Abstract
Increasing evidence suggests a significant association between periodontal disease and the occurrence of various cancers. The carcinogenic potential of several periodontal pathogens has been substantiated in vitro and in vivo. This review provides a comprehensive overview of the diverse mechanisms employed by different periodontal pathogens in the development of cancer. These mechanisms induce chronic inflammation, inhibit the host's immune system, activate cell invasion and proliferation, possess anti‐apoptotic activity, and produce carcinogenic substances. Elucidating these mechanisms might provide new insights for developing novel approaches for tumor prevention, therapeutic purposes, and survival improvement.
Keywords: cancer, carcinogenesis, cell invasion, cell proliferation, immune response, inflammation, periodontal pathogens
1. INTRODUCTION
Periodontitis is a complex inflammatory disease with many etiologic and contributing factors. 1 It affects up to 90% of the global population. 2 The pathogenic processes primarily arise from the host's reaction to an oral microbial biofilm. 3 Oral microbiomes contain hundreds of aerobic and anaerobic bacteria that metabolically and physically interact. These interactions create a complex biofilm community. Certain viruses, bacteria, and parasites have been recognized as crucial risk factors for specific types of cancer. 4 Microbes existing in mucosal locations can integrate into the tumor microenvironment, impacting cancer growth and dissemination through various means. This includes influencing cell growth rates and affecting cancer genomic stability, metabolism, and immune responsiveness. 5 The link between periodontal disease and the overall cancer risk is attracting increasing attention. Research has shown a statistically significant correlation between periodontal disease and the risk of esophageal, breast, lung, pancreatic, prostate, colorectal, digestive tract, and head and neck cancers. 6 The complex interaction of periodontal pathogens with the host still warrants further investigation of the underlying mechanisms. This article reviews the properties of periodontal pathogens in association with cancer development.
2. PERIODONTAL PATHOGENS
The oral microbiota is a substantial component of the human microbiota, comprising several hundreds to several thousands of diverse species. 7 The oral cavity is inhabited by almost 700 bacterial species, which are located in saliva, soft tissues like mucosa and tongue surface, hard tissues, and hard materials. 7 , 8 The Human Oral Microbiome Database (HOMD) lists 774 oral bacterial species and 2123 genomes representing 539 taxa (https://www.homd.org/). The oral cavity harbors a variety of bacterial genera, including Treponema, Bacteroides, Porphyromonas, Prevotella, Capnocytophaga, Peptostreptococcus, Fusobaterium, Actinobacillus, and Eikenella. 9 Notable high‐risk periodontal pathogens associated with periodontitis include Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans), Porphyromonas gingivalis (P. gingivalis), Tannerella forsythia (T. forsythia), Treponema denticola (T. denticola), and Fusobacterium nucleatum (F. nucleatum). 10
Head and neck squamous cell carcinoma (HNSCC) originates from the squamous epithelium of the upper aerodigestive tract, with over 800 000 new cases and over 400 000 deaths in 2020, making it the most common cancer in this specific region. 11 Cancers of the oral cavity and oropharynx constitute the majority of cases of HNSCC. 12 Notably, oral squamous cell carcinoma (OSCC) is a significant contributor to morbidity and mortality in patients with head and neck cancers. 13 Globally, an estimated 377 713 new cases and 177 757 fatalities attributed to oral cavity cancer (including lip cancer) were reported in 2020. 11 Primary risk factors associated with the development of oral cavity cancer include inadequate oral hygiene, smoking, alcohol consumption, utilization of smokeless tobacco, and human papillomavirus (HPV) infection. 14 , 15 , 16
Infection, chronic inflammation, or autoimmunity precede around 15%–20% of all cancer cases. 17 , 18 , 19 Chronic inflammation is primarily induced by bacterial infection. Associations between oral cancer and epithelial precursor lesions and bacteria from Fusobacterium, Veillonella, Actinomyces, Clostridium, Haemophilus, and Enterobacteriaceae genera have been established. 20 Chang et al. 21 tested 61 cancer tissues and their adjacent paracancerous tissues and 30 normal oral tissues. The findings indicated elevated levels of P. gingivalis and F. nucleatum in cancer tissue compared to normal tissues, consistent with the results of Nagy's and Schmidt. 20 , 22
Dating back to the 1980s, Marshall and Warren's 23 publication provided the initial evidence of bacteria playing a direct role in cancer development. Figure 1 illustrates the timeline of notable discoveries and milestones in cancer microbiome research. 24 In the 19th century, the German pathologist Virchow proposed that inflammation was a promoter of carcinogenesis. 25 Inflammation triggered by microbes, although protective against pathogens, can paradoxically cause substantial secondary damage to host tissues. If this inflammation persists, it has the potential to lead to tissue fibrosis and carcinogenesis. 26 In vitro and in vivo studies have illustrated the carcinogenic potential of numerous bacteria. Enzymatic attacks by bacterial products have the capacity to inflict DNA damage, eliciting an inflammatory response that generates free radicals and may impact DNA repair mechanisms. Disruption of signaling by bacterial products can disturb the delicate equilibrium of growth, cell division, and apoptosis, potentially providing support for tumor promotion. 27 Despite most developments in understanding HNSCCs occurring in recent decades, clinically meaningful discoveries have yet to be fully realized, as depicted in Figure 2. 28
FIGURE 1.

Timeline: Some significant discoveries and events in cancer microbiome research. Reprinted from Ref. [24].
FIGURE 2.

Timeline of molecular characterization, therapeutic innovations in head and neck cancers, and future perspectives. 28
3. THE ASSOCIATION BETWEEN PERIODONTAL PATHOGENS AND CANCER
Substantial evidence highlights the connection between alterations in the oral microbiome and specific cancer types (see Table 1, Figure 3). Breast cancer (BC) is the most commonly diagnosed cancer and the leading cause of cancer mortality in women. By analyzing the microbiota of breast skin tissue, breast skin swabs, and buccal swabs, Hieken et al. 29 identified the malignancy correlated with enrichment in taxa of lower abundance including the genera Fusobacterium, Atopobium, Gluconacetobacter, Hydrogenophaga, and Lactobacillus. In another study, Wang et al. 30 reported that Methylobacterium was decreased in cancer patients relative to non‐cancer patient samples. Breast cancer patients had increased levels of gram‐positive organisms including Corynebacterium, Staphylococcus, Actinomyces, and Propionibacteriaceae in urine. Parhi et al. 31 showed that the occurrence of F. nucleatum gDNA in breast cancer samples correlates with high Gal‐GalNAc levels.
TABLE 1.
Detection of oral microbiota in cancerous sites.
| Cancer type | Oral microbiome | Study design | Sample type | Numbers of samples | Microbial assessment platform | Major findings | References |
|---|---|---|---|---|---|---|---|
| Breast cancer | Fusobacterium, Atopobium, Gluconacetobacter, Hydrogenophaga, Lactobacillus | Prospective cohort | Breast skin tissue, breast skin swabs, and buccal swabs | Invasive breast cancer (n = 15), benign breast disease without atypia (n = 13) | 16S rDNA hypervariable tag sequencing | Malignancy correlated with enrichment in taxa of lower abundance including the genera Fusobacterium, Atopobium, Gluconacetobacter, Hydrogenophaga and Lactobacillus | Hieken et al. 29 |
| Methylobacterium, Corynebacterium, Staphylococcus, Actinomyces, Propionibacteriaceae | Cross‐sectional | Urine, oral rinse and surgically collected breast tissue | Invasive breast cancer patients (n = 57), healthy contorls (n = 21) | 16S rRNA gene sequencing |
There were no significant differences in oral rinse samples. Methylobacterium was decreased in cancer relative to non‐cancer patient samples. Independent of menopausal status, however, cancer patients had increased levels of gram‐positive organisms including Corynebacterium, Staphylococcus, Actinomyces, and Propionibacteriaceae in urine |
Wang et al. 30 | |
| F. nucleatum | Cross‐sectional | FFPE samples from breast tumors, fresh forzen colon tumors | 50 Breast tumors, 21 colon tumors | 16S rRNA gene sequencing, flow cytometry, immunofluorescence microscopy | Gal‐GalNAc levels increase as human BC progresses, and that occurrence of F. nucleatum gDNA in BC samples correlates with high Gal‐GalNAc levels | Parhi et al. 31 | |
| Esophageal cancer | T. denticola, S. mitis, S. anginousus | Cross‐sectional | Tissue, saliva | 20 Esophageal cancer tissues, 20 healthy volunteers | 16S rRNA gene sequencing | T. denticola, S. mitis, and S. anginousus could have significant roles in the carcinogenic process of many cases of esophageal cancer by causing inflammation and by promoting the carcinogenic process, and that eradication of these three bacteria may decrease the risk of recurrence | Narikiyo et al. 33 |
| P. gingivalis | Cross‐sectional | Tissue | 100 patients, 30 controls | Immunohistochemistry, qRT‐PCR | P. gingivalis infects the epithelium of the esophagus of ESCC patients, establish an association between infection with P. gingivalis and the progression of ESCC, and suggest P. gingivalis infection could be a biomarker for this disease | Gao et al. 35 | |
| F. nucleatum | Longitudinal cohort | Tissue | 325 FFPE EC specimens, 60 paired adjacent non‐tumor tissues | Immunohistochemistry, qPCR | F. nucleatum in esophageal cancer tissues was associated with shorter survival, suggesting a potential role as a prognostic biomarker. F. nucleatum might also contribute to aggressive tumor behavior through activation of chemokines, such as CCL20 | Yamamura et al. 34 | |
| T. forsythia, Neisseria, Streptococcus pneumoniae, P. gingivalis | Prospective cohort | Pre‐diagnostic oral wash sample | n = 81/160 EAC/matched controls, n = 25/50 ESCC/matched controls | 16S rRNA gene sequencing | T. forsythia to be associated with higher risk of EAC. The commensal genus Neisseria and the species Streptococcus pneumoniae were associated with lower EAC risk. Lastly, the abundance of the periodontal pathogen P. gingivalis trended with higher risk of ESCC | Peters et al. 65 | |
| A. actinomycetemcomitans, F. nucleatum, P. gingivalis, P. intermedia, T. forsythia, T. denticola, S. anginosus | Cross‐sectional | Subgingival dental plaque and unstimulated saliva | 61 Patients, 62 matched controls | RT‐PCR |
In subgingival plaque, A. actinomycetemcomitans, P. gingivalis, P. intermedia, T. forsythia, T. denticola, and S. anginosus were more abundant and prevalent in the cancer group versus the control group A. actinomycetemcomitans and S. anginosus were more abundant or prevalent in the salivary microbiota of the cancer group The prevalence of T. forsythia and S. anginosus in dental plaque and of A. actinomycetemcomitans in saliva, as well as a drinking habit, were associated with a high risk of esophageal cancer |
Kawasaki et al. 36 | |
| Bladder cancer, colorectal cancer | T. forsythia, T. denticola | Prospective cohort | Blood sample | Among the 621 participants with no prior cancer diagnoses, 221 men developed cancer | ELISA | Lowest levels of antibodies for the two oral bacteria T. forsythia and T. denticola had a higher risk of bladder cancer. Low levels of T. denticola were also associated with increased risk of colon cancer | Håheim et al. 37 |
| Colorectal cancer | F. nucleatum | Cross‐sectional | Tissue | 9 Tumor/normal pairs | qPCR, 16s rRNA gene sequencing, FISH | Fusobacterium sequences were enriched in CRC | Kostic et al. 38 |
| Fusobacterium, Porphyromonas | Cross sectional | Fecal | 47 CRC case subjects and 94 control subjects | 16S rRNA sequencing | Increased carriage of Fusobacterium and Porphyromonas were found in case subjects compared with control subjects | Ahn et al. 40 | |
| Fusobacterium, B. fragilis, Gemella, Peptostreptococcus, Parvimonas | Cross‐sectional | Tissue | 47 Paired samples of adenoma and adenoma‐adjacent mucosae, 52 paired samples of carcinoma and carcinoma‐adjacent mucosae and 61 healthy controls | 16S rRNA gene sequencing | In early‐stage CRC, Fusobacterium, Parvimonas, Gemella and Leptotrichia were most significantly enriched | Nakatsu et al. 42 | |
| F. nucleatum | Prospective cohort | Tissue | 1069 CRCs | PCR | The amount of F. nucleatum DNA in colorectal cancer tissue is associated with shorter survival, and may potentially serve as a prognostic biomarker | Mima et al. 39 | |
| Mogibacterium | Cross sectional | Fecal | 233 Adenomas, 547 without adenomas | 16S rRNA gene sequencing | Multiple taxa were significantly more abundant in patients with adenomas, including Bilophila, Desulfovibrio, proinflammatory bacteria in the genus Mogibacterium, and multiple Bacteroidetes species. Patients without adenomas had greater abundances of Veillonella, Firmicutes (Order Clostridia), and Actinobacteria (family Bifidobacteriales) | Hale et al. 43 | |
| F. nucleatum, Peptostreptococcus stomatitis, and Parvimonas micra | Cross sectional | Oral swab, colonic mucosae, stool | CRC (99 subjects), colorectal polyps (32) or controls (103) | 16S rRNA gene sequencing | Combining the data from fecal microbiota and oral swab microbiota increased the sensitivity of the model to 76% (CRC)/88% (polyps) | Flemer et al. 44 | |
| P. gingivalis | Cross sectional | Fecal and mucosal samples | 247 CRC patients and 89 control subjects | qRT‐PCR | P. gingivalis could be linked to patients with CRC and to a worse patient prognosis | Kerdreux et al. 41 | |
| Gastric cancer | P. stomatis, D. pneumosintes, S. exigua, P. micra, S. anginosus | Cross sectional | Gastric mucosal | 81 Cases including superficial gastritis, atrophic gastritis, intestinal metaplasia and gastric cancer | 16S rRNA sequencing | P. stomatis, D. pneumosintes, S. exigua, P. micra and S. anginosus play important roles in GC progression | Coker et al. 47 |
| Peptostreptococcus, Streptococcus, Fusobacterium | Cross sectional | Tissue | 62 Pairs of matched GC and adjacent non‐cancerous | 16S rRNA gene sequencing | The bacterial taxa enriched in the cancer samples were predominantly represented by oral bacteria (such as Peptostreptococcus, Streptococcus, and Fusobacterium), while lactic acid‐producing bacteria (such as Lactococcus lactis and Lactobacillus brevis) were more abundant in adjacent non‐tumor tissues | Chen et al. 46 | |
| Pancreatic cancer | Neisseria elongata, Streptococcus mitis | Cross sectional | Saliva |
Discovery phase: 10 PCs and 10 healthy controls Validation phase: 28 PCs, 28 healthy controls, and 27 CPs |
qPCR | Neisseria elongata and Streptococcus mitis shown significant variation between patients with pancreatic cancer and controls. The combination of two bacterial biomarkers (N. elongata and S. mitis) yielded a receiver operating characteristic plot area under the curve value of 0.90 (95% CI 0.78 to 0.96, p < 0.0001) with a 96.4% sensitivity and 82.1% specificity in distinguishing patients with pancreatic cancer from healthy subjects | Farrell et al. 52 |
| P. gingivalis | Prospective cohort | Blood samples | 405 PCs and 416 matched controls | Immunoblot array | Individuals with high levels of antibodies against Porphyromonas gingivalis ATTC 53978 had a twofold higher risk of PC than individuals with lower levels of these antibodies | Michaud et al. 49 | |
| P. gingivalis, A. actinomycetemcomitans | Prospective cohort | Pre‐diagnostic oral wash samples | 361 PCs and 371 matched controls | 16S rRNA gene sequencing | P. gingivalis and A. actinomycetemcomitans were associated with higher risk of PC. Phylum Fusobacteria and its genus Leptotrichia were associated with decreased pancreatic cancer risk | Fan et al. 50 | |
| Haemophilus, Enterobacteriaceae, Lachnospiraceae G7, Bacteroidaceae, Staphylococcaceae | Case–control | Saliva | 273 PCs and 285 controls | 16S rRNA gene sequencing | Increasing relative levels of Haemophilus were associated with decreased odds of pancreatic cancer, while the presence of Enterobacteriaceae, Lachnospiraceae G7, Bacteroidaceae, or Staphylococcaceae were associated with increased odds of pancreatic cancer | Vogtmann et al. 51 | |
| Liver cancer | Oribacterium, Fusobacterium | Cross sectional | Tongue coat | 35 Liver cancers, 25 healthy controls | 16S rRNA gene sequencing | Oribacterium and Fusobacterium could distinguish liver cancer patients from healthy subjects | Lu et al. 53 |
| Lung cancer | Capnocytophaga, Veillonella | Cross sectional | Saliva |
Discovery phase: 20 lung cancer and 10 controls Validation phase: 41 lung cancer and 15 controls |
16S rRNA gene sequencing, qPCR | The levels of Capnocytophaga and Veillonella were significantly higher in the saliva from lung cancer patients, which may serve as potential biomarkers for the disease detection/classification | Yan et al. 54 |
| Streptococcus, Veillonella | Cross sectional | Airway brushings | 39 Lung cancer, 36 non‐cancer, and 10 healthy controls | 16S rRNA gene sequencing | A higher abundance of Streptococcus and Veillonella was enriched in the lower airways of lung cancer patients | Tsay et al. 55 | |
| Veillonella, Streptococcus, Lautropia, Leptotrichia, Rothia, Aggregatibacter, Fusobacterium, Prevotella, Bacteroides, Faecalibacterium | Cross sectional | Saliva, blood | 39 NSCLCs and 20 healthy controls | 16S rRNA gene sequencing, ELISA | The relative abundance of Veillonella, Streptococcus, Lautropia, Leptotrichia, Rothia, and Aggregatibacter was significantly higher in the NSCLC group compared with the healthy control group. Additionally, the relative abundances of Fusobacterium, Prevotella, Bacteroides, and Faecalibacterium in NSCLC group were generally decreased | Zhang et al. 56 | |
| P. gingivalis | Prospective cohort | Tissue | 100 Patients with small cell lung cancer, 119 patients with lung adenocarcinoma and 100 patients with lung squamous cell carcinoma | Immunohistochemistry | The colonization rate of P. gingivalis in carcinoma tissues was significantly higher than that in adjacent lung tissues. And the survival rate and median survival time of patients with P. gingivalis infection were significantly shortened | Liu et al. 57 | |
| Head and neck squamous cell carcinomas | Capnocytophaga gingivalis, Prevotella melaninogenica, Streptococcus mitis | Cross sectional | Saliva | 229 OSCC‐free and 45 OSCC subjects | Checkerboard DNA–DNA hybridization | Capnocytophaga gingivalis, Prevotella melaninogenica and Streptococcus mitis, were elevated in the saliva of individuals with OSCC | Mager et al. 61 |
| P. gingivalis | Cross sectional | Tissue | 10 Gingival squamous cell carcinoma, 5 normal gingiva | Immunohistochemical staining | P. gingivalis is abundantly present in malignant oral epithelium suggesting a potential association of the bacteria with gingival squamous cell carcinoma | Katz et al. 62 | |
| Campylobacter, Eikenella, Alloprevotella, Fusobacterium, Selenomonas, Dialister, Peptostreptococcus, Filifactor, Peptococcus, Catonella, Parvimonas, Capnocytophaga, Peptostreptococcaceae | Cross sectional | Oral swab | 40 OSCC lesion and 40 anatomically matched normal sites | 16S rRNA gene sequencing | Campylobacter, Eikenella, Alloprevotella, Fusobacterium, Selenomonas, Dialister, Peptostreptococcus, Filifactor, Peptococcus, Catonella, Parvimonas, Capnocytophaga, and Peptostreptococcaceae was significantly enriched in OSCC samples. Additionally, several operational taxonomic units (OTUs) associated with Fusobacterium were highly involved in OSCC and demonstrated good diagnostic power | Zhao et al. 60 | |
| F. nucleatum subsp. Polymorphum, P. aeruginosa, Campylobacter sp.oral taxon 44 | Cross sectional | Tissue and swabs | 20 Fresh OSCC and 20 deep‐epithelium swabs matched control subjects | 16S rRNA gene sequencing |
F. nucleatum subsp. polymorphum was the most significantly overrepresented species in the tumors followed by P. aeruginosa and Campylobacter sp. oral taxon 44, while Streptococcus mitis, Rothia mucilaginosa, and Haemophilus parainfluenzae were the most significantly abundant in the controls. Functional prediction showed that genes involved in bacterial mobility, flagellar assembly, bacterial chemotaxis, and LPS synthesis were enriched in the tumors while those responsible for DNA repair and combination, purine metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, ribosome biogenesis, and glycolysis/gluconeogenesis were significantly associated with the controls |
AI‐Hebshi et al. 63 | |
| Acinetobacter, Fusobacterium, Streptococcus, Prevotella | Cross sectional | Tissue, saliva, mouthwash | 30 OSCC | 16S rRNA gene sequencing | Proteobacteria were most enriched in the tissue group at the phylum level, while Firmicutes were predominant in the groups saliva and mouthwash. At the genus level, the predominant taxa of group tissue were Acinetobacter and Fusobacterium, and for group saliva and mouthwash, the predominant taxa were Streptococcus and Prevotella. The genera related to late stage tumors were Acinetobacter and Fusobacterium, suggesting microbiota may be implicated in OSCC developing | Zhang et al. 59 | |
| F. nucleatum, Prevotella intermedia, Aggregatibacter segnis, Capnocytophaga leadbetteri, Peptostreptococcus stomatis, Catonella morbi, Porphyromonas catoniae, Campylobacter rectus, Gemella morbillorum, Peptococcus sp. | Cross sectional | Tissue | 50 OSCC | 16S rDNA sequencing | At the species level, the abundances of F. nucleatum, Prevotella intermedia, Aggregatibacter segnis, Capnocytophaga leadbetteri, Peptostreptococcus stomatis, and another five species were significantly increased, suggesting a potential association between these bacteria and OSCC | Zhang et al. 64 |
Abbreviations: BC, breast cancer; CCL, chemokine (C‐C motif) ligand; CP, chronic pancreatitis; CRC, colorectal cancer; EAC, esophageal adenocarcinoma; EC, esophageal cancer; ELISA, enzyme‐linked immunosorbent assay; ESCC, esophageal squamous cell carcinoma; FFPE, formalin‐fixed, paraffin‐embedded; FISH, fluorescence in situ hybridization; GC, gastric cancer; HNSCC, head and neck squamous cell carcinomas; LPS, lipopolysaccharide; NSCLC, non‐small cell lung cancer; OSCC, oral squamous cell carcinoma; PC, pancreatic cancer; PDAC, pancreatic adenocarcinoma; qRT‐PCR, quantitative real time polymerase chain reaction.
FIGURE 3.

