Skip to main content

Cellular sensitivity to EGF receptor inhibitors

  • Chapter
  • First Online:
EGFR Signaling Networks in Cancer Therapy

Abstract

The EGFR pathway is a critical signaling pathway regulation cell proliferation and survival. As such, it is frequently deregulated in cancer through over expression of both EGF family ligands and receptors and by mutation of critical components within the pathway. These characteristics have made this signaling axis an attractive target for the development of molecularly targeted therapies in the treatment of cancer. To date there are numerous small molecule inhibitors and antibodies, either already in clinical use or in late stage clinical trials, that specifically target EGFR. These inhibitors have achieved great success in treating cancer patients and have generated a large amount of interest in identifying molecular markers that predict clinical benefit and mechanisms of resistance to such treatments. The first major breakthrough in this line of research was the identification of mutations in the EGFR kinase domain, which rendered the receptor hypersensitive to the actions of small molecule kinase inhibitors. However, the mutation rate was insufficient to explain the overall clinical benefit observed with these inhibitors, suggesting patients with wild-type EGFR also received some benefit. Subsequently, numerous efforts have been made to identify biomarkers of response and resistance other than EGFR mutational status.

Here we will summarize the current literature describing attempts to identify such markers, with particular emphasis on markers of sensitivity and resistance to small molecule EGFR tyrosine kinase inhibitors (TKIs). These approaches have encompassed the analysis of expression levels, both at the protein and genomic level, of EGFR and the closely related family members HER2 and HER3 and the analysis of the mutational status of downstream components of the EGFR pathway. In addition, we will highlight the role of the epithelial to mesenchymal transition (EMT) in sensitivity to small molecule EGFR TKIs and finally the potential role of alternative signaling cascades as a mode of cellular resistance to EGFR inhibition.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 249.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nature reviews 2001;2:127-37.

    Google Scholar 

  2. Siegel-Lakhai WS, Beijnen JH, Schellens JH. Current knowledge and future directions of the selective epidermal growth factor receptor inhibitors erlotinib (Tarceva) and gefitinib (Iressa). The oncologist 2005;10:579-89.

    Google Scholar 

  3. Konecny GE, Pegram MD, Venkatesan N, et al. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer research 2006;66:1630-9.

    Google Scholar 

  4. Shepherd FA, Rodrigues Pereira J, Ciuleanu T, et al. Erlotinib in previously treated non-small-cell lung cancer. The New England journal of medicine 2005;353:123-32.

    Google Scholar 

  5. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. The New England journal of medicine 2004;350:2129-39.

    Google Scholar 

  6. Pao W, Miller V, Zakowski M, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proceedings of the National Academy of Sciences of the United States of America 2004;101:13306-11.

    Google Scholar 

  7. Sordella R, Bell DW, Haber DA, Settleman J. Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways. Science 2004;305:1163-7.

    Google Scholar 

  8. Carey KD, Garton AJ, Romero MS, et al. Kinetic analysis of epidermal growth factor receptor somatic mutant proteins shows increased sensitivity to the epidermal growth factor receptor tyrosine kinase inhibitor, erlotinib. Cancer research 2006;66:8163-71.

    Google Scholar 

  9. Ritchie E, Nichols G. Mechanisms of resistance to imatinib in CML patients: a paradigm for the advantages and pitfalls of molecularly targeted therapy. Current cancer drug targets 2006;6:645-57.

    Google Scholar 

  10. Han SW, Kim TY, Hwang PG, et al. Predictive and prognostic impact of epidermal growth factor receptor mutation in non-small-cell lung cancer patients treated with gefitinib. J Clin Oncol 2005;23:2493-501.

    Google Scholar 

  11. Tokumo M, Toyooka S, Kiura K, et al. The relationship between epidermal growth factor receptor mutations and clinicopathologic features in non-small cell lung cancers. Clin Cancer Res 2005;11:1167-73.

    Google Scholar 

  12. Tsao MS, Sakurada A, Cutz JC, et al. Erlotinib in lung cancer - molecular and clinical predictors of outcome. The New England journal of medicine 2005;353:133-44.

    Google Scholar 

  13. Fuster LM, Sandler AB. Select clinical trials of erlotinib (OSI-774) in non-small-cell lung cancer with emphasis on phase III outcomes. Clinical lung cancer 2004;6 Suppl 1:S24-9.

