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.

  • Article
  • Published:

A TNF–JNK–Axl–ERK signaling axis mediates primary resistance to EGFR inhibition in glioblastoma

Abstract

Aberrant epidermal growth factor receptor (EGFR) signaling is widespread in cancer, making the EGFR an important target for therapy. EGFR gene amplification and mutation are common in glioblastoma (GBM), but EGFR inhibition has not been effective in treating this tumor. Here we propose that primary resistance to EGFR inhibition in glioma cells results from a rapid compensatory response to EGFR inhibition that mediates cell survival. We show that in glioma cells expressing either EGFR wild type or the mutant EGFRvIII, EGFR inhibition triggers a rapid adaptive response driven by increased tumor necrosis factor (TNF) secretion, which leads to activation in turn of c-Jun N-terminal kinase (JNK), the Axl receptor tyrosine kinase and extracellular signal–regulated kinases (ERK). Inhibition of this adaptive axis at multiple nodes rendered glioma cells with primary resistance sensitive to EGFR inhibition. Our findings provide a possible explanation for the failures of anti-EGFR therapy in GBM and suggest a new approach to the treatment of EGFR-expressing GBM using a combination of EGFR and TNF inhibition.

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: EGFR inhibition triggers an adaptive response in glioma cells.
Figure 2: EGFR inhibition–induced Axl and ERK activation is mediated by JNK.
Figure 3: EGFR inhibition leads to an increase in GAS6 via a JNK-dependent mechanism.
Figure 4: EGFR inhibition leads to increased TNF signaling that triggers an adaptive signaling pathway.
Figure 5: Inhibition of JNK and ERK renders glioma cells sensitive to EGFR inhibition.
Figure 6: TNF inhibition sensitizes glioma cells to EGFR inhibition.
Figure 7: JNK or TNF inhibition sensitizes mouse tumors to EGFR inhibition in vivo.

Similar content being viewed by others

References

  1. Sharma, S.V., Bell, D.W., Settleman, J. & Haber, D.A. Epidermal growth factor receptor mutations in lung cancer. Nat. Rev. Cancer 7, 169–181 (2007).

    CAS  PubMed  Google Scholar 

  2. Arteaga, C.L. & Engelman, J.A. ERBB receptors: from oncogene discovery to basic science to mechanism-based cancer therapeutics. Cancer Cell 25, 282–303 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Chong, C.R. & Jänne, P.A. The quest to overcome resistance to EGFR-targeted therapies in cancer. Nat. Med. 19, 1389–1400 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Fan, W. et al. MET-independent lung cancer cells evading EGFR kinase inhibitors are therapeutically susceptible to BH3 mimetic agents. Cancer Res. 71, 4494–4505 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Lee, H.J. et al. Drug resistance via feedback activation of Stat3 in oncogene-addicted cancer cells. Cancer Cell 26, 207–221 (2014).

    CAS  PubMed  Google Scholar 

  6. Verhaak, R.G. et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17, 98–110 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Hatanpaa, K.J., Burma, S., Zhao, D. & Habib, A.A. Epidermal growth factor receptor in glioma: signal transduction, neuropathology, imaging, and radioresistance. Neoplasia 12, 675–684 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Mellinghoff, I.K. et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N. Engl. J. Med. 353, 2012–2024 (2005).

    CAS  PubMed  Google Scholar 

  9. Karpel-Massler, G., Schmidt, U., Unterberg, A. & Halatsch, M.E. Therapeutic inhibition of the epidermal growth factor receptor in high-grade gliomas: where do we stand? Mol. Cancer Res. 7, 1000–1012 (2009).

    CAS  PubMed  Google Scholar 

  10. Akhavan, D. et al. De-repression of PDGFRβ transcription promotes acquired resistance to EGFR tyrosine kinase inhibitors in glioblastoma patients. Cancer Discov. 3, 534–547 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Nathanson, D.A. et al. Targeted therapy resistance mediated by dynamic regulation of extrachromosomal mutant EGFR DNA. Science 343, 72–76 (2014).

