Oncotarget

Research Papers:

Dichloroacetate blocks aerobic glycolytic adaptation to attenuated measles virus and promotes viral replication leading to enhanced oncolysis in glioblastoma

Chunyan Li, Gang Meng, Lei Su, Aiping Chen, Mao Xia, Chun Xu, Decai Yu, Aiqin Jiang and Jiwu Wei _

PDF  |  HTML  |  How to cite

Oncotarget. 2015; 6:1544-1555. https://doi.org/10.18632/oncotarget.2838

Metrics: PDF 2885 views  |   HTML 2710 views  |   ?  


Abstract

Chunyan Li1,4,*, Gang Meng1,2,*, Lei Su3, Aiping Chen1, Mao Xia1, Chun Xu1, Decai Yu3, Aiqin Jiang1 and Jiwu Wei1,2

1 Jiangsu Key Laboratory of Molecular Medicine, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, China

2 Nanjing University Hightech Institute at Suzhou, Suzhou, China

3 Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China

4 Zhongda Hospital, Medical School of Southeast University, Nanjing, China

* These authors contributed equally to this work

Correspondence:

Jiwu Wei, email:

Aiqin Jiang, email:

Keywords: measles virus, glycolysis, dichloroacetate, glioma, oncolysis

Received: August 21, 2014 Accepted: December 01, 2014 Published: December 02, 2014

Abstract

Targeting reprogrammed energy metabolism such as aerobic glycolysis is a potential strategy for cancer treatment. However, tumors exhibiting low-rate glycolysis or metabolic heterogeneity might be resistant to such treatment. We hypothesized that a therapeutic modality that drove cancer cells to high-rate glycolysis might sensitize cancer cells to interference directed against metabolic flux. In this study, we found that attenuated oncolytic measles virus Edmonston strain (MV-Edm) caused glioblastoma cells to shift to high-rate aerobic glycolysis; this adaptation was blocked by dichloroacetate (DCA), an inhibitor of glycolysis, leading to profound cell death of cancer cells but not of normal cells. DCA enhanced viral replication by mitigating mitochondrial antiviral signaling protein (MAVS)-mediated innate immune responses. In a subcutaneous glioblastoma (GBM) xenograft mouse model, low-dose MV-Edm and DCA significantly inhibited tumor growth in vivo. We found that DCA impaired glycolysis (blocking bioenergetic generation) and enhanced viral replication (increasing bioenergetic consumption), which, in combination, accelerated bioenergetic exhaustion leading to necrotic cell death. Taken together, oncolytic MV-Edm sensitized cancer cells to DCA, and in parallel, DCA promoted viral replication, thus, improving oncolysis. This novel therapeutic approach should be readily incorporated into clinical trials.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 2838