DOI:
10.1039/D5PM00045A
(Review Article)
RSC Pharm., 2025,
2, 865-881
Intratumoral microbiota: a new perspective in cancer initiation, development, and therapy
Received
15th February 2025
, Accepted 31st July 2025
First published on 1st August 2025
Abstract
Microbes have been identified as significantly impacting human health. Considerable attention has been focused on how microbiota affects cancer initiation, development, and therapeutic response. Currently, the biological functions of intratumoral microbiota have been preliminarily elucidated in tumors with high microbial abundance. However, the biological roles of the microbiota and their clinical significance in tumors with low microbial abundance, to our knowledge, remain largely unexplored. This gap in understanding is primarily due to the limited sensitivity of current detection technologies. This review provides a detailed examination of intratumoral microbiota characteristics and their interactions with the tumor microenvironment, focusing on the microbiota composition in various systems and its clinical role in different tumor types. Furthermore, the review explores the potential applications of intratumoral microbiota in cancer immunotherapy, including their role as immune enhancers, new drug delivery targets, and anticancer therapeutic agents. In conclusion, these insights may facilitate the use of microbiota for cancer diagnosis, prognosis, and the development of new therapeutic strategies.
 Huiling Liu | Huiling Liu is an undergraduate student at the School of Pharmacy Science at the University of South China. She is currently conducting scientific research at Prof. Cui-Yun Yu's lab. |
 Zhonghui Luo | Zhonghui Luo is currently a doctoral candidate at the School of Pharmacy Science at the University of South China. He is currently conducting scientific research at Prof. Cui-Yun Yu's lab. His current research focuses on the mechanisms associated with intestinal injury and anti-tumor immunotherapy. |
 Fangzhen Luo | Fangzhen Luo is an associate professor at the Institute of Pharmacy & Pharmacology at the University of South China. She received her Ph.D. in Medicine under the supervision of Prof. Zhongyu Li in 2022 from the University of South China. Her research focuses on the pathogenesis and drug resistance of pathogenic microorganisms. |
 Xilian Wang | Xilian Wang is a deputy chief physician of the Pain Department at the First Affiliated Hospital of the University of South China. Her research focuses on the mechanism of neuropathic pain. |
 Hua Wei | Hua Wei is a full professor at the School of Pharmaceutical Science at the University of South China. He received his PhD in polymer science under the supervision of Prof. Xianzheng Zhang and Prof. Renxi Zhuo in 2009 from Wuhan University, China. He later worked with Prof. Suzie H. Pun as a postdoctoral fellow in the Department of Bioengineering at the University of Washington. He was selected as one of the Journal of Materials Chemistry B Emerging Investigators (2017) and Biomaterials Science Emerging Investigators (2021) and has been serving as an editorial advisory board member of ACS Biomaterials Science & Engineering since 2019. |
 Cui-Yun Yu | Cui-Yun Yu is a full professor at the Institute of Pharmacy & Pharmacology and Dean of the School of Pharmaceutical Science at the University of South China. She received her PhD in polymer science under the supervision of Prof. Sixue Cheng and Prof. Renxi Zhuo in 2009 from Wuhan University, China. She has set up her own research group at the University of South China since her PhD graduation. Her research interests focus on the design and development of novel biomaterials for targeted drug delivery and tissue engineering applications. |
1. Introduction
Cancer is a significant global health issue.1 Tumor therapy initially targets molecules like genes, DNA, and proteins and then proceeds to precise subcellular organelles, including mitochondria, endoplasmic reticulum, lysosomes, and nuclei. Mitochondria likely originated from an ancient endosymbiotic event wherein proteobacteria were engulfed to facilitate energy production and cellular metabolism,2 which implies that bacteria can integrate into host cells and fulfill specific biological functions. The human microbiome consists of bacteria, fungi, viruses, and archaea. These organisms are abundant in the digestive tract and also present in smaller numbers in the skin, upper respiratory tract, eye, and urogenital tract, which play a key role in various tumors. To date, considerable research has focused on the effect of non-resident bacteria on cancer development and therapeutic response.
Recent advancements in detection methodologies, particularly next-generation sequencing for the analysis of trace DNA, have significantly improved the precision of bacterial detection within tumors and enabled the identification of specific bacterial species. These technological developments challenge the previously held assumption that the presence of bacteria was merely a consequence of contamination during detection processes. Intratumoral microbiota may either promote or inhibit cancer initiation, progression, and response to immunotherapy. The role of intratumoral microbiota in tumorigenesis depends on its composition and abundance, tumor stage, and the host's immune response. Thus, understanding the microbiota composition across various tumors and their role in cancer initiation and progression can aid in identifying new therapeutic strategies and targets, enhancing treatment efficacy.
In this review, we discuss the characteristics and composition of intratumoral microbiota, their critical role in various tumor tissues, and potential applications in cancer immunotherapy. We emphasize the progress of intratumoral microbiota across various tumors. This work identifies the microbiota as a tool for cancer diagnosis or prognosis, as well as a new therapeutic strategy.
2. Characteristics of intratumoral microbiota in tumor tissue
2.1 Diverse sources
Intratumoral microbiota can originate from three sources: (i) the primary tumor site, where the microorganisms reside in the tissue that gives rise to the tumor. For instance, Porphyromonas gingivalis in the oral cavity can promote oral squamous cell carcinoma progression3 and Helicobacter pylori in the stomach can promote gastric cancer development;4 (ii) normal adjacent tissues (NATs), from which the microbiota can migrate into tumor tissues. Nejman et al. found that the bacterial composition of tumor tissues closely resembles that of NATs;5 (iii) circulation, through which intratumoral microbiota, primarily found in tumor and immune cells, can migrate to distant tumor tissues via the bloodstream. Intestinal bacteria can reach different tumor sites via different organ–gut axes.
2.2 High heterogeneity
Intratumoral microbiota compositions exhibit variation across different tumors. Analyses of the Cancer Genome Atlas (TCGA) database, including genome-wide and transcriptome-wide approaches, reveal characteristics of intratumoral microbiota. Nejman et al. demonstrated microbial composition variation in different tumors by analyzing more than 1500 patient tumor samples.5 Galeano Niño et al. used 16S rDNA sequencing on 44 tumor tissues isolated from 11 patients with colorectal cancer (CRC) to reveal varying degrees of heterogeneity in CRC's intratumoral microbiota.6 Furthermore, microorganisms were confirmed to exhibit heterogeneous spatial distribution. In addition, intratumoral microbiota composition may even vary among different subtypes of the same tumor type.5 Intratumoral microbiota is primarily bacterial, but similar characteristics are observed in intratumoral fungi. The diversity and abundance of cancer-type-specific fungi are generally lower than those of the corresponding bacterial populations.7 Notably, bacterial and fungal abundances, diversities, and co-occurrences are strongly positively correlated in several tumors. The tumor microenvironment (TME) may provide non-competitive spaces for microbial colonization. This contrasts with the gut, where bacterial and fungal populations compete for resources, especially under antitumor or antibiotic therapies.
2.3 Spatiotemporal dynamics of the intratumoral microbiome
The intratumoral microbiota demonstrates spatiotemporal dynamics, characterized by systematic reprogramming of its diversity, abundance, and functionality in response to tumor progression, therapeutic interventions, and alterations in the host microenvironment. Temporally, the lung cancer microbiome undergoes evolution with advancing disease stages: early-stage tumors exhibit greater microbial diversity with a predominance of Actinobacteria, whereas late-stage lesions are enriched with butyrate-producing bacteria, such as Roseburia spp. Microbial-derived butyrate functions by inhibiting histone deacetylase HDAC2, enhancing H3K27 acetylation in the promoter region of the long non-coding RNA H19, and inducing M2 macrophage polarization, collectively facilitating metastatic progression.8 Spatially, the microbiota establish heterogeneous microniches within tumors. For example, in oral squamous cell carcinoma and colorectal cancer, bacterial communities colonize hypovascularized and highly immunosuppressive microenvironments associated with malignant cells.6 Therapeutically, interventions significantly alter the composition of the microbiome: immune checkpoint blockade (ICB) decreases microbial richness across various tumor models.9 In contrast, shifts in the intratumoral microbiota play a critical role in modulating the efficacy of ICB. For instance, the translocation of the probiotic Limosilactobacillus reuteri to melanoma sites enhances the response to ICB through the production of indole-3-aldehyde (I3A), a catabolite of tryptophan.10 In conclusion, the dynamic evolution and biological functions of intratumoral microbiota are governed by a tripartite regulation involving stage progression, therapy-induced perturbations, and metabolic feedback. Their spatiotemporal heterogeneity may offer new insights for prognostic stratification and the development of targeted interventions.
3. Progress of the intratumoral microbiota in various types of tumors
3.1 Digestive system neoplasm
3.1.1 Esophageal cancer.
Esophageal cancer (ESCA) is divided into two main histological subtypes: esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma.11 Microbes such as Fusobacteria, Lactobacillales, Clostridia, Proteobacteria, and Negativicutes are correlated with the clinical characteristics of patients with ESCA.12 Intratumoral microbiota enhances antitumor immunity by recruiting the infiltration of immune cells. Streptococcus enrichment is often linked to increased CD8+ T cell infiltration and a favorable response to anti-PD-1 therapy.13 In addition, Fusobacterium, identified as a pathogen, accelerates ESCC tumorigenesis and metastasis via inducing DNA damage, recruiting myeloid-derived suppressor cells (MDSCs),14 activating the NF-κβ pathway,15 and increasing METTL3-mediated m6A methylation.16Fusobacterium nucleatum invades senescent ESCC cells, enhancing senescence-associated secretory phenotype secretion, and thereby promoting ESCC progression.17 Li et al. reported that Fusobacterium nucleatum inhibits T cell proliferation and cytokine secretion, attenuating antitumor immunity in ESCC.18 These findings highlight the potential for intratumoral microbes and their associated metabolites to influence the tumor immune microenvironment and the efficacy of immunotherapy.
