In vivo molecular imaging and radionuclide (131I) therapy of human nasopharyngeal carcinoma cells transfected with a lentivirus expressing sodium iodide symporter

PLoS One. 2015 Jan 26;10(1):e0116531. doi: 10.1371/journal.pone.0116531. eCollection 2015.

Abstract

Introduction: Despite recent improvements in the survival rates for nasopharyngeal carcinoma (NPC), novel treatment strategies are required to improve distant metastasis-free survival. The sodium iodine symporter (NIS) gene has been applied for in vivo imaging and cancer therapy. In this study, we examined the potential of NIS gene therapy as a therapeutic approach in NPC by performing non-invasive imaging using 125I and 131I therapy in vivo.

Methods: We constructed a lentiviral vector expressing NIS and enhanced green fluorescent protein (EGFP) under the control of the human elongation factor-1α (EF1α) promoter, and stably transfected the vector into CNE-2Z NPC cells to create CNE-2Z-NIS cells. CNE-2Z and CNE-2Z-NIS tumor xenografts were established in nude mice; 125I uptake, accumulation and efflux were measured using micro-SPECT/CT imaging; the therapeutic effects of treatment with 131I were assessed over 25 days by measuring tumor volume and immunohistochemical staining of the excised tumors.

Results: qPCR, immunofluorescence and Western blotting confirmed that CNE-2Z-NIS cells expressed high levels of NIS mRNA and protein. CNE-2Z-NIS cells and xenografts took up and accumulated significantly more 125I than CNE-2Z cells and xenografts. In vitro, 131I significantly reduced the clonogenic survival of CNE-2Z-NIS cells. In vivo, 131I effectively inhibited the growth of CNE-2Z-NIS xenografts. At the end of 131I therapy, CNE-2Z-NIS xenograft tumor cells expressed higher levels of NIS and caspase-3 and lower levels of Ki-67.

Conclusion: Lentiviruses effectively delivered and mediated long-lasting expression of NIS in CNE-2Z cells which enabled uptake and accumulation of radioisotopes and provided a significant therapeutic effect in an in vivo model of NPC. NIS-mediated radioiodine treatment merits further investigation as a potentially effective, low toxicity therapeutic strategy for NPC.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Carcinoma
  • Cell Line, Tumor
  • Genetic Therapy
  • Humans
  • Iodine Radioisotopes / pharmacology*
  • Iodine Radioisotopes / therapeutic use
  • Lentivirus / genetics
  • Male
  • Mice, Inbred BALB C
  • Mice, Nude
  • Nasopharyngeal Carcinoma
  • Nasopharyngeal Neoplasms / therapy*
  • Radioisotopes / pharmacology*
  • Radioisotopes / therapeutic use
  • Symporters / biosynthesis
  • Symporters / genetics*
  • Tissue Distribution
  • Transfection
  • Xenograft Model Antitumor Assays

Substances

  • Iodine Radioisotopes
  • Radioisotopes
  • Symporters
  • sodium-iodide symporter

Grants and funding

This work was supported by grants from the National Natural Science Foundation of China (No. 81071181) (http://isisn.nsfc.gov.cn/egrantweb/), Shanghai Outstanding Academic Leaders Project (11XD1403700) (http://www.stcsm.gov.cn/). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.