Skip to main content
Oncology Letters logoLink to Oncology Letters
. 2025 Sep 16;30(5):527. doi: 10.3892/ol.2025.15273

Tumor-associated microbiota: Multi-cancer landscape, mechanistic insights and clinical translation (Review)

Jindi Yang 1, Yige Dong 1, Yi Chen 1, Hong Liang 2, Shengyu Rong 1, Zhe Liu 1, Qiulei Lang 1,2,
PMCID: PMC12457931  PMID: 41000593

Abstract

Intratumoral microbiota, a critical component of the tumor microenvironment, notably impacts tumor progression through various complex mechanisms such as metabolic regulation, immune system remodeling and genotoxicity. The present review focuses on eight prevalent solid tumors (breast, colorectal, lung, pancreatic, gastric, ovarian, prostate cancers and melanoma), detailing the intratumoral microbial compositional heterogeneity within these malignancies. The present review analyzes the heterogeneous carcinogenic mechanisms mediated by these microorganisms, including gene instability, immune microenvironment remodeling and metabolic intervention. The diagnostic value of microbial markers in liquid biopsy and in situ tissue detection is explored, and the potential for synergistic strategies combining microbial-targeted therapy and immunotherapy is discussed. Finally, the review suggests future research directions, such as spatiotemporal dynamic analysis and organoid-microorganism co-culture, offering new insights for precise cancer treatment.

Keywords: tumor microorganisms, tumor microenvironment, biomarkers, cancer, immunotherapy

1. Introduction

The human microbiota, consisting of bacteria, fungi, viruses and protists, numbers in the tens of trillions, matching the count of human cells (1). The traditional theory is that human microorganisms mainly colonize mucosa-associated niches, including the gut, skin, oral cavity and reproductive tract (2). Previous studies have reported that previously considered sterile deep tissues (such as the lungs, mammary glands, pancreas, prostate and kidneys) also harbor low-biomass microbial communities (3). In normal physiological states, these microorganisms support the host's microecological balance by modulating local immunity, metabolism and barrier functions (4).

The intratumoral microbiota refers to the microbial community colonizing the tumor tissue (5). Previous groundbreaking research has demonstrated the presence of diverse microorganisms within tumor tissues, which were previously considered sterile environments, and their abundance is notably associated with tumorigenesis (6,7). These microorganisms colonize the tumor microenvironment (TME) through various routes, including mucosal disruption, migration from adjacent tissues and hematogenous invasion (8). The TME, composed of immune cells, stromal cells, cytokines and hypoxic regions, provides a colonization niche for intratumoral microorganisms due to its pathological characteristics (9). The colonization of intratumoral microorganisms depends on the TME's abnormal vascular leakage, immunosuppressive state, local hypoxic microdomains and eutrophic niches (10). As an endogenous component of the TME, tumor microorganisms drive malignant progression through mechanisms such as cell phenotype reprogramming, paracrine signal mediation and immune microenvironment regulation (8,11).

Beyond microbial influences, intrinsic host factors such as RNA helicases also critically shape the TME. A recent pan-cancer analysis identified DEAD-box helicase 1 (DDX1), an RNA helicase, as a prognostic marker with dual roles across cancers: Low DDX1 expression is associated with improved survival in renal carcinomas, while high expression drives poor prognosis in breast, liver and adrenal cancers by modulating immune infiltration, DNA repair pathways and phosphorylation-dependent signaling (12). This highlights the complex interplay between host-derived molecular machinery and microbial ecosystems in tumor evolution. Components in the TME can promote tumor invasion and metastasis by inducing epithelial-mesenchymal transition, enhancing the proteolytic activity of matrix metalloproteinases and inhibiting antitumor immunity (13). The interaction between tumor cells and the TME creates a vicious cycle that facilitates both local invasion and metastasis (14). Tumor-associated microorganisms influence tumor susceptibility, treatment response and malignant progression via host cell-microorganism interactions (15). The core carcinogenic mechanisms of intratumoral microorganisms include genetic instability, immune microenvironment remodeling and metabolic intervention (16,17).

The study of intratumoral microorganisms has a long history, dating back to the mid-20th century. Microorganisms were first detected in tumor tissues in the 1950s (5). In 1911, Rous (18) discovered that the Rous sarcoma virus could induce malignant tumors, and the Epstein-Barr virus was later identified in Burkitt lymphoma. A notable milestone was reached in 1983 when Warren and Marshall (19) successfully cultured Helicobacter pylori and demonstrated its pathogenic role in gastric cancer. The advent of high-throughput sequencing technology at the start of the 21st century has propelled rapid advancements in this field. In 2020, Nejman et al (7) analyzed 1,526 samples to reveal tumor-type-specific microbial compositions. More recently, in 2022, Narunsky-Haziza et al (20) mapped the fungal distribution across 17 different tumor types. During this period, microbial intervention strategies have evolved from early erysipelas therapy to engineered strain treatments (21,22).

The present review examines eight prevalent solid tumors: Breast, colorectal, lung, pancreatic, gastric, ovarian, prostate cancers and melanoma. These cancers are pivotal in tumor microbiome research due to their epidemiological impact, sample availability, defined mechanisms and clinical relevance. This selectivity not only reflects the current development status of the field, but also reflects the efficiency of academic communication. These eight types of cancer cover a notable body of the global cancer burden, and the research results have far-reaching impact on public health and have clear evidence of microbial associations. Investigating tumor-associated microorganisms has challenged the notion of tumors as sterile environments, introducing a paradigm shift from targeting cancer cells alone to regulating the host-microbe symbiotic network. Consequently, tumor-associated microorganisms have emerged as a crucial area in cancer therapy research, with numerous studies exploring their diagnostic value and therapeutic potential.

2. Differences in microbial communities among different cancer types

The intratumoral microbiota displays considerable heterogeneity across cancer types, with compositional variations primarily influenced by the TME, host characteristics, dietary habits, environmental exposures and the intrinsic properties of the microorganisms (23). Proteobacteria, Firmicutes and Actinobacteria are commonly enriched in most tumors (7,24,25). However, the same microbial genus may have opposite effects in different cancer types. For example, Pseudomonas serves a pro-tumorigenic role in breast cancer, colorectal cancer (CRC), lung cancer and gastric cancer, but colonizes the prostate as a commensal bacterium (7,26,27). Acinetobacter spp. promotes melanoma progression, exhibits commensalism in lung tissue and is enriched in prostate cancer where it appear to suppress tumorigenesis (28,29). Notably, commensal Veillonella spp. species from healthy lung tissues exhibit aberrant enrichment in gastric cancer tissues, with elevated abundance notable associating with adverse patient prognosis, which may facilitate gastric mucosal barrier disruption through lipopolysaccharide-mediated activation of the toll-like receptor (TLR) 2/4-NF-κB signaling pathway (27,30).

Breast cancer

Previous research has revealed that the microbial diversity and abundance in breast cancer tissues surpass those in corresponding normal breast tissues, with adjacent normal tissues exhibiting an intermediate composition (7). Various factors such as ethnicity, tumor stage and molecular subtype contribute to the heterogeneity of the breast microbiota (31). Methylobacterium radiotolerans and Staphylococcus consistently emerge as the predominant bacteria in breast tumors based on multi-omics analyses (7,3234). Additionally, Tzeng et al (26) further demonstrated that Lacibacter and Ezakiella are notably enriched in breast cancer tissues, and their abundance increases with the elevation of tumor stage. By contrast, the relative abundances of Sphingomonas yanoikuyae and Acetobacter aceti are higher in normal breast tissues, while Anaerococcus, Caulobacter and Streptococcus are notably absent in breast cancer (33,34). Despite these insights into microbial community characteristics, further research is required to elucidate the precise mechanisms by which these microorganisms influence processes such as breast cancer cell dissemination, intravasation and extravasation.

CRC

Meta-genomic research has revealed substantial alterations in the microbial community composition within tumor tissues of patients with CRC compared with those of healthy individuals (35). Fusobacterium nucleatum has consistently emerged as a prominent member of the microbiota associated with CRC (36). Additionally, Bacteroides fragilis, Parvimonas and Bacteroides have been notably observed to be more abundant in CRC tissues (37,38). Nevertheless, Repass (39) performed a comparative analysis of tumor and adjacent normal tissues from the same cohort of patients, demonstrating that Fusobacterium nucleatum was not prevalent in the majority of CRC samples. Moreover, the authors found no marked distinction in the overall microbiota composition between the two tissue types. This suggests that discrepancies in findings may be attributed to variations in sample origins, population diversity and methodological disparities.

Lung cancer

The lungs undergo continuous gas exchange with the external environment, making their microbial communities vulnerable to influences from the oral, nasal and gut microbiota through ‘ascending’ or ‘hematogenous’ routes (40). Meta-genomic studies have verified marked dysbiosis in the lung tissues of individuals with lung cancer, characterized by heightened local inflammatory responses, elevated microbial quantities and the proliferation of specific pro-inflammatory bacteria (41). Notably, potential pathogens such as Brevundimonas and Escherichia are markedly more abundant in cancerous tissues compared with adjacent normal tissues, whereas Corynebacterium, Lachnoanaerobaculum and Halomonas, prevalent in healthy individuals, are markedly diminished (28). Subsequent investigations indicated that Streptococcus and Peptoniphilus are associated with an increased lung cancer risk, whereas the presence of Aggregatibacter offers a protective effect (42).

