Elsevier

Clinical Colorectal Cancer

Volume 16, Issue 4, December 2017, Pages 247-251
Clinical Colorectal Cancer

Review
HER2 as an Emerging Oncotarget for Colorectal Cancer Treatment After Failure of Anti-Epidermal Growth Factor Receptor Therapy

https://doi.org/10.1016/j.clcc.2017.03.001Get rights and content

Abstract

Amplification of the HER2 gene is an indicator of poor prognosis for several kinds of malignancies such as breast and gastric cancer, and anti-HER2 targeting therapies provide clinical benefits in these patients. In 2011, HER2 was identified as a resistance molecule for de novo and secondary anti-epidermal growth factor receptor (EGFR) antibody therapy. HER2 activation provides a bypass signaling pathway after anti-EGFR antibody treatment of colorectal cancer. Cell line-based screening revealed that HER2 genomic amplification induces resistance to the anti-EGFR antibody cetuximab in colorectal cancer. Recently, HER2 itself has been recognized as a target for oncotherapy in colorectal cancer. The first part of this review provides an update on the present state of knowledge about the role of HER2 in colorectal cancer, including its prognostic relevance and role in resistance to anti-EGFR antibody treatment. In the second part of the review, we discuss the results of preclinical and clinical studies that examined the potential utility of anti-HER2 targeted therapy in colorectal cancer. Although it acts as a barrier for other molecular targeting agents such as cetuximab, HER2 itself is a promising target for oncotherapy. Current research indicates that anti-HER2 drugs will be developed further and introduced into clinical practice for the treatment of patients with HER2-positive colorectal cancer.

Introduction

Colorectal cancer (CRC) is the third most common cancer worldwide. More than 1.2 million new cases of CRC are diagnosed every year.1 Approximately 25% of these patients have metastatic lesions at diagnosis and almost 50% of these patients will eventually develop metastases.2 The standard therapy for advanced CRC is systemic chemotherapy. Until the 2000s, 5-fluorouracil was the only drug approved by the US Food and Drug Administration (FDA) for the treatment of metastatic CRC; however, median overall survival after treatment was <1.5 years.3 Subsequently, the development and the US FDA approval of new agents such as irinotecan, oxaliplatin, and bevacizumab further improved patient survival.4, 5, 6 The anti-epidermal growth factor receptor (EGFR) antibody cetuximab was approved in the United States and Europe in 2004, and another anti-EGFR antibody panitumumab was approved in 2006. Anti-EGFR antibodies bind to the extracellular domain of the EGFR, and prevent its activation by EGFR ligands such as epidermal growth factor and amphiregulin. Consequently, they inhibit the subsequent downstream signaling mediated by the RAS-RAF-extracellular signal-related kinase (ERK) and phosphoinositide 3-kinase (PI3K)-phosphatase and tensin homolog (PTEN)-AKT pathways. Early clinical trials revealed significant but limited efficacy of anti-EGFR antibodies in patients with CRC because some of these patients had a primary resistance to these agents, and patients who initially responded to treatment inevitably became refractory to those agents.7, 8 However, these anti-EGFR antibodies improved overall survival in some patients with CRC, especially in those with wild type RAS.9, 10 Since then, several mechanisms for resistance to anti-EGFR antibody therapy have been discovered in CRC. For example, mutations in KRAS, NRAS, BRAF, and PIC3CA, and loss of PTEN results in resistance to anti-EGFR therapies.11, 12, 13, 14 Furthermore HER2 amplification was identified in anti-EGFR antibody-resistant CRC, and HER2 was identified as the mediator of the bypass signaling.15

Section snippets

Effect of HER2 Amplification on Anti-EGFR Treatment in CRC

HER2 is a member of the human EGFR (HER/EGFR/ERBB) family. Its homodimerization or heterodimerization with any other receptor of the HER family activates its intracellular kinase, resulting in autophosphorylation of tyrosine residues on its intracellular cytoplasmic domain. Activated HER2 stimulates the downstream signaling pathway RAS-RAF-ERK and PI3K-PTEN-AKT, which regulate cell proliferation, apoptosis, and drug resistance. HER2 overexpression is associated with an increased risk of

