Gut microbiome is associated with the clinical response to anti-PD-1 based immunotherapy in hepatobiliary cancers

Jinzhu Mao, Dongxu Wang, Junyu Long, Xu Yang, Jianzhen Lin, Yiwei Song, Fucun Xie, Ziyu Xun, Yanyu Wang, Yunchao Wang, Yiran Li, Huishan Sun, Jingnan Xue, Yang Song, Bangyou Zuo, Junwei Zhang, Jin Bian, Ting Zhang, Xiaobo Yang, Lei Zhang, Xinting Sang, Haitao Zhao, Jinzhu Mao, Dongxu Wang, Junyu Long, Xu Yang, Jianzhen Lin, Yiwei Song, Fucun Xie, Ziyu Xun, Yanyu Wang, Yunchao Wang, Yiran Li, Huishan Sun, Jingnan Xue, Yang Song, Bangyou Zuo, Junwei Zhang, Jin Bian, Ting Zhang, Xiaobo Yang, Lei Zhang, Xinting Sang, Haitao Zhao

Abstract

Background: The gut microbiome is associated with the response to immunotherapy for different cancers. However, the impact of the gut microbiome on hepatobiliary cancers receiving immunotherapy remains unknown. This study aims to investigate the relationship between the gut microbiome and the clinical response to anti-programmed cell death protein 1 (PD-1) immunotherapy in patients with advanced hepatobiliary cancers.

Methods: Patients with unresectable hepatocellular carcinoma or advanced biliary tract cancers who have progressed from first-line chemotherapy (gemcitabine plus cisplatin) were enrolled. Fresh stool samples were collected before and during anti-PD-1 treatment and analyzed with metagenomic sequencing. Significantly differentially enriched taxa and prognosis associated taxa were identified. The Kyoto Encyclopedia of Genes and Genomes database and MetaCyc database were further applied to annotate the differentially enriched taxa to explore the potential mechanism of the gut microbiome influencing cancer immunotherapy.

Results: In total, 65 patients with advanced hepatobiliary cancers receiving anti-PD-1 treatment were included in this study. Seventy-four taxa were significantly enriched in the clinical benefit response (CBR) group and 40 taxa were significantly enriched in the non-clinical benefit (NCB) group. Among these taxa, patients with higher abundance of Lachnospiraceae bacterium-GAM79 and Alistipes sp Marseille-P5997, which were significantly enriched in the CBR group, achieved longer progression-free survival (PFS) and overall survival (OS) than patients with lower abundance. Higher abundance of Ruminococcus calidus and Erysipelotichaceae bacterium-GAM147 enriched in the CBR group was also observed in patients with better PFS. In contrast, worse PFS and OS were found in patients with higher abundance of Veillonellaceae, which was significantly enriched in the NCB group. Functional annotation indicated that the taxa enriched in the CBR group were associated with energy metabolism while the taxa enriched in the NCB group were associated with amino acid metabolism, which may modulate the clinical response to immunotherapy in hepatobiliary cancers. In addition, immunotherapy-related adverse events were affected by the gut microbiome diversity and relative abundance.

Conclusions: We demonstrate that the gut microbiome is associated with the clinical response to anti-PD-1 immunotherapy in patients with hepatobiliary cancers. Taxonomic signatures enriched in responders are effective biomarkers to predict the clinical response and survival benefit of immunotherapy, which might provide a new therapeutic target to modulate the response to cancer immunotherapy.

Keywords: immunotherapy; liver neoplasms; tumor biomarkers.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Study workflow. BTC, biliary tract cancer; CBR, clinical benefit response; HCC, hepatocellular carcinoma; NCB, non-clinical benefit; OS, overall survival; PD-1, programmed cell death protein 1; PFS, progression-free survival.
Figure 2
Figure 2
Gut microbiome composition of 65 patients with hepatobiliary cancers. (A) Gut microbiome composition at the order level in the 65 patients with hepatobiliary cancers (ordered by the most abundant taxa, Bacteroideles order). (B) Relative abundance comparison of Bacteroidales, Enterobacterales and Veillonellales in the CBR group and NCB group at the order level (Wilcoxon test). (C) Relative abundance comparison of Lachnospiraceae bacterium-GAM79, Ruminococcus callidus, Eubacterium siraeum, Gemmiger formicilis and Faecalibacterium genus in the CBR group and NCB group (Wilcoxon test). (D) Dynamic microbial composition in the CBR group and NCB group at the phylum level. CBR, clinical benefit response; NCB, non-clinical benefit.
Figure 3
Figure 3
Differential taxa were enriched in the CBR group and NCB group of the 65 patients with hepatobiliary cancers. (A) PCoA showed the beta diversity evaluated by Bray-Curtis distance between the CBR group and NCB group. (B) Common and unique taxa at the specie level between the CBR group and NCB group. (C) Taxonomic cladogram from LEfSe showed different taxa enriched in the CBR group and NCB group (LDA>3, p3, p

