Original Article | Published:

Targeting MET and AXL overcomes resistance to sunitinib therapy in renal cell carcinoma

Oncogene volume 35, pages 26872697 (2016) | Download Citation

Abstract

Antiangiogenic therapy resistance occurs frequently in patients with metastatic renal cell carcinoma (RCC). The purpose of this study was to understand the mechanism of resistance to sunitinib, an antiangiogenic small molecule, and to exploit this mechanism therapeutically. We hypothesized that sunitinib-induced upregulation of the prometastatic MET and AXL receptors is associated with resistance to sunitinib and with more aggressive tumor behavior. In the present study, tissue microarrays containing sunitinib-treated and untreated RCC tissues were stained with MET and AXL antibodies. The low malignant RCC cell line 786-O was chronically treated with sunitinib and assayed for AXL, MET, epithelial–mesenchymal transition (EMT) protein expression and activation. Co-culture experiments were used to examine the effect of sunitinib pretreatment on endothelial cell growth. The effects of AXL and MET were evaluated in various cell-based models by short hairpin RNA or inhibition by cabozantinib, the multi-tyrosine kinases inhibitor that targets vascular endothelial growth factor receptor, MET and AXL. Xenograft mouse models tested the ability of cabozantinib to rescue sunitinib resistance. We demonstrated that increased AXL and MET expression was associated with inferior clinical outcome in patients. Chronic sunitinib treatment of RCC cell lines activated both AXL and MET, induced EMT-associated gene expression changes, including upregulation of Snail and β-catenin, and increased cell migration and invasion. Pretreatment with sunitinib enhanced angiogenesis in 786-0/human umbilical vein endothelial cell co-culture models. The suppression of AXL or MET expression and the inhibition of AXL and MET activation using cabozantinib both impaired chronic sunitinib treatment-induced prometastatic behavior in cell culture and rescued acquired resistance to sunitinib in xenograft models. In summary, chronic sunitinib treatment induces the activation of AXL and MET signaling and promotes prometastatic behavior and angiogenesis. The inhibition of AXL and MET activity may overcome resistance induced by prolonged sunitinib therapy in metastatic RCC.

  • Subscribe to Oncogene for full access:

    $2190

    Subscribe

Additional access options:

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

References

  1. 1.

    , , . Histopathology and classification of renal cell tumors (adenomas, oncocytomas and carcinomas). The basic cytological and histopathological elements and their use for diagnostics. Pathol Res Pract 1986; 181: 125–143.

  2. 2.

    , . Targeted therapy for renal cell carcinoma: The next lap. J Carcinog 2014; 13: 3.

  3. 3.

    , , , . Emerging antiangiogenics for renal cancer. Expert Opin Emerg Drugs 2013; 18: 495–511.

  4. 4.

    , , , , , et al. Rapid angiogenesis onset after discontinuation of sunitinib treatment of renal cell carcinoma patients. Clin Cancer Res 2012; 18: 3961–3971.

  5. 5.

    , , , , , et al. Interleukin-8 mediates resistance to antiangiogenic agent sunitinib in renal cell carcinoma. Cancer Res 2010; 70: 1063–1071.

  6. 6.

    , , , , , et al. Upfront, randomized, phase 2 trial of sorafenib versus sorafenib and low-dose interferon alfa in patients with advanced renal cell carcinoma: clinical and biomarker analysis. Cancer 2010; 116: 57–65.

  7. 7.

    , , , , , et al. Gene and protein expression markers of response to combined antiangiogenic and epidermal growth factor targeted therapy in renal cell carcinoma. Ann Oncol 2010; 21: 1599–1606.

  8. 8.

    , , , , , et al. Cytoplasmic sequestration of p27 via AKT phosphorylation in renal cell carcinoma. Clin Cancer Res 2009; 15: 81–90.

  9. 9.

    , , , , , et al. Direct regulation of GAS6/AXL signaling by HIF promotes renal metastasis through SRC and MET. Proc Natl Acad Sci USA 2014; 111: 13373–13378.

  10. 10.

    , . MET: a promising anticancer therapeutic target. Nat Rev Clin Oncol 2012; 9: 314–326.

