Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

邮发代号 80-967

2019 Impact Factor: 3.421

Frontiers of Medicine  2023, Vol. 17 Issue (4): 729-746   https://doi.org/10.1007/s11684-022-0978-2
  本期目录
Dihydroartemisinin increased the abundance of Akkermansia muciniphila by YAP1 depression that sensitizes hepatocellular carcinoma to anti-PD-1 immunotherapy
Zhiqin Zhang, Xinli Shi(), Jingmin Ji, Yinglin Guo, Qing Peng, Liyuan Hao, Yu Xue, Yiwei Liu, Caige Li, Junlan Lu, Kun Yu
Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
 全文: PDF(13410 KB)   HTML
Abstract

The effect of anti-programmed cell death 1 (anti-PD-1) immunotherapy is limited in patients with hepatocellular carcinoma (HCC). Yes-associated protein 1 (YAP1) expression increased in liver tumor cells in early HCC, and Akkermansia muciniphila abundance decreased in the colon. The response to anti-PD-1 treatment is associated with A. muciniphila abundance in many tumors. However, the interaction between A. muciniphila abundance and YAP1 expression remains unclear in HCC. Here, anti-PD-1 treatment decreased A. muciniphila abundance in the colon, but increased YAP1 expression in the tumor cells by mice with liver tumors in situ. Mechanistically, hepatocyte-specific Yap1 knockout (Yap1LKO) maintained bile acid homeostasis in the liver, resulting in an increased abundance of A. muciniphila in the colon. Yap1 knockout enhanced anti-PD-1 efficacy. Therefore, YAP1 inhibition is a potential target for increasing A. muciniphila abundance to promote anti-PD-1 efficacy in liver tumors. Dihydroartemisinin (DHA), acting as YAP1 inhibitor, increased A. muciniphila abundance to sensitize anti-PD-1 therapy. A. muciniphila by gavage increased the number and activation of CD8+ T cells in liver tumor niches during DHA treatment or combination with anti-PD-1. Our findings suggested that the combination anti-PD-1 with DHA is an effective strategy for liver tumor treatment.

Key wordshepatocellular carcinoma    YAP1    Akkermansia muciniphila    anti-PD-1    dihydroartemisinin    bile acid
收稿日期: 2022-10-07      出版日期: 2023-10-12
Corresponding Author(s): Xinli Shi   
 引用本文:   
. [J]. Frontiers of Medicine, 2023, 17(4): 729-746.
Zhiqin Zhang, Xinli Shi, Jingmin Ji, Yinglin Guo, Qing Peng, Liyuan Hao, Yu Xue, Yiwei Liu, Caige Li, Junlan Lu, Kun Yu. Dihydroartemisinin increased the abundance of Akkermansia muciniphila by YAP1 depression that sensitizes hepatocellular carcinoma to anti-PD-1 immunotherapy. Front. Med., 2023, 17(4): 729-746.
