Abstract

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.

Highlights

  • The cytokine interferon-α (IFNα) has been used for a long time for treatment of many types of cancers, such as renal cell carcinoma, malignant melanoma or chronic myeloid leukemia [1, 2]

  • We showed for the first time that PD-L1 molecule was expressed and could be up-regulated in the majority of specific immune cell populations by IFNα

  • Taking into account the ubiquitous expression of PD-L1 receptor (PD-1) on non-myeloid specific immune cells (CD8, CD4 and Treg), our results indicate that immunotherapy with IFNα could lead to an undesirable side effect of general immunosuppression and to increased tumor immune evasion or chronification of infection

Read more

Summary

Introduction

The cytokine interferon-α (IFNα) has been used for a long time for treatment of many types of cancers, such as renal cell carcinoma, malignant melanoma or chronic myeloid leukemia [1, 2]. Utilized as an anti-viral agent and meant for use in tumors where a viral origin was suspected, IFNα initiated a variety of biological activities that warranted further investigation. This cytokine manifests direct suppressive effects on tumor cell growth in vitro and in vivo [4] and enhances tumor recognition by the increase in MHC-1 expression. Essers and colleagues have showed that the cytokine activated dormant hematopoietic stem cells in vivo [7] We have confirmed this phenomenon in pancreatic cancer, where we found that IFNα exhibited the ability to activate stem cell markers [8]. IFNα2b (trade name Intron-A), a well-known IFN-based therapeutic [13] that is approved for the treatment of various infectious diseases as well as for many types of cancer including leukemia, lymphoma, multiple myeloma and malignant melanoma, is actively used in multiple clinical trials (http://www.druglib.com/ druginfo/intron-a/trials/)

Objectives
Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.