  11. 11.

    , , , . Met metastasis, motility and more. Nat Rev Mol Cell Biol 2003; 4: 915–925.

  12. 12.

    , , , . Targeting MET in cancer: rationale and progress. Nat Rev Cancer 2012; 12: 89–103.

  13. 13.

    , , , , , et al. c-Met is a prognostic marker and potential therapeutic target in clear cell renal cell carcinoma. Ann Oncol 2013; 24: 343–349.

  14. 14.

    , , . Presence of phosphorylated hepatocyte growth factor receptor/c-Met is associated with tumor progression and survival in patients with conventional renal cell carcinoma. Clin Cancer Res 2006; 12: 4876–4881.

  15. 15.

    , , , , , et al. Clinical relevance of hepsin and hepatocyte growth factor activator inhibitor type 2 expression in renal cell carcinoma. Cancer Sci 2007; 98: 491–498.

  16. 16.

    , , , , , et al. Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discov 2012; 2: 270–287.

  17. 17.

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

  18. 18.

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

  19. 19.

    , , , , , et al. Multiple roles for the receptor tyrosine kinase axl in tumor formation. Cancer Res 2005; 65: 9294–9303.

  20. 20.

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

  21. 21.

    , , , . Taking aim at Mer and Axl receptor tyrosine kinases as novel therapeutic targets in solid tumors. Expert Opin Ther Targets 2010; 14: 1073–1090.

  22. 22.

    , , , , , et al. Profiling phospho-signaling networks in breast cancer using reverse-phase protein arrays. Oncogene 2012; 32: 3470–3476.

  23. 23.

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

  24. 24.

    , . Axl is essential for VEGF-A-dependent activation of PI3K/Akt. EMBO J 2012; 31: 1692–1703.

  25. 25.

    , , , , . Expression of the proto-oncogene Axl in renal cell carcinoma. DNA Cell Biol 2003; 22: 533–540.

  26. 26.

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

  27. 27.

    , , , . Cytoplasmic ACK1 interaction with multiple receptor tyrosine kinases is mediated by Grb2: an analysis of ACK1 effects on Axl signaling. J Biol Chem 2009; 284: 34954–34963.

  28. 28.

    , , , , , et al. Preclinical validation of AXL receptor as a target for antibody-based pancreatic cancer immunotherapy. Oncogene 2014; 33: 5405–5414.

  29. 29.

    , , , , . Gas6 and the receptor tyrosine kinase Axl in clear cell renal cell carcinoma. PLoS One 2009; 4: e7575.

  30. 30.

    , , , , , . Biological role of HGF/MET pathway in renal cell carcinoma. J Urol 1999; 161: 990–997.

  31. 31.

    , , . Potentiation of human estrogen receptor alpha-mediated gene expression by steroid receptor coactivator-1 (SRC-1) in Saccharomyces cerevisiae. J Steroid Biochem Mol Biol 2003; 86: 15–26.

  32. 32.

    , , , , . c-Src kinase activity is required for hepatocyte growth factor-induced motility and anchorage-independent growth of mammary carcinoma cells. J Biol Chem 1998; 273: 33714–33721.

  33. 33.

    , . Hepatocyte growth factor-induced c-Src-phosphatidylinositol 3-kinase-AKT-mammalian target of rapamycin pathway inhibits dendritic cell activation by blocking IkappaB kinase activity. Int J Biochem Cell Biol 2011; 43: 1134–1146.

  34. 34.

    , , , , , et al. TNF-alpha induces epithelial-mesenchymal transition of renal cell carcinoma cells via a GSK3beta-dependent mechanism. Mol Cancer Res 2012; 10: 1109–1119.

  35. 35.

    , , , , , et al. EGFR and MET receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and gefitinib resistance in lung cancers. Nat Med 2012; 18: 74–82.

  36. 36.

    , . Cancer invasion and the microenvironment: plasticity and reciprocity. Cell 2011; 147: 992–1009.

  37. 37.

    , . Targeting the hepatocyte growth factor/c-Met signaling pathway in renal cell carcinoma. Cancer J 2013; 19: 316–323.

