Abstract

BackgroundBoth tumor cells and their supporting endothelial cells should be considered for targeted cell killing when designing cancer treatments. Here we investigated the feasibility of combining radioiodide and antiangiogenic therapies after baculovirus-mediated transfer of genes encoding the sodium iodide symporter (NIS) and plasminogen kringle 5 (K5).MethodsA recombinant baculovirus containing the NIS gene under control of the human telomerase reverse transcriptase (hTERT) promoter and the K5 gene driven by the early growth response 1 (Egr1) promoter was developed. Dual-luciferase reporter assay was performed to confirm the activation of hTERT transcription. NIS and K5 gene expression were identified by Western blot and Real-Time PCR. Functional NIS activity in baculovirus-infected Hela cells was confirmed by the uptake of 125I and cytotoxicity of 131I. The apoptotic effect of 131I-induced K5 on baculovirus-infected human umbilical vein endothelial cells (HUVECs) was analyzed by a flow cytometry-based assay. In vivo, NIS reporter gene imaging and therapeutic experiments with 131I were performed. Finally, the microvessel density (MVD) in tumors after treatment was determined by CD31 immunostaining.ResultsThe activation of hTERT transcription was specifically up-regulated in tumor cells. NIS gene expression markedly increased in baculovirus-infected HeLa cells, but not in MRC5 cells. The Hela cells showed a significant increase of 125I uptake, which was inhibited by NaClO4, and a notably decreased cell survival rate by 131I treatment. Expression of the K5 gene induced by 131I was elevated in a dose- and time-dependent manner and resulted in the apoptosis of HUVECs. Furthermore, 131I SPECT imaging clearly showed cervical tumor xenografts infected with recombinant baculovirus. Following therapy, tumor growth was significantly retarded. CD31 immunostaining confirmed a significant decrease of MVD.ConclusionThe recombinant baculovirus supports a promising strategy of NIS-based raidoiodide therapy combined with K5-based antiangiogenic therapy by targeting both the tumor and its supporting vessels.

Highlights

  • The cloning of the sodium iodide symporter (NIS) gene and subsequent studies of its properties have led to a new approach of targeted radioiodide therapy for malignant cancers

  • Expression of NIS gene in baculovirus-infected hela cells The NIS gene in the baculovirus dual expression system was designed to be driven by the Human telomerase reverse transcriptase (hTERT) promoter for tumor-specific gene expression

  • A band with a molecular mass of approximately 97 kDa recognized by the anti-NIS antibody was detected in lysates derived from Human cervical cancer cells (Hela) cells infected with Bac-hTERT-NIS-early growth response 1 (Egr1)-kringle 5 (K5) or BacCMV-NIS-Egr1-K5

Read more

Summary

Introduction

The cloning of the sodium iodide symporter (NIS) gene and subsequent studies of its properties have led to a new approach of targeted radioiodide therapy for malignant cancers. The transcriptional activity of the hTERT gene promoter has been observed exclusively in telomerase-positive cells [5]. These observations have highlighted the potential of targeted transfer of genes for expression under the hTERT promoter [6] Both tumor cells and their supporting endothelial cells should be considered for targeted cell killing when designing cancer treatments. We investigated the feasibility of combining radioiodide and antiangiogenic therapies after baculovirus-mediated transfer of genes encoding the sodium iodide symporter (NIS) and plasminogen kringle 5 (K5)

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call