Skip to main content
NPJ Biofilms and Microbiomes logoLink to NPJ Biofilms and Microbiomes
. 2025 Sep 30;11:190. doi: 10.1038/s41522-025-00818-3

Harnessing gut microbiota for colorectal cancer therapy: from clinical insights to therapeutic innovations

Chaofan Chen 1, Qi Su 2, Min Zi 3, Xiaokun Hua 1,, Zhiyun Zhang 1,
PMCID: PMC12485087  PMID: 41028744

Abstract

Colorectal cancer (CRC) remains a leading cause of cancer morbidity and mortality worldwide, yet improvements in survival have been modest despite advances in conventional therapies. The gut microbiota has emerged as a critical player in CRC pathogenesis and a promising therapeutic target to enhance clinical outcomes. Mounting evidence implicates specific microorganisms, notably Escherichia coli, Fusobacterium nucleatum, and Bacteroides fragilis, in tumor initiation and progression through DNA damage, inflammatory modulation, and immunosuppressive mechanisms. Clinical trials investigating microbiome modulators—including faecal microbiota transplantation, probiotics, prebiotics, and engineered biotherapeutics—highlight their potential to augment chemotherapy, radiotherapy, immunotherapy, and surgical recovery, with encouraging preliminary efficacy in treatment-resistant CRC subtypes. Nonetheless, translating microbiome interventions into standardized clinical practice requires rigorous mechanistic validation, robust biomarker development, and careful management of safety concerns. Future research must focus on integrating high-resolution multi-omics, spatial microbiome mapping, artificial intelligence analytics, and innovative microbiome-targeted nanotechnologies to precisely reshape gut microbial communities, thereby ushering in a new era of precision oncology in colorectal cancer management.

Subject terms: Cellular microbiology, Clinical microbiology

Introduction

Colorectal cancer (CRC) is the third most common cancer worldwide, with over 1.9 million new cases and 900,000 deaths reported in 20221. The USA and China exhibit the highest incidence rates, followed by Japan, Russia, India, Germany, Brazil, the UK, Italy, and France2. Although CRC rates are higher in Western nations, they are rapidly increasing in developing countries. Early-onset CRC, diagnosed at age 50 or younger, has also emerged as a significant global concern3.

CRC is classified based on its spread: stage I or II when limited to the primary tumor, stage III when it involves regional lymph nodes, and stage IV when it metastasizes to distant sites. Treatment for stages I–III typically involves surgical resection, with stage III often requiring adjuvant chemotherapy. In metastatic CRC (stage IV), systemic therapies, including chemotherapy, targeted therapy, immunotherapy, or their combinations, are commonly employed4. Despite advances like laparoscopic surgery, overall cure rates and long-term survival have improved only modestly over recent decades5.

Since 2004, adjuvant chemotherapy for stage III colon cancer has generally included oxaliplatin and a fluoropyrimidine6. However, only 20% of patients benefit from this regimen, leaving 80% exposed to unnecessary toxicity7. Fluoropyrimidines are metabolized by dihydropyrimidine dehydrogenase (DPYD); variants in the DPYD gene can lead to truncated proteins, prolonged drug exposure, and increased toxicity. Treatment-related mortality occurs in 0.1% of patients without DPYD variants but rises to 2.3% in those with such variants8. Additional side effects include oxaliplatin-induced neuropathy and chemotherapy-associated diarrhea6. Targeted therapies, which address specific molecular characteristics, have become vital, although drug resistance remains a significant challenge9.

Immune checkpoint blockade (ICB) has transformed cancer therapy, with pembrolizumab approved as first-line treatment for the subset of CRC patients whose tumors exhibit microsatellite-instability-high (MSI-H) owing to their elevated mutational burden and neoantigen load10,11. However, approximately 85% of CRCs are proficient in mismatch repair (pMMR) and display microsatellite-instability-low (MSI-L) or microsatellite-stable (MSS) status. These pMMR-MSI-L/MSS “cold” tumors—characterized by low tumor mutation burden, scarce neoantigen presentation, minimal CD8⁺ T-cell infiltration, and activation of immunosuppressive pathways (e.g., WNT/β-catenin and TGF-β)—are largely resistant to PD-1/PD-L1 blockade12. Despite advances in surgery and adjuvant chemotherapy, long-term survival gains have been modest, underscoring a pressing need for effective adjunctive therapies to improve CRC outcomes—especially in MSS CRC.

Given the limitations of current CRC therapies—including chemoresistance, immunotherapy resistance, radiotoxicity, and post-surgical complications—there is growing interest in the gut microbiota as a complementary therapeutic target. Rather than acting independently, microbial interventions may enhance the efficacy and reduce the toxicity of chemotherapy, immunotherapy, radiotherapy, and surgery. For instance, specific gut commensals like Roseburia intestinalis have been shown to sensitize CRC cells to radiotherapy by promoting radiation-induced autophagy13, while perioperative microbiota composition has been linked to postoperative recovery and long-term complications following CRC resection14. As mechanistic understanding deepens, integrating microbiota-based strategies into established treatment regimens offers a promising path to improved outcomes. This Review first explores the role of gut microbiota in CRC pathogenesis, followed by an examination of their therapeutic relevance across immunotherapy, radiotherapy, and surgical management.

Role of gut microbial communities in CRC

Extending from the foundational roles of the gut microbiome in maintaining host metabolic, immune, and epithelial homeostasis, accumulating evidence implicates its disruption—termed gut dysbiosis—as a contributing factor in CRC1517. Over the past decade, advances in metagenomic sequencing have enabled large-scale comparisons between CRC patients and healthy individuals, aimed at identifying microbial patterns that distinguish cancer-associated from normative gut communities. However, this endeavor is challenged by the intrinsic variability of the microbiota among healthy individuals, as well as by population-level heterogeneity driven by diet, lifestyle, geography, and antibiotic exposure18,19. Moreover, technical inconsistencies in sample collection, preservation, and DNA extraction further complicate cross-study comparisons20.

Despite these limitations, meta-analyses integrating data across diverse cohorts and methodologies have revealed consistent microbial alterations in CRC4. These include enrichment of oral-origin taxa—such as Fusobacterium, Porphyromonas, Parvimonas, and Peptostreptococcus—and depletion of beneficial butyrate-producing genera including Faecalibacterium and Roseburia21,22. Microbial profiles also appear to vary with tumor location and molecular subtype23, although reports of stage-specific taxa enrichment have shown poor reproducibility across studies24. Early evidence established associations between tumor microbiota and CRC molecular subtypes. CMS1 tumors—typically right-sided, MSI-H, and immunologically active—were shown to be enriched for oral pathogens including Fusobacterium nucleatum, Parvimonas micra, and Porphyromonas gingivalis, alongside upregulation of interferon-γ and TNF signaling25. Building on this, a recent large-scale analysis identified three oncomicrobial community subtypes (OCS1–3) with distinct taxonomic and clinicomolecular profiles. OCS1, dominated by Fusobacterium, corresponded to CMS1 tumors and was associated with CIMP-positivity, BRAF^V600E mutations, and MSI-related mutational signatures. OCS2 and OCS3 aligned with left-sided, CIN-type tumors and distinct microbial ecologies. Among patients with MSS disease, OCS1 and OCS3 subtypes were associated with worse prognosis. These data underscore the alignment between microbial composition and tumor phenotype, reflecting the biological heterogeneity of CRC23. Functionally, CRC-associated microbiota exhibit increased abundance of amino acid fermentation and polyamine biosynthesis pathways (e.g., arginine and ornithine metabolism), alongside reduced carbohydrate degradation capacity21. These shifts have been linked to altered production of microbial metabolites such as secondary bile acids and short-chain fatty acids (SCFAs), which have been implicated in modulating tumor-promoting inflammation, DNA damage, and epithelial proliferation. However, whether such dysbiotic shifts actively drive tumorigenesis or merely reflect adaptive responses to the neoplastic microenvironment remains unclear, underscoring the need for mechanistic studies in well-controlled experimental models to delineate causal microbial contributors from bystander taxa4.

In addition, the gut virome, mycobiome, and archaeome remain underexplored, though emerging evidence suggests roles in tumor immunity and microbial synergy26,27. To advance the field, it will be essential to move beyond descriptive and correlative studies toward mechanistic investigations that delineate the functional contributions of intratumoural microorganisms to tumor progression, metastasis, and immune regulation.

Role of specific microorganisms in CRC pathogenesis

The connection between specific microorganisms and CRC was first identified over 50 years ago with the discovery that Streptococcus gallolyticus subspecies gallolyticus (Sgg) frequently colonizes patients with colorectal neoplasia28. Dysbiosis, increasingly defined as a weakening of host functions that regulate microbial growth by controlling resource availability, is frequently observed in CRC29,30. This dysbiotic state is often characterized by the overgrowth of specific bacterial taxa31. However, the influence of any individual microorganism must be understood within the broader ecological framework of the gut microbiome, where immunometabolic cues, circadian rhythms, epithelial integrity, and interbacterial interactions collectively shape microbial behavior and host response32. The evidence is particularly strong for three microorganisms—pks⁺ Escherichia coli, Fusobacterium nucleatum, and Bacteroides fragilis—which have been consistently linked to CRC through both mechanistic studies and correlative human data (Fig. 1)4. Although these bacteria are the most well-characterized, data implicating other taxa as potential CRC drivers are accumulating rapidly (Table 1)33. Nevertheless, despite extensive preclinical evidence, no microorganism or group of microorganisms has been definitively established as causative of human CRC, due to the absence of longitudinal studies demonstrating microbial presence preceding tumor onset or showing that microbial eradication reduces cancer risk34.

Fig. 1. Microbiota-associated mechanisms involved in the pathogenesis of colorectal cancer.

Fig. 1

Escherichia coli produces colibactin, causing DNA interstrand cross-links and double-strand breaks with a unique mutational signature. Fusobacterium nucleatum’s Fap2 binds Gal-GalNAc, while FadA engages E-cadherin to activate Wnt–β-catenin and MYC, and its LPS triggers NF-κB–driven proliferation. Formate from F. nucleatum activates AhR, boosting invasiveness and SOX2-dependent stemness. Bacteroides fragilis secretes BFT to cleave E-cadherin and disrupt tight junctions, inducing MYC and proliferation; its LPS via TLR4 upregulates JMJD2B, SOX2, and NANOG to promote stemness. AhR aryl hydrocarbon receptor, BFT B. fragilis toxin, E-cadherin epithelial cadherin, FadA Fusobacterium adhesin A, Fap2 Fusobacterium autotransporter protein 2, Gal-GalNAc galactose-α1,3-N-acetylgalactosamine, JMJD2B jumonji domain-containing protein 2B, LPS lipopolysaccharide, MYC v-myc avian myelocytomatosis viral oncogene homolog, NF-κB nuclear factor kappa-light-chain-enhancer of activated B cells, SOX2 SRY-box transcription factor 2, TLR4 Toll-like receptor 4.

Table 1.

Evidence for microorganisms as drivers of CRC: key bacteria, other bacteria, virome, and fungi

Microorganism Epidemiological evidence Mouse model evidence Type of mouse model Notes
Key Bacterial Escherichia coli Yes36,37,39,43 Yes

APCmin/+38

Zeb2IEC-Tg/+41

The oncogenic potential of pks+ E. coli critically depends on bacterial adhesion to host epithelial cells
Fusobacterium nucleatum Yes44,46 Yes

APCmin/+45

AOM/DSS51

Fn generates abundant butyric acid, which inhibits histone deacetylase 3/8 in CD8+ T cells, inducing Tbx21 promoter H3K27 acetylation and expression. Furthermore, Fn-induced PD-L1 expression by activating STING signaling.
Bacteroides fragilis Yes64 Yes

AOM60

AOM/DSS62,65

BRAF V600E Lgr5 CreMin66

ETBF promoted CRC cell proliferation by downregulating miR-149-3p both in vitro and in vivo.
Other Bacterial Campylobacter Species Yes67,69 Limited Germ-free ApcMin/+68 Potentially relevant human species are C. jejuni and Campylobacter coli; evidence for the involvement of these species in human CRC is not available.
Peptostreptococcus anaerobius Yes71 Limited APCmin/+74,75,77 Its tryptophan metabolite IDA activates the aryl hydrocarbon receptor to induce ALDH1A3 expression and FSP1-mediated CoQ10 reduction, inhibiting ferroptosis via the AHR–ALDH1A3–FSP1–CoQ10 axis.
Clostridioides difficile Yes78,79 Limited APCmin/+80 C. difficile promotes tumorigenesis by activating Wnt/β-catenin signaling, inducing ROS production, and eliciting a procarcinogenic mucosal immune response marked by IL-17 secretion and myeloid infiltration.
Morganella morganii Limited83 Limited AOM83 Small molecules such as indolimines, produced by M. morganii, induce DNA damage and promote tumorigenesis in preclinical models.
Streptococcus gallolyticus subspecies gallolyticus Yes84 Limited AOM85 Sgg may support CRC progression through niche adaptation mechanisms
Akkermansia muciniphila Yes92 Limited APCmin/+87,89,90 A. muciniphila facilitated enrichment of M1-like macrophages in an NLRP3-dependent manner in vivo and in vitro.
virome Yes25,26 Limited Apc+/1638N98 Their direct involvement in tumorigenesis remains inconclusive.
Fungi Yes102104 Limited Limited Despite their lower abundance and high variability among individuals, they also exhibit distinct profiles in CRC patients.

Limited” in the Mouse model evidence column indicates that the microorganism has been investigated in vivo, but the evidence remains preliminary, with limited mechanistic clarity, a lack of consistent tumorigenic outcomes, or insufficient validation across multiple CRC models.