    Google Scholar 

  14. Eberhard DA, Johnson BE, Amler LC, et al. Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol 2005;23:5900-9.

    Google Scholar 

  15. Herbst RS, Prager D, Hermann R, et al. TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol 2005;23:5892-9.

    Google Scholar 

  16. Fornier M, Risio M, Van Poznak C, Seidman A. HER2 testing and correlation with efficacy of trastuzumab therapy. Oncology (Williston Park, NY 2002;16:1340-8, 51-2; discussion 52, 55-8.

    Google Scholar 

  17. Hirsch FR, Varella-Garcia M, Bunn PA, Jr., et al. Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol 2003;21:3798-807.

    Google Scholar 

  18. Ono M, Hirata A, Kometani T, et al. Sensitivity to gefitinib (Iressa, ZD1839) in non-small cell lung cancer cell lines correlates with dependence on the epidermal growth factor (EGF) receptor/extracellular signal-regulated kinase 1/2 and EGF receptor/Akt pathway for proliferation. Molecular cancer therapeutics 2004;3:465-72.

    Google Scholar 

  19. Thomson S, Buck E, Petti F, et al. Epithelial to mesenchymal transition is a determinant of sensitivity of non-small-cell lung carcinoma cell lines and xenografts to epidermal growth factor receptor inhibition. Cancer research 2005;65:9455-62.

    Google Scholar 

  20. Yauch RL, Januario T, Eberhard DA, et al. Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin Cancer Res 2005;11:8686-98.

    Google Scholar 

  21. Meert AP, Martin B, Delmotte P, et al. The role of EGF-R expression on patient survival in lung cancer: a systematic review with meta-analysis. Eur Respir J 2002;20:975-81.

    Google Scholar 

  22. Bunn PA, Jr., Dziadziuszko R, Varella-Garcia M, et al. Biological markers for non-small cell lung cancer patient selection for epidermal growth factor receptor tyrosine kinase inhibitor therapy. Clin Cancer Res 2006;12:3652-6.

    Google Scholar 

  23. Toschi L, Cappuzzo F. Understanding the new genetics of responsiveness to epidermal growth factor receptor tyrosine kinase inhibitors. The oncologist 2007;12:211-20.

    Google Scholar 

  24. Clark GM, Zborowski DM, Santabarbara P, et al. Smoking history and epidermal growth factor receptor expression as predictors of survival benefit from erlotinib for patients with non-small-cell lung cancer in the National Cancer Institute of Canada Clinical Trials Group study BR.21. Clinical lung cancer 2006;7:389-94.

    Google Scholar 

  25. Perez-Soler R, Chachoua A, Hammond LA, et al. Determinants of tumor response and survival with erlotinib in patients with non--small-cell lung cancer. J Clin Oncol 2004;22:3238-47.

    Google Scholar 

  26. Chung KY, Shia J, Kemeny NE, et al. Cetuximab shows activity in colorectal cancer patients with tumors that do not express the epidermal growth factor receptor by immunohistochemistry. J Clin Oncol 2005;23:1803-10.

    Google Scholar 

  27. Cappuzzo F, Hirsch FR, Rossi E, et al. Epidermal growth factor receptor gene and protein and gefitinib sensitivity in non-small-cell lung cancer. Journal of the National Cancer Institute 2005;97:643-55.

    Google Scholar 

  28. Hirsch FR, Varella-Garcia M, Bunn PA, Jr., et al. Molecular predictors of outcome with gefitinib in a phase III placebo-controlled study in advanced non-small-cell lung cancer. J Clin Oncol 2006;24:5034-42.

    Google Scholar 

  29. Hirsch FR, Witta S. Biomarkers for prediction of sensitivity to EGFR inhibitors in non-small cell lung cancer. Current opinion in oncology 2005;17:118-22.

    Google Scholar 

  30. Comis RL. The current situation: erlotinib (Tarceva) and gefitinib (Iressa) in non-small cell lung cancer. The oncologist 2005;10:467-70.

    Google Scholar 

  31. Takano T, Ohe Y, Sakamoto H, et al. Epidermal growth factor receptor gene mutations and increased copy numbers predict gefitinib sensitivity in patients with recurrent non-small-cell lung cancer. J Clin Oncol 2005;23:6829-37.

    Google Scholar 

  32. Johnson BE, Janne PA. Selecting patients for epidermal growth factor receptor inhibitor treatment: A FISH story or a tale of mutations? J Clin Oncol 2005;23:6813-6.

