Opinion - (2025) Volume 9, Issue 1
Received: 01-Feb-2025, Manuscript No. jmbp-25-168771;
Editor assigned: 03-Feb-2025, Pre QC No. P-168771;
Reviewed: 15-Feb-2025, QC No. Q-168771;
Revised: 20-Feb-2025, Manuscript No. R-168771;
Published:
27-Feb-2025
, DOI: 10.37421/2684-4931.2025.9.246
Citation: Bordi, Emborg. “Microbiome-driven Modulation of the Tumor Microenvironment.” J Microbiol Patho 9 (2025): 246.
Copyright: © 2025 Bordi E. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited.
The tumor microenvironment is a highly heterogeneous and dynamic milieu comprising not only malignant cells but also fibroblasts, endothelial cells, immune cells (including T cells, macrophages, dendritic cells, and myeloid-derived suppressor cells), cytokines, chemokines, and extracellular matrix components. These constituents interact in a symbiotic yet often immunosuppressive fashion that supports tumor survival and growth. Recent studies reveal that microbial signals-particularly those from the gut microbiome-profoundly influence TME composition, behavior, and immunogenicity, extending the concept of the "host factor" to include the microbial community [3].
Microbial metabolites, such as short-chain fatty acids- notably butyrate, acetate, and propionate-are key mediators of microbiome-TME interactions. These metabolites can affect the epigenetic landscape of tumor and immune cells by modulating histone acetylation, DNA methylation, and gene expression. Butyrate, for example, is known to act as a histone deacetylase inhibitor, enhancing the expression of genes involved in T-cell effector function and tumor cell apoptosis. In colorectal cancer, butyrate has been shown to inhibit tumor proliferation and promote antitumor immunity through regulatory effects on T-cell differentiation and macrophage polarization [4].
The microbiome also influences the vascular and metabolic components of the TME. By modulating host metabolism, microbial communities can alter nutrient availability, hypoxia responses, and angiogenesis. Certain gut microbes produce polyamines and nitric oxide, which can enhance tumor vascularization, while others influence hypoxia-inducible factor signaling pathways. Furthermore, microbial regulation of systemic glucose and lipid metabolism affects the metabolic programming of tumor-associated immune cells, influencing their function and polarization. One of the most clinically significant aspects of microbiome-TME interaction is its impact on cancer therapy, particularly immunotherapy. The efficacy of immune checkpoint inhibitors, such as anti-PD-1 and anti-CTLA-4 antibodies, has been shown to depend in part on the composition of the gut microbiota. Pioneering studies demonstrated that patients with a higher abundance of Akkermansia muciniphila, Bifidobacterium spp., or Faecalibacterium prausnitzii in their gut had improved responses to ICIs. Mechanistically, these microbes enhance dendritic cell activity, promote effector T-cell recruitment to the tumor, and upregulate MHC class I expression on tumor cells. In contrast, antibiotic use or gut microbial dysbiosis prior to or during treatment is associated with poorer outcomes [5].
Google Scholar Cross Ref Indexed at
Google Scholar Cross Ref Indexed at
Google Scholar Cross Ref Indexed at
Google Scholar Cross Ref Indexed at
Journal of Microbiology and Pathology received 18 citations as per Google Scholar report