AOM azoxymethane, DSS dextran sodium sulfate, APC^Min/+Apc^Min heterozygous mouse model, Zeb2^IEC-Tg/+ intestinal epithelial cell-specific Zeb2 transgenic mouse, BRAF^V600E Lgr5^CreMinBraf^V600E mutant mouse under Lgr5 promoter, Germ-free Apc^Min/+ germ-free Apc^Min mouse, ETBF enterotoxigenic Bacteroides fragilis, pks+E. coliEscherichia coli strains harboring the pks genomic island encoding colibactin, FnFusobacterium nucleatum, IDA indoleacrylic acid, AHR aryl hydrocarbon receptor, ALDH1A3 aldehyde dehydrogenase 1 family member A3, FSP1 ferroptosis suppressor protein 1, CoQ10 coenzyme Q10, ROS reactive oxygen species, IL-17 interleukin-17, SggStreptococcus gallolyticus subsp. gallolyticus, Apc^+/1638NApc^+/1638N colorectal cancer mouse model.

Key bacterial players in CRC pathogenesis

Escherichia coli

E. coli strains harboring the pks genomic island—denoted pksE. coli—encode the biosynthetic machinery responsible for colibactin production35. Colibactin inflicts DNA double-strand breaks, engendering a spectrum of mutagenic lesions36,37. Accumulating evidence indicates that pksE. coli is overrepresented in CRC and that its colibactin contributes to intestinal tumorigenesis in murine models38. In particular, experiments in CRC-prone mice have shown that colonization with pksE. coli fosters an immunosuppressive tumor microenvironment (TME) by diminishing CD3⁺ and CD8⁺ tumor-infiltrating lymphocytes, thereby impairing responses to anti-PD-1 therapy39. Furthermore, a European cohort study linked seropositivity to both pks⁺ E. coli and enterotoxigenic Bacteroides fragilis (ETBF) with heightened CRC incidence, suggesting a cooperative role in neoplastic initiation40. Recent studies also highlight that the pathogenicity of enteropathogenic E. coli is influenced by its microbial context. In particular, mucin-degrading bacteria such as Akkermansia muciniphila facilitate E. coli encroachment into the epithelial niche by disrupting the mucus barrier, enabling access to host-derived nutrients and enhancing colonization under dietary restriction conditions41.

Building on this, pks⁺ E. coli has been shown to employ two lectin-like adhesins—FimH on type I pili and FmlH on F9 pili—to bind distinct glycan ligands (terminal D-mannose and T/Tn antigens, respectively), thereby orchestrating spatially resolved colonization of the tumor epithelium; deletion of either adhesin markedly attenuates CRC progression in murine models42. Concomitantly, colibactin-induced DNA damage triggers a senescence-associated secretory phenotype that selects for chemotherapy-resistant cancer stem cells, activates epithelial-to-mesenchymal transition, upregulates NANOG and OCT-3/4, and drives metabolic reprogramming with lipid droplet accumulation—further impeding CD8⁺ T-cell infiltration and fostering both chemoresistance and immune evasion43,44.

Fusobacterium nucleatum

Comprehensive metagenomic profiling of 1375 fecal samples from six cohorts confirmed enrichment of Fusobacterium nucleatum (Fn) across all colorectal tumor sites45. Neonatal inoculation of ApcMin/+ mice with the Fn strain Fn7-1 overcomes colonization resistance, triggers colonic Il17a upregulation prior to tumor onset, and accelerates intestinal tumorigenesis46. Within the colon, Fn invades deep gut crypts and, by single-cell transcriptomics, is shown to reprogram epithelial cells and activate LY6A⁺ revival stem cells that hyperproliferate into tumor-initiating cells47. At the molecular level, its FadA adhesin binds E-cadherin to trigger IκBα degradation and NF-κB signaling, while secreted ADP-heptose engages ALPK1 to induce CXCL8 release from epithelial cells48,49. In vivo, Fn or its metabolite formate increases tumor incidence and Th17 expansion, and recruits CX3CR1⁺PD-L1⁺ phagocytes to reshape the microenvironment50,51.

Recent studies have reported conflicting effects of Fn on immune checkpoint therapy outcomes in CRC. In preclinical models of PD-L1 blockade, Fn activated cGAS–STING signaling and increased IFN-γ⁺ CD8⁺ T-cell infiltration, thereby sensitizing tumors to immunotherapy52. Similarly, in microsatellite-stable colorectal cancer (MSS CRC), intratumoral Fn enhanced anti-PD-1 efficacy via butyrate-mediated inhibition of HDAC3/8 in CD8⁺ T cells, leading to TBX21 upregulation and reduced T-cell exhaustion; strains lacking butyrate synthesis lost this benefit, and high intratumoral Fn predicted favorable responses10. Conversely, Fn colonization in the intestinal lumen has been linked to immunotherapy resistance: in metastatic CRC, elevated Fn abundance correlated with succinate production, which suppressed the cGAS–interferon-β axis and impeded CD8⁺ T-cell trafficking; antibiotic depletion using metronidazole reversed these effects and restored PD-1 sensitivity53.

In addition, in KRAS p.G12D-mutant CRC, tumor-colonizing Fn drives tumorigenesis via DHX15-dependent ERK/STAT3 signaling, whereas targeted enrichment of Parabacteroides distasonis can displace Fn and attenuate tumor progression, highlighting the therapeutic promise of microbiota modulation54. Fn also promotes chemoresistance by inhibiting pyroptosis and ferroptosis through BCL2–GSDME and β-catenin–GPX4 pathways55,56. Metformin reverses Fn-induced stemness and drug resistance via MYC/miR-361-5p suppression57. Moreover, the antimicrobial peptide Br-J-I disrupts FadA-mediated adhesion and enhances 5-FU efficacy58. These findings underscore the multifaceted roles of Fn in CRC progression, immune escape, and therapy resistance, and highlight its potential as a microbiota-targeted therapeutic axis.

Bacteroides fragilis

Bacteroides fragilis constitutes approximately 0.1–0.5% of the colonic microbiota, and a subset of strains harbors a pathogenicity island encoding the zinc-dependent metalloproteinase toxin B. fragilis toxin (BFT or fragilysin)59. ETBF is enriched in CRC and drives tumorigenesis by BFT-mediated cleavage of E-cadherin, which activates β-catenin and either NF-κB or Wnt signaling, increases epithelial permeability, and provokes mucosal immune responses, while also promoting cancer-stem-cell traits via TLR4 activation60,61. In vitro and in vivo, ETBF treatment of CRC cell lines (SW480, HCT-116) enhances proliferation, invasion, and migration by downregulating miR-139-3p—an effect reversed by miR-139-3p overexpression—and mechanistically ETBF upregulates HDAC3, which represses miR-139-3p to facilitate malignancy; HDAC3 knockdown or miR-139-3p restoration attenuates ETBF-driven CRC progression and reduces tumor growth in xenografts62. ETBF and BFT disrupt the intestinal barrier—impairing tight junction proteins (MUC2, Occludin, ZO-1), increasing permeability, reducing TEER and villus integrity—and activate the STAT3/ZEB2 axis to drive EMT and SW480 malignancy; these effects, as well as ETBF-exacerbated tumor load and barrier injury in AOM/DSS models, are mitigated by the STAT3 inhibitor Brevilin A or ZEB2 knockdown63. KRAS-mutant CRCs exhibit increased Bacteroides abundance, and mutant KRAS downregulates miR-3655—thereby upregulating SURF6, which inhibits IRF7 nuclear translocation and IFNβ transcription—to promote intratumoral ETBF colonization; targeting the miR-3655/SURF6/IRF7/IFNβ axis restores IFNβ-mediated ETBF clearance and offers a novel therapeutic avenue64. Human studies report higher ETBF prevalence in patients with colorectal neoplasia versus healthy individuals, though the specific stages of neoplasia associated with enrichment vary across cohorts65. Exosomes from ETBF-treated cells augment Th17 differentiation and CRC cell proliferation by downregulating miR-149-3p, indicating that the ETBF/miR-149-3p pathway could be a promising target for treating ETBF-associated intestinal inflammation and CRC66. Preliminary evidence suggests that ETBF may enhance anti-PD-1 immunotherapy by recruiting IFNγ-producing CD8⁺ T cells into tumors, but further investigation is required to define this mechanism and its therapeutic potential67.

Other bacterial taxa implicated in CRC

Beyond well-documented bacteria like E. coli, Fusobacterium nucleatum, and Bacteroides fragilis, recent studies suggest that less-studied bacterial species may also influence CRC development. These findings highlight the intricate and dynamic role of the gut microbiota in CRC, paving the way for further exploration.

Campylobacter species

While primarily associated with gastroenteritis, Campylobacter jejuni has emerged as a potential contributor to intestinal tumorigenesis through the production of cytolethal distending toxin (CDT), a genotoxin capable of causing DNA double-strand breaks68. Experimental evidence in the ApcMin/+ mouse model demonstrates that CDT activity amplifies carcinogenesis69. Interestingly, Campylobacter concisus, an oral pathogen linked to inflammatory bowel disease (IBD), has also been associated with CRC, suggesting broader involvement of the Campylobacter genus in gastrointestinal malignancies70.

Moreover, mechanistic studies reveal that C. jejuni–derived CDT promotes CRC metastasis by activating the JAK2–STAT3–MMP9 axis, enhancing matrix degradation and tumor cell invasiveness in liver and lung models, human colonic explants, and patient-derived tumoroids71. Ablation of cdtB or inhibition with purified CdtB protein abolishes this effect, underscoring intratumoural CDT’s central role in driving metastatic dissemination and its promise as a therapeutic target71.

Peptostreptococcus anaerobius

Peptostreptococcus anaerobius is enriched in CRC and has been implicated in tumorigenesis via diverse72, experimentally supported mechanisms: it engages TLR2 and TLR4 on colonic epithelial cells to increase ROS production, thereby stimulating cholesterol synthesis and cellular proliferation73; its tryptophan metabolite IDA activates the aryl hydrocarbon receptor to induce ALDH1A3 expression and FSP1-mediated CoQ10 reduction, inhibiting ferroptosis via the AHR–ALDH1A3–FSP1–CoQ10 axis74,75. Additionally, Pa remodels the TME by recruiting CXCR2⁺ MDSCs through integrin α2β1–NF-κB–CXCL1 signaling and by secreting lytC_22 to engage Slamf4 on intratumoral MDSCs, which upregulates ARG1 and iNOS, activates STAT3-dependent EMT, and contributes to chemoresistance in CRC7678.

Clostridioides difficile

Traditionally known for its role in antibiotic-associated diarrhea, Clostridioides difficile has been identified as a potential contributor to CRC79. While one epidemiological analysis paradoxically found an inverse association between CDI and CRC incidence80, Sears et al. confirmed in mouse models that chronic colonization by toxigenic C. difficile drives colorectal tumorigenesis via TcdB-dependent activation of Wnt signaling, ROS generation, and pro-tumor mucosal immune responses—marked by myeloid cell activation and infiltration of IL-17–secreting innate lymphocyte81.

Beyond these pathways, recent findings indicate that TcdB can also activate Pyrin inflammasomes in intestinal epithelial cells, triggering ASC oligomer formation and robust inflammatory responses82. This activation disrupts tight junction proteins such as claudins and occludins, thereby impairing epithelial barrier integrity and promoting colitis83. These insights reveal that TcdB not only reprograms epithelial and immune signaling but also structurally compromises the mucosal barrier, collectively contributing to a tumor-favoring microenvironment.

Morganella morganii

Emerging evidence points to Morganella morganii, a gram-negative bacterium enriched in IBD and CRC patients, as a source of genotoxic compounds. Small molecules such as indolimines, produced by M. morganii, induce DNA damage and promote tumorigenesis in preclinical models84. These findings link microbial metabolite production to cancer progression, adding another layer of complexity to the gut microbiota–CRC axis.

Streptococcus gallolyticus subspecies gallolyticus

Sgg is frequently enriched in CRC tissue and strongly associated with colorectal neoplasia in epidemiological studies85. Despite its correlation with CRC, the question remains whether Sgg is a driver or merely a passenger in the TME. Experimental models suggest that Sgg may support CRC progression through niche adaptation mechanisms, such as gallocin production, which inhibits competing enterococci86,87. However, more robust evidence is needed to establish its role in tumor initiation.

Akkermansia muciniphila

Akkermansia muciniphila, a mucin-degrading commensal, exerts multifaceted effects on CRC. Its abundance is typically reduced in CRC patients, and supplementation suppresses tumorigenesis by enriching M1-like macrophages through the TLR2–NF-κB–NLRP3 axis, a mechanism essential for its antitumour efficacy88. It also promotes CD8⁺ T-cell-mediated immunity via outer membrane vesicles containing Amuc_1434, which downregulate PD-L1, and through the acetyltransferase Amuc_2172, which induces H3K14 acetylation and upregulation of HSP70 in cancer cells89,90. Additionally, A. muciniphila impairs tryptophan–AhR–β-catenin signaling, contributing to tumor suppression in both chemical and genetic CRC models91. However, overcolonization can disrupt the mucosal barrier by degrading mucin and reducing tight junction proteins, thereby exacerbating CRC progression92. Furthermore, by thinning the mucus layer, A. muciniphila facilitates nutrient scavenging by pathogenic E. coli, highlighting a cooperative metabolic adaptation under dietary restriction41. Clinically, its depletion is associated with gut dysbiosis and reduced survival in CRC patients, underscoring its potential as both a therapeutic target and prognostic biomarker93.

Klebsiella species

Klebsiella species have been implicated in CRC through multifactorial mechanisms. Oral Klebsiella, enriched during periodontitis, can translocate to the gut and promote CRC via inflammasome activation and Th17 cell recruitment94. In murine models, Klebsiella aerogenes exacerbates colitis-associated tumorigenesis by disrupting the mucosal barrier and inducing inflammatory responses and microbial dysbiosis95. Clinical isolates of Klebsiella pneumoniae from CRC patients often harbor polyketide synthase and siderophore genes, and enhance tumor cell proliferation in vitro96. Mendelian randomization further links Klebsiella abundance to CRC risk via immune modulation, particularly CD4⁺ T-cell pathways97. These findings highlight Klebsiella as a key oral–gut pathobiont in CRC development.

Fungal and viral contributions to CRC

While bacteria dominate the gut microbiota, non-bacterial components such as viruses and fungi also play a role in CRC pathogenesis98. Advances in sequencing technologies and metagenomic analyses have shed light on these understudied microbial kingdoms, revealing their potential contributions through trans-kingdom interactions and dysbiosis.

Virome alterations in CRC patients are marked by shifts in Siphoviridae and Myoviridae populations and an enrichment of Microviridae27. Virome dynamics—predominantly driven by bacteriophages—regulate gut bacterial communities and underpin microbiome stability. In H. pylori–infected Apc^+/1638N mice, an early-stage virome expansion of temperate phages—including those predicted to infect CRC-associated bacteria such as Enterococcus faecalis—was observed. Co-occurrence network analysis revealed strong viral–bacterial associations preceding tumor development, indicating that H. pylori–induced prophage activation and subsequent phage proliferation may promote colorectal carcinogenesis by reshaping the bacterial ecosystem99. Apart from their influence on bacterial ecosystems, Caudovirales-derived phages can breach the epithelial barrier, enter colonic cells, and trigger proinflammatory signaling that enhances tumor growth and invasiveness in CRC models100,101. Functionally, CRC patients’ viromes also display increased fatty acid biosynthesis pathways alongside depletion of viral metabolites such as L-methionine and acetate—compounds known to inhibit CRC cell proliferation and maintain mucosal homeostasis—indicating that virome shifts may directly impact tumor risk26,102.

Fungi, despite their lower abundance and high inter-individual variability, exhibit distinct alterations in CRC, including an increased Basidiomycota-to-Ascomycota ratio and enrichment of highly variable fungi103105. They contribute to tumourigenesis through biofilm formation, secretion of mycotoxins (e.g., aflatoxins, candidalysin), induction of proinflammatory cytokines such as IL-1β and IL-17, and disruption of epithelial barrier integrity—collectively fostering a pro-tumor, inflammation-prone microenvironment106. For example, Candida tropicalis activates the NLRP3 inflammasome in myeloid-derived suppressor cells to drive excessive IL-1β release, which in turn amplifies MDSC-mediated immunosuppression and promotes CRC progression107. Preliminary evidence also suggests that a compound ethanol extract of Ganoderma lucidum spores and Sanghuangporus vaninii may exert anti-CRC effects via the CYP24A1–VDR and TERT–Wnt signaling pathways108. Moreover, commensal bacteria limit fungal overgrowth by producing antifungal toxins and metabolites and by engaging host immune pathways, while fungal–bacterial interactions can themselves elicit unique immune responses109.

Role of microbiome modulators in CRC clinical trials

Current targeted therapies and immunotherapies exhibit limited efficacy due to the immunosuppressive TME and gut dysbiosis-driven drug resistance. Microbiome modulators (MMs), encompassing fecal microbiota transplantation (FMT), probiotics/prebiotics/synbiotics therapy, live biotherapeutic product (LBP), have emerged as paradigm-shifting adjuvants110. These agents restore gut eubiosis, enhance antitumor immunity via multiple mechanisms of action, demonstrating unprecedented therapeutic potentials in pan-cancers111. Herein, to unravel critical gaps in CRC treatment, this study delineates the evolving MM clinical trial landscape while bridging emerging preclinical insights to optimize therapeutic research and development.

We conducted a comprehensive search of the Trialtrove database (https://clinicalintelligence.citeline.com), an authoritative platform that integrates clinical trial data from dozens of countries and regions worldwide, including ClinicalTrials.gov, the WHO International Clinical Trials Registry Platform, and national registries across North America, Europe, and Asia. As of June 30, 2025, we applied the retrieval strategy: (Disease is Oncology: Colorectal) AND (Mechanism of Action is Microbiome modulator) to identify relevant studies. Analytical parameters included trial phase and status distribution, MM types, sponsor classifications, disease stage and line of therapy, primary/secondary endpoints, and combination strategies. Two independent investigators systematically reviewed and verified all retrieved records and analytical results (Fig. 2), including the extraction of unstructured information, the resolution of data gaps, and the minimization of classification bias, to ensure methodological rigor and strict quality control. Any residual limitations following these procedures do not affect the overall interpretation or observed trends of this study.

Fig. 2. The clinical investigation panorama of microbiome modulators in CRC treatment.

Fig. 2

A Trial phase distribution of clinical trials by start years. B Trial status distribution of clinical trials. C Microbiome modulator type distribution of clinical trials. D Sponsor type distribution of clinical trials. E Cancer staging and line of therapy distribution of clinical trials. F Endpoint distribution of clinical trials.

To date, 29 trials were conducted to evaluate MMs in CRC, spanning FMT (n = 11), probiotics/prebiotics therapy (n = 11), and LBP (n = 7) (Table S1/Fig. 2C). Phase II trials dominated in pipelines (n = 9), followed by Phase I (n = 7) and Phase I/II (n = 7) (Fig. 2A). Most of the trials are being planned, concurrently with a considerable proportion of completed trials (Fig. 2B). Academic sponsors dominated in this field with industry-initiated trials followed (Fig. 2D). Later-line therapies of advanced CRC patients are the predominant indication selections for this fresh therapeutic (Fig. 2E). And the primary endpoints focused mainly on the overall response rate (ORR) of this therapeutic strategy in CRC patients currently, with some novel microbiome indicators innovatively applied (Fig. 2F). In addition, combination with CRC standard of care (SOC) regimens represent the mainstream clinical trend (Table S1). More importantly, some trials disclosed the positive outcomes, implying the promising therapeutic potentials of diverse microbiome modulation strategies in CRC treatment.

While the clinical pipeline for MMs in CRC remains predominantly early-phase, selected studies have begun to demonstrate biologically meaningful signals that extend beyond descriptive endpoints. In the RENMIN-215 trial, FMT from healthy donors was combined with the anti-PD-1 antibody tislelizumab and the VEGFR inhibitor fruquintinib in patients with refractory, MSS metastatic CRC. The study reported an objective response rate of 20% and a disease control rate (DCR) of 95%, with a median progression-free survival (mPFS) of 9.6 months and a median overall survival (mOS) of 13.7 months—figures rarely observed in this resistant population112. Notably, responders exhibited increased microbial diversity and sustained microbiota engraftment, implying that gut ecosystem restoration may overcome immunotherapy resistance through modulation of the TME113.

More broadly, meta-analyses across solid tumors indicate that combining FMT with immune checkpoint inhibitors (ICIs) improves objective response rates compared to immunotherapy alone, with pooled ORRs of 43.1% versus 36.9% in early-phase trials114. Mechanistic studies have implicated key commensals—such as Akkermansia muciniphila, Faecalibacterium prausnitzii, and Bacteroides uniformis—in enhancing antitumour immune activation, through increased antigen presentation, interferon-γ production, and modulation of regulatory T cells115.

Despite these promising findings, most MM trials to date have enrolled patients with advanced, heavily pre-treated disease, a context in which microbial dysbiosis and chronic antigenic stimulation may drive CD8⁺ T-cell exhaustion, thereby attenuating the efficacy of microbiome-based interventions116. Furthermore, the absence of integrated microbial, metabolic, or immunologic biomarkers in many protocols restricts mechanistic interpretation and patient stratification. Addressing these gaps will require trial designs that incorporate microbiome profiling, functional readouts (e.g., SCFA production), and immune correlatives to identify predictive signatures and optimize rational combinations.

Collectively, this analysis delineates the burgeoning clinical investigation landscape of MMs in CRC, with 29 trials evaluating FMT, probiotics/prebiotics, and LBPs. These findings underscore the translational promise of microbiome modulation as a paradigm-shifting adjuvant in CRC. Future efforts should prioritize optimizing optimal MM combination regimens, validating microbiome-based biomarkers, and expanding trials to broader patient cohorts to accelerate clinical translation.

Despite its strengths, this study has several limitations. The Trialtrove database, curated by a team of therapeutically focused expert analysts, is widely recognized as a comprehensive and authoritative source of global clinical trial intelligence and has contributed to numerous peer-reviewed publications117119. Nevertheless, delays in data updates and occasional classification inconsistencies may affect the completeness of trial capture. To address this, two additional investigators were assigned to manually verify key records through timely tracking of multiple data sources, including official clinical trial registries across different countries and regions, conference abstracts, and published literature. These measures were intended to improve data coverage within the constraints of available resources.

Role of gut microbiota in CRC treatment

Therapeutic implications for chemotherapy

Increasing research suggests that gut microbiota can influence the anticancer effects of certain chemotherapeutic agents, such as 5-fluorouracil120, cyclophosphamide121, and oxaliplatin122, through various mechanisms, including microbial translocation, immunomodulation, metabolism, enzymatic degradation, and reduced ecological diversity123. Also, Gut microbial metabolites, particularly butyrate, may enhance the effectiveness of oxaliplatin by regulating the function of CD8+ T cells within the TME124. In vitro studies indicate that Lactobacillus species can either boost the activity of or counteract resistance to 5-FU and irinotecan in CRC by secreting metabolites like butyrate125, gamma-aminobutyric acid126, and extracellular vesicles127. The DM#1 mixture, which includes Bifidobacterium breve DM8310, L. acidophilus DM8302, Lactobacillus casei DM8121, and S. thermophilus DM8309, has been shown to restore intestinal integrity and reduce the activation of proinflammatory cytokines following 5-FU treatment128. Similar beneficial effects have been observed when Lactobacillus acidophilus, Lactobacillus paracasei, Lactobacillus rhamnosus, and Bifidobacterium lactis are used as probiotics in conjunction with 5-FU129. Furthermore, L. rhamnosus GG was associated with a reduction in the severity of diarrhea in a cohort of 150 crc patients undergoing 5-FU treatment130. A study also suggested that Bifidobacterium infantis might mitigate chemotherapy-induced toxicity (from 5-FU and oxaliplatin) by suppressing Th1 and Th17 activation while boosting the activity of cytotoxic regulatory T cells (CD4+ CD25+ Foxp3+) in Sprague-Dawley rats131. Recent evidence indicates that tumor-colonizing Bacteroides fragilis mediates resistance to 5-fluorouracil and oxaliplatin via SusD/RagB-dependent activation of Notch1 signaling, culminating in EMT and stemness acquisition132. Lang et al. developed a Capecitabine-loaded nanoparticle using a prebiotic xylan-stearic acid conjugate (SCXN). This SCXN nanoparticle significantly enhances antitumor immunity and increases the tumor inhibition rate from 5.29% to 71.78% compared to free Capecitabine133.

In addition to boosting antitumor effects, the gut microbiota also impacts the toxicity associated with chemotherapy. A frequent complication for cancer patients undergoing treatment is intestinal mucositis and diarrhea134,135. The active metabolite of irinotecan, SN-38, is detoxified in the liver through glucuronidation and then excreted into the gut via bile136. However, gut bacteria can reactivate SN-38 by producing β-glucuronidase, leading to intestinal damage and severe diarrhea137. Using a selective inhibitor to block this reactivation could prevent the associated toxicity in mice, offering a strategy to alter microbial activity and mitigate the adverse side effects of chemotherapy138.

Therapeutic implications for immunotherapy

Immunotherapy has emerged as a cornerstone in cancer treatment, demonstrating effectiveness across various types of cancer139. ICIs function by lifting the inhibitory signals on T-cell activation, enabling tumor-reactive T cells to generate a robust antitumor response140. Despite this, ICIs are generally ineffective in CRC, with the exception of a small percentage (~5%) of patients who have MMR-deficient or MSI-H tumors141,142. The gut microbiota is essential for a successful immune response during immunotherapy and can influence the efficacy of ICIs targeting the PD-1/PD-L1 and CTLA-4 pathways143145. Certain bacteria, including Akkermansia muciniphila143, B. fragilis145, Bifidobacterium spp.146, Eubacterium limosum147, Faecalibacterium spp.148, and Alistipes shahii122, have been linked to improved outcomes with immunotherapy. However, as CTLA-4 and PD-1 are also highly expressed on tumor-infiltrating regulatory T cells (Tregs), their blockade may paradoxically disrupt immune regulation, potentially exacerbating autoimmunity or undermining therapeutic efficacy149. Considering the gut microbiota’s role in modulating responses to ICIs, this area offers promising translational opportunities for advancing CRC treatment (Fig. 3).

Fig. 3. Current strategies leveraging the gut microbiota to enhance responsiveness to ICIs in CRC.

Fig. 3

These strategies include FMT and emerging approaches such as probiotics, prebiotics, probiotic metabolites, and genetically engineered probiotics, all of which hold great potential in improving patient outcomes.

Interestingly, pathobionts that promote CRC may have mixed effects on ICIs. For instance, while F. nucleatum is known to drive tumor growth and chemoresistance, it can also boost the effectiveness of anti-PD-L1 antibodies and extend survival in mouse models150. It activates STING–NF-κB signaling and enhances PD-L1 expression, yet paradoxically increases CD8+ T-cell activation when PD-L1 is blocked150. Similarly, ETBF in multiple intestinal neoplasia (Min) mice induces serrated-like tumors and increases sensitivity to anti-PD-L1 antibodies151. Thus, pathogenic bacteria can have conflicting effects on ICI therapy.

Prebiotics

Higher dietary fiber intake is linked to better responses to ICIs and fewer immune-related adverse events in cancer patients from diverse global cohorts152. Additionally, prebiotics and soluble fibers like inulin153, pectin154, polyphenols155, and polysaccharides156 have been found to boost the effectiveness of anti-PD-1 antibodies in several mouse models. Mechanistically, these substances promote the enrichment of specific bacterial genera, such as those in the Ruminococcaceae family, individual bacteria like Akkermansia muciniphila, and their metabolites, including SCFAs and cyclic diadenosine monophosphate (cyclic di-AMP)157. SCFAs enhance the function of cytotoxic CD8+ T cells153, while cyclic di-AMP triggers STING-dependent type I interferon production, leading to the activation of innate immune responses (including monocytes, NK cells, and dendritic cells) and improved ICI efficacy158. However, under fiber deprivation, mucin-degrading bacteria (Akkermansia muciniphila) may degrade host-derived mucins, compromising the mucus barrier and increasing colitis susceptibility. Thus, maintaining adequate dietary fiber intake is crucial to leverage beneficial microbial activities while avoiding potential detrimental effects associated with excessive mucin degradation159,160. In a transgenic mouse model of CRC, a combination of inulin gel and α-PD-1 therapy inhibited the growth of dextran sulfate sodium (DSS)-accelerated colon tumors in CDX2-Cre NLS-APCfl/fl mice153. Since prebiotics depend on the presence of beneficial taxa already existing in the host’s microbiota, synbiotics—which involve the combined administration of both prebiotics and probiotics—may offer greater effectiveness than using either alone161.

Probiotics and their products

Certain gut bacteria, such as A. muciniphila143, B. fragilis145, Bifidobacterium146, L. rhamnosus GG162, Lacticaseibacillus paracasei163, and various probiotic mixtures164, have been shown to enhance the efficacy of ICIs in mouse models. Recent studies have explored the effects of CBM588, which contains C. butyricum, on metastatic renal cell carcinoma treatments. One Phase I trial found that adding CBM588 to nivolumab and ipilimumab improved response rates (58% vs. 20%, P = 0.06), though not significantly165. Another trial showed a significant increase in response rates when CBM588 was combined with cabozantinib and nivolumab (74% vs. 20%, P = 0.01)166.

The optimal dose of probiotics is crucial for treatment efficacy, as an excessive presence of a beneficial bacterial species could disrupt the gut microbiota’s balance and diversity, potentially leading to worse outcomes. A study on NSCLC patients receiving ICB therapy found that while the presence of Akkermansia muciniphila correlated with improved responses, patients with a relative abundance of A. muciniphila over 5% had worse outcomes than those without it167. Also, selected reports have described context-specific interventions that failed to confer benefit or even exacerbated disease. In a cohort of 158 melanoma patients receiving ICB, self-reported use of commercially available probiotics was associated with a trend toward reduced response rates and shorter progression-free survival, although differences did not reach statistical significance. Preclinical validation in mice supported these findings, showing that probiotic supplementation impaired anti-PD-1 efficacy by reducing intratumoral interferon-γ⁺ cytotoxic T-cell accumulation168. Separately, in a murine model of colitis-associated CRC (AOM/Il10⁻/⁻), interventional treatment with the VSL#3 probiotic cocktail significantly increased tumor incidence, multiplicity, and histologic dysplasia, accompanied by loss of protective Clostridial taxa in the mucosa-associated microbiota169. These findings underscore the need for cautious, evidence-based selection of probiotic regimens in oncological settings, particularly when inflammation or dysbiosis is present. Current research is exploring the use of well-balanced microbial consortia, such as VE800, a probiotic cocktail of 11 strains, in combination with nivolumab in a Phase I/II trial for MSS CRC164. Additionally, a 30-bacteria consortium (MET4) was found to be safe and capable of modifying the gut microbiota and serum metabolome in ICB-naive patients, reinforcing the potential benefits of microbial consortia170. However, a study demonstrated that a mix of four Clostridiales strains (CC4) could prevent and treat CRC in mice, with the effect reliant on CD8+ T-cell activation. Interestingly, single applications of Roseburia intestinalis or Anaerostipes caccae were even more effective than the CC4 mix171. These findings suggest that while consortia may be beneficial, certain individual strains could provide superior therapeutic effects.

Several probiotic-derived therapeutics, informed by an understanding of their active components, are currently being evaluated as adjuvants to ICIs. These include bacterial lysates172, extracellular vesicles173, and more defined components such as exopolysaccharides174, muropeptides175, and metabolites124,176,177. These components have the potential to either fully or partially mimic the effects of live probiotics in enhancing the efficacy of ICIs.

Genetically engineered probiotics

Probiotics engineered with improved functionality could serve as a safe and efficient method for delivering adjuvant therapy. Investigators engineered a Lactobacillus lactis strain expressing either wild-type, catalytically inactive (C443A), or secretion-deficient (ΔSS) SagA, with the wild-type and C443A mutants showing higher signals in the secreted fraction; animals supplemented with L. lactis expressing wild-type SagA demonstrated tumor growth inhibition comparable to that of E. faecium-treated animals, effectively augmenting the activity of anti-PD-L1 antibodies to a similar extent as natural SagA-expressing Enterococcus strains in MC38 CRC allograft mice175. Genetically engineered bacteria could be employed as vehicles to support the metabolism of intratumoral T cells. In an immunocompetent mouse model of CRC (MC38), researchers developed a probiotic strain of E. coli Nissle 1917 (EcN) that converts tumor-derived ammonia into L-arginine178,179. This L-arginine plays a critical role in enhancing the metabolic fitness and survival of T cells, which are vital for effective antitumor responses180 (Fig. 4).

Fig. 4. Genetically engineered probiotics for tumor-targeted immunotherapy.

Fig. 4

Orally delivered Lactobacillus plantarum expressing SagA generates muropeptides that engage macrophage NOD2, activating the NLRP3 inflammasome and IL-1β release. Concurrently, tumor-colonizing Escherichia coli Nissle secretes anti-PD-L1 nanobodies in situ, blocking PD-1/PD-L1 interactions and restoring CD8⁺ T-cell activity. NOD2 nucleotide-binding oligomerization domain-containing protein 2, SagA secreted antigen A.

To evaluate the versatility of probiotic systems in treating immunologically “cold” cancers, researchers have developed engineered probiotics designed for targeted therapy delivery. This approach involves engineering probiotic bacteria to produce and release nanobodies targeting PD-L1 and CTLA-4 directly within tumors, utilizing a controlled lysing release mechanism. In a mouse model of MSS CRC, combining this system with a probiotic-produced cytokine, granulocyte-macrophage colony-stimulating factor, led to improved antitumor effects and increased survival181. These studies demonstrate the potential of genetically engineered probiotics as adaptable tools to enhance the effectiveness of ICIs.

Fecal microbiota transplantation

FMT is a clinical approach to modulating the microbiome, which has received FDA approval for treating recurrent C. difficile infections182. FMT has also been explored in combination with ICIs in clinical trials, based on evidence that administering FMT to germ-free animals can replicate the ICI responses observed in human donors183,184. However, one study found that mice receiving FMT from patients with IBD experienced a significant reduction in colonic TH1 cells compared to those receiving FMT from healthy donors, and exhibited resistance to treatment185, suggesting that a healthy microbiome is crucial for ICI efficacy.

Initial clinical studies have shown promising results, such as overcoming resistance to anti-PD-1 antibodies in some patients with ICI-refractory metastatic melanoma through FMT from anti-PD-1-responsive donors186,187. Moreover, a multicenter phase I trial evaluated the combination of FMT from cancer-free donors with nivolumab or pembrolizumab in patients with previously untreated advanced-stage melanoma188.

In a study by Huang, the combination of FMT and PD-1 inhibitors led to better survival outcomes and tumor control compared to either treatment alone in CRC tumor-bearing mice. The study highlighted that FMT-increased Bacteroides thetaiotaomicron and Bacteroides fragilis, along with FMT-decreased Bacteroides ovatus, contributed to the enhanced efficacy of ICIs in CRC184. In a phase II trial, MSS mCRC patients received FMT plus tislelizumab and fruquintinib as a third-line or beyond treatment, resulting in a mPFS of 9.6 months and a median overall survival (mOS) of 13.7 months, with an ORR of 20% and a DCR of 95%, and no treatment-related deaths112. Another clinical trial suggests that FMT with beneficial microbiota can help overcome resistance to anti-PD-1 inhibitors in advanced solid cancers, particularly gastrointestinal cancers113. Additional trials using similar approaches in CRC patients are ongoing (NCT04729322 and NCT04130763).

However, FMT has notable limitations, including safety concerns and challenges related to the generalizability and standardization of microbiota profiles needed for efficacy, both in donors and recipients, which makes it less suitable for widespread use. Since FMT involves transplanting the entire viable gut microbiota from a donor, it carries inherent risks of transmitting known and unknown pathogens. Reported complications include norovirus enteritis, cytomegalovirus reactivation, fungal and parasitic infections, and even the potential horizontal transfer of disease-associated traits such as obesity or metabolic dysfunctions189,190. In 2019, it was reported that two patients developed bacteremia with extended-spectrum β-lactamase-producing E. coli after receiving FMT from the same donor, resulting in one fatality191. This incident prompted the US Food and Drug Administration to issue a safety warning about the infection risks associated with FMT. Furthermore, a recent retrospective cohort study found that 9% of tested donor feces contained multidrug-resistant organisms at some point, highlighting another significant safety concern192.

Therapeutic implications for radiotherapy

Radiotherapy (RT) is a widely used antitumor treatment, with over 50% of newly diagnosed cancer patients with solid tumors undergoing RT at some stage of their treatment193. The intestine, however, is highly sensitive to radiation due to its rapid epithelial turnover194. Studies in mouse models, including those focused on segmental small bowel radiation195 and graft-versus-host disease196,197, have demonstrated significant shifts in gut microbiota following RT. There is a bidirectional interaction between intestinal microbiota and radiotherapy. On one side, ionizing radiation alters the composition and function of the gut microbiome, reducing microbial diversity and richness, increasing the abundance of pathogenic bacteria such as Proteobacteria and Fusobacteria, and decreasing beneficial species like Faecalibacterium and Bifidobacterium198. Prophylactic administration of B. infantis with its specific antibody has been shown to enhance radiosensitization and suppress tumor growth in a murine lung cancer model199, while absolute quantification via 16S rRNA qPCR demonstrates an approximately 90% reduction in total bacterial load after (chemo)radiotherapy, indicating that relative increases in pathobionts often reflect loss of dominant commensals rather than their true overgrowth200. On the other side, the microbiota influences the clinical efficacy of RT by enhancing treatment outcomes and mitigating radiotoxicity.

Improving the efficacy of radiotherapy

Research has shown that oral vancomycin enhances both the direct and abscopal antitumor effects of hypofractionated RT in preclinical models of melanoma and lung/cervical tumors201. Other studies have revealed that commensal bacteria are essential for effective antitumor immune responses, while commensal fungi regulate the immunosuppressive microenvironment after RT. Targeting these fungi has been shown to improve RT outcomes202. FMT has been found to increase levels of Roseburia intestinalis in the gastrointestinal tract, with this gut commensal bacterium promoting radiation-induced autophagy and enhancing CRC cell death via the butyrate/OR51E1/RALB axis. This mechanism offers a promising radiosensitizer for CRC in preclinical settings203. Additionally, ETBF, a bacterial strain enriched in CRC patients, produces Bacteroides fragilis toxin (BFT) and IL-17, which synergistically activate the STAT3 signaling pathway in immune cells, potentially promoting cancer growth204. Studies suggest that ETBF may also enhance radioresistance in CRC by generating BFT, potentially activating the JAK2/STAT3/CCND2 axis, a known mediator of radioresistance205.

Reducing radiotoxicity

Several strategies have been identified to reduce the harmful side effects of RT. A randomized clinical trial demonstrated that the probiotic Streptococcus salivarius K12 (SsK12) significantly reduced the incidence, onset, and duration of severe oral mucositis in RT patients, with a favorable safety profile206. Additionally, experimental studies have shown that FMT mitigates acute radiation syndrome (ARS), improving peripheral white blood cell counts and preserving gastrointestinal function and intestinal epithelial integrity in irradiated mice207. Further research found that FMT increases levels of indole-3-propionic acid (IPA) in irradiated mice, a compound that alleviates gastrointestinal toxicity after total abdominal irradiation without accelerating tumor growth208.

Therapeutic implications for surgical resection

Compelling experimental data from rodent studies highlight the vital role of gut microbes in intestinal mucosal vascularization and wound healing209. Specific components of the microbiota, especially butyrate-producing bacteria, are instrumental in epithelial repair210. Additionally, research has shown that Bacteroides thetaiotaomicron promotes angiogenesis by effectively doubling the capillary network in the small intestine through the activation of Paneth cells211.

The dynamics of bacterial competition and cooperation within microbial communities can either facilitate or hinder wound healing. For instance, Lactobacillus fermentum RC-14 has been found to inhibit infections caused by Staphylococcus aureus in a rat model of surgical implants212. Conversely, the collaboration among siderophore-producing bacteria, like Pseudomonas aeruginosa, can increase virulence by enabling the sharing of iron-scavenging compounds with other bacteria213.

In a landmark study in 1997, Schardey et al. suggested a potential link between P. aeruginosa and anastomotic leaks, leading to the first randomized double-blind trial that confirmed the involvement of microbes in human anastomotic complications214. Subsequent research found that a novel virulence-suppressing compound, PEG/Pi (high-molecular-weight PEG), prevented P. aeruginosa from adopting a tissue-destructive phenotype, thus mitigating anastomotic leaks in rat models215. Research by Shogan et al. examined the shifts in microbiota at anastomotic sites in a rodent model using 16S rRNA gene sequencing. Notably, their study avoided the use of antibiotics and bowel preparation, revealing significant changes in tissue-associated microbiota at these sites, while the luminal microbiota (stool) remained relatively stable216.

Comparative analyses of gut microbiota revealed that post-surgery CRC patients (A1) exhibit significant differences in microbial diversity compared to pre-surgery patients (A0) and healthy individuals (H). Specifically, both Shannon and Simpson diversity indices were notably lower in A1 (P < 0.05), underscoring the gut microbiota’s role in post-surgical outcomes217. Additionally, a recent study identified that an increased abundance of specific microbiota clusters, including pathobiont genera such as Fusobacterium, Granulicatella, and Gemella, correlates with better outcomes following primary resection, challenging conventional views on the gut microbiota’s role in CRC prognosis218.

In a clinical investigation, Komen et al. developed a rapid screening method for early detection of colorectal anastomotic leaks based on the detection of E. coli and Enterococcus faecalis in drained fluid using RT-PCR. Their results indicate that higher levels of E. faecalis may serve as a reliable indicator for early detection, while negative results strongly suggest the absence of anastomotic leaks219. Reviews on the use of probiotics in the perioperative care of CRC patients indicate that probiotics can reduce bacterial translocation, enhance intestinal mucosal integrity, and promote a favorable balance between beneficial and harmful microorganisms220. Furthermore, both probiotics and synbiotics are emerging as effective strategies to prevent postoperative complications in CRC surgery221.

Conclusion and outlooks

Unlike the relatively static human genome, the microbiome demonstrates remarkable plasticity, continually evolving in response to environmental, dietary, and lifestyle factors222. While the host genome is largely inherited from parental DNA, the microbiome is highly personalized, characterized by significant variability across individuals and over time223,224. Understanding the drivers of this complexity offers a unique opportunity to improve prevention, diagnosis, and treatment strategies, paving the way for more precise and personalized healthcare solutions225.

In CRC, metagenomic sequencing is increasingly employed to characterize tumor-associated microbial communities45,226; However, notable discrepancies across studies limit their clinical application, often arising from technical differences (such as sample processing and sequencing methods) and confounding factors like geography, diet, gender, and sampling time227,228. To address these issues, the Strengthening the Organization and Reporting of Microbiome Studies guideline was introduced, providing a 17-item checklist designed to promote consistency and transparency in microbiome research229. Moreover, rigorous a priori power and sample-size calculations—accounting for microbiome data’s compositionality, sparsity, and high dimensionality—are essential to ensure valid hypothesis testing and robust, generalizable conclusions230. Complementing these efforts, integrative multi-omics frameworks—combining metagenomics with metabolomics, transcriptomics, epigenomics, and proteomics at bulk and single-cell resolution in patient-derived tissues and experimental models—are being deployed to unravel the complexity of host–microbe crosstalk231233. Recent developments in spatial meta-omics profiling—employing targeted nucleic-acid hybridization probes to resolve microbial taxa distributions within TMEs—have begun to delineate the architecture of the intratumoural microbiome and its modulatory effects on adjacent cellular phenotypes234,235. However, these approaches remain unable to integrate microbial spatial mapping with concurrent in situ functional assays (eg, microbial toxin or cytokine expression). Addressing the constraints imposed by low microbial biomass and rapid RNA degradation will be pivotal for true interactome reconstruction and high-resolution spatial community profiling222.

Moreover, to address limitations in low-biomass clinical specimens, a tailored metatranscriptomics workflow has been developed that integrates optimized taxonomic profiling (Kraken 2/Bracken) with functional annotation (HUMAnN 3), enabling accurate detection of microbial species and their expressed pathways even in gastric tissues with a high host RNA background236. Complementing this, the advent of long-read sequencing platforms has significantly enhanced metagenome-assembled genome completeness, strain-level discrimination, and variant calling, overcoming the assembly and profiling biases inherent to short-read approaches237.

The increasing depth and scale of metagenomic and metatranscriptomic datasets can inadvertently amplify artefacts—as exemplified by Parida et al.’s report of extremophilic taxa in breast tumors—prompting rigorous revisions to contamination controls, batch-effect corrections, and validation pipelines238,239. In response, machine-learning and artificial-intelligence pipelines have been refined with stricter cross-validation, larger hold-out cohorts, and tailored feature-engineering to extract true signals from complex microbiome data240. Integrating explainable AI—such as Random Forests coupled with SHAP—yielded ~0.73 precision and a ~0.67 precision-recall AUC in CRC classification, while highlighting Fusobacterium, Peptostreptococcus, and Parvimonas as the most predictive taxa241.

However, many AI models—despite reported superiority—often show only moderate or unstable predictive power in cancer studies, largely due to overfitting on small cohorts, poor-quality or limited microbiome datasets, and inadequate validation242,243. To enhance generalizability and accuracy in microbiome-based CRC applications, strategies such as multimodal data integration (clinical, microbial, and longitudinal)244, leveraging microbiome signatures for immunotherapy response prediction, and incorporating high-resolution single-cell and spatial multi-omics have been proposed245,246, underscoring AI’s critical role in deciphering complex gut microbial ecosystems for precision oncology.

Nanotechnology has revolutionized CRC therapy by enabling nanoscale drug delivery systems that overcome poor solubility and stability in the gastrointestinal tract, while enhancing tumor accumulation and minimizing systemic exposure247,248. These platforms also offer precise modulation of the gut microbiota: bacteria-targeting ligands guide drug-loaded carriers to tumors, and self-thermophoretic nanoparticles cloaked in bacterial membranes achieve deeper mucus penetration and reduced microbial interception, yielding near-complete tumor regression in orthotopic CRC models247,249.

Building on these advances, bioengineered probiotics and prebiotic–nanoparticle composites have been developed to selectively colonize CRC lesions and reshape the microbial ecosystem133. Tumor-targeted E. coli Nissle and Pediococcus pentosaceus expressing therapeutic proteins inhibit CRC cell growth and regress adenomas250,251, while prebiotic-encapsulated spores and xylan-stearic acid nanoparticles promote SCFA production, augment CD8⁺ T-cell activity, and boost antitumour efficacy from 5% to over 70% in vivo133,252. Despite these promising strategies, key challenges—intestinal infiltration, probiotic engraftment, intratumoural targeting, and immune regulation—must be addressed to realize precision microbiota editing in CRC management.

In the future, rigorous study designs, standardized multi-omics frameworks, and the innovative integration of spatial omics, artificial intelligence, and microbiome-targeted nanotechnologies will be essential to unravel microbial mechanisms underpinning CRC, thereby accelerating the translation of microbiome insights into precision oncology.

Supplementary information

41522_2025_818_MOESM1_ESM.xlsx (119.9KB, xlsx)

Table S1: The clinical trials of microbiome modulators in CRC

Author contributions

C.C. performed the statistical analysis and drafted the manuscript. Q.S. and Z.M. conducted the literature search and data collection. C.C. and Q.S. contributed to data interpretation and provided critical revisions. Y.D. assisted with methodology and analysis. Z.Z. and X.H. designed the study, conceptualized the research framework, and revised the manuscript. All authors contributed to the article and approved the final version.

Data availability

No datasets were generated or analyzed during the current study.

Competing interests

The authors declare no competing interests.

Footnotes

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Contributor Information

Xiaokun Hua, Email: 43256647@qq.com.

Zhiyun Zhang, Email: jianchidaodi_2010@163.com.

Supplementary information

The online version contains supplementary material available at 10.1038/s41522-025-00818-3.

References

  • 1.Bray, F. et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin.74, 229–263 (2024). [DOI] [PubMed] [Google Scholar]
  • 2.Eng, C. et al. Colorectal cancer. Lancet404, 294–310 (2024). [DOI] [PubMed] [Google Scholar]
  • 3.Sinicrope, F. A. Increasing incidence of early-onset colorectal cancer. N. Engl. J. Med.386, 1547–1558 (2022). [DOI] [PubMed] [Google Scholar]
  • 4.White, M. T. & Sears, C. L. The microbial landscape of colorectal cancer. Nat. Rev. Microbiol.22, 240–254 (2024). [DOI] [PubMed] [Google Scholar]
  • 5.Kuipers, E. J. et al. Colorectal cancer. Nat. Rev. Dis. Prim.1, 15065 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Grothey, A. et al. Duration of adjuvant chemotherapy for stage III colon cancer. N. Engl. J. Med.378, 1177–1188 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Auclin, E. et al. Subgroups and prognostication in stage III colon cancer: future perspectives for adjuvant therapy. Ann. Oncol.28, 958–968 (2017). [DOI] [PubMed] [Google Scholar]
  • 8.Benson, A. B. et al. Colon Cancer, Version 3.2024, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw.22, e240029 (2024). [DOI] [PubMed]
  • 9.Zheng, H. et al. Targeted activation of ferroptosis in colorectal cancer via LGR4 targeting overcomes acquired drug resistance. Nat. Cancer5, 572–589 (2024). [DOI] [PubMed] [Google Scholar]
  • 10.Wang, X. et al. Fusobacterium nucleatum facilitates anti-PD-1 therapy in microsatellite stable colorectal cancer. Cancer Cell42, 1729–1746.e1728 (2024). [DOI] [PubMed] [Google Scholar]
  • 11.Casak, S. J. et al. FDA Approval Summary: pembrolizumab for the first-line treatment of patients with MSI-H/dMMR advanced unresectable or metastatic colorectal carcinoma. Clin. Cancer Res.27, 4680–4684 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chen, E. & Zhou, W. Immunotherapy in microsatellite-stable colorectal cancer: Strategies to overcome resistance. Crit. Rev. Oncol. Hematol.212, 104775 (2025). [DOI] [PubMed] [Google Scholar]
  • 13.Dong, J. et al. Roseburia intestinalis sensitizes colorectal cancer to radiotherapy through the butyrate/OR51E1/RALB axis. Cell Rep.43, 113846 (2024). [DOI] [PubMed] [Google Scholar]
  • 14.Zheng, Z. et al. The implication of gut microbiota in recovery from gastrointestinal surgery. Front. Cell. Infect. Microbiol.13, 1110787 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Fernandes, M. R., Aggarwal, P., Costa, R. G. F., Cole, A. M. & Trinchieri, G. Targeting the gut microbiota for cancer therapy. Nat. Rev. Cancer22, 703–722 (2022). [DOI] [PubMed] [Google Scholar]
  • 16.Fan, Y. & Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol.19, 55–71 (2021). [DOI] [PubMed] [Google Scholar]
  • 17.Li, M. et al. Gut microbiota metabolite indole-3-acetic acid maintains intestinal epithelial homeostasis through mucin sulfation. Gut Microbes16, 2377576 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Keohane, D. M. et al. Microbiome and health implications for ethnic minorities after enforced lifestyle changes. Nat. Med.26, 1089–1095 (2020). [DOI] [PubMed] [Google Scholar]
  • 19.Zhang, J. & Sears, C. L. Antibiotic use impacts colorectal cancer: a double-edged sword by tumor location?. J. Natl. Cancer Inst.114, 1–2 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bartolomaeus, T. U. P. et al. Quantifying technical confounders in microbiome studies. Cardiovasc. Res.117, 863–875 (2021). [DOI] [PubMed] [Google Scholar]
  • 21.Liu, N. N. et al. Multi-kingdom microbiota analyses identify bacterial-fungal interactions and biomarkers of colorectal cancer across cohorts. Nat. Microbiol.7, 238–250 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Fernandez-Rozadilla, C. et al. Deciphering colorectal cancer genetics through multi-omic analysis of 100,204 cases and 154,587 controls of European and East Asian ancestries. Nat. Genet.5‘5, 89–99 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Mouradov, D. et al. Oncomicrobial community profiling identifies clinicomolecular and prognostic subtypes of colorectal cancer. Gastroenterology165, 104–120 (2023). [DOI] [PubMed] [Google Scholar]
  • 24.Gao, R. et al. Integrated analysis of colorectal cancer reveals cross-cohort gut microbial signatures and associated serum metabolites. Gastroenterology163, 1024–1037.e1029 (2022). [DOI] [PubMed] [Google Scholar]
  • 25.Purcell, R. V., Visnovska, M., Biggs, P. J., Schmeier, S. & Frizelle, F. A. Distinct gut microbiome patterns associate with consensus molecular subtypes of colorectal cancer. Sci. Rep.7, 11590 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Zuo, W., Michail, S. & Sun, F. Metagenomic analyses of multiple gut datasets revealed the association of phage signatures in colorectal cancer. Front. Cell. Infect. Microbiol.12, 918010 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chen, F. et al. Meta-analysis of fecal viromes demonstrates high diagnostic potential of the gut viral signatures for colorectal cancer and adenoma risk assessment. J. Adv. Res.49, 103–114 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Klein, R. S. et al. Association of Streptococcus bovis with carcinoma of the colon. N. Engl. J. Med.297, 800–802 (1977). [DOI] [PubMed] [Google Scholar]
  • 29.Van Hul, M. et al. What defines a healthy gut microbiome?. Gut73, 1893–1908 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Winter, S. E. & Bäumler, A. J. Gut dysbiosis: ecological causes and causative effects on human disease. Proc. Natl. Acad. Sci. USA120, e2316579120 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Tilg, H., Adolph, T. E., Gerner, R. R. & Moschen, A. R. The intestinal microbiota in colorectal cancer. Cancer Cell33, 954–964 (2018). [DOI] [PubMed] [Google Scholar]
  • 32.Martínez-García, J. J. et al. Diurnal interplay between epithelium physiology and gut microbiota as a metronome for orchestrating immune and metabolic homeostasis. Metabolites12, 390 (2022). [DOI] [PMC free article] [PubMed]
  • 33.Wong, C. C. & Yu, J. Gut microbiota in colorectal cancer development and therapy. Nat. Rev. Clin. Oncol.20, 429–452 (2023). [DOI] [PubMed] [Google Scholar]
  • 34.Knippel, R. J., Drewes, J. L. & Sears, C. L. The cancer microbiome: recent highlights and knowledge gaps. Cancer Discov.11, 2378–2395 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Chagneau, C. V. et al. The pks island: a bacterial Swiss army knife? Colibactin: beyond DNA damage and cancer. Trends Microbiol.30, 1146–1159 (2022). [DOI] [PubMed] [Google Scholar]
  • 36.Dziubańska-Kusibab, P. J. et al. Colibactin DNA-damage signature indicates mutational impact in colorectal cancer. Nat. Med.26, 1063–1069 (2020). [DOI] [PubMed] [Google Scholar]
  • 37.Rosendahl Huber, A., Pleguezuelos-Manzano, C. & Puschhof, J. A bacterial mutational footprint in colorectal cancer genomes. Br. J. Cancer124, 1751–1753 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Rosendahl Huber, A. et al. Improved detection of colibactin-induced mutations by genotoxic E. coli in organoids and colorectal cancer. Cancer Cell42, 487–496.e486 (2024). [DOI] [PubMed] [Google Scholar]
  • 39.Lopès, A. et al. Colibactin-positive Escherichia coli induce a procarcinogenic immune environment leading to immunotherapy resistance in colorectal cancer. Int. J. Cancer146, 3147–3159 (2020). [DOI] [PubMed] [Google Scholar]
  • 40.Butt, J. et al. Association of pre-diagnostic antibody responses to Escherichia coli and bacteroides fragilis toxin proteins with colorectal cancer in a European cohort. Gut Microbes13, 1–14 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Sugihara, K. et al. Mucolytic bacteria license pathobionts to acquire host-derived nutrients during dietary nutrient restriction. Cell Rep.40, 111093 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jans, M. et al. Colibactin-driven colon cancer requires adhesin-mediated epithelial binding. Nature635, 472–480 (2024). [DOI] [PubMed] [Google Scholar]
  • 43.Dalmasso, G. et al. Colibactin-producing Escherichia coli enhance resistance to chemotherapeutic drugs by promoting epithelial to mesenchymal transition and cancer stem cell emergence. Gut Microbes16, 2310215 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.de Oliveira Alves, N. et al. The colibactin-producing Escherichia coli alters the tumor microenvironment to immunosuppressive lipid overload facilitating colorectal cancer progression and chemoresistance. Gut Microbes16, 2320291 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lin, Y. et al. Multi-cohort analysis reveals colorectal cancer tumor location-associated fecal microbiota and their clinical impact. Cell Host Microbe33, 589–601.e583 (2025). [DOI] [PubMed] [Google Scholar]
  • 46.Brennan, C. A. et al. Fusobacterium nucleatum drives a pro-inflammatory intestinal microenvironment through metabolite receptor-dependent modulation of IL-17 expression. Gut Microbes13, 1987780 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wang, Q. et al. Fusobacterium nucleatum promotes colorectal cancer through neogenesis of tumor stem cells. J. Clin. Investig.135, e181595 (2025). [DOI] [PMC free article] [PubMed]
  • 48.Li, D., Li, Z., Wang, L., Zhang, Y. & Ning, S. Oral inoculation of Fusobacterium nucleatum exacerbates ulcerative colitis via the secretion of virulence adhesin FadA. Virulence15, 2399217 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Duizer, C. et al. Fusobacterium nucleatum upregulates the immune inhibitory receptor PD-L1 in colorectal cancer cells via the activation of ALPK1. Gut Microbes17, 2458203 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ternes, D. et al. The gut microbial metabolite formate exacerbates colorectal cancer progression. Nat. Metab.4, 458–475 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chen, F. et al. Fusobacterium nucleatum-driven CX3CR1(+) PD-L1(+) phagocytes route to tumor tissues and reshape tumor microenvironment. Gut Microbes17, 2442037 (2025). [DOI] [PubMed] [Google Scholar]
  • 52.Gao, Y. et al. Fusobacterium nucleatum enhances the efficacy of PD-L1 blockade in colorectal cancer. Signal Transduct. Target. Ther.6, 398 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Jiang, S. S. et al. Fusobacterium nucleatum-derived succinic acid induces tumor resistance to immunotherapy in colorectal cancer. Cell Host Microbe31, 781–797.e789 (2023). [DOI] [PubMed] [Google Scholar]
  • 54.Zhu, H. et al. Fusobacterium nucleatum promotes tumor progression in KRAS p.G12D-mutant colorectal cancer by binding to DHX15. Nat. Commun.15, 1688 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wang, N. et al. Fusobacterium nucleatum induces chemoresistance in colorectal cancer by inhibiting pyroptosis via the Hippo pathway. Gut Microbes16, 2333790 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Li, B. et al. Fusobacterium nucleatum induces oxaliplatin resistance by inhibiting ferroptosis through E-cadherin/β-catenin/GPX4 axis in colorectal cancer. Free Radic. Biol. Med.220, 125–138 (2024). [DOI] [PubMed] [Google Scholar]
  • 57.Hong, X. L. et al. Metformin abrogates Fusobacterium nucleatum-induced chemoresistance in colorectal cancer by inhibiting miR-361-5p/sonic hedgehog signaling-regulated stemness. Br. J. Cancer128, 363–374 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Jia, F. et al. Optimized antimicrobial peptide Jelleine-I derivative Br-J-I inhibits Fusobacterium nucleatum to suppress colorectal cancer progression. Int. J. Mol. Sci.24, 1469 (2023). [DOI] [PMC free article] [PubMed]
  • 59.Valguarnera, E. & Wardenburg, J. B. Good gone bad: one toxin away from disease for bacteroides fragilis. J. Mol. Biol.432, 765–785 (2020). [DOI] [PubMed] [Google Scholar]
  • 60.Lee, C. G. et al. Bacteroides fragilis toxin induces intestinal epithelial cell secretion of interleukin-8 by the E-cadherin/β-catenin/NF-κB dependent pathway. Biomedicines. 10, 827 (2022). [DOI] [PMC free article] [PubMed]
  • 61.Liu, Q. Q. et al. Enterotoxigenic Bacteroides fragilis induces the stemness in colorectal cancer via upregulating histone demethylase JMJD2B. Gut Microbes12, 1788900 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wu, X. et al. Enterotoxigenic Bacteroides fragilis (ETBF) enhances colorectal cancer cell proliferation and metastasis through HDAC3/miR-139-3p pathway. Biochem. Genet.62, 3904–3919 (2024). [DOI] [PubMed] [Google Scholar]
  • 63.Yang, J. et al. Entero-toxigenic Bacteroides fragilis contributes to intestinal barrier injury and colorectal cancer progression by mediating the BFT/STAT3/ZEB2 pathway. Cell Cycle23, 70–82 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chen, Y. et al. KRAS mutations promote the intratumoral colonization of enterotoxigenic bacteroides fragilis in colorectal cancer through the regulation of the miRNA3655/SURF6/IRF7/IFNβ axis. Gut Microbes16, 2423043 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Pleguezuelos-Manzano, C. et al. Mutational signature in colorectal cancer caused by genotoxic pks(+) E. coli. Nature580, 269–273 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Cao, Y. et al. Enterotoxigenic Bacteroides fragilis promotes intestinal inflammation and malignancy by inhibiting exosome-packaged miR-149-3p. Gastroenterology161, 1552–1566.e1512 (2021). [DOI] [PubMed] [Google Scholar]
  • 67.DeStefano Shields, C. E. et al. Bacterial-driven inflammation and mutant BRAF expression combine to promote murine colon tumorigenesis that is sensitive to immune checkpoint therapy. Cancer Discov.11, 1792–1807 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Faïs, T., Delmas, J., Barnich, N., Bonnet, R. & Dalmasso, G. Colibactin: more than a new bacterial toxin. Toxins10, 151 (2018). [DOI] [PMC free article] [PubMed]
  • 69.He, Z. et al. Campylobacter jejuni promotes colorectal tumorigenesis through the action of cytolethal distending toxin. Gut68, 289–300 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Man, S. M. The clinical importance of emerging Campylobacter species. Nat. Rev. Gastroenterol. Hepatol.8, 669–685 (2011). [DOI] [PubMed] [Google Scholar]
  • 71.He, Z. et al. Campylobacter jejuni-derived cytolethal distending toxin promotes colorectal cancer metastasis. Cell Host Microbe32, 2080–2091.e2086 (2024). [DOI] [PubMed] [Google Scholar]
  • 72.Yu, J. et al. Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut66, 70–78 (2017). [DOI] [PubMed] [Google Scholar]
  • 73.Tsoi, H. et al. Peptostreptococcus anaerobius induces intracellular cholesterol biosynthesis in colon cells to induce proliferation and causes dysplasia in mice. Gastroenterology152, 1419–1433.e1415 (2017). [DOI] [PubMed] [Google Scholar]
  • 74.Zhang, Q., Goswami, S. & Yilmaz, O. Microbial regulation of ferroptosis in cancer. Nat. Cell Biol.26, 41–42 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Cui, W. et al. Gut microbial metabolite facilitates colorectal cancer development via ferroptosis inhibition. Nat. Cell Biol.26, 124–137 (2024). [DOI] [PubMed] [Google Scholar]
  • 76.Long, X. et al. Peptostreptococcus anaerobius promotes colorectal carcinogenesis and modulates tumour immunity. Nat. Microbiol.4, 2319–2330 (2019). [DOI] [PubMed] [Google Scholar]
  • 77.Gu, J. et al. Deciphering the mechanism of Peptostreptococcus anaerobius-induced chemoresistance in colorectal cancer: the important roles of MDSC recruitment and EMT activation. Front. Immunol.14, 1230681 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Liu, Y. et al. Peptostreptococcus anaerobius mediates anti-PD1 therapy resistance and exacerbates colorectal cancer via myeloid-derived suppressor cells in mice. Nat. Microbiol.9, 1467–1482 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Magat, E. M. et al. Clostridioides difficile antibody response of colorectal cancer patients versus clinically healthy individuals. Biosci. Microbiota Food Health39, 123–127 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Patel, R. K. et al. Incidence of colorectal cancer after intestinal infection due to Clostridioides difficile. World J. Oncol.15, 279–286 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Drewes, J. L. et al. Human colon cancer-derived Clostridioides difficile strains drive colonic tumorigenesis in mice. Cancer Discov.12, 1873–1885 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Loeven, N. A., Medici, N. P. & Bliska, J. B. The pyrin inflammasome in host-microbe interactions. Curr. Opin. Microbiol.54, 77–86 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sharma, D. et al. Pyrin Inflammasome regulates tight junction integrity to restrict colitis and tumorigenesis. Gastroenterology154, 948–964.e948 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cao, Y. et al. Commensal microbiota from patients with inflammatory bowel disease produce genotoxic metabolites. Science378, eabm3233 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Pasquereau-Kotula, E., Martins, M., Aymeric, L. & Dramsi, S. Significance of Streptococcus gallolyticus subsp. gallolyticus association with colorectal cancer. Front. Microbiol.9, 614 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kumar, R. et al. Streptococcus gallolyticus subsp. gallolyticus promotes colorectal tumor development. PLoS Pathog.13, e1006440 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Aymeric, L. et al. Colorectal cancer specific conditions promote Streptococcus gallolyticus gut colonization. Proc. Natl. Acad. Sci. USA115, E283–e291 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Fan, L. et al. A. Muciniphila suppresses colorectal tumorigenesis by inducing TLR2/NLRP3-mediated M1-Like TAMs. Cancer Immunol. Res.9, 1111–1124 (2021). [DOI] [PubMed] [Google Scholar]
