1. Academic Validation
  2. Autophagy Inhibition Dysregulates TBK1 Signaling and Promotes Pancreatic Inflammation

Autophagy Inhibition Dysregulates TBK1 Signaling and Promotes Pancreatic Inflammation

  • Cancer Immunol Res. 2016 Jun;4(6):520-30. doi: 10.1158/2326-6066.CIR-15-0235.
Shenghong Yang 1 Yu Imamura 2 Russell W Jenkins 1 Israel Cañadas 1 Shunsuke Kitajima 1 Amir Aref 3 Arthur Brannon 4 Eiji Oki 5 Adam Castoreno 6 Zehua Zhu 1 Tran Thai 1 Jacob Reibel 3 Zhirong Qian 7 Shuji Ogino 7 Kwok K Wong 3 Hideo Baba 8 Alec C Kimmelman 9 Marina Pasca Di Magliano 4 David A Barbie 10
Affiliations

Affiliations

  • 1 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • 2 Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan. Department of Gastroenterological Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan. Department of Surgery and Science, Graduate of Medical Sciences, Kyushu University, Fukuoka, Japan.
  • 3 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • 4 Department of Surgery, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan. Cell and Developmental Biology, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan.
  • 5 Department of Surgery and Science, Graduate of Medical Sciences, Kyushu University, Fukuoka, Japan.
  • 6 Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • 7 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts. Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts.
  • 8 Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
  • 9 Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • 10 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts. [email protected].
Abstract

Autophagy promotes tumor progression downstream of oncogenic KRAS, yet also restrains inflammation and dysplasia through mechanisms that remain incompletely characterized. Understanding the basis of this paradox has important implications for the optimal targeting of Autophagy in Cancer. Using a mouse model of cerulein-induced pancreatitis, we found that loss of Autophagy by deletion of Atg5 enhanced activation of the IκB kinase (IKK)-related kinase TBK1 in vivo, associated with increased neutrophil and T-cell infiltration and PD-L1 upregulation. Consistent with this observation, pharmacologic or genetic inhibition of Autophagy in pancreatic ductal adenocarcinoma cells, including suppression of the Autophagy receptors NDP52 or p62, prolonged TBK1 activation and increased expression of CCL5, IL6, and several other T-cell and neutrophil chemotactic cytokines in vitro Defective Autophagy also promoted PD-L1 upregulation, which is particularly pronounced downstream of IFNγ signaling and involves JAK pathway activation. Treatment with the TBK1/IKKε/JAK Inhibitor CYT387 (also known as momelotinib) not only inhibits Autophagy, but also suppresses this feedback inflammation and reduces PD-L1 expression, limiting KRAS-driven pancreatic dysplasia. These findings could contribute to the dual role of Autophagy in oncogenesis and have important consequences for its therapeutic targeting. Cancer Immunol Res; 4(6); 520-30. ©2016 AACR.

Figures
Products