Suppression of Tumor Energy Supply by Liposomal Nanoparticle-Mediated Inhibition of Aerobic Glycolysis

ACS Appl Mater Interfaces. 2018 Jan 24;10(3):2347-2353. doi: 10.1021/acsami.7b16685. Epub 2018 Jan 9.

Abstract

Aerobic glycolysis enables cancer cells to rapidly take up nutrients (e.g., nucleotides, amino acids, and lipids) and incorporate them into the biomass needed to produce a new cell. In contrast to existing chemotherapy/radiotherapy strategies, inhibiting aerobic glycolysis to limit the adenosine 5'-triphosphate (ATP) yield is a highly efficient approach for suppressing tumor cell proliferation. However, most, if not all, current inhibitors of aerobic glycolysis cause significant adverse effects because of their nonspecific delivery and distribution to nondiseased organs, low bioavailability, and a narrow therapeutic window. New strategies to enhance the biosafety and efficacy of these inhibitors are needed for moving them into clinical applications. To address this need, we developed a liposomal nanocarrier functionalized with a well-validated tumor-targeting peptide to specifically deliver the aerobic glycolysis inhibitor 3-bromopyruvate (3-BP) into the tumor tissue. The nanoparticles effectively targeted tumors after systemic administration into tumor-bearing mice and suppressed tumor growth by locally releasing 3-BP to inhibit the ATP production of the tumor cells. No overt side effects were observed in the major organs. This report demonstrates the potential utility of the nanoparticle-enabled delivery of an aerobic glycolysis inhibitor as an anticancer therapeutic agent.

Keywords: 3-bromopyruvate; ATP; Warburg effect; liposomal nanoparticles; tumor-targeting peptide.

MeSH terms

  • Adenosine Triphosphate
  • Animals
  • Cell Line, Tumor
  • Cell Proliferation
  • Glycolysis
  • Liposomes
  • Mice
  • Nanoparticles
  • Neoplasms*

Substances

  • Liposomes
  • Adenosine Triphosphate