4.8 Article

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

Journal

THERANOSTICS
Volume 9, Issue 20, Pages 5924-5936

Publisher

IVYSPRING INT PUBL
DOI: 10.7150/thno.36320

Keywords

iron oxide nanoclusters; TRAIL; photothermal therapy; magnetic hyperthermia; apoptosis

Funding

  1. National Research Agency program DECRET [ANR ANR-13-SECU-0001]
  2. Labex SEAM actions [ANR 11 LABX 086, ANR 11 IDEX 05 02]
  3. ANR (Agence Nationale de la Recherche) program Investissements d'Avenir Labex LipSTIC [ANR-11-LABX-0021-01]
  4. COFECUB/CAMPUS FRANCE [Me 888-17]
  5. European commission FEDER (Fonds Europeen de Developpement Regional) [BG0004892]
  6. RISE (DISCOVER) [777995]
  7. ARC (Association pour la Recherche sur le Cancer)
  8. La Ligue contre le Cancer

Ask authors/readers for more resources

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.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available