Pancreatic cancer

The local pancreatic microbiota is closely associated with the susceptibility, progression and treatment response of pancreatic ductal adenocarcinoma (PDAC) (43,44). Propionibacterium acnes, one of the earliest bacteria isolated from pancreatic cancer tissues, can induce chronic inflammation and provide an inflammatory microenvironment for tumorigenesis (45). Meta-genomic analysis has revealed that Gammaproteobacteria is the dominant bacterial class in human pancreatic tumors (46). In a PDAC mouse model, Bacteroides and Parabacteroides were notably enriched in tumor tissues, suggesting their potential involvement in cancer progression (47).

It was further found that intratumoral microbial diversity is associated with prognosis in patients with PDAC. Riquelme et al (48) reported that long-term survivors exhibited markedly higher tumor microbial diversity compared with short-term survivors. Moreover, the co-existence and high abundance of Pseudoxanthomonas, Streptomyces, Saccharopolyspora and Bacillus clausii could serve as an independent marker for predicting long-term survival.

Gastric cancer

Gastric microbiota dysbiosis serves a crucial role in the progression of gastric cancer. Helicobacter pylori (H. pylori) has been designated as a Group I carcinogen by the World Health Organization. However, only ~3% of individuals infected with H. pylori ultimately develop gastric cancer (49,50). Apart from H. pylori, various meta-genomic studies have identified specific microbiota associated with gastric cancer, including Peptostreptococcus stomatis, Streptococcus anginosus, Parvimonas micra, Slackia exigua and Dialister pneumosintes (51). Throughout the continuum from gastritis-gastric adenoma-early-stage gastric cancer-advanced stage gastric cancer, the microbiota composition undergoes dynamic changes. Akkermansia and Lachnospiraceae NK4A136 dominate the gastritis stage, whereas Lactobacillus and Veillonella markedly increase in abundance in gastric cancer tissues compared with normal tissues (30,52). The aforementioned distinctive microbial dysbiosis may expedite the carcinogenic process through mechanisms such as inflammation promotion, metabolic reprogramming and immune suppression. Therefore, monitoring the composition and function of the gastric microbiota holds notable clinical value for the early diagnosis and risk assessment of gastric cancer.

Ovarian cancer

Ovarian cancer ranks among the most aggressive gynecological malignancies. High-grade serous ovarian cancer is considered to originate from either the ovarian surface epithelium or fallopian tubes (53). Previous studies have revealed that dysregulation of the local microbiota may serve a regulatory role in the development and progression of ovarian cancer (54,55). Meta-genomic analyses have identified Roseomonas mucosa and Sphingomonas US_602 as being notably enriched in ovarian tumor tissues, indicating their potential as key microbial players in this disease (24). Additionally, human papillomavirus (HPV) infection has been proposed as a risk factor for ovarian cancer, with higher expression levels of high-risk HPV observed in malignant tissues compared with adjacent normal tissues (56). However, a study by Ingerslev et al (57) found no notable association between high-risk HPV and epithelial ovarian cancer in Caucasian patients, suggesting it may not be related to ethnic differences.

Prostate cancer

Prostate cancer is the most prevalent malignant tumor in men (58). Previous research has demonstrated that prostate tissue is not sterile, with microbial DNA identified in tumor samples from 87% of patients with prostate cancer, implicating the local microbiota in the disease's onset and progression (59,60). Multi-omics analyses reveal elevated levels of Escherichia and Propionibacterium in prostate cancer, suggesting their potential role in tumor development (61). Additionally, liquid biopsy indicates markedly increased levels of Streptococcus, Peptostreptococcus and Haemophilus in the urine of patients with prostate cancer (62). Furthermore, markers of parasites such as Toxoplasma and Plasmodium are detected in prostate cancer tissues, potentially contributing to tumorigenesis through chromosomal damage and reactive oxygen species generation (63).

Melanoma

Melanoma arises from the malignant transformation of melanocytes and is the deadliest form of skin cancer (64). The skin microbiota varies notably by location: Staphylococcus dominates sebaceous regions, Corynebacterium is prevalent in moist areas and β-Proteobacteria are found in dry regions (65,66). In melanoma tissues, Enterobacter and Streptococcus are notably enriched, facilitating tumor immune evasion by inhibiting CD8+ T cell infiltration and reducing chemokine expression (29,67). Notably, Kozmin et al (68) discovered that a commensal Staphylococcus epidermidis strain producing 6-N-hydroxyaminopurine could markedly inhibit B16F10 melanoma growth and decrease ultraviolet-induced tumor incidence, suggesting a novel approach for microbiota-driven prevention and treatment of skin cancer.

3. Mechanisms of heterogeneity of different cancer microorganisms

Intratumoral microbiota varies markedly across tumor types, affecting key biological functions such as metabolic reprogramming, immune regulation, metastatic potential and epigenetic modification. These microorganisms can either promote or inhibit tumor progression by activating or suppressing specific signaling pathways, modulating the immunosuppressive microenvironment, enhancing inflammatory responses and altering metabolic processes.

Genetic instability

Intratumoral microorganisms can drive genomic instability through a dual-pathway mechanism involving direct genotoxicity and indirect activation, with the specific mechanisms being strain-dependent (23). In CRC, Escherichia coli carrying the pks gene island secretes the genotoxin colibactin, inducing somatic base substitutions and insertion or deletion mutations, while the cyto-lethal distending toxin produced by Campylobacter can cause DNA double-strand breaks (69). Aflatoxin B1 (AFB1) is metabolized by cytochrome p450 to the reactive AFBO metabolite, which blocks nucleotide excision repair and induces TP53 mutations, promoting carcinogenesis in multiple organs (70). In CRC, Fusobacterium nucleatum activates the E-cadherin/β-catenin signaling pathway via the FadA adhesin, upregulates checkpoint kinase 2 and induces DNA damage (71). Elevated β-glucuronidase in the breast cancer microenvironment can catalyze the release of reactive intermediates, indirectly causing DNA damage (8). In the intestinal mucosa of patients with familial adenomatous polyposis, enterotoxigenic Bacteroides fragilis and Escherichia coli were found to colonize synergistically, accelerating early-stage CRC development through DNA damage and inflammatory pathways (72).

Remodeling of the immune microenvironment

Intratumoral microorganisms colonize TME, influencing immune remodeling and accelerating tumor progression (73). Their viable bacteria, residues and metabolites collectively regulate immune cell functions and inflammatory responses, enhancing immune suppression and promoting pro-tumor phenotypes (74). Enterobacteriaceae and Pseudomonadaceae in breast and pancreatic cancers directly promote tumor cell proliferation by activating the NF-κB/TLR4 inflammatory axis and upregulating the PI3K/Akt signaling pathway (34,75). In CRC, Fusobacterium nucleatum increases intracellular Ca2+, facilitates E-cadherin and Krüppel-like factor 4 (KLF4) interaction, promotes KLF4 nuclear translocation and upregulates integrin a5 transcription, driving proliferation, invasion and metastasis (76). In pancreatic cancer, Campylobacter, Selenomonas and Clostridium difficile inhibit immune cell activity via the MET proto-oncogene, receptor tyrosine kinase-protein tyrosine kinase 2 and programmed cell death protein 1 (PD-1) pathways, enhancing tumor invasiveness; these genera, rare in normal tissues, are potential diagnostic markers (77). Fusobacterium upregulates pathways such as cytotoxic T-lymphocyte associated protein 4, JAK-STAT, TNF and PI3K-AKT-mTOR, creating an immunosuppressive microenvironment and remodeling the transcriptome to promote oral squamous cell carcinoma progression (11). Conversely, microorganisms can inhibit tumor progression; for instance, Propionibacterium in normal breast tissue directly inhibits tumor growth by secreting antimicrobial peptides and produces short-chain fatty acids to activate free fatty acid receptors 2 and 3, reducing inflammation and tumorigenesis risk (34).

Metabolic reprogramming

Intratumoral microorganisms modify the metabolic networks of glucose, lipids, and amino acids within tumors via the ‘metabolite-receptor-signal axis’, influencing the immune microenvironment and thereby affecting tumor progression. In CRC, the anaerobic bacteria Fusobacterium nucleatum, Clostridium spp. and Bacteroides spp. produce short-chain fatty acids that activate pro-inflammatory pathways such as NF-κB, elevating tumorigenesis risk (38). In gastric cancer, Peptostreptococcus spp. enhance phosphatidylcholine hydrolysis and triglyceride synthesis by upregulating phospholipase C and 1-acyl-sn-glycerol-3-phosphate acyltransferase, driving lipid metabolic reprogramming and tumor progression (78). Additionally, the study by Flores-García et al (79) demonstrated that long non-coding RNAs boost glycolysis by upregulating 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4, phosphoglycerate kinase 1 and lactate dehydrogenase A, while promoting glutamine catabolism through increased glutamate dehydrogenase 1 and Golgi transport 1A expression, supplying extra energy to tumor cells. These enzymes further activate the hypoxia inducible factor 1α/PI3K/Akt/mTOR pathway, enhancing tumor cell proliferation, survival and invasion. Table I summarizes the compositional differences and action mechanisms of the tumor microbiome (80,81).

Table I.

