Repurposing propofol for breast cancer therapy through promoting apoptosis and arresting cell cycle

Oncol Rep. 2024 Nov;52(5):155. doi: 10.3892/or.2024.8814. Epub 2024 Oct 4.

Abstract

Breast cancer is the most prevalent cancer among women worldwide, characterized by a high mortality rate and propensity for metastasis. Although surgery is the standard treatment for breast cancer, there is still no effective method to inhibit tumor metastasis and improve the prognosis of patients with breast cancer after surgery. Propofol, one of the most widely used intravenous anesthetics in surgery, has exhibited a positive association with improved survival outcomes in patients with breast cancer post‑surgery. However, the underlying molecular mechanism remains to be elucidated. The present study revealed that triple negative breast cancer cells, MDA‑MB‑231 and 4T1, exposed to propofol exhibited a significant decrease in cell viability. Notably, propofol exhibited minimal cytotoxic effects on HUVECs under the same conditions. Furthermore, propofol significantly inhibited the migration and invasion ability of MDA‑MB‑231 and 4T1 cells. Propofol promoted apoptosis in 4T1 cells through upregulation of Bax and cleaved caspase 3, while downregulating B‑cell lymphoma‑extra large. Concomitantly, propofol induced cell cycle arrest of 4T1 cells by downregulating cyclin E2 and phosphorylated cell division cycle 6. Furthermore, propofol exhibited excellent anticancer efficacy in a 4T1 breast cancer allograft mouse model. The present study sheds light on the potential of propofol as an old medicine with a novel use for breast cancer treatment.

Keywords: apoptosis; cell cycle arrest; propofol; repurposed drug; triple‑negative breast cancer.

MeSH terms

  • Anesthetics, Intravenous / pharmacology
  • Anesthetics, Intravenous / therapeutic use
  • Animals
  • Apoptosis* / drug effects
  • Breast Neoplasms / drug therapy
  • Breast Neoplasms / pathology
  • Cell Cycle Checkpoints* / drug effects
  • Cell Line, Tumor
  • Cell Movement / drug effects
  • Cell Proliferation / drug effects
  • Cell Survival / drug effects
  • Drug Repositioning*
  • Female
  • Humans
  • Mice
  • Mice, Inbred BALB C
  • Propofol* / pharmacology
  • Propofol* / therapeutic use
  • Triple Negative Breast Neoplasms / drug therapy
  • Triple Negative Breast Neoplasms / pathology
  • Xenograft Model Antitumor Assays

Substances

  • Propofol
  • Anesthetics, Intravenous

Grants and funding

The present study was supported by ‘Science and Technology Program of Guangzhou, China’ (grant no. 2024A04J6603).