    CAS  PubMed  Google Scholar 

  12. Barkovich, K.J. et al. Kinetics of inhibitor cycling underlie therapeutic disparities between EGFR-driven lung and brain cancers. Cancer Discov. 2, 450–457 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Wykosky, J. et al. A urokinase receptor-Bim signaling axis emerges during EGFR inhibitor resistance in mutant EGFR glioblastoma. Cancer Res. 75, 394–404 (2015).

    CAS  PubMed  Google Scholar 

  14. Chakraborty, S. et al. Constitutive and ligand-induced EGFR signalling triggers distinct and mutually exclusive downstream signalling networks. Nat. Commun. 5, 5811 (2014).

    CAS  PubMed  Google Scholar 

  15. Guo, G. et al. Ligand-independent EGFR signaling. Cancer Res. 75, 3436–3441 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Acquaviva, J. et al. Chronic activation of wild-type epidermal growth factor receptor and loss of Cdkn2a cause mouse glioblastoma formation. Cancer Res. 71, 7198–7206 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Endres, N.F. et al. Conformational coupling across the plasma membrane in activation of the EGF receptor. Cell 152, 543–556 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Ramnarain, D.B. et al. Differential gene expression analysis reveals generation of an autocrine loop by a mutant epidermal growth factor receptor in glioma cells. Cancer Res. 66, 867–874 (2006).

    CAS  PubMed  Google Scholar 

  19. Wong, A.J. et al. Structural alterations of the epidermal growth factor receptor gene in human gliomas. Proc. Natl. Acad. Sci. USA 89, 2965–2969 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Gil Del Alcazar, C.R., Todorova, P.K., Habib, A.A., Mukherjee, B. & Burma, S. Augmented HR repair mediates acquired temozolomide resistance in glioblastoma. Mol. Cancer Res. 14, 928–940 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Pingle, S.C. et al. In silico modeling predicts drug sensitivity of patient-derived cancer cells. J. Transl. Med. 12, 128 (2014).

    PubMed  PubMed Central  Google Scholar 

  22. Puliyappadamba, V.T. et al. Opposing effect of EGFRWT on EGFRvIII-mediated NF-κB activation with RIP1 as a cell death switch. Cell Rep. 4, 764–775 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Sarkaria, J.N. et al. Use of an orthotopic xenograft model for assessing the effect of epidermal growth factor receptor amplification on glioblastoma radiation response. Clin. Cancer Res. 12, 2264–2271 (2006).

    CAS  PubMed  Google Scholar 

  24. Stommel, J.M. et al. Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science 318, 287–290 (2007).

    CAS  PubMed  Google Scholar 

  25. Huang, P.H. et al. Quantitative analysis of EGFRvIII cellular signaling networks reveals a combinatorial therapeutic strategy for glioblastoma. Proc. Natl. Acad. Sci. USA 104, 12867–12872 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Pillay, V. et al. The plasticity of oncogene addiction: implications for targeted therapies directed to receptor tyrosine kinases. Neoplasia 11, 448–458 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Greenall, S.A. et al. EGFRvIII-mediated transactivation of receptor tyrosine kinases in glioma: mechanism and therapeutic implications. Oncogene 34, 5277–5287 (2015).

    CAS  PubMed  Google Scholar 

  28. Awad, A.J., Burns, T.C., Zhang, Y. & Abounader, R. Targeting MET for glioma therapy. Neurosurg. Focus 37, E10 (2014).

    PubMed  Google Scholar 

  29. Zhu, Y. & Shah, K. Multiple lesions in receptor tyrosine kinase pathway determine glioblastoma response to pan-ERBB inhibitor PF-00299804 and PI3K/mTOR dual inhibitor PF-05212384. Cancer Biol. Ther. 15, 815–822 (2014).

    PubMed  PubMed Central  Google Scholar 

  30. Blakely, C.M. et al. NF-κB-activating complex engaged in response to EGFR oncogene inhibition drives tumor cell survival and residual disease in lung cancer. Cell Rep. 11, 98–110 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Wajant, H., Pfizenmaier, K. & Scheurich, P. Tumor necrosis factor signaling. Cell Death Differ. 10, 45–65 (2003).