Intratumoral microorganisms and their mechanisms of action in multiple cancers.

Tumor types Microorganisms Functions Mechanisms (Refs.)
Breast cancer Methylobacterium radiotolerans Pro-tumor Suppression of T-cell infiltration and promotion of inflammatory responses (7)
Staphylococcus Pro-tumor Inhibition of the RhoA-ROCK signaling pathway (31)
Fusobacterium Pro-tumor Modulation of estrogen metabolism (26)
Anaerococcus Antitumor Suppression of M2 macrophage polarization, reducing immunosuppressive tumor-associated macrophages and enhancing CD8+ T-cell infiltration (33,34)
Caulobacter Antitumor Secretion of antimicrobial peptides that inhibit pro-carcinogenic bacteria (34)
Streptococcus Antitumor Induction of early apoptosis in MCF-7 breast cancer cells, with activation of the PI3K-AKT-mTOR pathway and inhibition of the JAK2-STAT3 pathway
Colorectal cancer Fusobacterium nucleatum Pro-tumor Activation of the E-cadherin/β-catenin axis, up-regulation of CHK2 and induction of DNA damage (71)
Escherichia coli Pro-tumor Base substitutions or deletions provoking DNA damage (69)
Enterotoxigenic Bacteroides fragilis Pro-tumor DNA damage elicited via base substitution (72)
Lung cancer Firmicutes Pro-tumor Upregulation of ERK and PI3K signaling pathways to induce inflammatory responses (28)
Prevotella Pro-tumor
Peptoniphilus Pro-tumor Attenuation of immune surveillance (42)
Aggregatibacter Antitumor Augmented CD8+ T-cell infiltration
Pancreatic cancer Propionibacterium acnes Pro-tumor Activation of NF-κB- and TLR4-mediated inflammatory pathways, thereby amplifying inflammation (28,45)
Gammaproteobacteria Pro-tumor
Clostridium difficile Pro-tumor Activation of MET-PTK2 and PD-1 signaling pathways (77)
Porphyromonas gingivalis Pro-tumor MAPK pathway activation inducing inflammatory responses (80)
Bacillus clausii Pro-tumor Promotion of CD8+ T-cell infiltration alongside down-regulation of MDSCs and Tregs, abrogating immunosuppression (48)
Pseudoxanthomonas Pro-tumor Activation of the NF-κB/TLR4 inflammatory axis coupled with PI3K/Akt upregulation (34,75)
Gastric cancer Helicobacter pylori Pro-tumor TLR2/4-NF-κB pathway activation and concurrent YAP/β-catenin stimulation (49,50)
Fusobacterium nucleatum Pro-tumor Activation of β-catenin/Wnt signaling pathways and impaired CD3+ T-cell infiltration (51)
Lactobacillus Pro-tumor Induction of DNA alkylation, gene mutation and hypermethylation of tumor-suppressor genes (30,52)
Veillonella Pro-tumor LPS-triggers TLR2/4-NF-κB activation, leading to barrier disruption (51)
Prostate cancer Actinobacteria Antitumor Induction of apoptosis, necrosis, autophagy and G2/M cell-cycle arrest (62)
Streptococcus Antitumor activation of TLR2/4-NF-κB pathway and enhancement of CD8+ T cell cytotoxicity (62)
Propionibacterium Pro-tumor TLR4-PI3K-Akt axis activation to induce COX-2 expression and sustain chronic inflammation (61)
Escherichia Pro-tumor TLR4-NF-κB pathway activation eliciting pro-inflammatory cytokines and ROS burst, resulting in DNA damage (61)
Acinetobacter Antitumor Competitive inhibition of pro-inflammatory pathobiont colonization to preserve local micro-ecological homeostasis (28,29)
Melanoma Acinetobacter Pro-tumor IL-17 axis activation to potentiate STAT3 signaling and induce PD-L1 upregulation with Tregs (23)
Corynebacterium Pro-tumor
Lachnospira Pro-tumor (30)
Staphylococcus epidermidis Antitumor Suppression of Th17 responses and blockade of the TLR4-NF-κB-ROS inflammatory cascade, diminishing DNA-damage-associated mutagenesis (68)
Liver cancer Bacteroides ovatus Pro-tumor Metabolite iso-lithocholic acid-mediated impairment of NK-cell-dependent antitumor immunity, accelerating hepatocellular carcinoma progression (81)
Pseudomonadaceae Antitumor Blockade of EGFR phosphorylation and downstream Akt/IκBβ/NF-κB signaling, driving cancer cell apoptosis (25)

Pro-tumor, microbes or mechanisms that promote oncogenesis or accelerate progression; antitumor, microbes or mechanisms that suppress malignant phenotypes or enhance anticancer immunity; ROCK, Rho associated coiled-coil containing protein kinase 1; CHK2, checkpoint kinase 2; TLR, toll-like receptor; MET, MET proto-oncogene, receptor tyrosine kinase; PD-1, programmed cell death protein 1; PTK2, protein tyrosine kinase 2; MDSC, myeloid-derived suppressor cell; Treg, regulator T cell; LPS, lipopolysaccharide; COX-2, cyclooxygenase-2; ROS, reactive oxygen species; PD-L1, programmed death-ligand 1; NK, natural killer; IκBβ, inhibitor of nuclear factor-κBβ.

4. Diagnosis of tumor microbial biomarkers

New dimensions of liquid biopsy

Liquid biopsy, a non-invasive technique, utilizes peripheral blood samples to monitor tumor characteristics. Its core detection targets include circulating tumor cells, circulating tumor DNA (ctDNA), cell-free RNA and extracellular vesicles, which have demonstrated applicability in the early diagnosis and dynamic monitoring of various cancers (82). Specifically, ctDNA enables real-time assessment of tumor burden by identifying tumor-specific somatic mutations, copy number variations and epigenetic modifications, such as methylation (83). Furthermore, ctDNA analysis can predict postoperative recurrence in patients with CRC (84).

The bacteremia microbial profile in patients with CRC was closely associated with tumor stage, as reported by Kwong et al (85). Specifically, the positive inflection point of Clostridium perfringens appeared 100 days after the diagnosis of bacteremia, while the peaks of Peptostreptococcus spp. and Fusobacterium spp. occurred at 121 days and 127 days, respectively. Notably, Fusobacterium spp. and Peptostreptococcus spp. were associated with the diagnosis of early-stage CRC, whereas Streptococcus spp. was associated with late-stage CRC. Furthermore, the abundances of bacterial and fungal DNA in peripheral blood were highly consistent with the characteristics of the intratumoral microbiome, suggesting their potential as circulating microbial biomarkers (20). The integration of high-throughput sequencing and machine learning algorithms has enabled the multi-cancer early detection technology based on cell-free circulating DNA to identify molecular signals shared by multiple tumors in a single blood sample (86). Wang et al (87) demonstrated that Lactobacillus and Streptococcus could serve as microbial markers for gastric cancer, aiding in its early non-invasive diagnosis. Collectively, these studies indicate that circulating tumor microbiome markers have the potential to become non-invasive and highly sensitive diagnostic tools, which could expand the application of liquid biopsy in early tumor screening (88).

In situ tissue detection

In situ tissue detection technology, including immunohistochemistry, immunofluorescence and fluorescence in situ hybridization (FISH), serves a crucial role in directly identifying microbial biomarkers within tumor tissues. These techniques are valuable for elucidating the spatial distribution of microorganisms in relation to tumor cells and their involvement in tumorigenesis and progression (89). Notably, FISH stands out as a well-established molecular cytogenetic approach that employs highly complementary fluorescent DNA probes to sensitively and specifically detect and localize bacterial 16S rRNA genes (90).

Studies have shown that analyzing the tumor-associated microbiome using various techniques such as 16S rRNA gene amplicon sequencing and metagenomic sequencing has the potential to target and eliminate tumor-promoting microorganisms or enrich microorganisms that enhance antitumor immunity, thereby regulating the composition of the microbiome and providing new strategies for the diagnosis, prevention and treatment of tumors (91). Chai et al (92) identified specific bacteria, such as Klebsiella pneumoniae and the fungus Paraburkholderia, in intrahepatic cholangiocarcinoma tissues using FISH. In pancreatic cancer, Fusobacterium was independently associated with poor prognosis, suggesting it has a role as a prognostic biomarker (93). Zhang et al (94) showed that the tumor microbiome influences treatment outcomes and survival rates. By examining microbial changes in patients with stage III–IV non-small cell lung cancer, the authors identified Haemophilus parainfluenzae, Serratia marcescens, Acinetobacter junii and Streptococcus constellatus as markers for predicting 2-year survival rates. Yamamura et al (95) reported that high Fusobacterium nucleatum levels in esophageal squamous cell carcinoma indicated poor prognosis, serving as a potential biomarker. Intratumoral microbiota thus holds promise for tumor diagnosis and prognosis, although large-scale, multi-center studies are necessary to confirm their clinical utility.

5. Treatment strategies based on the tumor microbiota

The effectiveness of traditional tumor treatments such as immunotherapy, chemotherapy and radiotherapy is notably affected by microorganisms. Consequently, innovative microorganism-based therapies, including probiotics, prebiotics, synbiotics, fecal microbiota transplantation (FMT), engineered microbiota, phage therapy and oncolytic virus therapy, are gaining traction in tumor prevention and treatment (72). Contemporary strategies frequently integrate these novel microbial therapies with conventional approaches to enhance therapeutic outcomes.

