Next Article in Journal
Diagnostic and Therapeutic Management of Mesothelioma of the Tunica Vaginalis Testis: A Population-Based Study in Italy
Previous Article in Journal
T-Cell Engagers in Acute Myeloid Leukemia: Molecular Targets, Structure, and Therapeutic Challenges
Previous Article in Special Issue
Infection Biomarkers in Children with Chemotherapy-Induced Severe Neutropenia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Fusobacterium nucleatum and Its Impact on Colorectal Cancer Chemoresistance: A Meta-Analysis of In Vitro Co-Culture Infections

1
Population Health and Reproduction, 100K Pathogen Genome Project, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
2
School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(19), 3247; https://doi.org/10.3390/cancers17193247
Submission received: 15 August 2025 / Revised: 29 September 2025 / Accepted: 2 October 2025 / Published: 7 October 2025
(This article belongs to the Special Issue Infectious Agents and Cancer in Children and Adolescents)

Abstract

Simple Summary

Bacteria from the mouth are increasingly found in the gut and linked to worse outcomes in colorectal cancer, one of the leading causes of cancer death worldwide. One such bacterium, Fusobacterium nucleatum, has been shown to interfere with cancer treatment, contributing to chemoresistance. This study brings together data from several independent experiments where colorectal cancer cells were grown with Fusobacterium to identify consistent patterns in how the bacteria influence cancer biology. The combined analysis revealed that the bacterium activates immune and antiviral pathways, alters how cells repair DNA, induces metastasis, and may reduce the effectiveness of common cancer drugs. These findings reinforce the role of bacteria in cancer progression and drug resistance, providing a valuable resource for future research into improving treatment outcomes.

Abstract

Introduction: Fusobacterium nucleatum, a common oral microbe associated with periodontal disease, has emerged as a significant prognostic indicator in colorectal cancer (CRC). This organism is notably enriched in CRC tissues and is associated with reduced survival times and relapse. Fusobacterium is implicated in encouraging the development of chemoresistance through diverse tumor-promoting pathways that are increasingly being elucidated across molecular domains. Methods: This work uses a combined analysis of public data examining the role of F. nucleatum in CRC by investigating multiple transcriptomic datasets derived from co-culture infections in vitro. Results: In tandem with previously identified mechanisms known to be influenced by F. nucleatum, this analysis revealed that the bacterium activates multiple chemoresistance-associated pathways, including those driving inflammation, immune evasion, DNA damage, and metastasis. Notably, this study uncovered a novel induction of type I and type II interferon signaling, suggesting activation of a pseudo-antiviral state. Furthermore, pathway analysis (IPA) predicted altered regulation of several therapeutic agents, suggesting that F. nucleatum may compromise drug efficacy through transcriptional reprogramming. Conclusions: These findings reinforce the role of F. nucleatum in modulating host cellular pathways and support the hypothesis that bacterial association potentiates chemoresistance.

1. Introduction

Globally, colorectal cancer (CRC) is the fourth most diagnosed cancer and the second most frequent cause of cancer-related death [1], with a relative 5-year survival rate of 17% for distant metastatic stages [2]. This high mortality is attributed to the incidence of metastasis and chemoresistance in CRC [3]. The gut microbiome plays a critical role in human health, particularly in immune regulation and maintaining intestinal homeostasis [4]. Disruptions to the gut microbial community are associated with reduced effectiveness of chemotherapy and poorer clinical outcomes [5,6]. In the context of CRC, tumor tissues frequently exhibit microbiome dysbiosis, harboring distinct bacterial populations not found in adjacent healthy mucosa. Among these, Fusobacterium nucleatum often emerges as one of the most enriched taxa within colorectal tumors, drawing particular attention for its potential role in cancer progression and resistance to therapy [7,8].
F. nucleatum is a common oral microbe that plays a central role in periodontal disease [9]. However, the clinical understanding of F. nucleatum has expanded due to its isolation from the microbiome of tissues distant from the oral environment. Notably, this organism has been associated with cancerous tissues, suggesting that this microbe may participate in multiple negative outcomes in cancer development and treatment [10]. F. nucleatum’s influence on cancer has most extensively been studied in CRC, where it is becoming one of the most important microbe-related risk factors for CRC prognosis [11]. F. nucleatum has been identified to be significantly enriched in CRC carcinoma tissues compared to healthy adjacent tissues [7]. This pathogen is known to increase tumor proliferation and invasive activities of CRC cells, encouraging inflammation, immune evasion, DNA damage, and metastasis [11,12,13,14]. F. nucleatum participates in these activities by activating TLR4 signaling [15,16,17] and subsequently inducing oncogenic pathways, including Wnt/β-catenin [18,19], PI3K/Akt [20], and NF-κB [21,22]. Activation of these pathways stimulates robust pro-inflammatory cytokine production, including IL-1β, IL-6, IL-8, IL-17A, and TNF-α [12,13]. Moreover, F. nucleatum impairs tight cell junctions and promotes epithelial-to-mesenchymal transition (EMT) by binding and invading epithelial cells through FadA and Fap2 adhesins [23,24,25]. Additionally, F. nucleatum recruits myeloid-derived suppressor cells (MDSCs) and modulates tumor-associated immune populations by suppressing cytotoxic activity to foster an immunosuppressive tumor microenvironment [14,15,25,26].
Due to these various molecular modifications, F. nucleatum colonization has been linked to the development of chemoresistance in CRC [13]. F. nucleatum enrichment in the tumor-associated microbiome from CRC was found in patients with shorter survival times and increased relapses [27,28], suggesting that this pathogen may be a marker of poor prognosis. The use of antibiotic treatment to target anaerobic bacteria, including F. nucleatum, in CRC patients prior to surgical resection had a reduced risk of recurrence or death by 25.5% [29], further suggesting that prognosis is associated with the microbiome. F. nucleatum induces chemoresistance through multiple mechanisms, including activating autophagy, suppressing apoptosis, and regulating the expression of genes critical to drug response [13,28,30]. Through the establishment of a pro-inflammatory and immunosuppressive tumor microenvironment that promotes metastasis and facilitates DNA damage, F. nucleatum enhances the tumor’s susceptibility to the development of chemoresistance [13].
Some emerging studies have suggested F. nucleatum plays a role in engaging in viral reactivation and host antiviral defense pathways, a relatively underexplored mechanism with potential implications for chemoresistance [31,32]. For example, oral coinfections are prevalent in HIV-1 patients, where F. nucleatum has been found to induce latent HIV-1 reactivation through TLR2 and TLR9 [33]. Pathogen recognition receptors (PRRs), activated by pathogen-associated molecular patterns (PAMPs), induce the interferon system to initiate an antiviral response [34]. Specifically, F. nucleatum activates RIG-I, a key cytosolic PRR that prompts type I interferon production, through intracellular invasion and secretion of its nucleic acids [32,35]. Although these responses are classically antiviral, persistent activation within the tumor microenvironment can paradoxically contribute to immune dysregulation, inflammation, and resistance to apoptosis [36,37]. In cancer models, chronic activation of the interferon pathway has been linked to immune evasion and a poor response to chemotherapeutic agents [38,39,40].
The widespread alteration of the cell signaling within CRC tissues clouds the mechanistic understanding of how this organism participates in the multiple and complex signals to the cell. This gap in understanding, coupled with increasing recognition that tissues once thought to be sterile have a robust and active microbiome, opens many questions about therapeutic options. It is estimated that microbial pathogens promote tumorigenesis in 15–20% of cancer cases [41], yet the complete repertoire for how the microbiome influences cancer progression and resistance remains underdeveloped. This study conducted a meta-analysis of all publicly available studies, allowing a direct comparison of the impact of F. nucleatum in-vitro. This work aims to help identify multi-step complex networks of signals that are important yet largely overlooked for F. nucleatum interactions with CRC. This was done by examining multiple transcriptomic datasets from in-vitro co-culture of F. nucleatum with CRC cell lines to find the capacity of F. nucleatum to disrupt key regulatory cell functions to initiate a multitude of cascades that lead to chemoresistance as well as antiviral responses, which may also contribute to chemoresistance. These findings reflect a growing understanding that orally derived F. nucleatum may disseminate to distal tissues and potentiate or exacerbate severe diseases like CRC. More specifically, this study highlights multi-step complex pathways related to chemoresistance that may be regulated by interactions with F. nucleatum.

2. Materials and Methods

2.1. Search Strategy

A systematic search of the NCBI Gene Expression Omnibus (GEO) database [42] was conducted on 20 March 2025, to identify transcriptomic datasets examining the impact of F. nucleatum in cancer-related contexts. The search terms used were: (“cancer” OR “tumor” OR “carcinoma”) AND (“Fusobacterium” OR “Fusobacteria” OR “F. nucleatum”).

2.2. Selection Criteria

Search results were filtered using the Top Organism: Homo sapiens and Entry Type: Series. Duplicate entries were removed prior to analysis. Studies were subsequently manually reviewed and selected based on the following criteria: (1) gene expression profiling from RNA sequencing; (2) CRC cell lines as the model system; and (3) co-culture infection of CRC cell lines with F. nucleatum. Datasets that did not meet these conditions or lacked appropriate control samples were excluded.