  • 89.Zhu, J. et al. Amuc_1434 From Akkermansia muciniphila enhances CD8+ T cell-mediated anti-tumor immunity by suppressing PD-L1 in colorectal cancer. FASEB J.39, e70540 (2025). [DOI] [PubMed] [Google Scholar]
  • 90.Jiang, Y. et al. Acetyltransferase from Akkermansia muciniphila blunts colorectal tumourigenesis by reprogramming tumour microenvironment. Gut72, 1308–1318 (2023). [DOI] [PubMed] [Google Scholar]
  • 91.Zhang, L. et al. Akkermansia muciniphila inhibits tryptophan metabolism via the AhR/β-catenin signaling pathway to counter the progression of colorectal cancer. Int. J. Biol. Sci.19, 4393–4410 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Qu, S. et al. Excessive consumption of mucin by over-colonized Akkermansia muciniphila promotes intestinal barrier damage during malignant intestinal environment. Front. Microbiol.14, 1111911 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Birebent, R. et al. Surrogate markers of intestinal dysfunction associated with survival in advanced cancers. Oncoimmunology14, 2484880 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kitamoto, S. et al. The intermucosal connection between the mouth and gut in commensal pathobiont-driven colitis. Cell182, 447–462.e414 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wang, X. et al. Klebsiella aerogenes exacerbates colon tumorigenesis in the AOM/DSS-induced C57BL/6J mouse. Biochem. Biophys. Res. Commun.694, 149410 (2024). [DOI] [PubMed] [Google Scholar]
  • 96.Kaur, C. P., et al. Presence of polyketide synthase (PKS) gene and counterpart virulence determinants in Klebsiella pneumoniae strains enhances colorectal cancer progression in-vitro. Microorganisms. 11, 443 (2023). [DOI] [PMC free article] [PubMed]
  • 97.Yu, X., Niu, J. & Hu, J. Multidimensional bioinformatics analysis reveals the potential carcinogenic role of acrylamide in colorectal cancer. Ecotoxicol. Environ. Saf.298, 118281 (2025). [DOI] [PubMed] [Google Scholar]
  • 98.Collins, D., Hogan, A. M. & Winter, D. C. Microbial and viral pathogens in colorectal cancer. Lancet Oncol.12, 504–512 (2011). [DOI] [PubMed] [Google Scholar]
  • 99.Luo, S. et al. Gut virome profiling identifies an association between temperate phages and colorectal cancer promoted by Helicobacter pylori infection. Gut Microbes15, 2257291 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Lehti, T. A., Pajunen, M. I., Skog, M. S. & Finne, J. Internalization of a polysialic acid-binding Escherichia coli bacteriophage into eukaryotic neuroblastoma cells. Nat. Commun.8, 1915 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Nguyen, S. et al. Bacteriophage transcytosis provides a mechanism to cross epithelial cell layers. mBio8, e01874-17 (2017). [DOI] [PMC free article] [PubMed]
  • 102.Tun, H. M. et al. Gut virome in inflammatory bowel disease and beyond. Gut73, 350–360 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lin, Y. et al. Altered mycobiota signatures and enriched pathogenic Aspergillus rambellii are associated with colorectal cancer based on multicohort fecal metagenomic analyses. Gastroenterology163, 908–921 (2022). [DOI] [PubMed] [Google Scholar]
  • 104.Coker, O. O. et al. Enteric fungal microbiota dysbiosis and ecological alterations in colorectal cancer. Gut68, 654–662 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Yuan, K. et al. Intraneoplastic fungal dysbiosis is associated with colorectal cancer progression and host gene mutation. EBioMedicine113, 105608 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kiran, N. S. et al. The gastrointestinal mycobiome in inflammation and cancer: unraveling fungal dysbiosis, pathogenesis, and therapeutic potential. Med. Oncol.42, 195 (2025). [DOI] [PubMed] [Google Scholar]
  • 107.Zhang, Z. et al. IL-1β mediates Candida tropicalis-induced immunosuppressive function of MDSCs to foster colorectal cancer. Cell Commun. Signal.22, 408 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Huo, J. et al. Network pharmacology combined with transcriptomics reveals that Ganoderma lucidum spore and Sanghuangporus vaninii compound extract exerts anti-colorectal cancer effects via CYP24A1-mediated VDR pathway and TERT-mediated Wnt signaling pathway. J. Ethnopharmacol.348, 119820 (2025). [DOI] [PubMed] [Google Scholar]
  • 109.Guglietta, S., Li, X. & Saxena, D. Role of fungi in tumorigenesis: promises and challenges. Annu. Rev. Pathol.20, 459–482 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Wang, Y. et al. Bugs as drugs: Extended clinical applications and potential translational trends of Akkermansia muciniphila in oncology and comprehensive clinical investigation panorama of therapeutic microbiome modulators in gastric cancer treatment. Pharm. Res.207, 107331 (2024). [DOI] [PubMed] [Google Scholar]
  • 111.Wang, Y., Wang, S. & Li, N. Decoding the potential microbiome modulation strategies from bugs to drugs in pancreatic cancer: Insights from clinical trials. Cancer Lett.612, 217465 (2025). [DOI] [PubMed] [Google Scholar]
  • 112.Zhao, W. et al. Fecal microbiota transplantation plus tislelizumab and fruquintinib in refractory microsatellite stable metastatic colorectal cancer: an open-label, single-arm, phase II trial (RENMIN-215). EClinicalMedicine66, 102315 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Kim, Y. et al. Fecal microbiota transplantation improves anti-PD-1 inhibitor efficacy in unresectable or metastatic solid cancers refractory to anti-PD-1 inhibitor. Cell Host Microbe32, 1380–1393.e1389 (2024). [DOI] [PubMed] [Google Scholar]
  • 114.Lin, A. et al. Microbiota boost immunotherapy? A meta-analysis dives into fecal microbiota transplantation and immune checkpoint inhibitors. BMC Med.23, 341 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Lei, W., Zhou, K., Lei, Y., Li, Q. & Zhu, H. Gut microbiota shapes cancer immunotherapy responses. npj Biofilms Microbiomes11, 143 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Yu, A. I. et al. Gut microbiota modulate CD8 T cell responses to influence colitis-associated tumorigenesis. Cell Rep.31, 107471 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kammula, A. V., Schäffer, A. A., Rajagopal, P. S., Kurzrock, R. & Ruppin, E. Outcome differences by sex in oncology clinical trials. Nat. Commun.15, 2608 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Huang, H. et al. Accelerating the integration of China into the global development of innovative anticancer drugs. Lancet Oncol.23, e515–e520 (2022). [DOI] [PubMed] [Google Scholar]
  • 119.Wang, Y., Wang, S. & Li, N. Overcoming the quality gap in clinical trial landscape analysis: paving the way for impactful research. Int. J. Surg.10.1097/JS9.0000000000002770 (2025). [DOI] [PMC free article] [PubMed]
  • 120.García-González, A. P. et al. Bacterial metabolism affects the C. elegans response to cancer chemotherapeutics. Cell169, 431–441.e438 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Viaud, S. et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science342, 971–976 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Iida, N. et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science342, 967–970 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Alexander, J. L. et al. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat. Rev. Gastroenterol. Hepatol.14, 356–365 (2017). [DOI] [PubMed] [Google Scholar]
  • 124.He, Y. et al. Gut microbial metabolites facilitate anticancer therapy efficacy by modulating cytotoxic CD8(+) T cell immunity. Cell Metab.33, 988–1000.e1007 (2021). [DOI] [PubMed] [Google Scholar]
  • 125.Encarnação, J. C. et al. Butyrate, a dietary fiber derivative that improves irinotecan effect in colon cancer cells. J. Nutr. Biochem.56, 183–192 (2018). [DOI] [PubMed] [Google Scholar]
  • 126.An, J., Seok, H. & Ha, E. M. GABA-producing Lactobacillus plantarum inhibits metastatic properties and induces apoptosis of 5-FU-resistant colorectal cancer cells via GABA(B) receptor signaling. J. Microbiol.59, 202–216 (2021). [DOI] [PubMed] [Google Scholar]
  • 127.An, J. & Ha, E. M. Extracellular vesicles derived from Lactobacillus plantarum restore chemosensitivity through the PDK2-mediated glucose metabolic pathway in 5-FU-resistant colorectal cancer cells. J. Microbiol.60, 735–745 (2022). [DOI] [PubMed] [Google Scholar]
  • 128.Tang, Y. et al. Administration of probiotic mixture DM#1 ameliorated 5-fluorouracil-induced intestinal mucositis and dysbiosis in rats. Nutrition33, 96–104 (2017). [DOI] [PubMed] [Google Scholar]
  • 129.Quaresma, M. et al. Probiotic mixture containing Lactobacillus spp. and Bifidobacterium spp. attenuates 5-fluorouracil-induced intestinal mucositis in mice. Nutr. Cancer72, 1355–1365 (2020). [DOI] [PubMed] [Google Scholar]
  • 130.Osterlund, P. et al. Lactobacillus supplementation for diarrhoea related to chemotherapy of colorectal cancer: a randomised study. Br. J. Cancer97, 1028–1034 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Mi, H. et al. Bifidobacterium infantis ameliorates chemotherapy-induced intestinal mucositis via regulating T cell immunity in colorectal cancer rats. Cell. Physiol. Biochem.42, 2330–2341 (2017). [DOI] [PubMed] [Google Scholar]
  • 132.Ding, X. et al. Bacteroides fragilis promotes chemoresistance in colorectal cancer, and its elimination by phage VA7 restores chemosensitivity. Cell Host Microbe33, 941–956.e910 (2025). [DOI] [PubMed] [Google Scholar]
  • 133.Lang, T. et al. Combining gut microbiota modulation and chemotherapy by capecitabine-loaded prebiotic nanoparticle improves colorectal cancer therapy. Nat. Commun.14, 4746 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.van Vliet, M. J., Harmsen, H. J., de Bont, E. S. & Tissing, W. J. The role of intestinal microbiota in the development and severity of chemotherapy-induced mucositis. PLoS Pathog.6, e1000879 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Stein, A., Voigt, W. & Jordan, K. Chemotherapy-induced diarrhea: pathophysiology, frequency and guideline-based management. Ther. Adv. Med. Oncol.2, 51–63 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Haaz, M. C., Rivory, L., Jantet, S., Ratanasavanh, D. & Robert, J. Glucuronidation of SN-38, the active metabolite of irinotecan, by human hepatic microsomes. Pharmacol. Toxicol.80, 91–96 (1997). [DOI] [PubMed] [Google Scholar]
  • 137.Cheng, K. W. et al. Pharmacological inhibition of bacterial β-glucuronidase prevents irinotecan-induced diarrhea without impairing its antitumor efficacy in vivo. Pharmacol. Res.139, 41–49 (2019). [DOI] [PubMed] [Google Scholar]
  • 138.Wallace, B. D. et al. Structure and inhibition of microbiome β-glucuronidases essential to the alleviation of cancer drug toxicity. Chem. Biol.22, 1238–1249 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Wong, S. H. & Yu, J. Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat. Rev. Gastroenterol. Hepatol.16, 690–704 (2019). [DOI] [PubMed] [Google Scholar]
  • 140.Wei, S. C., Duffy, C. R. & Allison, J. P. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov.8, 1069–1086 (2018). [DOI] [PubMed] [Google Scholar]
  • 141.Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science357, 409–413 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Le, D. T. et al. PD-1 Blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med.372, 2509–2520 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science359, 91–97 (2018). [DOI] [PubMed] [Google Scholar]
  • 144.Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science359, 97–103 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Vétizou, M. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science350, 1079–1084 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Matson, V. et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science359, 104–108 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Peled, J. U. et al. Intestinal microbiota and relapse after hematopoietic-cell transplantation. J. Clin. Oncol.35, 1650–1659 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Chaput, N. et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann. Oncol.30, 2012 (2019). [DOI] [PubMed] [Google Scholar]
  • 149.Tay, C., Tanaka, A. & Sakaguchi, S. Tumor-infiltrating regulatory T cells as targets of cancer immunotherapy. Cancer Cell41, 450–465 (2023). [DOI] [PubMed] [Google Scholar]
  • 150.Gao, Y. et al. Correction to: Fusobacterium nucleatum enhances the efficacy of PD-L1 blockade in colorectal cancer. Signal Transduct. Target. Ther.6, 434 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Goodwin, A. C. et al. RA, Jr. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc. Natl. Acad. Sci. USA108, 15354–15359 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Blake, S. J., Wolf, Y., Boursi, B. & Lynn, D. J. Role of the microbiota in response to and recovery from cancer therapy. Nat. Rev. Immunol.24, 308–325 (2024). [DOI] [PubMed] [Google Scholar]
  • 153.Han, K. et al. Generation of systemic antitumour immunity via the in situ modulation of the gut microbiome by an orally administered inulin gel. Nat. Biomed. Eng.5, 1377–1388 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Zhang, S. L. et al. Pectin supplement significantly enhanced the anti-PD-1 efficacy in tumor-bearing mice humanized with gut microbiota from patients with colorectal cancer. Theranostics11, 4155–4170 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Messaoudene, M. et al. A natural polyphenol exerts antitumor activity and circumvents anti-PD-1 resistance through effects on the gut microbiota. Cancer Discov.12, 1070–1087 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Huang, J. et al. Ginseng polysaccharides alter the gut microbiota and kynurenine/tryptophan ratio, potentiating the antitumour effect of antiprogrammed cell death 1/programmed cell death ligand 1 (anti-PD-1/PD-L1) immunotherapy. Gut71, 734–745 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Naz, S. S. & Zafar, S. Analyzing potential of next-generation probiotics in cancer management. Mol. Biol. Rep.52, 646 (2025). [DOI] [PubMed] [Google Scholar]
  • 158.Lam, K. C. et al. Microbiota triggers STING-type I IFN-dependent monocyte reprogramming of the tumor microenvironment. Cell184, 5338–5356.e5321 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Derrien, M., Vaughan, E. E., Plugge, C. M. & de Vos, W. M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol.54, 1469–1476 (2004). [DOI] [PubMed] [Google Scholar]
  • 160.Desai, M. S. et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell167, 1339–1353.e1321 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Swanson, K. S. et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat. Rev. Gastroenterol. Hepatol.17, 687–701 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Si, W. et al. Lactobacillus rhamnosus GG induces cGAS/STING- dependent type I interferon and improves response to immune checkpoint blockade. Gut71, 521–533 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Zhang, S. L. et al. Lacticaseibacillus paracasei sh2020 induced antitumor immunity and synergized with anti-programmed cell death 1 to reduce tumor burden in mice. Gut Microbes14, 2046246 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Tanoue, T. et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature565, 600–605 (2019). [DOI] [PubMed] [Google Scholar]
  • 165.Dizman, N. et al. Nivolumab plus ipilimumab with or without live bacterial supplementation in metastatic renal cell carcinoma: a randomized phase 1 trial. Nat. Med.28, 704–712 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Ebrahimi, H. et al. Cabozantinib and nivolumab with or without live bacterial supplementation in metastatic renal cell carcinoma: a randomized phase 1 trial. Nat. Med.30, 2576–2585 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Derosa, L. et al. Intestinal Akkermansia muciniphila predicts clinical response to PD-1 blockade in patients with advanced non-small-cell lung cancer. Nat. Med.28, 315–324 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Spencer, C. N. et al. Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Science374, 1632–1640 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Arthur, J. C. et al. VSL#3 probiotic modifies mucosal microbial composition but does not reduce colitis-associated colorectal cancer. Sci. Rep.3, 2868 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Spreafico, A. et al. First-in-class Microbial Ecosystem Therapeutic 4 (MET4) in combination with immune checkpoint inhibitors in patients with advanced solid tumors (MET4-IO trial). Ann. Oncol.34, 520–530 (2023). [DOI] [PubMed] [Google Scholar]
  • 171.Montalban-Arques, A. et al. Commensal Clostridiales strains mediate effective anti-cancer immune response against solid tumors. Cell Host Microbe29, 1573–1588.e1577 (2021). [DOI] [PubMed] [Google Scholar]
  • 172.Zhuo, Q. et al. Lysates of Lactobacillus acidophilus combined with CTLA-4-blocking antibodies enhance antitumor immunity in a mouse colon cancer model. Sci. Rep.9, 20128 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Morishita, M. et al. Characterizing different probiotic-derived extracellular vesicles as a novel adjuvant for immunotherapy. Mol. Pharm.18, 1080–1092 (2021). [DOI] [PubMed] [Google Scholar]
  • 174.Kawanabe-Matsuda, H. et al. Dietary lactobacillus-derived exopolysaccharide enhances immune-checkpoint blockade therapy. Cancer Discov.12, 1336–1355 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Griffin, M. E. et al. Enterococcus peptidoglycan remodeling promotes checkpoint inhibitor cancer immunotherapy. Science373, 1040–1046 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Kang, X. et al. Roseburia intestinalis generated butyrate boosts anti-PD-1 efficacy in colorectal cancer by activating cytotoxic CD8(+) T cells. Gut72, 2112–2122 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Fong, W. et al. Lactobacillus gallinarum-derived metabolites boost anti-PD1 efficacy in colorectal cancer by inhibiting regulatory T cells through modulating IDO1/Kyn/AHR axis. Gut72, 2272–2285 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Canale, F. P. et al. Metabolic modulation of tumours with engineered bacteria for immunotherapy. Nature598, 662–666 (2021). [DOI] [PubMed] [Google Scholar]
  • 179.Cubillos-Ruiz, J. R. & Cubillos-Ruiz, A. Engineered bacteria recycle tumor metabolic waste to boost immunotherapy. Cell Host Microbe29, 1725–1727 (2021). [DOI] [PubMed] [Google Scholar]
  • 180.Geiger, R. et al. L-Arginine Modulates T Cell Metabolism and Enhances Survival and Anti-tumor Activity. Cell167, 829–842.e813 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Gurbatri, C. R. et al. Engineered probiotics for local tumor delivery of checkpoint blockade nanobodies. Sci. Transl. Med.12, eaax0876 (2020). [DOI] [PMC free article] [PubMed]
  • 182.Borody, T. J. & Khoruts, A. Fecal microbiota transplantation and emerging applications. Nat. Rev. Gastroenterol. Hepatol.9, 88–96 (2011). [DOI] [PubMed] [Google Scholar]
  • 183.Shaikh, F. Y. et al. Murine fecal microbiota transfer models selectively colonize human microbes and reveal transcriptional programs associated with response to neoadjuvant checkpoint inhibitors. Cancer Immunol. Immunother.71, 2405–2420 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Huang, J. et al. Metagenomic and metabolomic analyses reveal synergistic effects of fecal microbiota transplantation and anti-PD-1 therapy on treating colorectal cancer. Front. Immunol.13, 874922 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Goc, J. et al. Dysregulation of ILC3s unleashes progression and immunotherapy resistance in colon cancer. Cell184, 5015–5030.e5016 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Baruch, E. N. et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science371, 602–609 (2021). [DOI] [PubMed] [Google Scholar]
  • 187.Davar, D. et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science371, 595–602 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Routy, B. et al. Fecal microbiota transplantation plus anti-PD-1 immunotherapy in advanced melanoma: a phase I trial. Nat. Med.29, 2121–2132 (2023). [DOI] [PubMed] [Google Scholar]
  • 189.Yang, Y. et al. Fecal microbiota transplantation: no longer cinderella in tumour immunotherapy. EBioMedicine100, 104967 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Bilinski, J. et al. Eosinophilic gastroenteritis and graft-versus-host disease induced by transmission of Norovirus with fecal microbiota transplant. Transpl. Infect. Dis.23, e13386 (2021). [DOI] [PubMed] [Google Scholar]
  • 191.DeFilipp, Z. et al. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N. Engl. J. Med.381, 2043–2050 (2019). [DOI] [PubMed] [Google Scholar]
  • 192.Vendrik, K. E. W. et al. Periodic screening of donor faeces with a quarantine period to prevent transmission of multidrug-resistant organisms during faecal microbiota transplantation: a retrospective cohort study. Lancet Infect. Dis.21, 711–721 (2021). [DOI] [PubMed] [Google Scholar]
  • 193.Jaffray, D. A. Image-guided radiotherapy: from current concept to future perspectives. Nat. Rev. Clin. Oncol.9, 688–699 (2012). [DOI] [PubMed] [Google Scholar]
  • 194.Gerassy-Vainberg, S. et al. Radiation induces proinflammatory dysbiosis: transmission of inflammatory susceptibility by host cytokine induction. Gut67, 97–107 (2018). [DOI] [PubMed] [Google Scholar]
  • 195.Johnson, L. B. et al. Radiation enteropathy and leucocyte-endothelial cell reactions in a refined small bowel model. BMC Surg.4, 10 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Heimesaat, M. M. et al. MyD88/TLR9 mediated immunopathology and gut microbiota dynamics in a novel murine model of intestinal graft-versus-host disease. Gut59, 1079–1087 (2010). [DOI] [PubMed] [Google Scholar]
  • 197.Jenq, R. R. et al. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J. Exp. Med.209, 903–911 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Fernandes, A., Oliveira, A., Soares, R. & Barata, P. The effects of ionizing radiation on gut microbiota, a systematic review. Nutrients13, 3025 (2021). [DOI] [PMC free article] [PubMed]
  • 199.Yang, J. et al. Pre-treatment with Bifidobacterium infantis and its specific antibodies enhance targeted radiosensitization in a murine model for lung cancer. J. Cancer Res. Clin. Oncol.147, 411–422 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Montassier, E. et al. Chemotherapy-driven dysbiosis in the intestinal microbiome. Aliment. Pharmacol. Ther.42, 515–528 (2015). [DOI] [PubMed] [Google Scholar]
  • 201.Uribe-Herranz, M. et al. Gut microbiota modulate dendritic cell antigen presentation and radiotherapy-induced antitumor immune response. J. Clin. Investig.130, 466–479 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Shiao, S. L. et al. Commensal bacteria and fungi differentially regulate tumor responses to radiation therapy. Cancer Cell39, 1202–1213.e1206 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Dong, J. et al. Roseburia intestinalis sensitizes colorectal cancer to radiotherapy through the butyrate/OR51E1/RALB axis. Cell Rep.43, 114407 (2024). [DOI] [PubMed] [Google Scholar]
  • 204.Chung, L. et al. Bacteroides fragilis toxin coordinates a pro-carcinogenic inflammatory cascade via targeting of colonic epithelial cells. Cell Host Microbe23, 421 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Park, S. Y. et al. The JAK2/STAT3/CCND2 axis promotes colorectal cancer stem cell persistence and radioresistance. J. Exp. Clin. Cancer Res.38, 399 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Peng, X. et al. Streptococcus salivarius K12 alleviates oral mucositis in patients undergoing radiotherapy for malignant head and neck tumors: a randomized controlled trial. J. Clin. Oncol.42, 1426–1435 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Cui, M. et al. Faecal microbiota transplantation protects against radiation-induced toxicity. EMBO Mol. Med.9, 448–461 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Xiao, H. W. et al. Gut microbiota-derived indole 3-propionic acid protects against radiation toxicity via retaining acyl-CoA-binding protein. Microbiome8, 69 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Reinhardt, C. et al. Tissue factor and PAR1 promote microbiota-induced intestinal vascular remodelling. Nature483, 627–631 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Scales, B. S. & Huffnagle, G. B. The microbiome in wound repair and tissue fibrosis. J. Pathol.229, 323–331 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Stappenbeck, T. S., Hooper, L. V. & Gordon, J. I. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. Proc. Natl. Acad. Sci. USA99, 15451–15455 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Gan, B. S., Kim, J., Reid, G., Cadieux, P. & Howard, J. C. Lactobacillus fermentum RC-14 inhibits Staphylococcus aureus infection of surgical implants in rats. J. Infect. Dis.185, 1369–1372 (2002). [DOI] [PubMed] [Google Scholar]
  • 213.Harrison, F., Paul, J., Massey, R. C. & Buckling, A. Interspecific competition and siderophore-mediated cooperation in Pseudomonas aeruginosa. ISME J.2, 49–55 (2008). [DOI] [PubMed] [Google Scholar]
  • 214.Schardey, H. M. et al. FW. The prevention of anastomotic leakage after total gastrectomy with local decontamination. A prospective, randomized, double-blind, placebo-controlled multicenter trial. Ann. Surg.225, 172–180 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Wu, L. et al. High-molecular-weight polyethylene glycol prevents lethal sepsis due to intestinal Pseudomonas aeruginosa. Gastroenterology126, 488–498 (2004). [DOI] [PubMed] [Google Scholar]
  • 216.Shogan, B. D. et al. Intestinal anastomotic injury alters spatially defined microbiome composition and function. Microbiome2, 35 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Cong, J. et al. A pilot study: changes of gut microbiota in post-surgery colorectal cancer patients. Front. Microbiol.9, 2777 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Alexander, J. L. et al. Pathobionts in the tumour microbiota predict survival following resection for colorectal cancer. Microbiome11, 100 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Komen, N. et al. Polymerase chain reaction for Enterococcus faecalis in drain fluid: the first screening test for symptomatic colorectal anastomotic leakage. The Appeal-study: analysis of parameters predictive for evident anastomotic leakage. Int. J. Colorectal Dis.29, 15–21 (2014). [DOI] [PubMed] [Google Scholar]
  • 220.Pitsillides, L., Pellino, G., Tekkis, P. & Kontovounisios, C. The effect of perioperative administration of probiotics on colorectal cancer surgery outcomes. Nutrients13, 1451 (2021). [DOI] [PMC free article] [PubMed]
  • 221.Araújo, M. M., Montalvão-Sousa, T. M., Teixeira, P. D. C., Figueiredo, A. & Botelho, P. B. The effect of probiotics on postsurgical complications in patients with colorectal cancer: a systematic review and meta-analysis. Nutr. Rev.81, 493–510 (2023). [DOI] [PubMed] [Google Scholar]
  • 222.Baas, F. S., Brusselaers, N., Nagtegaal, I. D., Engstrand, L. & Boleij, A. Navigating beyond associations: opportunities to establish causal relationships between the gut microbiome and colorectal carcinogenesis. Cell Host Microbe32, 1235–1247 (2024). [DOI] [PubMed] [Google Scholar]
  • 223.Chen, L. et al. The long-term genetic stability and individual specificity of the human gut microbiome. Cell184, 2302–2315.e2312 (2021). [DOI] [PubMed] [Google Scholar]
  • 224.Valles-Colomer, M. et al. The person-to-person transmission landscape of the gut and oral microbiomes. Nature614, 125–135 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Schupack, D. A., Mars, R. A. T., Voelker, D. H., Abeykoon, J. P. & Kashyap, P. C. The promise of the gut microbiome as part of individualized treatment strategies. Nat. Rev. Gastroenterol. Hepatol.19, 7–25 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Hu, M. et al. Gut microbial-derived N-acetylmuramic acid alleviates colorectal cancer via the AKT1 pathway. Gut74, 1230–1245 (2025). [DOI] [PubMed]
  • 227.Allaband, C. et al. Time of sample collection is critical for the replicability of microbiome analyses. Nat. Metab.6, 1282–1293 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Schloss, P. D. Identifying and overcoming threats to reproducibility, replicability, robustness, and generalizability in microbiome research. mBio9, e00525-18 (2018). [DOI] [PMC free article] [PubMed]
  • 229.Mirzayi, C. et al. Reporting guidelines for human microbiome research: the STORMS checklist. Nat. Med.27, 1885–1892 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Ferdous, T. et al. The rise to power of the microbiome: power and sample size calculation for microbiome studies. Mucosal Immunol.15, 1060–1070 (2022). [DOI] [PubMed] [Google Scholar]
  • 231.Baysoy, A., Bai, Z., Satija, R. & Fan, R. The technological landscape and applications of single-cell multi-omics. Nat. Rev. Mol. Cell Biol.24, 695–713 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Chetty, A. & Blekhman, R. Multi-omic approaches for host-microbiome data integration. Gut Microbes16, 2297860 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Li, R. et al. Single-cell transcriptomic analysis deciphers heterogenous cancer stem-like cells in colorectal cancer and their organ-specific metastasis. Gut73, 470–484 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Galeano Niño, J. L. et al. Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer. Nature611, 810–817 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Lötstedt, B., Stražar, M., Xavier, R., Regev, A. & Vickovic, S. Spatial host-microbiome sequencing reveals niches in the mouse gut. Nat. Biotechnol.42, 1394–1403 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Pereira-Marques, J., Ferreira, R. M. & Figueiredo, C. A metatranscriptomics strategy for efficient characterization of the microbiome in human tissues with low microbial biomass. Gut Microbes16, 2323235 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Agustinho, D. P. et al. Unveiling microbial diversity: harnessing long-read sequencing technology. Nat. Methods21, 954–966 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Parida, S., Siddharth, S., Xia, Y. & Sharma, D. Concomitant analyses of intratumoral microbiota and genomic features reveal distinct racial differences in breast cancer. npj Breast Cancer9, 4 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Gihawi, A. et al. Major data analysis errors invalidate cancer microbiome findings. mBio14, e0160723 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Sepich-Poore, G. D. et al. Robustness of cancer microbiome signals over a broad range of methodological variation. Oncogene43, 1127–1148 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Novielli, P. et al. Explainable artificial intelligence for microbiome data analysis in colorectal cancer biomarker identification. Front. Microbiol.15, 1348974 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Freeman, K. et al. Algorithm based smartphone apps to assess risk of skin cancer in adults: systematic review of diagnostic accuracy studies. BMJ368, m127 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Chang, T. G., Park, S., Schäffer, A. A., Jiang, P. & Ruppin, E. Hallmarks of artificial intelligence contributions to precision oncology. Nat. Cancer6, 417–431 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Lipkova, J. et al. Artificial intelligence for multimodal data integration in oncology. Cancer Cell40, 1095–1110 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Derosa, L. et al. Custom scoring based on ecological topology of gut microbiota associated with cancer immunotherapy outcome. Cell187, 3373–3389.e3316 (2024). [DOI] [PubMed] [Google Scholar]
  • 246.Teixeira, M. et al. A review of machine learning methods for cancer characterization from microbiome data. npj Precis. Oncol.8, 123 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Wu, S. et al. Therapeutic m(6)A eraser ALKBH5 mRNA-loaded exosome-liposome hybrid nanoparticles inhibit progression of colorectal cancer in preclinical tumor models. ACS Nano17, 11838–11854 (2023). [DOI] [PubMed] [Google Scholar]
  • 248.Wu, S. et al. Matairesinol nanoparticles restore chemosensitivity and suppress colorectal cancer progression in preclinical models: role of lipid metabolism reprogramming. Nano Lett.23, 1970–1980 (2023). [DOI] [PubMed] [Google Scholar]
  • 249.Wei, Y. et al. The challenge and opportunity of gut microbiota-targeted nanomedicine for colorectal cancer therapy. iMeta3, e213 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Gurbatri, C. R. et al. Engineering tumor-colonizing E. coli Nissle 1917 for detection and treatment of colorectal neoplasia. Nat. Commun.15, 646 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.An, B. C. et al. Colorectal cancer therapy using a Pediococcus pentosaceus SL4 drug delivery system secreting lactic acid bacteria-derived protein p8. Mol. Cells42, 755–762 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Zheng, D. W. et al. Prebiotics-encapsulated probiotic spores regulate gut microbiota and suppress colon cancer. Adv. Mater.32, e2004529 (2020). [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

41522_2025_818_MOESM1_ESM.xlsx (119.9KB, xlsx)

Table S1: The clinical trials of microbiome modulators in CRC

Data Availability Statement

No datasets were generated or analyzed during the current study.


Articles from NPJ Biofilms and Microbiomes are provided here courtesy of Nature Publishing Group

RESOURCES