Targeting ALK averts ribonuclease 1-induced immunosuppression and enhances antitumor immunity in hepatocellular carcinoma

Abstract Tumor-secreted factors contribute to the development of a microenvironment that facilitates the escape of cancer cells from immunotherapy. In this study, we conduct a retrospective comparison of the proteins secreted by hepatocellular carcinoma (HCC) cells in responders and non-responders a...

Full description

Bibliographic Details
Main Authors: Chunxiao Liu, Chenhao Zhou, Weiya Xia, Yifan Zhou, Yufan Qiu, Jialei Weng, Qiang Zhou, Wanyong Chen, Ying-Nai Wang, Heng-Huan Lee, Shao-Chun Wang, Ming Kuang, Dihua Yu, Ning Ren, Mien-Chie Hung
Format: Article
Language:English
Published: Nature Portfolio 2024-02-01
Series:Nature Communications
Online Access:https://doi.org/10.1038/s41467-024-45215-0
_version_ 1797274001335123968
author Chunxiao Liu
Chenhao Zhou
Weiya Xia
Yifan Zhou
Yufan Qiu
Jialei Weng
Qiang Zhou
Wanyong Chen
Ying-Nai Wang
Heng-Huan Lee
Shao-Chun Wang
Ming Kuang
Dihua Yu
Ning Ren
Mien-Chie Hung
author_facet Chunxiao Liu
Chenhao Zhou
Weiya Xia
Yifan Zhou
Yufan Qiu
Jialei Weng
Qiang Zhou
Wanyong Chen
Ying-Nai Wang
Heng-Huan Lee
Shao-Chun Wang
Ming Kuang
Dihua Yu
Ning Ren
Mien-Chie Hung
author_sort Chunxiao Liu
collection DOAJ
description Abstract Tumor-secreted factors contribute to the development of a microenvironment that facilitates the escape of cancer cells from immunotherapy. In this study, we conduct a retrospective comparison of the proteins secreted by hepatocellular carcinoma (HCC) cells in responders and non-responders among a cohort of ten patients who received Nivolumab (anti-PD-1 antibody). Our findings indicate that non-responders have a high abundance of secreted RNase1, which is associated with a poor prognosis in various cancer types. Furthermore, mice implanted with HCC cells that overexpress RNase1 exhibit immunosuppressive tumor microenvironments and diminished response to anti-PD-1 therapy. RNase1 induces the polarization of macrophages towards a tumor growth-promoting phenotype through activation of the anaplastic lymphoma kinase (ALK) signaling pathway. Targeting the RNase1/ALK axis reprograms the macrophage polarization, with increased CD8+ T- and Th1- cell recruitment. Moreover, simultaneous targeting of the checkpoint protein PD-1 unleashes cytotoxic CD8+ T-cell responses. Treatment utilizing both an ALK inhibitor and an anti-PD-1 antibody exhibits enhanced tumor regression and facilitates long-term immunity. Our study elucidates the role of RNase1 in mediating tumor resistance to immunotherapy and reveals an RNase1-mediated immunosuppressive tumor microenvironment, highlighting the potential of targeting RNase1 as a promising strategy for cancer immunotherapy in HCC.
first_indexed 2024-03-07T14:52:09Z
format Article
id doaj.art-fec4a1384c9b4097a6c58e39022e975c
institution Directory Open Access Journal
issn 2041-1723
language English
last_indexed 2024-03-07T14:52:09Z
publishDate 2024-02-01
publisher Nature Portfolio
record_format Article
series Nature Communications
spelling doaj.art-fec4a1384c9b4097a6c58e39022e975c2024-03-05T19:39:45ZengNature PortfolioNature Communications2041-17232024-02-0115111810.1038/s41467-024-45215-0Targeting ALK averts ribonuclease 1-induced immunosuppression and enhances antitumor immunity in hepatocellular carcinomaChunxiao Liu0Chenhao Zhou1Weiya Xia2Yifan Zhou3Yufan Qiu4Jialei Weng5Qiang Zhou6Wanyong Chen7Ying-Nai Wang8Heng-Huan Lee9Shao-Chun Wang10Ming Kuang11Dihua Yu12Ning Ren13Mien-Chie Hung14Department of Liver Surgery, Center of Hepato-Pancreato-biliary Surgery, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen UniversityDepartment of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterDepartment of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterDepartment of laboratory medicine, Zhujiang Hospital of Southern Medical UniversityDepartment of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterDepartment of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan UniversityDepartment of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan UniversityDepartment of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan UniversityDepartment of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterDepartment of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterGraduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical UniversityDepartment of Liver Surgery, Center of Hepato-Pancreato-biliary Surgery, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen UniversityDepartment of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterDepartment of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan UniversityDepartment of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterAbstract Tumor-secreted factors contribute to the development of a microenvironment that facilitates the escape of cancer cells from immunotherapy. In this study, we conduct a retrospective comparison of the proteins secreted by hepatocellular carcinoma (HCC) cells in responders and non-responders among a cohort of ten patients who received Nivolumab (anti-PD-1 antibody). Our findings indicate that non-responders have a high abundance of secreted RNase1, which is associated with a poor prognosis in various cancer types. Furthermore, mice implanted with HCC cells that overexpress RNase1 exhibit immunosuppressive tumor microenvironments and diminished response to anti-PD-1 therapy. RNase1 induces the polarization of macrophages towards a tumor growth-promoting phenotype through activation of the anaplastic lymphoma kinase (ALK) signaling pathway. Targeting the RNase1/ALK axis reprograms the macrophage polarization, with increased CD8+ T- and Th1- cell recruitment. Moreover, simultaneous targeting of the checkpoint protein PD-1 unleashes cytotoxic CD8+ T-cell responses. Treatment utilizing both an ALK inhibitor and an anti-PD-1 antibody exhibits enhanced tumor regression and facilitates long-term immunity. Our study elucidates the role of RNase1 in mediating tumor resistance to immunotherapy and reveals an RNase1-mediated immunosuppressive tumor microenvironment, highlighting the potential of targeting RNase1 as a promising strategy for cancer immunotherapy in HCC.https://doi.org/10.1038/s41467-024-45215-0
spellingShingle Chunxiao Liu
Chenhao Zhou
Weiya Xia
Yifan Zhou
Yufan Qiu
Jialei Weng
Qiang Zhou
Wanyong Chen
Ying-Nai Wang
Heng-Huan Lee
Shao-Chun Wang
Ming Kuang
Dihua Yu
Ning Ren
Mien-Chie Hung
Targeting ALK averts ribonuclease 1-induced immunosuppression and enhances antitumor immunity in hepatocellular carcinoma
Nature Communications
title Targeting ALK averts ribonuclease 1-induced immunosuppression and enhances antitumor immunity in hepatocellular carcinoma
title_full Targeting ALK averts ribonuclease 1-induced immunosuppression and enhances antitumor immunity in hepatocellular carcinoma
title_fullStr Targeting ALK averts ribonuclease 1-induced immunosuppression and enhances antitumor immunity in hepatocellular carcinoma
title_full_unstemmed Targeting ALK averts ribonuclease 1-induced immunosuppression and enhances antitumor immunity in hepatocellular carcinoma
title_short Targeting ALK averts ribonuclease 1-induced immunosuppression and enhances antitumor immunity in hepatocellular carcinoma
title_sort targeting alk averts ribonuclease 1 induced immunosuppression and enhances antitumor immunity in hepatocellular carcinoma
url https://doi.org/10.1038/s41467-024-45215-0
work_keys_str_mv AT chunxiaoliu targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT chenhaozhou targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT weiyaxia targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT yifanzhou targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT yufanqiu targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT jialeiweng targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT qiangzhou targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT wanyongchen targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT yingnaiwang targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT henghuanlee targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT shaochunwang targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT mingkuang targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT dihuayu targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT ningren targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma
AT mienchiehung targetingalkavertsribonuclease1inducedimmunosuppressionandenhancesantitumorimmunityinhepatocellularcarcinoma