Biomolecules (Sep 2022)

Tumor Treating Fields (TTFields) Reversibly Permeabilize the Blood–Brain Barrier In Vitro and In Vivo

  • Ellaine Salvador,
  • Almuth F. Kessler,
  • Dominik Domröse,
  • Julia Hörmann,
  • Clara Schaeffer,
  • Aiste Giniunaite,
  • Malgorzata Burek,
  • Catherine Tempel-Brami,
  • Tali Voloshin,
  • Alexandra Volodin,
  • Adel Zeidan,
  • Moshe Giladi,
  • Ralf-Ingo Ernestus,
  • Mario Löhr,
  • Carola Y. Förster,
  • Carsten Hagemann

DOI
https://doi.org/10.3390/biom12101348
Journal volume & issue
Vol. 12, no. 10
p. 1348

Abstract

Read online

Despite the availability of numerous therapeutic substances that could potentially target CNS disorders, an inability of these agents to cross the restrictive blood–brain barrier (BBB) limits their clinical utility. Novel strategies to overcome the BBB are therefore needed to improve drug delivery. We report, for the first time, how Tumor Treating Fields (TTFields), approved for glioblastoma (GBM), affect the BBB’s integrity and permeability. Here, we treated murine microvascular cerebellar endothelial cells (cerebEND) with 100–300 kHz TTFields for up to 72 h and analyzed the expression of barrier proteins by immunofluorescence staining and Western blot. In vivo, compounds normally unable to cross the BBB were traced in healthy rat brain following TTFields administration at 100 kHz. The effects were analyzed via MRI and immunohistochemical staining of tight-junction proteins. Furthermore, GBM tumor-bearing rats were treated with paclitaxel (PTX), a chemotherapeutic normally restricted by the BBB combined with TTFields at 100 kHz. The tumor volume was reduced with TTFields plus PTX, relative to either treatment alone. In vitro, we demonstrate that TTFields transiently disrupted BBB function at 100 kHz through a Rho kinase-mediated tight junction claudin-5 phosphorylation pathway. Altogether, if translated into clinical use, TTFields could represent a novel CNS drug delivery strategy.

Keywords