    CAS  PubMed  Google Scholar 

  32. Higuchi, M. & Aggarwal, B.B. TNF induces internalization of the p60 receptor and shedding of the p80 receptor. J. Immunol. 152, 3550–3558 (1994).

    CAS  PubMed  Google Scholar 

  33. Deng, L., Ding, W. & Granstein, R.D. Thalidomide inhibits tumor necrosis factor-alpha production and antigen presentation by Langerhans cells. J. Invest. Dermatol. 121, 1060–1065 (2003).

    CAS  PubMed  Google Scholar 

  34. Gross, N.D. et al. Inhibition of Jun NH2-terminal kinases suppresses the growth of experimental head and neck squamous cell carcinoma. Clin. Cancer Res. 13, 5910–5917 (2007).

    CAS  PubMed  Google Scholar 

  35. Groves, M.D. et al. A North American Brain Tumor Consortium (NABTC 99-04) phase II trial of temozolomide plus thalidomide for recurrent glioblastoma multiforme. J. Neurooncol. 81, 271–277 (2007).

    CAS  PubMed  Google Scholar 

  36. Gu, X. et al. Intraperitoneal injection of thalidomide attenuates bone cancer pain and decreases spinal tumor necrosis factor-α expression in a mouse model. Mol. Pain 6, 64 (2010).

    PubMed  PubMed Central  Google Scholar 

  37. Prahallad, A. et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 483, 100–103 (2012).

    CAS  PubMed  Google Scholar 

  38. Duncan, J.S. et al. Dynamic reprogramming of the kinome in response to targeted MEK inhibition in triple-negative breast cancer. Cell 149, 307–321 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Huang, P.H., Xu, A.M. & White, F.M. Oncogenic EGFR signaling networks in glioma. Sci. Signal 2, re6 (2009).

    PubMed  Google Scholar 

  40. Li, L. et al. An EGFR wild type-EGFRvIII-HB-EGF feed-forward loop regulates the activation of EGFRvIII. Oncogene 33, 4253–4264 (2014).

    CAS  PubMed  Google Scholar 

  41. Fan, Q.W. et al. EGFR phosphorylates tumor-derived EGFRvIII driving STAT3/5 and progression in glioblastoma. Cancer Cell 24, 438–449 (2013).

    CAS  PubMed  Google Scholar 

  42. Jahani-Asl, A. et al. Control of glioblastoma tumorigenesis by feed-forward cytokine signaling. Nat. Neurosci. 19, 798–806 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. de la Iglesia, N. et al. Identification of a PTEN-regulated STAT3 brain tumor suppressor pathway. Genes Dev. 22, 449–462 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Rich, J.N. et al. Phase II trial of gefitinib in recurrent glioblastoma. J. Clin. Oncol. 22, 133–142 (2004).

    CAS  PubMed  Google Scholar 

  45. Brown, P.D. et al. Phase I/II trial of erlotinib and temozolomide with radiation therapy in the treatment of newly diagnosed glioblastoma multiforme: North Central Cancer Treatment Group Study N0177. J. Clin. Oncol. 26, 5603–5609 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Peereboom, D.M. et al. Phase II trial of erlotinib with temozolomide and radiation in patients with newly diagnosed glioblastoma multiforme. J. Neurooncol. 98, 93–99 (2010).

    CAS  PubMed  Google Scholar 

  47. Schuster, J. et al. A phase II, multicenter trial of rindopepimut (CDX-110) in newly diagnosed glioblastoma: the ACT III study. Neuro-oncol. 17, 854–861 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Inda, M.D. et al. Tumor heterogeneity is an active process maintained by a mutant EGFR-induced cytokine circuit in glioblastoma. Genes Dev. 24, 1731–1745 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Furnari, F.B., Cloughesy, T.F., Cavenee, W.K. & Mischel, P.S. Heterogeneity of epidermal growth factor receptor signalling networks in glioblastoma. Nat. Rev. Cancer 15, 302–310 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Stupp, R. et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 352, 987–996 (2005).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank F. Furnari (University of California, San Diego), C.D. James (Northwestern University) and E. Burstein (UT Southwestern Medical Center) for providing cell lines and reagents. This work was supported in part by NIH grants RO1 NS062080 and by the Office of Medical Research, Departments of Veterans Affairs to A.A.H. (I01BX002559). S.B. is supported by grants from the National Institutes of Health (RO1CA149461, RO1CA197796 and R21CA202403) and the National Aeronautics and Space Administration (NNX16AD78G). This work was also supported by NIH grant 1R01CA194578 to D.Z.

