Carrier-free multifunctional nanomedicine for intraperitoneal disseminated ovarian cancer therapy

J Nanobiotechnology. 2022 Feb 22;20(1):93. doi: 10.1186/s12951-022-01300-4.

Abstract

Background: Ovarian cancer is the most lethal gynecological cancer which is characterized by extensive peritoneal implantation metastasis and malignant ascites. Despite advances in diagnosis and treatment in recent years, the five-year survival rate is only 25-30%. Therefore, developing multifunctional nanomedicine with abilities of promoting apoptosis and inhibiting migration on tumor cells would be a promising strategy to improve the antitumor effect.

Methods and results: In this study, we developed a novel ACaT nanomedicine composed of alendronate, calcium ions and cyclin-dependent kinase 7 (CDK7) inhibitor THZ1. With the average size of 164 nm and zeta potential of 12.4 mV, the spherical ACaT nanoparticles were selectively internalized by tumor cells and effectively accumulated in the tumor site. Results of RNA-sequencing and in vitro experiments showed that ACaT promoted tumor cell apoptosis and inhibited tumor cell migration by arresting the cell cycle, increasing ROS and affecting calcium homeostasis. Weekly intraperitoneally administered of ACaT for 8 cycles significantly inhibited the growth of tumor and prolonged the survival of intraperitoneal xenograft mice.

Conclusion: In summary, this study presents a new self-assembly nanomedicine with favorable tumor targeting, antitumor activity and good biocompatibility, providing a novel therapeutic strategy for advanced ovarian cancer.

Keywords: Alendronate; Nanomedicine; Ovarian cancer; Self-assembly; THZ1.

MeSH terms

  • Animals
  • Apoptosis
  • Cell Line, Tumor
  • Humans
  • Mice
  • Nanomedicine*
  • Ovarian Neoplasms* / drug therapy
  • Protein Kinase Inhibitors / pharmacology
  • Xenograft Model Antitumor Assays

Substances

  • Protein Kinase Inhibitors