Skip to main content

Utilizing combinations of molecular targeted agents to sensitize tumor cells to EGFR inhibitors

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

Abstract

EGFR inhibitors have achieved clinical antitumor activity as single agents. The specific inhibition of EGFR and associated pathways provides a mechanism for efficacious inhibition of tumor cell growth while minimizing toxicities often associated with chemotherapeutic drugs. The challenge of using targeted cancer drugs as single agents is the potential for de novo or acquired resistance due to established or adapted alternate signal transduction pathways. In this chapter, we will describe how cancer drug combinations with EGFR inhibitors can be rationally identified by utilizing our understanding of the molecular mechanism and related biomarkers of EGFR inhibitor sensitivity. Such combinations may ultimately provide better efficacy and reduced toxicity for patients.

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. 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 Res 2005;65:9455-62.

    Google Scholar 

  2. 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 Res 2006;66:944-50.

    Google Scholar 

  3. 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 

  4. 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. Mol Cancer Ther 2007;6:532-41.

    Google Scholar 

  5. 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. Mol Cancer Ther 2006;5:2051-9.

    Google Scholar 

  6. 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. Proc Natl Acad Sci U S A 2005;102:3788-93.

    Google Scholar 

  7. 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 Res 2001;61:7184-8.

    Google Scholar 

  8. De Craene B, Gilbert B, Stove C, Bruyneel E, van Roy F, Berx G. The transcription factor snail induces tumor cell invasion through modulation of the epithelial cell differentiation program. Cancer Res 2005;65:6237-44.

    Google Scholar 

  9. Andl CD, Rustgi AK. No one-way street: cross-talk between e-cadherin and receptor tyrosine kinase (RTK) signaling: a mechanism to regulate RTK activity. Cancer Biol Ther 2005;4:28-31.

    Google Scholar 

  10. Comoglio PM, Boccaccio C, Trusolino L. Interactions between growth factor receptors and adhesion molecules: breaking the rules. Curr Opin Cell Biol 2003;15:565-71.

    Google Scholar 

  11. Fedor-Chaiken M, Hein PW, Stewart JC, Brackenbury R, Kinch MS. E-cadherin binding modulates EGF receptor activation. Cell Commun Adhes 2003;10:105-18.

    Google Scholar 

  12. Pece S, Chiariello M, Murga C, Gutkind JS. Activation of the protein kinase Akt/PKB by the formation of E-cadherin-mediated cell-cell junctions. Evidence for the association of phosphatidylinositol 3-kinase with the E-cadherin adhesion complex. J Biol Chem 1999;274:19347-51.

    Google Scholar 

  13. Bachman KE, Argani P, Samuels Y, et al. The PIK3CA gene is mutated with high frequency in human breast cancers. Cancer Biol Ther 2004;3:772-5.

    Google Scholar 

  14. 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 

  15. Levine DA, Bogomolniy F, Yee CJ, et al. Frequent mutation of the PIK3CA gene in ovarian and breast cancers. Clin Cancer Res 2005;11:2875-8.

    Google Scholar 

  16. 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 

  17. Bianco R, Troiani T, Tortora G, Ciardiello F. Intrinsic and acquired resistance to EGFR inhibitors in human cancer therapy. Endocr Relat Cancer 2005;12 Suppl 1:S159-71.

    Google Scholar 

  18. Jones HE, Gee JM, Taylor KM, et al. Development of strategies for the use of anti-growth factor treatments. Endocr Relat Cancer 2005;12 Suppl 1:S173-82.

    Google Scholar 

  19. Miller KD. The role of ErbB inhibitors in trastuzumab resistance. Oncologist 2004;9 Suppl 3:16-9.

    Google Scholar 

  20. Kaiser U, Schardt C, Brandscheidt D, Wollmer E, Havemann K. Expression of insulin-like growth factor receptors I and II in normal human lung and in lung cancer. J Cancer Res Clin Oncol 1993;119:665-8.