Figure 4

Differentially enriched taxa were associated…

Figure 4

Differentially enriched taxa were associated with the survival benefit of anti-programmed cell death…

Figure 4
Differentially enriched taxa were associated with the survival benefit of anti-programmed cell death protein 1 immunotherapy in patients with hepatobiliary cancers. The Kaplan-Meier method with log-rank test estimates the median progression-free survival (A) and median overall survival (B) for patients with higher or lower abundance of Lachnospiraceae bacterium-GAM79, Alistipes sp Marseille-P5997 and Veillonellaceae family.

Figure 5

Functional annotation of the gut…

Figure 5

Functional annotation of the gut microbiome metagenomic sequencing data. (A) Different KOs enriched…

Figure 5
Functional annotation of the gut microbiome metagenomic sequencing data. (A) Different KOs enriched in the CBR group and NCB group identified by LEfSe (LDA>2, p2, p

Figure 6

Immunotherapy-related adverse events were affected…

Figure 6

Immunotherapy-related adverse events were affected by the gut microbiome. (A) The Kaplan-Meier method…

Figure 6
Immunotherapy-related adverse events were affected by the gut microbiome. (A) The Kaplan-Meier method with log-rank test estimates the mPFS for patients with severe or mild diarrhea. (B) Gut microbiome alpha diversity comparison between the severe and mild diarrhea groups (Wilcoxon test). (C) Relative abundance comparison of significantly different taxa between patients with severe diarrhea and mild diarrhea (Wilcoxon test). mPFS, median progression-free survival.

Figure 7

The gut microbiome distribution was…

Figure 7

The gut microbiome distribution was affected by clinical factors. (A) RDA with permutation…

Figure 7
The gut microbiome distribution was affected by clinical factors. (A) RDA with permutation test showed the clinical factors associated with the distribution of patients with hepatobiliary cancers. (B) RDA with permutation test showed the clinical factors associated with the different taxa enriched in the CBR group and NCB group. (C) Relative abundance comparison of Eubacterium hallii in patients with large tumors (≥5 cm) and small tumors (≥5 cm) (Wilcoxon test). (D) Relative abundance comparison of the Clostridium genus in patients with elevated bile acid and normal bile acid (Wilcoxon test). CBR, clinical benefit response; ECOG-PS, Eastern Cooperative Oncology Group performance status; NCB, non-clinical benefit; RDA, redundancy analysis.
All figures (7)
Similar articles
Cited by
References
    1. Sung H, Ferlay J, Siegel RL, et al. . Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209–49. 10.3322/caac.21660 - DOI - PubMed
    1. Rizvi S, Khan SA, Hallemeier CL, et al. . Cholangiocarcinoma - evolving concepts and therapeutic strategies. Nat Rev Clin Oncol 2018;15:95–111. 10.1038/nrclinonc.2017.157 - DOI - PMC - PubMed
    1. Finn RS, Qin S, Ikeda M, et al. . Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med 2020;382:1894–905. 10.1056/NEJMoa1915745 - DOI - PubMed
    1. Finn RS, Qin S, Ikeda M, et al. . IMbrave150: updated overall survival (OS) data from a global, randomized, open-label phase III study of atezolizumab (atezo) + bevacizumab (bev) versus sorafenib (SOR) in patients (PTS) with unresectable hepatocellular carcinoma (HCC). Journal of Clinical Oncology 2021;39:267. 10.1200/JCO.2021.39.3_suppl.267 - DOI
    1. NCCN Clinical Practice Guidelines in Oncology . Hepatobiliary cancers. Version 2 2021.
Show all 57 references
Publication types
MeSH terms
Related information
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4
Figure 4
Differentially enriched taxa were associated with the survival benefit of anti-programmed cell death protein 1 immunotherapy in patients with hepatobiliary cancers. The Kaplan-Meier method with log-rank test estimates the median progression-free survival (A) and median overall survival (B) for patients with higher or lower abundance of Lachnospiraceae bacterium-GAM79, Alistipes sp Marseille-P5997 and Veillonellaceae family.
Figure 5
Figure 5
Functional annotation of the gut microbiome metagenomic sequencing data. (A) Different KOs enriched in the CBR group and NCB group identified by LEfSe (LDA>2, p2, p