Oral microbiota associations with common cancers. A schematic representation illustrating the correlation between oral microbiota and various cancer types, including head and neck, colorectal, lung, breast, bladder, pancreatic, gastric, liver, and esophageal cancers.
Esophageal cancer (EC) poses a significant global health challenge, ranking as the seventh most prevalent and sixth most fatal malignancy worldwide. 32 Narikiyo et al. 33 observed a prevalent and frequent infection of the oral periodontopathic spirochete T. denticola, Streptococcus mitis (S. mitis), and Streptococus anginosus (S. anginosus), in EC across various geographic regions. These bacteria may exert notable influence on the carcinogenic process of numerous esophageal cancer cases by instigating inflammation and facilitating carcinogenesis. Also, F. nucleatum and P. gingivalis are reported to be associated with EC and could be the biomarkers for this disease. 34 , 35 Kawasaki et al. 36 evaluated the prevalence and abundance of oral bacteria from subgingival dental plaque and saliva samples in patients with EC and individuals without any cancer. They found that in subgingival plaque, A. actinomycetemcomitans, P. gingivalis, Prevotella intermedia (P. intermedia), T. forsythia, T. denticola, and S. anginosus were more abundant and prevalent in the cancer group versus the control group. A. actinomycetemcomitans and S. anginosus were more abundant or prevalent in the salivary microbiota of the cancer group. The prevalence of T. forsythia and S. anginosus in dental plaque and of A. actinomycetemcomitans in saliva, as well as a drinking habit, was associated with a high risk of EC.
Measurements of antibodies against oral bacteria indicate previous infection and the level of immune response mounted by an individual's immune system. Håheim et al. 37 found that the lowest levels of antibodies for T. forsythia and T. denticola had a higher risk for bladder cancer. Low levels of T. denticola were also associated with increased risk of colon cancer. Moreover, F. nucleatum and P. gingivalis sequences were enriched in colorectal cancer (CRC) and are associated with shorter survival. 38 , 39 , 40 , 41 Besides, other oral microbiota have been reported to be associated with CRC, including Gemella, Mogibacterium, Peptostreptococcus stomatitis, and Parvimonas micra. 42 , 43 , 44
Gastric cancer (GC) is the fifth most common cancer and the third most common cause of cancer death worldwide. 45 The oral bacterial taxa enriched in the GC samples were predominantly represented by oral bacteria (such as Peptostreptococcus, Streptococcus, and Fusobacterium), while lactic acid‐producing bacteria (such as Lactococcus lactis and Lactobacillus brevis) were more abundant in adjacent non‐tumor tissues. 46 Parvimonas micra, Slackia exigua and Dialister also played important roles in GC progression. 47
Pancreatic cancer's (PC) global burden has seen a significant surge in recent decades and is expected to remain a prominent contributor to cancer‐related deaths. 48 P. gingivalis and A. actinomycetemcomitans were associated with a higher risk of PC. 49 , 50 Vogtmann et al. 51 and Farrell et al. 52 measured variations of salivary microbita in PC cases and controls, they found the presence of Enterobacteriaceae, Lachnospiraceae G7, Bacteroidaceae, Staphylococcaceae, Neisseria elongata, and Streptococcus mitis shown significant variation between patients with pancreatic cancer and controls.
The association between oral disease and increased risk of lung cancer (LC) has been reported in lots of studies. In 2016, a study investigated the tongue coat microbiome of liver cancer patients with cirrhosis based on 16S rRNA gene sequencing. The study found Oribacterium and Fusobacterium could distinguish LC patients from healthy subjects. 53 Yan et al. 54 found that the levels of Capnocytophaga and Veillonella were significantly higher in the saliva of LC patients. A higher abundance of Streptococcus and Veillonella was enriched in the lower airways of LC patients. 55 Furthermore, the non‐small cell lung cancer (NSCLC) group exhibited significantly higher relative abundances of Lautropia, Leptotrichia, Rothia, and Aggregatibacter compared to the healthy control group. 56 Lu et al. 57 reported that the colonization rate of P. gingivalis in carcinoma tissues was significantly higher than that in adjacent lung tissues. The survival rate and median survival time of patients with P. gingivalis infection were significantly shortened.
HNSCCs originate from the mucosal epithelium in the oral cavity, pharynx, and larynx, representing the most prevalent malignancies occurring in the head and neck region. 58 At the genus level, the predominant taxa of OSCC tissue and saliva were: Acinetobacter, Fusobacterium, Streptococcus, Prevotella Dialister, Peptococcus, Catonella and Parvimonas. 59 , 60 Capnocytophaga gingivalis, Prevotella melaninogenica and Streptococcus mitis were enriched in the saliva of patients with OSCC and they can predict 80% of cancer cases while excluding 83% of controls in the non‐matched group. 61 Additionally, the most common bacteria associated with periodontitis, P. gingivalis, were abundantly present in malignant oral epithelium. 62 The first epidemiological evidence linking F. nucleatum and Pseudomonas aeruginosa to OSCC was reported by Al‐Hebshi et al. 63 Another cross sectional study involving 50 patients with OSCC using the 16S rDNA sequencing, reported significantly increased abundances of various bacteria, including F. nucleatum, Prevotella intermedia, Aggregatibacter segnis, and others. These findings suggest a potential association between these bacteria and OSCC. 64
4. MECHANISTIC ROLES OF PERIODONTAL PATHOGENS IN VARIOUS CANCERS
This segment of the review provides an in‐depth analysis of the multifaceted roles played by periodontal pathogens, including P. gingivalis, F. nucleatum, A. actinomycetemcomitans, T. denticola, and T. forsythia, in the pathogenesis of various cancers. Table 2 displays their respective roles in tumor proliferation, invasion, metastasis, and modulation of the tumor microenvironment.
TABLE 2.
Peiodontal pathogens and cancer: Summary of mechanistic evidence.
| Periondontal pathogens | Associated cancer | Study design | Sample type | Effect | References |
|---|---|---|---|---|---|
| Porphyromonas gingivalis | CRC | In vitro | Human CRC cell line S1 and murine colon cancer MC38 cells | P. gingivalis can invade cells and promote the proliferation of colorectal cancer cells by activating the MAPK/ERK signaling pathway | Mu et al. 108 |
| CRC | In vitro | CL‐11 | Peptidoglycans of P. gingivalis plays a major role in PD‐L1 up‐regulation in colon cancer cells. In addition, the mechanism of PD‐L1 up‐regulation depends on NOD 1 and NOD 2 and involves activation of RIP2 and MAPK signaling pathways | Adel‐Khattab et al. 107 | |
| CRC | In vitro, animal model, clinical | CRC tissue samples and feces; colorectal adenoma mouse model, orthotopic and subcutaneous colorectal xenograft mouse model | P. gingivalis is associated with a poor prognosis in human colorectal cancer because this bacterium colonizes and becomes enriched in tumor tissue, resulting in activation of the NLRP3 inflammasome in the immune microenvironment, which ultimately promotes colorectal carcinoma progression | Wang et al. 109 | |
| EC | In vitro, animal model, clinical | ESCC samples; human ESCC cell lines Kyse‐410, Kyse‐150, Kyse‐180, and Yes‐2; humanized mouse model for ESCC | Highlighted the importance of reciprocal regulation of B7‐H4– and KDM5B‐related pathways involved at different stages in the lymphocyte compartment during P. gingivalis infection | Yuan et al. 100 | |
| EC | In vitro, clinical | ESCC saliva samples; ESCC cell lines Eca109 and KYSE‐150 | P. gingivalis triggered NF‐ĸB signaling and promoted the proliferation and metastasis of ESCC cells | Meng et al. 103 | |
| EC | In vitro, animal model | Human embryonic kidney 293T (HEK293T) cell line and the human ESCC cell lines KYSE‐30 and KYSE‐150; xenograft models in nude mice | P. gingivalis could promote the proliferation and migration of ESCC cells through the miR‐194/GRHL3/PTEN/Akt negative feedback signaling pathway mediated by miR‐194 | Liang et al. 104 | |
| EC | In vitro, animal model, clinical | ESCC samples; human ESCC cell lines CE81T and TE2; 4NQO‐induced ESCC model in mice | P. gingivalis may promote esophageal cancer development and progression. Direct targeting of P. gingivalis or concomitant IL‐6 signaling may be a promising strategy to prevent and/or treat ESCC associated with P. gingivalis infection | Chen et al. 102 | |
| EC | In vitro, clinical | ESCC samples; ESCC cell lines KYSE‐30, KYSE‐70, KYSE‐140, and KYSE‐150 | P. gingivalis infection reduces the sensitivity of ESCC cells to chemotherapy drugs through activation of the STAT3 signaling pathway both in vivo and in vitro | Gao et al. 105 | |
| EC | In vitro, animal model, clinical | ESCC samples; Human ESCC cell lines NE6‐T and KYSE30; mouse xenograft tumor model | P. gingivalis activates the TGF‐β signaling pathway through GARP via upregulation of TLR4/MyD88, at least partially, in ESCC cells. FimA contributed partly to the tumor‐promoting role of P. gingivalis through TGF‐β/Smad signaling. The clinical data showed that P. gingivalis infection and the phosphorylation of Smad2/3 were significantly associated with the overall survival of patients with ESCC. Inhibiting TGF‐β signaling or eliminating P. gingivalis suppresses tumor growth and metastasis, suggesting the potential clinical application of targeting the P. gingivalis‐related signaling network for treating ESCC patients | Gao et al. 106 | |
| HNC | In vitro | SCC‐25 and BHY cells | After infection with P. gingivalis, both B7‐H1 and B7‐DC receptors were up‐regulated, which might facilitate immune evasion by oral cancers | Groeger et al. 86 | |
| HNC | In vitro | SAS and Ca9‐22 cells | P. gingivalis activates theERK1/2‐Ets1, p38/HSP27, and PAR2/NF‐ĸB pathways to induce proMMP‐9 expression and to promote cellular invasion of OSCC cell lines | Inaba et al. 94 | |
| HNC | In vitro, animal model | SCC‐25 and CAL 27;4NQO‐induced OSCC model in mice | Chronic bacterial infection has been observed to promote OSCC, with enhanced signaling along the IL‐6‐STAT3 axis believed to be responsible for this effect. Both P. gingivalis and F. nucleatum have been shown to stimulate tumorigenesis through direct interaction with oral epithelial cells via TLR. Additionally, these oral pathogens have been found to stimulate proliferation of human OSCC cells and induce expression of key molecules associated with tumorigenesis | Gallimidi et al. 93 | |
| HNC | In vitro | SAS cells | P. gingivalis activates PAR4 signaling pathways, leading proMMP‐9 over‐expression and cellular invasion in OSCC cells | Inaba et al. 95 | |
| HNC | In vitro, animal model | 4NQO‐induced OSCC model in mice, SCC‐25 and CAL 27 human tongue SCC cell lines | P. gingivalis stimulate tumorigenesis via direct interaction with oral epithelial cells through Toll‐like receptors and stimulate human OSCC proliferation and induce expression of key molecules implicated in tumorigenesis | Binder et al. 93 | |
| HNC | In vitro | Ca9‐22 OSCC cells | OSCC cells infected with P. gingivalis exhibited elevated MMP‐1 and MMP‐10 expression, a response triggered by the release of IL‐8, consequently amplifying the aggressiveness of OSCC cells | Ha et al. 97 | |
| HNC | In vitro | Tissue biopsies of healthy gingiva and HNC tumors, Human bone invasive oral squamous cell carcinoma BHY cells | P. gingivalis and human ɑ‐defensins could led to an increase in oral tumor cell proliferation | Hoppe et al. 90 | |
| HNC | In vitro | SCC‐25 cells | P. gingivalis membrane up‐regulates the expression of genes of TLR, NF‐ĸB, and MAPK signaling pathways which inducting pro‐inflammatory cytokines and inducing cancer proliferation | Groeger et al. 91 | |
| HNC | In vitro | H400 cells | Heat‐killed P. gingivalis triggered EMT‐signaling pathways and up‐regulated the expression of MMP‐2, ‐3 and ‐9 | Abdulkareem et al. 96 | |
| HNC | In vitro | Tca8113 cells | P. gingivalis could promotes OSCC proliferation by inducing cyclin D1 expression via the miR‐21/PDCD4/AP‐1 signaling pathway | Chang et al. 92 | |
| HNC | In vitro, animal model | Cal‐27 cells, s.c. transplantation model of OSCC in mice | P. gingivalis possessed the capability to hinder macrophage‐mediated phagocytosis of Cal‐27 cells, with membrane‐component molecules speculated, such as proteins, that can act as the effector. Furthermore, persistent exposure to antibiotics‐inactivated P. gingivalis fostered OSCC progression in mice and prompted the polarization of macrophages toward M2 tumor‐associated macrophages, primarily exhibiting pro‐tumor characteristics | Liu et al. 87 | |
| HNC | In vitro, animal model, clinical | OSCC samples, 4NQO‐induced OSCC model in mice | The cohort study showed that the localization of P. gingivalis in tumor tissues was related to poor survival of patients with OSCC. The promotion of tumor progression by P. gingivalis involves the recruitment of MDSCs via increasing secretion of IL‐6, IL‐8, CCL2, and CXCL2 from infected oral dysplastic keratinocytes | Wen et al. 88 | |
| HNC | In vitro, animal model | The human OSCC cell lines, UM‐SCC‐14A and HSC‐3; nude mice | P. gingivalis enhance the migration, invasion, tumorsphere formation, and tumorigenesis of OSCC cells in vivo. Pathogen‐induced OSCC cell migration occurred through the activation of integrin alpha V and FAK. These effects were abolished by stably inhibiting the expression of alpha V or FAK. Moreover, the process was inhibited by nisin | Kamarajan et al. 134 | |
| HNC | In vitro | H400 cells | P. gingivalis increased IL‐1β by upregulating AIM2, NLRP3 and downregulating POP1 | Aral et al. 89 | |
| HNC | In vitro, animal model, clinical | OSCC samples; human OSCC cell line TSCCa; OSCC mouse model created with SCC7 cell lines | P. gingivalis promotes OSCC progression by recruiting tumor‐associated neutrophils (TANs) through activation of the CXCL2/CXCR2 axis in the TME of OSCC | Guo et al. 99 | |
| PC | In vitro, animal model | Human pancreatic carcinoma cell lines MIA PaCa‐2 and PANC1; injected severe combined immunodeficient mice with PANC1 cells | Live P. gingivalis prompted the proliferation of pancreatic cancer cells and augmented tumor growth in murine models | Gnanasekaran et al. 111 | |
| PC | Animal model | PG‐LPS (lipopolysaccharide from P. gingivalis ATCC 33277) was prepared in physiological saline and intraperitoneally administered to C57BL/J mice | The upregulated expression levels of Reg3A and G might play a key role in PG‐LPS‐related pancreatic cancer in mouse | Hiraki et al. 110 | |
| PC | In vitro, animal model, clinical | Murine pancreatic cancer cell line Pan02; orthotopic and subcutaneous PC mouse model; PC samples | P. gingivalis promoted PC progression via elevating the secretion of neutrophilic chemokines and neutrophil elastase (NE) | Tan et al. 112 | |
| PC | In vitro, animal model, clinical | PDAC mouse model; human PDAC cell lines, Panc‐1, MIA‐PaCa‐2, AsPC‐1, BxPC‐3 and murine PDAC cell line Panc02; pancreatic tissue | Viable P. gingivalis was recovered from the pancreas of healthy mice after application of the bacteria to the oral mucosa, and chronic application of the bacteria to the oral cavity induced acinar cell‐to‐ductal metaplasia in the pancreas, and led to a shift in the pancreatic microbiome. Oral cavity application of P. gingivalis accelerated progression of pancreatic intraepithelial neoplasia to PDAC in mice expressing inducible acinar cell oncogenic KRAS. Oncogenic KRAS enabled intracellular survival of P. gingivalis, and intracellular bacteria promoted PDAC cell survival in hypoxic and nutrient‐depleted conditions | Saba et al. 113 | |
| Fusobacterium nucleatum | BC | In vitro, animal model, clinical | Human breast cancer cell line MCF‐7 and MDA‐MB‐231, mouse BALB/c breast cancer model cell line 4T1, mouse C57BL/6 breast cancer model cell line AT3, and mouse C57BL/6 melanoma model cell line B16; Balb/C, C57BL/c, and SCID‐beige mice; BC samples | This study provide strong evidence for a model whereby F. nucleatum generally reaches tumors via the hematogenous route and specifically attaches to them via a bacterial lectin‐host sugar (Fap2‐Gal‐GalNAc) interaction | Parhi et al. 31 |
| BC | In vitro, animal model, clinical | BC cell lines MDA‐MB‐231 and MCF‐7; BC tissues; BALB/c nude mice | F. nucleatum plays an important role in BC tumor growth and metastasis by regulating TLR4 through Fn‐EVs | Li et al. 152 | |
| CRC | In vitro, clinical | HCT116; CRC tissues | F. nucleatum adhesin FadA binds E‐cadherin and promotes CRC cell proliferation. FadA promotes inflammation and E‐cadherin‐mediated CRC tumor growth in xenograft mice | Rubinstein et al. 140 | |
| CRC | In vitro, animal model, clinical | C57BL/6J–Apc Min/J, BALB/c Il‐10 −/− and BALB/c T‐bet −/− Rag2 −/−; CD3ε+ CD4+ T cells fromApc Min mice; colonic adenocarcinoma, adenomas and stool samples | Fusobacteria recruit tumor‐infiltrating immune cells, fostering a pro‐inflammatory microenvironment that supports the progression of colorectal neoplasia | Kostic et al. 146 | |
| CRC | In vitro, animal model, clinical | CT26‐luc, human colon adenocarcinoma cell line HT29, RKO, and HCT116; murine CRC model; CRC samples; NSG mice | Gal‐GalNAc is highly expressed in human CRC, metastases, and a preclinical CRC model. Fap2 is a fusobacterial Gal‐GalNAc‐binding lectin. Fap2 mediates F. nucleatum binding to Gal‐GalNAc overexpressed in CRC. Blood‐borne Fap2‐expressing F. nucleatum localizes to orthotopic mouse colon tumors | Abed et al. 142 | |
| CRC | In vitro, clinical | CRC tissues; human colon cancer cell lines SW480 and Caco‐2 | Invasive F. nucleatum activates β‐catenin signaling via a TLR4/P‐PAK1/P‐β‐catenin S675 cascade in the carcinogenesis of CRC; TLR4 and PAK1 could be potential pharmaceutical targets for the treatment of F. nucleatum‐related CRCs | Chen et al. 143 | |
| CRC | In vitro, animal model, clinical | CRC tissues; The CRC cell lines (HCT116, SW480, HT29, Caco2, LoVo, SW620, DLD1, RKO, Colo205) and IEC; C57BL/6J‐Adenomatous polyposis coli (APC)min/J mice | CRC cells infected with F. nucleatum increased cells proliferation, invasive activity, and ability to form xenograft tumors in mice. The activation of TLR4 signaling by F. nucleatum leads to MYD88 activation, subsequently triggering the nuclear factor NF‐ĸB and upregulating miR21 expression. Patients exhibiting both high levels of tissue F. nucleatum DNA and miR21 expression showed a heightened risk for poor outcomes | Yang et al. 144 | |
| CRC | In vitro, animal model, clinical | CRC tissues; male C57BL/6‐Apc Min/+ mice; mouse macrophage cell line RAW 264.7 | F. nucleatum infection increased M2 macrophage polarization in vitro and in vivo, enhanced colorectal tumor growth, which is likely TLR4‐dependent involving the IL‐6/p‐STAT3/c‐Myc signaling pathway | Chen et al. 145 | |
| CRC | In vitro, animal model | C1, SB, 10C, HCT116, DLD1, SW480, HT29, RKO, PC‐9, 22RV1, UMUC3, and MCF‐7; nude mice | The FadA adhesin from F. nucleatum up‐regulates Annexin A1 expression through E‐cadherin | Rubinstein et al. 141 | |
| EC | Clinical | EC samples | F. nucleatum was detected in esophageal cancer tissues and was associated with shorter survival, suggesting that it may serve as a useful prognostic biomarker. F. nucleatum might also contribute to the acquisition of aggressive tumor behavior through the activation of chemokines, such as CCL20 | Yamamura et al. 34 | |
| EC | In vitro, animal model, clinical | ESCC samples; Human esophageal cancer cell lines TE‐8 and TE‐10; TE‐8 cells was injected subcutaneously and bilaterally into BALB/c mice | F. nucleatum invaded ESCC cells and induced the NF‐ĸB pathway through the NOD1/RIPK2 pathway, leading to tumor progression | Nomoto et al. 136 | |
| EC | In vitro, animal model, clinical | The human ESCC KYSE150 cell line and 293 T cell line; ESCC tissues; NSG mice | F. nucleatum could induce the enrichment of MDSCs in the tumor microenvironment by activating NLRP3 in ESCC cells. F. nucleatum infection could cause CDDP resistance in ESCC | Liang et al. 137 | |
| EC | In vitro, animal model, clinical | ESCC tissues; CD8+ T cells, KYSE150; NSG mice | F. nucleatum may induce high expression of KIR2DL1 on the surface of CD8+ T cells in ESCC tissues to provide a favorable microenvironment for self‐sustaining infection, thus promoting tumor progression | Wang et al. 138 | |
| GC | In vitro, clinical | GC tissues and peripheral blood; human erythroleukemia cell K562 and promyelocytic cell HL‐60 | F. nucleatum infection promotes NET formation, which releases EVs containing 14‐3‐3ε. These EVs could deliver 14‐3‐3ε to HPCs and promote their differentiation into MKs via activation of PI3K‐Akt signaling | Liu et al. 153 | |
| GC | In vitro, animal model, clinical | GC tissues; BGC‐823 (C6123) and SGC‐7901 (C6795) cell lines; BALB/c nude mice | Fn‐EVs facilitated GC tumor proliferation, DNA damage repair, stemness and metastasis in vitro and in vivo. Fn‐EVs also conferred GC cells resistance to oxaliplatin, which might rely on its regulation on DNA damage repair and stemness | Meng et al. 154 | |
| HNC | In vitro, animal model | 4NQO‐induced OSCC model in mice, SCC‐25 and CAL 27 human tongue SCC cell lines | F. nucleatum stimulate tumorigenesis via direct interaction with oral epithelial cells through Toll‐like receptors and stimulate human OSCC proliferation and induce expression of key molecules implicated in tumorigenesis | Binder et al. 93 | |
| HNC | In vitro, animal model | SCC‐25 and CAL 27;4NQO‐induced OSCC model in mice | Chronic bacterial infection has been observed to promote OSCC, with enhanced signaling along the IL‐6‐STAT3 axis believed to be responsible for this effect. Both P. gingivalis and F. nucleatum have been shown to stimulate tumorigenesis through direct interaction with oral epithelial cells via TLR. Additionally, these oral pathogens have been found to stimulate proliferation of human OSCC cells and induce expression of key molecules associated with tumorigenesis | Gallimidi et al. 93 | |
| HNC | In vitro | H400 cells | Heat‐killed F. nucleatum triggered EMT‐signaling pathways and increased MMP‐2, ‐3, and ‐9 expression | Abdulkareem et al. 96 | |
| HNC | In vitro, clinical | OSCC tissues; human squamous cell carcinoma cell lines SCC‐9 and HSC‐4 | F. nucleatum infection could initiate EMT via the lncRNA MIR4435‐2HG/miR‐296‐5p/Akt2/SNAI1 signaling pathway, potentially linking F. nucleatum infection to the onset of oral epithelial carcinomas | Zhang et al. 130 | |
| HNC | In vitro, animal model | The human OSCC cell lines, UM‐SCC‐14A and HSC‐3; nude mice | F. nucleatum enhance the migration, invasion, tumorsphere formation, and tumorigenesis of OSCC cells in vivo. Pathogen‐induced OSCC cell migration occurred through the activation of integrin alpha V and FAK. These effects were abolished by stably inhibiting the expression of alpha V or FAK. Moreover, the process was inhibited by nisin | Kamarajan et al. 134 | |
| HNC | In vitro, animal model | HN and BHY cells; 4NQO‐induced OSCC model in mice | Fusobacteria could potentially enhance cancer cell invasiveness, survival, and EMT when presented in the oral tumor microenvironment | Harrandah et al. 131 | |
| HNC | In vitro | Tca8113 tongue squamous cell carcinoma cell | F. nucleatum infection promoted the proliferation ability of Tca8113 by causing DNA damage via the Ku70/p53 pathway | Geng et al. 132 | |
| HNC | In vitro | H400 cells | F. nucleatum promoted IL‐1β by increasing AIM2 and downregulating POP1 | Aral et al. 89 | |
| HNC | In vitro | Human oral squamous cell carcinoma lines Cal‐27 and HSC‐3 | F. nucleatum promotes cisplatin resistance and migration in oral squamous cell carcinoma cells through the Wnt/NFAT pathway | Da et al. 133 | |
| HNC | In vitro, animal model, clinical | OSCC tissues; C3H mice; human primary oral keratinocytes, human OSCC cell lines CAL27, SCC15, and SCC25, human monocyte cell line THP‐1 | F. nucleatum‐induced acidification renders a protumorigenic immune microenvironment, which in turn causes cell malignant transformation. F. nucleatum colonized in OSCC tissues could induce cancer cells to produce much more lactate via the GalNAc‐Autophagy‐TBC1D5‐GLUT1 signaling axis | Sun et al. 129 | |
| Aggregatibacter actinomycetemcomitans | HNC | In vitro | Human gingival squamous carcinoma cell line Ca9‐22 | Delivering the CdtB protein and transfecting the cdtB gene could induce cell cycle arrest and apoptosis in Ca9‐22 cells in vitro | Yamamoto et al. 176 |
| HNC | In vitro | Tissue biopsies of healthy gingiva and HNC tumors, Human bone invasive oral squamous cell carcinoma BHY cells | A. actinomycetemcomitans enhanced oral tumor cell death | Hoppe et al. 90 | |
| Treponema denticola | HNC | In vitro | Tumor tissues | In addition, Td‐CTLP positivity was significantly associated with invasion depth, tumor diameter and the expression of TLR‐7, TLR‐9 and c‐Myc in early‐stage mobile tongue squamous cell carcinoma | Listyarifah et al. 177 |
| HNC | In vitro, animal model | The human OSCC cell lines, UM‐SCC‐14A and HSC‐3; nude mice | T. denticola enhance the migration, invasion, tumorsphere formation, and tumorigenesis of OSCC cells in vivo. Pathogen‐induced OSCC cell migration occurred through the activation of integrin alpha V and FAK. These effects were abolished by stably inhibiting the expression of alpha V or FAK. Moreover, the process was inhibited by nisin. T. denticola induced the expression of TLR2, TLR4, and MyD88. Stable suppression of MyD88 significantly inhibited FAK activation induced by Treponema denticola and abolished the pathogen‐induced migration | Kamarajan et al. 134 | |
| HNC | In vitro, animal model | The OSCC cell line Cal‐27; female BALB/c‐nu nude mice | T. denticola could promote OSCC cell proliferation directly, and the mechanism was associated with intracellular TGF‐β pathway activation | Peng et al. 184 | |
| Orodigestive carcinogenesis | In vitro, animal model, clinical | Orodigestive tumor tissue (squamous cell carcinomas of tongue, tonsil, esophagus, and adenocarcinoma of stomach, pancreas, and colon); New Zealand White rabbit | Td‐CTLP could activate pro MMP‐8 and ‐9 into their active forms and degrade proteinase inhibitors such as TIMP‐1, TIMP‐2, and α‐1‐antichymotrypsin, along with complement C1q | Nieminen et al. 185 |
Abbreviations: 4NQO, 4‐nitroquinoline 1‐oxide; AIM2, absent in melanoma 2; AP‐1, activator protein‐1; BC, breast cancer; CCL, chemokine (C‐C motif) ligand; CRC, colorectal cancer; CXCL, chemokine (C‐X‐C motif) ligand; CXCR, C‐X‐C chemokine receptor; EC, eosphageal cancer; EMT, epithelial‐mesenchymal transition; ERK, extracellular signal‐regulated kinase; ESCC, esophageal squamous cell carcinoma; EVs, extracellular vesicles; FAK, focal adhesion kinase; GC, gastric cancer; GLUT, glucose transporter; HNC, head and neck cancer; HPCs, hematopoietic progenitor cells; HSP27, heat shock protein 27; IL, interleukin; lncRNA, long non‐coding RNA; MAPK, Mitogen‐activated protein kinase; MDSCs, myeloid‐derived suppressor cells; miR, microRNA; MMP, matrix metalloproteinase; MyD88, myeloid differentiation primary response 88; NFAT, nuclear factor of activated T cells; NF‐ĸB, nuclear Factor‐kappa B; NLRP, nucleotide‐binding oligomerization domain, leucine‐rich repeat‐containing receptor (NLR) family pyrin domain‐containing; NOD, nucleotide‐binding oligomerization domain; OSCC, oral squamous cell carcinoma; PAR, protease‐activated receptor; PC, pancreatic cancer; PDAC, pancreatic ductal adenocarcinoma; PDCD4, programmed cell death protein 4; PD‐L1, programmed death‐ligand 1; PI3K, phosphoinositide 3‐kinase; PTEN, phosphatase and tensin homolog; RIPK, receptor‐interacting protein kinase; STAT3, signal transducer and activator of transcription 3; TGF‐β, Transforming Growth Factor β; TIMP, tissue inhibitor of metalloproteinases; TLR, toll like receptor; TME, tumor microenvironment.
4.1. Porphyromonas gingivalis
4.1.1. Characteristics of P. gingivalis
P. gingivalis, a gram‐negative bacterium, is considered a primary etiologic factor in the pathogenesis of periodontal disease. 66 Furthermore, this bacterium has been linked to several extraoral infection‐related diseases, including cardiovascular disease, diabetes, pulmonary disease, and rheumatoid arthritis. 67 Increasing evidence suggests the involvement of P. gingivalis in the etiology of oral, gastrointestinal, and pancreatic cancers. 49 , 68 The investigation of gut microbiota, comparing cancer cases and control subjects for the presence and relative abundance of taxa, also identified an association between an elevated risk of CRC and increased presence of proinflammatory genera Fusobacterium and Porphyromonas. 40
P. gingivalis possesses diverse virulence factors that facilitate its reproduction within the host's reservoir. The most significant virulence factors of P. gingivalis include fimbriae, lipopolysaccharide (LPS), outer membrane vesicles (OMV), and gingipains. 69 Fundamental pathogenic mechanisms involve generating dysregulated microbiota and impairing immune defenses. Among these virulence factors, fimbriae play a pivotal role in bacterial colonization. Two distinct types of fimbriae exist, with the significant type encoded by the FimA gene and the minor type encoded by the mfa1 gene. 70 , 71 Six FimA genotypes have been identified, with FimA II and FimA IV genotypes predominating in patients with periodontitis. In contrast, the FimA I genotype is most frequently observed in healthy patients. 70 , 71 These findings suggest that microbial danger may be discerned at the subspecies level, and the outcome could contribute to varying degrees of predispositions. 72
The LPS, present on the outer membrane of P. gingivalis, consists of lipid A, core oligosaccharide, and O‐antigen, serving as another virulence factor capable of activating macrophages through toll‐like receptors (TLRs). 73 , 74 It suppresses alkaline phosphatase, α1 collagen, and osteocalcin differentiation and mineralization in periodontal ligament stem cells. 70 Lipid A, in particular, can stimulate TLR4 on host cells, initiating signaling cascades that result in the production of pro‐inflammatory cytokines. The enzymes generated by P. gingivalis represent an additional virulence factor. The key enzymes facilitating the growth of this bacterium are known as proteases, specifically referred to as gingipains. These enzymes play a crucial role in degrading extracellular matrix proteins, including fibronectin and collagen. 75 To evade the host's defense mechanism, they dismantle immunoglobulins, complement factors, T cell receptors, and the ɑ‐defensins mechanism expressed by neutrophils. 76 For example, these enzymes can cleave surface proteins of T cells, such as CD4 and CD8, thereby influencing T cells functionality. 77 Gingipains induce the secretion of interleukin‐6 (IL‐6) by oral epithelial cells 78 and the expression of IL‐8 by gingival fibroblasts. 79 Additionally, they can hydrolyze and deactivate both anti‐inflammatory cytokines (IL‐4, IL‐5) and pro‐inflammatory cytokines (IL‐12, IFN‐γ). 80 , 81
Biofilms and OMVs are two crucial structures produced by microorganisms, playing pivotal roles in the survival, dissemination, and pathogenicity of microbes. 82 Biofilms consist of water, bacterial cells, and extracellular polymeric substances, and serve as protective layers, making them resistant to clinical interventions, including antibiotics. 82 OMVs, spherical membrane‐enclosed nanostructures released from the outer membrane of gram‐negative bacteria, play a crucial role in bacterial survival, virulence, and pathogenicity. 83 OMVs from P. gingivalis contribute to biofilm development by interacting with other periodontopathogens. They foster bacterial adhesion and invasion into host cells, adaptation to stress, and evasion of immune defense mechanisms. 82 , 83 , 84 Figure 4 summarizes the structure and representative components of P. gingivalis.
FIGURE 4.

