A novel preclinical strategy for identifying cardiotoxic kinase inhibitors and mechanisms of cardiotoxicity

Circ Res. 2011 Dec 9;109(12):1401-9. doi: 10.1161/CIRCRESAHA.111.255695. Epub 2011 Oct 13.

Abstract

Rationale: Despite intense interest in strategies to predict which kinase inhibitor (KI) cancer therapeutics may be associated with cardiotoxicity, current approaches are inadequate. Sorafenib is a KI of concern because it inhibits growth factor receptors and Raf-1/B-Raf, kinases that are upstream of extracellular signal-regulated kinases (ERKs) and signal cardiomyocyte survival in the setting of stress.

Objectives: To explore the potential use of zebrafish as a preclinical model to predict cardiotoxicity and to determine whether sorafenib has associated cardiotoxicity, and, if so, define the mechanisms.

Methods and results: We find that the zebrafish model is readily able to discriminate a KI with little or no cardiotoxicity (gefitinib) from one with demonstrated cardiotoxicity (sunitinib). Sorafenib, like sunitinib, leads to cardiomyocyte apoptosis, a reduction in total myocyte number per heart, contractile dysfunction, and ventricular dilatation in zebrafish. In cultured rat cardiomyocytes, sorafenib induces cell death. This can be rescued by adenovirus-mediated gene transfer of constitutively active MEK1, which restores ERK activity even in the presence of sorafenib. Whereas growth factor-induced activation of ERKs requires Raf, α-adrenergic agonist-induced activation of ERKs does not require it. Consequently, activation of α-adrenergic signaling markedly decreases sorafenib-induced cell death. Consistent with these in vitro data, inhibition of α-adrenergic signaling with the receptor antagonist prazosin worsens sorafenib-induced cardiomyopathy in zebrafish.

Conclusions: Zebrafish may be a valuable preclinical tool to predict cardiotoxicity. The α-adrenergic signaling pathway is an important modulator of sorafenib cardiotoxicity in vitro and in vivo and appears to act through a here-to-fore unrecognized signaling pathway downstream of α-adrenergic activation that bypasses Raf to activate ERKs.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Animals
  • Animals, Genetically Modified
  • Apoptosis / drug effects*
  • Benzenesulfonates / pharmacology*
  • Cardiotoxins / pharmacology*
  • Cell Survival / drug effects
  • Cells, Cultured
  • Extracellular Signal-Regulated MAP Kinases / antagonists & inhibitors
  • Female
  • Gefitinib
  • Indoles / pharmacology*
  • Male
  • Models, Animal
  • Myocytes, Cardiac / cytology
  • Myocytes, Cardiac / drug effects*
  • Niacinamide / analogs & derivatives
  • Phenylurea Compounds
  • Protein Kinase Inhibitors / pharmacology*
  • Proto-Oncogene Proteins B-raf / antagonists & inhibitors
  • Proto-Oncogene Proteins c-raf / antagonists & inhibitors
  • Pyridines / pharmacology*
  • Pyrroles / pharmacology*
  • Quinazolines / pharmacology*
  • Rats
  • Rats, Sprague-Dawley
  • Signal Transduction / drug effects
  • Sorafenib
  • Sunitinib
  • Zebrafish

Substances

  • Benzenesulfonates
  • Cardiotoxins
  • Indoles
  • Phenylurea Compounds
  • Protein Kinase Inhibitors
  • Pyridines
  • Pyrroles
  • Quinazolines
  • Niacinamide
  • Sorafenib
  • Proto-Oncogene Proteins B-raf
  • Proto-Oncogene Proteins c-raf
  • Extracellular Signal-Regulated MAP Kinases
  • Gefitinib
  • Sunitinib