Cargando…

In vivo Molecular Imaging and Radionuclide ((131)I) Therapy of Human Nasopharyngeal Carcinoma Cells Transfected with a Lentivirus Expressing Sodium Iodide Symporter

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 e...

Descripción completa

Detalles Bibliográficos
Autores principales: Shi, Shuo, Zhang, Min, Guo, Rui, Miao, Ying, Hu, Jiajia, Xi, Yun, Li, Biao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4306548/
https://www.ncbi.nlm.nih.gov/pubmed/25621996
http://dx.doi.org/10.1371/journal.pone.0116531
Descripción
Sumario: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 (125)I and (131)I 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; (125)I uptake, accumulation and efflux were measured using micro-SPECT/CT imaging; the therapeutic effects of treatment with (131)I 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 (125)I than CNE-2Z cells and xenografts. In vitro, (131)I significantly reduced the clonogenic survival of CNE-2Z-NIS cells. In vivo, (131)I effectively inhibited the growth of CNE-2Z-NIS xenografts. At the end of (131)I 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.