[HTML][HTML] Interferon-α up-regulates the expression of PD-L1 molecules on immune cells through STAT3 and p38 signaling

AV Bazhin, K Von Ahn, J Fritz, J Werner… - Frontiers in …, 2018 - frontiersin.org
AV Bazhin, K Von Ahn, J Fritz, J Werner, S Karakhanova
Frontiers in immunology, 2018frontiersin.org
Interferon-α (IFNα) has one of the longest histories of use amongst cytokines in clinical
oncology and has been applied for the treatment of many types of cancers. Due to its
immune-activating properties, IFNα is also an attractive candidate for combinatory anti-
cancer therapies. Despite its extensive use in animal tumor models as well as in several
clinical trials, the different mechanisms underlying patient responses and affecting desirable
clinical benefits are still under investigation. Here we show that in addition to its immune …
Interferon-α (IFNα) has one of the longest histories of use amongst cytokines in clinical oncology and has been applied for the treatment of many types of cancers. Due to its immune-activating properties, IFNα is also an attractive candidate for combinatory anti-cancer therapies. Despite its extensive use in animal tumor models as well as in several clinical trials, the different mechanisms underlying patient responses and affecting desirable clinical benefits are still under investigation. Here we show that in addition to its immune-activating properties, IFNα induces the expression of a key negative regulator, immunosuppressive PD-L1 molecule, in the majority of the specific immune cell populations, particularly in the dendritic cells (DC). DC can modulate immune responses by a variety of mechanisms, including expression of T-cell regulatory molecules and cytokines. Our results showed that treatment of DC with IFNα-2b led to pronounced up-regulation of surface expression of PD-L1 molecules, increased IL-6 and decreased IL-12 production. Moreover, we present evidence that IFNα-treated DC exhibited a reduced capacity to stimulate interferon-γ production in T cells compared to control DC. This T-cell response after treatment of DC with IFNα was recovered by a pre-treatment with an anti-PD-L1 blocking antibody. Further analyses revealed that IFNα regulated PD-L1 expression through the STAT3 and p38 signaling pathways, since blocking of STAT3 and p38 activation with specific inhibitors prevented PD-L1 up-regulation. Our findings underline the important roles of p38 and STAT3 in the regulation of PD-L1 expression and prove that IFNα induces STAT3/p38-mediated expression of PD-L1 and thereby a reduced stimulatory ability of DC. The augmentation of PD-L1 expression in immune cells through IFNα treatment should be considered by use of IFNα in an anti-cancer therapy.
Frontiers