TRAIL acts synergistically with iron oxide nanocluster-mediated magneto- and photothermia

Theranostics. 2019 Aug 14;9(20):5924-5936. doi: 10.7150/thno.36320. eCollection 2019.

Abstract

Targeting TRAIL (Tumor necrosis factor (TNF)-Related Apoptosis-Inducing Ligand) receptors for cancer therapy remains challenging due to tumor cell resistance and poor preparations of TRAIL or its derivatives. Herein, to optimize its therapeutic use, TRAIL was grafted onto iron oxide nanoclusters (NCs) with the aim of increasing its pro-apoptotic potential through nanoparticle-mediated magnetic hyperthermia (MHT) or photothermia (PT). Methods: The nanovector, NC@TRAIL, was characterized in terms of size, grafting efficiency, and potential for MHT and PT. The therapeutic function was assessed on a TRAIL-resistant breast cancer cell line, MDA-MB-231, wild type (WT) or TRAIL-receptor-deficient (DKO), by combining complementary methylene blue assay and flow cytometry detection of apoptosis and necrosis. Results: Combined with MHT or PT under conditions of "moderate hyperthermia" at low concentrations, NC@TRAIL acts synergistically with the TRAIL receptor to increase the cell death rate beyond what can be explained by the mere global elevation of temperature. In contrast, all results are consistent with the idea that there are hotspots, close to the nanovector and, therefore, to the membrane receptor, which cause disruption of the cell membrane. Furthermore, nanovectors targeting other membrane receptors, unrelated to the TNF superfamily, were also found to cause tumor cell damage upon PT. Indeed, functionalization of NCs by transferrin (NC@Tf) or human serum albumin (NC@HSA) induces tumor cell killing when combined with PT, albeit less efficiently than NC@TRAIL. Conclusions: Given that magnetic nanoparticles can easily be functionalized with molecules or proteins recognizing membrane receptors, these results should pave the way to original remote-controlled antitumoral targeted thermal therapies.

Keywords: TRAIL; apoptosis; iron oxide nanoclusters; magnetic hyperthermia; photothermal therapy.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Apoptosis / drug effects
  • Cell Death / physiology
  • Cell Line, Tumor
  • Cell Survival / drug effects*
  • Ferric Compounds / chemistry*
  • Ferric Compounds / pharmacology*
  • Flow Cytometry
  • Humans
  • Hyperthermia, Induced / methods*
  • Microscopy, Electron, Transmission
  • TNF-Related Apoptosis-Inducing Ligand / chemistry*
  • TNF-Related Apoptosis-Inducing Ligand / pharmacology*
  • Tumor Necrosis Factor-alpha / metabolism

Substances

  • Ferric Compounds
  • TNF-Related Apoptosis-Inducing Ligand
  • Tumor Necrosis Factor-alpha
  • ferric oxide