    Google Scholar 

  33. Shigematsu H, Takahashi T, Nomura M, et al. Somatic mutations of the HER2 kinase domain in lung adenocarcinomas. Cancer research 2005;65:1642-6.

    Google Scholar 

  34. Wang SE, Narasanna A, Perez-Torres M, et al. HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors. Cancer cell 2006;10:25-38.

    Google Scholar 

  35. Han SW, Kim TY, Jeon YK, et al. Optimization of patient selection for gefitinib in non-small cell lung cancer by combined analysis of epidermal growth factor receptor mutation, K-ras mutation, and Akt phosphorylation. Clin Cancer Res 2006;12:2538-44.

    Google Scholar 

  36. Moasser MM, Basso A, Averbuch SD, Rosen N. The tyrosine kinase inhibitor ZD1839 (“Iressa”) inhibits HER2-driven signaling and suppresses the growth of HER2-overexpressing tumor cells. Cancer research 2001;61:7184-8.

    Google Scholar 

  37. Moulder SL, Yakes FM, Muthuswamy SK, Bianco R, Simpson JF, Arteaga CL. Epidermal growth factor receptor (HER1) tyrosine kinase inhibitor ZD1839 (Iressa) inhibits HER2/neu (erbB2)-overexpressing breast cancer cells in vitro and in vivo. Cancer research 2001;61:8887-95.

    Google Scholar 

  38. Hendriks BS, Opresko LK, Wiley HS, Lauffenburger D. Coregulation of epidermal growth factor receptor/human epidermal growth factor receptor 2 (HER2) levels and locations: quantitative analysis of HER2 overexpression effects. Cancer research 2003;63:1130-7.

    Google Scholar 

  39. Graus-Porta D, Beerli RR, Daly JM, Hynes NE. ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. The EMBO journal 1997;16:1647-55.

    Google Scholar 

  40. Hendriks BS, Opresko LK, Wiley HS, Lauffenburger D. Quantitative analysis of HER2-mediated effects on HER2 and epidermal growth factor receptor endocytosis: distribution of homo- and heterodimers depends on relative HER2 levels. The Journal of biological chemistry 2003;278:23343-51.

    Google Scholar 

  41. Anido J, Matar P, Albanell J, et al. ZD1839, a specific epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, induces the formation of inactive EGFR/HER2 and EGFR/HER3 heterodimers and prevents heregulin signaling in HER2-overexpressing breast cancer cells. Clin Cancer Res 2003;9:1274-83.

    Google Scholar 

  42. Kim HH, Sierke SL, Koland JG. Epidermal growth factor-dependent association of phosphatidylinositol 3-kinase with the erbB3 gene product. The Journal of biological chemistry 1994;269:24747-55.

    Google Scholar 

  43. Soltoff SP, Carraway KL, 3rd, Prigent SA, Gullick WG, Cantley LC. ErbB3 is involved in activation of phosphatidylinositol 3-kinase by epidermal growth factor. Molecular and cellular biology 1994;14:3550-8.

    Google Scholar 

  44. Engelman JA, Janne PA, Mermel C, et al. ErbB-3 mediates phosphoinositide 3-kinase activity in gefitinib-sensitive non-small cell lung cancer cell lines. Proceedings of the National Academy of Sciences of the United States of America 2005;102:3788-93.

    Google Scholar 

  45. Buck E, Eyzaguirre A, Haley JD, Gibson NW, Cagnoni P, Iwata KK. Inactivation of Akt by the epidermal growth factor receptor inhibitor erlotinib is mediated by HER-3 in pancreatic and colorectal tumor cell lines and contributes to erlotinib sensitivity. Molecular cancer therapeutics 2006;5:2051-9.

    Google Scholar 

  46. Sergina NV, Rausch M, Wang D, et al. Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature 2007;445:437-41.

    Google Scholar 

  47. Fujimoto N, Wislez M, Zhang J, et al. High expression of ErbB family members and their ligands in lung adenocarcinomas that are sensitive to inhibition of epidermal growth factor receptor. Cancer research 2005;65:11478-85.

    Google Scholar 

  48. Suzuki M, Shigematsu H, Hiroshima K, et al. Epidermal growth factor receptor expression status in lung cancer correlates with its mutation. Human pathology 2005;36:1127-34.