2.3. Data Extraction and Synthesis

The ‘Series RNA-seq raw counts matrix’ file was downloaded from GEO for each dataset. The raw count matrices were combined into a single file, with sample labels assigned as either the control group or the F. nucleatum treatment group based on the original study design. This combined raw counts matrix containing all four datasets was then used for differential expression analysis and interpretation. DESeq2 [43] was used within R (version 4.4.1) to analyze the merged dataset. Counts were first normalized using count normalization within DESeq2, then the normalized count table was used to determine differentially expressed genes between F. nucleatum-treated host cells compared to no bacteria added controls. This resulted in a table of log2FC values and associated significance values for F. nucleatum-treated cells compared to non-infected controls, which was used as the input for downstream analyses.
Merging datasets using this strategy strengthens the ability to detect genes that were consistently and significantly altered across multiple conditions, increasing the biological relevance of the findings. This filtering approach reduced noise but may exclude moderately changing genes that may be biologically important. The dataset was not included as a covariate, since the condition was fully nested within the dataset, and modeling it would eliminate the treatment signal. We acknowledge that unmodeled dataset effects remain a limitation of this approach, although consistent patterns across datasets support the biological relevance of the results. Datasets that could not have RNAseq counts downloaded from the GEO database were excluded from downstream analysis. Public metadata from each study were used to identify control and treatment samples (i.e., which samples were treated with F. nucleatum).

2.4. Data Synthesis and Analysis

The combined dataset was imported into Ingenuity Pathway Analysis (IPA) (Qiagen, Redwood City, CA, USA) for enrichment analysis and signal transduction visualization. Gene identifiers were mapped, and a cutoff of |p-value| ≥ 0.1 was applied. Core analysis was performed within IPA (Qiagen) to identify enriched canonical pathways, upstream regulators, causal networks, and disease/function annotations relevant to F. nucleatum interactions in CRC.

2.5. Statistical Analysis

Statistical significance and pathway predictions were evaluated using IPA (Qiagen). Enrichment and differential expression analyses were assessed using −log(p-value), z-score, and log2FC, reflecting the statistical significance and magnitude of gene expression changes. In addition to enrichment analysis, IPA (Qiagen) was used to determine predicted upstream regulator activity and pathway dynamics, computed through activation z-scores and −log(p-value), which quantify the direction and strength of predicted activation or inhibition based on concordance between observed and expected gene expression patterns. Hierarchical clustering and its accompanying dendrograms were created using MetaboAnalyst 6.0. Figures were made and designed in IPA, Adobe Illustrator (version 29.5; San Jose, CA, USA), and bioRender (bioRender, Toronto, ON, Canada).

3. Results

3.1. F. nucleatum Infection Outcome Depends on Experimental Parameters

After screening all public datasets, four eligible datasets were identified that met the quality control settings used in this study and were available on NCBI GEO (Figure 1). Each dataset had its own cell line, infection time, multiplicity of infection (MOI), and overall concluding results, as reported in its associated paper (Appendix A, Table A1), establishing initial variability among the datasets. As these datasets contained only human RNA sequences, we could not directly confirm the presence or abundance of F. nucleatum. However, because each study was conducted in-vitro under controlled conditions with a defined MOI, we relied on the authors’ experimental design and documentation to assert bacterial exposure as defined in the publication. Hierarchical clustering was performed with the expectation that control and infection groups from these individual studies would cluster. This analysis revealed that datasets grouped together rather than treatment with F. nucleatum, indicating experimental design influenced the ultimate study outcome (Appendix A, Figure A1). Therefore, findings that were statistically significant represent those genes and pathways that were above the variation introduced by differences in the study design.

3.2. F. nucleatum Promotes Chemoresistance in Colorectal Cancer

A combined dataset consisting of the four selected studies was run through the core analysis pipeline in IPA, provided in Supplemental File S1. Within this combined dataset, cancer-related inflammatory pathways were the most significantly activated pathways in response to F. nucleatum treatment (Figure 2a). Localized digestive tract-related pathways like those related to digestive system cancers (−log(p-value) = 19.247, z-score = +1.40) and to irritable bowel syndrome (−log(p-value) = 10.934, z-score = +3.528) were significantly activated with F. nucleatum addition. Further supporting this observation of broad inflammatory activation was the more specific induction of cytokine genes such as CSF2, IL2, IL1A, IL1B, IL17A, IL18, IFNG, and TNF (Figure 2b). Together, these observations indicate that F. nucleatum addition modulated the host infectious disease response, immune response, activation of inflammatory signaling, and stimulation of tumor proliferation, all of which may affect cancer onset, progression, and prognosis.
While the activation of inflammatory pathways from F. nucleatum association appeared as a consistent trend, the regulation of multiple cell death and migration pathways by F. nucleatum was inconsistent. Despite the significant activation of pathways involved in apoptosis and organismal death (−log(p-value) = 13.947, z-score = +1.807), those for survival (−log(p-value) = 10.055, z-score = +1.853) and migration of tumor cells (−log(p-value) = 6.777, z-score = +2.301) were also induced. Additionally, the oncogenic pathways Wnt/β-catenin signaling (−log(p-value) = 0.482, z-score = NaN) and PI3K/AKT signaling (−log(p-value) = 1.484, z-score = 0) were expected to be upregulated based on previous work [18,19,20]. However, this analysis found that F. nucleatum did not significantly affect their regulation at the pathway level. These seemingly contradictory findings suggest a complex interplay between cell death and tumor-promoting pathways that was not captured in the models used. Such complexity presents challenges when attempting to define this host–pathogen relationship and perhaps in part explains the confusion and contradictory evidence from in vivo studies [44].

3.2.1. F. nucleatum Induces Chronic Inflammation

Analysis of differentially regulated canonical pathways revealed a prominent inflammatory signature in response to bacterial treatment, with notable enrichment of pathogen-induced cytokine storm signaling (−log(p-value) = 11.92), z-score = +2.502). Despite lacking clear enrichment at the individual gene level, IPA also predicted activation of various inflammatory signaling, including TLR signaling (−log(p-value) = 6.845, activation of z-score = +3.526), NF-κB signaling (−log(p-value) = 14.480, activation of z-score = +5.858), and MAPK signaling (−log(p-value) = 8.013, activation of z-score = +2.456). Consistent with these pathway predictions, a suite of cytokines and chemokines associated with inflammatory recruitment and immune modulation were significantly upregulated. These include TNF (−log(p-value) = 2.438, log2FC = +3.76) and IL1B (−log(p-value) = 1.848, log2FC = +2.07), both implicated in acute phase inflammation and colitis-associated cancer. Chemokines such as CCL2 (−log(p-value) = 1.662, log2FC = +3.58), CCL5 (−log(p-value) = 2.364, log2FC = +1.98), CCL20 (−log(p-value) = 1.646, log2FC = +2.79), CXCL8 (IL8) (−log(p-value) = 6.094, log2FC = +4.39), CXCL1 (−log(p-value) = 2.245, log2FC = +2.48), and CXCL10 (−log(p-value) = 1.456, log2FC = +2.21) also had elevated expression. Functional predictions suggest that these factors contribute to the recruitment of T cells (CXCL8, CXCL10, CCL2, CCL5), monocytes (CCL2), and neutrophils (CXCL8), while IL7 enhanced Th1 cell differentiation, further shaping the immune landscape toward a pro-inflammatory state (Figure 3). Additionally, the top upstream regulator was identified as CSF2 (−log(p-value) = 3.041, log2FC = +3.92), a cytokine central to the recruitment and activation of myeloid-derived cells, including neutrophils, macrophages, and other phagocytes.

3.2.2. F. nucleatum Encourages Immune Evasion

Immune modulation by F. nucleatum exposure was further evidenced by significant enrichment of the Interleukin-10 (IL-10) signaling pathway (−log(p-value) = 9.788, z-score = +3.464).
Several cytokines upregulated in the cross-study comparison are implicated in the induction of immunosuppressive mechanisms. Specifically, elevated expression of CSF2, CCL2, and IL1B was associated with the recruitment and accumulation of myeloid-derived suppressor cells (MDSCs), while CCL2 was predicted to stimulate the proliferation of tumor-associated macrophages (TAMs).

3.2.3. F. nucleatum Influences DNA Damage and Epigenetic Modification

F. nucleatum exposure also triggered a significant shift in host DNA repair and chromatin regulation pathways. Most notably, IPA predicted inhibition of the core base excision repair enzyme POLB (−log(p-value) = 19.629, activation of z-score = −5.63) while predicted activation of components of the polycomb repressive complex 2 (PRC2), EZH2 (−log(p-value) = 18.606, activation of z-score = +3.46,) and SUZ12 (−log(p-value) = 18.932, activation of z-score = +3.35).

3.2.4. F. nucleatum Drives Metastasis

Several pathways related to the motility and invasiveness of malignant cells were significantly enriched, including those involved in the migration of tumor cells (−log(p-value) = 6.777, z-score = +2.301), migration of cancer cells (−log(p-value) = 6.728, z-score = +1.954), movement of tumor cells (−log(p-value) = 6.578, z-score = +1.779), movement of cancer cells (−log(p-value) = 6.187, z-score = +1.105), and invasion of tumor cell lines (−log(p-value) = 6.025, z-score = +1.28). These pathways are fueled through cytokine and chemokine enrichment and may be coupled to the cytokine storm response (Figure 2).
This metastatic signature was accompanied by the concurrent upregulation of matrix metalloproteinases (MMPs), such as MMP3 (−log(p-value) = 2.146, log2FC = +3.02) and MMP13 (−log(p-value) = 1.103, log2FC = +1.37). IPA also predicted the activation of several pro-metastatic regulators, including the small GTPase RHO (−log(p-value) = 20.423, activation of z-score = +3.35).