Microbial targeted therapy

Microorganism-based adjuvant therapies have demonstrated notable potential to enhance the efficacy of tumor immunotherapy. A study has shown that in patients with CRC receiving anti-PD-1 monoclonal antibody (mAb) immune checkpoint blockade, the abundance of Fusobacterium nucleatum is markedly increased in non-responders, and its metabolite succinate can induce CRC cells to develop resistance to immunotherapy (96). A clinical trial on locally advanced rectal cancer found that the anti-PD-1 mAb dostarlimab exhibited promising therapeutic effects, with some patients avoiding chemotherapy, radiotherapy or surgery (97). Furthermore, a phase I trial reported complete remission in a metastatic melanoma patient resistant to anti-PD-1 therapy after FMT (98).

Emerging strategies, such as oncolytic viruses (OVs) and engineered bacteria, have shown promise in overcoming the limitations of CAR-T cell therapy for solid tumors, owing to their tumor selectivity and programmable immunogenicity (99). For instance, the OV CD19t can induce CD19 expression on tumor cells, thereby enhancing the antitumor response of CD19-CAR-T cells in mouse models (100). Additionally, the combined use of attenuated Brucella strains and adoptive transfer of antigen-specific CD8+ CAR-T cells has demonstrated near-complete elimination of tumor growth and proliferation, achieving a 100% host survival rate and effectively overcoming CAR-T cell resistance (101). However, intratumoral microorganisms may also weaken the efficacy of traditional chemotherapy. 5-fluorouracil (5-FU) is a first-line chemotherapy drug for CRC, but Fusobacterium nucleatum and Escherichia coli can metabolically deplete 5-FU and activate the autophagy pathway, reducing the local drug concentration and efficacy (102). Therefore, precise regulation of the intratumoral microbiota is of great importance for optimizing comprehensive tumor treatment.

Enhancement of immunotherapy efficacy

The commensal microbiota and essential nutrients have demonstrated notable potential in cancer treatment. Multiple studies have revealed the promoting effects of commensal bacteria and their metabolites on the efficacy of immune checkpoint blockade (ICB). For example, Jia et al (103) found that the abundance of commensal Lactobacillus spp. was positively associated with ICB responsiveness, and its metabolite, indolepropionic acid, could improve the immunotherapy outcomes of melanoma, breast cancer and CRC by regulating the stemness program of intratumoral CD8+ T cells. Bifidobacterium spp. has been shown to enhance the immunotherapy response by activating the STING signaling pathway (104). Oral administration of live Lactobacillus rhamnosus GG increased the numbers of tumor-infiltrating dendritic cells and T cells, thereby enhancing the antitumor activity of anti-PD-1 therapy (105). Kalaora et al (29) identified human leukocyte antigen-presenting peptide fragments derived from intratumoral bacteria in melanoma, which could be co-presented by antigen-presenting cells and tumor cells. This can increase the diversity of immunogenic antigens and promote T cell activation, thus enhancing the benefits of immune checkpoint inhibitors.

Probiotics can mitigate the side effects of conventional cancer treatments. Linn et al (106) demonstrated that a probiotic mix of Lactobacillus acidophilus LA-5 and Bifidobacterium animalis subsp. lactis BB-12 reduces acute diarrhea in patients with cervical cancer undergoing radiotherapy. Certain commensal microbiota also have direct therapeutic potential. Montalban-Arques et al (107) showed that oral administration of four Clostridium spp. strains prevented and effectively treated CRC in a mouse model, outperforming anti-PD-1 monotherapy. Engineered microbial therapy is an emerging cancer treatment strategy. Canale et al (108) developed an engineered Escherichia coli Nissle1917 that converts ammonia-nitrogen to L-arginine within tumors, enhancing CD8+ T cell infiltration and achieving antitumor effects when combined with programmed death-ligand 1 antibodies. Commensal bacteria, their metabolites and engineered strains can reshape the TME through various pathways, offering new strategies and targets for combined immunotherapy.

6. Limitations

The present review discusses foundational knowledge on intratumoral microbiota, while acknowledging several limitations. Firstly, cancer type coverage remains incomplete, and while eight major solid tumors were analyzed, emerging models such as hepatocellular carcinoma (HCC) and renal cell carcinoma were underrepresented, reflecting current research disparities. HCC exclusion, for instance, stems from unresolved challenges in discriminating true tumoral microbiota from hepatic translocation of gut bacteria (24). Furthermore, methodological heterogeneity across studies, including inconsistent sample processing, DNA extraction protocols and bioinformatic approaches (such as 16S rRNA gene sequencing vs. shotgun metagenomics) compromises the comparability of reported microbial profiles (4). Critically, the majority of cited evidence establishes associative rather than causal relationships, leaving underlying mechanisms largely unvalidated; observed microbial enrichments, such as Veillonella in gastric tumors, could potentially be a consequence rather than a driver of tumor-induced microenvironmental changes (30). Finally, notable translational barriers exist: Promising microbial biomarkers (such as Fusobacterium nucleatum in CRC) lack validation of clinical utility in large prospective trials, and microbiota-targeted therapeutic strategies, including engineered bacteria, currently lack phase III efficacy data.

7. Challenges and prospects

Bottlenecks in key technologies

The differences in the local microenvironments of microbial colonization and the heterogeneity of host cells form a complex interaction network, because both microbial communities and host cells exhibit notable spatiotemporal dynamic changes (109,110). However, current technological constraints hinder the concurrent acquisition of microbial spatial distribution and host single-cell transcriptome information (111,112).

The causal relationship between microbial colonization and tumor progression is difficult to clarify, mainly due to unclear temporal relationships and interference from confounding factors (111). Most existing studies are based on cross-sectional analyses, making it difficult to distinguish whether microbial colonization is a driving factor, a concomitant phenomenon or a secondary result of tumors (8). The dynamic changes in the TME (such as hypoxia or immune suppression) may reversely shape the microbial community, forming a complex feedback loop of bidirectional interaction (70). In addition, individual differences (including genetic background, diet and antibiotic use) and the spatiotemporal heterogeneity of sample collection (such as differences in intratumoral/peritumoral microbiota) need further investigation (70,113).

Research on microbe-host interactions faces a notable lack of standardization, mainly manifested in the absence of unified specifications for sampling methods and bioinformatics analysis, resulting in poor data comparability (69). The complexity of clinical sample collection poses challenges in guaranteeing the quality and quantity of tumor samples (114). Ensuring sample purity and devising strategies to effectively mitigate environmental microbial contamination are crucial objectives during the detection process (4).

Future research directions

Firstly, future research should prioritize the integration of tumor-microbiome-immune multi-omics through systematic analysis of interaction mechanisms using dynamic network modeling (4). To achieve this, advanced technologies such as single-cell transcriptomics, spatial metabolomics and high-throughput sequencing should be combined for synchronous data collection across different scales (115). Machine learning algorithms should be employed to integrate metagenomic, metabolomic and immune cell interaction data to identify key hub molecules (116). Integrating organoid models with dynamic perturbation experiments could validate the predicted networks. This approach aims to develop a quantifiable and controllable ‘microbiome-metabolism-immunity’ computational model, serving as a predictive platform for targeted interventions and advancing research from association to causal mechanisms (117).

Secondly, future research should focus on the development of precision intervention strategies based on the characteristics of the tumor microbiome. Through the integration of multi-omics data such as intratumoral microbiota sequencing, metabolomics and immune microenvironment analysis, a system of predictive biomarkers should be established to guide individualized treatment (118). Utilizing machine learning to analyze tumor-specific microorganisms and validating the interaction between microorganisms and drugs through organoid co-cultures and high-throughput drug screening are essential (119). Moreover, the advancement of microorganism-targeted delivery technologies such as nanocarriers and the design of microbiome combination therapies are crucial (97,120). Ultimately, the goal is to establish a closed-loop transition from microbial diagnostic profiling to treatment decision-making, thereby shifting the paradigm of tumor treatment from a generalized approach to precise microbiota regulation.

Finally, future research should also focus on establishing a highly biomimetic organoid-microbe co-culture system to construct a standardized and high-throughput research and translation platform for the interaction between microorganisms and the host (121,122). Long-term co-culture systems that pair specific organ-derived organoids with complex microbial communities should be developed, and microfluidic-chip technologies should be integrated to simulate key in vivo microenvironmental parameters such as oxygen gradients (123,124). Building on these systems, real-time multimodal monitoring should be employed to analyze the regulatory mechanisms by which microbial colonization influences barrier function, immune responses and drug sensitivity in organoids (125). Additionally, an automated culture-and-detection platform should be established and coupled with artificial-intelligence algorithms to predict the therapeutic effects of microbiota-targeted interventions, thereby accelerating the translation of mechanistic discoveries into clinical treatment regimens (126). This platform will bridge the technological gap between in vitro models and clinical trials and provide accurate prediction tools for microbial targeted therapy.

Acknowledgements

Not applicable.

Funding Statement

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

JY drafted the manuscript and summarized the tables. QL was involved in the conception of the study. HL supervised methodology and theoretical framework and critically reviewed and edited the manuscript for intellectual content and language accuracy. YD, YC, SR and ZL reviewed the manuscript. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

  • 1.El-Sayed A, Aleya L, Kamel M. Microbiota's role in health and diseases. Environ Sci Pollut Res Int. 2021;28:36967–36983. doi: 10.1007/s11356-021-14593-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Cho I, Blaser MJ. The human microbiome: At the interface of health and disease. Nat Rev Genet. 2012;13:260–270. doi: 10.1038/nrg3182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Wong-Rolle A, Wei HK, Zhao C, Jin C. Unexpected guests in the tumor microenvironment: Microbiome in cancer. Protein Cell. 2021;12:426–435. doi: 10.1007/s13238-020-00813-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sepich-Poore GD, Zitvogel L, Straussman R, Hasty J, Wargo JA, Knight R. The microbiome and human cancer. Science. 2021;371:eabc4552. doi: 10.1126/science.abc4552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Cogdill AP, Gaudreau PO, Arora R, Gopalakrishnan V, Wargo JA. The impact of intratumoral and gastrointestinal microbiota on systemic cancer therapy. Trends Immunol. 2018;39:900–920. doi: 10.1016/j.it.2018.09.007. [DOI] [PubMed] [Google Scholar]
  • 6.Gong Y, Huang X, Wang M, Liang X. Intratumor microbiota: A novel tumor component. J Cancer Res Clin Oncol. 2023;149:6675–6691. doi: 10.1007/s00432-023-04576-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, Geller LT, Rotter-Maskowitz A, Weiser R, Mallel G, Gigi E, et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science. 2020;368:973–980. doi: 10.1126/science.aay9189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cao Y, Xia H, Tan X, Shi C, Ma Y, Meng D, Zhou M, Lv Z, Wang S, Jin Y. Intratumoural microbiota: A new frontier in cancer development and therapy. Signal Transduct Target Ther. 2024;9:15. doi: 10.1038/s41392-023-01693-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hanus M, Parada-Venegas D, Landskron G, Wielandt AM, Hurtado C, Alvarez K, Hermoso MA, López-Köstner F, De la Fuente M. Immune system, microbiota, and microbial metabolites: The unresolved triad in colorectal cancer microenvironment. Front Immunol. 2021;12:612826. doi: 10.3389/fimmu.2021.612826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Walker SP, Tangney M, Claesson MJ. Sequence-based characterization of intratumoral Bacteria-A guide to best practice. Front Oncol. 2020;10:179. doi: 10.3389/fonc.2020.00179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Galeano Niño JL, Wu H, LaCourse KD, Kempchinsky AG, Baryiames A, Barber B, Futran N, Houlton J, Sather C, Sicinska E, et al. Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer. Nature. 2022;611:810–817. doi: 10.1038/s41586-022-05435-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gao B, Li X, Li S, Wang S, Wu J, Li J. Pan-cancer analysis identifies RNA helicase DDX1 as a prognostic marker. Phenomics. 2022;2:33–49. doi: 10.1007/s43657-021-00034-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wang H, Chen L. Tumor microenviroment and hepatocellular carcinoma metastasis. J Gastroenterol Hepatol. 2013;28((Suppl 1)):S43–S48. doi: 10.1111/jgh.12091. [DOI] [PubMed] [Google Scholar]
  • 14.Zhou S, Lu J, Liu S, Shao J, Liu Z, Li J, Xiao W. Role of the tumor microenvironment in malignant melanoma organoids during the development and metastasis of tumors. Front Cell Deve Biol. 2023;11:1166916. doi: 10.3389/fcell.2023.1166916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gao F, Yu B, Rao B, Sun Y, Yu J, Wang D, Cui G, Ren Z. The effect of the intratumoral microbiome on tumor occurrence, progression, prognosis and treatment. Front Immunol. 2022;13:1051987. doi: 10.3389/fimmu.2022.1051987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Yang L, Li A, Wang Y, Zhang Y. Intratumoral microbiota: Roles in cancer initiation, development and therapeutic efficacy. Signal Transduct Target Ther. 2023;8:35. doi: 10.1038/s41392-022-01304-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Garrett WS. Cancer and the microbiota. Science. 2015;348:80–86. doi: 10.1126/science.aaa4972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Rous P. A sarcoma of the fowl transmissible by an agent separable from the tumor cells. J Exp Med. 1911;13:397–411. doi: 10.1084/jem.13.4.397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Warren JR, Marshall B. Unidentified curved bacilli on gastric epithelium in active chronic gastritis. Lancet. 1983;1:1273–1275. [PubMed] [Google Scholar]
  • 20.Narunsky-Haziza L, Sepich-Poore GD, Livyatan I, Asraf O, Martino C, Nejman D, Gavert N, Stajich JE, Amit G, González A, et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell. 2022;185:3789–3806.e17. doi: 10.1016/j.cell.2022.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Felgner S, Kocijancic D, Frahm M, Weiss S. Bacteria in cancer therapy: Renaissance of an old concept. Int J Microbiol. 2016;2016:8451728. doi: 10.1155/2016/8451728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Budynek P, Dabrowska K, Skaradziński G, Górski A. Bacteriophages and cancer. Arch Microbiol. 2010;192:315–320. doi: 10.1007/s00203-010-0559-7. [DOI] [PubMed] [Google Scholar]
  • 23.Xiong X, Zheng LW, Ding Y, Chen YF, Cai YW, Wang LP, Huang L, Liu CC, Shao ZM, Yu KD, et al. Breast cancer: Pathogenesis and treatments. Signal Transduct Target Ther. 2025;10:49. doi: 10.1038/s41392-024-02108-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zhou B, Sun C, Huang J, Xia M, Guo E, Li N, Lu H, Shan W, Wu Y, Li Y, et al. The biodiversity composition of microbiome in ovarian carcinoma patients. Sci Rep. 2019;9:1691. doi: 10.1038/s41598-018-38031-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Qu D, Wang Y, Xia Q, Chang J, Jiang X, Zhang H. Intratumoral microbiome of human primary liver cancer. Hepatol Commun. 2022;6:1741–1752. doi: 10.1002/hep4.1908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Tzeng A, Sangwan N, Jia M, Liu CC, Keslar KS, Downs-Kelly E, Fairchild RL, Al-Hilli Z, Grobmyer SR, Eng C. Human breast microbiome correlates with prognostic features and immunological signatures in breast cancer. Genome Med. 2021;13:60. doi: 10.1186/s13073-021-00874-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Sommariva M, Le Noci V, Bianchi F, Camelliti S, Balsari A, Tagliabue E, Sfondrini L. The lung microbiota: Role in maintaining pulmonary immune homeostasis and its implications in cancer development and therapy. Cell Mol Life Sci. 2020;77:2739–2749. doi: 10.1007/s00018-020-03452-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Najafi S, Abedini F, Azimzadeh Jamalkandi S, Shariati P, Ahmadi A, Gholami Fesharaki M. The composition of lung microbiome in lung cancer: A systematic review and meta-analysis. BMC Microbiol. 2021;21:315. doi: 10.1186/s12866-021-02375-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kalaora S, Nagler A, Nejman D, Alon M, Barbolin C, Barnea E, Ketelaars SLC, Cheng K, Vervier K, Shental N, et al. Identification of bacteria-derived HLA-bound peptides in melanoma. Nature. 2021;592:138–143. doi: 10.1038/s41586-021-03368-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Park JY, Seo H, Kang CS, Shin TS, Kim JW, Park JM, Kim JG, Kim YK. Dysbiotic change in gastric microbiome and its functional implication in gastric carcinogenesis. Scie Rep. 2022;12:4285. doi: 10.1038/s41598-022-08288-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Smith A, Pierre JF, Makowski L, Tolley E, Lyn-Cook B, Lu L, Vidal G, Starlard-Davenport A. Distinct microbial communities that differ by race, stage, or breast-tumor subtype in breast tissues of non-Hispanic Black and non-Hispanic White women. Sci Rep. 2019;9:11940. doi: 10.1038/s41598-019-48348-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wang Q, Liu Z, Ma A, Li Z, Liu B, Ma Q. Computational methods and challenges in analyzing intratumoral microbiome data. Trends Microbiol. 2023;31:707–722. doi: 10.1016/j.tim.2023.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Guo H. Interactions between the tumor microbiota and breast cancer. Front Cell Infect Microbiol. 2024;14:1499203. doi: 10.3389/fcimb.2024.1499203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Thu MS, Chotirosniramit K, Nopsopon T, Hirankarn N, Pongpirul K. Human gut, breast, and oral microbiome in breast cancer: A systematic review and meta-analysis. Front Oncol. 2023;13:1144021. doi: 10.3389/fonc.2023.1144021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zalila-Kolsi I, Dhieb D, Osman HA, Mekideche H. The gut microbiota and colorectal cancer: Understanding the link and exploring therapeutic interventions. Biology (Basel) 2025;14:251. doi: 10.3390/biology14030251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, Barnes R, Watson P, Allen-Vercoe E, Moore RA, Holt RA. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012;22:299–306. doi: 10.1101/gr.126516.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Matsuda T, Fujimoto A, Igarashi Y. Colorectal cancer: Epidemiology, risk factors, and public health strategies. Digestion. 2025;106:91–99. doi: 10.1159/000543921. [DOI] [PubMed] [Google Scholar]