3.1.2 Gastric cancer.
The gastric cancer microbiota is characterized by reduced diversity and enriched Oceanobacter, Methylobacterium, and Syntrophomonas genera. Helicobacter pylori, a common inhabitant, is classified as a class I carcinogen by the WHO.19 However, Helicobacter pylori is not the only microbe accelerating gastric cancer progression. Fu et al. identified Streptococcus anginosus as a gastric tumorigenesis-promoting pathogen (Fig. 1A).20 Li et al. have demonstrated that the Streptococcus anginosus group consistently upregulates all metabolites associated with arginine metabolism in gastric cancer tumor specimens. This metabolic reprogramming facilitates tumor cell proliferation, migration, and invasion, while simultaneously inhibiting the differentiation and infiltration of CD8+ T lymphocytes within the tumor immune microenvironment (TIME). These processes collectively contribute to the tumorigenesis and progression of gastric cancer.21
 |
| Fig. 1 (A) Schematic illustration of Streptococcus anginosus promoting gastric inflammation, atrophy, and tumorigenesis in mice. Reproduced with permission from ref. 20. Copyright, 2024, Elsevier Inc. (B) Schematic depiction of Helicobacter pylori CagA eliciting BRCAness to induce genome instability. Reproduced with permission from ref. 25. Copyright, 2021, Elsevier Inc. (C) Schematic depiction of the fungal mycobiome driving IL-33 secretion and type 2 immunity in pancreatic cancer. Reproduced with permission from ref. 52. Copyright, 2022, Elsevier Inc. (D) Schematic depiction of PDAC LTS displaying high tumor microbial diversity and immunoactivation. Reproduced with permission from ref. 54. Copyright, 2019, Elsevier Inc. | |
Helicobacter pylori promotes gastric tumorigenesis via mechanisms such as DNA damage,22 oncogenic pathway activation,23 and induction of chronic inflammation and apoptosis.24 Specific virulence factors, including the cytotoxin-associated gene A (CagA) and vacuolating cytotoxin A (VacA), are critical in inducing host cell DNA damage. CagA induces DNA double-strand breaks and disrupts error-free DNA repair via homologous recombination, contributing to gastric carcinogenesis (Fig. 1B).25 VacA induces vacuolization, necrosis, and apoptosis.26 The link between gastric cancer and Helicobacter pylori is one of the strongest between a single bacterium and cancer causation. Prospective data indicate that Helicobacter pylori precedes tumorigenesis, and antibiotic eradication may reduce gastric cancer incidence.
3.1.3 Colorectal cancer.
The gut microbiota, found in CRC, engages in structured crosstalk with the host, influencing multiple physiological processes. CRC-promoting bacterial species like Fusobacterium nucleatum, Escherichia coli, and Bacteroides fragilis, and CRC-protecting bacterial species like Clostridium butyricum, Streptococcus thermophilus, and Lacticaseibacillus paracasei are part of the gut microbiota and play critical roles in the development of CRC.27
Intratumoral microbiota produces genotoxins that damage colonic epithelial cell DNA, promoting CRC development. Escherichia coli harboring polyketide synthetase pathogenicity island, known to encode the genotoxin colibactin, are increasingly associated with CRC.28–30Escherichia coli that produce colibactin induce DNA breaks, cell cycle arrest, and senescence, promoting tumor growth.31Campylobacter jejuni produces a cytolethal-distending toxin that causes DNA double-strand breaks.32 Moreover, the toxin produced by enterotoxigenic Bacteroides fragilis compromises colonic epithelial and barrier integrity, causing inflammation and inducing epithelial cell proliferation through activating the NF-κβ and STAT3 signaling pathways, promoting CRC progression.33 Recent evidence suggests that the microbiota may drive tumor metastasis by regulating CRC metabolism. Fusobacterium nucleatum, a key periodontal pathogen, is enriched in CRC. Fusobacterium nucleatum subspecies animalis (Fna) exists in two distinct clades: Fna C1 and Fna C2, with only the latter shown to induce tumors and promote oxidative stress in intestinal metabolism in mouse models.34Fusobacterium nucleatum activates the TLR4/Keap1/NRF2 pathway, increasing CYP2J2 and 12,13-EpOME levels, which promote tumor metastasis.35 Cui et al. reported that trans-3-indoleacrylic acid, a tryptophan metabolite from Peptostreptococcus anaerobius, promotes colorectal carcinogenesis via inhibiting ferroptosis.36 These studies indicate that gut microbes can indirectly affect the tumor microenvironment through metabolites or the immune system, potentially altering the composition and function of the intratumoral microbiota.
3.1.4 Liver cancer.
Primary liver cancer is classified into three main histological subtypes: hepatocellular carcinoma (HCC), intrahepatic cholangiocarcinoma (ICC), and the combined HCC-ICC.37 HCC is the most prevalent type of liver cancer, characterized by high recurrence rates and poor prognosis.38 Microbial diversity in HCC tissues significantly exceeds that in adjacent tissues. Bacteroidetes, Firmicutes, and Proteobacteria are more abundant in HCC tissues, whereas antitumor bacteria like Pseudomonas are less so.39 Cai et al. identified Burkholderiales, Pseudomonadales, Xanthomonadales, Bacillales, and Clostridiales as the predominant bacterial orders in ICC. An increased presence of Paraburkholderia fungorum in paracancerous tissues suggests a role in ICC pathogenesis.40
Microorganisms contribute to HCC development via direct and indirect mechanisms.41Stenotrophomonas maltophilia is found in HCC patients with cirrhosis. Stenotrophomonas maltophilia activates the TLR4/NF-κβ/NLRP3 pathway, resulting in a senescence-associated secretory phenotype in hepatic stellate cells, which induces inflammation and promotes cirrhosis and HCC progression.42 Sun et al. found that CD68+ macrophages were more prevalent in areas rich in intratumoral microbiota.43 Comparative analysis of HCC subtypes shows that the bacteria-dominant subtype increased M2 macrophage infiltration and upregulated metabolic pathways compared to the virus-dominated subtype. This infiltration of M2 macrophages correlates positively with amino acid metabolism. Notably, HCC metabolic patterns differ significantly from those in normal liver tissues, indicating that the unique microbial environments in HCC patients may influence metabolic variations.44 These findings suggest that the intratumoral microbiota could modulate the tumor immune microenvironment through metabolic reprogramming.
3.1.5 Pancreatic cancer.
The pancreatic cancer microbiome, communicating with the gut microbiome, affects the host immune response and the disease's natural history. The predominant bacterial genera in the pancreas are Proteobacteria, Bacteroidetes, and Firmicutes. Enterobacteriaceae and Pseudomonadaceae, identified in pancreatic ductal adenocarcinoma (PDAC), may modulate tumor sensitivity to gemcitabine (Gem).45
Intratumoral microbiota can remodel the tumor immune microenvironment, accelerating pancreatic cancer development. Bacteria linked to PDAC trigger innate and adaptive immune suppression, reducing the amount of MDSCs and enhancing M1 macrophage and TH1 differentiation, thereby activating CD8+ T cells.46 Indole-producing bacteria, including Lactobacillus murinus, elevate aryl hydrocarbon receptor transcriptional responses and promote an immunosuppressive TME in PDAC, promoting tumor growth.47 Moreover, colonization within the microenvironment of PDAC results in the metabolic production of butyrate. This meta Clostridium butyricum bolite increases the susceptibility of PDAC tumor cells to ferroptosis inducers, such as RSL3. The combined therapeutic approach, utilizing either Clostridium butyricum colonization or exogenous butyrate supplementation alongside RSL3, synergistically amplifies the inhibitory impact on the proliferation of PDAC tumor cells.48Porphyromonas gingivalis, a key periodontal pathogen, is strongly associated with pancreatic cancer.49 Ma et al. reported that Porphyromonas gingivalis boosts neutrophil chemokines and elastase secretion, creating a proinflammatory TME and promoting pancreatic cancer progression.50 Saba et al. found that Porphyromonas gingivalis protects tumor cells from ROS-induced cell death due to nutrient stress, accelerating PDAC progression.51 Intratumoral fungi stimulate IL-33 secretion and type 2 immunity, potentially promoting tumor growth (Fig. 1C).52Malassezia activates mannose-binding lectin, initiating the complement cascade and promoting PDAC progression.53 However, the intratumoral microbiota in pancreatic cancer may not be invariably harmful, with some microbes linked to better clinical outcomes. Intratumoral microbiota may modulate immune infiltration in pancreatic cancer long-term survivors, thereby inhibiting tumor progression. Riquelme et al. demonstrate that gut bacteria from long-term survival patients modulate pancreatic intratumoral bacterial composition, enhance tumor CD8+ T cell activation, and inhibit MDSCs and regulatory T cells (Tregs) accumulation, thereby inhibiting tumor growth (Fig. 1D).54 Furthermore, Ghaddar et al. found that a subset of tumors contains somatic-cell-associated bacteria, primarily associated with tumor cells and rarely found in nonmalignant tissues.55 This suggests that microbiome targeting may prevent oncogenesis, reverse intratumoral immune tolerance, and enhance the effectiveness of checkpoint-based immunotherapy.
3.2 Respiratory system neoplasms
3.2.1 Lung cancer.
Microorganisms are closely associated with lung cancer,56 and the predominant bacteria in lung cancer are Proteobacteria and Actinobacteria.5 Fungi have been identified within lung cancer cells. Smokers with lung cancer have more abundant intratumoral fungi, including higher levels of Aspergillus and Umbelliferae fungi. Intratumoral fungi may serve as markers to effectively distinguish lung cancer from healthy controls, holding potential for early diagnosis. In addition, Goto et al. suggest that the John Cunningham virus may contribute to lung cancer progression.57
Intratumoral microbiota in lung cancer can modulate cytokine production and foster a chronic inflammatory microenvironment that promotes tumorigenesis. Tyler et al. reported that lung microbiota activates γδ T cells, triggering the production of IL-17 and other effector molecules that promote inflammation and tumor cell proliferation.58 Intratumoral microbiota can induce an immunosuppressive TME and promote lung cancer progression. Notably, reduced bacterial loads correlate with decreased Tregs, enhanced NK cell activity, and reduced lung cancer metastasis.59 This finding highlights the importance of microbiota–immune interactions in cancer progression. Studies have revealed a Treg cell-driven mechanism underlying lung-specific immunosuppression.60,61 Additionally, Liu et al. reported that the tumor-resident fungus Aspergillus sydowi induces IL-1β secretion and MDSC activation via the β-glucan-mediated Dectin-1/CARD9 pathway, thereby advancing lung cancer progression.62 Low concentration of butyrate from the intratumoral microbiome may promote lung cancer progression and metastasis by enhancing M2 macrophage polarization and function.63 In summary, the microbiome contributes to lung cancer through multiple biological pathways, including genotoxicity, inflammation, immune response, and angiogenesis.
3.2.2 Nasopharyngeal cancer.
Nasopharyngeal carcinoma (NPC) originates from the nasopharyngeal mucosa as an epithelial carcinoma in NPC tumor tissues, where the microbiota is present, with Corynebacterium and Staphylococcus being predominant.64,65 The NPC microbiota plays a role in intratumoral infiltration and TME remodeling. Qiao et al. reported a strong association between high bacterial load and reduced CD8+ T cell infiltration, which contributes to an immunosuppressive environment in NPC.66 Furthermore, patients with a high bacterial load exhibited significant proliferation dependent on cell cycling. Oral-derived microbes were significantly enriched in the nasopharynx and closely associated with epithelial EBV infection.67 This finding suggests that blocking the translocation of microbes from the oral cavity to the nasopharynx could be a potential preventative intervention for NPC.
3.3 Reproductive system neoplasms
3.3.1 Cervical cancer.
Intratumoral microbiota may promote cervical carcinogenesis by inducing immune response drivers. Human papillomavirus (HPV) is established as the primary causative agent in cervical carcinogenesis. Chlamydia trachomatis is recognized as a cofactor of HPV in cervical cancer. Coinfections with Chlamydia trachomatis are more frequently observed in patients with invasive cervical cancers.68 Cervical cancer development involves a substantial decrease in Lactobacillus species and an increase in Gardnerella vaginalis, Prevotella bivia, Porphyromonas spp., and Streptococcus spp.69Lactobacillus depletion may foster a proinflammatory environment, inducing malignant cell proliferation, and increasing HPV E6 and E7 oncogene expression, promoting cervical cancer development.70Lactobacillus iners, a common species, appears to be an opportunistic pathogen.71 Colebert et al. illustrate that the intratumoral colonization of Lactobacillus iners in cervical cancer contributes to chemoradiotherapy resistance viaL-lactate-mediated metabolic reprogramming, which is significantly associated with decreased patient survival rates.72 Furthermore, higher levels of Robiginitomaculum, Klebsiella, Micromonospora, and Microbispora are linked to cervical cancer mortality, while Methylobacter levels show an inverse relationship.73 Microbiome relative abundance and tumor classification can predict cervical cancer prognosis.
3.3.2 Prostate cancer.
Prostate cancer is the second most frequent malignancy among men worldwide.74 Current evidence indicates that the microbiota induces an inflammatory prostate microenvironment, promoting prostate cancer development and progression.75 Infection with Propionibacterium acnes activates the COX2-prostaglandin and plasminogen-matrix metalloproteinase pathways, triggering a strong inflammatory response.76 Additionally, Ma et al. found Pediococcus pentosaceus, Listeria monocytogenes, and Lactobacillus crispatus in prostate cancer.77 Therefore, further investigation into microbiome interaction with prostate cancer cells is warranted.
3.4 Other cancers
3.4.1 Breast cancer.
Breast cancer is the most common type of cancer in women. The metabolic status of breast cancer is highly plastic, and certain microbial genera are significantly correlated with metabolic activity in cancer. Researchers found that Porphyromonas, Lacibacter, Ezakiella, Fusobacterium, and Pseudomonas are more abundant in higher-stage breast tumors compared to lower-stage tumors and healthy breast microbiota. These genera exhibit distinct features in normal and tumor tissues.78 Additionally, Banerjee et al. discovered that breast cancer contains fungi, viruses, Chlamydiae, and parasites (Fig. 2A).79
 |
| Fig. 2 (A) Bar graphs showing breast cancer containing fungi, viruses, Chlamydiae, and parasites. Reproduced with permission from ref. 79. Copyright, 2021. (B) Schematic illustration of the microbial metabolite trimethylamine N-oxide promoting antitumor immunity in TNBC. Reproduced with permission from ref. 82. Copyright, 2022, Elsevier Inc. (C) Schematic illustration of tumor-resident intracellular microbiota promoting metastatic colonization in breast cancer. Reproduced with permission from ref. 78. Copyright, 2022, Elsevier Inc. (D) Schematic illustration of the dietary tryptophan metabolite released by Lactobacillus reuteri facilitates ICI treatment. Reproduced with permission from ref. 10. Copyright, 2023, Elsevier Inc. | |
Intratumoral microbiota may affect the tumor immune microenvironment. Propionibacterium is more abundant in healthy controls and NATs but is scarce in tumor tissues. Higher Propionibacterium levels correlate with increased T-cell activation and decreased oncogenic growth, suggesting that its absence could promote tumorigenesis by inhibiting an adaptive antitumor response and fostering a pro-tumorigenic environment. Besides directly affecting host immune responses, breast microbiota produces substances that can boost the antitumor immunity response. With a higher prevalence in non-breast cancer tissues, the Streptococcus genus generates cadaverine, which inhibits breast cancer invasion and epithelial–mesenchymal transition.80 Moreover, bile acids from the microbiota in breast tumors are associated with high cell proliferation and poorer survival rates.81 Shao et al. found that the microbial metabolite trimethylamine N-oxide activates the endoplasmic reticulum stress kinase PERK, inducing pyroptosis in malignant cells and strengthening the immune response against triple-negative breast cancer (TNBC) (Fig. 2B).82 Furthermore, in a mouse model of spontaneous breast tumors, the presence of bacteria induces a reorganization of the actin cytoskeleton in circulating tumor cells (CTCs) in the blood. This restructuring enhances CTCs’ ability to withstand fluid shear stress, promoting CTC survival and breast cancer metastasis (Fig. 2C).78 Although present in low biomass, tumor-resident microbiota plays a crucial role in cancer metastasis, making their manipulation a potentially valuable avenue for advancing oncology care.
3.4.2 Melanoma.
The cutaneous microbiota, with its potential role in melanoma development and treatment response, has emerged as a promising research area. Melanoma, a highly malignant skin tumor, has seen dramatically increased incidence rates globally.83 Compared to other tumors, such as bone, pancreatic, and breast, melanoma has a relatively lower bacterial biomass. The predominant bacterial genera in melanoma have been identified as Acinetobacter, Actinomyces, Corynebacterium, Enterobacter, Roseomonas, and Streptococcus.5
Intratumoral microbiota may inhibit T cell and NK cell activation, thereby prompting melanoma development. In a mouse melanoma model, antibiotic treatment in the lung reduced bacterial load, decreased regulatory T cells, and enhanced T cells and NK cell activation, resulting in fewer lung metastases. Furthermore, recent studies implied a link between Corynebacterium species and melanoma progression, possibly through an IL-17-dependent pathway. Zitvogel et al. reported a stage-associated increase in Corynebacterium among 27 patients with acral melanoma by analyzing culture-based skin swabs and the patients exhibited a higher frequency of Th17 cells. These findings highlight the potential impact of the skin microbiota on local cancers.84 Changes in microbiome composition affect immunotherapy treatment outcomes.85 The probiotic Lactobacillus reuteri, which colonizes and persists within melanoma, secretes dietary tryptophan catabolite indole-3-aldehyde, stimulating an immune response. Indole-3-aldehyde activates AhR signaling in CD8+ T cells, promoting interferon-γ production and enhancing the efficacy of immune checkpoint inhibitors (Fig. 2D).10 Emerging evidence indicates that the composition and diversity of the skin microbiota critically modulate the efficacy of immune checkpoint inhibitors for skin cancer treatment.
4. Crosstalk between the intratumoral microbiota and TME
The relationship between the microbiota and tumors is highly complex, involving an intricate network of microbial infection, dysbiosis, tumorigenesis, and the TME.86–88 The TME, encompassing tumor cells, immune cells, stromal cells, and a dense microvascular network, is the critical internal milieu essential for tumor cell existence and proliferation. Intratumoral microbiota has been identified as a crucial component of the TME.6 The unique features of the TME including hypoxia, angiogenesis, low pH, and immunosuppression facilitate microbial invasion and colonization. In turn, intratumoral microbiota can affect the function of tumor and immune cells and their TME, playing a critical role in cancer initiation, development, and metastasis.
4.1 Effect of the TME on microbial colonization
Several studies suggest that the microbiota migrates after cancer formation. Specific tumor microenvironments may facilitate microbial invasion and colonization. The hypoxic environment in tumors is highly favorable for the growth of facultative anaerobes and anammox bacteria, including Clostridium perfringens, Escherichia coli, Listeria monocytogenes, and Bifidobacterium bifidum.89 Additionally, increased endothelial leakiness in tumor neovascularization facilitates microbiota entry into the tumor tissue through the bloodstream.90 Furthermore, constantly necrotic tumor tissue provides ample nutrients for microbial reproduction. Most cancer cells depend on aerobic glycolysis, known as the Warburg effect, to provide energy for tumor growth.91 Unlike most normal tissues, tumor cells tend to generate lactate from glucose even when oxygen is sufficient to support mitochondrial oxidative phosphorylation. The acidic environment promotes a highly immunosuppressive state in the TME. Deregulations in the tumor immune system may permit unhindered pathogen growth. These features create conducive conditions for microbiota colonization and survival.
4.2 Role of intratumoral microbiota in the TME
The International Agency for Cancer Research estimates that about 3.7 × 1030 microorganisms inhabit the earth, contributing to about 20% of human malignancies, with only 12 identified as human carcinogens.92 Microorganisms within tumors have a dual role. On the one hand, intratumoral microbiota can promote tumor progression by inducing immunosuppression, inflammation, oncogenic signaling pathways, and host gene mutations. On the other hand, intratumoral microbiota can enhance antitumor immunity and therapeutic efficacy by activating STING, promoting tertiary lymphoid structure maturation,93,94 and presenting microbial antigens. The role of intratumoral microbiota as a promoter or suppressor of tumors depends on its composition and abundance, tumor stage, and the host immune system response. Considering the diverse microbiota composition across tumors, targeting specific intratumoral microbiota may enable precision diagnosis, treatment, and prognosis in clinical settings.
5. Potential applications of intratumoral microbiota in cancer therapy
Given the crucial role played by intratumoral microbiota in cancer initiation and development, intratumoral microbiota can be utilized to stimulate the immune response, thus enhancing the efficacy of immunotherapy. On the other hand, intratumoral microbiota can be targeted to eliminate microorganisms at specific sites as targets, thereby enhancing precision cancer treatments.
5.1 Intratumoral microbiota as immune enhancers in immunotherapy
In recent years, immunotherapy has become a critical component in cancer treatment.95–97 The role of intratumoral microbiota in cancer immunotherapy is increasingly recognized as studies expand. As immune enhancers, intratumoral microbiota can improve antitumor immune responses by remodeling the TME, inducing pyroptosis, and prompting tumor and immune cells to present microbial antigens, thereby enhancing the efficacy of immune checkpoint inhibitors (ICIs).98 Intratumoral microbiota is closely linked to the host immune system response, potentially resolving current dilemmas in immunotherapy.
Eliminating certain intratumoral microbiota can convert a cold TME, thereby enhancing the immunotherapy efficacy. Qu et al. effectively reversed the cold TME by eliminating Fusobacterium nucleatum, which was then repurposed as an immune enhancer for triple-negative breast cancer (TNBC) immunotherapy. Furthermore, dead Fusobacterium nucleatum and non-cellular outer membrane vesicles served as immune enhancers, prompting dendritic cell maturation and T-cell infiltration, which significantly boosted the efficacy of TNBC immunotherapy (Fig. 3A).99 Gastric cancer associated with Helicobacter pylori infection exhibits a higher density of PD-L1+ cells and non-exhausted CD8+ T cells, suggesting that Helicobacter pylori promotes a hot TME and is a favorable prognostic factor for gastric cancer immunotherapy.100 The accumulation of Bifidobacterium bifidum in tumors, which occurs in an interferon-dependent and STING pathway, facilitates anti-CD47 immunotherapy.101 Zhang et al. analyzed 1296 intratumoral microbiome genera across samples from the TCGA database, offering a comprehensive view of the links between the intratumoral microbiome and immune features (Fig. 3B).102 Their findings revealed that Eudoraea increases the abundance of active immune cells in the TME, potentially improving immune checkpoint blockade (ICB) treatment outcomes.
 |
| Fig. 3 (A) Schematic depiction of intratumoral bacteria reversing cold tumors for enhanced therapy of TNBC. Reproduced with permission from ref. 99. Copyright, 2023, American Chemical Society. (B) Associations between intratumoral microbiome abundance and the objective response rate at the genus, family, and order levels. Reproduced with permission from ref. 102. Copyright, 2023. (C) Schematic illustration of the synthesis of BSA-Cu SAN and its function of destroying pathogen-tumor symbionts for antitumor therapy. Reproduced with permission from ref. 126. Copyright 2023, Springer Protocols. (D) Schematic illustration of the synergistic target of the intratumoral microbiome and tumor via MTI-FDU. Reproduced with permission from ref. 135. Copyright 2023, American Chemical Society. | |
Intratumoral microbiota may trigger pyroptosis, thereby enhancing the efficacy of immunotherapy. Pyroptosis, a regulated form of cell death, is mediated by the pore-forming, membrane-targeting gastrin family of proteins.103 Pyroptosis is a critical regulator in physiological processes such as inflammation, cell development, tissue homeostasis, and stress response.104 Shen et al. demonstrated that intravenous administration of Listeria monocytogenes induces gasdermin C-dependent pyroptosis, which modifies the immunosuppressive TME and enhances the efficacy of immunotherapy by prompting a robust antitumor immune response.105 Sun et al. developed an oral bacterial pyroptosis amplifier for CRC. They utilized hyaluronic acid-coated Shewanella oneidensis MR-1 and engineered Escherichia coli for tumor-specific accumulation, leveraging hyaluronic acid's role in bacterial survival in the harsh gastrointestinal tract. This approach enhances oncolytic microbe-triggered pyroptosis, activates the gut mucosal immune response, and inhibits colon tumors and metastasis.106
Furthermore, the display of intratumoral bacterial peptides on host cell human leukocyte antigens (HLAs) may enable the utilization of the intratumoral microbiome in cancer immunotherapy. Microbial antigens, sharing antigenic epitopes with tumor antigens, recruit T cells specific to the microbe, which then recognize and eliminate tumor cells. Given that bacterial antigens are considered non-autologous, they may serve as targets for immunotherapy to stimulate immune responses. Kalaora et al. used 16S rRNA gene sequencing and HLA peptidomics to profile peptide libraries from intratumoral bacteria, revealing that tumor cells can present peptides from these bacteria to provoke an immune response.107 Naghavian et al. also found that microbial peptides activate tumor-infiltrating lymphocytes (TILs) and peripheral blood memory cells, initiating an immune response.108 TILs are essential for the success of immunotherapy and improved survival in various tumors.109 Consequently, intratumoral microbiota can trigger an immune response via microbial peptides.
5.2 Intratumoral microbiota as a new target for drug delivery
Researchers are developing drug delivery systems to enhance the effectiveness of antitumor treatments.110–113 These systems, such as liposomes, hydrogels, and nanoparticles, offer biocompatibility, prolonged circulation, and high drug-loading capacity for anticancer drugs.114–116 Given the critical role of the intratumoral microbiota in carcinogenesis, eliminating specific pathogenic microbiota (Table 1) and restoring tumor sensitivity to ICB therapy could enhance precision cancer treatments and prevent recurrence.
Table 1 The intratumoral microbiota serves as a target for nanodrug delivery
|
Nanoplatform |
Microbiota |
Work |
Ref. |
Delivery of antimicrobial agents |
LipoAgTNZ |
Fusobacterium nucleatum, Escherichia coli Nissle |
Eliminating intratumoral bacteria in the primary tumor and liver metastases. |
121
|
Generating microbial neoantigens. |
|
MTZ/Ftn-DOX@HM |
Fusobacterium nucleatum
|
Eliminating intratumoral bacteria. Remodeling the TME. |
122
|
|
BSA-Cu-SAN |
Fusobacterium nucleatum
|
Disrupting the pathogen–tumor symbiosis |
126
|
|
N-CSs |
|
Reversing Gem resistance. |
127
|
Generating ˙OH radicals. |
|
|
Au@BSA-CuPpIX |
Fusobacterium nucleatum
|
Enhancing ROS-induced apoptosis and the therapeutic efficacy of SDT for orthotopic CRC and inhibiting lung metastasis. |
128
|
|
F. nucleatum-mimicking nanomedicine |
Fusobacterium nucleatum
|
Killing intratumoral bacteria |
129
|
Restoring ICB therapies. |
Co-delivery of antitumor drugs and antimicrobial agents |
sNP@G/IR |
Escherichia coli Nissle 1917 |
Eliminating tumor-resident intracellular bacteria |
134
|
Augmenting drug delivery efficacy. |
|
MTI-FDU |
Fusobacterium nucleatum
|
Achieving the dual target of the intratumoral microbiota and tumor cells. |
135
|
|
PG-Pt-LA/CB(7) |
Fusobacterium nucleatum
|
Eliminating Fusobacterium nucleatum. |
117
|
Enhancing chemotherapeutic efficacy. |
|
GC-DCPA-H2O |
|
Eliminating intratumoral microbiota. |
118
|
Enhancing chemotherapeutic efficacy. |
|
OLP/PP nanoassembly |
Fusobacterium nucleatum
|
Eliminating Fusobacterium nucleatum. |
119
|
Inhibiting tumor growth. |
5.2.1 Delivery of antimicrobial agents.
Antibiotics are routinely used in clinical settings for bacterial infections. However, a single antibiotic's ability to permeate tumor cells and address bacterial infections is impeded by cell membrane barriers and the complex TME. Systemic administration of antibiotics targeting pathogenic bacteria in tumor may disrupt other microbiota in the body. Hence, precise antibiotic delivery to deplete intratumoral pathogenic microbiota without upsetting systemic microbiota imbalance is essential. Drug carrier nanoparticles can selectively target microbiota colonized within tumor cells.120 Huang et al. recently developed a liposome-encapsulated silver-tinidazole complex to eliminate intratumoral bacteria using a remote loading technique. The treatment generates microbial neoantigens that enhance immune system recognition of both infected and uninfected tumor cells, triggering an antitumor immune response.121 Yang et al. also developed a biomimetic nanovehicle mimicking Fusobacterium nucleatum for targeted antibiotic delivery.122 This approach effectively eliminates intratumoral Fusobacterium nucleatum and restores tumor sensitivity to ICB therapy.
The misuse of antibiotics has fostered the emergence of multidrug-resistant strains, complicating antibiotic-dependent treatments.123 Recently, nanozymes have emerged as alternatives to traditional antibiotics, catalyzing reactive oxygen species (ROS) generation to kill bacteria or disrupt biofilms without inducing drug resistance.124,125 Qin et al. designed protein-supported copper single-atom nanozymes (BSA-Cu-SAN) that can passively target tumors, producing ROS and depleting glutathione to promote cancer cell apoptosis. This strategy efficiently eliminates intratumoral Fusobacterium nucleatum and disrupts pathogen-tumor symbionts, blocking the interaction between intratumoral microbiota and tumors (Fig. 3C).126 Gao et al. used nitrogen-doped carbon nanospheres (N-CSs) as nanozymes to generate hydroxyl radicals for catalytic tumor therapy. By inhibiting cytidine deaminase, the N-CSs effectively reverse Gem resistance induced by bacterial cytidine deaminase in mouse models.127 Furthermore, Qu et al. designed an albumin-based nanoplatform responsive to ultrasonic stimulation. This strategy eradicated Fusobacterium nucleatum and promoted cancer cell apoptosis by increasing ROS levels.128
In addition, nanomedicines that mimic bacteria have been used to target intratumoral bacteria specifically, preserving the intestinal microbiota. Chen et al. created a nanomedicine mimicking Fusobacterium nucleatum by combining its cytoplasmic membranes with liposomes containing polymyxins. This treatment has shown promise in restoring sensitivity to ICB therapy in tumors colonized by Fusobacterium nucleatum.129
5.2.2 Co-delivery of antitumor drugs and antimicrobial agents.
Some chemotherapy drugs, known to induce immunogenic cell death (ICD), can enhance the efficacy of tumor immunotherapy.130 Observations indicate that the microbiota may contribute to resistance to chemotherapeutic drugs.131–133 Specifically, Gammaproteobacteria residing in tumors can convert the chemotherapeutic drug Gem into an inactive form, reducing chemotherapy efficacy. Combining chemotherapy with immunotherapy to eliminate intratumoral microbiota may enhance the efficacy of tumor treatment. Nano-drug delivery systems hold promise for targeting lesion sites and achieving intelligent, sustained drug release. Therefore, integrating antimicrobial agents and chemotherapeutic drugs into nano-drug delivery systems could target the elimination of intratumoral microbiota and overcome drug resistance, enhancing the efficacy of antitumor therapy.
Eliminating intratumoral microbiota can increase drug delivery efficacy and, subsequently, the release of antitumor drugs, achieving a synergistic antitumor effect by dual targeting of intratumoral microbiota and tumors. Wang et al. designed a dual-cascade responsive nanoparticle (sNP@G/IR) containing Gem and a photothermal agent (IR1048) to enhance antitumor efficacy, with the polymer core serving an antimicrobial function. In addition, the hyperthermic effect of IR1048 aids in further eliminating tumors and bacteria.134 Ma et al. designed metronidazole–fluorouridine nanoparticles (MTI-FDU) to achieve a synergistic antitumor effect. The nanoparticles metronidazole target intratumoral bacteria with minimal disruption to gut microbial homeostasis (Fig. 3D).135 Li et al. developed size-tunable nanogels that integrate zinc-imidazolate frameworks with encapsulated doxorubicin (DOX) and folate grafting (f-ZIFD), combined with metronidazole. The sequential release of f-ZIFD nanoparticles from NGs promotes effective tumor penetration and precise tumor cell targeting, while the acidic-triggered intracellular release of doxorubicin enhances the antitumor effect.136
5.3 Intratumoral microbiota as an anticancer therapeutic agent
In the past decades, engineered strains such as Salmonella and Clostridium have effectively slowed tumor growth and metastasis,137 enhancing survival in preclinical models and clinical cases. Bacterial-based therapies, often used as drug carriers, benefit from nanotechnology,138 synthetic bioengineering,139 and genetic engineering140 to attenuate bacteria and enhance drug efficacy in oncology treatments. Recently, intratumoral bacteria have emerged as promising natural anticancer therapeutic agents. Miyako et al. isolated three types of intratumoral bacteria, namely Rhodopseudomonas palustris, Proteus mirabilis, and a complex bacterium of these two from CRC tissue. These isolated bacteria possess inherent biocompatibility and potent immunogenic anticancer efficacies. They selectively grow and proliferate within the tumor environment, effectively prompting immune cells to infiltrate and eliciting robust anticancer responses in mice.141Staphylococcus epidermidis, producing 6-N-hydroxyaminopurine, inhibits skin tumor growth.142 Modifying these bacteria to enhance their tumor-targeting capabilities represents a promising new research direction.
6. Conclusions
Recent studies highlight the significant role of intratumoral microbiota in influencing tumor therapy outcomes. The microbiota residing within tumors can alter the tumor microenvironment, potentially impacting therapeutic efficacy. Here, we summarize the characteristics and interactions of intratumoral microbiota with various cancers, emphasizing their composition and the critical roles they play. Intratumoral microbiota consist of diverse microbial communities, which can differ markedly between tumor types and even between patients with the same cancer. For example, certain bacterial species may be more prevalent in specific tumors, influencing tumor behavior and patient response to treatment. Research has shown that these microbes can modulate immune responses, affect drug metabolism, and even alter the tumor microenvironment, potentially enhancing or inhibiting therapeutic effects.
The therapeutic applications of intratumoral microbiota are emerging as a promising area of research. Potential strategies include: (1) microbiota-based therapies: modulating the intratumoral microbiota through probiotics or targeted therapies to enhance immune responses; (2) biomarker development: identifying specific microbial signatures that correlate with treatment responses could help tailor therapies to individual patients; and (3) combination therapies: integrating microbiota modulation with existing cancer therapies, such as immunotherapy or chemotherapy, may enhance treatment efficacy. However, it is crucial to maximize the benefits of intratumoral microbiota while minimizing potential adverse effects. This requires a thorough understanding of how these microbial communities interact with cancer therapies.
Several limitations hinder the advancement of research on intratumoral microbiota. First, the low abundance of intratumoral microbiota presents challenges in obtaining adequate samples from tumor tissues. This scarcity can lead to difficulties in accurately assessing microbial diversity and composition. Secondly, current sequencing methods, such as 16S rRNA gene sequencing, primarily provide relative abundance data. This can lead to misunderstandings regarding microbial community structures and interactions. Advanced techniques like metagenomic sequencing are needed for more comprehensive insights. Additionally, enhancing the detection rate of intratumoral bacteria presents a significant challenge, as the inability to accurately identify bacterial components undermines the comprehensive assessment of microbial diversity and composition while exacerbating contamination artifacts. To address this issue, the use of microbial DNA pre-amplification through multiple displacement amplification (MDA) lowers the detection threshold to an abundance of 0.1%. Simultaneously, the application of MolYsis™ reagent kits facilitates the selective lysis of eukaryotic cells while preserving intact microbial cells. Complementary techniques, such as laser microdissection (LMD), enable precise isolation of tumor parenchymal regions, thereby reducing stromal contamination and ensuring data fidelity in studies of microbe–host interactions. Finally, establishing suitable cell and small animal models for studying intratumoral microbiota is essential for basic and preclinical research. The advancement of patient-derived organoid–bacteria co-culture systems offers a promising approach for addressing this requirement. The inherent heterogeneity of tumors and their associated microbiota complicates model development, requiring innovative approaches for replicating tumor conditions accurately.
Despite existing preclinical evidence linking intratumoral microbial modulation with immunotherapy outcomes, further research is necessary to validate these findings in clinical settings. To enhance our understanding, future investigations should focus on exploring the specific mechanisms through which intratumoral microbiota influence tumor progression, metastasis, and therapeutic responses and utilizing multi-omics technologies and refined reference databases to ensure comprehensive and accurate analyses of intratumoral microbiota. By integrating insights from microbiota research with conventional cancer therapies, we may pave the way for more effective and personalized cancer treatment strategies.
Author contributions
Huiling Liu: writing – original draft and review & editing. Zhonghui Luo: writing – original draft and review & editing. Fangzhen Luo: writing – original draft and review & editing. Xilian Wang: writing – original draft and review & editing. Hua Wei: writing – original draft and review & editing. Cui-Yun Yu: writing – original draft and review & editing.
Conflicts of interest
All authors confirmed that they have no conflicts of interest.
Data availability
No primary research results, software or code have been included and no new data were generated or analysed as part of this review (PM-REV-02-2025-000045.R1).
Acknowledgements
This work was financially supported by the Hunan Science and Technology Innovation Leading Talent Project (2022RC3080), the National Natural Science Foundation of China (82373826 and 82202557), the Key R&D Program of Hunan Province (2021SK2036 and 2023SK2043), the Hunan Provincial Natural Science Foundation (2023JJ50138, 2023JJ50140, and 2022JJ40359), and the Health Research Project of the Hunan Provincial Health Commission (B202313020111 and W20243061).
Declaration of generative AI and AI-assisted technologies in the writing process: during the preparation of this work, the authors used ChatGPT in order to improve the grammatical structure in some paragraphs; all the research cited came from PubMed. The authors reviewed all the references cited to ensure the accurate representation of the original research findings. After using ChatGPT, the authors reviewed and edited the content as needed and take full responsibility for the content of the publication.
References
- D. Crosby, S. Bhatia, K. M. Brindle, L. M. Coussens, C. Dive and M. Emberton,
et al., Early detection of cancer, Science, 2022, 375(6586), eaay9040 CrossRef CAS.
- N. Borcherding and J. R. Brestoff, The power and potential of mitochondria transfer, Nature, 2023, 623(7986), 283–291 CrossRef CAS.
- L. Wen, W. Mu, H. Lu, X. Wang, J. Fang and Y. Jia,
et al., Porphyromonas gingivalis Promotes Oral Squamous Cell Carcinoma Progression in an Immune Microenvironment, J. Dent. Res., 2020, 99(6), 666–675 CrossRef CAS.
- A. Taglialegna, When Helicobacter pylori spells gastric cancer, Nat. Rev. Microbiol., 2023, 21(10), 628 CrossRef CAS.
- D. Nejman, I. Livyatan, G. Fuks, N. Gavert, Y. Zwang and L. T. Geller,
et al., The human tumor microbiome is composed of tumor type-specific intracellular bacteria, Science, 2020, 368(6494), 973–980 CrossRef CAS.
- J. L. Galeano Niño, H. Wu, K. D. LaCourse, A. G. Kempchinsky, A. Baryiames and B. Barber,
et al., Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer, Nature, 2022, 611(7937), 810–817 CrossRef.
- L. Narunsky-Haziza, G. D. Sepich-Poore, I. Livyatan, O. Asraf, C. Martino and D. Nejman,
et al., Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions, Cell, 2022, 185(20), 3789–806 CrossRef CAS.
- Y. Ma, H. Chen, H. Li, M. Zheng, X. Zuo and W. Wang,
et al., Intratumor microbiome-derived butyrate promotes lung cancer metastasis, Cell Rep. Med., 2024, 5(4), 101488 CrossRef CAS.
- T. W. Battaglia, I. L. Mimpen, J. J. H. Traets, A. van Hoeck, L. J. Zeverijn and B. S. Geurts,
et al., A pan-cancer analysis of the microbiome in metastatic cancer, Cell, 2024, 187(9), 2324–35 CrossRef CAS PubMed.
- M. J. Bender, A. C. McPherson, C. M. Phelps, S. P. Pandey, C. R. Laughlin and J. H. Shapira,
et al., Dietary tryptophan metabolite released by intratumoral Lactobacillus reuteri facilitates immune checkpoint inhibitor treatment, Cell, 2023, 186(9), 1846–62 CrossRef CAS PubMed.
- A. P. Thrift, Global burden and epidemiology of Barrett oesophagus and oesophageal cancer, Nat. Rev. Gastroenterol. Hepatol., 2021, 18(6), 432–443 CrossRef PubMed.
- K. L. Greathouse, J. K. Stone, A. J. Vargas, A. Choudhury, R. N. Padgett and J. R. White,
et al., Co-enrichment of cancer-associated bacterial taxa is correlated with immune cell infiltrates in esophageal tumor tissue, Sci. Rep., 2024, 14(1), 2574 CrossRef CAS.
- H. Wu, X. F. Leng, Q. S. Liu, T. Q. Mao, T. Jiang and Y. Q. Liu,
et al., Intratumoral Microbiota Composition Regulates Chemoimmunotherapy Response in Esophageal Squamous Cell Carcinoma, Cancer Res., 2023, 83(18), 3131–3144 CrossRef CAS.
- M. Liang, Y. Liu, Z. Zhang, H. Yang, N. Dai and N. Zhang,
et al., Fusobacterium nucleatum induces MDSCs enrichment via activation the NLRP3 inflammosome in ESCC cells, leading to cisplatin resistance, Ann. Med., 2022, 54(1), 989–1003 CrossRef CAS PubMed.
- D. Nomoto, Y. Baba, Y. Liu, H. Tsutsuki, K. Okadome and K. Harada,
et al., Fusobacterium nucleatum promotes esophageal squamous cell carcinoma progression via the NOD1/RIPK2/NF-κB pathway, Cancer Lett., 2022, 530, 59–67 CrossRef CAS.
- S. Guo, F. Chen, L. Li, S. Dou, Q. Li and Y. Huang,
et al., Intracellular Fusobacterium nucleatum infection increases METTL3-mediated m6A methylation to promote the metastasis of esophageal squamous cell carcinoma, J. Adv. Res., 2024, 61, 165–178 CrossRef CAS PubMed.
- J.-W. Zhang, D. Zhang, H.-S. Yin, H. Zhang, K.-Q. Hong and J.-P. Yuan,
et al., Fusobacterium nucleatum promotes esophageal squamous cell carcinoma progression and chemoresistance by enhancing the secretion of chemotherapy-induced senescence-associated secretory phenotype via activation of DNA damage response pathway, Gut Microbes, 2023, 15(1), 2197836 CrossRef PubMed.
- Y. Li, S. Xing, F. Chen, Q. Li, S. Dou and Y. Huang,
et al., Intracellular Fusobacterium nucleatum infection attenuates antitumor immunity in esophageal squamous cell carcinoma, Nat. Commun., 2023, 14(1), 5788 CrossRef CAS.
- J. G. Navashenaq, A. G. Shabgah, M. Banach, T. Jamialahmadi, P. E. Penson and T. P. Johnston,
et al., The interaction of Helicobacter pylori with cancer immunomodulatory stromal cells: New insight into gastric cancer pathogenesis, Semin. Cancer Biol., 2022, 86, 951–959 CrossRef CAS.
- K. Fu, A. H. K. Cheung, C. C. Wong, W. Liu, Y. Zhou and F. Wang,
et al., Streptococcus anginosus promotes gastric inflammation, atrophy, and tumorigenesis in mice, Cell, 2024, 187(4), 882–96 CrossRef CAS PubMed.
- L. Yuan, L. Pan, Y. Wang, J. Zhao, L. Fang and Y. Zhou,
et al., Characterization of the landscape of the intratumoral microbiota reveals that Streptococcus anginosus increases the risk of gastric cancer initiation and progression, Cell Discovery, 2024, 10(1), 117 CrossRef CAS PubMed.
- N. Murata-Kamiya and M. Hatakeyama, Helicobacter pylori-induced DNA double-stranded break in the development of gastric cancer, Cancer Sci., 2022, 113(6), 1909–1918 CrossRef CAS PubMed.
- X. Yong, B. Tang, Y.-F. Xiao, R. Xie, Y. Qin and G. Luo,
et al., Helicobacter pylori upregulates Nanog and Oct4 via Wnt/β-catenin signaling pathway to promote cancer stem cell-like properties in human gastric cancer, Cancer Lett., 2016, 374(2), 292–303 CrossRef CAS.
- M. C. C. Lim, P. Jantaree and M. Naumann, The conundrum of Helicobacter pylori-associated apoptosis in gastric cancer, Trends Cancer, 2023, 9(8), 679–690 CrossRef CAS.
- S. Imai, T. Ooki, N. Murata-Kamiya, D. Komura, K. Tahmina and W. Wu,
et al., Helicobacter pylori CagA elicits BRCAness to induce genome instability that may underlie bacterial gastric carcinogenesis, Cell Host Microbe, 2021, 29(6), 941–58 CrossRef CAS.
- S. Salvatori, I. Marafini, F. Laudisi, G. Monteleone and C. Stolfi, Helicobacter pylori and Gastric Cancer: Pathogenetic Mechanisms, Int. J. Mol. Sci., 2023, 24(3), 2895 CrossRef CAS.
- E. M. Park, M. Chelvanambi, N. Bhutiani, G. Kroemer, L. Zitvogel and J. A. Wargo, Targeting the gut and tumor microbiota in cancer, Nat. Med., 2022, 28(4), 690–703 CrossRef CAS PubMed.
- K. Arima, R. Zhong, T. Ugai, M. Zhao, K. Haruki and N. Akimoto,
et al., Western-Style Diet, pks Island-Carrying Escherichia coli, and Colorectal Cancer: Analyses From Two Large Prospective Cohort Studies, Gastroenterology, 2022, 163(4), 862–874 CrossRef CAS.
- C. Pleguezuelos-Manzano, J. Puschhof, A. Rosendahl Huber, A. van Hoeck, H. M. Wood and J. Nomburg,
et al., Mutational signature in colorectal cancer caused by genotoxic pks+ E. coli, Nature, 2020, 580(7802), 269–273 CrossRef CAS.
- B. Chen, D. Ramazzotti, T. Heide, I. Spiteri, J. Fernandez-Mateos and C. James,
et al., Contribution of pks+ E. coli mutations to colorectal carcinogenesis, Nat. Commun., 2023, 14(1), 7827 CrossRef CAS PubMed.
- M. R. Wilson, Y. Jiang, P. W. Villalta, A. Stornetta, P. D. Boudreau and A. Carrá,
et al., The human gut bacterial genotoxin colibactin alkylates DNA, Science, 2019, 363(6428), eaar7785 CrossRef CAS PubMed.
- Z. He, R. Z. Gharaibeh, R. C. Newsome, J. L. Pope, M. W. Dougherty and S. Tomkovich,
et al., Campylobacter jejuni promotes colorectal tumorigenesis through the action of cytolethal distending toxin, Gut, 2019, 68(2), 289–300 CrossRef CAS.
- L. R. Lopez, R. M. Bleich and J. C. Arthur, Microbiota Effects on Carcinogenesis: Initiation, Promotion, and Progression, Annu. Rev. Med., 2021, 72(1), 243–261 Search PubMed.
- M. Zepeda-Rivera, S. S. Minot, H. Bouzek, H. Wu, A. Blanco-Míguez and P. Manghi,
et al., A distinct Fusobacterium nucleatum clade dominates the colorectal cancer niche, Nature, 2024, 628(8007), 424–432 CrossRef CAS.
- C. Kong, X. Yan, Y. Zhu, H. Zhu, Y. Luo and P. Liu,
et al., Fusobacterium Nucleatum Promotes the Development of Colorectal Cancer by Activating a Cytochrome P450/Epoxyoctadecenoic Acid Axis via TLR4/Keap1/NRF2 Signaling, Cancer Res., 2021, 81(17), 4485–4498 CrossRef CAS PubMed.
- W. Cui, M. Guo, D. Liu, P. Xiao, C. Yang and H. Huang,
et al., Gut microbial metabolite facilitates colorectal cancer development via ferroptosis inhibition, Nat. Cell Biol., 2024, 26(1), 124–137 CrossRef CAS PubMed.
- M. Feng, Y. Pan, R. Kong and S. Shu, Therapy of Primary Liver Cancer, Innovation, 2020, 1(2), 100032 CAS.
- J. M. Llovet, R. K. Kelley, A. Villanueva, A. G. Singal, E. Pikarsky and S. Roayaie,
et al., Hepatocellular carcinoma, Nat. Rev. Dis. Primers, 2021, 7(1), 6 CrossRef PubMed.
- S. Komiyama, T. Yamada, N. Takemura, N. Kokudo, K. Hase and Y. I. Kawamura, Profiling of tumour-associated microbiota in human hepatocellular carcinoma, Sci. Rep., 2021, 11(1), 10589 CrossRef CAS PubMed.
- X. Chai, J. Wang, H. Li, C. Gao, S. Li and C. Wei,
et al., Intratumor microbiome features reveal antitumor potentials of intrahepatic cholangiocarcinoma, Gut Microbes, 2022, 15(1), 2156255 CrossRef PubMed.
- Y. Feng, Z. Han, H. Zhou, W. Wang, Y. Wang and T. Sun,
et al. The multifaceted role of microbiota in liver cancer: pathogenesis, therapy, prognosis, and immunotherapy, Front Immunol., 2025, 16, 1575963 Search PubMed.
- B. Liu, Z. Zhou, Y. Jin, J. Lu, D. Feng and R. Peng,
et al., Hepatic stellate cell activation and senescence induced by intrahepatic microbiota disturbances drive progression of liver cirrhosis toward hepatocellular carcinoma, J. Immunotherap. Cancer, 2022, 10(1), e003069 CrossRef PubMed.
- L. Sun, X. Ke, A. Guan, B. Jin, J. Qu and Y. Wang,
et al., Intratumoural microbiome can predict the prognosis of hepatocellular carcinoma after surgery, Clin. Transl. Med., 2023, 13(7), e1331 CrossRef PubMed.
- C. Xue, J. Jia, X. Gu, L. Zhou, J. Lu and Q. Zheng,
et al., Intratumoral bacteria interact with metabolites and genetic alterations in hepatocellular carcinoma, Signal Transduction Targeted Ther., 2022, 7(1), 335 CrossRef.
- L. T. Geller, M. Barzily-Rokni, T. Danino, O. H. Jonas, N. Shental and D. Nejman,
et al., Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine, Science, 2017, 357(6356), 1156–1160 CrossRef CAS.
- S. Pushalkar, M. Hundeyin, D. Daley, C. P. Zambirinis, E. Kurz and A. Mishra,
et al., The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression, Cancer Discovery, 2018, 8(4), 403–416 CrossRef CAS.
- K. Hezaveh, R. S. Shinde, A. Klötgen, M. J. Halaby, S. Lamorte and M. T. Ciudad,
et al., Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity, Immunity, 2022, 55(2), 324–40 CrossRef CAS PubMed.
- X. Yang, Z. Zhang, X. Shen, J. Xu, Y. Weng and W. Wang,
et al., Clostridium butyricum and its metabolite butyrate promote ferroptosis susceptibility in pancreatic ductal adenocarcinoma, Cell. Oncol., 2023, 46(6), 1645–1658 CrossRef CAS.
- M. Stasiewicz and T. M. Karpiński, The oral microbiota and its role in carcinogenesis, Semin. Cancer Biol., 2022, 86, 633–642 CrossRef CAS PubMed.
- Q. Tan, X. Ma, B. Yang, Y. Liu, Y. Xie and X. Wang,
et al., Periodontitis pathogen Porphyromonas gingivalis promotes pancreatic tumorigenesis via neutrophil elastase from tumor-associated neutrophils, Gut Microbes, 2022, 14(1), 2073785 CrossRef.
- E. Saba, M. Farhat, A. Daoud, A. Khashan, E. Forkush and N. H. Menahem,
et al., Oral bacteria accelerate pancreatic cancer development in mice, Gut, 2024, 73(5), 770–786 CrossRef CAS PubMed.
- A. Alam, E. Levanduski, P. Denz, H. S. Villavicencio, M. Bhatta and L. Alhorebi,
et al., Fungal mycobiome drives IL-33 secretion and type 2 immunity in pancreatic cancer, Cancer Cell, 2022, 40(2), 153–67 CrossRef CAS PubMed.
- B. Aykut, S. Pushalkar, R. Chen, Q. Li, R. Abengozar and J. I. Kim,
et al., The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL, Nature, 2019, 574(7777), 264–267 CrossRef CAS.
- E. Riquelme, Y. Zhang, L. Zhang, M. Montiel, M. Zoltan and W. Dong,
et al., Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes, Cell, 2019, 178(4), 795–806 Search PubMed.
- B. Ghaddar, A. Biswas, C. Harris, M. B. Omary, D. R. Carpizo and M. J. Blaser,
et al., Tumor microbiome links cellular programs and immunity in pancreatic cancer, Cancer Cell, 2022, 40(10), 1240–53 Search PubMed.
- A. A. Thai, B. J. Solomon, L. V. Sequist, J. F. Gainor and R. S. Heist, Lung cancer, Lancet, 2021, 398(10299), 535–554 Search PubMed.
- T. Goto, Microbiota and lung cancer, Semin. Cancer Biol., 2022, 86, 1–10 CrossRef PubMed.
- C. Jin, G. K. Lagoudas, C. Zhao, S. Bullman, A. Bhutkar and B. Hu,
et al., Commensal Microbiota Promote Lung Cancer Development via γδ T Cells, Cell, 2019, 176(5), 998–1013 CrossRef CAS PubMed.
- V. Le Noci, S. Guglielmetti, S. Arioli, C. Camisaschi, F. Bianchi and M. Sommariva,
et al., Modulation of Pulmonary Microbiota by Antibiotic or Probiotic Aerosol Therapy: A Strategy to Promote Immunosurveillance against Lung Metastases, Cell Rep., 2018, 24(13), 3528–3538 Search PubMed.
- M. Zagorulya, L. Yim, D. M. Morgan, A. Edwards, E. Torres-Mejia and N. Momin,
et al., Tissue-specific abundance of interferon-gamma drives regulatory T cells to restrain DC1-mediated priming of cytotoxic T cells against lung cancer, Immunity, 2023, 56(2), 386–405 CrossRef CAS PubMed.
- M. J. Watson, P. D. A. Vignali, S. J. Mullett, A. E. Overacre-Delgoffe, R. M. Peralta and S. Grebinoski,
et al., Metabolic support of tumour-infiltrating regulatory T cells by lactic acid, Nature, 2021, 591(7851), 645–651 CrossRef CAS PubMed.
- N.-N. Liu, C.-X. Yi, L.-Q. Wei, J.-A. Zhou, T. Jiang and C.-C. Hu,
et al., The intratumor mycobiome promotes lung cancer progression via myeloid-derived suppressor cells, Cancer Cell, 2023, 41(11), 1927–44 Search PubMed.
- Y. Ma, H. Chen, H. Li, M. Zheng, X. Zuo and W. Wang,
et al., Intratumor microbiome-derived butyrate promotes lung cancer metastasis, Cell Rep. Med., 2024, 5(4), 101488 CrossRef CAS PubMed.
- S. Jin, R. Li, M.-Y. Chen, C. Yu, L.-Q. Tang and Y.-M. Liu,
et al., Single-cell transcriptomic analysis defines the interplay between tumor cells, viral infection, and the microenvironment in nasopharyngeal carcinoma, Cell Res., 2020, 30(11), 950–965 CrossRef CAS PubMed.
- J. He, L. Liu, F. Tang, Y. Zhou, H. Liu and C. Lu,
et al., Paradoxical effects of DNA tumor virus oncogenes on epithelium-derived tumor cell fate during tumor progression and chemotherapy response, Signal Transduction Targeted Ther., 2021, 6(1), 408 CrossRef CAS PubMed.
- H. Qiao, X.-R. Tan, H. Li, J.-Y. Li, X.-Z. Chen and Y.-Q. Li,
et al., Association of Intratumoral Microbiota With Prognosis in Patients With Nasopharyngeal Carcinoma From 2 Hospitals in China, JAMA Oncol., 2022, 8(9), 1301–1309 CrossRef.
- Y. Liao, Y.-X. Wu, M. Tang, Y.-W. Chen, J.-R. Xie and Y. Du,
et al., Microbes translocation from oral cavity to nasopharyngeal carcinoma in patients, Nat. Commun., 2024, 15(1), 1645 CrossRef CAS PubMed.
- S. Koster, R. K. Gurumurthy, N. Kumar, P. G. Prakash, J. Dhanraj and S. Bayer,
et al., Modelling Chlamydia and HPV co-infection in patient-derived ectocervix organoids reveals distinct cellular reprogramming, Nat. Commun., 2022, 13(1), 1030 CrossRef CAS PubMed.
- V. P. Nikitina, T. A. Zykova, E. A. Shevyakova, O. E. Zhenilo, E. V. Verenikina and V. A. Ivanova,
et al., Vaginal biocenosis in patients with gynecological cancers, J. Clin. Oncol., 2021, 39(15_suppl), e17574 Search PubMed.
- M. Kyrgiou and A.-B. Moscicki, Vaginal microbiome and cervical cancer, Semin. Cancer Biol., 2022, 86, 189–198 CrossRef CAS PubMed.
- N. Zheng, R. Guo, J. Wang, W. Zhou and Z. Ling, Contribution of Lactobacillus iners to Vaginal Health and Diseases: A Systematic Review, Front. Cell. Infect. Microbiol., 2021, 11, 792787 CrossRef CAS PubMed.
- L. E. Colbert, M. B. El Alam, R. Wang, T. Karpinets, D. Lo and E. J. Lynn,
et al., Tumor-resident Lactobacillus iners confer chemoradiation resistance through lactate-induced metabolic rewiring, Cancer Cell, 2023, 41(11), 1945–62 Search PubMed.
- L. Jiang, B. Duan, P. Jia, Y. Zhang and X. Yan, The Role of Intratumor Microbiomes in Cervical Cancer Metastasis, Cancers, 2023, 15(2), 509 CrossRef CAS PubMed.
- I. Rastogi, A. Muralidhar and D. G. McNeel, Vaccines as treatments for prostate cancer, Nat. Rev. Urol., 2023, 20(9), 544–559 CrossRef CAS PubMed.
- K. S. Sfanos, S. Yegnasubramanian, W. G. Nelson and A. M. De Marzo, The inflammatory microenvironment and microbiome in prostate cancer development, Nat. Rev. Urol., 2018, 15(1), 11–24 CrossRef PubMed.
- S. Radej, M. Szewc and R. Maciejewski, Prostate Infiltration by Treg and Th17 Cells as an Immune Response to Propionibacterium acnes Infection in the Course of Benign Prostatic Hyperplasia and Prostate Cancer, Int. J. Mol. Sci., 2022, 23(16), 8849 CrossRef CAS PubMed.
- J. Ma, A. Gnanasekar, A. Lee, W. T. Li, M. Haas and J. Wang-Rodriguez,
et al., Influence of Intratumor Microbiome on Clinical Outcome and Immune Processes in Prostate Cancer, Cancers, 2020, 12(9), 2524 CrossRef CAS PubMed.
- A. Fu, B. Yao, T. Dong, Y. Chen, J. Yao and Y. Liu,
et al., Tumor-resident intracellular microbiota promotes metastatic colonization in breast cancer, Cell, 2022, 185(8), 1356–72 Search PubMed.
- S. Banerjee, Z. Wei, T. Tian, D. Bose, N. N. C. Shih and M. D. Feldman,
et al., Prognostic correlations with the microbiome of breast cancer subtypes, Cell Death Dis., 2021, 12(9), 831 CrossRef PubMed.
- T. Kovács, E. Mikó, A. Vida, É. Sebő, J. Toth and T. Csonka,
et al., Cadaverine, a metabolite of the microbiome, reduces breast cancer aggressiveness through trace amino acid receptors, Sci. Rep., 2019, 9(1), 1300 CrossRef.
- W. Tang, V. Putluri, C. R. Ambati, T. H. Dorsey, N. Putluri and S. Ambs, Liver- and Microbiome-derived Bile Acids Accumulate in Human Breast Tumors and Inhibit Growth and Improve Patient Survival, Clin. Cancer Res., 2019, 25(19), 5972–5983 CrossRef CAS PubMed.
- H. Wang, X. Rong, G. Zhao, Y. Zhou, Y. Xiao and D. Ma,
et al., The microbial metabolite trimethylamine N-oxide promotes antitumor immunity in triple-negative breast cancer, Cell Metab., 2022, 34(4), 581–94 CrossRef CAS PubMed.
- G. V. Long, S. M. Swetter, A. M. Menzies, J. E. Gershenwald and R. A. Scolyer, Cutaneous melanoma, Lancet, 2023, 402(10400), 485–502 Search PubMed.
- B. Routy, T. Jackson, L. Mählmann, C. K. Baumgartner, M. Blaser and A. Byrd,
et al., Melanoma and microbiota: Current understanding and future directions, Cancer Cell, 2024, 42(1), 16–34 Search PubMed.
- J. R. Björk, L. A. Bolte, A. Maltez Thomas, K. A. Lee, N. Rossi and T. T. Wind,
et al., Longitudinal gut microbiome changes in immune checkpoint blockade-treated advanced melanoma, Nat. Med., 2024, 30(3), 785–796 Search PubMed.
- A. Wong-Rolle, H. K. Wei, C. Zhao and C. Jin, Unexpected guests in the tumor microenvironment: microbiome in cancer, Protein Cell, 2020, 12(5), 426–435 Search PubMed.
- A. York, Tumour-specific microbiomes, Nat. Rev. Microbiol., 2020, 18(8), 413 Search PubMed.
- M. A. D. Silveira, S. Bilodeau, T. F. Greten, X. W. Wang and G. Trinchieri, The gut–liver axis: host microbiota interactions shape hepatocarcinogenesis, Trends Cancer, 2022, 8(7), 583–597 CrossRef CAS PubMed.
- X. Zhang, D. Yu, D. Wu, X. Gao, F. Shao and M. Zhao,
et al., Tissue-resident Lachnospiraceae family bacteria protect against colorectal carcinogenesis by promoting tumor immune surveillance, Cell Host Microbe, 2023, 31(3), 418–32 CrossRef CAS PubMed.
- X. Wei, Y. Chen, X. Jiang, M. Peng, Y. Liu and Y. Mo,
et al., Mechanisms of vasculogenic mimicry in hypoxic tumor microenvironments, Mol. Cancer, 2021, 20(1), 7 CrossRef.
- J. Pouysségur, I. Marchiq, S. K. Parks, J. Durivault, M. Ždralević and M. Vucetic, ‘Warburg effect’ controls tumor growth, bacterial, viral infections and immunity – Genetic deconstruction and therapeutic perspectives, Semin. Cancer Biol., 2022, 86, 334–346 CrossRef.
- P. Zhou, Y. Hu, X. Wang, L. Shen, X. Liao and Y. Zhu,
et al., Microbiome in cancer: An exploration of carcinogenesis, immune responses and immunotherapy, Front. Immunol., 2022, 13, 877939 CrossRef CAS.
- A. E. Overacre-Delgoffe, H. J. Bumgarner, A. R. Cillo, A. H. P. Burr, J. T. Tometich and A. Bhattacharjee,
et al., Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer, Immunity, 2021, 54(12), 2812–24 Search PubMed.
- R. Cabrita, M. Lauss, A. Sanna, M. Donia, M. Skaarup Larsen and S. Mitra,
et al., Tertiary lymphoid structures improve immunotherapy and survival in melanoma, Nature, 2020, 577(7791), 561–565 Search PubMed.
- L. L. Cao and J. C. Kagan, Targeting innate immune pathways for cancer immunotherapy, Immunity, 2023, 56(10), 2206–2217 CrossRef CAS PubMed.
- M. Yi, T. Li, M. Niu, Q. Mei, B. Zhao and Q. Chu,
et al., Exploiting innate immunity for cancer immunotherapy, Mol. Cancer, 2023, 22(1), 187 CrossRef CAS PubMed.
- D. Lee, K. Huntoon, Y. Wang, W. Jiang and B. Y. S. Kim, Harnessing Innate Immunity Using Biomaterials for Cancer Immunotherapy, Adv. Mater., 2021, 33(27), 2007576 CrossRef CAS PubMed.
- Y. Gao, D. Bi, R. Xie, M. Li, J. Guo and H. Liu,
et al., Correction To: Fusobacterium nucleatum enhances the efficacy of PD-L1 blockade in colorectal cancer, Signal Transduction Targeted Ther., 2021, 6(1), 434 CrossRef.
- X. Liu, M. Sun, F. Pu, J. Ren and X. Qu, Transforming Intratumor Bacteria into Immunopotentiators to Reverse Cold Tumors for Enhanced Immuno-chemodynamic Therapy of Triple-Negative Breast Cancer, J. Am. Chem. Soc., 2023, 145(48), 26296–26307 CrossRef CAS PubMed.
- K. Jia, Y. Chen, Y. Xie, X. Wang, Y. Hu and Y. Sun,
et al., Helicobacter pylori and immunotherapy for gastrointestinal cancer, Innovation, 2024, 5(2), 100561 CAS.
- Y. Shi, W. Zheng, K. Yang, K. G. Harris, K. Ni and L. Xue,
et al., Intratumoral accumulation of gut microbiota facilitates CD47-based immunotherapy via STING signaling, J. Exp. Med., 2020, 217(5), e20192282 CrossRef CAS.
- Z. Zhang, Q. Gao, X. Ren, M. Luo, Y. Liu and P. Liu,
et al., Characterization of intratumor microbiome in cancer immunotherapy, Innovation, 2023, 4(5), 100482 Search PubMed.
- E. E. Elias, B. Lyons and D. A. Muruve, Gasdermins and pyroptosis in the kidney, Nat. Rev. Nephrol., 2023, 19(5), 337–350 CrossRef.
- H. Wang, X. Zhou, C. Li, S. Yan, C. Feng and J. He,
et al., The emerging role of pyroptosis in pediatric cancers: from mechanism to therapy, J. Hematol. Oncol., 2022, 15(1), 140 CrossRef.
- Y. Liu, Y. Lu, B. Ning, X. Su, B. Yang and H. Dong,
et al., Intravenous Delivery of Living Listeria monocytogenes Elicits Gasdmermin-Dependent Tumor Pyroptosis and Motivates Anti-Tumor Immune Response, ACS Nano, 2022, 16(3), 4102–4115 CrossRef CAS.
