↓ Skip to main content

Recombinant oncolytic Newcastle disease virus displays antitumor activities in anaplastic thyroid cancer cells

Overview of attention for article published in BMC Cancer, July 2018
Altmetric Badge

Readers on

mendeley
34 Mendeley
You are seeing a free-to-access but limited selection of the activity Altmetric has collected about this research output. Click here to find out more.
Title
Recombinant oncolytic Newcastle disease virus displays antitumor activities in anaplastic thyroid cancer cells
Published in
BMC Cancer, July 2018
DOI 10.1186/s12885-018-4522-3
Pubmed ID
Authors

Ke Jiang, Cuiping Song, Lingkai Kong, Lulu Hu, Guibin Lin, Tian Ye, Gang Yao, Yupeng Wang, Haibo Chen, Wei Cheng, Martin P. Barr, Quentin Liu, Guirong Zhang, Chan Ding, Songshu Meng

Abstract

Anaplastic thyroid cancer (ATC) is one of the most aggressive of all solid tumors for which no effective therapies are currently available. Oncolytic Newcastle disease virus (NDV) has shown the potential to induce oncolytic cell death in a variety of cancer cells of diverse origins. However, whether oncolytic NDV displays antitumor effects in ATC remains to be investigated. We have previously shown that the oncolytic NDV strain FMW (NDV/FMW) induces oncolytic cell death in several cancer types. In the present study, we investigated the oncolytic effects of NDV/FMW in ATC. In this study, a recombinant NDV expressing green fluorescent protein (GFP) was generated using an NDV reverse genetics system. The resulting virus was named after rFMW/GFP and the GFP expression in infected cells was demonstrated by direct fluorescence and immunoblotting. Viral replication was evaluated by end-point dilution assay in DF-1 cell lines. Oncolytic effects were examined by biochemical and morphological experiments in cultural ATC cells and in mouse models. rFMW/GFP replicated robustly in ATC cells as did its parent virus (NDV/FMW) while the expression of GFP protein was detected in lungs and spleen of mice intravenously injected with rFMW/GFP. We further showed that rFMW/GFP infection substantially increased early and late apoptosis in the ATC cell lines, THJ-16 T and THJ-29 T and increased caspase-3 processing and Poly (ADP-ribose) polymerase (PARP) cleavage in ATC cells as assessed by immunoblotting. In addition, rFMW/GFP induced lyses of spheroids derived from ATC cells in three-dimensional (3D) cultures. We further demonstrated that rFMW/GFP infection resulted in the activation of p38 MAPK signaling, but not Erk1/2 or JNK, in THJ-16 T and THJ-29 T cells. Notably, inhibition of p38 MAPK activity by SB203580 decreased rFMW/GFP-induced cleavage of caspase-3 and PARP in THJ-16 T and THJ-29 T cells. Finally, both rFMW/GFP and its parent virus inhibited tumor growth in mice bearing THJ-16 T derived tumors. Taken together, these data indicate that both the recombinant reporter virus rFMW/GFP and its parent virus NDV/FMW, display oncolytic activities in ATC cells in vitro and in vivo and suggest that oncolytic NDV may have potential as a novel therapeutic strategy for ATC.

Timeline

Login to access the full chart related to this output.

If you don’t have an account, click here to discover Explorer

Mendeley readers

Mendeley readers

The data shown below were compiled from readership statistics for 34 Mendeley readers of this research output. Click here to see the associated Mendeley record.

Geographical breakdown

Country Count As %
Unknown 34 100%

Demographic breakdown

Readers by professional status Count As %
Student > Ph. D. Student 7 21%
Researcher 6 18%
Student > Bachelor 5 15%
Student > Master 4 12%
Unspecified 1 3%
Other 2 6%
Unknown 9 26%
Readers by discipline Count As %
Biochemistry, Genetics and Molecular Biology 9 26%
Medicine and Dentistry 5 15%
Agricultural and Biological Sciences 3 9%
Veterinary Science and Veterinary Medicine 2 6%
Unspecified 1 3%
Other 4 12%
Unknown 10 29%