  38. 38.

    , , , , , et al. Sunitinib significantly suppresses the proliferation, migration, apoptosis resistance, tumor angiogenesis and growth of triple-negative breast cancers but increases breast cancer stem cells. Vasc Cell 2014; 6: 12.

  39. 39.

    , , , , , et al. A phase I study of cabozantinib (XL184) in patients with renal cell cancer. Ann Oncol 2014; 25: 1603–1608.

  40. 40.

    , , , , , et al. In vitro and in vivo activity of cabozantinib (XL184), an inhibitor of RET, MET, and VEGFR2, in a model of medullary thyroid cancer. Thyroid 2013; 23: 1569–1577.

  41. 41.

    , , , , . XL-184, a MET, VEGFR-2 and RET kinase inhibitor for the treatment of thyroid cancer, glioblastoma multiforme and NSCLC. IDrugs 2010; 13: 112–121.

  42. 42.

    , , , , , et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 2009; 15: 220–231.

  43. 43.

    , , , , , et al. Myelosuppression and kinase selectivity of multikinase angiogenesis inhibitors. Br J Cancer 2009; 101: 1717–1723.

  44. 44.

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

  45. 45.

    , , , , . Patterns of disease progression in metastatic renal cell carcinoma patients treated with antivascular agents and interferon: impact of therapy on recurrence patterns and outcome measures. Cancer 2009; 115: 1859–1866.

  46. 46.

    , , , , , et al. Cabozantinib in patients with advanced prostate cancer: results of a phase II randomized discontinuation trial. J Clin Oncol 2013; 31: 412–419.

  47. 47.

    , , , , , et al. Phase II presurgical feasibility study of bevacizumab in untreated patients with metastatic renal cell carcinoma. J Clin Oncol 2009; 27: 4076–4081.

  48. 48.

    . Intratumor microvessel density as a prognostic factor in cancer. Am J Pathol 1995; 147: 9–19.

Download references

Acknowledgements

This work was supported by Exelixis, Renee Kaye Cure Fur Cancer Foundation, the Monteleone Family Foundation and the UT MD Anderson Cancer Center P30 Cancer Center Support Grant.

Author information

Affiliations

  1. Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA

    • L Zhou
    • , X-D Liu
    • , M Sun
    • , X Zhang
    • , P German
    • , S Bai
    • , Z Ding
    • , N Tannir
    •  & E Jonasch
  2. Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA

    • C G Wood
    • , S F Matin
    •  & J A Karam
  3. Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA

    • P Tamboli
    • , K Sircar
    • , P Rao
    •  & A G Hoang
  4. Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA

    • E B Rankin
    •  & A J Giaccia
  5. Exelixis Inc., South San Francisco, CA, USA

    • D A Laird
  6. Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA

    • C L Walker

Authors

  1. Search for L Zhou in:

  2. Search for X-D Liu in:

  3. Search for M Sun in:

  4. Search for X Zhang in:

  5. Search for P German in:

  6. Search for S Bai in:

  7. Search for Z Ding in:

  8. Search for N Tannir in:

  9. Search for C G Wood in:

  10. Search for S F Matin in:

  11. Search for J A Karam in:

  12. Search for P Tamboli in:

  13. Search for K Sircar in:

  14. Search for P Rao in:

  15. Search for E B Rankin in:

  16. Search for D A Laird in:

  17. Search for A G Hoang in:

  18. Search for C L Walker in:

  19. Search for A J Giaccia in:

  20. Search for E Jonasch in:

Competing interests

Dr Jonasch has received research funding from Pfizer, Novartis, GSK, Onyx and Exelixis. Dr Jonasch is a consultant for Pfizer, GSK, Novartis and Genentech. The other authors declare no conflict of interest.

Corresponding author

Correspondence to E Jonasch.

Supplementary information

About this article

Publication history

Received

Revised

Accepted

Published

DOI

https://doi.org/10.1038/onc.2015.343

Rights and permissions

To obtain permission to re-use content from this article visit RightsLink.

Supplementary Information accompanies this paper on the Oncogene website (http://www.nature.com/onc)