Author information

Authors and Affiliations

Authors

Contributions

A.A.H., G.G. and K.G. designed experiments. G.G., K.G., S.A., N.A., S.S. and D.H.W. performed or assisted with experiments. B.M., K.J.H., J.N.S. and S.K. provided tumor samples. E.P., S.B., D.Z., J.N.S., G.G. and A.A.H. analyzed data. A.A.H. and G.G. wrote the manuscript with contributions from K.G., E.P. and K.J.H. A.A.H. supervised the study.

Corresponding author

Correspondence to Amyn A Habib.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 EGFR inhibition activates signaling pathways

(a-b) EGFR inhibition activates signaling pathways (a) U251EGFR cells were treated with erlotinib (1μM) for the indicated times followed by Western blot with the indicated antibodies. (b) GBM9 neurospheres were treated with afatinib (100nM) for the indicated time points followed by Western blot with the indicated antibodies. (c-g) EGFR inhibition decreases pSTAT3 and pAKT activation. Glioma cells were treated with high dosage of erlotinib (10μM) or afatinib (10μM). Western blot showing that pSTAT3 and pAKT decrease upon treatment with erlotinib or afatinib. Western blots are representative of at least three independent replicates. (h-j) pERK is activated in U87EGFRwt and blocked by cetuximab. (h) U87EGFRwt cells were cultured in medium with (10%) or without serum in DMEM. pERK is activated under both conditions. (i) Cells were serum starved and treated overnight with cetuximab (100 μg/ml). Cetuximab treatment blocks both EGFR and ERK activation. (j) Cells were treated wtith EGF (50 ng/ml) for 15 min. EGF increases pERK level in U87EGFRwt cells. SF:Serum free. Western blots are representative of at least three independent replicates. Full-length blots are presented in Supplementary Figure 15.

Supplementary Figure 2 Intracellular signaling networks and biological responses to EGFR inhibition in glioma cells.

