Abstract

BackgroundIt is important to simultaneously induce strong cell death and antitumor immunity in cancer patients for successful cancer treatment. Here, we investigated the cytotoxic and phenotypic modulation effects of the combination of ANT2 shRNA and human sodium iodide symporter (hNIS) radioiodine gene therapy in vitro and in vivo and visualized the antitumor effects in an immunocompromised mouse colon cancer model.MethodsA mouse colon cancer cell line co-expressing hNIS and the luciferase gene (CT26/hNIS-Fluc, named CT26/NF) was established. CT26/NF cells and tumor-bearing mice were treated with HBSS, scramble, ANT2 shRNA, I-131, and ANT2 shRNA + I-131. The apoptotic rates (%) and MHC class I and Fas gene expression levels were determined in treated CT26/NF cells using flow cytometry. Concurrently, the level of caspase-3 activation was determined in treated cells in vitro. For in vivo therapy, tumor-bearing mice were treated with scramble, ANT2 shRNA, I-131, and the combination therapy, and the anti-tumor effects were monitored using bioluminescence. The killing activity of cytotoxic T cells (CTLs) was measured with a lactate dehydrogenase (LDH) assay.ResultsFor the in vitro experiments, the combination of ANT2 shRNA and I-131 resulted in a higher apoptotic cell death rate compared with ANT2 shRNA or I-131 alone, and the levels of MHC class I and Fas-expressing cancer cells were highest in the cells receiving combination treatment, while single treatment modestly increased the level of MHC class I and Fas gene expression. The combination of ANT2 shRNA and I-131 resulted in a higher caspase-3 activation than single treatments. Interestingly, in vivo combination treatment led to increased gene expression of MHC class I and Fas than the respective mono-therapies; furthermore, bioluminescence showed increased antitumor effects after combination treatment than monotherapies. The LDH assay revealed that the CTL killing activity against CT26/NF cells was most effective after combination therapy.ConclusionsIncreased cell death and phenotypic modulation of cancer cells in vitro and in vivo were achieved simultaneously after combination therapy with ANT2 shRNA and I-131, and this combination therapy induced remarkable antitumor outcomes through improvements in CTL immunity against CT26/NF. Our results suggest that combination therapy can be used as a new therapeutic strategy for cancer patients who show resistance to single therapy such as radiation or immunotherapy.

Highlights

  • It is important to simultaneously induce strong cell death and antitumor immunity in cancer patients for successful cancer treatment

  • Our results suggest that combination therapy can be used as a new therapeutic strategy for cancer patients who show resistance to single therapy such as radiation or immunotherapy

  • Similar to Adenine nucleotide translocase-2 (ANT2) Short hairpin RNA (shRNA) treatment, we showed that human sodium iodide symporter (hNIS) radioiodine gene therapy modulates the phenotype of cancer cells in vitro and in vivo, resulting in an increased susceptibility of cancer cells to cytotoxic T cells (CTLs) [14]

Read more

Summary

Introduction

It is important to simultaneously induce strong cell death and antitumor immunity in cancer patients for successful cancer treatment. We investigated the cytotoxic and phenotypic modulation effects of the combination of ANT2 shRNA and human sodium iodide symporter (hNIS) radioiodine gene therapy in vitro and in vivo and visualized the antitumor effects in an immunocompromised mouse colon cancer model. To achieve successful cancer treatment, it is important to overcome obstacles that occur when cancer patients receive single treatment such as chemotherapy, radiation therapy and immunotherapy. Due to the limitations of single therapy, new combined therapies that can simultaneously induce strong cytotoxic effects and enhance anti-tumor immunity should be explored. ANT2 inhibition with RNAi induces a phenotypic modulation of cancer cells such as alterations in Fas, MHC class I, and ICAM-I expression levels, and sequentially, these modulations enhance anti-tumor immunity when combined with hMUC1 DNA vaccination

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