Inhibition of TCA cycle improves the anti-PD-1 immunotherapy efficacy in melanoma cells via ATF3-mediated PD-L1 expression and glycolysis

Background anti-Programmed Death-1 (anti-PD-1) immunotherapy has shown promising manifestation in improving the survival rate of patients with advanced melanoma, with its efficacy closely linked to Programmed cell death-Ligand 1 (PD-L1) expression. However, low clinical efficacy and drug resistance...

Full description

Bibliographic Details
Main Authors: Jing Chen, Jie Wu, Xiang Chen, Qian Tao, Nian Liu, Mingjie Yan, Cong Peng
Format: Article
Language:English
Published: BMJ Publishing Group 2023-09-01
Series:Journal for ImmunoTherapy of Cancer
Online Access:https://jitc.bmj.com/content/11/9/e007146.full
_version_ 1797669495770185728
author Jing Chen
Jie Wu
Xiang Chen
Qian Tao
Nian Liu
Mingjie Yan
Cong Peng
author_facet Jing Chen
Jie Wu
Xiang Chen
Qian Tao
Nian Liu
Mingjie Yan
Cong Peng
author_sort Jing Chen
collection DOAJ
description Background anti-Programmed Death-1 (anti-PD-1) immunotherapy has shown promising manifestation in improving the survival rate of patients with advanced melanoma, with its efficacy closely linked to Programmed cell death-Ligand 1 (PD-L1) expression. However, low clinical efficacy and drug resistance remain major challenges. Although the metabolic alterations from tricarboxylic acid (TCA) cycle to glycolysis is a hallmark in cancer cells, accumulating evidence demonstrating TCA cycle plays critical roles in both tumorigenesis and treatment.Methods The plasma levels of metabolites in patients with melanoma were measured by nuclear magnetic resonance (NMR) spectroscopy. The effect of pyruvate dehydrogenase subunit 1 (PDHA1) and oxoglutarate dehydrogenase (OGDH) on immunotherapy was performed by B16F10 tumor-bearing mice. Flow cytometry analyzed the immune microenvironment. RNA sequencing analyzed the global transcriptome alterations in CPI613-treated melanoma cells. The regulation of PD-L1 and glycolysis by PDHA1/OGDH-ATF3 signaling were confirmed by Quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, dual-luciferase reporter gene, Chromatin immunoprecipitation (ChIP)-quantitative PCR and Seahorse assay. The relationship between PDHA1/OGDH-ATF3-glycolysis and the efficacy of melanoma anti-PD-1 immunotherapy was verified in the clinical database and single-cell RNA-seq (ScRNA-Seq).Results In our study, the results showed that significant alterations in metabolites associated with glycolysis and the TCA cycle in plasma of patients with melanoma through NMR technique, and then, PDHA1 and OGDH, key enzymes for regulation TCA cycle, were remarkable raised in melanoma and negatively related to anti-PD-1 efficacy through clinical database analysis as well as ScRNA-Seq. Inhibition of PDHA1 and OGDH by either shRNA or pharmacological inhibitor by CPI613 dramatically attenuated melanoma progression as well as improved the therapeutic efficacy of anti-PD-1 against melanoma. Most importantly, suppression of TCA cycle remarkably raises PD-L1 expression and glycolysis flux through AMPK-CREB-ATF3 signaling.Conclusions Taken together, our results demonstrated the role of TCA cycle in immune checkpoint blockade and provided a novel combination strategy for anti-PD-1 immunotherapy in melanoma treatment.
first_indexed 2024-03-11T20:45:13Z
format Article
id doaj.art-430016a38d034d219fd1789fd04ff458
institution Directory Open Access Journal
issn 2051-1426
language English
last_indexed 2024-03-11T20:45:13Z
publishDate 2023-09-01
publisher BMJ Publishing Group
record_format Article
series Journal for ImmunoTherapy of Cancer
spelling doaj.art-430016a38d034d219fd1789fd04ff4582023-10-01T16:20:07ZengBMJ Publishing GroupJournal for ImmunoTherapy of Cancer2051-14262023-09-0111910.1136/jitc-2023-007146Inhibition of TCA cycle improves the anti-PD-1 immunotherapy efficacy in melanoma cells via ATF3-mediated PD-L1 expression and glycolysisJing Chen0Jie Wu1Xiang Chen2Qian Tao3Nian Liu4Mingjie Yan5Cong Peng6National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, ChinaDepartment of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, ChinaDepartment of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, ChinaNational Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, ChinaDepartment of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, ChinaFurong Laboratory, Central South University, Changsha, Hunan, ChinaDepartment of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, ChinaBackground