Advanced Search

Submit Manuscript

ADVANCE ONLINE PUBLICATION  |  Open Access

ORIGINAL ARTICLES

Targeting ATAD3A-PINK1-mitophagy axis overcomes chemoimmunotherapy resistance by redirecting PD-L1 to mitochondria

Xiao-Qing Xie1 , Yi Yang1 , Qiang Wang1,2 , Hao-Fei Liu1 , Xuan-Yu Fang1 , Cheng-Long Li1 , Yi-Zhou Jiang3 , Shuai Wang1 , Hong-Yu Zhao4,5 , Jing-Ya Miao1 , Shuai-Shuai Ding1 , Xin-Dong Liu1 , Xiao-Hong Yao1 , Wen-Tao Yang6 , Jun Jiang7 , Zhi-Ming Shao3 , Guoxiang Jin1,* , Xiu-Wu Bian1,*

1Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
2Department of Oncology, Shandong Second Provincial General Hospital, Jinan, Shandong, China
3Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Shanghai, China
4National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
5College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
6Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
7Department of Breast Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
* Correspondence: Guoxiang Jin(gxjinking@hotmail.com)Xiu-Wu Bian(bianxiuwu@263.net)

Only a small proportion of patients with triple-negative breast cancer benefit from immune checkpoint inhibitor (ICI) targeting PD-1/PD-L1 signaling in combination with chemotherapy. Here, we discovered that therapeutic response to ICI plus paclitaxel was associated with subcellular redistribution of PD-L1. In our immunotherapy cohort of ICI in combination with nab-paclitaxel, tumor samples from responders showed significant distribution of PD-L1 at mitochondria, while non-responders showed increased accumulation of PD-L1 on tumor cell membrane instead of mitochondria. Our results also revealed that the distribution pattern of PD-L1 was regulated by an ATAD3A-PINK1 axis. Mechanistically, PINK1 recruited PD-L1 to mitochondria for degradation via a mitophagy pathway. Importantly, paclitaxel increased ATAD3A expression to disrupt proteostasis of PD-L1 by restraining PINK1-dependent mitophagy. Clinically, patients with tumors exhibiting high expression of ATAD3A detected before the treatment with ICI in combination with paclitaxel had markedly shorter progression-free survival compared with those with ATAD3A-low tumors. Preclinical results further demonstrated that targeting ATAD3A reset a favorable antitumor immune microenvironment and increased the efficacy of combination therapy of ICI plus paclitaxel. In summary, our results indicate that ATAD3A serves not only as a resistant factor for the combination therapy of ICI plus paclitaxel through preventing PD-L1 mitochondrial distribution, but also as a promising target for increasing the therapeutic responses to chemoimmunotherapy.

https://doi.org/10.1038/s41422-022-00766-z

FULL TEXT | PDF

Browse 288