(a-b) EGFR inhibition does not activate NF-κB transcriptional activity. (a) A luciferase reporter assay shows that EGFR inhibition with erlotinib does not result in an increase in NF-κB reporter activity in GBM9 neurospheres and U87EGFRwt cells. Erlotinib was used for 24h at a concentration of 100 nM for GBM9 neurospheres and 1uM for U87EGFRwt cells. GBM9: Ctrl vs erlotinib: P =0.88, t=0.16, d.f.=4; U87EGFRwt: Ctrl vs erlotinib: P =0.60, t=0.57, d.f.=4. (b) As a positive control we used LPS (1μg/ml) which activates the NF-κB reporter. Renilla luciferase was used as an internal control. Ctrl vs LPS: P =0.0012, t=8.27, d.f.=4. Data are presented as mean±s.e.m; **P < 0.01 from two-tailed unpaired Student's t-test (n = 3 biologically independent experimental replicates). n.s. not significant. (c-d) Upregulation of TNF in response to EGFR inhibition in multiple patient derived samples and established cell lines. (c) SK987, SK748. SK1422, and GBM622 are patient derived primary GBM cells cultured as neurospheres and treated with erlotinib (100nM) for 24h followed by extraction of RNA and qRT-PCR for TNF. SB19vIII and U251EGFRwt cells are established GBM cell lines that were treated with erlotinib (1μM) for 24h followed by qRT-PCR for TNF. SB19vIII: Ctrl vs Erlotinib: P =0.0014,t=7.88, d.f.=4; SK987: Ctrl vs erlotinib: P =0.0024,t=6.83, d.f.=4; SK748: Ctrl vs Erlotinib: P =0.0019, t=7.32, d.f.=4; SK1422: Ctrl vs erlotinib: P =0.0030,t=6.45, d.f.=4; GBM622: Ctrl vs erlotinib: P =0.21,t=1.5, d.f.=4; U251EGFRwt: Ctrl vs erlotinib: P =0.0005, t=10.4, d.f.=4. Data are presented as mean±s.e.m; **P < 0.01, ***P<0.001 from two-tailed unpaired Student's t-test (n = 3 biologically independent experimental replicates). n.s. not significant. (d) Western blot showing EGFR levels in various neurospheres and cell lines. No increase in TNF is detected in GBM622 cells expressing a very low level of EGFR. Western blots are representative of at least three independent replicates. Full-length blots are presented in Supplementary Figure 15. (e) Downregulation of TNFR1 in response to EGFR inhibition in patient derived primary GBM neurosphere cultures and established cell lines. GBM9, SK987 and U87EGFRwt cells were treated with erlotinib for the indicated time points followed by Western blot with TNFR1 antibodies. Erlotinib was used at a concentration of 100 nM for GBM9 and SK987 and 1uM for U87EGFRwt cells. Western blots are representative of at least three independent replicates. (f) EGFR inhibition induced effects on cell viability are influenced by specific inhibitors of downstream signaling pathways. In this experiment primary GBM9 neurospheres were exposed to various inhibitors alone or in combination with erlotinib (100 nM) for 72h followed by Alamarblue Cell Viability assay. The concentrations of the various drugs were: SB203850 (10uM), SP600125 (1uM), U0126 (1uM), BMS-345541 (10uM), XL765 (10uM), Necrostatin-1 (300nM). Erlotinib vs erlotinib +SP600125: P =0.0001,t=14.96, d.f.=4; Erlotinib vs erlotinib +U0126: P =0.0018, t=7.42, d.f.=4. Data are presented as mean±s.e.m; **P < 0.01, ***P<0.001 from two-tailed unpaired Student's t-test (n = 3 biologically independent experimental replicates). (g-i) Western blot showing efficient silencing of JNK1/2, Axl and TNFR1. For cell viability experiments involving siRNA knockdown of JNK1/2, Axl and TNFR1, we confirmed silencing by Western blot. Western blots are representative of at least three independent replicates. Full-length blots are presented in Supplementary Figure 15.

Supplementary Figure 3 EGFR inhibition induces a biologically significant upregulation of TNF in glioma cells.

(a-b) Thalidomide blocks EGFR inhibition induced upregulation of TNF. Patient derived GBM9 neurospheres or established GBM cell line U87EGFRwt cells were treated with erlotinib with or without thalidomide (1μM) followed by collection of supernatants and TNF ELISA. (a) Erlotinib vs erlotinib +thalidomide: P =0.0015, t=7.77, d.f.=4. (b) Erlotinib vs erlotinib +thalidomide: P =0.0008, t=8.98, d.f.=4. Data are presented as mean±s.e.m; **P < 0.01, ***P<0.001 from two-tailed unpaired Student's t-test (n = 3 biologically independent experimental replicates). (c-d) A neutralizing antibody to TNF sensitizes cells to EGFR inhibition. Patient derived GBM9 neurospheres or established GBM cell line U87EGFRwt cells were exposed to erlotinib plus either normal mouse IgG (Ctrl) or a neutralizing antibody to TNF. The TNF neutralizing antibody sensitizes cells to EGFR inhibition as shown by the Alamarblue Cell Viability assay. (c) Erlotinib vs erlotinib+TNF Ab: P =0.0013,t=8.07, d.f.=4. (d) Erlotinib vs erlotinib+TNF Ab: P =0.0012, t=8.13, d.f.=4. Data are presented as mean±s.e.m; **P < 0.01 from two-tailed unpaired Student's t-test (n =3 biologically independent experimental replicates). (e-f) Thalidomide sensitizes patient derived primary GBM9 and GBM39 neurospheres to EGFR inhibiton. GBM9 or GBM39 neurospheres were exposed to EGFR inhibition using afatinib (100nM) and thalidomide (1μM) alone or in combination concurrently for 72h followed by Alamarblue Cell Viability Assay. DMSO was used as a control (Ctrl). (e) Afatinib vs afatinib+thalidomide: P =0.0001,t=14.47, d.f.=4. (f) Afatinib vs afatinib+thalidomide: P =0.0005, t=10.4, d.f.=4. Data are presented as mean±s.e.m; ***P<0.001 from two-tailed unpaired Student's t-test (n =3 biologically independent experimental replicates).