3.3. New Discoveries

3.3.1. F. nucleatum Influences Chemoresistance Genes and Drug Responses

Several transcriptional shifts associated with chemoresistance genes were observed following F. nucleatum exposure, indicating a possible interference with the efficacy of therapeutic agents, including the differential expression of BIRC3 (Table 1).
Causal network analysis revealed a significant predicted inhibition and activation of various biological and chemical agents, including etanercept, adalimumab, infliximab, GSK2816126, and poly I:C RNA (Table 2). These compounds span anti-inflammatory biologics and epigenetic or immune-stimulating therapeutics, suggesting a broad reprogramming of host response pathways to reshape the intracellular signaling environment in a manner antagonistic to treatment efficacy.

3.3.2. F. nucleatum Induces an Antiviral Response

Several key sensors and mediators of the type I interferon pathway were predicted to be activated, including TLR3 (−log(p-value) = 7.695, activation of z-score = +3.284), TLR7 (−log(p-value) = 7.465, activation of z-score = +3.9), and IFIH1 (MDA5) (−log(p-value) = 19.366, activation of z-score = +6.50). Importantly, expression of DDX58 (RIG-I), a cytosolic sensor of viral RNA, was also modestly enriched (−log(p-value) = 1.254, log2FC = +0.673), suggesting heightened surveillance through RIG-I–like receptor pathways (Figure 4).
Concurrently, type II interferon signaling was strongly activated, as evidenced by the IPA-predicted activation of IFNG (−log(p-value) = 20.947, activation of z-score = +5.91), along with the observed upregulation of its receptor IFNGR (−log(p-value) = 1.342, log2FC = +1.210) and key downstream transcription factors IRF1 (−log(p-value) = 1.346, log2FC = +0.523) and IRF9 (−log(p-value) = 2.593, log2FC = +0.573). While IRF9 expression was modestly increased, IPA further predicted its functional involvement in antiviral activity and pro-inflammatory cytokine production (Figure 3).

4. Discussion

This study leveraged publicly available transcriptomic datasets to investigate the host response to F. nucleatum infection in CRC. Specifically, four independent RNAseq studies were selected based on stringent inclusion criteria. While this focused selection limits the breadth of comparative analysis, it ensures biological relevance and reflects the current availability of suitable datasets in the public domain. Although the datasets differed in cell line, MOI, and infection time, introducing variability and batch effects, this heterogeneity was strategically leveraged to identify transcriptional changes that were conserved across diverse conditions. As a result, only genes with highly significant and large differential changes in expression among all datasets were retained in the analysis. This explains why modest but biologically meaningful gene enrichments, including Wnt/β-catenin, NF-κB, and PI3K/AKT pathways that have been reported in other studies [18,45], were not identified.
Nevertheless, the integrated dataset produced here reflects core host processes consistently modulated by F. nucleatum, emphasizing that these mechanisms are not artifacts of a single experimental setup but likely represent fundamental features of the host–microbe interaction. While the use of in vitro cell lines does not fully represent the complexity of the tumor microenvironment, this study addresses fundamental molecular signaling pathways to inspire further in vivo explorations. This study confirms previous mechanisms for chemoresistance, being inflammation, immune evasion, DNA damage, and metastasis, as well as proposes F. nucleatum’s role in drug efficacy and the activation of a pseudo-antiviral state.
Our findings support and build on existing literature proposing F. nucleatum promotes inflammation-driven tumorigenesis, with the further implication of fostering a pro-inflammatory microenvironment that initiates chemoresistance. Previous studies report that F. nucleatum triggers Toll-like receptor (TLR) signaling, particularly TLR4, which in turn activates inflammatory cascades via central proteins that include NF-κB and MAPK, resulting in elevated cytokine and chemokine expression that supports immune evasion, epithelial proliferation, and carcinogenesis [13,15,46]. Although canonical components of these pathways, such as TLRs, NF-κB, or MAPKs, were not significantly differentially expressed in the integrated datasets, the downstream signaling cascades were predicted to be enriched.
Correspondingly, the expression of several downstream pro-inflammatory mediators were significantly upregulated, including TNF, IL1B, CXCL8 (IL8), CCL2, CCL5, CCL20, and CXCL10. These cytokines and chemokines orchestrate immune cell recruitment and amplify the inflammatory response [14,47,48]. This potent recruitment signaling could establish a self-sustaining cycle of chronic inflammation that continuously attracts immune cells. Over time, instead of mounting an anti-tumor response, these cells reinforce tumor-promoting conditions. This is supported by the observation of CSF2, a cell growth-initiating cytokine, being the most significant regulatory network among these studies. CSF2 is a known biomarker and prognostic factor that influences the host immune response [49,50], and this analysis confirmed induction for the known role in recruiting and activating immune stimulatory cells. Inflammatory dysregulation of this nature not only contributes to therapeutic resistance but also facilitates tumor cell plasticity and epithelial-to-mesenchymal transition, thereby enhancing metastatic potential [14]. Collectively, these findings reinforce F. nucleatum’s prominent role in promoting a chronic, immune-mediated inflammatory microenvironment that may contribute to both tumor progression and resistance to therapy. Further detailed work is needed to verify this specific activity directly in vivo, perhaps with gnotobiotic animal models or organoids.
Beyond initiating inflammation, F. nucleatum has increasingly been recognized for its ability to manipulate the tumor immune microenvironment, and this study further supports its role in promoting immune evasion among the datasets compared. The induction of IL-10 signaling may represent a mechanism by which F. nucleatum dampens antitumor immune responses, thereby fostering an immunosuppressive microenvironment conducive to chemoresistance. IL-10 can inhibit pro-inflammatory cytokine production and suppress the Th1-mediated immune response, ultimately supporting tumor immune evasion. Yet, its role in cancer remains inconclusive, with studies indicating a dual role in tumor suppression and promotion. In CRC, IL-10 is significantly elevated, while in other cancers, IL-10 signaling has been shown to impair responsiveness to chemotherapy [51,52,53]. This immunomodulatory shift may enable persistent tumor cell survival during treatment, reinforcing the development of chemoresistant subpopulations and metastatic progression. The recruitment of myeloid-derived suppressor cells (MDSCs), which suppress T cell proliferation and effector function [54], has been documented as a key immunosuppressive strategy in F. nucleatum-associated colorectal tumors [12]. Prior studies have shown that elevated levels of IL1B, CSF2, and CCL2, all of which had increased expression in our dataset, enhance the mobilization and suppressive function of MDSCs [54,55]. CCL2 is particularly central to this process, not only recruiting MDSCs but also expanding tumor-associated macrophages (TAMs), which can shift toward an M2-like phenotype and suppress cytotoxic T cell activity [15,21,56]. Taken together, these findings underscore how F. nucleatum infection may foster an immunosuppressive niche by subverting both innate and adaptive immune components. By enabling immune evasion and establishing a suppressive microenvironment, F. nucleatum effectively shields tumor cells from immunogenic cell death, reducing the efficacy of immunotherapies and chemotherapeutic agents that rely on immune system engagement.
F. nucleatum has been implicated in promoting genomic instability and epigenetic modification in host epithelial cells, and this comparative analysis further suggests that this pathogen may alter host DNA repair responses. Although the genes within these pathways were not significantly enriched in this analysis, they were predicted to be activated based on downstream activity, suggesting coordinated upstream regulation and downstream induction. Prior studies have shown that F. nucleatum exposure increases DNA damage in colorectal epithelial cells, particularly through the generation of ROS and subsequent oxidative stress [12,13]. This damage often necessitates base excision repair (BER), a process heavily dependent on DNA polymerase β (POLB) [57]. Inhibition of POLB, as observed in this analysis, may impair BER efficiency and allow the accumulation of mutations, potentially driving tumorigenesis. While proposed in various studies, this work highlights the potential that a bacterium may induce mutations in mammals. Compounding this, the activation of epigenetic silencers EZH2 and SUZ12, core members of the polycomb repressive complex 2 (PRC2), suggests repression of DNA repair and tumor suppressor genes. EZH2 has been shown to transcriptionally silence key repair mediators and checkpoint regulators, contributing to genomic instability and poor prognosis in cancer [58,59]. Moreover, PRC2 activation has been linked to EMT, the acquisition of stem-like features, and silencing tumor suppressor genes that further promote cancer progression [60]. These mechanisms align with recent reports that F. nucleatum infection promotes a pro-carcinogenic epigenetic landscape, including altered chromatin accessibility and gene silencing at DNA repair loci [11,12]. The coordinated repression of repair enzymes and activation of epigenetic modifiers observed here highlights a dual strategy by which F. nucleatum may foster genomic instability, by both damaging DNA and simultaneously suppressing the cellular machinery required to correct it. These disruptions in DNA repair pathways may compromise the long-term effectiveness of DNA-damaging chemotherapies like 5-FU and oxaliplatin, common treatments for CRC [61], not by reducing their cytotoxicity, but by synergistically enhancing mutation accumulation during treatment. This may potentiate tumor evolution and relapse through this unchecked genomic instability, accelerating clonal diversification and disease progression. Considering this organism is a common inhabitant of the oral cavity and promotes mutagenic activity in the host tissue, further studies are needed that specifically examine how, where, and when this modulation occurs, as it will likely have an impact on many tissues and diseases.
As observed in analysis, F. nucleatum promotes a metastatic phenotype in CRC cells through the coordinated activation of cytoskeletal motility and extracellular matrix (ECM) remodeling programs. Infection is observed to induce a transcriptional state conducive to metastatic progression and EMT, with significant enrichment of pathways regulating cell migration and invasion. This metastatic gene expression profile was accompanied by the robust activation of RHO, a small GTPase that drives cytoskeletal remodeling and cell motility. This activation points to dysregulated cytoskeletal dynamics and cellular migration, processes known to be hijacked during EMT and metastatic spread [62]. Furthermore, enrichment of MMPs such as MMP3 and MMP13, which support ECM degradation and dissemination through tissue barriers, was observed in this analysis, as well as being reported in other studies [63]. MMPs also drive cancer migration through initiating EMT. MMPs have been implicated in mediating resistance to chemotherapy by altering drug penetration, modulating the tumor microenvironment, and activating pro-survival signaling pathways [63,64,65]. MMPs have been associated with F. nucleatum infection, facilitating microbial invasion and colonization but also contributing to tumor cell motility and immune evasion [63,65], being stimulated by cytokines such as TNF [66]. Elevated TNF expression in this context may further contribute to endothelial permeability and barrier dysfunction, promoting not only local invasion but also potential distant metastasis [67,68]. This metastatic reprogramming may enable tumor cells to evade drug delivery, contribute to heterogeneity in drug response, and establish resistant niches at secondary sites.
Collectively, this analysis suggests that F. nucleatum may contribute to the development of chemoresistance in CRC through multifaceted reprogramming of oncogenic signaling that leads to inflammation, immune evasion, DNA damage, and metastasis. Additionally, several chemoresistance-associated genes were significantly altered in response to F. nucleatum exposure, suggesting that this microbe may contribute in other ways directly towards therapeutic failure in host cells. Several pro-survival and chemoresistance genes were upregulated, including TNF (log2FC = +3.76, p = 0.0036) [69]. CYP3A4 (log2FC = +1.54, p = 0.0143), a key enzyme involved in xenobiotic metabolism, was also significantly induced, potentially accelerating the degradation and inactivation of chemotherapeutic agents [70]. The upregulation of ESR1 (log2FC = +1.80, p = 0.0398), encoding estrogen receptor alpha, is notable given its known involvement in resistance to hormone-targeted therapies when mutated and in CRC is linked to poor prognosis [71,72]. BIRC3 (log2FC = +2.85, p = 0.000692), a member of the inhibitor of apoptosis proteins (IAP) family, was significantly upregulated, confirming previous studies indicating BIRC3 enrichment in CRC. BIRC3 enrichment is also associated with chemoresistance to 5-FU [30]. These transcriptomic shifts suggest that F. nucleatum orchestrates a coordinated reprogramming of host defense, apoptosis regulation, and drug response pathways, tipping the balance toward chemoresistance. This complexity indicates that narrowly focused analyses will likely miss important regulatory triggers induced by F. nucleatum, indicating that a broader assessment of interconnected networks is likely needed to fully appreciate the impact of F. nucleatum.
Considering that therapeutic drugs have molecular targets in pathways described in this work, we examined how these signaling networks would influence drug impact. Network analysis in this study indicated that F. nucleatum infection modulates mechanisms that are associated with a broad panel of biologic and chemical drugs, indicating that the activity for these drugs may be affected. Etanercept, infliximab, and adalimumab are TNF-α inhibitors and were significantly inhibited among the datasets, consistent with the upregulation of TNF signaling that was observed. These drugs block TNF-mediated inflammation and are often used in immunosuppressive settings [73]. Their inhibition implies that F. nucleatum may amplify TNF-dependent inflammation that directly opposes and could reduce the activity for those drugs.
Multiple epigenetic and metabolic regulators were also targeted. GSK2816126, an EZH2 inhibitor [74], was inhibited, implying that F. nucleatum may maintain pro-tumor epigenetic states by preventing PRC2 complex inhibition. Additionally, poly dA-dT and poly rI:rC-RNA, nucleic acids that trigger innate immune stimuli through cytosolic DNA and RNA sensors such as STING1 and TLR3 [35,75], were activated, indicating an engagement of innate antiviral responses. The widespread inhibition or activation of drugs targeting inflammation, epigenetics, and metabolic pathways underscores F. nucleatum’s capacity to remodel the tumor environment and reinforce resistance against diverse therapeutic modalities. By influencing the activity of pathways targeted by these drugs, F. nucleatum may actively diminish their therapeutic efficacy, suggesting a direct microbial role in shaping host drug responsiveness and chemoresistance.
The observation that Fusobacterium nucleatum induces both type I and type II interferon signaling adds a novel layer to our understanding of host–microbe interactions in CRC. However, while F. nucleatum is traditionally studied in the context of inflammation and immune evasion, there is little research investigating whether it can also engage antiviral pathways [32,33]. The antiviral response is initiated through the interferon system, comprising secreted cytokines such as type I interferons IFN-α and IFN-β and type II interferon IFN-γ. These cytokines can be induced through recognition of pathogen-associated molecular patterns (PAMPs) by pattern-recognition receptors (PRRs), including TLR2, TLR3, TLR7, TLR9, and IFIH1, which then induce interferon-regulatory factors such as IRF-1, IRF-3, and IRF-9 [34]. The activation of innate RNA sensors such as TLR3, TLR7, MDA5, and RIG-I, observed in this comparative analysis, mirrors mechanisms typically reserved for viral infections and may represent an immune response that fuels chronic inflammation via the induction of interferons [34,37]. RIG-I specifically is a cytosol PRR that senses nucleic acids, and F. nucleatum has been identified to secrete nucleic acids to activate RIG-I and trigger a pro-inflammatory response [32,33]. Sustained activation of type I interferon responses has been shown to contribute to immune exhaustion, metastasis, and tumor-promoting inflammation in several cancers [37,40].
Moreover, F. nucleatum was also observed to activate type II interferon signaling, with the activation of IFNG (IFN-γ) and enrichment of its corresponding receptor IFNGR. Type II interferon signaling, while not typically antiviral, plays a crucial role in shaping immune responses that support antiviral-like states through enhanced pro-inflammatory signaling, antigen presentation, and the induction of interferon-stimulated genes via STAT1-dependent pathways [76]. IFNG is known to activate immune effector pathways, but when chronically induced, can also promote immunoregulatory programs that paradoxically support tumor survival [76].