HER2 Evaluation in Clinical Samples

The frequency of HER2 overexpression and amplification was lower in CRC than in breast cancer and gastric cancer.18, 21, 22, 23 Because of differences in examination methods and objective criteria, the reported frequency of HER2 amplification and overexpression in CRC varies between studies, ranging from 1.5% to 20.0% and from 1.3% to 47.4%, respectively (Table 1).15, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33 Notably, HER2 amplification and KRAS, NRAS, and BRAF mutations were mutually exclusive in

Preclinical Investigations of Anti-HER2 Therapy

Bertotti et al generated large xenograft cohorts from 85 patient-derived, genetically characterized metastatic CRC samples (“xenopatients”) and investigated predictors of therapeutic response and new therapeutic targets.24 Their results indicated that HER2 amplification plays an important role in cetuximab-resistance, specifically in a subset of cetuximab-resistant cases with wild type KRAS/NRAS/BRAF/PIK3CA (quadruple-negative), which are sensitive to dual EGFR/HER2 inhibitors such as lapatinib

Clinical Trials in Patients With HER2 Overexpression

HER2-targeting agents such as trastuzumab, pertuzumab, and lapatinib are part of the standard therapy for patients with HER2-positive cancer, including breast and gastric cancer. Several clinical trials were conducted to assess the efficacy of anti-HER2 therapy in CRC patients with HER2 overexpression (Table 2).35, 41, 42, 43, 44, 45, 46 The effects of combining trastuzumab with standard chemotherapy were assessed in patients with HER2-positive CRC. Trastuzumab was also evaluated in combination

Future Challenges for Optimization of HER2-Targeted Oncotherapy in CRC

HER2 amplification is observed less frequently in CRC than in breast or gastric cancer; therefore, efficient screening of HER2 status is critical. In 2 retrospective studies, HER2 amplification was reported to be mutually exclusive with KRAS, NRAS, and BRAF mutations in tumors from chemotherapy-naive patients.24, 34 This finding suggests that HER2 status should mainly be evaluated in patients with wild type RAS and RAF. Furthermore, HER2 amplification was reported to be highly prevalent in

Conclusions

In the past decade, systemic therapy for CRC has advanced markedly because of the development of molecular targeting agents, especially anti-EGFR antibody. Elucidation of its underlying molecular mechanisms will help optimize anti-EGFR antibody treatment for a subpopulation of CRC patients. Because bypass signaling through HER2 amplification was identified as a mechanism underlying the development of resistance to anti-EGFR therapy, patients with HER2 amplification will likely not benefit from

Disclosure

The authors have stated that they have no conflicts of interest.

Acknowledgments

We thank the medical staff in our department for reviewing an earlier version of this report.

References (47)

  • L.B. Saltz et al.

    Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study

    J Clin Oncol

    (2008)
  • A. de Gramont et al.

    Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer

    J Clin Oncol

    (2000)
  • R.G. Amado et al.

    Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer

    J Clin Oncol

    (2008)
  • C.S. Karapetis et al.

    K-ras mutations and benefit from cetuximab in advanced colorectal cancer

    N Engl J Med

    (2008)
  • L.S. Schwartzberg et al.

    PEAK: a randomized, multicenter phase II study of panitumumab plus modified fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) or bevacizumab plus mFOLFOX6 in patients with previously untreated, unresectable, wild type KRAS exon 2 metastatic colorectal cancer

    J Clin Oncol

    (2014)
  • F. Di Nicolantonio et al.

    Wild type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer

    J Clin Oncol

    (2008)
  • A. Sartore-Bianchi et al.

    PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies

    Cancer Res

    (2009)
  • Z.Y. Yang et al.

