Q6, a novel hypoxia-targeted drug, regulates hypoxia-inducible factor signaling via an autophagy-dependent mechanism in hepatocellular carcinoma

Autophagy. 2014 Jan;10(1):111-22. doi: 10.4161/auto.26838. Epub 2013 Nov 11.

Abstract

Tumor hypoxia underlies treatment failure and yields more aggressive and metastatic cancer phenotypes. Although therapeutically targeting these hypoxic environments has been proposed for many years, to date no approaches have shown the therapeutic value to gain regulatory approval. Here, we demonstrated that a novel hypoxia-activated prodrug, Q6, exhibits potent antiproliferative efficacy under hypoxic conditions and induces caspase-dependent apoptosis in 2 hepatocellular carcinoma (HCC) cell lines, with no obvious toxicity being detected in 2 normal liver cell lines. Treatment with Q6 markedly downregulated HIF1A [hypoxia inducible factor 1, α subunit (basic helix-loop-helix transcription factor)] expression and transcription of the downstream target gene, VEGFA (vascular endothelial growth factor A). This dual hypoxia-targeted modulation mechanism leads to high potency in suppressing tumor growth and vascularization in 2 in vivo models. Intriguingly, it is the autophagy-dependent degradation pathway that plays a crucial role in Q6-induced attenuation of HIF1A expression, rather than the proteasome-dependent pathway, which is normally regarded as the predominant mechanism underlying posttranslational regulation of HIF1A. Inhibition of autophagy, either by short interfering RNA (siRNA) or by chemical inhibitors, blocked Q6-induced HIF1A degradation. Autophagic degradation of HIF1A was further confirmed by the observation that HIF1A coimmunoprecipitated with the ubiquitin-binding adaptor protein, SQSTM1, which is degraded through autophagy. Additionally, silencing of SQSTM1 inhibited Q6-induced HIF1A degradation. These findings suggest that the novel hypoxia-targeted agent, Q6, has potential clinical value in the therapy of HCC. Furthermore, the identification of autophagy as a crucial regulator of HIF1A provides new insights into hypoxia-related treatments.

Keywords: autophagy; dual modulation mechanisms; hypoxia-activated prodrug; posttranslational regulation; targeting treatment.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adaptor Proteins, Signal Transducing / metabolism
  • Animals
  • Antineoplastic Agents / pharmacology
  • Apoptosis / drug effects
  • Autophagy / drug effects*
  • Autophagy-Related Protein 5
  • Carcinoma, Hepatocellular / metabolism
  • Carcinoma, Hepatocellular / pathology
  • Carcinoma, Hepatocellular / ultrastructure
  • Caspases / metabolism
  • Cell Hypoxia / drug effects
  • Cell Line, Tumor
  • Cell Proliferation / drug effects
  • Down-Regulation / drug effects
  • Hypoxia-Inducible Factor 1, alpha Subunit / antagonists & inhibitors
  • Hypoxia-Inducible Factor 1, alpha Subunit / genetics
  • Hypoxia-Inducible Factor 1, alpha Subunit / metabolism*
  • Liver Neoplasms / metabolism
  • Liver Neoplasms / pathology*
  • Liver Neoplasms / ultrastructure
  • Lysosomes / drug effects
  • Lysosomes / metabolism
  • Lysosomes / ultrastructure
  • Mice
  • Mice, Nude
  • Microtubule-Associated Proteins / metabolism
  • Proteolysis / drug effects
  • Quinoxalines / pharmacology*
  • Sequestosome-1 Protein
  • Signal Transduction / drug effects
  • Signal Transduction / genetics
  • Transcription, Genetic / drug effects

Substances

  • ATG5 protein, human
  • Adaptor Proteins, Signal Transducing
  • Antineoplastic Agents
  • Autophagy-Related Protein 5
  • Hypoxia-Inducible Factor 1, alpha Subunit
  • MAP1LC3B protein, human
  • Microtubule-Associated Proteins
  • Quinoxalines
  • SQSTM1 protein, human
  • Sequestosome-1 Protein
  • Caspases