Characteristics of Porphyromonas gingivalis (P. gingivalis) and its outer membrane vesicles (OMVs). P. gingivalis consists of cell membrane and genetic material. The outer layer of P. gingivalis' cell membrane encompasses fimbriae, proteins, and channels. The primary virulence factors of P. gingivalis primarily stem from both its structural components, such as lipopolysaccharide (LPS) and fimbriae, and its secretory components, including gingipains and outer membrane vesicles (OMVs). Adapted from Ref. [85].
4.1.2. Proposed carcinogenic mechanisms
Head and neck cancer
P. gingivalis strains have been shown to induce the expression of B7‐H1 and B7‐DC receptors in squamous cell carcinoma cells, potentially aiding in immune evasion in oral cancer. 86 Liu et al. 87 investigated the impact of P. gingivalis on the phagocytosis of Cal‐27 cells (OSCC cell line) by bone marrow‐derived macrophages and its effects on OSCC growth in vivo. They found that P. gingivalis had the capability to hinder macrophage‐mediated phagocytosis of Cal‐27 cells, with membrane‐component molecules, such as proteins, speculated to act as the effector. Furthermore, persistent exposure to antibiotics‐inactivated P. gingivalis fostered OSCC progression in mice and prompted the polarization of macrophages toward M2 tumor‐associated macrophages, primarily exhibiting pro‐tumor characteristics. 87 These results suggest that P. gingivalis may enhance immune evasion in oral cancer by protecting cancer cells from macrophage attack. Moreover, P. gingivalis promotes tumor progression by generating a cancer‐promoting microenvironment. In a mouse model of carcinogenesis induced by 4‐nitroquinoline‐1 oxide (4NQO), P. gingivalis infection exacerbates oral lesions and promotes tumor progression through invasion of oral lesions. Increased infiltration of CD11b + myeloid cells and myeloid‐derived suppressor cells (MDSCs) is observed in these lesions. In vitro experiments reveal that exposure of human‐derived dysplastic oral keratinocytes (DOKs) to P. gingivalis leads to the accumulation of MDSCs. Chemokines such as Chemokine (C‐X‐C motif) ligand 2 (CXCL2), chemokine (C‐C motif) ligand 2 (CCL2), IL‐6, and IL‐8 are implicated in facilitating the recruitment of MDSCs, suggesting their potential role in tumor progression. 88 Moreover, P. gingivalis upregulates IL‐1β by upregulating the inflammasome complexes nucleotide‐binding oligomerization domain, leucine‐rich repeat‐containing receptor (NLR) family pyrin domain‐containing 3 (NLRP3) and absent in melanoma 2 (AIM2), while downregulats pyrin domain (PYD)‐only protein 1 (POP1). 89
P. gingivalis and human ɑ‐defensins could lead to an increase in oral tumor cell proliferation. 90 Groeger et al. 91 utilized a profiler PCR array and found that P. gingivalis membrane upregulates the expression of genes of TLR, nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NF‐κB), and mitogen‐activated protein kinases (MAPK) signaling pathways, leading to the induction of pro‐inflammatory cytokines and promoting cancer proliferation. Moreover, P. gingivalis promotes OSCC proliferation by inducing cyclin D1 expression via the miR‐21/programmed cell death protein 4 (PDCD4)/activator protein‐1 (AP‐1) signaling pathway. 92 Furthermore, another study observed that certain activators of TLR2/1 and TLR4, or when exposed to P. gingivalis, trigger essential molecular components intricately associated with the progression and virulence of oral tumors, such as IL‐6, cyclin D1, TNF‐ɑ, and heparanase. 93 Matrix metalloproteinase 9 (MMP‐9) is implicated in the invasion and metastasis of cancer cells. Inaba et al. 94 , 95 demonstrated that P. gingivalis activates the extracellular signal‐regulated kinase (ERK)1/2‐Ets1, p38/heat shock protein27 (HSP27), protease‐activated receptor (PAR) 4, and PAR2/NF‐ĸB pathways to induce proMMP‐9 expression and to promote cellular invasion of OSCC cell lines. Additionally, heat‐killed P. gingivalis upregulates the expression of MMP‐2, MMP‐3, and MMP‐9 in OSCCs. Epithelial‐mesenchymal transition (EMT) is closely associated with invasion and metastasis. Heat‐killed P. gingivalis increased the levels of TGF‐β1, TNF‐ɑ, and epidermal growth factor (EGF), which are involved in EMT‐signaling pathways, in OCSS cell lines. Furthermore, exposure to heat‐killed P. gingivalis enhances migration and the rate of wound closure. 96 OSCC cells infected with P. gingivalis also exhibit elevated MMP‐1 and MMP‐10 expression, triggered by the release of IL‐8, thereby amplifying the aggressiveness of OSCC cells. 97 Physiologically, bacteria and other microorganisms in the oral cavity are recognized by TLR2. Ikehata et al. 98 reported that the progression of OSCC can be accelerated by the activation of TLR2 through bacterial components. This activation potentially contributes to acquired resistance against cisplatin‐induced apoptosis via modulation of the miR‐146a pathway.
In tumor‐bearing animal experiments, tumor samples with P. gingivalis infection in the tumor microenvironment (TME) of OSCC displayed enhanced cell invasion and proliferation, along with larger tumor volumes. Additionally, the expression of P. gingivalis, CXCL2, and tumor‐associated neutrophils (TANs) serve as independent risk factors for poor prognosis in OSCC patients. Treatment with a CXCL2/CXCR2 signaling axis inhibitor significantly attenuated cell invasion and proliferation and reduced tumor volume in mice. Lentivirus‐mediated blockade of the CXCL2/CXCR2 signaling axis results in decreased activity of the JAK1/signal transducer and activator of transcription (STAT) signaling pathway and reversal of the EMT phenotype 99 (Figure 5).
FIGURE 5.

Mechanisms by which Porphyromonas gingivalis may contribute to head and neck cancers. This figure illustrates the potential mechanisms by which the periodontal pathogen Porphyromonas gingivalis (P. gingivalis) contributes to the development and progression of head and neck cancers. It depicts a series of signaling pathways and molecular interactions initiated by P. gingivalis, leading to immune evasion, proliferation, invasion, metastasis, inflammation, and increased aggressiveness of cancer cells. P. gingivalis induces the expression of B7‐H1 and B7‐DC receptors on host cells, leading to immune evasion. It also promotes OSCC proliferation by inducing epidermal growth factor receptor (EGFR) and cyclin D1 expression via the miR‐21/PDCD4/AP‐1 signaling pathway. P. gingivalis induces the expression of MMP‐9 through activate the protease‐activated receptor (PAR) 2/nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NF‐κB), p38/heat shock protein27 (HSP27), and ERK1/2‐Ets1 pathway, ultimately promoting invasion. The activation of the Chemokine (C‐X‐C motif) ligand 2 (CXCL2)/C‐X‐C chemokine receptor 2 (CXCR2) axis by P. gingivalis triggers the epithelial‐mesenchymal transition (EMT) phenotype through the JAK1/signal transducer and activator of transcription 3 (STAT3) signaling pathway. P. gingivalis elevates TGF‐β1, TNF‐ɑ, and epidermal growth factor (EGF) levels, associated with EMT signaling pathways, closely linked to invasion and metastasis. The activation of MMP‐2, matrix metalloproteinase (MMP)‐3 by P. gingivalis is shown to facilitate metastasis. Additionally, the bacteria induce the production of IL‐6 and IL‐8, leading to inflammation, which further enhances the aggressiveness of cancer cells through MMP‐1 activation.
Eosphageal cancer
P. gingivalis isolated from esophageal squamous cell carcinoma (ESCC) lesions is recognized as a major pathogen associated with this fatal disease. Yuan et al. 100 investigated host immune responses during P. gingivalis infection, revealing increased expression of B7‐H4 and lysine demethylase 5B (KDM5B) in experimentally infected ESCC. B7‐H4, also known as B7S1, B7x, or VTCN1, plays a negative regulatory role in T cell activation. 101 Anti‐B7‐H4 and histone demethylase inhibitors were investigated for their immunotherapeutic effects against chronic infection and xenografted human tumors. Through combined therapy in three distinct preclinical mouse models, enhanced resistance against P. gingivalis infection and tumor challenge was observed, possibly attributed to T cell‐mediated responses and immune memory formation. The coexpression of B7‐H4 and KDM5B correlated significantly with bacterial load in ESCC subjects, suggesting potential therapeutic targets for improving control of P. gingivalis infection and associated neoplasia. 100 Additionally, another study found that P. gingivalis was associated with elevated EC incidence in a 4‐nitroquinoline 1‐oxide‐induced mouse model and increased xenograft tumor growth. P. gingivalis infection promoted IL‐6 production and MDSCs recruitment. Inhibition of IL‐6 signaling attenuated the tumor‐promoting effects of P. gingivalis in both mouse models. Moreover, ESCC cells infected with P. gingivalis exhibited enhanced characteristics associated with EMT, including increased expression levels of β‐catenin and MMP‐9, while simultaneously showing decreased expression levels of E‐cadherin. 102
In vitro studies revealed that P. gingivalis enhances the proliferation and motility of ESCC cells, upregulating key molecules such as cyclin D1, MMP‐2, c‐Myc, and BCL‐XL, associated with the NF‐ĸB signaling pathway. 103 Gao et al. examined the impact of P. gingivalis infection on ESCC and found that it significantly enhanced cellular proliferation, invasion, and migration in the ESCC cell lines KYSE‐30 and KYSE‐150. High‐throughput sequencing identified upregulation of miR‐194 in these infected cells, with miR‐194 modulation affecting cell migration and invasion. Bioinformatics analysis identified GRHL3 as a direct target of miR‐194, with decreased expression of GRHL3 observed in P. gingivalis‐infected ESCC cells and patients. Downregulation of both GRHL3 and PTEN, along with upregulation of p‐Akt, suggests a potential signaling pathway involving miR‐194/GRHL3/PTEN/Akt in ESCC progression promoted by P. gingivalis. 104
Gao et al. 105 have found that P. gingivalis infection is associated with a worse prognosis in patients with ESCC, reduced efficacy of neoadjuvant chemotherapy (NACT), and promotion of apoptosis resistance and proliferation in ESCC cells. Furthermore, it has been reported that TGF‐β/Smad signaling mediates the oncogenic function of P. gingivalis in ESCC. 106 Global transcriptomic analysis revealed that P. gingivalis infection induces elevated secretion of TGF‐β and activates TGF‐β/Smad signaling in both cultured cells and clinical ESCC samples. Moreover, this study demonstrated that P. gingivalis enhances the expression of Glycoprotein A repetitions predominant (GARP), thus activating TGF‐β/Smad signaling, with this effect partially dependent on P. gingivalis fimbriae (FimA). Xenograft models further demonstrated that P. gingivalis infection activated TGF‐β signaling, thereby promoting tumor growth and lung metastasis. 106 (Figure 6).
FIGURE 6.