    Google Scholar 

  49. Lievre A, Bachet JB, Le Corre D, et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer research 2006;66:3992-5.

    Google Scholar 

  50. Pao W, Wang TY, Riely GJ, et al. KRAS mutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib. PLoS medicine 2005;2:e17.

    Google Scholar 

  51. Cully M, You H, Levine AJ, Mak TW. Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat Rev Cancer 2006;6:184-92.

    Google Scholar 

  52. Kokubo Y, Gemma A, Noro R, et al. Reduction of PTEN protein and loss of epidermal growth factor receptor gene mutation in lung cancer with natural resistance to gefitinib (IRESSA). British journal of cancer 2005;92:1711-9.

    Google Scholar 

  53. Mellinghoff IK, Wang MY, Vivanco I, et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. The New England journal of medicine 2005;353:2012-24.

    Google Scholar 

  54. Bianco R, Shin I, Ritter CA, et al. Loss of PTEN/MMAC1/TEP in EGF receptor-expressing tumor cells counteracts the antitumor action of EGFR tyrosine kinase inhibitors. Oncogene 2003;22:2812-22.

    Google Scholar 

  55. She QB, Solit D, Basso A, Moasser MM. Resistance to gefitinib in PTEN-null HER-overexpressing tumor cells can be overcome through restoration of PTEN function or pharmacologic modulation of constitutive phosphatidylinositol 3′-kinase/Akt pathway signaling. Clin Cancer Res 2003;9:4340-6.

    Google Scholar 

  56. Haas-Kogan DA, Prados MD, Tihan T, et al. Epidermal growth factor receptor, protein kinase B/Akt, and glioma response to erlotinib. Journal of the National Cancer Institute 2005;97:880-7.

    Google Scholar 

  57. Coldren CD, Helfrich BA, Witta SE, et al. Baseline gene expression predicts sensitivity to gefitinib in non-small cell lung cancer cell lines. Mol Cancer Res 2006;4:521-8.

    Google Scholar 

  58. Witta SE, Gemmill RM, Hirsch FR, et al. Restoring E-cadherin expression increases sensitivity to epidermal growth factor receptor inhibitors in lung cancer cell lines. Cancer research 2006;66:944-50.

    Google Scholar 

  59. Buck E, Eyzaguirre A, Barr S, et al. Loss of homotypic cell adhesion by epithelial-mesenchymal transition or mutation limits sensitivity to epidermal growth factor receptor inhibition. Molecular cancer therapeutics 2007;6:532-41.

    Google Scholar 

  60. Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2002;2:442-54.

    Google Scholar 

  61. Brabletz T, Jung A, Spaderna S, Hlubek F, Kirchner T. Opinion: migrating cancer stem cells - an integrated concept of malignant tumour progression. Nat Rev Cancer 2005;5:744-9.

    Google Scholar 

  62. Halbleib JM, Nelson WJ. Cadherins in development: cell adhesion, sorting, and tissue morphogenesis. Genes & development 2006;20:3199-214.

    Google Scholar 

  63. Christofori G, Semb H. The role of the cell-adhesion molecule E-cadherin as a tumour-suppressor gene. Trends in biochemical sciences 1999;24:73-6.

    Google Scholar 

  64. Perl AK, Wilgenbus P, Dahl U, Semb H, Christofori G. A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature 1998;392:190-3.

    Google Scholar 

  65. Peinado H, Ballestar E, Esteller M, Cano A. Snail mediates E-cadherin repression by the recruitment of the Sin3A/histone deacetylase 1 (HDAC1)/HDAC2 complex. Molecular and cellular biology 2004;24:306-19.

    Google Scholar 

  66. Moreno-Bueno G, Cubillo E, Sarrio D, et al. Genetic profiling of epithelial cells expressing e-cadherin repressors reveals a distinct role for snail, slug, and e47 factors in epithelial-mesenchymal transition. Cancer research 2006;66:9543-56.

    Google Scholar 

  67. Olmeda D, Jorda M, Peinado H, Fabra A, Cano A. Snail silencing effectively suppresses tumour growth and invasiveness. Oncogene 2006.

    Google Scholar 

  68. Qian X, Karpova T, Sheppard AM, McNally J, Lowy DR. E-cadherin-mediated adhesion inhibits ligand-dependent activation of diverse receptor tyrosine kinases. The EMBO journal 2004;23:1739-48.

    Google Scholar 

  69. Barrallo-Gimeno A, Nieto MA. The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development (Cambridge, England) 2005;132:3151-61.

