Review
Resistance to EGF-R (erbB-1) and VEGF-R modulating agents

https://doi.org/10.1016/j.ejca.2008.11.038Get rights and content

Abstract

In an effort to improve the survival of cancer patients, new therapeutic approaches focusing on the molecular mechanisms that mediate tumour cell growth or survival have gained much attention. In particular, EGF-R and VEGF/VEGF-R have been extensively investigated as targets for anti-neoplastic therapy. Agents that selectively target EGF-R, erbB-2, VEGF-R-2 or VEGF have shown promising activity in clinical trials, and several are now approved for use in selected cancer indications. However, all patients ultimately develop resistance to these drugs. Thus, there is a great need to understand how patients become resistant to effective therapies for these cancers since this approach may lead to improvements in therapies that target EGF-R and VEGF/VEGF-R. Pre-clinical studies have begun to shed light on the mechanisms of resistance to anti-angiogenetic drugs and to date four mechanisms of resistance have been identified (1) upregulation of bFGF, (2) overexpression of MMP-9, (3) increased levels of SDF-1α and (4) HIF-1α-induced recruitment of bone marrow-derived CD45+ myeloid cells. In addition, the molecular mechanisms of resistance to EGF-R modulating agents can be attributed to several general processes: (1) activation of alternative tyrosine kinase inhibitors that bypass the EGF-R pathway (e.g. c-MET and IGF-1R), (2) increased angiogenesis, (3) constitutive activation of downstream mediators (e.g. PTEN and K-ras) and (4) the existence of specific EGF-R mutations. K-ras mutations have been significantly associated with a lack of response to EGF-R tyrosine kinase inhibitors in patients with NSCLC and with a lack of response to cetuximab or to panitumumab in patients with advanced colorectal cancer. The identification of these resistance mechanisms has led to clinical trials using newly designed targeted therapies that can overcome resistance and have shown promise in laboratory studies. Ongoing research efforts will likely continue to identify additional resistance mechanisms, and these findings will hopefully translate into effective therapies for different cancers.

Introduction

Despite advances in chemotherapy, most patients with cancer that has metastasised will succumb to the disease within 2 years of diagnosis. In an effort to improve survival, new therapeutic approaches focusing on the molecular mechanisms that mediate tumour cell growth or survival have gained much attention. In particular, the epidermal growth factor receptor (EGF-R) and the vascular endothelium growth factor receptor (VEGF-R) have been extensively investigated as targets for anti-neoplastic therapy.

Receptor tyrosine kinases (RTKs) such as EGF-R or VEGF-R are transmembrane proteins with an extracellular ligand binding domain and an intracellular tyrosine kinase catalytic domain. On binding to their cognate ligands, most RTKs dimerise and become activated through autophosphorylation of intracellular tyrosine residues. Activation of RTKs results in upregulation of multiple cellular signalling pathways that promote cell growth, survival and angiogenesis or environmental stimuli. Inappropriate activation of RTKs via mutation, overexpression or ectopic ligand production is a frequent feature of human tumour development and progression, and is thought to be a major mechanism by which cancer cells subvert normal growth controls.1, 2, 3 Consequently, in recent years modulation of RTK signal transduction has been an active area in oncology drug discovery. EGF-R (also called erbB1) and other erbB family RTKs (erbB-2/HER-2-neu, erbB-3/HER-3 and erbB-4/HER-4) encoded by the c-erbB proto-oncogenes have been strongly implicated in cancer development and progression as reviewed by 4, 2. Several mechanisms can cause aberrant receptor activation, resulting in tyrosine kinase activity, which is observed in cancer, including receptor overexpression, mutation, ligand-dependent receptor dimerisation and ligand-independent activation. For erbB-2, where a specific ligand has not been identified, activation occurs by homo- or hetero-dimerisation alone, whereas erbB-3 does not have significant kinase activity.4, 5 However, on activation, all 4 receptors are capable of signal transduction, causing activation of the ras/MAP kinase pathway, the PI3K/Akt pathway, src family kinases and STAT proteins. Activation of these pathways promotes cell proliferation, survival and angiogenesis.6

