Elsevier

Hearing Research

Volume 328, October 2015, Pages 59-66
Hearing Research

Research paper
Canertinib induces ototoxicity in three preclinical models

https://doi.org/10.1016/j.heares.2015.07.002Get rights and content

Highlights

  • Canertinib, a new cancer drug candidate blocking ERBB signaling, causes toxicity in hair cells of zebrafish lateral line.

  • Canertinib ototoxicity is confirmed in two mouse models.

  • One major target of its ototoxicity in mice is outer hair cells.

  • Ototoxicity should be monitored for cancer drug candidates with pan-ERBB inhibition properties.

Abstract

Neuregulin-1 (NRG1) ligand and its epidermal growth factor receptor (EGFR)/ERBB family regulate normal cellular proliferation and differentiation in many tissues including the cochlea. Aberrant NRG1 and ERBB signaling cause significant hearing impairment in mice. Dysregulation of the same signaling pathway in humans is involved in certain types of cancers such as breast cancer or non-small cell lung cancer (NSCLC). A new irreversible pan-ERBB inhibitor, canertinib, has been tested in clinical trials for the treatment of refractory NSCLC. Its possible ototoxicity was unknown. In this study, a significant dose-dependent canertinib ototoxicity was observed in a zebrafish model. Canertinib ototoxicity was further confirmed in two mouse models with different genetic backgrounds. The data strongly suggested an evolutionally preserved ERBB molecular mechanism underlying canertinib ototoxicity. Thus, these results imply that clinical monitoring of hearing loss should be considered for clinical testing of canertinib or other pan-ERBB inhibitors.

Introduction

Lung cancer is the number one cause of cancer mortality globally with few screening and treatment options available (Jemal et al., 2011). Non-small cell lung cancer (NSCLC) accounts for 80% of all lung cancer cases (Jemal et al., 2010).

Advanced NSCLCs (Stage IIIb and IV) are treated with cisplatin or combination therapies; however, the five-year survival rate is under 5% (de Castria et al., 2013). Cisplatin, as first-line treatment of many solid tumors, has been extensively studied for its ototoxicity (Lobarinas et al., 2013, Harrison et al., 2014), however, its effectiveness against NSCLC is often subverted (Lipp and Hartmann, 2005, Yorgason et al., 2006, Schacht et al., 2012). Recent research into novel anti-NSCLC drugs has focused on inhibition of the epidermal growth factor receptors (EGFRs/ERBB), which belong to the receptor family of tyrosine kinases (RTKs).

RTK inhibitors, which block ERBB receptors, present novel and exciting therapies for NSCLC (Cowan-Jacob, 2006, Pytel et al., 2009, Awada et al., 2012, Ou, 2012, de Castria et al., 2013, Majem and Pallares, 2013). Most ERBB family members have three domains: an extracellular N-terminus responsible for ligand binding, a transmembrane hydrophobic region, and a conserved intracellular domain with a ligand-activated tyrosine kinase on the C-terminal end (Allen et al., 2003). The ERBB family of receptors includes ERBB1 (EGFR), ERBB2 (Her-2/neu), ERBB3 (Her-3), and ERBB4 (Her-4) (Janne et al., 2007). Aberrant signaling of ERBB family members (Hynes and Stern, 1994, Leung et al., 1997) plays an important role in many cancers including breast (Sainsbury et al., 1985), ovarian (Meden and Kuhn, 1997), prostate (Schwartz et al., 1999), gastric (Hirono et al., 1995), laryngeal (Almadori et al., 1999), and indicate a more aggressive cancer phenotype (Klijn et al., 1994, Zinner et al., 2007). ERBB1 is overexpressed in the majority of NSCLC and is found frequently in lung squamous cell carcinoma (57–100%) whereas ERBB2 is found in lung adenocarcinomas (33%).

Gefitinib and erlotinib are first generation EGFR RTK inhibitors that result in 10–20% lung tumor suppression but do not inhibit other ERBB family members (Wakeling et al., 2002, Barbacci et al., 2003). After treatment with first generation RTK inhibitors, 50% of patients have a secondary missense T790 mutation resulting in acquired resistance due to steric hindrance with a bulkier methionine (Pao et al., 2005, Shepherd et al., 2005, Engelman and Janne, 2008, Sequist et al., 2011, Majem and Pallares, 2013). Although no preclinical studies of their ototoxicity, one clinical case was reported with erlotinib ototoxicity (Koutras et al., 2008). The success of this first generation inhibitors coupled with the known acquired resistance has resulted in a need for new RTK inhibitor therapies with more sustained effects than gefitinib and erlotinib (Han et al., 2008, Butts et al., 2010, Mukherjea et al., 2011, Allen et al., 2012, Yang et al., 2013, Agustoni et al., 2014). Second and third generation ERBB inhibitors are irreversible, have higher binding affinities, inhibit multiple ERBB family members, and have better activity against T790 mutation. Therefore, these new inhibitors, such as canertinib, may provide additional clinical benefits by functioning as a pan-ERBB inhibitor (Fry, 2000, Slichenmyer et al., 2001, Allen et al., 2003, Janne et al., 2007). Although adverse effects such as diarrhea, asthenia, stomatitis, and persistent rash were observed in canertinib clinical trials (Janne et al., 2007), no clinical studies of ototoxicity for these new inhibitors are reported so far.

The NRG1-ERBB signaling pathway plays a significant role in the development and mature function of the inner ear. In the cochlea, NRG1 is expressed in spiral ganglion neurons (SGNs) (Morley et al., 1998, Bao et al., 2003, Bao et al., 2004); whereas hair cells, Schwann cells, and supporting cells express ERBB (Morley, 1998, Hansen et al., 2001, Zhang et al., 2002, Hume et al., 2003). Cochlear innervation is abnormal in NRG1 and ERBB2 null mice and these adult mice have progressive hearing loss (Lee et al., 1995, Meyer and Birchmeier, 1995, Fritzsch et al., 1997, Morley, 1998, Adlkofer and Lai, 2000, Chen et al., 2003, Fritzsch et al., 2004, Hellard et al., 2004, Stankovic et al., 2004, Morris et al., 2006). One chemical compound blocking ERBB signaling could lead to progressive hearing loss in mature female pigmented guinea pigs (Watanabe et al., 2010). Since canertinib can block ERBB receptors, we first assessed possible hair cell toxicity of canertinib in a zebrafish model, and then in two mouse models (C57BL/6J and CBA/CaJ) by auditory brainstem recording (ABR) thresholds, distortion product otoacoustic emissions (DPOAE), and hair cell counts. Canertinib resulted in significant ototoxicity in all three preclinical animal models.

Section snippets

Zebrafish studies

Zebrafish (Danio rerio) embryos were produced by paired matings of adult fish maintained at the University of Washington zebrafish facility. We used AB wild-type zebrafish strains. Embryos were maintained in fish embryo media (EM; 1 mM MgSO4, 120 μM KH2PO4, 74 μM Na2HPO4, 1 mM CaCl2, 500 μM KCl, 15 μM NaCl, and 500 μM NaHCO3 in dH2O) at a density of 50 animals per 100 mm2 Petri dish and kept in an incubator at 28.5 °C. At 4 days post-fertilization (dpf), larvae were fed live paramecia. All

Toxicity to zebrafish lateral line hair cells

Since the zebrafish lateral line allows rapid assessment and quantification of hair cell toxicity (Hirose et al., 2011, Ou et al., 2012, Thomas et al., 2013), we first tested possible canertinib toxicity in zebrafish (Fig. 1). At the lower concentrations from 0 to 50 μM, canertinib showed no obvious hair cell toxicity, while at the higher doses, it clearly demonstrated a dose-dependent loss of hair cell at 100 μM (p = 0.28 × 10−3), or 200 μM (p = 0.18 × 10−10). Doses above 200 μM were lethal to

Discussion

Based on a possible shared RTK molecular signaling mechanism, we examined possible canertinib ototoxicity in three preclinical animal models. The zebrafish studies clearly showed dose-dependent canertinib ototoxicity, which was further confirmed in two mouse models with different genetic backgrounds. Thus, our study strongly suggested an evolutionally preserved ERBB molecular mechanism underlying canertinib ototoxicity. Because canertinib is a promising NSCLC drug candidate for one of the

Conclusions

RTK inhibitors blocking ERBB signaling are a promising new treatment for advanced NSCLC. Given the importance of ERBB signaling in the cochlea, it is important to consider the potential ototoxicity of these new cancer drug candidates. Here, we discovered the ototoxic effects of canertinib, indicating the importance of assessing human ototoxicity in future therapies that inhibit ERBB family members.

Conflicts of interest

All authors state that they have no conflicts of interests.

Acknowledgments

The project was supported by grants to J.B. from the National Institute of Health (DC010489 and DC011793). We thank Debin Lei for his laboratory management and assistance with mouse husbandry.

References (74)

  • N.E. Hynes et al.

    The biology of erbB-2/neu/HER-2 and its role in cancer

    Biochim. Biophys. Acta

    (1994)
  • E. Lobarinas et al.

    Insensitivity of the audiogram to carboplatin induced inner hair cell loss in chinchillas

    Hear Res.

    (2013)
  • H. Meden et al.

    Overexpression of the oncogene c-erbB-2 (HER2/neu) in ovarian cancer: a new prognostic factor

    Eur. J. Obstet. Gynecol. Reprod. Biol.

    (1997)
  • B.J. Morley

    ARIA is heavily expressed in rat peripheral auditory and vestibular ganglia

    Brain Res. Mol. Brain Res.

    (1998)
  • B.J. Morley et al.

    Identification of the subunits of the nicotinic cholinergic receptors in the rat cochlea using RT-PCR and in situ hybridization

    Brain Res. Mol. Brain Res.

    (1998)
  • J.K. Morris et al.

    A disorganized innervation of the inner ear persists in the absence of ErbB2

    Brain Res.

    (2006)
  • K.K. Ohlemiller et al.

    Vulnerability to noise-induced hearing loss in ‘middle-aged’ and young adult mice: a dose-response approach in CBA, C57BL, and BALB inbred strains

    Hear. Res.

    (2000)
  • H. Ou et al.

    Screening for chemicals that affect hair cell death and survival in the zebrafish lateral line

    Hear. Res.

    (2012)
  • S.H. Ou

    Second-generation irreversible epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs): a better mousetrap? A review of the clinical evidence

    Crit. Rev. Oncol. Hematol.

    (2012)
  • W.J. Slichenmyer et al.

    CI-1033, a pan-erbB tyrosine kinase inhibitor

    Semin. Oncol.

    (2001)
  • T. Yang et al.

    The molecular basis of making spiral ganglion neurons and connecting them to hair cells of the organ of corti

    Hear. Res.

    (2011)
  • M. Zhang et al.

    Expression of heregulin and ErbB/Her receptors in adult chinchilla cochlear and vestibular sensory epithelium

    Hear. Res.

    (2002)
  • K. Adlkofer et al.

    Role of neuregulins in glial cell development

    Glia

    (2000)
  • C.T. Allen et al.

    The clinical implications of antitumor immunity in head and neck cancer

    Laryngoscope

    (2012)
  • G. Almadori et al.

    Epidermal growth factor receptor expression in primary laryngeal cancer: an independent prognostic factor of neck node relapse

    Int. J. Cancer

    (1999)
  • J. Bao et al.

    Requirement of nicotinic acetylcholine receptor subunit beta2 in the maintenance of spiral ganglion neurons during aging

    J. Neurosci.

    (2005)
  • J. Bao et al.

    Activity-dependent transcription regulation of PSD-95 by neuregulin-1 and Eos

    Nat. Neurosci.

    (2004)
  • J. Bao et al.

    Back signaling by the Nrg-1 intracellular domain

    J. Cell. Biol.

    (2003)
  • E.G. Barbacci et al.

    The biological and biochemical effects of CP-654577, a selective erbB2 kinase inhibitor, on human breast cancer cells

    Cancer Res.

    (2003)
  • S. Chen et al.

    Disruption of ErbB receptor signaling in adult non-myelinating Schwann cells causes progressive sensory loss

    Nat. Neurosci.

    (2003)
  • S.W. Cowan-Jacob

    Structural biology of protein tyrosine kinases

    Cell. Mol. Life Sci.

    (2006)
  • T.B. de Castria et al.

    Cisplatin versus carboplatin in combination with third-generation drugs for advanced non-small cell lung cancer

    Cochrane Database Syst. Rev.

    (2013)
  • D. Ding et al.

    Cochlear Hair Cell Densities and Inner-ear Staining techniques. James F. Willott. Handbook of Mouse Auditory Research

    (2001)
  • D. Ding et al.

    Addition of exogenous NAD(+) prevents mefloquine-induced neuroaxonal and hair cell degeneration through reduction of caspase-3-mediated apoptosis in cochlear organotypic cultures

    PLoS One

    (2013)
  • D.L. Ding et al.

    Selective loss of inner hair cells and type-I ganglion neurons in carboplatin-treated chinchillas. Mechanisms of damage and protection

    Ann. N. Y. Acad. Sci.

    (1999)
  • J.A. Engelman et al.

    Mechanisms of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer

    Clin. Cancer Res.

    (2008)
  • I. Farinas et al.

    Spatial shaping of cochlear innervation by temporally regulated neurotrophin expression

    J. Neurosci.

    (2001)
  • Cited by (0)

    View full text