Elsevier

Cellular Signalling

Volume 19, Issue 10, October 2007, Pages 2013-2023
Cellular Signalling

Review
Mechanisms for oncogenic activation of the epidermal growth factor receptor

https://doi.org/10.1016/j.cellsig.2007.06.023Get rights and content

Abstract

The Epidermal growth factor receptor (EGFR) is a membrane spanning glycoprotein, which frequently has been implicated in various cancer types. The mechanisms by which EGFR becomes oncogenic are numerous and are often specific for each cancer type. In some tumors, EGFR is activated by autocrine/paracrine growth factor loops, whereas in others activating mutations promote EGFR signaling. Overexpression and/or amplification of the EGFR gene are prevalent in many cancer types leading to aberrant EGFR signaling. In addition, failure to attenuate receptor signaling by receptor downregulation can also lead to cellular transformation. Heterodimerization of EGFR with ErbB2 inhibits downregulation of EGFR and thereby prolongs growth factor signaling. This also indicates that cross-talk between EGFR and heterologous receptor systems serves as another mechanism for oncogenic activation of EGFR.

Because of its role in tumor promotion, the EGFR has been intensely studied as a therapeutic target. There are currently two major mechanisms by which the EGFR is targeted: antibodies binding to the extracellular domain of EGFR and small-molecule tyrosine-kinase inhibitors. However, tumorigenesis is a multi-step process involving several mutations, which might explain why EGFR therapeutics has only been partially successful. This highlights the importance of pinpointing the mechanisms by which EGFR becomes oncogenic in a particular cancer. In this review, each of the above mentioned mechanisms will be discussed, as a detailed molecular and genetic understanding of how EGFR contributes to the malignant phenotype might offer new promise for the design, development and clinical evaluation of future tumor-specific anticancer approaches.

Introduction

Carcinogenesis is a multi-step process that requires the accumulation of several genetic alterations in a single cell. Among numerous factors, carcinogenesis involves the activation of oncogenes such as the epidermal growth factor receptor (EGFR), also known as ErbB1/HER1. EGFR is a receptor tyrosine kinase (RTK) that is catalytically active and under tight regulatory control. This receptor belongs to the ErbB/HER family of ligand-activated RTKs, which in addition include ErbB2/Neu/HER2, ErbB3/HER3 and ErbB4/HER4 [1]. EGFR and its family members play essential roles in regulating a number of cellular processes including cell proliferation, survival and migration [2]. Dysregulation of their activity is therefore strongly associated with tumorigenesis, which classifies them as some of the most frequent implicated cell-surface markers of human cancer [3].

The involvement of increased and/or aberrant EGFR activity in human cancers is well documented [4], [5], [6] and cancer patients with altered EGFR activity tend to have a more aggressive disease, associated with a poor clinical outcome [1]. Therefore, EGFR is a rational target for anti-tumor strategies. There is a range of mechanisms leading to dysregulated EGFR activity and it is important to know which one is prevalent in a particular tumor in order to give the patients the most appropriate treatment.

Since the vast majority of reviews largely focus on anti-EGFR agents and their clinical outcome, this review will mainly focus on the mechanisms by which EGFR becomes oncogenic and briefly summarize some of the clinical approaches currently used to target tumors with aberrant EGFR activation. A detailed molecular and genetic understanding of how EGFR contributes to the malignant phenotype is important for future design and development of tumor-specific anticancer approaches.

Section snippets

EGFR: structure and regulation of activity

The EGFR is a highly glycosylated 170 kDa membrane spanning protein, which consists of a single polypeptide chain of 1186 amino acids [7], [8], [9]. Like all RTKs, the EGFR is characterized by three main domains: an extracellular ligand-binding domain, a transmembrane domain and a cytoplasmic domain containing the tyrosine kinase (Fig. 1A) [1], [10]. The extracellular domain of the EGFR can be further divided into four subdomains designated I, II, III and IV (Fig. 1A). Crystallographic studies

EGFR and cancer: mechanisms leading to the constitutive activation of EGFR

As EGFR is involved in the regulation of cell proliferation, motility and survival an imbalance in the EGFR-ligand system giving rise to increased EGFR signaling can lead to neoplastic transformation. Indeed, dysregulated EGFR has frequently been found in various forms of human cancers [19], [20].

There are quite a few mechanisms by which the tight regulation of the EGFR-ligand system can be abrogated (Fig. 2). These include: 1) increased production of ligands, 2) increased levels of EGFR

Conclusion

EGFR has been intensively studied as a therapeutic target. New agents are entering the clinic and new combinatorial approaches with anti-EGFR agents are being explored. Ongoing clinical trials continue to investigate EGFR therapeutics in various treatment settings and major efforts are being made to identify molecular markers that can select patients more likely to respond to anti-EGFR therapy. However, as EGFR therapeutics have so far only been partially successful, it is important to have a

References (148)

  • T. Holbro et al.

    Exp. Cell Res.

    (2003)
  • H. Ogiso et al.

    Cell

    (2002)
  • S. Bishayee

    Biochem. Pharmacol.

    (2000)
  • J. Schlessinger

    Cell

    (2002)
  • T.P. Garrett et al.

    Cell

    (2002)
  • J. Schlessinger

    Cell

    (2000)
  • S. Bogdan et al.

    Curr. Biol.

    (2001)
  • S.R. Datta et al.

    Cell

    (1997)
  • K. Shtiegman et al.

    Semin. Cancer Biol.

    (2003)
  • D.W. Cohen et al.

    J. Urol.

    (1994)
  • E.P. Sandgren et al.

    Cell

    (1990)
  • A. Rosenthal et al.

    Cell

    (1986)
  • S. Li et al.

    Cancer Cell

    (2005)
  • R. Bhargava et al.

    Mod. Path.

    (2005)
  • V.J. Bykov et al.

    Eur. J. Cancer

    (2003)
  • F. Gebhardt et al.

    J. Biol. Chem.

    (1999)
  • M. Moroni et al.

    Lancet Oncol.

    (2005)
  • P.A. Humphrey et al.

    Biochem. Biophys. Res. Commun.

    (1991)
  • M.A. Antonyak et al.

    J. Biol. Chem.

    (1998)
  • M.W. Pedersen et al.

    Ann.Oncol.

    (2001)
  • T.G. Johns et al.

    J. Biol. Chem.

    (2004)
  • C. Metayer et al.

    Lung Cancer

    (2002)
  • G. Levkowitz et al.

    Mol. Cell

    (1999)
  • Y. Mosesson et al.

    J. Biol. Chem.

    (2003)
  • P. Peschard et al.

    Cancer Cell

    (2003)
  • R. Worthylake et al.

    J. Biol. Chem.

    (1999)
  • A.R. French et al.

    J. Biol. Chem.

    (1995)
  • R. De Wit et al.

    Free Radic. Biol. Med.

    (2000)
  • T. Ravid et al.

    J. Biol. Chem.

    (2002)
  • N.E. Hynes et al.

    Endocr. Relat. Cancer.

    (2001)
  • C.T. Kuan et al.

    Endocr. Relat. Cancer

    (2001)
  • Y. Umekita et al.

    Int. J. Cancer

    (2000)
  • F.R. Hirsch et al.

    J. Clin. Oncol.

    (2003)
  • A.J. Wong et al.

    Proc. Natl. Acad. Sci. U.S.A.

    (1987)
  • T. Hunter

    Nature

    (1984)
  • G. Pages et al.

    Proc. Natl. Acad. Sci. U.S.A.

    (1993)
  • P. Chen et al.

    J. Cell Biol.

    (1994)
  • S. Lipkowitz

    Breast Cancer Res.

    (2003)
  • N. Prenzel et al.

    Endocr. Relat Cancer

    (2001)
  • C.L. Arteaga

    Oncologist

    (2002)
  • Y.C. Cai et al.

    Virchows Arch.

    (1999)
  • Y. Umekita et al.

    Int. J. Cancer

    (2000)
  • H. Riedel et al.

    Proc. Natl. Acad. Sci. U.S.A.

    (1988)
  • Y. Yamanaka et al.

    Anticancer Res.

    (1993)
  • Y. Yonemura et al.

    Oncology

    (1992)
  • T. Narita et al.

    Jpn. J. Cancer Res.

    (2001)
  • N.I. Goldstein et al.

    Clin. Cancer Res.

    (1995)
  • J. Baselga

    Eur. J. Cancer

    (2001)
  • M.C. Prewett et al.

    Clin. Cancer Res.

    (2002)
  • R.I. Nicholson et al.

    Eur. J. Cancer

    (2001)
  • Cited by (0)

    View full text