anti-Programmed Death-1 (anti-PD-1) immunotherapy has shown promising manifestation in improving the survival rate of patients with advanced melanoma, with its efficacy closely linked to Programmed cell death-Ligand 1 (PD-L1) expression. However, low clinical efficacy and drug resistance remain major challenges. Although the metabolic alterations from tricarboxylic acid (TCA) cycle to glycolysis is a hallmark in cancer cells, accumulating evidence demonstrating TCA cycle plays critical roles in both tumorigenesis and treatment.Methods The plasma levels of metabolites in patients with melanoma were measured by nuclear magnetic resonance (NMR) spectroscopy. The effect of pyruvate dehydrogenase subunit 1 (PDHA1) and oxoglutarate dehydrogenase (OGDH) on immunotherapy was performed by B16F10 tumor-bearing mice. Flow cytometry analyzed the immune microenvironment. RNA sequencing analyzed the global transcriptome alterations in CPI613-treated melanoma cells. The regulation of PD-L1 and glycolysis by PDHA1/OGDH-ATF3 signaling were confirmed by Quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, dual-luciferase reporter gene, Chromatin immunoprecipitation (ChIP)-quantitative PCR and Seahorse assay. The relationship between PDHA1/OGDH-ATF3-glycolysis and the efficacy of melanoma anti-PD-1 immunotherapy was verified in the clinical database and single-cell RNA-seq (ScRNA-Seq).Results In our study, the results showed that significant alterations in metabolites associated with glycolysis and the TCA cycle in plasma of patients with melanoma through NMR technique, and then, PDHA1 and OGDH, key enzymes for regulation TCA cycle, were remarkable raised in melanoma and negatively related to anti-PD-1 efficacy through clinical database analysis as well as ScRNA-Seq. Inhibition of PDHA1 and OGDH by either shRNA or pharmacological inhibitor by CPI613 dramatically attenuated melanoma progression as well as improved the therapeutic efficacy of anti-PD-1 against melanoma. Most importantly, suppression of TCA cycle remarkably raises PD-L1 expression and glycolysis flux through AMPK-CREB-ATF3 signaling.Conclusions Taken together, our results demonstrated the role of TCA cycle in immune checkpoint blockade and provided a novel combination strategy for anti-PD-1 immunotherapy in melanoma treatment.https://jitc.bmj.com/content/11/9/e007146.full
spellingShingle Jing Chen
Jie Wu
Xiang Chen
Qian Tao
Nian Liu
Mingjie Yan
Cong Peng
Inhibition of TCA cycle improves the anti-PD-1 immunotherapy efficacy in melanoma cells via ATF3-mediated PD-L1 expression and glycolysis
Journal for ImmunoTherapy of Cancer
title Inhibition of TCA cycle improves the anti-PD-1 immunotherapy efficacy in melanoma cells via ATF3-mediated PD-L1 expression and glycolysis
title_full Inhibition of TCA cycle improves the anti-PD-1 immunotherapy efficacy in melanoma cells via ATF3-mediated PD-L1 expression and glycolysis
title_fullStr Inhibition of TCA cycle improves the anti-PD-1 immunotherapy efficacy in melanoma cells via ATF3-mediated PD-L1 expression and glycolysis
title_full_unstemmed Inhibition of TCA cycle improves the anti-PD-1 immunotherapy efficacy in melanoma cells via ATF3-mediated PD-L1 expression and glycolysis
title_short Inhibition of TCA cycle improves the anti-PD-1 immunotherapy efficacy in melanoma cells via ATF3-mediated PD-L1 expression and glycolysis
title_sort inhibition of tca cycle improves the anti pd 1 immunotherapy efficacy in melanoma cells via atf3 mediated pd l1 expression and glycolysis
url https://jitc.bmj.com/content/11/9/e007146.full
work_keys_str_mv AT jingchen inhibitionoftcacycleimprovestheantipd1immunotherapyefficacyinmelanomacellsviaatf3mediatedpdl1expressionandglycolysis
AT jiewu inhibitionoftcacycleimprovestheantipd1immunotherapyefficacyinmelanomacellsviaatf3mediatedpdl1expressionandglycolysis
AT xiangchen inhibitionoftcacycleimprovestheantipd1immunotherapyefficacyinmelanomacellsviaatf3mediatedpdl1expressionandglycolysis
AT qiantao inhibitionoftcacycleimprovestheantipd1immunotherapyefficacyinmelanomacellsviaatf3mediatedpdl1expressionandglycolysis
AT nianliu inhibitionoftcacycleimprovestheantipd1immunotherapyefficacyinmelanomacellsviaatf3mediatedpdl1expressionandglycolysis
AT mingjieyan inhibitionoftcacycleimprovestheantipd1immunotherapyefficacyinmelanomacellsviaatf3mediatedpdl1expressionandglycolysis
AT congpeng inhibitionoftcacycleimprovestheantipd1immunotherapyefficacyinmelanomacellsviaatf3mediatedpdl1expressionandglycolysis