  • 38.Li J, Ma X, Chakravarti D, Shalapour S, DePinho RA. Genetic and biological hallmarks of colorectal cancer. Genes Dev. 2021;35:787–820. doi: 10.1101/gad.348226.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Repass J. Replication Study: Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. ELife. 2018;7:e25801. doi: 10.7554/eLife.25801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ramírez-Labrada AG, Isla D, Artal A, Arias M, Rezusta A, Pardo J, Gálvez EM. The Influence of lung microbiota on lung carcinogenesis, immunity, and immunotherapy. Trends Cancer. 2020;6:86–97. doi: 10.1016/j.trecan.2019.12.007. [DOI] [PubMed] [Google Scholar]
  • 41.Liu NN, Yi CX, Wei LQ, Zhou JA, Jiang T, Hu CC, Wang L, Wang YY, Zou Y, Zhao YK, et al. The intratumor mycobiome promotes lung cancer progression via myeloid-derived suppressor cells. Cancer Cell. 2023;41:1927–1944.e9. doi: 10.1016/j.ccell.2023.08.012. [DOI] [PubMed] [Google Scholar]
  • 42.Vogtmann E, Hua X, Yu G, Purandare V, Hullings AG, Shao D, Wan Y, Li S, Dagnall CL, Jones K, et al. The oral microbiome and lung cancer risk: An analysis of 3 prospective cohort studies. J Natl Cancer Inst. 2022;114:1501–1510. doi: 10.1093/jnci/djac149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Klein AP. Pancreatic cancer epidemiology: Understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol. 2021;18:493–502. doi: 10.1038/s41575-021-00457-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Stoffel EM, Brand RE, Goggins M. Pancreatic cancer: Changing epidemiology and new approaches to risk assessment, early detection, and prevention. Gastroenterology. 2023;164:752–765. doi: 10.1053/j.gastro.2023.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Cohen RJ, Shannon BA, McNeal JE, Shannon T, Garrett KL. Propionibacterium acnes associated with inflammation in radical prostatectomy specimens: A possible link to cancer evolution? J Urol. 2005;173:1969–1974. doi: 10.1097/01.ju.0000158161.15277.78. [DOI] [PubMed] [Google Scholar]
  • 46.McAllister F, Khan MAW, Helmink B, Wargo JA. The tumor microbiome in pancreatic cancer: Bacteria and beyond. Cancer Cell. 2019;36:577–579. doi: 10.1016/j.ccell.2019.11.004. [DOI] [PubMed] [Google Scholar]
  • 47.Kaune T, Griesmann H, Theuerkorn K, Hämmerle M, Laumen H, Krug S, Plumeier I, Kahl S, Junca H, Gustavo Dos Anjos Borges L, et al. Gender-specific changes of the gut microbiome correlate with tumor development in murine models of pancreatic cancer. iScience. 2023;26:106841. doi: 10.1016/j.isci.2023.106841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Riquelme E, Zhang Y, Zhang L, Montiel M, Zoltan M, Dong W, Quesada P, Sahin I, Chandra V, San Lucas A, et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell. 2019;178:795–806.e12. doi: 10.1016/j.cell.2019.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Stewart OA, Wu F, Chen Y. The role of gastric microbiota in gastric cancer. Gut Microbes. 2020;11:1220–1230. doi: 10.1080/19490976.2020.1762520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ianiro G, Molina-Infante J, Gasbarrini A. Gastric microbiota. Helicobacter. 2015;20((Suppl 1)):S68–S71. doi: 10.1111/hel.12260. [DOI] [PubMed] [Google Scholar]
  • 51.Coker OO, Dai Z, Nie Y, Zhao G, Cao L, Nakatsu G, Wu WK, Wong SH, Chen Z, Sung JJY, Yu J. Mucosal microbiome dysbiosis in gastric carcinogenesis. Gut. 2018;67:1024–1032. doi: 10.1136/gutjnl-2017-314281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Engstrand L, Graham DY. Microbiome and Gastric Cancer. Dig Dis Sci. 2020;65:865–873. doi: 10.1007/s10620-020-06101-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Stewart C, Ralyea C, Lockwood S. Ovarian cancer: An integrated review. Semin Oncol Nurs. 2019;35:151–156. doi: 10.1016/j.soncn.2019.02.001. [DOI] [PubMed] [Google Scholar]
  • 54.Sipos A, Ujlaki G, Mikó E, Maka E, Szabó J, Uray K, Krasznai Z, Bai P. The role of the microbiome in ovarian cancer: Mechanistic insights into oncobiosis and to bacterial metabolite signaling. Mol Med. 2021;27:33. doi: 10.1186/s10020-021-00295-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Dhingra A, Sharma D, Kumar A, Singh S, Kumar P. Microbiome and development of ovarian cancer. Endocr Metab Immune Disord Drug Targets. 2022;22:1073–1090. doi: 10.2174/1871530322666220509034847. [DOI] [PubMed] [Google Scholar]
  • 56.Sun J, Xiang J, An Y, Xu J, Xiong Y, Wang S, Xia Q. Unveiling the association between HPV and Pan-cancers: A bidirectional two-sample mendelian randomization study. Cancers Basel. 2023;15:5147. doi: 10.3390/cancers15215147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ingerslev K, Hogdall E, Skovrider-Ruminski W, Schnack TH, Karlsen MA, Nedergaard L, Hogdall C, Blaakær J. High-risk HPV is not associated with epithelial ovarian cancer in a Caucasian population. Infect Agent Cancer. 2016;11:39. doi: 10.1186/s13027-016-0087-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Rizzo A, Santoni M, Mollica V, Fiorentino M, Brandi G, Massari F. Microbiota and prostate cancer. Semin Cancer Biol. 2022;86:1058–1065. doi: 10.1016/j.semcancer.2021.09.007. [DOI] [PubMed] [Google Scholar]
  • 59.Massari F, Mollica V, Di Nunno V, Gatto L, Santoni M, Scarpelli M, Cimadamore A, Lopez-Beltran A, Cheng L, Battelli N, et al. The human microbiota and prostate cancer: Friend or Foe? Cancers (Basel) 2019;11:459. doi: 10.3390/cancers11040459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Cavarretta I, Ferrarese R, Cazzaniga W, Saita D, Lucianò R, Ceresola ER, Locatelli I, Visconti L, Lavorgna G, Briganti A, et al. The microbiome of the prostate tumor microenvironment. Eur Urol. 2017;72:625–631. doi: 10.1016/j.eururo.2017.03.029. [DOI] [PubMed] [Google Scholar]
  • 61.Feng Y, Ramnarine VR, Bell R, Volik S, Davicioni E, Hayes VM, Ren S, Collins CC. Metagenomic and metatranscriptomic analysis of human prostate microbiota from patients with prostate cancer. BMC Genomics. 2019;20:146. doi: 10.1186/s12864-019-5457-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Shrestha E, White JR, Yu SH, Kulac I, Ertunc O, De Marzo AM, Yegnasubramanian S, Mangold LA, Partin AW, Sfanos KS. Profiling the urinary microbiome in men with positive versus negative biopsies for prostate cancer. J Urol. 2018;199:161–171. doi: 10.1016/j.juro.2017.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Banerjee S, Alwine JC, Wei Z, Tian T, Shih N, Sperling C, Guzzo T, Feldman MD, Robertson ES. Microbiome signatures in prostate cancer. Carcinogenesis. 2019;40:749–764. doi: 10.1093/carcin/bgz008. [DOI] [PubMed] [Google Scholar]
  • 64.Ahmed B, Qadir MI, Ghafoor S. Malignant melanoma: Skin cancer-diagnosis, prevention, and treatment. Crit Rev Eukaryot Gene Expr. 2020;30:291–297. doi: 10.1615/CritRevEukaryotGeneExpr.2020028454. [DOI] [PubMed] [Google Scholar]
  • 65.Byrd AL, Belkaid Y, Segre JA. The human skin microbiome. Nat Rev Microbiol. 2018;16:143–155. doi: 10.1038/nrmicro.2017.157. [DOI] [PubMed] [Google Scholar]
  • 66.Grice EA, Kong HH, Conlan S, Deming CB, Davis J, Young AC, NISC Comparative Sequencing Program. Bouffard GG, Blakesley RW, Murray PR, et al. Topographical and temporal diversity of the human skin microbiome. Science. 2009;324:1190–1192. doi: 10.1126/science.1171700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Zhu G, Su H, Johnson CH, Khan SA, Kluger H, Lu L. Intratumour microbiome associated with the infiltration of cytotoxic CD8+ T cells and patient survival in cutaneous melanoma. Eur J Cancer. 2021;151:25–34. doi: 10.1016/j.ejca.2021.03.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kozmin SG, Rogozin IB, Moore EA, Abney M, Schaaper RM, Pavlov YI. Comment on A commensal strain of Staphylococcus epidermidis protects against skin neoplasia by Nakatsuji et al. Sci Adv. 2019;5:eaaw3915. doi: 10.1126/sciadv.aaw3915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Long J, Wang J, Xiao C, You F, Jiang Y, Li X. Intratumoral microbiota in colorectal cancer: Focus on specific distribution and potential mechanisms. Cell Commun Signal. 2024;22:455. doi: 10.1186/s12964-024-01831-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Ding T, Liu C, Li Z. The mycobiome in human cancer: Analytical challenges, molecular mechanisms, and therapeutic implications. Mol Cancer. 2025;24:18. doi: 10.1186/s12943-025-02227-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Guo P, Tian Z, Kong X, Yang L, Shan X, Dong B, Ding X, Jing X, Jiang C, Jiang N, Yu Y. FadA promotes DNA damage and progression of Fusobacterium nucleatum-induced colorectal cancer through up-regulation of chk2. J Exp Clin Cancer Res. 2020;39:202. doi: 10.1186/s13046-020-01677-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Dejea CM, Fathi P, Craig JM, Boleij A, Taddese R, Geis AL, Wu X, DeStefano Shields CE, Hechenbleikner EM, Huso DL, et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science. 2018;359:592–597. doi: 10.1126/science.aah3648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zhang H, Fu L, Leiliang X, Qu C, Wu W, Wen R, Huang N, He Q, Cheng Q, Liu G, Cheng Y. Beyond the Gut: The intratumoral microbiome's influence on tumorigenesis and treatment response. Cancer Commun (Lond) 2024;44:1130–1167. doi: 10.1002/cac2.12597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Fu Y, Li J, Cai W, Huang Y, Liu X, Ma Z, Tang Z, Bian X, Zheng J, Jiang J, Li C. The emerging tumor microbe microenvironment: From delineation to multidisciplinary approach-based interventions. Acta Pharm Sin B. 2024;14:1560–1591. doi: 10.1016/j.apsb.2023.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chen X, Sun F, Wang X, Feng X, Aref AR, Tian Y, Ashrafizadeh M, Wu D. Inflammation, microbiota, and pancreatic cancer. Cancer Cell Int. 2025;25:62. doi: 10.1186/s12935-025-03673-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Yan X, Qu X, Wang J, Lu L, Wu W, Mao J, Li D, Wang Y, Wei Q, Liu J. Fusobacterium nucleatum promotes the growth and metastasis of colorectal cancer by activating E-Cadherin/Krüppel-Like Factor 4/Integrin α5 Signaling in a Calcium-dependent manner. MedComm. 2025;6:e70137. doi: 10.1002/mco2.70137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ghaddar B, Biswas A, Harris C, Omary MB, Carpizo DR, Blaser MJ, De S. Tumor microbiome links cellular programs and immunity in pancreatic cancer. Cancer Cell. 2022;40:1240–1253.e5. doi: 10.1016/j.ccell.2022.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Jiang XW, Zhang L, Liu ZC, Zhou T, Li WQ, Liu WD, Zhang LF, You WC, Zhang Y, Pan KF. Integrative metabolomics and microbiomics analysis reveals distinctive microbiota-metabolites interactions in gastric carcinogenesis. Int J Cancer. 2025;156:2389–2400. doi: 10.1002/ijc.35392. [DOI] [PubMed] [Google Scholar]
  • 79.Flores-García LC, García-Castillo V, Pérez-Toledo E, Trujano-Camacho S, Millán-Catalán O, Pérez-Yepez EA, Coronel-Hernández J, Rodríguez-Dorantes M, Jacobo-Herrera N, Pérez-Plasencia C. HOTAIR participation in glycolysis and glutaminolysis through lactate and glutamate production in colorectal cancer. Cells. 2025;14:388. doi: 10.3390/cells14050388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hayashi M, Ikenaga N, Nakata K, Luo H, Zhong P, Date S, Oyama K, Higashijima N, Kubo A, Iwamoto C, et al. Intratumor Fusobacterium nucleatum promotes the progression of pancreatic cancer via the CXCL1-CXCR2 axis. Cancer Sci. 2023;114:3666–3678. doi: 10.1111/cas.15901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wei H, Suo C, Gu X, Shen S, Lin K, Zhu C, Yan K, Bian Z, Chen L, Zhang T, et al. AKR1D1 suppresses liver cancer progression by promoting bile acid metabolism-mediated NK cell cytotoxicity. Cell Metab. 2025;37:1103–1118.e7. doi: 10.1016/j.cmet.2025.01.011. [DOI] [PubMed] [Google Scholar]
  • 82.Martignano F, Munagala U, Crucitta S, Mingrino A, Semeraro R, Del Re M, Petrini I, Magi A, Conticello SG. Nanopore sequencing from liquid biopsy: Analysis of copy number variations from cell-free DNA of lung cancer patients. Mol Cancer. 2021;20:32. doi: 10.1186/s12943-021-01327-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Dao J, Conway PJ, Subramani B, Meyyappan D, Russell S, Mahadevan D. Using cfDNA and ctDNA as oncologic markers: A path to clinical validation. Int J Mol Sci. 2023;24:13219. doi: 10.3390/ijms241713219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Malla M, Loree JM, Kasi PM, Parikh AR. Using circulating tumor DNA in colorectal cancer: Current and evolving practices. J Clin Oncol. 2022;40:2846–2857. doi: 10.1200/JCO.21.02615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kwong TNY, Wang X, Nakatsu G, Chow TC, Tipoe T, Dai RZW, Tsoi KKK, Wong MCS, Tse G, Chan MTV, et al. Association between bacteremia from specific microbes and subsequent diagnosis of colorectal cancer. Gastroenterology. 2018;155:383–390.e8. doi: 10.1053/j.gastro.2018.04.028. [DOI] [PubMed] [Google Scholar]
  • 86.Vittone J, Gill D, Goldsmith A, Klein EA, Karlitz JJ. A multi-cancer early detection blood test using machine learning detects early-stage cancers lacking USPSTF-recommended screening. NPJ Precis Oncol. 2024;8:91. doi: 10.1038/s41698-024-00568-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wang Y, Wang Y, Han W, Han M, Liu X, Dai J, Dong Y, Sun T, Xu J. Intratumoral and fecal microbiota reveals microbial markers associated with gastric carcinogenesis. Front Cell Infect Microbiol. 2024;14:1397466. doi: 10.3389/fcimb.2024.1397466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Poore GD, Kopylova E, Zhu Q, Carpenter C, Fraraccio S, Wandro S, Kosciolek T, Janssen S, Metcalf J, Song SJ, et al. Microbiome analyses of blood and tissues suggest cancer diagnostic approach. Nature. 2020;579:567–574. doi: 10.1038/s41586-020-2095-1. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 89.Wang N, Wu S, Huang L, Hu Y, He X, He J, Hu B, Xu Y, Rong Y, Yuan C, et al. Intratumoral microbiome: Implications for immune modulation and innovative therapeutic strategies in cancer. J Biomed Sci. 2025;32:23. doi: 10.1186/s12929-025-01117-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Chrzanowska NM, Kowalewski J, Lewandowska MA. Use of fluorescence in situ hybridization (FISH) in diagnosis and tailored therapies in solid tumors. Molecules. 2020;25:1864. doi: 10.3390/molecules25081864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.El Tekle G, Garrett WS. Bacteria in cancer initiation, promotion and progression. Nat Rev Cancer. 2023;23:600–618. doi: 10.1038/s41568-023-00594-2. [DOI] [PubMed] [Google Scholar]
  • 92.Chai X, Wang J, Li H, Gao C, Li S, Wei C, Huang J, Tian Y, Yuan J, Lu J, et al. Intratumor microbiome features reveal antitumor potentials of intrahepatic cholangiocarcinoma. Gut Microbes. 2023;15:2156255. doi: 10.1080/19490976.2022.2156255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Mitsuhashi K, Nosho K, Sukawa Y, Matsunaga Y, Ito M, Kurihara H, Kanno S, Igarashi H, Naito T, Adachi Y, et al. Association of Fusobacterium species in pancreatic cancer tissues with molecular features and prognosis. Oncotarget. 2015;6:7209–7220. doi: 10.18632/oncotarget.3109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zhang M, Zhang Y, Sun Y, Wang S, Liang H, Han Y. Intratumoral microbiota impacts the First-line treatment efficacy and survival in Non-Small cell lung cancer patients free of lung infection. J Healthc Eng. 2022;2022:5466853. doi: 10.1155/2022/5466853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Yamamura K, Izumi D, Kandimalla R, Sonohara F, Baba Y, Yoshida N, Kodera Y, Baba H, Goel A. Intratumoral Fusobacterium nucleatum levels predict therapeutic response to neoadjuvant chemotherapy in esophageal squamous cell carcinoma. Clin Cancer Res. 2019;25:6170–6179. doi: 10.1158/1078-0432.CCR-19-0318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Jiang SS, Xie YL, Xiao XY, Kang ZR, Lin XL, Zhang L, Li CS, Qian Y, Xu PP, Leng XX, et al. Fusobacterium nucleatum-derived succinic acid induces tumor resistance to immunotherapy in colorectal cancer. Cell Host Microbe. 2023;31:781–797.e9. doi: 10.1016/j.chom.2023.04.010. [DOI] [PubMed] [Google Scholar]