Supplementary Figure 4 Inhibition of JNK and ERK sensitizes glioma cells to EGFR inhibition.

(a-b) GBM9 cells were treated with erlotinib and/or JNK inhibitor SP600125 (1μM), ERK inhibitor U0126 (1μM) for 72 hours followed by Annexin-FACS assay. Unstained cells represent viable cells. Annexin positive cells are undergoing apoptosis. PI (Propidium iodide) positive cells are undergoing late apoptosis. In “Control” cells are treated with control vehicle (DMSO). The combination treatment decreases the number of viable cells and there is an increase in double stained cells (Annexin+PI Positive). (b) Unstained: Erlotinib vs Erlotinib+SP600125: P =0.0008, t=9.10, d.f.=4; Erlotinib vs erlotinib+U0126: P =0.0036, t=6.14, d.f.=4. Annexin+PI: Erlotinib vs erlotinib+SP600125: P =0.0003, t=11.37, d.f.=4; Erlotinib vs erlotinib+U0126: P =0.0021,t=7.04, d.f.=4. (c-d) A similar experiment was conducted in U87EGFR cells with a similar result. (d) Unstained: Erlotinib vs erlotinib+SP600125: P =0.0045, t=5.77, d.f.=4; Erlotinib vs erlotinib+U0126: P =0.0013,t=8.06, d.f.=4. Annexin+PI: Erlotinib vs erlotinib+SP600125: P =0.0017, t=7.53, d.f.=4; Erlotinib vs erlotinib+U0126: P =0.0006, t=10.04, d.f.=4. Data are presented as mean±s.e.m; **P<0.01, ***P<0.001 using two-tailed unpaired Student's t-test (n =3 biologically independent experimental replicates).

Supplementary Figure 5 Axl and TNF inhibition renders glioma cells sensitive to EGFR inhibition.

(a-b) GBM9 cells were treated with erlotinib and/or Axl inhibitor R428 (1μM), Thalidomide (1μM) for 72 hours followed by Annexin-FACS assay. Unstained cells represent viable cells. Annexin positive cells are undergoing apoptosis. PI (Propidium iodide) positive cells are undergoing late apoptosis. In “Control” cells are treated with control vehicle (DMSO). The combination treatment decreases the number of viable cells and there is an increase in both PI and double stained cells (Annexin+PI Positive). (b) Unstained: Erlotinib vs erlotinib+R428: P =0.0051,t=5.58, d.f.=4; Erlotinib vs erlotinib+thalidomide: P =0.0009,t=8.75, d.f.=4. Annexin+PI: Erlotinib vs erlotinib+R428: P =0.015, t=4.10, d.f.=4; Erlotinib vs erlotinib+thalidomide: P =0.0011,t=8.32, d.f.=4. (c-d) A similar experiment was conducted in U87EGFR cells with a similar result. (d) Unstained: Erlotinib vs erlotinib+R428: P =0.0045, t=5.71, d.f.=4; Erlotinib vs erlotinib+thalidomide: P =0.0036, t=6.13, d.f.=4. Annexin+PI: Erlotinib vs erlotinib+R428: P =0.0014, t=7.83, d.f.=4; Erlotinib vs erlotinib+thalidomide: P =0.0022, t=6.98, d.f.=4. Data are presented as mean±s.e.m; *P<0.05, **P<0.01, ***P<0.001 using two-tailed unpaired Student's t-test (n =3 biologically independent experimental replicates).

