Introduction
The intratumoral microbiota is a dynamic ecosystem of bacteria, fungi, and viruses in tumor tissues, and it is increasingly recognized as a significant factor in cancer progression and therapeutic outcomes. Emerging research highlights the dual impact of the intratumoral microbiota on anti-tumor immunity, similar to the gut microbiome.[1,2] Specific microbial communities can activate immune responses, driving inflammation and promoting cytotoxic immune cell infiltration, enhancing the efficacy of immunotherapy. In contrast, other microbial populations can create an immunosuppressive environment that is not conducive to tumor immunotherapy. Understanding these mechanisms is vital for improving current therapies and developing new strategies. The complex interplay between microbiota, the tumor microenvironment, and the host immune system offers potential for personalized, more effective cancer immunotherapy. The complex interplay between microbiota, the tumor microenvironment, and the host immune system offers potential for personalized, more effective cancer immunotherapy (Figure 1).

Mechanisms of intratumoral microbiota in orchestrating cancer immune response. Intratumoral microbiota can either enhance anti-tumor immunity (e.g., via STING activation, T/NK cell activation, antigen presentation) or suppress anti-tumor immunity (e.g., anti-inflammatory response, ROS production, T cell inhibition).
The promotive role of intratumoral microbiota in anti-tumor immunity
Intratumoral microbiota might modulate anti-tumor immunity in many ways, including activating the STING and PERK pathways, promoting lymph node maturation, and enhancing cellular immunity mediated by bacterial peptides (References and citations are displayed in Supplementary Table 1). Shi et al. showed that treating Bifidobacterium by oral route promotes anti-CD47 immunotherapy through the STING pathway in mice. Bifidobacterium activated the STING pathway in dendritic cells, thereby enhancing the production of type I interferon for further stimulation of anti-tumor responses mediated by CD8+ T cells in combination with anti-CD47 therapy. Moreover, Akkermansia muciniphila contributes to the interferon (IFN)-I-natural killer axis by induction the STING pathway by producing cyclic dimeric adenosine monophosphate (di-AMP). Such activation releases IFN-I, chemokines CXCL10 and CCL5, and cytokines interleukin-15 (IL-15)/IL-15R and IL-18, thereby allowing crosstalk of NK cells and dendritic cells for reprogramming of macrophages for an improved therapeutic outcome in melanoma patients.
Beyond STING pathway activation, intratumoral bacteria can regulate other immune pathways. In triple-negative breast cancer, Clostridiales-derived trimethylamine oxide (TMAO) , also an important metabolite within the gut microbiota, induces tumor cell pyroptosis via the PERK pathway.[3] This process, characterized by increased cleaved caspase-3 and gasdermin E (GSDME), activates CD8+ T cell-mediated anti-tumor immunity. Combining intratumoral TMAO injection with systemic anti-PD-1 therapy further enhances anti-tumor effects macroscopically. Moreover, intratumoral bacteria can influence the formation of lymphoid structures. In Helicobacter hepaticus-colonized colorectal cancer (CRC), increased T follicular helper (Tfh) cells promoted the maturation of tertiary lymphoid structures (TLS) near the tumor, enhancing lymphatic transport and activating Tfh cells. This resulted in significant shrinkage of the tumor in H. hepaticus-colonized mice. Lastly, tumor immunity is contributed to through bacterial peptide presentation. Kalaora et al. identified immunogenic bacterial peptides derived from Staphylococcus caprae and Actinomyces odontolyticus within melanoma cells. These peptides, presented on MHC class I and II molecules, activate T-cell responses, pointing out bacterial peptide presentation’s role in stimulating cellular immunity.
The suppressive role of intratumoral microbiota in anti-tumor immunity
Despite these beneficial roles, the intratumoral microbiota can also hinder anti-tumor immunity and reduce cancer immunotherapy efficacy (Table 1). Targeting these detrimental components offers a strategy to enhance treatment responses. When mucosal barriers are compromised, Bacteroides fragilis and Fusobacterium drive intestinal tumorigenesis by generating reactive oxygen species (ROS), which regulate localized inflammation but can impair T cell function and anti-tumor responses.
Similarly, Nevskia ramosa and S. aureus promote regulatory T cell (Treg) activation, facilitating prostate and liver cancer progression. Additionally, Aspergillus sydowii enhances lung tumor growth by IL-1β-dependent activation of myeloid-derived suppressor cells, which inhibit cytotoxic T cell function and increase PD-1-expressing CD8+ T cells.
Chronic antigen exposure and a microbial metabolite-augmented immunosuppressive milieu can lead to T cell exhaustion, characterized by loss of effector function and increased PD-1 expression. This exhausted state reduces T cell responsiveness to immunotherapies like checkpoint inhibitors. In pancreatic ductal adenocarcinoma (PDAC), the microbiome promotes immune tolerance by modulating tumor-associated macrophages (TAMs) via TLR signaling, fostering an immunosuppressive environment.[4] Recent studies show that PDAC-derived tryptophan metabolites activate the aryl hydrocarbon receptor (AhR) in macrophages, promoting tumor-supportive polarization, reducing inflammatory T-cell infiltration, and enhancing PDAC growth.[5]
The intratumoral microbiota can induce anti-inflammatory properties and T-cell dysfunction, reducing anti-tumor immune responses. In PDAC, intratumoral fungi stimulate IL-33 secretion, recruiting T helper 2 (Th2) and innate lymphoid cells (ILC2). Antifungal therapy has been shown to contribute to PDAC regression, offering a novel treatment strategy. Fusobacterium nucleatum binds CRC cells via FadA, inducing IL-6, IL-8, and IL-18, while Escherichia coli virulence factor CNF1 triggers IL-6 and TNF-α, sustaining chronic inflammation that promotes tumor progression.[6] Intratumoral bacteria also stimulate myeloid cells to produce MyD88-dependent IL-1β and IL-23, activating γδT cells to secrete IL-17, which promotes B cell infiltration and tumor progression. In breast cancer, F. nucleatum correlates negatively with CD3+ T cell infiltration, inhibiting T-cell aggregation and promoting TAM and fibroblast recruitment.[7] A new tellurium-containing polycarbonate with cisplatin (PTE@ CDDP) inhibits F. nucleatum by disrupting its membrane integrity, reducing its load in colorectal cancer, and reversing inflammation and intestinal barrier damage. PTE@CDDP enhances cisplatin bioavailability, enables ROS-responsive drug release, and overcomes chemotherapy resistance. This antimicrobial polymer-based drug delivery approach offers a promising strategy for synergistic immunotherapy by modulating the tumor microenvironment and mitigating microbial-induced immunosuppression.[8]
Future perspectives in targeting the intratumoral microbiota
The interplay between intratumoral microbiota and cancer immunotherapy offers new therapeutic strategies. Future strategies could involve using targeted antibiotics, engineered probiotics, RNA vaccines, or bacteria as potential approaches to precisely modulate the tumor microenvironment.[9] Advanced delivery systems, like nanoparticles triggered by microbial signals, may enhance precision.[10] Identifying microbiome-based biomarkers and targeting microbial-host interactions could refine immune responses. Artificial intelligence may further clarify complex microbiome-tumor-immune interactions. Addressing specificity and off-target effects across tumor heterogeneity is essential to realize the potential of microbiome-based cancer therapies.
Supplementary Information
Supplementary materials are only available at the official site of the journal (www.intern-med.com).
Funding statement: This research was funded by the Natural Science Foundation of Hunan Province (NO. 2023JJ30524).
Acknowledgements
None.
-
Author Contributions
Zou YT: Conceptualization, Data interpretation, Writing—Original draft, Writing—Review and Editing. Zhang HQ: Literature search, Data interpretation, Writing—Original draft. Liu F: Literature search, Writing—Original draft. Chen ZS: Writing—Original draft. Tang HL: Conceptualization, Writing—Review and Editing. All authors read and approve the manuscript.
-
Ethical Approval
Not applicable.
-
Informed Consent
Not applicable.
-
Conflict of Interest
The authors declare no conflicts of interest.
-
Data Availability Statement
No additional data is available.