    Google Scholar 

  21. LeRoith D, Roberts CT, Jr. The insulin-like growth factor system and cancer. Cancer Lett 2003;195:127-37.

    Google Scholar 

  22. Rubin R, Baserga R. Insulin-like growth factor-I receptor. Its role in cell proliferation, apoptosis, and tumorigenicity. Lab Invest 1995;73:311-31.

    Google Scholar 

  23. Adams TE, McKern NM, Ward CW. Signalling by the type 1 insulin-like growth factor receptor: interplay with the epidermal growth factor receptor. Growth Factors 2004;22:89-95.

    Google Scholar 

  24. Gooch JL, Van Den Berg CL, Yee D. Insulin-like growth factor (IGF)-I rescues breast cancer cells from chemotherapy-induced cell death--proliferative and anti-apoptotic effects. Breast Cancer Res Treat 1999;56:1-10.

    Google Scholar 

  25. Lu Y, Zi X, Zhao Y, Mascarenhas D, Pollak M. Insulin-like growth factor-I receptor signaling and resistance to trastuzumab (Herceptin). Journal of the National Cancer Institute 2001;93:1852-7.

    Google Scholar 

  26. Nahta R, Yuan LX, Zhang B, Kobayashi R, Esteva FJ. Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells. Cancer research 2005;65:11118-28.

    Google Scholar 

  27. Turner BC, Haffty BG, Narayanan L, et al. Insulin-like growth factor-I receptor overexpression mediates cellular radioresistance and local breast cancer recurrence after lumpectomy and radiation. Cancer research 1997;57:3079-83.

    Google Scholar 

  28. 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. Endocr Relat Cancer 2004;11:793-814.

    Google Scholar 

  29. Knowlden JM, Hutcheson IR, Barrow D, Gee JM, Nicholson RI. Insulin-like growth factor-I receptor signaling in tamoxifen-resistant breast cancer: a supporting role to the epidermal growth factor receptor. Endocrinology 2005;146:4609-18.

    Google Scholar 

  30. 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 

  31. 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 

  32. Goetsch L, Gonzalez A, Leger O, et al. A recombinant humanized anti-insulin-like growth factor receptor type I antibody (h7C10) enhances the antitumor activity of vinorelbine and anti-epidermal growth factor receptor therapy against human cancer xenografts. Int J Cancer 2005;113:316-28.

    Google Scholar 

  33. Irie HY, Pearline RV, Grueneberg D, et al. Distinct roles of Akt1 and Akt2 in regulating cell migration and epithelial-mesenchymal transition. The Journal of cell biology 2005;171:1023-34.

    Google Scholar 

  34. Morali OG, Delmas V, Moore R, Jeanney C, Thiery JP, Larue L. IGF-II induces rapid beta-catenin relocation to the nucleus during epithelium to mesenchyme transition. Oncogene 2001;20:4942-50.

    Google Scholar 

  35. Iwao K, Miyoshi Y, Ooka M, et al. Quantitative analysis of estrogen receptor-alpha and -beta messenger RNA expression in human pancreatic cancers by real-time polymerase chain reaction. Cancer Lett 2001;170:91-7.

    Google Scholar 

  36. Stabile LP, Davis AL, Gubish CT, et al. Human non-small cell lung tumors and cells derived from normal lung express both estrogen receptor alpha and beta and show biological responses to estrogen. Cancer research 2002;62:2141-50.

    Google Scholar 

  37. Stabile LP, Lyker JS, Gubish CT, Zhang W, Grandis JR, Siegfried JM. Combined targeting of the estrogen receptor and the epidermal growth factor receptor in non-small cell lung cancer shows enhanced antiproliferative effects. Cancer research 2005;65:1459-70.

    Google Scholar 

  38. Levin ER. Bidirectional signaling between the estrogen receptor and the epidermal growth factor receptor. Mol Endocrinol 2003;17:309-17.