5. Conclusions

This integrative transcriptomic analysis, using independent in vitro inoculation studies, identified multiple integrated signaling pathways initiated by F. nucleatum. These analyses uncovered observations that lead to specific molecular mechanisms to allow compelling conclusions that F. nucleatum plays a significant role in chemoresistance development in CRC, through mechanisms of inflammation, immune evasion, DNA damage, and metastasis. Furthermore, these findings broaden the paradigm of F. nucleatum pathogenicity, suggesting that its presence may alter drug efficacy and trigger a pseudo-viral immune state. The broad impact of F. nucleatum on the cellular response highlights the controversial role of this bacterium. Further exploration of these pathways could provide new insights into how microbial pattern recognition intersects with tumor immunology and how disrupting these signals might restore immune equilibrium and improve therapeutic outcomes.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers17193247/s1, File S1: A combined dataset.

Author Contributions

Conceptualization, K.R.R., C.A.S., J.C. and B.C.W.; methodology, K.R.R., C.A.S. and B.C.W.; formal analysis, K.R.R. and C.A.S.; investigation, K.R.R., C.A.S., J.C. and B.C.W.; resources, B.C.W.; data curation, K.R.R. and C.A.S.; writing—original draft preparation, K.R.R.; writing—review and editing, K.R.R., C.A.S., J.C. and B.C.W.; visualization, K.R.R. and C.A.S.; project administration, B.C.W.; funding acquisition, B.C.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original datasets are available at NCBI. The specific data used for this analysis are available as Supplementary Files.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
BERBase Excision Repair
CRCColorectal Cancer
ECMExtracellular Matrix
EMTEpithelial-to-Mesenchymal Transition
GEOGene Expression Omnibus
IPAIngenuity Pathway Analysis
MDSCsMyeloid-Derived Suppressor Cells
MMPsMatrix Metalloproteinases
MOIMultiplicity of Infection
PAMPsPathogen-Associated Molecular Patterns
PRRsPattern-Recognition Receptors
TAMsTumor-Associated Macrophages

Appendix A

Table A1. Paper characteristics for the selected datasets. This table indicates their accession number on NCBI GEO, paper publication date, cancer model, cancer cell line, Fusobacterium strain, infection time in hours, MOI, and paper conclusions. Based on these experimental conditions, there is initial diversity between datasets as different CRC cell lines and infection times were used among the different datasets.
Table A1. Paper characteristics for the selected datasets. This table indicates their accession number on NCBI GEO, paper publication date, cancer model, cancer cell line, Fusobacterium strain, infection time in hours, MOI, and paper conclusions. Based on these experimental conditions, there is initial diversity between datasets as different CRC cell lines and infection times were used among the different datasets.
AccessionPub DateCancerCell LineFusobacterium StrainInfection Time (hrs)MOIPaper ConclusionsCitation
GSE245617August, 2024CRCHCT116F. nucleatum 223,7263100Adhesion RadD directly binds to CD147 and induces a PI3K–AKT–NF–κB–MMP9 cascade that heightens tumorigenesis[77]
GSE173549October, 2021CRCLOVOF. nucleatum 25,58624100F. nucleatum influences the miR-1322/CCL20 axis and M2 macrophage polarization to encourage metastasis and reprogram the tumor microenvironment[78]
GSE90944July, 2017CRCHT29F. nucleatum 25,5862–4100F. nucleatum induces autophagy to encourage chemoresistance[28]
GSE175593October, 2021CRCDLD-1F. nucleatum 25,586Overnight10F. nucleatum induces ANGPTL4 to increase glycolysis activity and thus initiate further F. nucleatum colonization[79]
Figure A1. Dendrogram of datasets to indicate hierarchical clustering. FN indicates CRC cell lines cocultured with F. nucleatum, while Control indicates CRC cell lines that were not infected. The numbers following their identified experimental condition indicate the dataset, being 1–4, followed by the identification of the biological replicate in that experiment. Replicates grouped together based on their dataset, rather than experimental condition.
Figure A1. Dendrogram of datasets to indicate hierarchical clustering. FN indicates CRC cell lines cocultured with F. nucleatum, while Control indicates CRC cell lines that were not infected. The numbers following their identified experimental condition indicate the dataset, being 1–4, followed by the identification of the biological replicate in that experiment. Replicates grouped together based on their dataset, rather than experimental condition.
Cancers 17 03247 g0a1