 链接本文:  
https://academic.hep.com.cn/fmd/CN/10.1007/s11684-022-0978-2
https://academic.hep.com.cn/fmd/CN/Y2023/V17/I4/729
Fig.1  
Fig.2  
Fig.3  
Fig.4  
Fig.5  
Fig.6  
Fig.7  
Fig.8  
Fig.9  
1 P Konyn, A Ahmed, D Kim. Current epidemiology in hepatocellular carcinoma. Expert Rev Gastroenterol Hepatol 2021; 15(11): 1295–1307
https://doi.org/10.1080/17474124.2021.1991792 pmid: 34624198
2 AB El-Khoueiry, B Sangro, T Yau, TS Crocenzi, M Kudo, C Hsu, TY Kim, SP Choo, J Trojan, TH 3rd Welling, T Meyer, YK Kang, W Yeo, A Chopra, J Anderson, Cruz C Dela, L Lang, J Neely, H Tang, HB Dastani, I Melero. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017; 389(10088): 2492–2502
https://doi.org/10.1016/S0140-6736(17)31046-2 pmid: 28434648
3 CG Kim, C Kim, SE Yoon, KH Kim, SJ Choi, B Kang, HR Kim, SH Park, EC Shin, YY Kim, DJ Kim, HC Chung, HJ Chon, HJ Choi, HY Lim. Hyperprogressive disease during PD-1 blockade in patients with advanced hepatocellular carcinoma. J Hepatol 2021; 74(2): 350–359
https://doi.org/10.1016/j.jhep.2020.08.010 pmid: 32810553
4 AX Zhu, RS Finn, J Edeline, S Cattan, S Ogasawara, D Palmer, C Verslype, V Zagonel, L Fartoux, A Vogel, D Sarker, G Verset, SL Chan, J Knox, B Daniele, AL Webber, SW Ebbinghaus, J Ma, AB Siegel, AL Cheng, M; KEYNOTE-224 investigators Kudo. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): a non-randomised, open-label phase 2 trial. Lancet Oncol 2018; 19(7): 940–952
https://doi.org/10.1016/S1470-2045(18)30351-6 pmid: 29875066
5 Z Ren, A Li, J Jiang, L Zhou, Z Yu, H Lu, H Xie, X Chen, L Shao, R Zhang, S Xu, H Zhang, G Cui, X Chen, R Sun, H Wen, JP Lerut, Q Kan, L Li, S Zheng. Gut microbiome analysis as a tool towards targeted non-invasive biomarkers for early hepatocellular carcinoma. Gut 2019; 68(6): 1014–1023
https://doi.org/10.1136/gutjnl-2017-315084 pmid: 30045880
6 B Routy, Chatelier E Le, L Derosa, CPM Duong, MT Alou, R Daillère, A Fluckiger, M Messaoudene, C Rauber, MP Roberti, M Fidelle, C Flament, V Poirier-Colame, P Opolon, C Klein, K Iribarren, L Mondragón, N Jacquelot, B Qu, G Ferrere, C Clémenson, L Mezquita, JR Masip, C Naltet, S Brosseau, C Kaderbhai, C Richard, H Rizvi, F Levenez, N Galleron, B Quinquis, N Pons, B Ryffel, V Minard-Colin, P Gonin, JC Soria, E Deutsch, Y Loriot, F Ghiringhelli, G Zalcman, F Goldwasser, B Escudier, MD Hellmann, A Eggermont, D Raoult, L Albiges, G Kroemer, L Zitvogel. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018; 359(6371): 91–97
https://doi.org/10.1126/science.aan3706 pmid: 29097494
7 V Matson, J Fessler, R Bao, T Chongsuwat, Y Zha, ML Alegre, JJ Luke, TF Gajewski. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018; 359(6371): 104–108
https://doi.org/10.1126/science.aao3290 pmid: 29302014
8 NJ Salgia, PG Bergerot, MC Maia, N Dizman, J Hsu, JD Gillece, M Folkerts, L Reining, J Trent, SK Highlander, SK Pal. Stool microbiome profiling of patients with metastatic renal cell carcinoma receiving anti-PD-1 immune checkpoint inhibitors. Eur Urol 2020; 78(4): 498–502
https://doi.org/10.1016/j.eururo.2020.07.011 pmid: 32828600
9 Y Zheng, T Wang, X Tu, Y Huang, H Zhang, D Tan, W Jiang, S Cai, P Zhao, R Song, P Li, N Qin, W Fang. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J Immunother Cancer 2019; 7(1): 193