    Google Scholar 

  39. Zhang Z, Maier B, Santen RJ, Song RX. Membrane association of estrogen receptor alpha mediates estrogen effect on MAPK activation. Biochemical and biophysical research communications 2002;294:926-33.

    Google Scholar 

  40. Dickson RB, Huff KK, Spencer EM, Lippman ME. Induction of epidermal growth factor-related polypeptides by 17 beta-estradiol in MCF-7 human breast cancer cells. Endocrinology 1986;118:138-42.

    Google Scholar 

  41. Nicholson RI, McClelland RA, Finlay P, et al. Relationship between EGF-R, c-erbB-2 protein expression and Ki67 immunostaining in breast cancer and hormone sensitivity. European journal of cancer (Oxford, England 1993;29A:1018-23.

    Google Scholar 

  42. Nicholson RI, McClelland RA, Gee JM, et al. Epidermal growth factor receptor expression in breast cancer: association with response to endocrine therapy. Breast Cancer Res Treat 1994;29:117-25.

    Google Scholar 

  43. van Agthoven T, van Agthoven TL, Portengen H, Foekens JA, Dorssers LC. Ectopic expression of epidermal growth factor receptors induces hormone independence in ZR-75-1 human breast cancer cells. Cancer research 1992;52:5082-8.

    Google Scholar 

  44. McClelland RA, Barrow D, Madden TA, et al. Enhanced epidermal growth factor receptor signaling in MCF7 breast cancer cells after long-term culture in the presence of the pure antiestrogen ICI 182,780 (Faslodex). Endocrinology 2001;142:2776-88.

    Google Scholar 

  45. Dutcher JP. Mammalian target of rapamycin inhibition. Clin Cancer Res 2004;10:6382S-7S.

    Google Scholar 

  46. Kim DH, Sarbassov DD, Ali SM, et al. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 2002;110:163-75.

    Google Scholar 

  47. Sarbassov DD, Guertin, D. A., Ali, S. M., and Sabatini, D. M. Phosphorylation and Regulation of Akt/PKB by the Rictor-mTOR Complex. Science 2005;307:1098-101.

    Google Scholar 

  48. Sarbassov DD, Ali SM, Kim DH, et al. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr Biol 2004;14:1296-302.

    Google Scholar 

  49. Sarbassov DD, Ali SM, Sabatini DM. Growing roles for the mTOR pathway. Curr Opin Cell Biol 2005.

    Google Scholar 

  50. Yonezawa K, Tokunaga C, Oshiro N, Yoshino K. Raptor, a binding partner of target of rapamycin. Biochem Biophys Res Commun 2004;313:437-41.

    Google Scholar 

  51. Harrington LS, Findlay GM, Gray A, et al. The TSC1-2 tumor suppressor controls insulin-PI3K signaling via regulation of IRS proteins. The Journal of cell biology 2004;166:213-23.

    Google Scholar 

  52. Tremblay F, Marette A. Amino acid and insulin signaling via the mTOR/p70 S6 kinase pathway. A negative feedback mechanism leading to insulin resistance in skeletal muscle cells. The Journal of biological chemistry 2001;276:38052-60.

    Google Scholar 

  53. Wan X, Harkavy B, Shen N, Grohar P, Helman LJ. Rapamycin induces feedback activation of Akt signaling through an IGF-1R-dependent mechanism. 2006.

    Google Scholar 

  54. Gual P, Le Marchand-Brustel Y, Tanti JF. Positive and negative regulation of insulin signaling through IRS-1 phosphorylation. Biochimie 2005;87:99-109.

    Google Scholar 

  55. Lee AV, Gooch JL, Oesterreich S, Guler RL, Yee D. Insulin-like growth factor I-induced degradation of insulin receptor substrate 1 is mediated by the 26S proteasome and blocked by phosphatidylinositol 3′-kinase inhibition. Molecular and cellular biology 2000;20:1489-96.

    Google Scholar 

  56. Liu YF, Herschkovitz A, Boura-Halfon S, et al. Serine phosphorylation proximal to its phosphotyrosine binding domain inhibits insulin receptor substrate 1 function and promotes insulin resistance. Molecular and cellular biology 2004;24:9668-81.

    Google Scholar 

  57. Paz K, Hemi R, LeRoith D, et al. A molecular basis for insulin resistance. Elevated serine/threonine phosphorylation of IRS-1 and IRS-2 inhibits their binding to the juxtamembrane region of the insulin receptor and impairs their ability to undergo insulin-induced tyrosine phosphorylation. The Journal of biological chemistry 1997;272:29911-8.

    Google Scholar 

  58. O'Reilly KE, Rojo F, She QB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res 2006;66:1500-8.

    Google Scholar 

  59. Atkins MB, Hidalgo M, Stadler WM, et al. Randomized phase II study of multiple dose levels of CCI-779, a novel mammalian target of rapamycin kinase inhibitor, in patients with advanced refractory renal cell carcinoma. J Clin Oncol 2004;22:909-18.

    Google Scholar 

  60. Gemmill RM, Zhou M, Costa L, Korch C, Bukowski RM, Drabkin HA. Synergistic growth inhibition by Iressa and Rapamycin is modulated by VHL mutations in renal cell carcinoma. Br J Cancer 2005;92:2266-77.

    Google Scholar 

  61. Goudar RK, Shi Q, Hjelmeland MD, et al. Combination therapy of inhibitors of epidermal growth factor receptor/vascular endothelial growth factor receptor 2 (AEE788) and the mammalian target of rapamycin (RAD001) offers improved glioblastoma tumor growth inhibition. Mol Cancer Ther 2005;4:101-12.

    Google Scholar 

  62. Buck E, Eyzaguirre A, Brown E, et al. Rapamycin synergizes with the epidermal growth factor receptor inhibitor erlotinib in non-small-cell lung, pancreatic, colon, and breast tumors. Mol Cancer Ther 2006;5:2676-84.

    Google Scholar 

  63. Huber MA, Kraut N, Beug H. Molecular requirements for epithelial-mesenchymal transition during tumor progression. Curr Opin Cell Biol 2005;17:548-58.

    Google Scholar 

  64. Kang Y, Massague J. Epithelial-mesenchymal transitions: twist in development and metastasis. Cell 2004;118:277-9.

    Google Scholar 

  65. Thiery JP. Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol 2003;15:740-6.

    Google Scholar 

  66. Vincent-Salomon A, Thiery JP. Host microenvironment in breast cancer development: epithelial-mesenchymal transition in breast cancer development. Breast Cancer Res 2003;5:101-6.

    Google Scholar 

  67. Saito A, Yamashita T, Mariko Y, et al. A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proceedings of the National Academy of Sciences of the United States of America 1999;96:4592-7.

    Google Scholar 

  68. Chinnaiyan P, Varambally S, Tomlins SA, et al. Enhancing the antitumor activity of ErbB blockade with histone deacetylase (HDAC) inhibition. International journal of cancer 2006;118:1041-50.

    Google Scholar 

  69. Nimmanapalli R, Fuino L, Bali P, et al. Histone deacetylase inhibitor LAQ824 both lowers expression and promotes proteasomal degradation of Bcr-Abl and induces apoptosis of imatinib mesylate-sensitive or -refractory chronic myelogenous leukemia-blast crisis cells. Cancer research 2003;63:5126-35.

    Google Scholar 

  70. Pao W, Miller VA, Politi KA, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med 2005;2:e73.

    Google Scholar 

  71. Marks PA, Richon VM, Rifkind RA. Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. Journal of the National Cancer Institute 2000;92:1210-6.

    Google Scholar 

  72. Yoshikawa M, Hishikawa K, Marumo T, Fujita T. Inhibition of histone deacetylase activity suppresses epithelial-to-mesenchymal transition induced by TGF-beta1 in human renal epithelial cells. J Am Soc Nephrol 2007;18:58-65.

    Google Scholar 

  73. Bolen JB. Nonreceptor tyrosine protein kinases. Oncogene 1993;8:2025-31.

    Google Scholar 

  74. Yeatman TJ. A renaissance for SRC. Nat Rev Cancer 2004;4:470-80.

    Google Scholar 

  75. Frame MC. Src in cancer: deregulation and consequences for cell behaviour. Biochim Biophys Acta 2002;1602:114-30.

    Google Scholar 

  76. Schlaepfer DD, Mitra SK, Ilic D. Control of motile and invasive cell phenotypes by focal adhesion kinase. Biochim Biophys Acta 2004;1692:77-102.

    Google Scholar 

  77. Fincham VJ, Frame MC. The catalytic activity of Src is dispensable for translocation to focal adhesions but controls the turnover of these structures during cell motility. The EMBO journal 1998;17:81-92.

    Google Scholar 

  78. Behrens J, Vakaet L, Friis R, et al. Loss of epithelial differentiation and gain of invasiveness correlates with tyrosine phosphorylation of the E-cadherin/beta-catenin complex in cells transformed with a temperature-sensitive v-SRC gene. The Journal of cell biology 1993;120:757-66.

    Google Scholar 

  79. Rodier JM, Valles AM, Denoyelle M, Thiery JP, Boyer B. pp60c-src is a positive regulator of growth factor-induced cell scattering in a rat bladder carcinoma cell line. The Journal of cell biology 1995;131:761-73.

    Google Scholar 

  80. Avizienyte E, Wyke AW, Jones RJ, et al. Src-induced de-regulation of E-cadherin in colon cancer cells requires integrin signalling. Nature cell biology 2002;4:632-8.

    Google Scholar 

  81. Boyer B, Bourgeois Y, Poupon MF. Src kinase contributes to the metastatic spread of carcinoma cells. Oncogene 2002;21:2347-56.

    Google Scholar 

  82. Nam JS, Ino Y, Sakamoto M, Hirohashi S. Src family kinase inhibitor PP2 restores the E-cadherin/catenin cell adhesion system in human cancer cells and reduces cancer metastasis. Clinical cancer research 2002;8:2430-6.

    Google Scholar 

  83. Calcagno AM, Fostel JM, Orchekowski RP, et al. Modulation of cell adhesion molecules in various epithelial cell lines after treatment with PP2. Mol Pharm 2005;2:170-84.

    Google Scholar 

  84. Dannenberg AJ, Lippman SM, Mann JR, Subbaramaiah K, DuBois RN. Cyclooxygenase-2 and epidermal growth factor receptor: pharmacologic targets for chemoprevention. Journal of clinical oncology 2005;23:254-66.

    Google Scholar 

  85. Dannenberg AJ, Subbaramaiah K. Targeting cyclooxygenase-2 in human neoplasia: rationale and promise. Cancer cell 2003;4:431-6.

    Google Scholar 

  86. Dubinett SM, Sharma S, Huang M, Dohadwala M, Pold M, Mao JT. Cyclooxygenase-2 in lung cancer. Prog Exp Tumor Res 2003;37:138-62.

    Google Scholar 

  87. Dohadwala M, Yang SC, Luo J, et al. Cyclooxygenase-2-dependent regulation of E-cadherin: prostaglandin E(2) induces transcriptional repressors ZEB1 and snail in non-small cell lung cancer. Cancer Res 2006;66:5338-45.

    Google Scholar 

  88. Chen Z, Zhang X, Li M, et al. Simultaneously targeting epidermal growth factor receptor tyrosine kinase and cyclooxygenase-2, an efficient approach to inhibition of squamous cell carcinoma of the head and neck. Clinical cancer research 2004;10:5930-9.

    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

Buck, E., Eyzaguirre, A., Iwata, K.K. (2008). Utilizing combinations of molecular targeted agents to sensitize tumor cells to EGFR 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_23

Download citation

  • DOI: https://doi.org/10.1007/978-1-59745-356-1_23

  • 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