    Google Scholar 

  70. Maestro R, Dei Tos AP, Hamamori Y, et al. Twist is a potential oncogene that inhibits apoptosis. Genes & development 1999;13:2207-17.

    Google Scholar 

  71. Vega F, Medeiros LJ, Leventaki V, et al. Activation of mammalian target of rapamycin signaling pathway contributes to tumor cell survival in anaplastic lymphoma kinase-positive anaplastic large cell lymphoma. Cancer research 2006;66:6589-97.

    Google Scholar 

  72. Moody SE, Perez D, Pan TC, et al. The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer cell 2005;8:197-209.

    Google Scholar 

  73. Samani AA, Yakar S, LeRoith D, Brodt P. The role of the IGF system in cancer growth and metastasis: overview and recent insights. Endocrine reviews 2007;28:20-47.

    Google Scholar 

  74. Moyer JD, Barbacci EG, Iwata KK, et al. Induction of apoptosis and cell cycle arrest by CP-358,774, an inhibitor of epidermal growth factor receptor tyrosine kinase. Cancer research 1997;57:4838-48.

    Google Scholar 

  75. Chakravarti A, Loeffler JS, Dyson NJ. Insulin-like growth factor receptor I mediates resistance to anti-epidermal growth factor receptor therapy in primary human glioblastoma cells through continued activation of phosphoinositide 3-kinase signaling. Cancer research 2002;62:200-7.

    Google Scholar 

  76. Morgillo F, Woo JK, Kim ES, Hong WK, Lee HY. Heterodimerization of insulin-like growth factor receptor/epidermal growth factor receptor and induction of survivin expression counteract the antitumor action of erlotinib. Cancer research 2006;66:10100-11.

    Google Scholar 

  77. Jones HE, Goddard L, Gee JM, et al. Insulin-like growth factor-I receptor signalling and acquired resistance to gefitinib (ZD1839; Iressa) in human breast and prostate cancer cells. Endocrine-related cancer 2004;11:793-814.

    Google Scholar 

  78. Desbois-Mouthon C, Cacheux W, Blivet-Van Eggelpoel MJ, et al. Impact of IGF-1R/EGFR cross-talks on hepatoma cell sensitivity to gefitinib. International journal of cancer 2006;119:2557-66.

    Google Scholar 

  79. Pietras K, Sjoblom T, Rubin K, Heldin CH, Ostman A. PDGF receptors as cancer drug targets. Cancer cell 2003;3:439-43.

    Google Scholar 

  80. Yu Y, Sweeney M, Zhang S, et al. PDGF stimulates pulmonary vascular smooth muscle cell proliferation by upregulating TRPC6 expression. American journal of physiology 2003;284:C316-30.

    Google Scholar 

  81. Kassouf W, Dinney CP, Brown G, et al. Uncoupling between epidermal growth factor receptor and downstream signals defines resistance to the antiproliferative effect of Gefitinib in bladder cancer cells. Cancer research 2005;65:10524-35.

    Google Scholar 

  82. Matei D, Emerson RE, Lai YC, et al. Autocrine activation of PDGFRalpha promotes the progression of ovarian cancer. Oncogene 2006;25:2060-9.

    Google Scholar 

  83. Jechlinger M, Sommer A, Moriggl R, et al. Autocrine PDGFR signaling promotes mammary cancer metastasis. The Journal of clinical investigation 2006;116:1561-70.

    Google Scholar 

  84. Carvalho I, Milanezi F, Martins A, Reis RM, Schmitt F. Overexpression of platelet-derived growth factor receptor alpha in breast cancer is associated with tumour progression. Breast Cancer Res 2005;7:R788-95.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2008 Humana Press, a part of Springer Science+Business Media, LLC

About this chapter

Cite this chapter

Thomson, S., Haley, J.D., Yauch, R. (2008). Cellular sensitivity to EGF receptor inhibitors. In: Haley, J., Gullick, W. (eds) EGFR Signaling Networks in Cancer Therapy. Cancer Drug Discovery and Development. Humana Press. https://doi.org/10.1007/978-1-59745-356-1_22

Download citation

  • DOI: https://doi.org/10.1007/978-1-59745-356-1_22

  • Published:

  • Publisher Name: Humana Press

  • Print ISBN: 978-1-58829-948-2

  • Online ISBN: 978-1-59745-356-1

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics