Deciphering the Role of Intratumoral Microbiota in Breast Cancer: Pathogenesis to Precision Therapeutics

Introduction

Breast cancer persists as the most frequently diagnosed malignancy among women globally, with over 2.4 million new cases annually and substantial mortality. Despite advancements in screening and multimodality treatment, prognosis for advanced and metastatic disease remains unsatisfactory. Notably, triple-negative breast cancer (TNBC) exemplifies the clinical challenge, given its aggressive nature and poor 5-year survival rates below 50%. Recent progress reveals that the tumor microenvironment harbors a diverse intratumoral microbiota comprising bacteria, viruses, and fungi, which critically influence tumor biology and therapeutic outcomes. Understanding this tumor-resident microbial ecosystem holds promise for novel diagnostic biomarkers and therapeutics, ushering precision medicine advancements in breast cancer management.

Intratumoral Microbiota Characteristics and Origins

Intratumoral microbiota in breast cancer constitute complex, dynamic communities that differ markedly by tumor subtype, spatial distribution, and patient demographics. High-throughput sequencing identifies predominant bacterial phyla including Proteobacteria, Firmicutes, Actinobacteria, and Bacteroidetes, with key genera such as Fusobacterium, Staphylococcus, and Streptococcus enriched variably across hormone receptor (HR), HER2, and TNBC subtypes. Viruses like HPV and EBV also contribute. Spatial heterogeneity within tumors influences local immune modulation and treatment responses. Temporal microbial shifts accompany tumor progression and treatment, necessitating longitudinal profiling. Intratumoral microbes derive from several routes: translocation from gut microbiota via compromised mucosal barriers (the gut-mammary axis), migration from adjacent skin or nipple-areolar complex via the ductal network, and hematogenous dissemination from oral pathogens during bacteremia.

Biological Mechanisms Mediated by Intratumoral Microbiota

Intratumoral microbiota actively participate in breast cancer pathogenesis by fostering chronic inflammation through cytokine dysregulation (e.g., IL-1β, TNF-α), modulating immune cell populations that encourage immunosuppression (via MDSCs and Tregs), and provoking tumorigenesis. Bacterial toxins (e.g., colibactin, FadA adhesin from Fusobacterium nucleatum) induce DNA damage by triggering pathways such as E-cadherin/β-catenin and CHK2 upregulation, accelerating genomic instability. These microbes further shape the tumor microenvironment by producing metabolites like short-chain fatty acids (SCFAs) and lactic acid, influencing immune tolerance and tumor proliferation.

Importantly, intratumoral microbiota facilitate metastatic processes by promoting pre-metastatic niche formation through intestinal vascular barrier disruption, enhancing cancer cell migration via cytoskeletal reprogramming (RhoA/Rock pathway), stimulating epithelial-mesenchymal transition (EMT), and altering stromal signaling. The presence of fungal species such as Candida albicans contributes to angiogenesis through VEGF induction and immunosuppression via lactic acid accumulation. Moreover, microbial enzymatic activities (β-glucuronidase) influence estrogen metabolism, impacting hormone-driven breast cancer growth.

Impact on Drug Resistance and Epigenetic Modifications

Chemoresistance poses a significant barrier to effective breast cancer therapy. Fusobacterium nucleatum exemplifies microbiota-induced resistance by activating autophagy through TLR4/MyD88 and microRNA-mediated pathways, protecting tumor cells from apoptosis prompted by agents like paclitaxel and oxaliplatin. Microbial enzymes degrade chemotherapeutics, reducing intratumoral drug concentrations and efficacy. Additionally, microbial shifts during chemotherapy can provoke adverse effects such as neuropathic pain via elevated deoxycholic acid (DCA) production interacting with bile acid receptors.

Intratumoral microbiota also affect epigenetic regulation. Microbial metabolites including butyrate modulate histone acetylation by inhibiting histone deacetylases (HDACs), affecting gene transcription relevant to carcinogenesis. Distinct correlations exist between microbial species and miRNA expression profiles linked to metastatic progression, suggesting microbiome-epigenome crosstalk as a potential therapeutic target.

Clinical Applications of Intratumoral Microbiota in Breast Cancer

The distinct microbial signatures between malignant and benign breast tissues, alongside their association with molecular subtypes and prognostic parameters, endorse intratumoral microbiota as promising diagnostic and prognostic biomarkers. Classification models based on bacterial profiles demonstrate high accuracy in distinguishing tumor tissue, and detection of microbial DNA (e.g., Fusobacterium nucleatum) in plasma offers minimally invasive early detection tools with superior performance compared to traditional markers.

Therapeutically, targeting intratumoral microbiota enhances chemotherapy efficacy and mitigates side effects. Antibiotic adjuncts can improve chemosensitivity by diminishing microbial-induced resistance, as demonstrated by increased paclitaxel effectiveness upon Fusobacterium clearance. Probiotic and synbiotic supplementation during chemotherapy have shown potential in reducing treatment-related gastrointestinal symptoms and systemic metabolic disturbances.

Intratumoral microbiota also modulate immunotherapy responses. Certain bacteria, such as Bifidobacterium species, potentiate anti-PD-1/PD-L1 efficacy by stimulating CD8+ T cell activation and the STING signaling pathway. Bacterial extracellular vesicles engineered as nanoplatforms can transform tumor microbes into immune adjuvants, enhancing immunotherapy effectiveness and suppressing metastasis, particularly in TNBC.

Emerging Therapeutic Strategies and Future Directions

Innovative approaches targeting intratumoral microbiota encompass precision antibiotics, engineered probiotic formulations tailored to tumor microbiomes, and bacteriophage therapies delivering chemotherapeutics or selectively eliminating pathogenic microbes. Genetically modified bacteria that localize to tumor sites can serve both for direct antitumor activity and modulation of the tumor immune microenvironment.

Microbial metabolism regulation via dietary interventions or metabolite supplementation, such as SCFAs or trimethylamine N-oxide (TMAO), offers additional avenues to enhance antitumor immunity and inhibit tumor growth. Ongoing clinical trials incorporating microbiota-based interventions and biomarker development are expanding the translational potential of this field.

Challenges remain including the low abundance and contamination risks during microbiome sampling, patient heterogeneity, and safety considerations of microbial therapeutics. Integration of multi-omics, advanced spatial analyses, and artificial intelligence will greatly enhance mechanistic insights and personalization of treatments.

Conclusion

Intratumoral microbiota have emerged as vital modulators of breast cancer biology, impacting tumor initiation, progression, metastasis, and therapeutic response. Their remarkable diversity and dynamic interplay with host pathways present unprecedented opportunities for novel biomarkers and targeted therapies. With advancing technologies and clinical validation, microbiome-guided precision oncology is poised to transform breast cancer diagnosis and management, ultimately improving patient outcomes.

References

1. Bray F et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide. CA Cancer J Clin. 2024;74(3):229–263.
2. Li Y et al. Recent advances in therapeutic strategies for triple-negative breast cancer. J Hematol Oncol. 2022;15(1):121.
3. Xiong X et al. Breast cancer: pathogenesis and treatments. Signal Transduct Target Ther. 2025;10(1):49.

126. Peng F et al. Intratumoral microbiota as a target for advanced cancer therapeutics. Adv Mater. 2024;36(38):e2405331.
photo
photo
photo

Comments

No comments yet. Why don’t you start the discussion?

Leave a Reply

Your email address will not be published. Required fields are marked *