Supplementary Figure 6 Etanercept renders glioma cells sensitive to EGFR inhibition.

(a-b) GBM9 cells were treated with erlotinib and/or enbrel (1μM) for 72 hours followed by Annexin-FACS assay. Unstained cells represent viable cells. Annexin positive cells are undergoing apoptosis. PI (Propidium iodide) positive cells are undergoing late apoptosis. In “Control” cells are treated with control vehicle (DMSO). The combination treatment decreases the number of viable cells and there is an increase in double stained cells (Annexin+PI Positive). (b) Unstained: Erlotinib vs erlotinib+Enbrel: P =0.0008, t=9.11, d.f.=4; Annexin+PI: Erlotinib vs erlotinib+Enbrel: P=0.0012, t=8.18, d.f.=4. (c-d) A similar experiment was conducted in U87EGFRwt cells with a similar result. (d) Unstained: Erlotinib vs erlotinib+Enbrel: P =0.0006, t=9.6, d.f.=4; Annexin+PI: Erlotinib vs erlotinib+Enbrel: P =0.0003, t=11.34, d.f.=4. Data are presented as mean±s.e.m; **P<0.01, ***P<0.001 using two-tailed unpaired Student's t-test (n =3 biologically independent experimental replicates).

Supplementary Figure 7 Combination treatment increases caspase 3/7 activity in glioma cells.

(a) GBM9 cells were treated by erlotinib and/or SP600125, U1026 for 72 hours followed by Caspase-Glo 3/7 Assay. Caspase3/7 activity was evaluated in GBM9 cells exposed to erlotinib plus SP600125. Similar results were obtained using a combination of erlotinib and U1026. Erlotinib vs erlotinib+SP600125: P =0.001, t=8.66, d.f.=4; Erlotinib vs erlotinib+U0126: P =0.0002, t=13.24, d.f.=4. Similar procedures were performed in b-f and similar results were obtained. (b) Erlotinib vs erlotinib+R428: P =0.0004, t=10.76, d.f.=4; Erlotinib vs erlotinib+thalidomide: P=0.0004, t=10.64, d.f.=4. (c) Erlotinib vs erlotinib+SP600125: P<0.0001, t=21.41, d.f.=4; Erlotinib vs erlotinib+U1026: P =0.0007, t=9.49, d.f.=4. (d) Erlotinib vs erlotinib+R428: P=0.0001, t=14.8, d.f.=4; Erlotinib vs erlotinib+T: P <0.0001, t=16.77, d.f.=4. (e) Erlotinib vs erlotinib+enbrel: P =0.0096, t=8.72, d.f.=4. (f) Erlotinib vs erlotinib+enbrel: P =0.0017, t=7.43, d.f.=4. Data are presented as mean±s.e.m; **P<0.01, ***P<0.001, ****P<0.0001 from two-tailed unpaired Student's t-test (n =3 biologically independent experimental replicates).

Supplementary Figure 8 Combination treatment inhibits glioma cell proliferation.

(a) GBM9 cells were plated in 6 well plated and treated with drugs as indicated. Cell proliferation was measured through cell counting using the trypan blue dye exclusion test. A combination of erlotinib and SP600125 (SP) significantly reduces cell growth after 72 hours. Similar results were shown using a combination of erlotinb and U1026 (U). Erlotinib vs erlotinib+SP: P =0.0016, t=7.60, d.f.=4; Erlotinib vs erlotinib+U: P =0.0012, t=8.24, d.f.=4. Similar procedures were performed in b-f and similar results were obtained. (b) Erlotinib vs erlotinib+R428: P =0.0008, t=9.24, d.f.=4; Erlotinib vs erlotinib+T: P =0.0004, t=11.18, d.f.=4. (c) Erlotinib vs erlotinib+SP: P =0.0004, t=10.78, d.f.=4; Erlotinib vs erlotinib+U: P =0.0014, t=7.85, d.f.=4. (d) Erlotinib vs erlotinib+R428: P =0.001, t=8.62, d.f.=4; Erlotinib vs erlotinib+T: P =0.0003, t=11.58, d.f.=4. (e) Erlotinib vs erlotinib+enbrel: P =0.0014, t=7.85, d.f.=4. (f) Erlotinib vs erlotinib+enbrel: P =0.0005, t=10.37, d.f.=4. SP: SP600125; U:U0126; T: Thalidomide. Data are presented as mean±s.e.m; **P<0.01, ***P<0.001 from two-tailed unpaired Student's t-test (n =3 biologically independent experimental replicates).