Figure 6

Immunotherapy-related adverse events were affected…

Figure 6

Immunotherapy-related adverse events were affected by the gut microbiome. (A) The Kaplan-Meier method…

Figure 6
Immunotherapy-related adverse events were affected by the gut microbiome. (A) The Kaplan-Meier method with log-rank test estimates the mPFS for patients with severe or mild diarrhea. (B) Gut microbiome alpha diversity comparison between the severe and mild diarrhea groups (Wilcoxon test). (C) Relative abundance comparison of significantly different taxa between patients with severe diarrhea and mild diarrhea (Wilcoxon test). mPFS, median progression-free survival.

Figure 7

The gut microbiome distribution was…

Figure 7

The gut microbiome distribution was affected by clinical factors. (A) RDA with permutation…

Figure 7
The gut microbiome distribution was affected by clinical factors. (A) RDA with permutation test showed the clinical factors associated with the distribution of patients with hepatobiliary cancers. (B) RDA with permutation test showed the clinical factors associated with the different taxa enriched in the CBR group and NCB group. (C) Relative abundance comparison of Eubacterium hallii in patients with large tumors (≥5 cm) and small tumors (≥5 cm) (Wilcoxon test). (D) Relative abundance comparison of the Clostridium genus in patients with elevated bile acid and normal bile acid (Wilcoxon test). CBR, clinical benefit response; ECOG-PS, Eastern Cooperative Oncology Group performance status; NCB, non-clinical benefit; RDA, redundancy analysis.
All figures (7)
Figure 6
Figure 6
Immunotherapy-related adverse events were affected by the gut microbiome. (A) The Kaplan-Meier method with log-rank test estimates the mPFS for patients with severe or mild diarrhea. (B) Gut microbiome alpha diversity comparison between the severe and mild diarrhea groups (Wilcoxon test). (C) Relative abundance comparison of significantly different taxa between patients with severe diarrhea and mild diarrhea (Wilcoxon test). mPFS, median progression-free survival.
Figure 7
Figure 7
The gut microbiome distribution was affected by clinical factors. (A) RDA with permutation test showed the clinical factors associated with the distribution of patients with hepatobiliary cancers. (B) RDA with permutation test showed the clinical factors associated with the different taxa enriched in the CBR group and NCB group. (C) Relative abundance comparison of Eubacterium hallii in patients with large tumors (≥5 cm) and small tumors (≥5 cm) (Wilcoxon test). (D) Relative abundance comparison of the Clostridium genus in patients with elevated bile acid and normal bile acid (Wilcoxon test). CBR, clinical benefit response; ECOG-PS, Eastern Cooperative Oncology Group performance status; NCB, non-clinical benefit; RDA, redundancy analysis.

