Mitochondria-targeted nitroxide, Mito-CP, suppresses medullary thyroid carcinoma cell survival in vitro and in vivo

J Clin Endocrinol Metab. 2013 Apr;98(4):1529-40. doi: 10.1210/jc.2012-3671. Epub 2013 Mar 18.

Abstract

Context: Medullary thyroid carcinoma (MTC) is a neuroendocrine tumor mainly caused by mutations in the RET proto-oncogene. For MTC therapy, the U.S. Food and Drug Administration recently approved vandetanib and cabozantinib, multikinase inhibitors targeting RET and other tyrosine kinase receptors of vascular endothelial growth factor, epidermal growth factor, or hepatocyte growth factor. Nevertheless, not all patients with the progressive MTC respond to these drugs, requiring the development of additional therapeutic modalities that have distinct activity.

Objective: We aimed to evaluate mitochondria-targeted carboxy-proxyl (Mito-CP), a mitochondria-targeted redox-sensitive agent, for its tumor-suppressive efficacy against MTC.

Design: In vitro cultures of 2 human MTC cell lines, TT and MZ-CRC-1, and TT xenografts in mice were treated with Mito-CP in comparison with vandetanib. The effects on cell survival/death, RET expression, mitochondrial integrity, and oxidative stress were determined.

Results: Contrary to vandetanib, Mito-CP induced RET downregulation and strong cytotoxic effects in both cell lines in vitro, including caspase-dependent apoptosis. These effects were accompanied by mitochondrial membrane depolarization, decreased oxygen consumption, and increased oxidative stress in cells. Intriguingly, Mito-CP-induced cell death, but not RET downregulation, was partially inhibited by the reactive oxygen species scavenger, N-acetyl-cysteine, indicating that Mito-CP mediates tumor-suppressive effects via redox-dependent as well as redox-independent mechanisms. Orally administered Mito-CP effectively suppressed TT xenografts in mice, with an efficacy comparable to vandetanib and relatively low toxicity to animals.

Conclusion: Our results suggest that Mito-CP can effectively suppress MTC cell growth/survival via a mechanism distinct from vandetanib effects. Mitochondrial targeting may be a potential strategy for MTC therapy.

Publication types

  • Comparative Study
  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antineoplastic Agents / pharmacology
  • Antineoplastic Agents / therapeutic use
  • Carcinoma, Neuroendocrine
  • Cell Line, Tumor
  • Cell Survival / drug effects
  • Cyclic N-Oxides / pharmacology*
  • Cyclic N-Oxides / therapeutic use*
  • Drug Delivery Systems
  • Female
  • Humans
  • Mice
  • Mice, Nude
  • Mitochondria / drug effects
  • Nitrogen Oxides / pharmacology
  • Nitrogen Oxides / therapeutic use
  • Organophosphorus Compounds / pharmacology*
  • Organophosphorus Compounds / therapeutic use*
  • Piperidines / pharmacology
  • Piperidines / therapeutic use
  • Proto-Oncogene Mas
  • Quinazolines / pharmacology
  • Quinazolines / therapeutic use
  • Thyroid Neoplasms / drug therapy*
  • Thyroid Neoplasms / pathology*
  • Xenograft Model Antitumor Assays

Substances

  • Antineoplastic Agents
  • Cyclic N-Oxides
  • MAS1 protein, human
  • Nitrogen Oxides
  • Organophosphorus Compounds
  • Piperidines
  • Proto-Oncogene Mas
  • Quinazolines
  • mito-carboxy proxyl
  • nitroxyl
  • vandetanib

Supplementary concepts

  • Thyroid cancer, medullary