[PDF][PDF] Energy status dictates PD-L1 protein abundance and anti-tumor immunity to enable checkpoint blockade

X Dai, X Bu, Y Gao, J Guo, J Hu, C Jiang, Z Zhang… - Molecular cell, 2021 - cell.com
X Dai, X Bu, Y Gao, J Guo, J Hu, C Jiang, Z Zhang, K Xu, J Duan, S He, J Zhang, L Wan
Molecular cell, 2021cell.com
Aberrant energy status contributes to multiple metabolic diseases, including obesity,
diabetes, and cancer, but the underlying mechanism remains elusive. Here, we report that
ketogenic-diet-induced changes in energy status enhance the efficacy of anti-CTLA-4
immunotherapy by decreasing PD-L1 protein levels and increasing expression of type-I
interferon (IFN) and antigen presentation genes. Mechanistically, energy deprivation
activates AMP-activated protein kinase (AMPK), which in turn, phosphorylates PD-L1 on …
Summary
Aberrant energy status contributes to multiple metabolic diseases, including obesity, diabetes, and cancer, but the underlying mechanism remains elusive. Here, we report that ketogenic-diet-induced changes in energy status enhance the efficacy of anti-CTLA-4 immunotherapy by decreasing PD-L1 protein levels and increasing expression of type-I interferon (IFN) and antigen presentation genes. Mechanistically, energy deprivation activates AMP-activated protein kinase (AMPK), which in turn, phosphorylates PD-L1 on Ser283, thereby disrupting its interaction with CMTM4 and subsequently triggering PD-L1 degradation. In addition, AMPK phosphorylates EZH2, which disrupts PRC2 function, leading to enhanced IFNs and antigen presentation gene expression. Through these mechanisms, AMPK agonists or ketogenic diets enhance the efficacy of anti-CTLA-4 immunotherapy and improve the overall survival rate in syngeneic mouse tumor models. Our findings reveal a pivotal role for AMPK in regulating the immune response to immune-checkpoint blockade and advocate for combining ketogenic diets or AMPK agonists with anti-CTLA4 immunotherapy to combat cancer.
cell.com