References

    1. Sung H, Ferlay J, Siegel RL, et al. . Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209–49. 10.3322/caac.21660
    1. Rizvi S, Khan SA, Hallemeier CL, et al. . Cholangiocarcinoma - evolving concepts and therapeutic strategies. Nat Rev Clin Oncol 2018;15:95–111. 10.1038/nrclinonc.2017.157
    1. Finn RS, Qin S, Ikeda M, et al. . Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med 2020;382:1894–905. 10.1056/NEJMoa1915745
    1. Finn RS, Qin S, Ikeda M, et al. . IMbrave150: updated overall survival (OS) data from a global, randomized, open-label phase III study of atezolizumab (atezo) + bevacizumab (bev) versus sorafenib (SOR) in patients (PTS) with unresectable hepatocellular carcinoma (HCC). Journal of Clinical Oncology 2021;39:267. 10.1200/JCO.2021.39.3_suppl.267
    1. NCCN Clinical Practice Guidelines in Oncology . Hepatobiliary cancers. Version 2 2021.
    1. Llovet JM, Kelley RK, Villanueva A, et al. . Hepatocellular carcinoma. Nat Rev Dis Primers 2021;7:6. 10.1038/s41572-020-00240-3
    1. Valle JW, Kelley RK, Nervi B, et al. . Biliary tract cancer. Lancet 2021;397:428–44. 10.1016/S0140-6736(21)00153-7
    1. Schwabe RF, Jobin C. The microbiome and cancer. Nat Rev Cancer 2013;13:800–12. 10.1038/nrc3610
    1. Skelly AN, Sato Y, Kearney S, et al. . Mining the microbiota for microbial and metabolite-based immunotherapies. Nat Rev Immunol 2019;19:305–23. 10.1038/s41577-019-0144-5
    1. Gopalakrishnan V, Helmink BA, Spencer CN, et al. . The influence of the gut microbiome on cancer, immunity, and cancer immunotherapy. Cancer Cell 2018;33:570–80. 10.1016/j.ccell.2018.03.015
    1. Matson V, Chervin CS, Gajewski TF. Cancer and the microbiome-influence of the commensal microbiota on cancer, immune responses, and immunotherapy. Gastroenterology 2021;160:600–13. 10.1053/j.gastro.2020.11.041
    1. Routy B, Gopalakrishnan V, Daillère R, et al. . The gut microbiota influences anticancer immunosurveillance and general health. Nat Rev Clin Oncol 2018;15:382–96. 10.1038/s41571-018-0006-2
    1. Helmink BA, Khan MAW, Hermann A, et al. . The microbiome, cancer, and cancer therapy. Nat Med 2019;25:377–88. 10.1038/s41591-019-0377-7
    1. Sivan A, Corrales L, Hubert N, et al. . Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015;350:1084–9. 10.1126/science.aac4255
    1. Vétizou M, Pitt JM, Daillère R, et al. . Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015;350:1079–84. 10.1126/science.aad1329
    1. Bhatt AP, Redinbo MR, Bultman SJ. The role of the microbiome in cancer development and therapy. CA Cancer J Clin 2017;67:326–44. 10.3322/caac.21398
    1. Routy B, Le Chatelier E, Derosa L, et al. . Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018;359:91–7. 10.1126/science.aan3706
    1. Gopalakrishnan V, Spencer CN, Nezi L, et al. . Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018;359:97–103. 10.1126/science.aan4236
    1. Matson V, Fessler J, Bao R, et al. . The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018;359:104–8. 10.1126/science.aao3290
    1. Baruch EN, Youngster I, Ben-Betzalel G, et al. . Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 2021;371:602–9. 10.1126/science.abb5920
    1. Davar D, Dzutsev AK, McCulloch JA, et al. . Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science 2021;371:595–602. 10.1126/science.abf3363
    1. Tanoue T, Morita S, Plichta DR, et al. . A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 2019;565:600–5. 10.1038/s41586-019-0878-z
    1. Schwabe RF, Greten TF. Gut microbiome in HCC - mechanisms, diagnosis and therapy. J Hepatol 2020;72:230–8. 10.1016/j.jhep.2019.08.016
    1. Yu L-X, Schwabe RF. The gut microbiome and liver cancer: mechanisms and clinical translation. Nat Rev Gastroenterol Hepatol 2017;14:527–39. 10.1038/nrgastro.2017.72
    1. Behary J, Amorim N, Jiang X-T, et al. . Gut microbiota impact on the peripheral immune response in non-alcoholic fatty liver disease related hepatocellular carcinoma. Nat Commun 2021;12:187. 10.1038/s41467-020-20422-7
    1. Zhang Q, Ma C, Duan Y, et al. . Gut microbiome directs hepatocytes to recruit MDSCs and promote cholangiocarcinoma. Cancer Discov 2021;11:1248–67. 10.1158/-20-0304
    1. Ma C, Han M, Heinrich B, et al. . Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 2018;36010.1126/science.aan5931
    1. Zheng Y, Wang T, Tu X, et al. . Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J Immunother Cancer 2019;7:193. 10.1186/s40425-019-0650-9
    1. Peng Z, Cheng S, Kou Y, et al. . The gut microbiome is associated with clinical response to anti-PD-1/PD-L1 immunotherapy in gastrointestinal cancer. Cancer Immunol Res 2020;8:1251–61. 10.1158/2326-6066.CIR-19-1014
    1. Health NIo . Common terminology criteria for adverse events (CTCAE). Version 5.0 2017.
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 2014;30:2114–20. 10.1093/bioinformatics/btu170
    1. Rosenbloom KR, Armstrong J, Barber GP, et al. . The UCSC genome browser database: 2015 update. Nucleic Acids Res 2015;43:D670–81. 10.1093/nar/gku1177
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012;9:357–9. 10.1038/nmeth.1923
    1. Wood DE, Lu J, Langmead B. Improved metagenomic analysis with Kraken 2. Genome Biol 2019;20:257. 10.1186/s13059-019-1891-0
    1. Lu J, Breitwieser FP, Thielen P, et al. . Bracken: estimating species abundance in metagenomics data. PeerJ Comput Sci 2017;3:e104. 10.7717/peerj-cs.104
    1. Jari Oksanen FGB, Friendly M, et al. . Package vegan. community ecology package (version2) 2020:5–7.
    1. Liu Y-X, Qin Y, Chen T, et al. . A practical guide to amplicon and metagenomic analysis of microbiome data. Protein Cell 2021;12:315–30. 10.1007/s13238-020-00724-8
    1. Segata N, Izard J, Waldron L, et al. . Metagenomic biomarker discovery and explanation. Genome Biol 2011;12:R60. 10.1186/gb-2011-12-6-r60
    1. Buchfink B, Xie C, Huson DH. Fast and sensitive protein alignment using DIAMOND. Nat Methods 2015;12:59–60. 10.1038/nmeth.3176
    1. Franzosa EA, McIver LJ, Rahnavard G, et al. . Species-level functional profiling of metagenomes and metatranscriptomes. Nat Methods 2018;15:962–8. 10.1038/s41592-018-0176-y
    1. Kanehisa M, Furumichi M, Tanabe M, et al. . KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res 2017;45:D353–61. 10.1093/nar/gkw1092
    1. Caspi R, Billington R, Ferrer L, et al. . The MetaCyc database of metabolic pathways and enzymes and the BioCyc collection of pathway/genome databases. Nucleic Acids Res 2016;44:D471–80. 10.1093/nar/gkv1164
    1. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol 2016;16:341–52. 10.1038/nri.2016.42
    1. Furusawa Y, Obata Y, Fukuda S, et al. . Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013;504:446–50. 10.1038/nature12721
    1. Roberti MP, Yonekura S, Duong CPM, et al. . Chemotherapy-induced ileal crypt apoptosis and the ileal microbiome shape immunosurveillance and prognosis of proximal colon cancer. Nat Med 2020;26:919–31. 10.1038/s41591-020-0882-8
    1. Sorbara MT, Littmann ER, Fontana E, et al. . Functional and genomic variation between human-derived isolates of Lachnospiraceae reveals inter- and intra-species diversity. Cell Host Microbe 2020;28:134–46. 10.1016/j.chom.2020.05.005
    1. Sinha SR, Haileselassie Y, Nguyen LP, et al. . Dysbiosis-induced secondary bile acid deficiency promotes intestinal inflammation. Cell Host Microbe 2020;27:659–70. 10.1016/j.chom.2020.01.021
    1. Peters BA, Wilson M, Moran U, et al. . Relating the gut metagenome and metatranscriptome to immunotherapy responses in melanoma patients. Genome Med 2019;11:61. 10.1186/s13073-019-0672-4
    1. Jia X, Lu S, Zeng Z, et al. . Characterization of gut microbiota, bile acid metabolism, and cytokines in intrahepatic cholangiocarcinoma. Hepatology 2020;71:893–906. 10.1002/hep.30852
    1. Campbell C, McKenney PT, Konstantinovsky D, et al. . Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells. Nature 2020;581:475–9. 10.1038/s41586-020-2193-0
    1. Song X, Sun X, Oh SF, et al. . Microbial bile acid metabolites modulate gut RORγ+ regulatory T cell homeostasis. Nature 2020;577:410–5. 10.1038/s41586-019-1865-0
    1. Huang H, Ren Z, Gao X, et al. . Integrated analysis of microbiome and host transcriptome reveals correlations between gut microbiota and clinical outcomes in HBV-related hepatocellular carcinoma. Genome Med 2020;12:102. 10.1186/s13073-020-00796-5
    1. Andrews MC, Duong CPM, Gopalakrishnan V, et al. . Gut microbiota signatures are associated with toxicity to combined CTLA-4 and PD-1 blockade. Nat Med 2021;27:1432–41. 10.1038/s41591-021-01406-6
    1. Dubin K, Callahan MK, Ren B, et al. . Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun 2016;7:10391. 10.1038/ncomms10391
    1. Chaput N, Lepage P, Coutzac C, et al. . Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol 2017;28:1368–79. 10.1093/annonc/mdx108
    1. Hakozaki T, Richard C, Elkrief A, et al. . The gut microbiome associates with immune checkpoint inhibition outcomes in patients with advanced non-small cell lung cancer. Cancer Immunol Res 2020;8:1243–50. 10.1158/2326-6066.CIR-20-0196
    1. Cortellini A, Di Maio M, Nigro O, et al. . Differential influence of antibiotic therapy and other medications on oncological outcomes of patients with non-small cell lung cancer treated with first-line pembrolizumab versus cytotoxic chemotherapy. J Immunother Cancer 2021;9:e002421. 10.1136/jitc-2021-002421

Source: PubMed

3
Abonner