Articles

< Previous         Next >  
RIG-I immunotherapy overcomes radioresistance in p53-positive malignant melanoma 
Silke Lambing1 , Yu Pan Tan1 , Paraskevi Vasileiadou1 , Stefan Holdenrieder1,2 , Patrick Müller1 , Christian Hagen1 , Stephan Garbe3 , Rayk Behrendt1 , Martin Schlee1 , Jasper G. van den Boorn1 , Eva Bartok1,4,5,†,* , Marcel Renn1,6,† , Gunther Hartmann1,†,*
1Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn 53127, Germany
2Institute of Laboratory Medicine, German Heart Centre, Munich 80636, Germany
3Department of Radiation Oncology, University Hospital Bonn, Bonn 53127, Germany
4Unit of Experimental Immunology, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp 2000, Belgium
5Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, Bonn 53127, Germany
6Mildred Scheel School of Oncology, Bonn, University Hospital Bonn, Medical Faculty, Bonn 53127, Germany
These authors contributed equally to this work.
*Correspondence to:Eva Bartok , Email:ebartok@uni-bonn.de Gunther Hartmann , Email:gunther.hartmann@uni-bonn.de
J Mol Cell Biol, Volume 15, Issue 1, January 2023, mjad001,  https://doi.org/10.1093/jmcb/mjad001
Keyword: RIG-I, p53, melanoma, immunotherapy, irradiation, radiotherapy, radioresistance

Radiotherapy induces DNA damage, resulting in cell cycle arrest and activation of cell-intrinsic death pathways. However, the radioresistance of some tumour entities such as malignant melanoma limits its clinical application. The innate immune sensing receptor retinoic acid-inducible gene I (RIG-I) is ubiquitously expressed and upon activation triggers an immunogenic form of cell death in a variety of tumour cell types including melanoma. To date, the potential of RIG-I ligands to overcome radioresistance of tumour cells has not been investigated. Here, we demonstrate that RIG-I activation enhanced the extent and immunogenicity of irradiation-induced tumour cell death in human and murine melanoma cells in vitro and improved survival in the murine B16 melanoma model in vivo. Transcriptome analysis pointed to a central role for p53, which was confirmed using p53–/– B16 cells. In vivo, the additional effect of RIG-I in combination with irradiation on tumour growth was absent in mice carrying p53–/– B16 tumours, while the antitumoural response to RIG-I stimulation alone was maintained. Our results identify p53 as a pivotal checkpoint that is triggered by RIG-I resulting in enhanced irradiation-induced tumour cell death. Thus, the combined administration of RIG-I ligands and radiotherapy is a promising approach to treating radioresistant tumours with a functional p53 pathway, such as melanoma.