Anti-angiogenic and anti-metastatic activity of synthetic phosphoethanolamine

PLoS One. 2013;8(3):e57937. doi: 10.1371/journal.pone.0057937. Epub 2013 Mar 14.

Abstract

Background: Renal cell carcinoma (RCC) is the most common type of kidney cancer, and represents the third most common urological malignancy. Despite the advent of targeted therapies for RCC and the improvement of the lifespan of patients, its cost-effectiveness restricted the therapeutic efficacy. In a recent report, we showed that synthetic phosphoethanolamine (Pho-s) has a broad antitumor activity on a variety of tumor cells and showed potent inhibitor effects on tumor progress in vivo.

Methodology/principal findings: We show that murine renal carcinoma (Renca) is more sensitive to Pho-s when compared to normal immortalized rat proximal tubule cells (IRPTC) and human umbilical vein endothelial cells (HUVEC). In vitro anti-angiogenic activity assays show that Pho-s inhibits endothelial cell proliferation, migration and tube formation. In addition, Pho-s has anti-proliferative effects on HUVEC by inducing a cell cycle arrest at the G2/M phase. It causes a decrease in cyclin D1 mRNA, VEGFR1 gene transcription and VEGFR1 receptor expression. Pho-s also induces nuclear fragmentation and affects the organization of the cytoskeleton through the disruption of actin filaments. Additionally, Pho-s induces apoptosis through the mitochondrial pathway. The putative therapeutic potential of Pho-s was validated in a renal carcinoma model, on which our remarkable in vivo results show that Pho-s potentially inhibits lung metastasis in nude mice, with a superior efficacy when compared to Sunitinib.

Conclusions/significance: Taken together, our findings provide evidence that Pho-s is a compound that potently inhibits lung metastasis, suggesting that it is a promising novel candidate drug for future developments.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Angiogenesis Inhibitors / chemical synthesis
  • Angiogenesis Inhibitors / pharmacology*
  • Angiogenesis Inhibitors / toxicity
  • Animals
  • Antineoplastic Agents / chemical synthesis
  • Antineoplastic Agents / pharmacology*
  • Antineoplastic Agents / toxicity
  • Apoptosis / drug effects
  • Carcinoma, Renal Cell / drug therapy
  • Carcinoma, Renal Cell / pathology
  • Caspase 3 / metabolism
  • Cell Cycle Checkpoints / drug effects
  • Cell Line, Tumor
  • Cell Movement / drug effects
  • Cell Proliferation / drug effects
  • Cyclin D1 / genetics
  • Cytoskeleton / drug effects
  • Ethanolamines / chemical synthesis
  • Ethanolamines / pharmacology*
  • Ethanolamines / toxicity
  • Gene Expression Regulation, Neoplastic / drug effects
  • Human Umbilical Vein Endothelial Cells / drug effects
  • Humans
  • Indoles / pharmacology
  • Indoles / toxicity
  • Kidney Tubules, Proximal / drug effects
  • Lung Neoplasms / drug therapy
  • Lung Neoplasms / secondary
  • Male
  • Mice
  • Mice, Inbred BALB C
  • Mitochondria / drug effects
  • Mitochondria / metabolism
  • Neoplasm Metastasis / drug therapy
  • Neovascularization, Pathologic / drug therapy
  • Oxidation-Reduction / drug effects
  • Pyrroles / pharmacology
  • Pyrroles / toxicity
  • Rats
  • Sunitinib
  • Transcription, Genetic
  • Vascular Endothelial Growth Factor Receptor-1 / genetics
  • Wound Healing / drug effects

Substances

  • Angiogenesis Inhibitors
  • Antineoplastic Agents
  • Ethanolamines
  • Indoles
  • Pyrroles
  • Cyclin D1
  • phosphorylethanolamine
  • Vascular Endothelial Growth Factor Receptor-1
  • Caspase 3
  • Sunitinib

Grants and funding

This work was supported by a Sao Paulo Research Foundation (FAPESP) - grant (2007/50571-3; Doctorate and Master Fellowship 2008/56089-1; 2010/50220-9). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.