References

  1. Colorectal Cancer. Available online: https://www.who.int/news-room/fact-sheets/detail/colorectal-cancer (accessed on 15 April 2025).
  2. Colon Cancer: Symptoms, Stages & Treatment. Available online: https://my.clevelandclinic.org/health/diseases/14501-colorectal-colon-cancer (accessed on 15 April 2025).
  3. Ashique, S.; Bhowmick, M.; Pal, R.; Khatoon, H.; Kumar, P.; Sharma, H.; Garg, A.; Kumar, S.; Das, U. Multi Drug Resistance in Colorectal Cancer- Approaches to Overcome, Advancements and Future Success. Adv. Cancer Biol. Metastasis 2024, 10, 100114. [Google Scholar] [CrossRef]
  4. Ding, G.; Yang, X.; Li, Y.; Wang, Y.; Du, Y.; Wang, M.; Ye, R.; Wang, J.; Zhang, Y.; Chen, Y.; et al. Gut Microbiota Regulates Gut Homeostasis, Mucosal Immunity and Influences Immune-Related Diseases. Mol. Cell Biochem. 2024, 480, 1969–1981. [Google Scholar] [CrossRef]
  5. Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal Bacteria Control Cancer Response to Therapy by Modulating the Tumor Microenvironment. Science 2013, 342, 967–970. [Google Scholar] [CrossRef]
  6. Viaud, S.; Saccheri, F.; Mignot, G.; Yamazaki, T.; Daillère, R.; Hannani, D.; Enot, D.P.; Pfirschke, C.; Engblom, C.; Pittet, M.J.; et al. The Intestinal Microbiota Modulates the Anticancer Immune Effects of Cyclophosphamide. Science 2013, 342, 971. [Google Scholar] [CrossRef] [PubMed]
  7. Kostic, A.D.; Gevers, D.; Pedamallu, C.S.; Michaud, M.; Duke, F.; Earl, A.M.; Ojesina, A.I.; Jung, J.; Bass, A.J.; Tabernero, J.; et al. Genomic Analysis Identifies Association of Fusobacterium with Colorectal Carcinoma. Genome Res. 2012, 22, 292. [Google Scholar] [CrossRef] [PubMed]
  8. Kovács, T.; Mikó, E.; Ujlaki, G.; Sári, Z.; Bai, P. The Microbiome as a Component of the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1225, 137–153. [Google Scholar] [CrossRef]
  9. Signat, B.; Roques, C.; Poulet, P.; Duffaut, D. Role of Fusobacterium Nucleatum in Periodontal Health and Disease. Curr. Issues Mol. Biol. 2011, 13, 25–36. [Google Scholar] [CrossRef]
  10. Akbari, E.; Epstein, J.B.; Samim, F. Unveiling the Hidden Links: Periodontal Disease, Fusobacterium Nucleatum, and Cancers. Curr. Oncol. Rep. 2024, 26, 1388–1397. [Google Scholar] [CrossRef]
  11. Wang, N.; Fang, J.Y. Fusobacterium Nucleatum, a Key Pathogenic Factor and Microbial Biomarker for Colorectal Cancer. Trends Microbiol. 2023, 31, 159–172. [Google Scholar] [CrossRef]
  12. Galasso, L.; Termite, F.; Mignini, I.; Esposto, G.; Borriello, R.; Vitale, F.; Nicoletti, A.; Paratore, M.; Ainora, M.E.; Gasbarrini, A.; et al. Unraveling the Role of Fusobacterium Nucleatum in Colorectal Cancer: Molecular Mechanisms and Pathogenic Insights. Cancers 2025, 17, 368. [Google Scholar] [CrossRef]
  13. Dadgar-Zankbar, L.; Elahi, Z.; Shariati, A.; Khaledi, A.; Razavi, S.; Khoshbayan, A. Exploring the Role of Fusobacterium Nucleatum in Colorectal Cancer: Implications for Tumor Proliferation and Chemoresistance. Cell Commun. Signal. 2024, 22, 1–16. [Google Scholar] [CrossRef] [PubMed]
  14. Wu, Z.; Ma, Q.; Guo, Y.; You, F. The Role of Fusobacterium Nucleatum in Colorectal Cancer Cell Proliferation and Migration. Cancers 2022, 14, 5350. [Google Scholar] [CrossRef] [PubMed]
  15. Hu, L.; Liu, Y.; Kong, X.; Wu, R.; Peng, Q.; Zhang, Y.; Zhou, L.; Duan, L. Fusobacterium Nucleatum Facilitates M2 Macrophage Polarization and Colorectal Carcinoma Progression by Activating TLR4/NF-ΚB/S100A9 Cascade. Front. Immunol. 2021, 12, 658681. [Google Scholar] [CrossRef] [PubMed]
  16. Liu, H.; Redline, R.W.; Han, Y.W. Fusobacterium Nucleatum Induces Fetal Death in Mice via Stimulation of TLR4-Mediated Placental Inflammatory Response. J. Immunol. 2007, 179, 2501–2508. [Google Scholar] [CrossRef]
  17. Jia, Y.P.; Wang, K.; Zhang, Z.J.; Tong, Y.N.; Han, D.; Hu, C.Y.; Li, Q.; Xiang, Y.; Mao, X.H.; Tang, B. TLR2/TLR4 Activation Induces Tregs and Suppresses Intestinal Inflammation Caused by Fusobacterium Nucleatum in Vivo. PLoS ONE 2017, 12, e0186179. [Google Scholar] [CrossRef]
  18. Rubinstein, M.R.; Baik, J.E.; Lagana, S.M.; Han, R.P.; Raab, W.J.; Sahoo, D.; Dalerba, P.; Wang, T.C.; Han, Y.W. Fusobacterium Nucleatum Promotes Colorectal Cancer by Inducing Wnt/Β-catenin Modulator Annexin A1. EMBO Rep. 2019, 20, e47638. [Google Scholar] [CrossRef]
  19. Rubinstein, M.R.; Wang, X.; Liu, W.; Hao, Y.; Cai, G.; Han, Y.W. Fusobacterium Nucleatum Promotes Colorectal Carcinogenesis by Modulating E-Cadherin/β-Catenin Signaling via Its FadA Adhesin. Cell Host Microbe 2013, 14, 195–206. [Google Scholar] [CrossRef]
  20. Fruman, D.A.; Rommel, C. PI3K and Cancer: Lessons, Challenges and Opportunities. Nat. Rev. Drug Discov. 2014, 13, 140–156. [Google Scholar] [CrossRef]
  21. Zheng, W.; Wang, Y.; Sun, H.; Bao, S.; Ge, S.; Quan, C.; Jantsch, J.; Schröder, A.; Weber, M. The Role of Fusobacterium Nucleatum in Macrophage M2 Polarization and NF-ΚB Pathway Activation in Colorectal Cancer. Front. Immunol. 2025, 16, 1549564. [Google Scholar] [CrossRef]
  22. Harrandah, A.M.; Chukkapalli, S.S.; Bhattacharyya, I.; Progulske-Fox, A.; Chan, E.K.L. Fusobacteria Modulate Oral Carcinogenesis and Promote Cancer Progression. J. Oral. Microbiol. 2021, 13, 1849493. [Google Scholar] [CrossRef]
  23. Guo, P.; Tian, Z.; Kong, X.; Yang, L.; Shan, X.; Dong, B.; Ding, X.; Jing, X.; Jiang, C.; Jiang, N.; et al. 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. [Google Scholar] [CrossRef]
  24. Fardini, Y.; Wang, X.; Témoin, S.; Nithianantham, S.; Lee, D.; Shoham, M.; Han, Y.W. Fusobacterium Nucleatum Adhesin FadA Binds Vascular Endothelial Cadherin and Alters Endothelial Integrity. Mol. Microbiol. 2011, 82, 1468–1480. [Google Scholar] [CrossRef] [PubMed]
  25. Gur, C.; Ibrahim, Y.; Isaacson, B.; Yamin, R.; Abed, J.; Gamliel, M.; Enk, J.; Bar-On, Y.; Stanietsky-Kaynan, N.; Coppenhagen-Glazer, S.; et al. Binding of the Fap2 Protein of Fusobacterium Nucleatum to Human Inhibitory Receptor TIGIT Protects Tumors from Immune Cell Attack. Immunity 2015, 42, 344–355. [Google Scholar] [CrossRef] [PubMed]
  26. Sakamoto, Y.; Mima, K.; Ishimoto, T.; Ogata, Y.; Imai, K.; Miyamoto, Y.; Akiyama, T.; Daitoku, N.; Hiyoshi, Y.; Iwatsuki, M.; et al. Relationship between Fusobacterium Nucleatum and Antitumor Immunity in Colorectal Cancer Liver Metastasis. Cancer Sci. 2021, 112, 4470–4477. [Google Scholar] [CrossRef] [PubMed]
  27. Mima, K.; Nishihara, R.; Qian, Z.R.; Cao, Y.; Sukawa, Y.; Nowak, J.A.; Yang, J.; Dou, R.; Masugi, Y.; Song, M.; et al. Fusobacterium Nucleatum in Colorectal Carcinoma Tissue and Patient Prognosis. Gut 2016, 65, 1973–1980. [Google Scholar] [CrossRef]
  28. Yu, T.C.; Guo, F.; Yu, Y.; Sun, T.; Ma, D.; Han, J.; Qian, Y.; Kryczek, I.; Sun, D.; Nagarsheth, N.; et al. Fusobacterium Nucleatum Promotes Chemoresistance to Colorectal Cancer by Modulating Autophagy. Cell 2017, 170, 548. [Google Scholar] [CrossRef]
  29. Wang, M.; Rousseau, B.; Qiu, K.; Huang, G.; Zhang, Y.; Su, H.; Le Bihan-Benjamin, C.; Khati, I.; Artz, O.; Foote, M.B.; et al. Killing Tumor-Associated Bacteria with a Liposomal Antibiotic Generates Neoantigens That Induce Anti-Tumor Immune Responses. Nat. Biotechnol. 2024, 42, 1263–1274. [Google Scholar] [CrossRef]