  • 97.Cercek A, Lumish M, Sinopoli J, Weiss J, Shia J, Lamendola-Essel M, El Dika IH, Segal N, Shcherba M, Sugarman R, et al. PD-1 blockade in mismatch Repair-deficient, locally advanced rectal cancer. N Engl J Med. 2022;386:2363–2376. doi: 10.1056/NEJMoa2201445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371:602–609. doi: 10.1126/science.abb5920. [DOI] [PubMed] [Google Scholar]
  • 99.Shan J, Han D, Shen C, Lei Q, Zhang Y. Mechanism and strategies of immunotherapy resistance in colorectal cancer. Front Immunoly. 2022;13:1016646. doi: 10.3389/fimmu.2022.1016646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Park AK, Fong Y, Kim SI, Yang J, Murad JP, Lu J, Jeang B, Chang WC, Chen NG, Thomas SH, et al. Effective combination immunotherapy using oncolytic viruses to deliver CAR targets to solid tumors. Sci Transl Med. 2020;12:eaaz1863. doi: 10.1126/scitranslmed.aaz1863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Guo F, Das JK, Kobayashi KS, Qin QM, A Ficht T, Alaniz RC, Song J, Figueiredo P. Live attenuated bacterium limits cancer resistance to CAR-T therapy by remodeling the tumor microenvironment. J Immunother Cancer. 2022;10:e003760. doi: 10.1136/jitc-2021-003760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.LaCourse KD, Zepeda-Rivera M, Kempchinsky AG, Baryiames A, Minot SS, Johnston CD, Bullman S. The cancer chemotherapeutic 5-fluorouracil is a potent Fusobacterium nucleatum inhibitor and its activity is modified by intratumoral microbiota. Cell Rep. 2022;41:111625. doi: 10.1016/j.celrep.2022.111625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Jia D, Wang Q, Qi Y, Jiang Y, He J, Lin Y, Sun Y, Xu J, Chen W, Fan L, et al. Microbial metabolite enhances immunotherapy efficacy by modulating T cell stemness in pan-cancer. Cell. 2024;187:1651–1665.e21. doi: 10.1016/j.cell.2024.02.022. [DOI] [PubMed] [Google Scholar]
  • 104.Shi Y, Zheng W, Yang K, Harris KG, Ni K, Xue L, Lin W, Chang EB, Weichselbaum RR, Fu YX. Intratumoral accumulation of gut microbiota facilitates CD47-based immunotherapy via STING signaling. J Exp Med. 2020;217:e20192282. doi: 10.1084/jem.20192282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Si W, Liang H, Bugno J, Xu Q, Ding X, Yang K, Fu Y, Weichselbaum RR, Zhao X, Wang L. Lactobacillus rhamnosus GG induces cGAS/STING-dependent type I interferon and improves response to immune checkpoint blockade. Gut. 2022;71:521–533. doi: 10.1136/gutjnl-2020-323426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Linn YH, Thu KK, Win NHH. Effect of probiotics for the prevention of acute Radiation-induced diarrhoea among cervical cancer patients: A randomized Double-Blind Placebo-controlled study. Probiotics Antimicrob Proteins. 2019;11:638–647. doi: 10.1007/s12602-018-9408-9. [DOI] [PubMed] [Google Scholar]
  • 107.Montalban-Arques A, Katkeviciute E, Busenhart P, Bircher A, Wirbel J, Zeller G, Morsy Y, Borsig L, Glaus Garzon JF, Müller A, et al. Commensal Clostridiales strains mediate effective anti-cancer immune response against solid tumors. Cell Host Microbe. 2021;29:1573–1588.e7. doi: 10.1016/j.chom.2021.08.001. [DOI] [PubMed] [Google Scholar]
  • 108.Canale FP, Basso C, Antonini G, Perotti M, Li N, Sokolovska A, Neumann J, James MJ, Geiger S, Jin W, et al. Metabolic modulation of tumours with engineered bacteria for immunotherapy. Nature. 2021;598:662–666. doi: 10.1038/s41586-021-04003-2. [DOI] [PubMed] [Google Scholar]
  • 109.Lam KC, Araya RE, Huang A, Chen Q, Di Modica M, Rodrigues RR, Lopès A, Johnson SB, Schwarz B, Bohrnsen E, et al. Microbiota triggers STING-type I IFN-dependent monocyte reprogramming of the tumor microenvironment. Cell. 2021;184:5338–5356.e21. doi: 10.1016/j.cell.2021.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Feng T, Li P, Li S, Wang Y, Lv J, Xia T, Lee HJ, Piao HL, Chen D, Ma Y. Metabolic state uncovers prognosis insights of esophageal squamous cell carcinoma patients. J Transl Med. 2025;23:342. doi: 10.1186/s12967-025-06087-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Xue C, Chu Q, Zheng Q, Yuan X, Su Y, Bao Z, Lu J, Li L. Current understanding of the intratumoral microbiome in various tumors. Cell Rep Med. 2023;4:100884. doi: 10.1016/j.xcrm.2022.100884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Elhanani O, Ben-Uri R, Keren L. Spatial profiling technologies illuminate the tumor microenvironment. Cancer Cell. 2023;41:404–420. doi: 10.1016/j.ccell.2023.01.010. [DOI] [PubMed] [Google Scholar]
  • 113.Meng YF, Fan ZY, Zhou B, Zhan HX. Role of the intratumoral microbiome in tumor progression and therapeutics implications. Biochim Biophys Acta Rev Cancer. 2023;1878:189014. doi: 10.1016/j.bbcan.2023.189014. [DOI] [PubMed] [Google Scholar]
  • 114.Liu H, Zhang J, Rao Y, Jin S, Zhang C, Bai D. Intratumoral microbiota: An emerging force in diagnosing and treating hepatocellular carcinoma. Med Oncol. 2024;41:300. doi: 10.1007/s12032-024-02545-9. [DOI] [PubMed] [Google Scholar]
  • 115.Wang N, Li X, Wang R, Ding Z. Spatial transcriptomics and proteomics technologies for deconvoluting the tumor microenvironment. Biotechnol J. 2021;16:e2100041. doi: 10.1002/biot.202100041. [DOI] [PubMed] [Google Scholar]
  • 116.Fang P, Yang J, Zhang H, Shuai D, Li M, Chen L, Liu L. Emerging roles of intratumoral microbiota: A key to novel cancer therapies. Front Oncol. 2025;15:1506577. doi: 10.3389/fonc.2025.1506577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Su ACY, Ding X, Lau HCH, Kang X, Li Q, Wang X, Liu Y, Jiang L, Lu Y, Liu W, et al. Lactococcus lactis HkyuLL 10 suppresses colorectal tumourigenesis and restores gut microbiota through its generated alpha-mannosidase. Gut. 2024;73:1478–1488. doi: 10.1136/gutjnl-2023-330835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Liu J, Xie J, Dong P. Editorial: Transcriptome analysis in tumor microenvironment and tumor heterogeneity. Oncol Res. 2023;32:99–100. doi: 10.32604/or.2023.045719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Horvath TD, Haidacher SJ, Engevik MA, Luck B, Ruan W, Ihekweazu F, Bajaj M, Hoch KM, Oezguen N, Spinler JK, et al. Interrogation of the mammalian gut-brain axis using LC-MS/MS-based targeted metabolomics with in vitro bacterial and organoid cultures and in vivo gnotobiotic mouse models. Nat Protoc. 2023;18:490–529. doi: 10.1038/s41596-022-00767-7. [DOI] [PubMed] [Google Scholar]
  • 120.Geng S, Guo P, Li X, Shi Y, Wang J, Cao M, Zhang Y, Zhang K, Li A, Song H, et al. Biomimetic nanovehicle-enabled targeted depletion of intratumoral Fusobacterium nucleatum synergizes with PD-L1 blockade against breast cancer. ACS Nano. 2024;18:8971–8987. doi: 10.1021/acsnano.3c12687. [DOI] [PubMed] [Google Scholar]
  • 121.Puschhof J, Pleguezuelos-Manzano C, Martinez-Silgado A, Akkerman N, Saftien A, Boot C, de Waal A, Beumer J, Dutta D, Heo I, Clevers H. Intestinal organoid cocultures with microbes. Nature Protoc. 2021;16:4633–4649. doi: 10.1038/s41596-021-00589-z. [DOI] [PubMed] [Google Scholar]
  • 122.Puschhof J, Pleguezuelos-Manzano C, Clevers H. Organoids and organs-on-chips: Insights into human gut-microbe interactions. Cell Host Microbe. 2021;29:867–878. doi: 10.1016/j.chom.2021.04.002. [DOI] [PubMed] [Google Scholar]
  • 123.Allam-Ndoul B, Castonguay-Paradis S, Veilleux A. Gut microbiota and intestinal Trans-epithelial permeability. Int J Mol Sci. 2020;21:6402. doi: 10.3390/ijms21176402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Dutta D, Heo I, Clevers H. Disease modeling in stem Cell-derived 3D organoid systems. Trends Mol Med. 2017;23:393–410. doi: 10.1016/j.molmed.2017.02.007. [DOI] [PubMed] [Google Scholar]
  • 125.Chen S, Zhou Z, Li Y, Du Y, Chen G. Application of single-cell sequencing to the research of tumor microenvironment. Fron Immunol. 2023;14:1285540. doi: 10.3389/fimmu.2023.1285540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Lau HCH, Kranenburg O, Xiao H, Yu J. Organoid models of gastrointestinal cancers in basic and translational research. Nat Rev Gastroenterol Hepatol. 2020;17:203–222. doi: 10.1038/s41575-019-0255-2. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Data Availability Statement

Not applicable.


Articles from Oncology Letters are provided here courtesy of Spandidos Publications

RESOURCES