VEGF is the prototype of a large family of angiogenic and lymphangiogenic growth factors, which includes 6 structurally homologous, secreted glycoproteins called VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E and placenta growth factor.1 VEGF-A (commonly referred to as VEGF) was the first such molecule to be identified by the virtue of its ability to induce vascular permeability.7 The VEGF ligands trigger biological effects on their interaction with specific cell-surface receptors. The diversity of these receptors also adds to the biological complexity of angiogenesis and lymphangiogenesis. Two receptors were originally identified on vascular endothelial cells: VEGF-R-1 (a 180-kD transmembrane protein, also called Flt-1) and VEGF-R-2 (a 200-kD transmembrane protein, also called KDR). A third structurally related tyrosine kinase receptor is the 180-kD VEGF-R-3 (also called Flt-4), which is expressed broadly on endothelial cells during early embryogenesis.8 VEGF-R-2 is expressed in most, if not all, adult vascular endothelial cells as well as on circulating endothelial progenitor cells. Interestingly, both epithelial and mesenchymal tumour cells more typically express VEGF-R-1 than VEGF-R-29; however, in several experimental tumour models tumour cell-specific VEGF-R-2 expression has been shown to be the critical driver in the pathogenesis of tumours.1, 3 VEGF binding induces conformational changes within VEGF-R-2 followed by receptor dimerisation and autophosphorylation of tyrosine residues in the intracellular kinase domain. These tyrosine residues (Tyr951, Tyr996, Tyr1054 and Tyr1059) serve as high-affinity docking sites for a variety of signalling proteins, including phospholipase Cγ, ras-GAP, focal adhesion kinase, src family of tyrosine kinases, PI3K. Akt, PK-C, Raf-1 and MAPs. The interaction of 1 or more of these molecules with VEGF-R-2 may lead to alterations in cell proliferation, migration, differentiation, tube formation, and increase in vascular permeability and vascular integrity.3

Agents that selectively target EGF-R, erbB-2, VEGF-R-2 or VEGF have shown promising activity in clinical trials, and several are now approved for use in selected cancer indications (Table 1, Table 2). All patients, however, ultimately develop resistance to anti-EGF-R-and anti-VEGF(-R)-targeted therapies. Thus, there is a great need to understand how patients become resistant to effective therapies for these cancers.

Section snippets

Resistance to VEGF-R modulating drugs

Most current antiangiogenic strategies for cancer therapy are based on blocking VEGF functions, and anti-VEGF agents have successfully been used for the treatment of certain types of human cancers (Fig. 1). However, tumours also produce multiple non-VEGF angiogenic factors, and anti-VEGF monotherapy could potentially encounter drug resistance, suggesting that tumours could use non-VEGF angiogenic factors to grow blood vessels.

Intrinsic and acquired resistance to anti-angiogenetic drugs are

Resistance to EGF-R (erbB-1) modulating drugs

Anti-EGF-R-targeted therapies have improved the efficacy of conventional chemotherapy in both pre-clinical and clinical studies. Although such therapies may lead to partial response or disease stabilisation in some patients, many patients do not benefit from anti-EGF-R therapy, to those who do eventually develop resistance to that therapy. Great interest therefore, exists in elucidating resistance mechanisms for anti-EGF-R therapies as well as those for chemotherapy agents. The molecular

Effect of K-ras mutation on response to anti-EGF-R therapy

The clinical relevance of K-ras mutations has been evaluated retrospectively in several clinical trials investigating the effect of EGF-R inhibitors such as cetuximab or panitumumab in the first-line treatment of metastatic colorectal cancer. Van Cutsem and colleagues80 reported the CRYSTAL trial in which 5-fluorouracil, folinic acid and irinotecan (FOLFIRI) plus cetuximab were compared to FOLFIRI alone. An analysis of 45% of the study population (540 of 1198 patients) revealed a K-ras mutation

Conflict of interest statement

None declared.

References (98)

  • X. Hong et al.

    SDF-1 and CXCR4 are up-regulated by VEGF and contribute to glioma cell invasion

    Cancer Lett

    (2006)
  • M. Grunewald et al.

    VEGF-induced neovascularization: recruitment, retention, and role of accessory cells

    Cell

    (2006)
  • D. Zagzag et al.

    Hypoxia-inducible factor-1 and VEGF upregulate CXCR4 in glioblastoma: implications for angiogenesis and glioma cell invasion

    Lab Invest

    (2006)
  • M. Selvakumaran et al.

    Antitumor effect of the angiogenesis inhibitor bevacizumab is dependent on susceptibility of tumors to hypoxia-induced apoptosis

    Biochem Pharmacol

    (2008)
  • G. Valabrega et al.

    Trastuzumab: mechanism of action, resistance and future perspectives in HER2-overexpressing breast cancer

    Ann Oncol

    (2007)
  • S. Corso et al.

    Cancer therapy: can the challenge be MET?

    Trends Mol Med

    (2005)
  • T.J. Lynch et al.

    Summary statement novel agents in the treatment of lung cancer: fifth Cambridge conference assessing opportunities for combination therapy

    J Thorac Oncol

    (2008)
  • K. Takeda et al.

    Role of phosphatidylinositol 3-kinases in chemotaxis in Dictyostelium

    J Biol Chem

    (2007)
  • Y.D. Jung et al.

    Effects of combination anti-vascular endothelial growth factor receptor and anti-epidermal growth factor receptor therapies on the growth of gastric cancer in a nude mouse model

    Eur J Cancer

    (2002)
  • M.D. Marmor et al.

    Signal transduction and oncogenesis by ErbB/HER receptors

    Int J Radiat Oncol Biol Phys

    (2004)
  • P.A. Jänne

    Challenges of detecting EGFR T790M in gefitinib/erlotinib-resistant tumours

    Lung Cancer

    (2008)
  • G.J. Riely

    Second-generation epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer

    J Thorac Oncol

    (2008)
  • H. Youssoufian et al.