  30. Zhang, S.; Yang, Y.; Weng, W.; Guo, B.; Cai, G.; Ma, Y.; Cai, S. Fusobacterium Nucleatum Promotes Chemoresistance to 5-Fluorouracil by Upregulation of BIRC3 Expression in Colorectal Caner. J. Exp. Clin. Cancer Res. 2019, 38, 14. [Google Scholar] [CrossRef]
  31. Khodarev, N.N.; Roizman, B.; Weichselbaum, R.R. Molecular Pathways: Interferon/Stat1 Pathway: Role in the Tumor Resistance to Genotoxic Stress and Aggressive Growth. Clin. Cancer Res. 2012, 18, 3015–3021. [Google Scholar] [CrossRef]
  32. Lee, P.; Tan, K.S. Fusobacterium Nucleatum Activates the Immune Response through Retinoic Acid-Inducible Gene i. J. Dent. Res. 2014, 93, 162–168. [Google Scholar] [CrossRef]
  33. González, O.A.; Li, M.; Ebersole, J.L.; Huang, C.B. HIV-1 Reactivation Induced by the Periodontal Pathogens Fusobacterium Nucleatum and Porphyromonas Gingivalis Involves Toll-like Receptor 4 and 9 Activation in Monocytes/Macrophages. Clin. Vaccine Immunol. 2010, 17, 1417–1427. [Google Scholar] [CrossRef]
  34. Randall, R.E.; Goodbourn, S. Interferons and Viruses: An Interplay between Induction, Signalling, Antiviral Responses and Virus Countermeasures. J. General. Virol. 2008, 89, 1–47. [Google Scholar] [CrossRef]
  35. Qiao, Y.; Zhu, S.; Deng, S.; Zou, S.S.; Gao, B.; Zang, G.; Wu, J.; Jiang, Y.; Liu, Y.J.; Chen, J. Human Cancer Cells Sense Cytosolic Nucleic Acids Through the RIG-I–MAVS Pathway and CGAS–STING Pathway. Front. Cell Dev. Biol. 2021, 8, 606001. [Google Scholar] [CrossRef]
  36. Musella, M.; Galassi, C.; Manduca, N.; Sistigu, A. The Yin and Yang of Type I IFNs in Cancer Promotion and Immune Activation. Biology 2021, 10, 856. [Google Scholar] [CrossRef]
  37. Kopitar-Jerala, N. The Role of Interferons in Inflammation and Inflammasome Activation. Front. Immunol. 2017, 8, 263291. [Google Scholar] [CrossRef]
  38. Minn, A.J. Interferons and the Immunogenic Effects of Cancer Therapy. Trends Immunol. 2015, 36, 725. [Google Scholar] [CrossRef]
  39. Padariya, M.; Sznarkowska, A.; Kote, S.; Gómez-Herranz, M.; Mikac, S.; Pilch, M.; Alfaro, J.; Fahraeus, R.; Hupp, T.; Kalathiya, U. Functional Interfaces, Biological Pathways, and Regulations of Interferon-Related DNA Damage Resistance Signature (IRDS) Genes. Biomolecules 2021, 11, 622. [Google Scholar] [CrossRef] [PubMed]
  40. Boukhaled, G.M.; Harding, S.; Brooks, D.G. Opposing Roles of Type I Interferons in Cancer Immunity. Annu. Rev. Pathol. 2020, 16, 167. [Google Scholar] [CrossRef] [PubMed]
  41. Bhatt, A.P.; Redinbo, M.R.; Bultman, S.J. The Role of the Microbiome in Cancer Development and Therapy. CA Cancer J. Clin. 2017, 67, 326–344. [Google Scholar] [CrossRef] [PubMed]
  42. Edgar, R.; Domrachev, M.; Lash, A.E. Gene Expression Omnibus: NCBI Gene Expression and Hybridization Array Data Repository. Nucleic Acids Res. 2002, 30, 207–210. [Google Scholar] [CrossRef]
  43. Love, M.I.; Huber, W.; Anders, S. Moderated Estimation of Fold Change and Dispersion for RNA-Seq Data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef]
  44. Queen, J.; Domingue, J.C.; White, J.R.; Stevens, C.; Udayasuryan, B.; Nguyen, T.T.D.; Wu, S.; Ding, H.; Fan, H.; McMann, M.; et al. Comparative Analysis of Colon Cancer-Derived Fusobacterium Nucleatum Subspecies: Inflammation and Colon Tumorigenesis in Murine Models. mBio 2022, 13, e02991-21. [Google Scholar] [CrossRef]
  45. Kang, W.; Jia, Z.; Tang, D.; Zhang, Z.; Gao, H.; He, K.; Feng, Q. Fusobacterium Nucleatum Facilitates Apoptosis, ROS Generation, and Inflammatory Cytokine Production by Activating AKT/MAPK and NF-ΚB Signaling Pathways in Human Gingival Fibroblasts. Oxid. Med. Cell Longev. 2019, 2019, 1681972. [Google Scholar] [CrossRef]
  46. Park, S.R.; Kim, D.J.; Han, S.H.; Kang, M.J.; Lee, J.Y.; Jeong, Y.J.; Lee, S.J.; Kim, T.H.; Ahn, S.G.; Yoon, J.H.; et al. Diverse Toll-like Receptors Mediate Cytokine Production by Fusobacterium Nucleatum and Aggregatibacter Actinomycetemcomitans in Macrophages. Infect. Immun. 2014, 82, 1914–1920. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, F.; Mears, J.R.; Shakib, L.; Beynor, J.I.; Shanaj, S.; Korsunsky, I.; Nathan, A.; Donlin, L.T.; Raychaudhuri, S. IFN-γ and TNF-α Drive a CXCL10+ CCL2+ Macrophage Phenotype Expanded in Severe COVID-19 Lungs and Inflammatory Diseases with Tissue Inflammation. Genome Med. 2021, 13, 64. [Google Scholar] [CrossRef] [PubMed]
  48. Zheng, Z.Q.; Yuan, G.Q.; Zhang, G.G.; Chen, Y.T.; Nie, Q.Q.; Wang, Z. Identification of CCL20 as a Key Biomarker of Inflammatory Responses in the Pathogenesis of Intracerebral Hemorrhage. Inflammation 2023, 46, 1290–1304. [Google Scholar] [CrossRef] [PubMed]
  49. Xu, Z.; Zhang, Y.; Xu, M.; Zheng, X.; Lin, M.; Pan, J.; Ye, C.; Deng, Y.; Jiang, C.; Lin, Y.; et al. Demethylation and Overexpression of CSF2 Are Involved in Immune Response, Chemotherapy Resistance, and Poor Prognosis in Colorectal Cancer. OncoTargets Ther. 2019, 12, 11255–11269. [Google Scholar] [CrossRef]
  50. Chen, Y.C.; Zheng, W.Z.; Liu, C.P.; Zhao, Y.Q.; Li, J.W.; Du, Z.S.; Zhai, T.T.; Lin, H.Y.; Shi, W.Q.; Cai, S.Q.; et al. Pan-Cancer Analysis Reveals CCL5/CSF2 as Potential Predictive Biomarkers for Immune Checkpoint Inhibitors. Cancer Cell Int. 2024, 24, 311. [Google Scholar] [CrossRef]
  51. Stanilov, N.; Miteva, L.; Deliysky, T.; Jovchev, J.; Stanilova, S. Advanced Colorectal Cancer Is Associated With Enhanced IL-23 and IL-10 Serum Levels. Lab. Med. 2010, 41, 159–163. [Google Scholar] [CrossRef]
  52. Yang, C.; He, L.; He, P.; Liu, Y.; Wang, W.; He, Y.; Du, Y.; Gao, F. Increased Drug Resistance in Breast Cancer by Tumor-Associated Macrophages through IL-10/STAT3/Bcl-2 Signaling Pathway. Med. Oncol. 2015, 32, 14. [Google Scholar] [CrossRef]
  53. Wu, D.; Wang, Z. Gastric Cancer Cell-Derived Kynurenines Hyperactive Regulatory T Cells to Promote Chemoresistance via the IL-10/STAT3/BCL2 Signaling Pathway. DNA Cell Biol. 2022, 41, 447–455. [Google Scholar] [CrossRef]
  54. Lu, J.; Luo, Y.; Rao, D.; Wang, T.; Lei, Z.; Chen, X.; Zhang, B.; Li, Y.; Liu, B.; Xia, L.; et al. Myeloid-Derived Suppressor Cells in Cancer: Therapeutic Targets to Overcome Tumor Immune Evasion. Exp. Hematol. Oncol. 2024, 13, 39. [Google Scholar] [CrossRef]
  55. Li, X.; Wang, J.; Wu, W.; Gao, H.; Liu, N.; Zhan, G.; Li, L.; Han, L.; Guo, X. Myeloid-Derived Suppressor Cells Promote Epithelial Ovarian Cancer Cell Stemness by Inducing the CSF2/p-STAT3 Signalling Pathway. FEBS J. 2020, 287, 5218–5235. [Google Scholar] [CrossRef]
  56. Huang, R.; Kang, T.; Chen, S. The Role of Tumor-Associated Macrophages in Tumor Immune Evasion. J. Cancer Res. Clin. Oncol. 2024, 150, 238. [Google Scholar] [CrossRef] [PubMed]
  57. Gulkis, M.; Martinez, E.; Almohdar, D.; Çaglayan, M. Unfilled Gaps by Polβ Lead to Aberrant Ligation by LIG1 at the Downstream Steps of Base Excision Repair Pathway. Nucleic Acids Res. 2024, 52, 3810–3822. [Google Scholar] [CrossRef] [PubMed]
  58. Gan, L.; Yang, Y.; Li, Q.; Feng, Y.; Liu, T.; Guo, W. Epigenetic Regulation of Cancer Progression by EZH2: From Biological Insights to Therapeutic Potential. Biomark. Res. 2018, 6, 1–10. [Google Scholar] [CrossRef] [PubMed]
  59. Duan, R.; Du, W.; Guo, W. EZH2: A Novel Target for Cancer Treatment. J. Hematol. Oncol. 2020, 13, 104. [Google Scholar] [CrossRef]
  60. Goleij, P.; Heidari, M.M.; Tabari, M.A.K.; Hadipour, M.; Rezaee, A.; Javan, A.; Sanaye, P.M.; Larsen, D.S.; Daglia, M.; Khan, H. Polycomb Repressive Complex 2 (PRC2) Pathway’s Role in Cancer Cell Plasticity and Drug Resistance. Funct. Integr. Genom. 2025, 25, 1–26. [Google Scholar] [CrossRef]
