Targeting Phosphatidylinositol 3-Kinase Signaling Pathway for Therapeutic Enhancement of Vascular-Targeted Photodynamic Therapy

Mol Cancer Ther. 2017 Nov;16(11):2422-2431. doi: 10.1158/1535-7163.MCT-17-0326. Epub 2017 Aug 23.

Abstract

Vascular-targeted photodynamic therapy (PDT) selectively disrupts vascular function by inducing oxidative damages to the vasculature, particularly endothelial cells. Although effective tumor eradication and excellent safety profile are well demonstrated in both preclinical and clinical studies, incomplete vascular shutdown and angiogenesis are known to cause tumor recurrence after vascular-targeted PDT. We have explored therapeutic enhancement of vascular-targeted PDT with PI3K signaling pathway inhibitors because the activation of PI3K pathway was involved in promoting endothelial cell survival and proliferation after PDT. Here, three clinically relevant small-molecule inhibitors (BYL719, BKM120, and BEZ235) of the PI3K pathway were evaluated in combination with verteporfin-PDT. Although all three inhibitors were able to synergistically enhance PDT response in endothelial cells, PDT combined with dual PI3K/mTOR inhibitor BEZ235 exhibited the strongest synergism, followed in order by combinations with pan-PI3K inhibitor BKM120 and p110α isoform-selective inhibitor BYL719. Combination treatments of PDT and BEZ235 exhibited a cooperative inhibition of antiapoptotic Bcl-2 family protein Mcl-1 and induced more cell apoptosis than each treatment alone. In addition to increasing treatment lethality, BEZ235 combined with PDT effectively inhibited PI3K pathway activation and consequent endothelial cell proliferation after PDT alone, leading to a sustained growth inhibition. In the PC-3 prostate tumor model, combination treatments improved treatment outcomes by turning a temporary tumor regrowth delay induced by PDT alone to a more long-lasting treatment response. Our study strongly supports the combination of vascular-targeted PDT and PI3K pathway inhibitors, particularly mTOR inhibitors, for therapeutic enhancement. Mol Cancer Ther; 16(11); 2422-31. ©2017 AACR.

MeSH terms

  • Aminopyridines / administration & dosage
  • Animals
  • Apoptosis / drug effects
  • Cell Line, Tumor
  • Cell Proliferation / drug effects
  • Combined Modality Therapy
  • Endothelial Cells / drug effects
  • Endothelial Cells / pathology
  • Humans
  • Imidazoles / administration & dosage
  • Mice
  • Morpholines / administration & dosage
  • Myeloid Cell Leukemia Sequence 1 Protein / genetics
  • Neoplasm Recurrence, Local / drug therapy*
  • Neoplasm Recurrence, Local / genetics
  • Neoplasm Recurrence, Local / pathology
  • Neoplasms / drug therapy*
  • Neoplasms / genetics
  • Neoplasms / pathology
  • Neovascularization, Pathologic / drug therapy*
  • Neovascularization, Pathologic / genetics
  • Neovascularization, Pathologic / pathology
  • Oxidative Stress / drug effects
  • Phosphatidylinositol 3-Kinases / genetics
  • Phosphoinositide-3 Kinase Inhibitors*
  • Photochemotherapy*
  • Porphyrins / administration & dosage
  • Protein Kinase Inhibitors / administration & dosage
  • Quinolines / administration & dosage
  • Signal Transduction / drug effects
  • Thiazoles / administration & dosage
  • Verteporfin
  • Xenograft Model Antitumor Assays

Substances

  • Aminopyridines
  • Imidazoles
  • MCL1 protein, human
  • Morpholines
  • Myeloid Cell Leukemia Sequence 1 Protein
  • NVP-BKM120
  • Phosphoinositide-3 Kinase Inhibitors
  • Porphyrins
  • Protein Kinase Inhibitors
  • Quinolines
  • Thiazoles
  • Alpelisib
  • Verteporfin
  • dactolisib