Login / Signup

Controversial Role of Transferrin in the Transport of Ruthenium Anticancer Drugs.

Aviva LevinaAnthony R M ChetcutiPeter A Lay
Published in: Biomolecules (2022)
Ruthenium complexes are at the forefront of developments in metal-based anticancer drugs, but many questions remain open regarding their reactivity in biological media, including the role of transferrin (Tf) in their transport and cellular uptake. A well-known anticancer drug, KP1019 ((IndH)[Ru III Cl 4 (Ind) 2 ], where Ind = indazole) and a reference complex, [Ru III (nta) 2 ] 3- (nta = nitrilotriacetato(3-)) interacted differently with human apoTf, monoFeTf, or Fe 2 Tf. These reactions were studied by biolayer interferometry (BLI) measurements of Ru-Fe-Tf binding to recombinant human transferrin receptor 1 (TfR1) in conjunction with UV-vis spectroscopy and particle size analysis. Cellular Ru uptake in human hepatoma (HepG2) cells was measured under the conditions of the BLI assays. The mode of Tf binding and cellular Ru uptake were critically dependent on the nature of Ru complex, availability of Fe(III) binding sites of Tf, and the presence of proteins that competed for metal binding, particularly serum albumin. Cellular uptake of KP1019 was not Tf-mediated and occurred mostly by passive diffusion, which may also be suitable for treatments of inoperable cancers by intratumoral injections. High cellular Ru uptake from a combination of [Ru III (nta) 2 ] 3- and Fe 2 Tf in the absence of significant Ru-Tf binding was likely to be due to trapping of Ru(III) species into the endosome during TfR1-mediated endocytosis of Fe 2 Tf.
Keyphrases
  • energy transfer
  • endothelial cells
  • high resolution
  • emergency department
  • binding protein
  • radiation therapy
  • mass spectrometry
  • quantum dots
  • drug induced
  • adverse drug
  • rectal cancer