  61. Marin, J.J.G.; Sanchez De Medina, F.; Castão, B.; Bujanda, L.; Romero, M.R.; Martinez-Augustin, O.; Del Moral-Avila, R.; Briz, O. Chemoprevention, Chemotherapy, and Chemoresistance in Colorectal Cancer. Drug Metab. Rev. 2012, 44, 148–172. [Google Scholar] [CrossRef]
  62. Parri, M.; Chiarugi, P. Rac and Rho GTPases in Cancer Cell Motility Control. Cell Commun. Signal. 2010, 8, 23. [Google Scholar] [CrossRef]
  63. Xin, B.; Tune, J.; Sim, M.S.; Poh, C.L.; Guad, R.M.; Woon, C.K.; Hazarika, I.; Das, A.; Gopinath, S.C.B.; Rajan, M.; et al. Review Article Matrix Metalloproteinases in Chemoresistance: Regulatory Roles, Molecular Interactions, and Potential Inhibitors. J. Oncol. 2022, 2022, 3249766. [Google Scholar] [CrossRef]
  64. Liu, J.; Li, Y.; Lian, X.; Zhang, C.; Feng, J.; Tao, H.; Wang, Z. Potential Target within the Tumor Microenvironment—MT1-MMP. Front. Immunol. 2025, 16, 1517519. [Google Scholar] [CrossRef] [PubMed]
  65. Ou, S.; Chen, H.; Wang, H.; Ye, J.; Liu, H.; Tao, Y.; Ran, S.; Mu, X.; Liu, F.; Zhu, S.; et al. Fusobacterium Nucleatum Upregulates MMP7 to Promote Metastasis-Related Characteristics of Colorectal Cancer Cell via Activating MAPK(JNK)-AP1 Axis. J. Transl. Med. 2023, 21, 704. [Google Scholar] [CrossRef]
  66. Uitto, V.J.; Baillie, D.; Wu, Q.; Gendron, R.; Grenier, D.; Putnins, E.E.; Kanervo, A.; Firth, J.D. Fusobacterium Nucleatum Increases Collagenase 3 Production and Migration of Epithelial Cells. Infect. Immun. 2005, 73, 1171. [Google Scholar] [CrossRef]
  67. Ababneh, O.; Nishizaki, D.; Kato, S.; Kurzrock, R. Tumor Necrosis Factor Superfamily Signaling: Life and Death in Cancer. Cancer Metastasis Rev. 2024, 43, 1137–1163. [Google Scholar] [CrossRef]
  68. Friedl, J.; Puhlmann, M.; Bartlett, D.L.; Libutti, S.K.; Turner, E.N.; Gnant, M.F.X.; Richard Alexander, H. Induction of Permeability across Endothelial Cell Monolayers by Tumor Necrosis Factor (TNF) Occurs via a Tissue Factor–Dependent Mechanism: Relationship between the Procoagulant and Permeability Effects of TNF. Blood 2002, 100, 1334–1339. [Google Scholar] [CrossRef]
  69. Zhang, Z.; Lin, G.; Yan, Y.; Li, X.; Hu, Y.; Wang, J.; Yin, B.; Wu, Y.; Li, Z.; Yang, X.P. Transmembrane TNF-Alpha Promotes Chemoresistance in Breast Cancer Cells. Oncogene 2018, 37, 3456–3470. [Google Scholar] [CrossRef]
  70. Olszewski, U.; Liedauer, R.; Ausch, C.; Thalhammer, T.; Hamilton, G. Overexpression of CYP3A4 in a COLO 205 Colon Cancer Stem Cell Model in Vitro. Cancers 2011, 3, 1467–1479. [Google Scholar] [CrossRef]
  71. Taya, M.; Merenbakh-Lamin, K.; Zubkov, A.; Honig, Z.; Kurolap, A.; Mayer, O.; Shomron, N.; Wolf, I.; Rubinek, T. Beyond Endocrine Resistance: Estrogen Receptor (ESR1) Activating Mutations Mediate Chemotherapy Resistance through the JNK/c-Jun MDR1 Pathway in Breast Cancer. Breast Cancer Res. Treat. 2024, 209, 431–449. [Google Scholar] [CrossRef]
  72. Topi, G.; Satapathy, S.R.; Ghatak, S.; Hellman, K.; Ek, F.; Olsson, R.; Ehrnström, R.; Lydrup, M.L.; Sjölander, A. High Oestrogen Receptor Alpha Expression Correlates with Adverse Prognosis and Promotes Metastasis in Colorectal Cancer. Cell Commun. Signal. 2024, 22, 198. [Google Scholar] [CrossRef] [PubMed]
  73. Scheinfeld, N. A Comprehensive Review and Evaluation of the Side Effects of the Tumor Necrosis Factor Alpha Blockers Etanercept, Infliximab and Adalimumab. J. Dermatol. Treat. 2004, 15, 280–294. [Google Scholar] [CrossRef]
  74. Yap, T.A.; Winter, J.N.; Giulino-Roth, L.; Longley, J.; Lopez, J.; Michot, J.M.; Leonard, J.P.; Ribrag, V.; McCabe, M.T.; Creasy, C.L.; et al. Phase I Study of the Novel Enhancer of Zeste Homolog 2 (EZH2) Inhibitor GSK2816126 in Patients with Advanced Hematologic and Solid Tumors. Clin. Cancer Res. 2019, 25, 7331–7339. [Google Scholar] [CrossRef]
  75. Alexopoulou, L.; Holt, A.C.; Medzhitov, R.; Flavell, R.A. Recognition of Double-Stranded RNA and Activation of NF-ΚB by Toll-like Receptor 3. Nature 2001, 413, 732–738. [Google Scholar] [CrossRef] [PubMed]
  76. Castro, F.; Cardoso, A.P.; Gonçalves, R.M.; Serre, K.; Oliveira, M.J. Interferon-Gamma at the Crossroads of Tumor Immune Surveillance or Evasion. Front. Immunol. 2018, 9, 337231. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, L.; Leng, X.X.; Qi, J.; Wang, N.; Han, J.X.; Tao, Z.H.; Zhuang, Z.Y.; Ren, Y.; Xie, Y.L.; Jiang, S.S.; et al. The Adhesin RadD Enhances Fusobacterium Nucleatum Tumour Colonization and Colorectal Carcinogenesis. Nat. Microbiol. 2024, 9, 2292–2307. [Google Scholar] [CrossRef] [PubMed]
  78. Xu, C.; Fan, L.; Lin, Y.; Shen, W.; Qi, Y.; Zhang, Y.; Chen, Z.; Wang, L.; Long, Y.; Hou, T.; et al. Fusobacterium Nucleatum Promotes Colorectal Cancer Metastasis through MiR-1322/CCL20 Axis and M2 Polarization. Gut Microbes 2021, 13, 1980347. [Google Scholar] [CrossRef]
  79. Zheng, X.; Liu, R.; Zhou, C.; Yu, H.; Luo, W.; Zhu, J.; Liu, J.; Zhang, Z.; Xie, N.; Peng, X.; et al. ANGPTL4-Mediated Promotion of Glycolysis Facilitates the Colonization of Fusobacterium Nucleatum in Colorectal Cancer. Cancer Res. 2021, 81, 6157. [Google Scholar] [CrossRef]
Figure 1. Dataset selection pipeline. After searching the NCBI GEO DataSets Database, filtering with our selected criteria, and removal of duplicates or inaccessible datasets, a final selection of four datasets continued with further analysis: GSE2454617, GSE173549, GSE90944, and GSE175593. n indicates the number of datasets.
Figure 1. Dataset selection pipeline. After searching the NCBI GEO DataSets Database, filtering with our selected criteria, and removal of duplicates or inaccessible datasets, a final selection of four datasets continued with further analysis: GSE2454617, GSE173549, GSE90944, and GSE175593. n indicates the number of datasets.
Cancers 17 03247 g001
Figure 2. (a) Pathways related to cancer processes. There were enrichments in cancer-related pathways such as pathogen-induced cytokine storm signaling, irritable bowel syndrome signaling, interleukin-10 signaling, colorectal cancer metastasis signaling, toll-like receptor signaling, TNF signaling, and autophagy. However, oncogenic pathways PI3K/AKT signaling, WNT/β-catenin signaling, and NK-κB signaling were insignificant or inconclusive; (b) Graphical summary of the combined dataset. This summary indicates the most significant pathways, the most activated or inhibited pathways, key molecules and interactions, and predicted biological effects, providing a high-level overview of the dataset. Various cytokines have been identified, along with the biological processes involved in inflammation, immune dysregulation, proliferation, and tissue damage.
Figure 2. (a) Pathways related to cancer processes. There were enrichments in cancer-related pathways such as pathogen-induced cytokine storm signaling, irritable bowel syndrome signaling, interleukin-10 signaling, colorectal cancer metastasis signaling, toll-like receptor signaling, TNF signaling, and autophagy. However, oncogenic pathways PI3K/AKT signaling, WNT/β-catenin signaling, and NK-κB signaling were insignificant or inconclusive; (b) Graphical summary of the combined dataset. This summary indicates the most significant pathways, the most activated or inhibited pathways, key molecules and interactions, and predicted biological effects, providing a high-level overview of the dataset. Various cytokines have been identified, along with the biological processes involved in inflammation, immune dysregulation, proliferation, and tissue damage.
