Gadolinium Oxide Nanoparticles Reinforce the Fractionated Radiotherapy-Induced Immune Response in Tri-Negative Breast Cancer via cGAS-STING Pathway

Int J Nanomedicine. 2023 Dec 15:18:7713-7728. doi: 10.2147/IJN.S428044. eCollection 2023.

Abstract

Introduction: Radiotherapy is a widely recognized first-line clinical treatment for cancer, but its efficacy may be impeded by the radioresistance of advanced tumors. It is urgent to improve the sensitivity of radioresistant tumors to radiotherapy. In this work, gadolinium oxide nanocrystals (GONs) were utilized as radiosensitizers to enhance the killing effect and reinforce the immune activation of X-ray irradiation on 4T1 breast cancer cells in vitro and in vivo.

Methods: 1.0 T small animal MR imaging (MRI) system was employed to trace GONs in vivo, while 225 kVp X-ray irradiation equipment was utilized for investigating the radiosensitization of GONs in 4T1 breast cancer cells in vitro and in vivo. Western blot, quantitative real-time PCR (RT-qPCR), immunohistochemistry, immunofluorescence, clonal survival assay, flow cytometry and reactive oxygen species assay were used to explore the biological mechanism of GON sensitization.

Results: GONs exhibited exceptional utility as contrast agents for both in vivo and in vitro MRI imaging. Interestingly, a single dose of 8.0 Gy X-rays together with GONs failed to confer superior therapeutic effects in tumor-bearing mice, while only 3.0 Gy × 3 fractions X-rays combined with GONs exhibited effective tumor growth inhibition. Moreover, fractionated X-ray irradiation with GONs demonstrated a superior capacity to activate the cGAS-STING pathway.

Discussion: Fractionated X-ray irradiation in the presence of GONs has demonstrated the most significant activation of the anti-tumor immune response by boosting the cGAS-STING pathway.

Keywords: 4T1 breast cancer cells; cGAS-STING pathway; gadolinium oxide nanoparticles; magnetic resonance imaging; radiosensitization.

MeSH terms

  • Animals
  • Cell Line, Tumor
  • Dose Fractionation, Radiation
  • Humans
  • Mice
  • Nanoparticles* / chemistry
  • Nucleotidyltransferases
  • Triple Negative Breast Neoplasms* / diagnostic imaging
  • Triple Negative Breast Neoplasms* / radiotherapy

Substances

  • gadolinium oxide
  • Nucleotidyltransferases

Grants and funding

This work was jointly supported by the Key Deployment Project of Chinese Academy of Sciences [Grant No. KFZD-SW-222], the National Natural Science Foundation of China [Grant No. 82272746], the Natural Science Foundation of Gansu Province [Grant No. 21JR7RA106], the West Light Foundation of the Chinese Academy of Sciences [Grant No. xbzg-zdsys-201920] and the Natural Science Foundation of Gansu Province [Grant No. 23JRRA569].