https://doi.org/10.1186/s40425-019-0650-9 pmid: 31337439
10 M Shibata, K Ham, MO Hoque. A time for YAP1: tumorigenesis, immunosuppression and targeted therapy. Int J Cancer 2018; 143(9): 2133–2144
https://doi.org/10.1002/ijc.31561 pmid: 29696628
11 A Perra, MA Kowalik, E Ghiso, GM Ledda-Columbano, L Di Tommaso, MM Angioni, C Raschioni, E Testore, M Roncalli, S Giordano, A Columbano. YAP activation is an early event and a potential therapeutic target in liver cancer development. J Hepatol 2014; 61(5): 1088–1096
https://doi.org/10.1016/j.jhep.2014.06.033 pmid: 25010260
12 J Cao, C Zhang, GQ Jiang, SJ Jin, Q Wang, AQ Wang, DS Bai. Identification of hepatocellular carcinoma-related genes associated with macrophage differentiation based on bioinformatics analyses. Bioengineered 2021; 12(1): 296–309
https://doi.org/10.1080/21655979.2020.1868119 pmid: 33380242
13 M Yu, Z Peng, M Qin, Y Liu, J Wang, C Zhang, J Lin, T Dong, L Wang, S Li, Y Yang, S Xu, W Guo, X Zhang, M Shi, H Peng, X Luo, H Zhang, L Zhang, Y Li, XP Yang, S Sun. Interferon-γ induces tumor resistance to anti-PD-1 immunotherapy by promoting YAP phase separation. Mol Cell 2021; 81(6): 1216–1230.e9
https://doi.org/10.1016/j.molcel.2021.01.010 pmid: 33606996
14 T Hagi, SY Geerlings, B Nijsse, C Belzer. The effect of bile acids on the growth and global gene expression profiles in Akkermansia muciniphila. Appl Microbiol Biotechnol 2020; 104(24): 10641–10653
https://doi.org/10.1007/s00253-020-10976-3 pmid: 33159542
15 L Van den Bossche, P Hindryckx, L Devisscher, S Devriese, S Van Welden, T Holvoet, R Vilchez-Vargas, M Vital, DH Pieper, J Vanden Bussche, L Vanhaecke, T Van de Wiele, M De Vos, D Laukens. Ursodeoxycholic acid and its taurine- or glycine-conjugated species reduce colitogenic dysbiosis and equally suppress experimental colitis in mice. Appl Environ Microbiol 2017; 83(7): e02766–16
https://doi.org/10.1128/AEM.02766-16 pmid: 28115375
16 X Zheng, F Huang, A Zhao, S Lei, Y Zhang, G Xie, T Chen, C Qu, C Rajani, B Dong, D Li, W Jia. Bile acid is a significant host factor shaping the gut microbiome of diet-induced obese mice. BMC Biol 2017; 15(1): 120
https://doi.org/10.1186/s12915-017-0462-7 pmid: 29241453
17 S Anakk, M Bhosale, VA Schmidt, RL Johnson, MJ Finegold, DD Moore. Bile acids activate YAP to promote liver carcinogenesis. Cell Rep 2013; 5(4): 1060–1069
https://doi.org/10.1016/j.celrep.2013.10.030 pmid: 24268772
18 T Wang, R Luo, W Li, H Yan, S Xie, W Xiao, Y Wang, B Chen, P Bai, J Xing. Dihydroartemisinin suppresses bladder cancer cell invasion and migration by regulating KDM3A and p21. J Cancer 2020; 11(5): 1115–1124
https://doi.org/10.7150/jca.36174 pmid: 31956358
19 Q Li, Q Ma, J Cheng, X Zhou, W Pu, X Zhong, X Guo. Dihydroartemisinin as a sensitizing agent in cancer therapies. OncoTargets Ther 2021; 14: 2563–2573
https://doi.org/10.2147/OTT.S297785 pmid: 33880035
20 Z LiG Tuteja J SchugKH Kaestner. Foxa1 and Foxa2 are essential for sexual dimorphism in liver cancer. Cell 2012; 148(1–2): 72–83 doi:10.1016/j.cell.2011.11.026
pmid: 22265403
21 J Zhang, X Bu, H Wang, Y Zhu, Y Geng, NT Nihira, Y Tan, Y Ci, F Wu, X Dai, J Guo, YH Huang, C Fan, S Ren, Y Sun, GJ Freeman, P Sicinski, W Wei. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature 2018; 553(7686): 91–95
https://doi.org/10.1038/nature25015 pmid: 29160310
22 Z Jiang, SO Lim, M Yan, JL Hsu, J Yao, Y Wei, SS Chang, H Yamaguchi, HH Lee, B Ke, JM Hsu, LC Chan, GN Hortobagyi, L Yang, C Lin, D Yu, MC Hung. TYRO3 induces anti-PD-1/PD-L1 therapy resistance by limiting innate immunity and tumoral ferroptosis. J Clin Invest 2021; 131(8): e139434
https://doi.org/10.1172/JCI139434 pmid: 33855973
23 L Hao, Y Guo, Q Peng, Z Zhang, J Ji, Y Liu, Y Xue, C Li, K Zheng, X Shi. Dihydroartemisinin reduced lipid droplet deposition by YAP1 to promote the anti-PD-1 effect in hepatocellular carcinoma. Phytomedicine 2022; 96: 153913
https://doi.org/10.1016/j.phymed.2021.153913 pmid: 35026515
24 Y Guo, Q Peng, L Hao, J Ji, Z Zhang, Y Xue, Y Liu, Y Gao, C Li, X Shi. Dihydroartemisinin promoted FXR expression independent of YAP1 in hepatocellular carcinoma. FASEB J 2022; 36(6): e22361
https://doi.org/10.1096/fj.202200171R pmid: 35616366
25 J Li, S Lin, PM Vanhoutte, CW Woo, A Xu. Akkermansia muciniphila protects against atherosclerosis by preventing metabolic endotoxemia-induced inflammation in Apoe–/– mice. Circulation 2016; 133(24): 2434–2446
https://doi.org/10.1161/CIRCULATIONAHA.115.019645 pmid: 27143680
26 MC Collado, M Derrien, E Isolauri, WM de Vos, S Salminen. Intestinal integrity and Akkermansia muciniphila, a mucin-degrading member of the intestinal microbiota present in infants, adults, and the elderly. Appl Environ Microbiol 2007; 73(23): 7767–7770
https://doi.org/10.1128/AEM.01477-07 pmid: 17933936
27 R Pieper, J Bindelle, B Rossnagel, A Van Kessel, P Leterme. Effect of carbohydrate composition in barley and oat cultivars on microbial ecophysiology and proliferation of Salmonella enterica in an in vitro model of the porcine gastrointestinal tract. Appl Environ Microbiol 2009; 75(22): 7006–7016
https://doi.org/10.1128/AEM.01343-09 pmid: 19783749
28 L Wang, L Tang, Y Feng, S Zhao, M Han, C Zhang, G Yuan, J Zhu, S Cao, Q Wu, L Li, Z Zhang. A purified membrane protein from Akkermansia muciniphila or the pasteurised bacterium blunts colitis associated tumourigenesis by modulation of CD8+ T cells in mice. Gut 2020; 69(11): 1988–1997
https://doi.org/10.1136/gutjnl-2019-320105 pmid: 32169907
29 X Jiu, Y Liu, J Wen. Artesunate combined with verteporfin inhibits uveal melanoma by regulation of the MALAT1/yes-associated protein signaling pathway. Oncol Lett 2021; 22(2): 597
https://doi.org/10.3892/ol.2021.12858 pmid: 34188699
30 R Liu, HS Choi, YC Ko, BS Yun, DS Lee. 5-Desmethylsinensetin isolated from Artemisia princeps suppresses the stemness of breast cancer cells via Stat3/IL-6 and Stat3/YAP1 signaling. Life Sci 2021; 280: 119729
https://doi.org/10.1016/j.lfs.2021.119729 pmid: 34146553
31 V Gopalakrishnan, CN Spencer, L Nezi, A Reuben, MC Andrews, TV Karpinets, PA Prieto, D Vicente, K Hoffman, SC Wei, AP Cogdill, L Zhao, CW Hudgens, DS Hutchinson, T Manzo, M Petaccia de Macedo, T Cotechini, T Kumar, WS Chen, SM Reddy, R Szczepaniak Sloane, J Galloway-Pena, H Jiang, PL Chen, EJ Shpall, K Rezvani, AM Alousi, RF Chemaly, S Shelburne, LM Vence, PC Okhuysen, VB Jensen, AG Swennes, F McAllister, E Marcelo Riquelme Sanchez, Y Zhang, E Le Chatelier, L Zitvogel, N Pons, JL Austin-Breneman, LE Haydu, EM Burton, JM Gardner, E Sirmans, J Hu, AJ Lazar, T Tsujikawa, A Diab, H Tawbi, IC Glitza, WJ Hwu, SP Patel, SE Woodman, RN Amaria, MA Davies, JE Gershenwald, P Hwu, JE Lee, J Zhang, LM Coussens, ZA Cooper, PA Futreal, CR Daniel, NJ Ajami, JF Petrosino, MT Tetzlaff, P Sharma, JP Allison, RR Jenq, JA Wargo. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018; 359(6371): 97–103
https://doi.org/10.1126/science.aan4236 pmid: 29097493
32 ZY Gao, Z Cui, YQ Yan, LJ Ning, ZH Wang, J Hong. Microbe-based management for colorectal cancer. Chin Med J (Engl) 2021; 134(24): 2922–2930
https://doi.org/10.1097/CM9.0000000000001887 pmid: 34855639
33 KS Sfanos, MC Markowski, LB Peiffer, SE Ernst, JR White, KJ Pienta, ES Antonarakis, AE Ross. Compositional differences in gastrointestinal microbiota in prostate cancer patients treated with androgen axis-targeted therapies. Prostate Cancer Prostatic Dis 2018; 21(4): 539–548
https://doi.org/10.1038/s41391-018-0061-x pmid: 29988102
34 PL Miller, TL Carson. Mechanisms and microbial influences on CTLA-4 and PD-1-based immunotherapy in the treatment of cancer: a narrative review. Gut Pathog 2020; 12(1): 43
https://doi.org/10.1186/s13099-020-00381-6 pmid: 32944086
35 EJ Snider, G Compres, DE Freedberg, H Khiabanian, YR Nobel, S Stump, AC Uhlemann, CJ Lightdale, JA Abrams. Alterations to the esophageal microbiome associated with progression from Barrett’s esophagus to esophageal adenocarcinoma. Cancer Epidemiol Biomarkers Prev 2019; 28(10): 1687–1693
https://doi.org/10.1158/1055-9965.EPI-19-0008 pmid: 31466948
36 NT Baxter, JP Zackular, GY Chen, PD Schloss. Structure of the gut microbiome following colonization with human feces determines colonic tumor burden. Microbiome 2014; 2(1): 20
https://doi.org/10.1186/2049-2618-2-20 pmid: 24967088
37 MA Osman, HM Neoh, NS Ab Mutalib, SF Chin, L Mazlan, RA Raja Ali, AD Zakaria, CS Ngiu, MY Ang, R Jamal. Parvimonas micra, Peptostreptococcus stomatis, Fusobacterium nucleatum and Akkermansia muciniphila as a four-bacteria biomarker panel of colorectal cancer. Sci Rep 2021; 11(1): 2925
https://doi.org/10.1038/s41598-021-82465-0 pmid: 33536501
38 F Wang, K Cai, Q Xiao, L He, L Xie, Z Liu. Akkermansia muciniphila administration exacerbated the development of colitis-associated colorectal cancer in mice. J Cancer 2022; 13(1): 124–133
https://doi.org/10.7150/jca.63578 pmid: 34976176
39 M Santoni, F Piva, A Conti, A Santoni, A Cimadamore, M Scarpelli, N Battelli, R Montironi. Re: gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Eur Urol 2018; 74(4): 521–522
https://doi.org/10.1016/j.eururo.2018.05.033 pmid: 29891391
40 ZW Luo, K Xia, YW Liu, JH Liu, SS Rao, XK Hu, CY Chen, R Xu, ZX Wang, H Xie. Extracellular vesicles from Akkermansia muciniphila elicit antitumor immunity against prostate cancer via modulation of CD8+ T cells and macrophages. Int J Nanomedicine 2021; 16: 2949–2963
https://doi.org/10.2147/IJN.S304515 pmid: 33907401
41 S Septer, G Edwards, S Gunewardena, A Wolfe, H Li, J Daniel, U Apte. Yes-associated protein is involved in proliferation and differentiation during postnatal liver development. Am J Physiol Gastrointest Liver Physiol 2012; 302(5): G493–G503
https://doi.org/10.1152/ajpgi.00056.2011 pmid: 22194415
42 H Wu, L Wei, F Fan, S Ji, S Zhang, J Geng, L Hong, X Fan, Q Chen, J Tian, M Jiang, X Sun, C Jin, ZY Yin, Q Liu, J Zhang, F Qin, KH Lin, JS Yu, X Deng, HR Wang, B Zhao, RL Johnson, L Chen, D Zhou. Integration of Hippo signalling and the unfolded protein response to restrain liver overgrowth and tumorigenesis. Nat Commun 2015; 6(1): 6239
https://doi.org/10.1038/ncomms7239 pmid: 25695629
43 G Xie, X Wang, F Huang, A Zhao, W Chen, J Yan, Y Zhang, S Lei, K Ge, X Zheng, J Liu, M Su, P Liu, W Jia. Dysregulated hepatic bile acids collaboratively promote liver carcinogenesis. Int J Cancer 2016; 139(8): 1764–1775
https://doi.org/10.1002/ijc.30219 pmid: 27273788
44 Y Fang, SI Han, C Mitchell, S Gupta, E Studer, S Grant, PB Hylemon, P Dent. Bile acids induce mitochondrial ROS, which promote activation of receptor tyrosine kinases and signaling pathways in rat hepatocytes. Hepatology 2004; 40(4): 961–971
https://doi.org/10.1002/hep.1840400427 pmid: 15382121
45 MJ Perez, O Briz. Bile-acid-induced cell injury and protection. World J Gastroenterol 2009; 15(14): 1677–1689
https://doi.org/10.3748/wjg.15.1677 pmid: 19360911
46 P Luo, P Yin, R Hua, Y Tan, Z Li, G Qiu, Z Yin, X Xie, X Wang, W Chen, L Zhou, X Wang, Y Li, H Chen, L Gao, X Lu, T Wu, H Wang, J Niu, G Xu. A large-scale, multicenter serum metabolite biomarker identification study for the early detection of hepatocellular carcinoma. Hepatology 2018; 67(2): 662–675
https://doi.org/10.1002/hep.29561 pmid: 28960374
47 T Chen, G Xie, X Wang, J Fan, Y Qiu, X Zheng, X Qi, Y Cao, M Su, X Wang, LX Xu, Y Yen, P Liu, W Jia. Serum and urine metabolite profiling reveals potential biomarkers of human hepatocellular carcinoma. Mol Cell Proteomics 2011; 10(7): M110.004945
https://doi.org/10.1074/mcp.M110.004945 pmid: 21518826
48 E Loftfield, JA Rothwell, R Sinha, P Keski-Rahkonen, N Robinot, D Albanes, SJ Weinstein, A Derkach, J Sampson, A Scalbert, ND Freedman. Prospective investigation of serum metabolites, coffee drinking, liver cancer incidence, and liver disease mortality. J Natl Cancer Inst 2020; 112(3): 286–294
https://doi.org/10.1093/jnci/djz122 pmid: 31168595
49 Geerlings SY, Kostopoulos I, de Vos WM, Belzer C. Akkermansia muciniphila in the human gastrointestinal tract: when, where, and how? Microorganisms 2018; 6(3): 75 doi:10.3390/microorganisms6030075
pmid: 30041463
50 MH Foley, S O’Flaherty, R Barrangou, CM Theriot. Bile salt hydrolases: gatekeepers of bile acid metabolism and host-microbiome crosstalk in the gastrointestinal tract. PLoS Pathog 2019; 15(3): e1007581
https://doi.org/10.1371/journal.ppat.1007581 pmid: 30845232
51 C Grajeda-Iglesias, S Durand, R Daillère, K Iribarren, F Lemaitre, L Derosa, F Aprahamian, N Bossut, N Nirmalathasan, F Madeo, L Zitvogel, G Kroemer. Oral administration of Akkermansia muciniphila elevates systemic antiaging and anticancer metabolites. Aging (Albany NY) 2021; 13(5): 6375–6405
https://doi.org/10.18632/aging.202739 pmid: 33653967
[1] FMD-22055-OF-SXL_suppl_1 Download
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed