine kinase stimulated by the vitamin-K–dependent protein encoded by growth-arrest-specific gene-6. Nature 373, 623–626 (1995).

  20. 20.

    , , & The receptor AXL diversifies EGFR signaling and limits the response to EGFR-targeted inhibitors in triple-negative breast cancer cells. Sci. Signal. 6, ra66 (2013).

  21. 21.

    , & Beyond antibodies: using biological principles to guide the development of next-generation protein therapeutics. Curr. Opin. Biotechnol. 24, 1072–1077 (2013).

  22. 22.

    et al. Cytokine traps: multi-component, high-affinity blockers of cytokine action. Nat. Med. 9, 47–52 (2003).

  23. 23.

    , & A tumor necrosis factor (TNF) receptor-IgG heavy chain chimeric protein as a bivalent antagonist of TNF activity. J. Exp. Med. 174, 1483–1489 (1991).

  24. 24.

    et al. VEGF-Trap: a VEGF blocker with potent antitumor effects. Proc. Natl. Acad. Sci. USA 99, 11393–11398 (2002).

  25. 25.

    et al. Structural basis for Gas6-Axl signalling. EMBO J. 25, 80–87 (2006).

  26. 26.

    & Kinetic exclusion assay technology: characterization of molecular interactions. Assay Drug Dev. Technol. 2, 647–657 (2004).

  27. 27.

    & Gas6 and protein S. Vitamin K–dependent ligands for the Axl receptor tyrosine kinase subfamily. FEBS J. 273, 5231–5244 (2006).

  28. 28.

    , , & The protein encoded by a growth arrest-specific gene (gas6) is a new member of the vitamin K–dependent proteins related to protein S, a negative coregulator in the blood coagulation cascade. Mol. Cell. Biol. 13, 4976–4985 (1993).

  29. 29.

    et al. Crystal structure of a C-terminal fragment of growth arrest-specific protein Gas6—receptor tyrosine kinase activation by laminin G–like domains. J. Biol. Chem. 277, 44164–44170 (2002).

  30. 30.

    , & Helix capping propensities in peptides parallel those in proteins. Proc. Natl. Acad. Sci. USA 90, 11332–11336 (1993).

  31. 31.

    et al. Exploiting a natural conformational switch to engineer an interleukin-2 'superkine'. Nature 484, 529–533 (2012).

  32. 32.

    , , & Rational design and engineering of therapeutic proteins. Drug Discov. Today 8, 212–221 (2003).

  33. 33.

    et al. Axl is an essential epithelial-to-mesenchymal transition-induced regulator of breast cancer metastasis and patient survival. Proc. Natl. Acad. Sci. USA 107, 1124–1129 (2010).

  34. 34.

    & Selective events in the metastatic process defined by analysis of the sequential dissemination of subpopulations of a mouse mammary tumor. Cancer Res. 52, 1399–1405 (1992).

  35. 35.

    , & Tumor metastasis: moving new biological insights into the clinic. Nat. Med. 19, 1450–1464 (2013).

  36. 36.

    et al. Tyro-3 family receptors are essential regulators of mammalian spermatogenesis. Nature 398, 723–728 (1999).

  37. 37.

    et al. Deficiency or inhibition of Gas6 causes platelet dysfunction and protects mice against thrombosis. Nat. Med. 7, 215–221 (2001).

  38. 38.

    et al. The E3 ligase Cbl-b and TAM receptors regulate cancer metastasis via natural killer cells. Nature 507, 508–512 (2014).

  39. 39.

    & Yeast surface display for screening combinatorial polypeptide libraries. Nat. Biotechnol. 15, 553–557 (1997).

  40. 40.

    et al. Isolating and engineering human antibodies using yeast surface display. Nat. Protoc. 1, 755–768 (2006).

  41. 41.

    Xds. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010).

  42. 42.

    & Molecular replacement with MOLREP. Acta Crystallogr. D Biol. Crystallogr. 66, 22–25 (2010).

  43. 43.

    & Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

  44. 44.

    , & Refinement of macromolecular structures by the maximum-likelihood method. Acta Crystallogr. D Biol. Crystallogr. 53, 240–255 (1997).

  45. 45.

    The PyMOL Molecular Graphics System. Version 1.3r1, Schrödinger, LLC (2010).

  46. 46.

    & Inference of macromolecular assemblies from crystalline state. J. Mol. Biol. 372, 774–797 (2007).

  47. 47.

    , & Flexibility analysis of enzyme active sites by crystallographic temperature factors. Protein Eng. 16, 109–114 (2003).

Download references

Acknowledgements

The authors thank E. Hohenester (Imperial College London) for his generous gift of the Gas6 plasmid and helpful discussions regarding protein production, L.L. Zheng for technical assistance with protein production and purification and the Stanford FACS Core Facility for assistance with the flow cytometric sorting. All in vivo and ex vivo imaging was conducted in the Stanford Small Animal Imaging Facility. Portions of this research were performed at the Stanford Synchrotron Radiation Laboratory, a national user facility operated by Stanford University on behalf of the US Department of Energy Office of Basic Energy Sciences. This work was supported by the Wallace H. Coulter Translational Research Grant Program (J.R.C. and A.J.G.), the Stanford Chemistry, Engineering & Medicine for Human Health (ChEM-H) Institute (J.R.C. and I.I.M.), Stanford Bio-X and ARCS Graduate Fellowships (M.S.K.), US National Institutes of Health (NIH) grants CA-088480 and CA-67166 (A.J.G. and Y.R.M.), NIH National Institute of General Medical Sciences Training Grant T32 GM008412 and the Siebel Graduate Fellowship (D.S.J.).

Author information

Affiliations

  1. Department of Bioengineering, Stanford University, Stanford, California, USA.

    • Mihalis S Kariolis
    • , Douglas S Jones II
    • , Shiven Kapur
    •  & Jennifer R Cochran
  2. Department of Radiation Oncology, Stanford University School of Medicine, Stanford University, Stanford, California, USA.

    • Yu Rebecca Miao
    •  & Amato J Giaccia
  3. Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California, USA.

    • Irimpan I Mathews
  4. Department of Chemical Engineering, Stanford University, Stanford, California, USA.

    • Jennifer R Cochran

Authors

  1. Search for Mihalis S Kariolis in:

  2. Search for Yu Rebecca Miao in:

  3. Search for Douglas S Jones in:

  4. Search for Shiven Kapur in:

  5. Search for Irimpan I Mathews in:

  6. Search for Amato J Giaccia in:

  7. Search for Jennifer R Cochran in:

Contributions

M.S.K., Y.R.M., D.S.J., S.K., I.I.M., A.J.G. and J.R.C. designed the research; M.S.K. and D.S.J. performed protein engineering and characterization; M.S.K., S.K. and I.I.M. did the crystallography; M.S.K. and Y.R.M. conducted the in vivo experiments; all of the authors analyzed the data; and M.S.K. prepared the manuscript with input from all co-authors.

Competing interests

Stanford University holds patent number US8618254 B2, 'Biologic inhibitors for therapeutic targeting the receptor tyrosine kinase AXL', which is related to the work described in this paper, with M.S.K., Y.R.M., D.S.J., A.J.G. and J.R.C. named as inventors. A.J.G. is a founder of Ruga, a company that discovers and develops targeted therapeutics in oncology that has licensed this patent.

Corresponding authors

Correspondence to Amato J Giaccia or Jennifer R Cochran.

Supplementary information

PDF files

  1. 1.

    Supplementary Text and Figures

    Supplementary Results, Supplementary Figures 1–14 and Supplementary Tables 1–7.