    Review: monoclonal antibodies to the vascular endothelial growth factor receptor-2 in cancer therapy

    Clin Cancer Res

    (2007)
  • F. Ciadiello et al.

    EGFR antagonists in cancer treatment

    N Engl J Med

    (2008)
  • R.S. Kerbel

    Tumor angiogenesis

    N Engl J Med

    (2008)
  • P.M. Harari et al.

    Biology of interactions: antiepidermal growth factor receptor agents

    J Clin Oncol

    (2007)
  • A. Kumar et al.

    Structure and clinical relevance of the epidermal growth factor receptor in human cancer

    J Clin Oncol

    (2008)
  • S. Grant et al.

    The role of signal transduction pathways in drug and radiation resistance

    Cancer Treat Res

    (2002)
  • D.R. Senger et al.

    Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid

    Science

    (1983)
  • D.J. Hicklin et al.

    Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis

    J Clin Oncol

    (2005)
  • E. Tassi et al.

    The angiogenic switch molecule, secreted FGF-binding protein, an indicator of early stages of pancreatic and colorectal adenocarcinoma

    Semin Oncol

    (2006)
  • V.D. Blanckaert et al.

    Basic fibroblast growth factor receptors and their prognostic value in human breast cancer

    Clin Cancer Res

    (1998)
  • J.L. Nissen et al.

    Angiogenic factors FGF2 and PDGF-BB synergistically promote murine tumor neovascularization and metastasis

    J Clin Invest

    (2007)
  • Dempke W Brivanib. A novel dual VEGF-R2/bFGF inhibitor. Anticancer Res 2009, in...
  • A.R. Nelson et al.

    Matrix metalloproteinases: biologic activity and clinical implications

    J Clin Oncol

    (2000)
  • L.A. Cornelius et al.

    Matrix metalloproteinases generate angiostatin: effects on neovascularization

    J Immunol

    (1998)
  • W.Y. Chung et al.

    Reduced expression of vascular endothelial growth factor paralleled with increased angiostatin expression resulting from the upregulated activities of matrix metalloproteinase-2 and -9 in human type 2 diabetic arterial vasculature

    Circ Res

    (2006)
  • M. Hollborn et al.

    Positive feedback regulation between MMP-9 and VEGF in human RPE cells

    Invest Ophthalmol Vis Sci

    (2007)
  • C.Z. Lee et al.

    Matrix metalloproteinase-9 inhibition attenuates vascular endothelial growth factor-induced intracerebral hemorrhage

    Stroke

    (2007)
  • C.R. Mackay

    Chemokines: immunology’s high impact factors

    Nat Immunol

    (2001)
  • R. Lima e Silva et al.

    The SDF-1/CXCR4 ligand/receptor pair is an important contributor to several types of ocular neovascularization

    FASEB J

    (2007)
  • I.S. Woo et al.

    Circulating stromal cell derived factor-1alpha (SDF-1alpha) is predictive of distant metastasis in gastric carcinoma

    Cancer Invest

    (2008)
  • K. Reddy et al.

    Stromal-cell-derived factor-1 stimulates vasculogenesis and enhances Ewing’s sarcoma tumor growth in the absence of vascular endothelial growth factor

    Int J Cancer

    (2008)
  • H.G. Kopp et al.

    Contribution of endothelial progenitors and proangiogenic hematopoietic cells to vascularization of tumor and ischemic tissue

    Curr Opin Hematol

    (2006)
  • B. Li et al.

    VEGF and PIGF promote adult vasculogenesis by enhancing EPC recruitment and vessel formation at the site of tumor neovascularization

    FASEB J

    (2006)
  • G.L. Semenza

    Regulation of mammalian O2 homeostasis by hypoxia-inducible factor-1

    Annu Rev Cell Dev Biol

    (1999)
  • G. Lucarini et al.

    Increased VEGF165 expression in HCT116 colon cancer cells after transient transfection with a GFP vector encoding HIF-1 gene

    J Exp Clin Cancer Res

    (2007)
  • D.T. Dang et al.

    Hypoxia-inducible factor-1 target genes as indicators of tumor vessel response to vascular endothelial growth factor inhibition

    Cancer Res

    (2008)
  • L.Q. Chow et al.

    Sunitinib: from rational design to clinical efficacy

    J Clin Oncol

    (2007)
  • Cited by (80)

    • Design, synthesis and biological evaluation of 2,3-dihydroimidazo[2,1-b]thiazoles as dual EGFR and IGF1R inhibitors

      2021, Bioorganic Chemistry
      Citation Excerpt :

      Just like EGFR, IGF1R is also overexpressed in a plethora of solid tumors and plays a role in pathogenesis, progression of multiple cancer types and anti-apoptotic signalling [5–8]. There is ample evidence of cross-talk between EGFR and IGF-1R cellular signalling pathways [9]. EGFR and IGF1R hetero-dimerization has also been reported in drug resistance [10].

    View all citing articles on Scopus
    View full text