References
1 Xie J, Liu M, Deng X, Tang Y, Zheng S, Ou X, et al. Gut microbiota reshapes cancer immunotherapy efficacy: Mechanisms and therapeutic strategies. Imeta 2024;3:e156.10.1002/imt2.156Search in Google Scholar PubMed PubMed Central
2 Zhang H, Wang X, Zhang J, He Y, Yang X, Nie Y, et al. Crosstalk between gut microbiota and gut resident macrophages in inflammatory bowel disease. J Transl Int Med 2023;11:382–392.10.2478/jtim-2023-0123Search in Google Scholar PubMed PubMed Central
3 Feng X, Deng M, Zhang L, Pan Q. Impact of gut microbiota and associated mechanisms on postprandial glucose levels in patients with diabetes. J Transl Int Med 2023;11:363–371.10.2478/jtim-2023-0116Search in Google Scholar PubMed PubMed Central
4 Gautam SK, Batra SK, Jain M. Molecular and metabolic regulation of immunosuppression in metastatic pancreatic ductal adenocarcinoma. Mol Cancer 2023;22:118.10.1186/s12943-023-01813-ySearch in Google Scholar PubMed PubMed Central
5 Hezaveh K, Shinde RS, Klötgen A, Halaby MJ, Lamorte S, Ciudad MT, et al. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 2022;55:324–340.10.1016/j.immuni.2022.01.006Search in Google Scholar PubMed PubMed Central
6 Fan S, Zhang W, Zhou L, Wang D, Tang D. Potential role of the intratumoral microbiota in colorectal cancer immunotherapy. Int Immunopharmacol 2024;137:112537.10.1016/j.intimp.2024.112537Search in Google Scholar PubMed
7 Parhi L, Alon-Maimon T, Sol A, Nejman D, Shhadeh A, Fainsod-Levi T, et al. Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat Commun 2020;11:3259.10.1038/s41467-020-16967-2Search in Google Scholar PubMed PubMed Central
8 Hu J, Ran S, Huang Z, Liu Y, Hu H, Zhou Y, et al. Antibacterial tellurium-containing polycarbonate drug carriers to eliminate intratumor bacteria for synergetic chemotherapy against colorectal cancer. Acta Biomater 2024;185:323–335.10.1016/j.actbio.2024.06.042Search in Google Scholar PubMed
9 Xie J, Ye F, Deng X, Tang Y, Liang JY, Huang X, et al. Circular RNA: A promising new star of vaccine. J Transl Int Med 2023;11:372–381.10.2478/jtim-2023-0122Search in Google Scholar PubMed PubMed Central
10 Gao C, Wang X, Yang B, Yuan W, Huang W, Wu G, et al. Synergistic Target of Intratumoral Microbiome and Tumor by Metronidazole-Fluorouridine Nanoparticles. ACS Nano 2023;17:7335–7351.10.1021/acsnano.2c11305Search in Google Scholar PubMed
© 2024 Yutian Zou, Hanqi Zhang, Feng Liu, Zhe-Sheng Chen, Hailin Tang, published by De Gruyter on behalf of the SMP
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Perspective
- Prospects for the application of artificial intelligence in geriatrics
- Artificial intelligence for disease X: Progress and challenges
- Secondary prevention for intracranial atherosclerotic stenosis: Where we stand and challenges ahead
- Intratumoral microbiota in orchestrating cancer immunotherapy response
- Review Article
- Gene therapy in polycystic kidney disease: A promising future
- Original Article
- A Notch signaling pathway-related gene signature: Characterizing the immune microenvironment and predicting prognosis in hepatocellular carcinoma
- Development and validation of web-based risk score predicting prognostic nomograms for elderly patients with primary colorectal lymphoma: A population-based study
- Association of life’s essential 8 with prevalence and all-cause mortality of chronic kidney disease among US adults: Results from the National Health and Nutrition Examination Survey (2015–2018)
- Selective HDAC8 inhibition by PCI-34051 attenuates inflammation and airway remodeling in asthma via miR-381-3p-TGFβ3 axis
- Computational analyses to reveal the key determinants of the high malignancy level of cholangiocarcinoma
- Letter to Editor
- Effectiveness and safety of Qixuekang Oral Liquid on vascular health
Articles in the same Issue
- Perspective
- Prospects for the application of artificial intelligence in geriatrics
- Artificial intelligence for disease X: Progress and challenges
- Secondary prevention for intracranial atherosclerotic stenosis: Where we stand and challenges ahead
- Intratumoral microbiota in orchestrating cancer immunotherapy response
- Review Article
- Gene therapy in polycystic kidney disease: A promising future
- Original Article
- A Notch signaling pathway-related gene signature: Characterizing the immune microenvironment and predicting prognosis in hepatocellular carcinoma
- Development and validation of web-based risk score predicting prognostic nomograms for elderly patients with primary colorectal lymphoma: A population-based study
- Association of life’s essential 8 with prevalence and all-cause mortality of chronic kidney disease among US adults: Results from the National Health and Nutrition Examination Survey (2015–2018)
- Selective HDAC8 inhibition by PCI-34051 attenuates inflammation and airway remodeling in asthma via miR-381-3p-TGFβ3 axis
- Computational analyses to reveal the key determinants of the high malignancy level of cholangiocarcinoma
- Letter to Editor
- Effectiveness and safety of Qixuekang Oral Liquid on vascular health