Supplementary Figure 9 TNF inhibition sensitizes mouse tumors to EGFR inhibition with afatinib in vivo.

(a) Combined treatment of afatinib and thalidomide prolonged survival and suppressed tumor growth in an orthotropic model. Kaplan-Maier survival curves were calculated using GraphPad Prism 7. Statistical significance verified by the log rank test, P=0.0015,**P<0.01. (b) Representative bioluminescence images from afatinib and afatinib plus thalidomide group at day 1, 10 and 20 post-treatment. Since all mice in vehicle and thalidomide group died within 20 days after transplant, images at day 20 post-treatment were not available. (c-d) Body weight of xenograft mice were monitored regularly and recorded every 2 days. No significant change of body weight was observed in subcutaneous mouse model between groups. (e) Body weight of orthotopic models. Body weight losses are found in vehicle, erlotinib and thalidomide group but not in combination therapy (erlotinib+thalidomide). Erlotinib vs erlotinib+thalidmoide: P<0.0001, t=6.32, d.f.=14; (f) Similar results were obtained in orthotopic models treated by afatinib and/or thalidmide. Afatinib vs afatinib+thalidomide: P =0.0003, t=4.79, d.f.=14.

Supplementary Figure 10 Erlotinib treatment activates Axl–JNK–ERK signaling in orthotopic models; this is suppressed by TNF inhibition.

(a) Immunostainig of pAXL, pJNK and pERK proteins from a representative brain section from vehicle, erlotinb and erlotinib plus thalidomide group. Cells with brown staining are considered as positive. Erlotinib group shows higher expression of pAxl, pJNK and pERK compared to vehicle group, whereas thaldimide inhibits erlotinib induced pAxl-pJNK-pERK activation. (b-d) Semi-quantitative analysis of pAxl, pJNK and pERK immunostaining. Four random fields in 3 tissue blocks at x200 magnification were scored semiquantitatively as: 0=No positive staining; 1=1-25% tumor cells stained, 2=26%-75% tumor cells stained and 3=>75% tumor cells stained. Differences between treatment groups were analyzed by the Mann-Whitney U test (n=12). (b) pAXL: Erlotinib vs. vehicle, p=0.0062, U=26; Erlotinib vs. erlotinib+thalidomide, p=0.0093, U=28. (c) pJNK: Erlotinib vs. vehicle, p=0.022, U=32; Erlotinib vs. erlotinib+thalidomide, p=0.014, U=29.5. (d) pERK: Erlotinib vs. vehicle, p=0.0040, U=23.5; Erlotinib vs. erlotinib+thalidomide, p=0.0026, U=21.

Supplementary Figure 11 Full-length western blots.

Full-length Western blots for cropped images in Fig. 1.

Supplementary Figure 12 Full-length western blots and DNA agarose gel.

Full-length Western blots for cropped images in Fig. 2 and Fig. 3f, 3h, 3i.

Supplementary Figure 13 Full-length western blots.

Full-length Western blots for cropped images in Fig. 4g, 4h, 4i.

Supplementary Figure 14 Full-length western blots.

Full-length Western blots for cropped images in Fig. 6j-l and Fig. 7f, 7g.

Supplementary Figure 15 Full-length western blots.

Full-length Western blots for cropped images in Supplementary Fig. 1 and 2d,e,g–i.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guo, G., Gong, K., Ali, S. et al. A TNF–JNK–Axl–ERK signaling axis mediates primary resistance to EGFR inhibition in glioblastoma. Nat Neurosci 20, 1074–1084 (2017). https://doi.org/10.1038/nn.4584

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4584

This article is cited by

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