    Promising biomarkers for predicting the outcomes of patients with KRAS wild-type metastatic colorectal cancer treated with anti-epidermal growth factor receptor monoclonal antibodies: a systematic review with meta-analysis

    Int J Cancer

    (2013)
  • K. Yonesaka et al.

    Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab

    Sci Transl Med

    (2011)
  • M.D. Begnami et al.

    Prognostic implications of altered human epidermal growth factor receptors (HERs) in gastric carcinomas: HER2 and HER3 are predictors of poor outcome

    J Clin Oncol

    (2011)
  • M.F. Press et al.

    HER2/neu gene amplification characterized by fluorescence in situ hybridization: poor prognosis in node-negative breast carcinomas

    J Clin Oncol

    (1997)
  • D.J. Slamon et al.

    Human breast cancer: correlation of relapse and survival with amplification of the HER2/neu oncogene

    Science

    (1987)
  • G.Z. Yu et al.

    Overexpression of Grb2/HER2 signaling in Chinese gastric cancer: their relationship with clinicopathological parameters and prognostic significance

    J Cancer Res Clin Oncol

    (2009)
  • Cited by (34)

    • Application of histology-agnostic treatments in metastatic colorectal cancer

      2022, Digestive and Liver Disease
      Citation Excerpt :

      The ERBB2 oncogene encodes for a transmembrane glycoprotein receptor (HER2) that functions as an intracellular tyrosine kinase. Despite the lack of a known ligand for HER2, its homodimerization or heterodimerization with other receptors in the EGFR family results in the activation of signal transduction pathways (mainly the MAPK and PI3K-AKT-mTOR pathways) fostering cell growth and proliferation [105]. The HER2 signaling cascade can be secondary to gene amplification (and mutations) or overexpression of its protein product, and has been observed in multiple cancer types [106].

    • Anemoside A3 activates TLR4-dependent M1-phenotype macrophage polarization to represses breast tumor growth and angiogenesis

      2021, Toxicology and Applied Pharmacology
      Citation Excerpt :

      Radiotherapy and chemotherapy, unfortunately, the therapeutic effects are still limited, particularly with advanced breast cancer. ( Takegawa and Yonesaka, 2017). It has been revealed that tumor-associated macrophages (TAMs) play a crucial role in breast cancer development and immunotherapy (Quail and Joyce, 2013).

    • Cardiotoxic mechanisms of cancer immunotherapy – A systematic review

      2021, International Journal of Cardiology
      Citation Excerpt :

      Trastuzumab represents the most prominent example of an Anti-EGFR-antibody, targeting Her2/neu [14]. This agent is widely used to treat Her2 positive breast cancer patients, but also for other solid tumours harbouring Her2 alterations, such gastric or colon cancer [14–16]. As more and more cardiotoxicities have become apparent, this aspect has been intensively studied in recent years [17].

    • Kinome scale profiling of venom effects on cancer cells reveals potential new venom activities

      2020, Toxicon
      Citation Excerpt :

      Reduction in EGFR expression and phosphorylation levels seen in response to C.durissus vegrandis, N.naja as well as scorpion H.swammerdami and theraphosid A.geniculata venoms could open up a large pool of venom-derived biological molecules, from a large diverse population of species, which may possess novel mechanisms and binding sites for the targeted suppression of EGFR and treatment of EGFR-expressing cancers where prior treatment has failed due to therapy resistance. HER2 over-expression has been shown to be linked to the induction and progression of a variety of cancer types including, prostate (Day et al., 2017), colon and colorectal (Takegawa and Yonesaka, 2017; Pirpour Tazehkand et al., 2018; Siena et al., 2018), gastric and oesophageal (Gerson et al., 2017; Wu et al., 2017), ovarian (Luo et al., 2018) and predominantly occurring in the HER2+ breast cancer subtypes (Baselga et al., 2017: Loibl and Gianni, 2017; Pondé et al., 2018). A two-fold reduction was observed in HER2 phosphorylation in response to treatment with N.naja (Fig. 3) and H.swammerdami (Fig. 2) venoms.

    View all citing articles on Scopus
    View full text