- X. Lou, J. Wang, X. Jin, X. Wang, B. Qin and D. Liu,
et al., An oral bacterial pyroptosis amplifier against malignant colon cancer, Nano Today, 2024, 54, 102091 CrossRef CAS.
- S. Kalaora, A. Nagler, D. Nejman, M. Alon, C. Barbolin and E. Barnea,
et al., Identification of bacteria-derived HLA-bound peptides in melanoma, Nature, 2021, 592(7852), 138–143 CrossRef CAS.
- R. Naghavian, W. Faigle, P. Oldrati, J. Wang, N. C. Toussaint and Y. Qiu,
et al., Microbial peptides activate tumour-infiltrating lymphocytes in glioblastoma, Nature, 2023, 617(7962), 807–817 CrossRef CAS.
- S. Klobuch, T. T. P. Seijkens, T. N. Schumacher and J. B. A. G. Haanen, Tumour-infiltrating lymphocyte therapy for patients with advanced-stage melanoma, Nat. Rev. Clin. Oncol., 2024, 21(3), 173–184 CrossRef CAS.
- C. Backlund, S. Jalili-Firoozinezhad, B. Kim and D. J. Irvine, Biomaterials-Mediated Engineering of the Immune System, Annu. Rev. Immunol., 2023, 41(1), 153–179 CrossRef CAS.
- X. Ma, S.-J. Li, Y. Liu, T. Zhang, P. Xue and Y. Kang,
et al., Bioengineered nanogels for cancer immunotherapy, Chem. Soc. Rev., 2022, 51(12), 5136–5174 RSC.
- N. Boehnke, J. P. Straehla, H. C. Safford, M. Kocak, M. G. Rees and M. Ronan,
et al., Massively parallel pooled screening reveals genomic determinants of nanoparticle delivery, Science, 2022,(6604), 377 Search PubMed.
- X. Zhu, J. Xu, G. Ling and P. Zhang, Tunable metal–organic frameworks assist in catalyzing DNAzymes with amplification platforms for biomedical applications, Chem. Soc. Rev., 2023, 52(21), 7549–7578 RSC.
- X. Han, A. Alu, H. Liu, Y. Shi, X. Wei and L. Cai,
et al., Biomaterial-assisted biotherapy: A brief review of biomaterials used in drug delivery, vaccine development, gene therapy, and stem cell therapy, Bioact. Mater., 2022, 17, 29–48 CAS.
- C. Pacheco, A. Baião, T. Ding, W. Cui and B. Sarmento, Recent advances in long-acting drug delivery systems for anticancer drug, Adv. Drug Delivery Rev., 2023, 194, 114724 CrossRef CAS PubMed.
- A. Erfani, A. E. Diaz and P. S. Doyle, Hydrogel-enabled, local administration and combinatorial delivery of immunotherapies for cancer treatment, Mater. Today, 2023, 65, 227–243 CrossRef CAS.
- X. Yan, F. Ma, Q. Chen, X. Gou, X. Li and L. Zhang,
et al., Construction of size-transformable supramolecular nano-platform against drug-resistant colorectal cancer caused by Fusobacterium nucleatum, Chem. Eng. J., 2022, 450, 137605 CrossRef CAS.
- D. Y. Wang, Y. Cao, G. Yang, S. Zhang, H. C. van der Mei and Y. Ren,
et al., Self–Targeted Co–Delivery of an Antibiotic and a Cancer–Chemotherapeutic from Synthetic Liposomes for the Treatment of Infected Tumors, Adv. Funct. Mater., 2023, 33(32), 2215153 CrossRef CAS.
- X. Li, Y. Ma, Y. Xin, F. Ma and H. Gao, Tumor-Targeting Nanoassembly for Enhanced Colorectal Cancer Therapy by Eliminating Intratumoral Fusobacterium nucleatum, ACS Appl. Mater. Interfaces, 2023, 15(11), 14164–14172 CAS.
- W.-F. Song, D. Zheng, S.-M. Zeng, X. Zeng and X.-Z. Zhang, Targeting to Tumor-Harbored Bacteria for Precision Tumor Therapy, ACS Nano, 2022, 16(10), 17402–17413 CrossRef CAS PubMed.
- M. Wang, B. Rousseau, K. Qiu, G. Huang, Y. Zhang and H. Su,
et al., Killing tumor-associated bacteria with a liposomal antibiotic generates neoantigens that induce anti-tumor immune responses, Nat. Biotechnol., 2023, 42(8), 1263–1274 CrossRef PubMed.
- S. Geng, P. Guo, X. Li, Y. Shi, J. Wang and M. Cao,
et al., Biomimetic Nanovehicle-Enabled Targeted Depletion of Intratumoral Fusobacterium nucleatum Synergizes with PD-L1 Blockade against Breast Cancer, ACS Nano, 2024, 18(12), 8971–8987 CrossRef CAS PubMed.
- C. Cao, T. Zhang, N. Yang, X. Niu, Z. Zhou and J. Wang,
et al., POD Nanozyme optimized by charge separation engineering for light/pH activated bacteria catalytic/photodynamic therapy, Signal Transduction Targeted Ther., 2022, 7(1), 86 CrossRef CAS PubMed.
- J. Hou and Y. Xianyu, Tailoring the Surface and Composition of Nanozymes for Enhanced Bacterial Binding and Antibacterial Activity, Small, 2023, 19(42), e2302640 CrossRef PubMed.
- N. Song, Y. Yu, Y. Zhang, Z. Wang, Z. Guo and J. Zhang,
et al., Bioinspired Hierarchical Self-Assembled Nanozyme for Efficient Antibacterial Treatment, Adv. Mater., 2024, 36(10), 2210455 CrossRef CAS.
- X. Wang, Q. Chen, Y. Zhu, K. Wang, Y. Chang and X. Wu,
et al., Destroying pathogen-tumor symbionts synergizing with catalytic therapy of colorectal cancer by biomimetic protein-supported single-atom nanozyme, Signal Transduction Targeted Ther., 2023, 8(1), 277 CrossRef CAS.
- J. Xi, Y. Wang, X. Gao, Y. Huang, J. Chen and Y. Chen,
et al., Reverse intratumor bacteria-induced gemcitabine resistance with carbon nanozymes for enhanced tumor catalytic-chemo therapy, Nano Today, 2022, 43, 101395 CrossRef CAS.
- X. Qu, F. Yin, M. Pei, Q. Chen, Y. Zhang and S. Lu,
et al., Modulation of Intratumoral Fusobacterium nucleatum to Enhance Sonodynamic Therapy for Colorectal Cancer with Reduced Phototoxic Skin Injury, ACS Nano, 2023, 17(12), 11466–11480 CrossRef CAS.
- L. Chen, R. Zhao, J. Shen, N. Liu, Z. Zheng and Y. Miao,
et al., Antibacterial Fusobacterium nucleatum–Mimicking Nanomedicine to Selectively Eliminate Tumor–Colonized Bacteria and Enhance Immunotherapy Against Colorectal Cancer, Adv. Mater., 2023, 35(45), e2306281 CrossRef.
- J. Guo, Y. Zou and L. Huang, Nano Delivery of Chemotherapeutic ICD Inducers for Tumor Immunotherapy, Small Methods, 2023, 7(5), 2201307 CrossRef CAS PubMed.
- G. Dalmasso, A. Cougnoux, T. Faïs, V. Bonnin, B. Mottet-Auselo and H. T. T. Nguyen,
et al., Colibactin-producing Escherichia coli enhance resistance to chemotherapeutic drugs by promoting epithelial to mesenchymal transition and cancer stem cell emergence, Gut Microbes, 2024, 16(1), 2310215 CrossRef.
- S.-S. Jiang, Y.-L. Xie, X.-Y. Xiao, Z.-R. Kang, X.-L. Lin and L. Zhang,
et al., Fusobacterium nucleatum-derived succinic acid induces tumor resistance to immunotherapy in colorectal cancer, Cell Host Microbe, 2023, 31(5), 781–97 CrossRef CAS.
- N. de Oliveira Alves, G. Dalmasso, D. Nikitina, A. Vaysse, R. Ruez and L. Ledoux,
et al., The colibactin-producing Escherichia coli alters the tumor microenvironment to immunosuppressive lipid overload facilitating colorectal cancer progression and chemoresistance, Gut Microbes, 2024, 16(1), 2320291 CrossRef.
- X. Kang, F. Bu, W. Feng, F. Liu, X. Yang and H. Li,
et al., Dual–Cascade Responsive Nanoparticles Enhance Pancreatic Cancer Therapy by Eliminating Tumor–Resident Intracellular Bacteria, Adv. Mater., 2022, 34(49), e2206765 CrossRef.
- C. Gao, X. Wang, B. Yang, W. Yuan, W. Huang and G. Wu,
et al., Synergistic Target of Intratumoral Microbiome and Tumor by Metronidazole–Fluorouridine Nanoparticles, ACS Nano, 2023, 17(8), 7335–7351 CrossRef CAS PubMed.
- S. Xie, L. Wei, Y. Liu, J. Meng, W. Cao and B. Qiu,
et al., Size-tunable nanogels for cascaded release of metronidazole and chemotherapeutic agents to combat Fusobacterium nucleatum-infected colorectal cancer, J. Controlled Release, 2024, 365, 16–28 CrossRef CAS.
- S. Zhou, Y. Lin, Z. Zhao, Y. Lai, M. Lu and Z. Shao,
et al., Targeted deprivation of methionine with engineered Salmonella leads to oncolysis and suppression of metastasis in broad types of animal tumor models, Cell Rep. Med., 2023, 4(6), 101070 CrossRef CAS.
- Z. Chen, Y. Liu, Y. Yu, S. Yang, J. Feng and Y. Zhu,
et al., Micro-to-Nano Oncolytic Microbial System Shifts from Tumor Killing to Tumor Draining Lymph Nodes Remolding for Enhanced Immunotherapy, Adv. Mater., 2024, 36(7), 2306488 CrossRef CAS.
- R. Liu, Z. Cao, L. Wang, X. Wang, S. Lin and F. Wu,
et al., Multimodal oncolytic bacteria by coating with tumor cell derived nanoshells, Nano Today, 2022, 45, 101537 CrossRef CAS.
- Y. Guo, M. Song, X. Liu, Y. Chen, Z. Xun and Y. Sun,
et al., Photodynamic therapy-improved oncolytic bacterial immunotherapy with FAP-encoding S. typhimurium, J. Controlled Release, 2022, 351, 860–871 CrossRef CAS.
- Y. Goto, S. Iwata, M. Miyahara and E. Miyako, Discovery of Intratumoral Oncolytic Bacteria Toward Targeted Anticancer Theranostics, Adv. Sci., 2023, 10(20), e2301679 CrossRef.
- T. Nakatsuji, T. H. Chen, A. M. Butcher, L. L. Trzoss, S.-J. Nam and K. T. Shirakawa,
et al., A commensal strain of Staphylococcus epidermidis protects against skin neoplasia, Sci. Adv., 2018, 4(2), eaao4502 CrossRef.
Footnote |
† These authors contributed equally to this work. |
|
This journal is © The Royal Society of Chemistry 2025 |
Click here to see how this site uses Cookies. View our privacy policy here.