Cancers 17 03247 g002
Figure 3. Pathogen-induced cytokine storm signaling pathway. Innate and adaptive immune pathways activated by pathogen-associated molecular patterns (PAMPs), cytokines, and chemokines. Upstream signals such as TNF, IL-6, IRF1, IRF9, and NF-κB stimulate downstream cascades that control tissue integrity disruption, cytokine production, cell recruitment, apoptosis, macrophage activation, and pro-inflammatory responses. Specifically, cytokines and chemokines TNF, IL1B, CCL2, CCL5, CCL20, CXCL8, CXCL1, and CXCL10 were differentially expressed. * next to a gene name indicates that node is a composite of multiple genes mapping to the same name in the dataset. C next to a line indicates that connection is a manually curated finding from scientific literature, while E next to a line represents a connection that was made through automated curation with secondary review by an expert.
Figure 3. Pathogen-induced cytokine storm signaling pathway. Innate and adaptive immune pathways activated by pathogen-associated molecular patterns (PAMPs), cytokines, and chemokines. Upstream signals such as TNF, IL-6, IRF1, IRF9, and NF-κB stimulate downstream cascades that control tissue integrity disruption, cytokine production, cell recruitment, apoptosis, macrophage activation, and pro-inflammatory responses. Specifically, cytokines and chemokines TNF, IL1B, CCL2, CCL5, CCL20, CXCL8, CXCL1, and CXCL10 were differentially expressed. * next to a gene name indicates that node is a composite of multiple genes mapping to the same name in the dataset. C next to a line indicates that connection is a manually curated finding from scientific literature, while E next to a line represents a connection that was made through automated curation with secondary review by an expert.
Cancers 17 03247 g003
Figure 4. Antiviral pathways. This figure illustrates the interconnected pathways activated during the antiviral immune response. Key upstream signals such as TNF, IFN-γ, and pattern recognition receptors (e.g., IFNGR, RIG-I) initiate downstream cascades that lead to the activation of transcription factors, including NF-κB, IRF3, IRF1, and STATs. These pathways regulate the expression of genes involved in antiviral responses, apoptosis, and innate immunity. Activation of key effectors promotes antiviral defense mechanisms, such as the induction of interferons.
Figure 4. Antiviral pathways. This figure illustrates the interconnected pathways activated during the antiviral immune response. Key upstream signals such as TNF, IFN-γ, and pattern recognition receptors (e.g., IFNGR, RIG-I) initiate downstream cascades that lead to the activation of transcription factors, including NF-κB, IRF3, IRF1, and STATs. These pathways regulate the expression of genes involved in antiviral responses, apoptosis, and innate immunity. Activation of key effectors promotes antiviral defense mechanisms, such as the induction of interferons.
Cancers 17 03247 g004
Table 1. Chemoresistance genes identified in IPA to be enriched or inhibited when co-cultured with F. nucleatum.
Table 1. Chemoresistance genes identified in IPA to be enriched or inhibited when co-cultured with F. nucleatum.
Genelog2FCExpr
p-Value
LocationFamily
BIRC32.8466.92 × 10−5Cytoplasmenzyme
CEBPB0.6900.0309Nucleustranscription regulator
CERS4−3.0020.0117Cytoplasmtranscription regulator
CSAG2−4.6650.00310Otherother
CYP3A41.5360.0143Cytoplasmenzyme
ESR11.8050.0398Nucleusligand-dependent nuclear receptor
IL71.3990.00405Extracellular Spacecytokine
KISS10.6020.0667Cytoplasmother
LGALS1−1.4910.0502Extracellular Spaceother
MAST1−1.8100.0125Cytoplasmkinase
MDK−1.8960.00187Extracellular Spacegrowth factor
MIR100HG−2.3000.0631Otherother
NFKBIA1.3090.0211Cytoplasmtranscription regulator
PHGDH0.4170.0427Cytoplasmenzyme
SOD20.7330.0881Cytoplasmenzyme
TMEM40−2.0150.0452Otherother
TNF3.7580.00365Extracellular Spacecytokine
TRIB30.6020.0824Nucleuskinase
TRIM9−2.4730.0154Cytoplasmenzyme
VEGFA0.6910.0951Extracellular Spacegrowth factor
Table 2. Top 20 drugs recognized in causal networks to be activated or inhibited. The activation z-score predicts the likely activation or inhibition of a pathway or regulator based on the direction and consistency of gene expression changes, while the p-value quantifies the statistical significance of the overlap between the dataset and known pathway targets.
Table 2. Top 20 drugs recognized in causal networks to be activated or inhibited. The activation z-score predicts the likely activation or inhibition of a pathway or regulator based on the direction and consistency of gene expression changes, while the p-value quantifies the statistical significance of the overlap between the dataset and known pathway targets.
Compound or DrugMolecule TypePredicted ActivationActivation z-Scorep-Value
etanerceptbiologic drugInhibited−5.4221.18 × 10−31
adalimumabbiologic drugInhibited−6.0754.12 × 10−31
infliximabbiologic drugInhibited−6.1395.08 × 10−31
tetrandrinechemical drugInhibited−6.0227.91 × 10−31
poly rI:rC-RNAbiologic drugActivated6.4014.1 × 10−25
GSK583chemical drugInhibited−4.7372.88 × 10−22
ferric hexacyanoferrate(II)chemical drugInhibited−4.5893.66 × 10−22
SKLB023chemical reagentInhibited−4.8517.64 × 10−22
dexamethasonechemical drugInhibited−3.4024.1 × 10−21
GSK2816126chemical drugInhibited−3.2131.7 × 10−20
ML385chemical reagentActivated3.5231.75 × 10−20
L 655238chemical reagentInhibited−4.51.9 × 10−20
MDK4882chemical reagentInhibited−4.4812.23 × 10−20
poly dA-dTchemical reagentActivated4.7817.24 × 10−20
2-(4-acetoxyphenyl)-2-chloro-N-methylethylaminechemical reagentInhibited−5.6577.96 × 10−20
manumycin Achemical reagentInhibited−2.839.08 × 10−20
epicatechinchemical drugInhibited−5.5849.19 × 10−20
imipramine bluechemical drugInhibited−4.7379.54 × 10−20
indoxamchemical reagentInhibited−4.9931.16 × 10−19
1-docosapentaenoylglycerolchemical reagentInhibited−4.2581.21 × 10−19
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Risoen, K.R.; Shaw, C.A.; Chien, J.; Weimer, B.C. Fusobacterium nucleatum and Its Impact on Colorectal Cancer Chemoresistance: A Meta-Analysis of In Vitro Co-Culture Infections. Cancers 2025, 17, 3247. https://doi.org/10.3390/cancers17193247

AMA Style

Risoen KR, Shaw CA, Chien J, Weimer BC. Fusobacterium nucleatum and Its Impact on Colorectal Cancer Chemoresistance: A Meta-Analysis of In Vitro Co-Culture Infections. Cancers. 2025; 17(19):3247. https://doi.org/10.3390/cancers17193247

Chicago/Turabian Style

Risoen, Katie R., Claire A. Shaw, Jeremy Chien, and Bart C. Weimer. 2025. "Fusobacterium nucleatum and Its Impact on Colorectal Cancer Chemoresistance: A Meta-Analysis of In Vitro Co-Culture Infections" Cancers 17, no. 19: 3247. https://doi.org/10.3390/cancers17193247

APA Style

Risoen, K. R., Shaw, C. A., Chien, J., & Weimer, B. C. (2025). Fusobacterium nucleatum and Its Impact on Colorectal Cancer Chemoresistance: A Meta-Analysis of In Vitro Co-Culture Infections. Cancers, 17(19), 3247. https://doi.org/10.3390/cancers17193247

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop