Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Review
. 2021 Aug 19:11:720842.
doi: 10.3389/fonc.2021.720842. eCollection 2021.

The Role of Gut Microbiota in Lung Cancer: From Carcinogenesis to Immunotherapy

Affiliations
Review

The Role of Gut Microbiota in Lung Cancer: From Carcinogenesis to Immunotherapy

Xiangjun Liu et al. Front Oncol. .

Abstract

The influence of microbiota on host health and disease has attracted adequate attention, and gut microbiota components and microbiota-derived metabolites affect host immune homeostasis locally and systematically. Some studies have found that gut dysbiosis, disturbance of the structure and function of the gut microbiome, disrupts pulmonary immune homeostasis, thus leading to increased disease susceptibility; the gut-lung axis is the primary cross-talk for this communication. Gut dysbiosis is involved in carcinogenesis and the progression of lung cancer through genotoxicity, systemic inflammation, and defective immunosurveillance. In addition, the gut microbiome harbors the potential to be a novel biomarker for predicting sensitivity and adverse reactions to immunotherapy in patients with lung cancer. Probiotics and fecal microbiota transplantation (FMT) can enhance the efficacy and depress the toxicity of immune checkpoint inhibitors by regulating the gut microbiota. Although current studies have found that gut microbiota closely participates in the development and immunotherapy of lung cancer, the mechanisms require further investigation. Therefore, this review aims to discuss the underlying mechanisms of gut microbiota influencing carcinogenesis and immunotherapy in lung cancer and to provide new strategies for governing gut microbiota to enhance the prevention and treatment of lung cancer.

Keywords: biomarker; gut microbiota; gut-lung axis; immunotherapy; lung cancer.

PubMed Disclaimer

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Figures

Figure 1
Figure 1
Major routes of communications within the gut–lung axis. (1) Ruminococcus gnavus stimulates secretion of IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) by colon tissues, those cytokines activate DCs and ILC2 to produce cytokines IL-4, IL-5, and IL-13 and travel through the bloodstream to the lungs and lead to infiltration of the lung parenchyma by eosinophils and mast cells. (2) SCFAs transform macrophage and DC progenitors (MDPs) and their commitment into Ly6C− monocytes in the bone marrow, which can differentiate into alternatively activated macrophages (AAMs) in the lungs, thus control the immunopathology caused by infiltrating neutrophils.
Figure 2
Figure 2
Gut dysbiosis regulates lung inflammation and immunity. (1) Gut dysbiosis causes damage to the intestinal mucosal barrier, invading gut bacteria and metabolites affect the host’s inflammation and immunity locally and systematically which in turn leads to the carcinogensis of lung cancer. Bile acid (BA) stimulates inflammatory markers such as IL-1β、IL-6 and IL-8 in the lung. (2) 12,13-diHOME decreases the number of regulatory T (Treg) cells in the lungs resulting in a reduced anti-inflammatory effect. (3) Bacteria-derived antigens activates TLR4 in the intestine immune cells, which increases the level of IL-1β in peripheral circulation that activates NF-κB pathway, leading to pulmonary inflammation. (4) SFB-induced gut Th17 cells are preferentially recruited to lung due to robust expression in the lung of CCL20. (5) Gut dysbiosis leads to a decrease in the expression of ICAM-1, which decreases the number of activated and effective CD8+ T cells in tumors.

Similar articles

Cited by

References

    1. Zhuang H, Cheng L, Wang Y, Zhang Y-K, Zhao M-F, Liang G-D, et al. . Dysbiosis of the Gut Microbiome in Lung Cancer. Front Cell Infect Microbiol (2019) 9:112. 10.3389/fcimb.2019.00112 - DOI - PMC - PubMed
    1. Yachida S, Mizutani S, Shiroma H, Shiba S, Nakajima T, Sakamoto T, et al. . Metagenomic and Metabolomic Analyses Reveal Distinct Stage-Specific Phenotypes of the Gut Microbiota in Colorectal Cancer. Nat Med (2019) 25(6):968–76. 10.1038/s41591-019-0458-7 - DOI - PubMed
    1. Franzosa EA, Sirota-Madi A, Avila-Pacheco J, Fornelos N, Haiser HJ, Reinker S, et al. . Gut Microbiome Structure and Metabolic Activity in Inflammatory Bowel Disease. Nat Microbiol (2019) 4(2):293–305. 10.1038/s41564-018-0306-4 - DOI - PMC - PubMed
    1. Virtue AT, Mccright SJ, Wright JM, Jimenez MT, Mowel WK, Kotzin JJ, et al. . The Gut Microbiota Regulates White Adipose Tissue Inflammation and Obesity via a Family of microRNAs. Sci Transl Med (2019) 11(496):eaav1892. 10.1126/scitranslmed.aav1892 - DOI - PMC - PubMed
    1. Yang JJ, Yu D, Xiang Y-B, Blot W, White E, Robien K, et al. . Association of Dietary Fiber and Yogurt Consumption With Lung Cancer Risk: A Pooled Analysis. JAMA Oncol (2020) 6(2):e194107. 10.1001/jamaoncol.2019.4107 - DOI - PMC - PubMed

LinkOut - more resources