Mechanisms by which Porphyromonas gingivalis may contribute to esophageal cancer. Porphyromonas gingivalis (P. gingivalis) up‐regulates B7‐H4 and KDM5B in esophageal squamous cell carcinoma, contributing to immune evasion. Additionally, P. gingivalis infection also promotes IL‐6 production and MDSCs recruitment. Furthermore, it upregulates miR‐194 while inhibiting GRHL3 and PTEN. This, in turn, activates the phosphoinositide 3‐kinase (PI3K)/Akt pathway, promoting cell proliferation and migration. By inducing the epithelial‐to‐mesenchymal transition (EMT) process, P. gingivalis enhances the invasiveness and metastasis of cancer cells through the upregulation of molecules such as β‐catenin and matrix metalloproteinase 9 (MMP‐9). It enhances the expression of glycoprotein A repetitions predominant (GARP), thus activating transforming growth factor‐β (TGF‐β)/Smad signaling pathway. This effect is partially dependent on P. gingivalis fimbriae (FimA), thereby promoting tumor growth and lung metastasis. P. gingivalis upregulates the expression of cyclin D1, c‐Myc, and MMP‐2, as well as activating the nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NF‐κB) pathway, which collectively promote cell proliferation, migration, and invasion.
Colorectal cancer
Adel‐Khattab et al. 107 investigated the impact of P. gingivalis on colon cancer cells (CL‐11). They stimulated the cells with P. gingivalis total membrane (TM) fractions and peptidoglycans (PDG), as well as viable P. gingivalis bacteria. Their findings revealed that both the TM fraction and PDG induced an upregulation of PD‐L1 in colon cancer cells, with PDG having a significant influence. The mechanism involved the activation of NOD 1 and NOD 2, along with the RIP2 and MAPK signaling pathways. Mu et al. 108 observed that P. gingivalis can adhere to and invade host cells, promoting cell proliferation. They found an increase in the percentage of S phase cells in the cell cycle assay. Through qPCR and western blot analyses, they suggested that the MAPK/ERK signaling pathway is significantly activated by P. gingivalis. Wang et al. 109 found that P. gingivalis enhanced tumor counts and volumes in the Apc Min/+ mouse model and boosts tumor growth in orthotopic rectal and subcutaneous carcinoma models. It also modulates the tumor immune microenvironment by selectively expanding myeloid‐derived immune cells and fostering a proinflammatory milieu. Furthermore, P. gingivalis contributes to the progression of colorectal cancer neoplasia by activating the hematopoietic NLRP3 inflammasome (Figure 7).
FIGURE 7.

Mechanisms by which Porphyromonas gingivalis may contribute to colorectal cancer. Porphyromonas gingivalis (P. gingivalis) has been demonstrated to induce programmed cell death 1 (PD‐L1) expression through the regulation of molecules such as non‐occlusive disease (NOD)1/NOD2, receptor‐interacting protein 2 (RIP2), and mitogen‐activated protein kinases (MAPK). This process serves to mediate the immune evasion of tumor cells. In vivo, P. gingivalis has been observed to stimulate the secretion of pro‐inflammatory factors by myeloid cells, including nucleotide‐binding oligomerization domain, leucine‐rich repeat‐containing receptor (NLR) family pyrin domain‐containing 3 (NLRP3), Caspase‐1, and interleukin (IL)‐1β. This leads to the promotion of inflammation and the acceleration of tumor growth. Activation of the MAPK/ERK signaling pathway by P. gingivalis has been demonstrated to accelerate cell proliferation and invasive behavior.
Pancreatic cancer
Hiraki et al. 110 estabilished a mouse model that is not affected by acute indlammation in various organs by intraperitoneally administering P. gingivalis‐LPS to mice. They analyzed pancreatic gene expression and observed elevated expression of Reg3A and G, which are previously associated with PC. Gnanasekaran et al. 111 discovered that live P. gingivalis prompted the proliferation of pancreatic cancer cells. Interestingly, this impact was not contingent upon TLR2. Furthermore, P. gingivalis augmented tumor growth in murine models, with the pro‐tumorigenic effects potentially attributable, at least in part, to the activation of the Akt signaling cascade. Tan et al. 112 confirmed the presence of a rich intratumor microbiota in human PC tissue, with the oral cavity bacterium P. gingivalis identified as colonizing and proliferating within the tumor microenvironment. Exposure to P. gingivalis accelerated tumor growth in both orthotopic and subcutaneous PC mouse models. Mechanistically, intratumoral P. gingivalis accelerated PC progression by enhancing the secretion of neutrophilic chemokines and neutrophil elastase (NE). Saba et al. 113 illustrated the migration of P. gingivalis from the oral cavity to the pancreas in a genetically engineered mouse model of pancreatic ductal adenocarcinoma (PDAC). Repetitive administration of P. gingivalis to wild‐type mice induced pancreatic acinar‐to‐ductal metaplasia (ADM) and altered the composition of the intrapancreatic microbiome. In iKC mice, P. gingivalis expedited the progression from pancreatic intraepithelial neoplasia (PanIN) to PDAC. In vitro, P. gingivalis infection induced the expression of acinar cell ADM markers SOX9 and CK19, while intracellular bacteria shielded PDAC cells from reactive oxygen species‐mediated cell death induced by nutrient stress.
4.2. Fusobacterium nucleatum
4.2.1. Characteristics of F. nucleatum
Fusobacterium nucleatum (F. nucleatum) is an obligate anaerobic gram‐negative bacterium commonly found in both healthy and diseased individuals within the oral cavity. 114 It is associated with periodontal diseases and has been isolated from clinical specimens of many diseases, such as appendicitis, brain abscesses, osteomyelitis, pericarditis, and chorioamnionitis. 114 , 115 There are reports indicating that F. nucleatum is also likely involved in the onset of gastrointestinal and oral cancer. 116
F. nucleatum has the ability to co‐aggregate with almost all bacterial species involved in oral plaque formation. 117 In addition, it can bind to and facilitate the transport of non‐invasive bacterial species into host cells, functioning as a shuttle during this process. 118 F. nucleatum also mediates essential biofilm organization and engages in interactions with host cells by expressing multiple adhesins, such as adhesion inducing determinant 1 (Aid1), coaggregation‐mediated protein A (CmpA), fibroblast activation protein 2 (Fap2), FomA, fusobacterial adhesin (FadA), and RadD. 119 Aid1, likely unique to Fusobacteria, can facilitate RadD‐mediated interaction with oral streptococci. 120 The interplay between F. nucleatum and Streptococcus gordonii is facilitated through CmpA. 121 FadA, the significant adhesin identified to bind host cells, stands out as the most extensively characterized virulence factor of F. nucleatum. 122 It plays a critical role in the tumorigenic response as well as in the binding and invasion of host cells by the organism. 119 By binding to E‐cadherin, FadA triggers the activation of β‐catenin signaling, influencing both inflammatory and oncogenic responses. 12 Fap2, a galactose‐sensitive hemagglutinin and adhesion, can bind with acetylgalactosamine (Gal‐GalNAc) to mediate CRC development. 123 The study showed that Fap2 could also bind to human inhibitory receptor T cell immunoglobulin and ITIM domain (TIGIT), including T cells and natural killing (NK) cells. 124 This Fap2‐TIGIT interaction shields both F. nucleatum and adjacent tumor cells from immune cell‐mediated killing. 124 In addition to TIGIT, F. nucleatum employs its surface protein CbpF to hinder T and NK cells function through the activation of carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1). 125 , 126 FomA exhibits the ability to attach to F. nucleatum‐specific IgA. Additionally, FomA can stimulate intestinal immunity, leading to increased secretion of IgA, which serves as a deterrent to its colonization. 127 These adhesins are also considered to potentially contribute to the carcinogenesis process. 128
4.2.2. Proposed carcinogenic mechanisms
Head and neck cancer
Sun et al. 129 observed the accumulation of F. nucleatum in the invasive margins of OSCC tissues, correlating with the formation of tumor‐associated macrophages (TAMs). They identified that F. nucleatum triggered GalNAc‐Autophagy‐TBC1D5 signaling, resulting in glucose transporter 1 (GLUT1) aggregation on the plasma membrane and extracellular lactate deposition. Concurrent functional inhibition of GalNAc and GLUT1 effectively suppressed TAMs formation and restrained OSCC progression. 129
Zhang et al. 130 found that F. nucleatum infection induces cell migration in both noncancerous human immortalized oral epithelial cells and OSCC cells, without affecting cell proliferation or cell cycle progression. They noted upregulation of mesenchymal markers such as N‐cadherin, Vimentin, and snail family transcriptional repressor 1 (SNAI1), along with decreased E‐cadherin expression, which translocated to the cytoplasm. Moreover, both FadA adhesin and heat‐inactivated F. nucleatum showed similar effects to viable bacterial cells. High‐throughput sequencing identified upregulated lncRNA MIR4435‐2HG, which negatively regulated miR‐296‐5p expression, reduced in F. nucleatum‐infected human immortalized oral epithelial cells (HIOECs) and SCC‐9 cells. Knockdown of MIR4435‐2HG with siRNA decreased SNAI1 expression, while miR‐296‐5p indirectly regulated SNAI1 expression via Akt2. Thus, their study suggested that F. nucleatum infection can initiate EMT via the lncRNA MIR4435‐2HG/miR‐296‐5p/Akt2/SNAI1 signaling pathway, potentially linking F. nucleatum infection to the onset of oral epithelial carcinomas. Harrandah et al. 131 demonstrated in vitro that F. nucleatum induces the expression of STAT3, janus kinase 1 (JAK1), MYC, and EMT markers in oral cancer cells. Additionally, it enhances the expression of MMP‐1, MMP‐9, and IL‐8, leading to increased cancer cell invasiveness. This in vivo study revealed that both P. gingivalis and F. nucleatum promote tumor progression. F. nucleatum was also observed to trigger TLR signaling, resulting in IL‐6 production that activates STAT3, which regulate SCC growth. 93 In HNCSS cell line (H400), F. nucleatum increased AIM2 and downregulated POP1, promoting IL‐1β. 89 The heat‐killed F. nucleatum increased cytokines expression which invoveld in EMT‐induction, such as TGF‐β1, EGF, and TNF‐ɑ, and increased MMP‐2, ‐3, and ‐9 expression in OSCC cell line H400. 96 Geng et al. 132 found that the infection with F. nucleatum led to increased expression of γH2AX, a marker for DNA double‐strand break, in Tca8113 tongue squamous cell carcinoma cells. This infection promoted cell proliferation and altered the cell cycle, downregulated p27 expression, and decreased the levels of Ku70 and wild‐type p53. Overexpression of Ku70 reversed these effects, leading to the upregulation of wild‐type p53 and inhibition of cell proliferation. These findings suggest that F. nucleatum infection promotes OSCC proliferation via the Ku70/p53 pathway by inducing DNA damage. Another study demonstrated that F. nucleatum enhances cisplatin resistance and migration in oral squamous carcinoma cells by downregulating p53 and E‐cadherin via the Wnt/nuclear factor of activated T cells (NFAT) pathway. Pretreatment of Cal‐27 and HSC‐3 cells with F. nucleatum significantly increased survival rates against cisplatin. Western blot analysis revealed decreased expression of migration and apoptosis‐related proteins E‐cadherin and p53. F. nucleatum activation of the Wnt/NFAT pathway was observed, with elevated expression levels of wnt5a and Nuclear factors of activated T cells 3 (NFATc3). These findings suggest that F. nucleatum promotes cisplatin resistance and migration in oral squamous cell carcinoma cells through the Wnt/NFAT pathway. 133
P. gingivalis, T. denticola, and F. nucleatum were observed to enhance the migration, invasion, tumorsphere formation, and tumorigenesis of OSCC cells in vivo. However, they did not significantly impact cell proliferation or apoptosis. Pathogen‐induced OSCC cell migration occurred through the activation of integrin alpha V and focal adhesion kinase (FAK). These effects were abolished by stably inhibiting the expression of alpha V or FAK. Moreover, the process was inhibited by nisin, a 34‐amino acid polycyclic antimicrobial peptide produced by Gram‐positive Lactococcus and Streptococcus species. 134 , 135 (Figure 8).
FIGURE 8.

Mechanisms by which Fusobacterium nucleatum may contribute to head and neck cancer. Fusobacterium nucleatum (F. nucleatum) infection initiates the epithelial‐to‐mesenchymal transition (EMT) through the lncRNA MIR4435‐2HG/miR‐296‐5p/Akt2/SNAI1 signaling pathway. The infection with F. nucleatum also increased cytokine expression, contributing to EMT induction and increased cancer cell invasiveness. Moreover, F. nucleatum triggered GalNAc‐Autophagy‐TBC1D5 signaling, leading to GLUT1 aggregation on the plasma membrane and extracellular lactate deposition. F. nucleatum promotes migration and apoptosis in oral squamous cell carcinoma (OSCC) cells via the Wnt/nuclear factor of activated T cells (NFAT) pathway. Additionally, F. nucleatum induces absent in melanoma 2 (AIM2) expression and downregulated pyrin domain (PYD)‐only protein 1 (POP1), promoting interleukin‐1β (IL‐1β) production and inflammation. It also triggers integrin alpha V and focal adhesion kinase (FAK), inducing OSCC cell proliferation. Furthermore, F. nucleatum activates toll like receptor (TLR) signaling, resulting in IL‐6 production that activates signal transducer and activator of transcription 3 (STAT3), thereby regulating squamous cell carcinoma growth. F. nucleatum induces the expression of janus kinase 1 (JAK1) and MYC in oral cancer cells, highlighting its role in promoting tumorigenesis.
Eosphageal cancer
The presence of F. nucleatum in EC tissues correlated with shorter survival, suggesting its potential utility as a prognostic biomarker. Through microarray data enrichment analyses of kyoto encyclopedia of genes and genomes (KEGG) pathways, it was identified that F. nucleatum may promote aggressive tumor behavior by activating chemokines such aschemokine (C‐C motif) ligand 20 (CCL20), a finding confirmed by immunohistochemistry. 34 Nomoto et al. 136 found that F. nucleatum invades ESCC cells and triggers the NF‐ĸB pathway, thereby promoting tumor progression both in vivo and in vitro. Moreover, F. nucleatum enhances the growth, migration, and invasion capabilities of ESCC cells. The activation of NF‐ĸB by F. nucleatum is facilitated through nucleotide oligomerization domain 1 (NOD1) and receptor‐interacting protein kinase 2 (RIPK2). Liang et al. 137 observed a correlation between F. nucleatum infection in ESCC tissues, heightened NLRP3 expression, and an increase in myeloid‐derived suppressor cells (MDSC). Both in vitro and in vivo models demonstrated that F. nucleatum prompts a significant accumulation of MDSCs by upregulating NLRP3 expression in ESCC cells and reducing their susceptibility to cisplatin (CDDP) therapy. Wang et al. 138 noted that F. nucleatum induced the expression of KIR2DL1, a significant inhibitory molecule, on the surface of CD8+ T cells, consequently attenuating their cytotoxic activity. This phenomenon facilitates tumor cell evasion from immune surveillance and diminishes the effectiveness of CDDP therapy. By examining the correlations between the elevated expression of KIR2DL1 on CD8+ T cell surfaces induced by F. nucleatum and the clinicopathological characteristics of ESCC patients, they observed that tumors with increased malignancy exhibited a microenvironment conducive to F. nucleatum survival and the induction of KIR2DL1 expression on CD8+ T cells (Figure 9).
FIGURE 9.

Mechanisms by which Fusobacterium nucleatum may contribute to esophageal cancer. Fusobacterium nucleatum (F. nucleatum) activates the nucleotide oligomerization domain 1 (NOD1)/receptor‐interacting protein kinase 2 (RIPK2)/nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NF‐κB) pathway, leading to cell growth, migration, and invasion in esophageal cancer. F. nucleatum interacts with myeloid‐derived suppressor cells (MDSCs) and CD8+ T cells through the nucleotide‐binding oligomerization domain, leucine‐rich repeat‐containing receptor (NLR) family pyrin domain‐containing 3 (NLRP3) and KIR2D1 receptors, respectively, resulting in immune evasion.
Colorectal cancer
Fusobacteria from the oral cavity could potentially migrate to CRC sites either through the digestive tract's descent or via the bloodstream during intermittent bacteremia triggered by routine oral activities like chewing, hygiene practices, or dental treatments. In a study by Abed et al., 139 CRC colonization by F. nucleatum was compared between mice inoculated through gavage and intravenous injection in the MC38 and CT26 orthotopic CRC models. The findings indicated that fusobacterial colonization of CRC tumors was more successful through hematogenous dissemination compared to oral administration. These results suggest that the bloodstream may serve as the primary route for oral fusobacteria to reach and colonize colon tumors.
Rubinstein et al. 140 unveiled the mechanism by which F. nucleatum adheres to and invades CRC cells, stimulating oncogenic cell proliferation and inflammatory responses both in vivo and in vitro through its distinct FadA adhesin. This adhesin binds specifically to E‐cadherin, activating β‐catenin signaling and modulating inflammatory and oncogenic pathways. An unrecognized modulator of Wnt/β‐catenin signaling, annexin A1, plays a key role in F. nucleatum's stimulatory effect by promoting Cyclin D1 activation in proliferating colorectal cancer cells. Subsequent work by Rubinstein et al. 141 further demonstrated that the FadA adhesin from F. nucleatum increased annexin A1 expression via E‐cadherin. Interestingly, this positive feedback loop between FadA and annexin A1 was exclusively detected in cancerous cells, while it was absent in non‐cancerous cells. Based on these findings, they proposed a “two‐hit” hypothesis for colorectal carcinogenesis, wherein the initial “hit” involves the accumulation of host driver mutations, such as increased annexin A1 expression. Subsequently, microbes like F. nucleatum represent the second “hit,” exacerbating the progression of cancer. 141 Abed et al. 142 identified a host polysaccharide and fusobacterial lectin responsible for the abundance of fusobacteria in CRC. Fusobacterial Fap2 recognizes Gal‐GalNAc, highly expressed in CRC, acting as a Gal‐GalNAc lectin. Strains lacking Fap2 or with inactivated Fap2 mutants exhibit reduced binding to Gal‐GalNAc‐expressing CRC cells and established CRCs in murine models. Moreover, intravenously administered F. nucleatum localizes to mouse tumor tissues in a Fap2‐dependent manner, suggesting fusobacteria utilize a hematogenous route to colon adenocarcinomas. Chen et al. 143 observed elevated levels of TLR4, p21‐activated kinase 1 (PAK1), and nuclear β‐catenin proteins in F. nucleatum‐positive CRCs. Exposure to F. nucleatum or its lipopolysaccharide resulted in increased protein abundance of TLR4/P‐PAK1/P‐β‐catenin S675/c‐Myc/CyclinD1, along with enhanced nuclear translocation of β‐catenin. Pre‐treatment with TLR4 or PAK1 inhibitors reduced the protein abundance of P‐β‐catenin S675, c‐Myc, and Cyclin D1, and nuclear β‐catenin accumulation. Inhibition of TLR4 also reduced P‐PAK1 protein abundance, suggesting invasive F. nucleatum activates β‐catenin signaling through a TLR4/P‐PAK1/P‐β‐catenin S675 cascade in CRC. TLR4 and PAK1 may serve as potential therapeutic targets for the treatment of F. nucleatum‐related CRCs. Besides, Yang et al. 144 discovered that activation of TLR4 signaling by F. nucleatum leads to MYD88 activation, subsequently triggering NF‐ĸB and upregulating miR21 expression. This miRNA decreases RAS GTPase RASA1 levels, which plays a crucial role in regulating cell proliferation, differentiation, and apoptosis. Patients exhibiting both high levels of tissue F. nucleatum DNA and miR21 expression showed a heightened risk for poor outcomes. Furthermore, F. nucleatum also plays a regulatory role in immune responses. Chen et al. 145 observed an increase in M2 polarization of macrophages upon F. nucleatum infection both in vitro and in vivo. Moreover, F. nucleatum infection augmented colorectal tumor growth through a TLR4‐dependent mechanism, involving the activation of the IL‐6/p‐STAT3/c‐Myc signaling. F. nucleatum also plays a selective role in recruiting tumor‐infiltrating myeloid cells, particularly CD11b+ cells such as dendritic cells (DCs), macrophages, and granulocytes, thereby promoting tumor progression in CRC mouse models. 146 These CD11b+ myeloid cells play a pivotal role in enhancing tumor development and angiogenesis. 147 Toor et al. 148 observed increased levels of granulocytic myeloid cells, including myeloid‐derived suppressor cells (MDSCs), in both the tumor microenvironment and peripheral blood of CRC patients. MDCSs, known for their potent immune suppressive activity, exhibit significant T cell suppressive capabilities. This aligns with findings that F. nucleatum impedes the accumulation of tumor‐infiltrating T cells in breast and colorectal cancer tissues 31 , 146 , 149 , 150 , 151 (Figure 10).
FIGURE 10.

Mechanisms by which Fusobacterium nucleatum may contribute to colorectal cancer. The invasion of Fusobacterium nucleatum (F. nucleatum) selectively recruits myeloid cells into the tumor microenvironment. Upon interaction with immune cells, F. nucleatum leads to reduced T cell density, heightened M2 macrophage polarization, suppression of natural killer (NK) cell activity, and increased dendritic cells and tumor‐associated neutrophils, collectively dampening anti‐tumor immunity. The adhesin FadA of F. nucleatum interacts with E‐cadherin on host cells, leading to the activation of annexin A1. This activation regulates β‐catenin and subsequent downstream signaling pathways in proliferating colorectal cancer cells. Exposure to F. nucleatum increases the protein abundance of toll like receptor (TLR) 4/P‐PAK1/P‐β‐catenin S675/c‐Myc/CyclinD1, promoting inflammation, and oncogenic transformation. F. nucleatum also activates the nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NF‐κB) pathway, upregulating miR‐21, which in turn decrease RASA1. This cascade ultimately increases proliferation, differentiation, and tumor growth. Additionally, F. nucleatum interacts with the Gal‐GalNAc pathway, contributing to its localization and enrichment within the tumor microenvironment. Adapted from Ref. [276].
Breast cancer
Parhi et al. 31 observed an increase in Gal‐GalNAc levels during the progression of human breast cancer. They found a correlation between the presence of F. nucleatum gDNA in breast cancer samples and elevated Gal‐GalNAc levels. The study demonstrated Fap2‐dependent binding of F. nucleatum to breast cancer samples, a process inhibited by GalNAc. Intravascular inoculation of Fap2‐expressing F. nucleatum ATCC 23726 specifically colonized mammary tumors in mice, while Fap2‐deficient bacteria exhibited impaired tumor colonization. Inoculation with F. nucleatum suppressed the accumulation of tumor‐infiltrating T cells and promoted tumor growth and metastatic progression. In a recent study, Li et al. 152 investigated the impact of extracellular vesicles (EVs) derived from F. nucleatum (Fn‐EVs) on BC cells. They observed a significant increase in cell viability, proliferation, migration, and invasion in BC cells upon administration of Fn‐EVs. Notably, knockdown of TLR4 in BC cells blocked these effects. Furthermore, in vivo experiments confirmed the contributory role of Fn‐EVs in promoting tumor growth and metastasis in BC, potentially through their modulation of TLR4 signaling.
Gastric cancer
Liu et al. 153 observed increased counts of neutrophil extracellular traps (NETs) and platelets in F. nucleatum‐positive GC patients. Extracellular vesicles (EVs) derived from F. nucleatum‐positive patients were found to promote the differentiation and maturation of megakaryocytes (MKs) and exhibited increased levels of 14‐3‐3 proteins, particularly 14‐3‐3ε. In vitro experiments demonstrated that the upregulation of 14‐3‐3ε promoted MK differentiation and maturation. Additionally, hematopoietic progenitor cells (HPCs) and K562 cells were capable of receiving 14‐3‐3ε from the EVs, which interacted with GP1BA and 14‐3‐3ζ to initiate phosphoinositide 3‐kinase (PI3K)‐Akt signaling. Meng et al. 154 discovered that Fn‐EVs significantly enhanced resistance to oxaliplatin and promoted cell proliferation, migration, invasion, and stemness in GC cells. Moreover, Fn‐EVs enhanced the stemness and DNA repair capabilities of GC cells. In vivo experiments revealed that administration of Fn‐EVs not only promoted the growth of GC tumors and liver metastasis but also conferred resistance to oxaliplatin treatment in GC tumors.
4.3. Aggregatibacter actinomycetemcomitans
4.3.1. Characteristics of A. actinomycetemcomitans
A. actinomycetemcomitans, a gram‐negative bacterium, is linked to periodontitis and other systemic diseases, like infective endocarditis and rheumatoid arthritis. 155 , 156 It generates many virulence factors capable of inducing cell death and either triggering or evading inflammation. 157 The virulence factors of the organism include the potent leukotoxin (LtxA), LPS, cell surface‐associated materials, enzymes, and less clearly defined virulence factors that modulate the host defense activity. 158 (Figure 11) LtxA plays a significant role in A. actinomycetemcomitans pathogenicity, specifically targeting human immune cells. 159 , 160 , 161 , 162 , 163 Monocytes/macrophages exhibit high susceptibility to LtxA, resulting in their lysis through the activation of caspase 1 and the secretion of IL‐1β. 158 , 164 , 165 A. actinomycetemcomitans is the only known oral species identified to express cytolethal distending toxin (CDT). CDT primarily induces DNA damage, cell cycle arrest, and eventual apoptosis in the affected host cells. 163 , 166 Additionally, it is able to impair macrophage function by inhibiting phagocytic activity and altering cytokine balance. 167 CDT can potentially activate another pathogenic mechanism by stimulating the production of pro‐inflammatory and osteolytic cytokines in the intoxicated host cells, leading to periodontal breakdown. 168 , 169 The LPS of A. actinomycetemcomitans triggers the production of proinflammatory mediators, including IL‐1β, IL‐6, IL‐8, and tumor necrosis factor‐α (TNF‐α). 170 It also influences nitric oxide production in macrophages, contributing to pathophysiological changes during both acute and chronic inflammation, ultimately leading to tissue damage. 158 , 171 Experiments have revealed that A. actinomycetemcomitans OMVs can transport virulence factors, including LtxA, LPS, CDT, and microRNA‐size small RNAs, to host cells, thereby influencing the immune response. 172 , 173 Investigations indicate that A. actinomycetemcomitans infection induces the formation of DNA double‐strand breaks in host cells, potentially elevating the risk of carcinogenesis. 174 Analysis of gingival crevicular samples showed a robust association between the pathogen A. actinomycetemcomitans and malignancy in 32 out of 99 patients. 175
FIGURE 11.

Characteristics of Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans). A. actinomycetemcomitans possesses several virulence factors, including the potent leukotoxin (LtxA), lipopolysaccharide (LPS), cytolethal distending toxin (CDT), cell surface‐associated materials, enzymes, and less well‐defined virulence factors capable of influencing the host's defense mechanisms. Additionally, A. actinomycetemcomitans OMVs have been observed to transport these virulence factors, ultimately impacting the immune response.
4.3.2. Proposed carcinogenic mechanisms
Stimulation of oral tumor cells with P. gingivalis resulted in increased cell proliferation, interestingly, A. actinomycetemcomitans enhanced oral tumor cell death. Both bacteria and antimicrobial peptides exerted diverse effects on the transcription levels of oncogenic defensin genes and epidermal growth factor receptor (EGFR) signaling. The primary impact of the two oral pathogens on the proliferation behavior of oral tumor cells was opposite. However, they both induced similar secondary effects on the proliferation rate by modulating the extent of oncogenic important α‐defensin gene expression. 90 A. actinomycetemcomitans produces the cytolethal distending toxin (Cdt), consisting three proteins: CdtA, CdtB, and CdtC, which induce cell cycle arrest and apoptosis. In a study by Yamamoto et al., 176 it was demonstrated that delivering the CdtB protein and transfecting the cdtB gene could lead to cell cycle arrest and apoptosis in Ca9‐22 cells in vitro. They proposed the potential use of electroporation to induce apoptosis in human gingival squamous cell carcinoma by introducing the cdtB gene.
4.4. Tannerella forsythia and Treponema denticola
Tannerella forsythia (T. forsythia) and Treponema denticola (T. denticola) are anaerobic gram‐negative bacteria that are associated with the risk of periodontitis and gingivitis. 177 , 178 , 179 T. forsythia expresses many virulence factors, including trypsin‐like and PrtH proteases, sialidases like SiaH and NanH, Bacteroides surface protein A (BspA), apoptosis‐inducing activity, ɑ‐D‐glucosidase and N‐acetyl‐β‐glucosaminidase, a hemagglutinin, components of the bacterial surface layer, and methylglyoxal. 180 Studies demonstrated that T. forsythia and other pathogens could enhance bone loss and the formation of abscesses in animals. 181 , 182 T. forsythia has the capability to stimulate pro‐inflammatory cytokines, including IL‐1ß and IL‐6, through CD4+ T helper cells and TNF‐ɑ. 180 Research has indicated an upregulation of Treponema in saliva and tissue samples of patients with oral or esophageal cancer. 33 , 183 However, the precise impact of T. denticola on OSCC cells remains unclear.
4.4.1. Proposed carcinogenic mechanisms
T. denticola was found to induce the expression of TLR2, TLR4, and MyD88. Stable suppression of MyD88 significantly inhibited FAK activation induced by T. denticola and abolished the pathogen‐induced migration. These findings highlight the contribution of periodontal pathogens to a highly aggressive cancer phenotype through the interplay between TLR/MyD88 and integrin/FAK signaling pathways. 134 Peng et al. 184 found that T. denticola could invade Cal‐27 cells and directly promote cell proliferation, regulate the cell cycle, and inhibit apoptosis. T. denticola also promoted the growth of OSCC tumors in mice, and it upregulated Ki67 expression. Mechanistically, T. denticola was found to promote the development of OSCC by activating the TGF‐β pathway. Researchers have taken interest in T. denticola chymotrypsin‐like proteinase (Td‐CTLP) as a significant virulence factor of T. denticola. Nieminen et al. 185 observed the presence of Td‐CTLP in the majority of orodigestive tumor samples. They found that Td‐CTLP could activate pro MMP‐8 and ‐9 into their active forms and degrade proteinase inhibitors such as TIMP‐1, TIMP‐2, and ɑ‐1‐antichymotrypsin, along with complement C1q. Furthermore, the presence of Td‐CTLP correlated significantly with the depth of invasion, tumor diameter, and the expression levels of TLR‐7, TLR‐9, and c‐Myc. Additionally, Td‐CTLP positivity was significantly associated with invasion depth, tumor diameter and the expression of TLR‐7, TLR‐9 and c‐Myc in early‐stage mobile tongue squamous cell carcinoma. 177 Overall, T. denticola and Td‐CTLP contribute to creating a tumor tissue microenvironment favorable for invasion and metastasis.
5. MECHANISMS RELATED TO PERIODONTAL PATHOGENS‐LINKED CARCINOGENESIS
In addition to their direct impact on cancer cells, periodontal pathogens have been found to exert various effects on other cell types. These pathogens have been associated with carcinogenesis through various potential mechanisms of action, including the induction of chronic inflammation, suppression of the host's immune system, facilitation of cell invasion and proliferation, anti‐apoptotic activity, and the presence of carcinogenic substances. Understanding these diverse effects is essential for unraveling the comprehensive mechanisms underlying cancer development. In this section, we delve into the multifaceted impact of periodontal pathogens on different cell types and potential contributions to carcinogenesis to provide a holistic understanding of how periodontal pathogens contribute to cancer progression.
5.1. Induction of chronic inflammation
It has become increasingly clear that chronic inflammation significantly increases the risk of carcinogenesis. Uncontrolled inflammation may disrupt stromal integrity, which can promote cancer progression and accelerate the processes of invasion and metastasis. 17 , 93 , 186 Periodontal pathogens induce chronic inflammation, especially Fusobacterium, Porphyromonas, and Prevotella. 187 , 188 These germs have been linked to the up‐regulation of various cytokines, including IL‐1β, IL‐6, IL‐17, IL‐23, TNF‐ɑ, and other inflammatory mediators such as the CXC family, MMP‐8, and MMP‐9, which may play a role in carcinogenesis. 189 , 190 , 191 Serum IL‐6 concentrations are elevated in patients with OSCC and are correlated with a worse prognosis. 192 Additionally, there is an increase in IL‐8 levels in the saliva of OSCC patients. 193 The activation of NF‐κB is a prominent characteristic in the development of bacteria‐associated tumor. During infection, LPS has been known to trigger a robust immune response in human with gram‐negative bacteria. Pattern recognition receptors (PRRs) like TLRs, triggering the NF‐κB signaling pathway and inducing the production of inflammatory‐associated cytokine. This activation plays a pivotal role in the inflammation induced by bacteria and contributes to the carcinogenesis process. 12 , 194 , 195
P. gingivalis activated MAPK and NF‐κB signaling pathways, leading to the production of IL‐8. Additionally, it stimulates the JAK/c‐Jun signaling axis, resulting in elevated levels of IL‐1β and IL‐6. 196 , 197 Furthermore, it promotes inflammatory responses through TLR4/TLR2 pathways in host cells. 198 , 199 , 200 In response to infective agents, the host's natural defense mechanism activates inflammatory signaling cascades, utilizing miRNA species as alternative genetic inhibitory transcriptional endpoints. 201 P. gingivalis has the capability to suppress the expression of miR‐205‐5p, leading to the activation of JAK/STAT by upregulating IL6ST 202 (Figure 12).
FIGURE 12.

Mechanisms linking P. gingivalis and inflammation. P. gingivalis can promote inflammatory responses via toll like receptor (TLR) 4/TLR2. It can activate mitogen‐activated protein kinases (MAPK) and kappa‐light‐chain‐enhancer of activated B cells (NF‐κB) signaling pathways to express interleukin‐8 (IL‐8) and activate the janus kinase (JAK)/c‐Jun signaling axis to elevate IL‐1β and IL‐6. It also can inhibit miR‐205‐5p expression from activating JAK/signal transducer and activator of transcription (STAT) by upregulating IL6ST.
F. nucleatum is a robust inducer of inflammatory cytokines. F. nucleatum infection in gingival epithelial cells (GECs) triggers the activation of NF‐κB, which translocates to the nucleus and enhances the expression of pro‐inflammatory genes, including those responsible for pro‐IL‐1β. 93 , 203 LPS derived from F. nucleatum is responsible for cellular‐inflammatory response and immune system activation in periodontal diseases. 12 , 204 This leads to the production of inflammatory cytokines such as IL‐1α, IL‐1β, IL‐6, IL‐8, and MMPs through the activation and translocation of NF‐κB into the nucleus. 12 , 205 , 206 , 207 F. nuleatum also intensifies TNF‐α production from GECs and macrophages, contributing to free radical generation during sepsis. 206 , 208 NLRP10, the smallest human NLRP, exhibits both anti‐ and pro‐inflammatory functions. 209 It activates the extracellular regulated protein kinases (ERK) signaling pathway in human epithelial cells infected with F. nuleatum, leading to an elevation in the levels of the pro‐inflammatory cytokine IL‐1α. 209 Krisanaprakornkit et al. 210 demonstrated that LPS extracted from F. nucleatum's cell wall, along with TNF‐α, induces human β‐defensin 2 (hBD2) and IL‐8. Another study demonstrated that the infection of gingival squamous cells with F. nucleatum induces the expression of hBD2 and hBD3. 211 As an adhesive bacterium, F. nuleatum engages in co‐aggregates with various microbial species in the oral cavity. 114 FadA assumes a pivotal role in inducing of tumorigenic responses by binding to E‐cadherin and activating β‐catenin signaling. This activation leads to a differential regulation of inflammatory and oncogenic responses. 140
In addition, F. nucleatum infection activates the nucleotide‐binding domain and leucine‐rich‐repeat‐containing family member X1 (NLRX1). This stimulation additionally boosts the NLRP3 inflammasome, initiating caspase‐1 activation, culminating in the processing and liberation of IL‐1β in GECs. 203 , 212 The inflammation is amplified through the release of the danger signals apoptosis‐associated speck‐like protein containing a carboxy‐terminal CARD (ASC) and caspase‐1, resulting in the secretion of high mobility group box‐1 (HMGB1). 203 HMGB1, a DNA‐binding nuclear protein, is implicated in various inflammatory disorders, cell adhesion, and migration. 213 The interaction between HMGB1 and advanced glycation end‐product (RAGE) may contribute to oral inflammation and the development of oral cancer 204 (Figure 13).
FIGURE 13.

Mechanisms that link inflammation and F. nucleatum, T. denticola, T. forsythia and A. actinomycetemcomitans. The interaction between fusobacterial adhesin (FadA) and E‐cadherin triggers the activation of the β‐catenin. F. nucleatum initiates the toll like receptor (TLR) pathway, activating nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NF‐κB), leading to an increase in inflammatory cytokine (interleukin‐6 (IL‐6), IL‐8, IL‐10, IL‐18, tumor necrosis factor‐α (TNF‐α), and human β‐defensins (hBDs)). Additionally, F. nucleatum can elevate IL‐1α level by modulating the extracellular regulated protein kinases (ERK) signaling pathway. Furthermore, F. nucleatum infection triggers the activation of the nucleotide‐binding domain and leucine‐rich‐repeat‐containing family member X1 (NLRX1), which further promotes the formation of NOD‐, LRR‐, and pyrin domain‐containing protein 3 (NLRP3) inflammasome, leading to the stimulation of IL‐1ß. Co‐infection with T. denticola and F. nucleatum induced expression of hBDs and IL‐8 by inhibiting endo‐lysosomal maturation and reactive oxygen species (ROS). T. forsythia induces various proinflammatory cytokines, including IL‐1α, IL‐1β, IL‐6, TNF‐α, IL‐8, and IL‐24 in different cell types. Clinical isolates of A. actinomycetemcomitans may interfere with neutrophil function by immunosuppressing IL‐8 responses.
T. denticola fails to stimulate the generation of innate immune mediators, such as IL‐6, IL‐8, and hBDs from epithelial cells. 214 Conversely, co‐infection with T. denticola disrupts endo‐lysosomal maturation of F. nucleatum‐containing endosomes, causing a buildup of intracellular reactive oxygen species (ROS). This accumulation leads to inhibition of hBDs and IL‐8 expression in GECs. 215 Upon colonization, T. denticola can establish its niche, creating a conducive environment for other plaque bacteria. 214 Pahumunto et al. 216 analysis of 50 clinical strains and 7 reference strains of A. actinomycetemcomitans revealed that JP2‐like leukotoxin promoter gene, NS1, and NS2 from clinical isolates may interfere with neutrophil function by inducing minimal and immunosuppressing IL‐8 responses, thereby enhancing bacteria survival and virulence. T. forsythia is recognized for inducing various proinflammatory cytokines, including IL‐1α, IL‐1β, IL‐6, TNF‐α, IL‐8, and IL‐24 in different cell types 217 (Figure 13).
5.2. Inhibition of the host's immune system
The interaction between the host immune system and microbiota may contribute to the progression of cancer. 187 Periodontal pathogens, such as P. gingivalis and F. nucleatum, not only initiate proinflammatory immune responses but also trigger or induce immunosuppressive reactions, undermining the anti‐tumor immunity. 218 , 219 Bacterial toxins, like CDT secreted by A. actinomycetemcomitans, disrupt the host response by influencing phagocytic activity and altering the balance of cytokines. 167 A. actinomycetemcomitans' outer membrane protein 29 (OMP29) and OMP29par can subvert the host immune response by suppressing C‐X‐C Motif Chemokine Ligand 8 (CXCL‐8), crucial in tumor angiogenesis, and modulating genes related to apoptosis and inflammatory response in GECs. 220 The tumor microenvironment, characterized by immunosuppression, plays a pivotal role in tumor immune escape mechanisms. 221 PD‐L1 is associated with cell‐mediated immune responses, exerting crucial immune‐regulatory functions during infection and self‐recognition. 222 High PD‐L1 expression levels were observed in tissue samples from OSCC. 223 Groeger et al. 224 illustrated that the membrane fraction of P. gingivalis stimulates the expression of the PD‐L1 in squamous carcinoma cells and gingival keratinocytes. Peptidoglycan (PDG) from P. gingivalis was found to induce PD‐L1 expression in various cancer cells, including OSCCs and colon cancer cells, dependent on NOD1 and NOD2, and the activation of RIP2 and MAPK signaling pathways. 107 While upregulation of the PD‐1/PD‐L1 pathway may be necessary to limit host damage, inhibiting T cell responses undoubtedly benefits invading pathogens. 225
P. gingivalis has been demonstrated to dampen the IFN‐γ‐stimulated release of CXCL9, CXCL10, and CXCL11 from epithelial cells. The inhibition of chemokine expression operates at the gene transcription level and correlates with the down‐regulation of IRF‐1 and a decrease in STAT1 levels. 226 The nucleoside diphosphate kinase (NDK) secreted by P. gingivalis has been implicated in promoting tumorigenesis. NDK inhibits ATP activation of purinergic receptor (P2X7), subsequently suppressing the production of IL‐1ß in the epithelium. 227 , 228 Arjunan et al. 229 demonstrated that P. gingivalis, through Mfa1 and FimA fimbriae, fosters immunosuppression and oncogenic cell proliferation in myeloid‐derived dendritic suppressor cells.
Neutrophils play a vital role in defending periodontal tissues through the innate immune system, engaging in phagocytosis, bacterial killing, and digestion. 230 , 231 P. gingivalis disrupts the antimicrobial response of macrophages, hindering antigen presentation and T‐cell activation by manipulating the C5aR/TLR2 crosstalk. This modulation involves the regulation of MyD88 and PI3K signaling pathways. The downstream molecule MyD88 undergoes degradation via ubiquitination, inhibiting the host‐protective antimicrobial response. 232 The crosstalk between C5aR and TLR2, triggered by P. gingivalis, also activates PI3K, leading to reduced phagocytosis of P. gingivalis by neutrophils. 232 , 233 , 234 Additionally, P. gingivalis citrullinates histone H3, impairing the bactericidal components in neutrophil extracellular traps (NETs) and thereby weakening their effectiveness. 87 , 232 NETs, web‐like structures released by neutrophils during infection, aid in capturing and killing pathogens. 232 Gingipains activate protease‐activated receptor 2 (PAR‐2), triggering Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation, increasing ROS levels, and promoting NETs formation. 232 , 235 However, NETs induced by P. gingivalis lack bactericidal activity. 235 A. actinomycetemcomitans can also induce NETs formation and the release of MMP‐8 and ‐9 in neutrophils alone, potentially contributing to tissue destruction and disease progression 236 (Figure 14).
FIGURE 14.

P. gingivalis and A. actinomycetemcomitans escape the killing by neutrophils. P. gingivalis triggers a crosstalk between toll like receptor 2 (TLR2) and C5aR, modulating the signaling pathways of myeloid differentiation primary response 88 (MyD88) and phosphatidylinositol 3‐kinase (PI3K). Furthermore, P. gingivalis employs histone H3 citrullination, facilitated by PPAD, to evade neutrophil extracellular traps (NETs). Additionally, gingipains from P. gingivalis activate protease‐activated receptor 2 (PAR‐2) and Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, resulting in the formation of NETs lacking bactericidal activity. A. actinomycetemcomitans similarly induces NETs formation and the secretion of matrix metalloprotease (MMP)‐8 and MMP‐9 in neutrophils, potentially contributing to tissue destruction and disease progression. Adapted from Ref. [232].
P. gingivalis employs various strategies to evade immune detection and manipulate host responses. The utilization of lipid A structures enables evasion from immune detection via TLR4. 237 Additionally, gingipains produced by P. gingivalis degrade CD14 on macrophage surfaces, hindering the phagocytosis of infected apoptotic cells. 232 , 238 , 239 Furthermore, the fimbria and sialidase of P. gingivalis contribute to immune evasion through the exploitation of CXCR4 and CR3. The binding to CXCR4 activates cAMP‐dependent protein kinase A (PKA) signaling, which inhibits TLR2‐mediated proinflammatory and antimicrobial responses. 240 , 241 , 242 During the inflammatory process, P. gingivalis triggers IL‐12 secretion, playing a crucial role in macrophage clearance of bacteria and stimulating T cells to produce IFN‐γ. 241 , 243 The release of sialidase enhances P. gingivalis virulence, reducing IL‐12 production and influencing T‐cell responses. 241 Moreover, P. gingivalis inhibits T‐cell antimicrobial responses by down‐regulating IL‐2 expression, achieved through the prevention of protein kinase C and p38 phosphorylation and AP‐1. 244 Additionally, the up‐regulation of IL‐10 by P. gingivalis in macrophages inhibits antigen presentation and T‐cell activation. 232 The increased IL‐10 production induced by P. gingivalis activates PD‐L1 on macrophages and PD‐1 on CD4+T cell surfaces, indicating multiple inhibitory mechanisms employed by P. gingivalis to evade the host's immune response. 245
5.3. Cell invasion and proliferation
In addition to prolonged cellular survival, P. gingivalis induces heightened proliferation in infected epithelial cells. 246 The FimA fimbriae of P. gingivalis elevate the proliferation rate of GECs, accelerating progression through the S‐phase. This effect is associated with a reduction in p53 levels and activation of the PI3K pathway. 247 P. ginglivalis LPS enhanced the proliferation of gingival stem/progenitor cells without compromising their regenerative capacity through NF‐κB. 248 Exposure of immortalized OKF6 cells to P. gingivalis resulted in changes in the molecular profile of proteins associated with cell proliferation, EMT, stem cell generation, migration, invasion, and resistance to anoikis. This alteration was accompanied by increased rates of proliferation, which correlated with the activation of the PI3K/Akt signaling pathway and the mTOR pathway. 249 F. nucleatum enhances cell proliferation by activating p38, followed by HSP27, which plays a significant role in cell proliferation, differentiation, and oncogenesis. 250 This activation is associated with the secretion of MMP‐9 and MMP‐13, both contributing to the invasion and metastasis phenotype. 251 Moreover, F. nucleatum stimulation promotes cellular migration through the activation of epithelial and endothelial tyrosine kinase/bone marrow X kinase (Etk/BMX), S6 kinase p70, and RhoA kinase. 251 This interaction with human epithelial cells during F. nucleatum infection results in the up‐regulation of 12 kinases, as evaluated by the Kinetworks immunoblotting system, which are involved in cell migration, proliferation, and cell survival signaling. 251 , 252 The interaction of F. nucleatum with the epithelial cell surface CD46, as observed by Mahtout et al., 253 may contribute to the elevation of proinflammatory mediators and MMPs in periodontal sites, subsequently modulating tissue destruction. Jia et al. 254 reported that the cultured media derived from P. gingivalis can lead to the malignant transformation of normal esophageal epithelial cells. This transformation is characterized by increased proliferation and migration of the cells, as well as the appearance of aneuploid cells. In addition, mouse esophageal epithelial cells exposed to P. gingivalis cultured media exhibited disordered arrangement, increased proliferation, and weakened expression of Claudin 1 and Claudin 4, both associated with dysplasia. Furthermore, the expression of cancer‐related genes was upregulated, while tight junction‐related genes were downregulated. Notably, aberrant activation of the sonic hedgehog pathway was observed, and its inhibition attenuated the pathogenic effect of P. gingivalis cultured media in normal esophageal epithelium (Figure 15).
FIGURE 15.

Mechanisms related to periodontal pathogens and cell invasion and proliferation. P. gingivalis induces cell proliferation by reducing p53 levels and activating the PI3K pathway, while F. nucleatum enhances proliferation through p38 activation and MMP secretion. Both bacteria promote migration via various kinase pathways. P. gingivalis cultured media lead to malignant transformation of esophageal cells by cancer‐related genes, involving aberrant sonic hedgehog pathway activation.
5.4. Anti‐apoptotic activity
Apoptosis plays a crucial role in various biological processes, including normal cell turnover, immune system development and function, embryological development, and responses to viral and bacterial infections. Dysregulation of apoptosis, either insufficient or excessive, contributes to the pathogenesis of many cancer types. 255 P. gingivalis exerts an anti‐apoptotic effect by influencing multiple pathways. The suppression of apoptosis is facilitated by the degradation of ATP mediated by NDK, an enzyme released by P. gingivalis, specifically inhibiting apoptosis dependent on ATP activation of P2X7 receptors. 256 Additionally, the phosphorylation of HSP27 by P. gingivalis‐derived NDK imparts an anti‐apoptotic phenotype to primary GECs, underscoring the significance of HSP27 in mitigating host cell apoptosis induced by P. gingivalis. 228 , 257 Another mechanism by which P. gingivalis can block the apoptotic pathway in GECs is through modulating the JAK/STAT pathway, leading to the blockade of caspase‐3 activation and subsequent control of the intrinsic mitochondrial cell death pathways. 258 Furthermore, P. gingivalis inhibits apoptosis and enhances the survival of GECs by activating Akt via PI3K, potentially through the inhibition of mitochondrial permeability alterations. 259 Jewett et al. 260 illustrated that F. nucleatum's immunosuppressive role primarily stems from its ability to trigger apoptotic cell death in peripheral blood mononuclear cells (PBMCs) and polymorphonuclear cells (PMNs).
5.5. Carcinogenic substances
Metabolic derivatives from certain periodontal pathogens, such as organic acids, volatile sulfur compounds (VSC), and ROS, possess the potential to induce DNA damage, mutagenesis, secondary hyperproliferation of the cells, metastasis, and cancer progression. 187 , 218 , 261 Microorganisms involved in alcohol metabolism to acetaldehyde can impact cancer development. 187 VSC‐producing periodontal pathogens like P. gingivalis, Prevotella intermedia, A. actinomycetemcomitans, and F. nucleatum generate VSCs like hydrogen sulfide, methyl mercaptan, dimethyl sulfide, and dimethyl disulfide. 188 Research indicates that these VSCs influence epithelial cell proliferation and induce apoptosis through the activation of the mitochondrial pathway. 262 , 263 Notably, VSCs, including methyl mercaptan, contribute to connective tissue breakdown and inflammation by stimulating the release of IL‐1ß from mononuclear cells. 264 , 265
ROS typically function as signals to regulate cell proliferation and survival, playing a pivotal role in biological processes and host innate immune responses when induced by pathogens. Both a lower and a higher percentage of ROS can have adverse effects. When ROS levels are low, they fail to support proper cellular functioning by regulating numerous biochemical reactions. 266 However, an excessive amount of ROS can damage cellular components, disrupt normal cellular processes, and lead to cell death. 267 , 268 , 269 Interestingly, ROS dynamically influences the tumor microenvironment, initiating cancer angiogenesis, metastasis, and survival at different concentrations. 270 P. gingivalis exhibits the capacity to stimulate ROS production, leading to JAK2 phosphorylation and the upregulation of proinflammatory cytokines such as IL‐6 and IL‐1ß. 196 The secretion of P. gingivalis NDK may modulate eATP‐induced cytosolic and mitochondrial ROS, and the antioxidant glutathione response (AGR) generated via the P2X7/NADPH‐oxidase interactome. 271 Additionally, P. gingivalis‐triggered ferritinophagy induces ROS production and inflammatory responses in periodontal ligament fibroblasts. 272 F. nucleatum has the ability to induce ROS in colon lining epithelial cells, OSCCs, and human gingival fibroblasts. 72 , 273 , 274 Previous study shows that F. nuleatum can inhibit human gingival fibroblast proliferation and promote cell apoptosis, ROS production, and inflammatory cytokine production, partly through the activation of the AKT/MAPK and NF‐κB signaling pathways. 273 Okinaga et al. 275 demonstrated that A. actinomycetemcomitans induces IL‐1β production in RAW 264 cells by generating ROS and cathepsin B. T. forsythia can stimulate ROS, subsequently inducing the expression of IL‐24 217 (Figure 16).
FIGURE 16.

Periodontal pathogen's metabolic derivatives affect cancer development. Periodontal pathogens (e.g., P. gingivalis, A. actinomycetemcomitans, and F. nucleatum) produce volatile sulfur compounds (VSCs) impact cell proliferation, induce apoptosis, and trigger the release of inflammatory cytokines. Additionally, P. gingivalis, A. actinomycetemcomitans, T. forsythia and F. nucleatum can stimulate the production of reactive oxygen species (ROS), exerting a dynamic influence on the tumor microenvironment.
6. CONCLUSIONS
Cancer, characterized by its multi‐step and generally slowly progressing, involves a complex interplay of genetic and environmental factors. 246 Periodontal pathogens exert direct or indirect effects on chronic inflammation, the human immune response, cell invasion and proliferation, anti‐apoptotic activity, and the presence of carcinogenic substances, all of which precede cancer development. Although establishing a clear link between periodontal pathogens and carcinoma development poses a challenge, specific pathogens such as P. gingivalis, F. nucleatum, A. actinomycetemcomitans, T. forsythia, and T. denticola have been identified to promote diverse signaling pathways that may contribute to carcinoma development. Further exploration of the pathogenic mechanisms employed by periodontal pathogens is essential, holding the potential to unveil various pathways crucial for diagnostic, preventive, and therapeutic strategies. These insights will not only enhance the efficacy of treatment but also contribute to advancements in survival outcomes.
AUTHOR CONTRIBUTIONS
All authors contributed to the article and approved the submitted version.
CONFLICT OF INTEREST STATEMENT
The authors declare that they have no conflicts of interest.
ACKNOWLEDGMENTS
Yuxi Zhou was supported by China Scholarship Council (CSC) under Grant CSC No.201908450033 and Förderprogramm der Forschungsgemeinschaft Dental e.V. (FGD). Figures were created with BioRender.com. Open Access funding enabled and organized by Projekt DEAL.
Zhou Y, Meyle J, Groeger S. Periodontal pathogens and cancer development. Periodontol 2000. 2024;96:112‐149. doi: 10.1111/prd.12590
DATA AVAILABILITY STATEMENT
Data sharing is not applicable to this article as no new data were created or analyzed in the current study.
REFERENCES
- 1. Meyle J, Chapple I. Molecular aspects of the pathogenesis of periodontitis. Periodontol 2000. 2015;69:7‐17. [DOI] [PubMed] [Google Scholar]
- 2. Pihlstrom BL, Michalowicz BS, Johnson NW. Periodontal diseases. Lancet. 2005;366:1809‐1820. [DOI] [PubMed] [Google Scholar]
- 3. Kinane DF. Causation and pathogenesis of periodontal disease. Periodontol 2000. 2001;25:8‐20. [DOI] [PubMed] [Google Scholar]
- 4. de Martel C, Ferlay J, Franceschi S, et al. Global burden of cancers attributable to infections in 2008: a review and synthetic analysis. Lancet Oncol. 2012;13:607‐615. [DOI] [PubMed] [Google Scholar]
- 5. Sears CL, Garrett WS. Microbes, microbiota, and colon cancer. Cell Host Microbe. 2014;15:317‐328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Nwizu N, Wactawski‐Wende J, Genco RJ. Periodontal disease and cancer: epidemiologic studies and possible mechanisms. Periodontol 2000. 2020;83:213‐233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Arweiler NB, Netuschil L. The oral microbiota. Adv Exp Med Biol. 2016;902:45‐60. [DOI] [PubMed] [Google Scholar]
- 8. Dodet B. Microbiota and representations of the human body. Med Sci. 2016;32:1003‐1008. [DOI] [PubMed] [Google Scholar]
- 9. Bui FQ, Almeida‐da‐Silva CLC, Huynh B, et al. Association between periodontal pathogens and systemic disease. Biom J. 2019;42:27‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Bale BF, Doneen AL, Vigerust DJ. High‐risk periodontal pathogens contribute to the pathogenesis of atherosclerosis. Postgrad Med J. 2017;93:215‐220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209‐249. [DOI] [PubMed] [Google Scholar]
- 12. Chattopadhyay I, Verma M, Panda M. Role of oral microbiome signatures in diagnosis and prognosis of oral cancer. Technol Cancer Res Treat. 2019;18:1533033819867354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Lindemann A, Takahashi H, Patel AA, Osman AA, Myers JN. Targeting the DNA damage response in OSCC with TP53 mutations. J Dent Res. 2018;97:635‐644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Blot WJ, McLaughlin JK, Winn DM, et al. Smoking and drinking in relation to oral and pharyngeal cancer. Cancer Res. 1988;48:3282‐3287. [PubMed] [Google Scholar]
- 15. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet‐Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87‐108. [DOI] [PubMed] [Google Scholar]
- 16. Danaei G, Vander Hoorn S, Lopez AD, Murray CJ, Ezzati M. Causes of cancer in the world: comparative risk assessment of nine behavioural and environmental risk factors. Lancet. 2005;366:1784‐1793. [DOI] [PubMed] [Google Scholar]
- 17. Greten FR, Grivennikov SI. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity. 2019;51:27‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883‐899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer‐related inflammation. Nature. 2008;454:436‐444. [DOI] [PubMed] [Google Scholar]
- 20. Nagy KN, Sonkodi I, Szöke I, Nagy E, Newman HN. The microflora associated with human oral carcinomas. Oral Oncol. 1998;34:304‐308. [PubMed] [Google Scholar]
- 21. Chang C, Geng F, Shi X, et al. The prevalence rate of periodontal pathogens and its association with oral squamous cell carcinoma. Appl Microbiol Biotechnol. 2019;103:1393‐1404. [DOI] [PubMed] [Google Scholar]
- 22. Schmidt BL, Kuczynski J, Bhattacharya A, et al. Changes in abundance of oral microbiota associated with oral cancer. PLoS One. 2014;9:e98741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Marshall BJ, Goodwin CS, Warren JR, et al. Prospective double‐blind trial of duodenal ulcer relapse after eradication of Campylobacter pylori . Lancet. 1988;2:1437‐1442. [DOI] [PubMed] [Google Scholar]
- 24. Irfan M, Delgado RZR, Frias‐Lopez J. The oral microbiome and cancer. Front Immunol. 2020;11:591088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Virchow R. Die krankhaften Geschwülste; dreissig Vorlesungen: gehalten während des Wintersemesters 1862–1863 an der Universität zu Berlin. Verlag von August Hirschwald; 1863. [Google Scholar]
- 26. Kipanyula MJ, Seke Etet PF, Vecchio L, Farahna M, Nukenine EN, Nwabo Kamdje AH. Signaling pathways bridging microbial‐triggered inflammation and cancer. Cell Signal. 2013;25:403‐416. [DOI] [PubMed] [Google Scholar]
- 27. Lax AJ. Opinion: bacterial toxins and cancer—a case to answer? Nat Rev Microbiol. 2005;3:343‐349. [DOI] [PubMed] [Google Scholar]
- 28. Alsahafi E, Begg K, Amelio I, et al. Clinical update on head and neck cancer: molecular biology and ongoing challenges. Cell Death Dis. 2019;10:540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Hieken TJ, Chen J, Hoskin TL, et al. The microbiome of aseptically collected human breast tissue in benign and malignant disease. Sci Rep. 2016;6:30751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Wang H, Altemus J, Niazi F, et al. Breast tissue, oral and urinary microbiomes in breast cancer. Oncotarget. 2017;8:88122‐88138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Parhi L, Alon‐Maimon T, Sol A, et al. Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat Commun. 2020;11:3259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Rogers JE, Sewastjanow‐Silva M, Waters RE, Ajani JA. Esophageal cancer: emerging therapeutics. Expert Opin Ther Targets. 2022;26:107‐117. [DOI] [PubMed] [Google Scholar]
- 33. Narikiyo M, Tanabe C, Yamada Y, et al. Frequent and preferential infection of Treponema denticola, Streptococcus mitis, and Streptococcus anginosus in esophageal cancers. Cancer Sci. 2004;95:569‐574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Yamamura K, Baba Y, Nakagawa S, et al. Human microbiome Fusobacterium nucleatum in esophageal cancer tissue is associated with prognosis. Clin Cancer Res. 2016;22:5574‐5581. [DOI] [PubMed] [Google Scholar]
- 35. Gao S, Li S, Ma Z, et al. Presence of Porphyromonas gingivalis in esophagus and its association with the clinicopathological characteristics and survival in patients with esophageal cancer. Infect Agent Cancer. 2016;11:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Kawasaki M, Ikeda Y, Ikeda E, et al. Oral infectious bacteria in dental plaque and saliva as risk factors in patients with esophageal cancer. Cancers. 2021;127:512‐519. [DOI] [PubMed] [Google Scholar]
- 37. Lund Håheim L, Thelle DS, Rønningen KS, Olsen I, Schwarze PE. Low level of antibodies to the oral bacterium Tannerella forsythia predicts bladder cancers and Treponema denticola predicts colon and bladder cancers: a prospective cohort study. PLoS One. 2022;17:e0272148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Kostic AD, Gevers D, Pedamallu CS, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012;22:292‐298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Mima K, Nishihara R, Qian ZR, et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut. 2016;65:1973‐1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Ahn J, Sinha R, Pei Z, et al. Human gut microbiome and risk for colorectal cancer. J Natl Cancer Inst. 2013;105:1907‐1911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Kerdreux M, Edin S, Löwenmark T, et al. Porphyromonas gingivalis in colorectal cancer and its association to patient prognosis. J Cancer. 2023;14:1479‐1485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Nakatsu G, Li X, Zhou H, et al. Gut mucosal microbiome across stages of colorectal carcinogenesis. Nat Commun. 2015;6:8727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Hale VL, Chen J, Johnson S, et al. Shifts in the fecal microbiota associated with adenomatous polyps. Cancer Epidemiol Biomarkers Prev. 2017;26:85‐94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Flemer B, Warren RD, Barrett MP, et al. The oral microbiota in colorectal cancer is distinctive and predictive. Gut. 2018;67:1454‐1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet. 2020;396:635‐648. [DOI] [PubMed] [Google Scholar]
- 46. Chen XH, Wang A, Chu AN, Gong YH, Yuan Y. Mucosa‐associated microbiota in gastric cancer tissues compared with non‐cancer tissues. Front Microbiol. 2019;10:1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Coker OO, Dai Z, Nie Y, et al. Mucosal microbiome dysbiosis in gastric carcinogenesis. Gut. 2018;67:1024‐1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Klein AP. Pancreatic cancer epidemiology: understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol. 2021;18:493‐502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Michaud DS, Izard J, Wilhelm‐Benartzi CS, et al. Plasma antibodies to oral bacteria and risk of pancreatic cancer in a large European prospective cohort study. Gut. 2013;62:1764‐1770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Fan X, Alekseyenko AV, Wu J, et al. Human oral microbiome and prospective risk for pancreatic cancer: a population‐based nested case‐control study. Gut. 2018;67:120‐127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Vogtmann E, Han Y, Caporaso JG, et al. Oral microbial community composition is associated with pancreatic cancer: a case‐control study in Iran. Cancer Med. 2020;9:797‐806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Farrell JJ, Zhang L, Zhou H, et al. Variations of oral microbiota are associated with pancreatic diseases including pancreatic cancer. Gut. 2012;61:582‐588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Lu H, Ren Z, Li A, et al. Deep sequencing reveals microbiota dysbiosis of tongue coat in patients with liver carcinoma. Sci Rep. 2016;6:33142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Yan X, Yang M, Liu J, et al. Discovery and validation of potential bacterial biomarkers for lung cancer. Am J Cancer Res. 2015;5:3111‐3122. [PMC free article] [PubMed] [Google Scholar]
- 55. Tsay JJ, Wu BG, Badri MH, et al. Airway microbiota is associated with upregulation of the PI3K pathway in lung cancer. Am J Respir Crit Care Med. 2018;198:1188‐1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Zhang W, Luo J, Dong X, et al. Salivary microbial dysbiosis is associated with systemic inflammatory markers and predicted oral metabolites in non‐small cell lung cancer patients. J Cancer. 2019;10:1651‐1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Liu Y, Yuan X, Chen K, et al. Clinical significance and prognostic value of Porphyromonas gingivalis infection in lung cancer. Transl Oncol. 2021;14:100972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE, Grandis JR. Head and neck squamous cell carcinoma. Nat Rev Dis Primers. 2020;6:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Zhang Z, Yang J, Feng Q, et al. Compositional and functional analysis of the microbiome in tissue and saliva of oral squamous cell carcinoma. Front Microbiol. 2019;10:1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Zhao H, Chu M, Huang Z, et al. Variations in oral microbiota associated with oral cancer. Sci Rep. 2017;7:11773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Mager DL, Haffajee AD, Devlin PM, Norris CM, Posner MR, Goodson JM. The salivary microbiota as a diagnostic indicator of oral cancer: a descriptive, non‐randomized study of cancer‐free and oral squamous cell carcinoma subjects. J Transl Med. 2005;3:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Katz J, Onate MD, Pauley KM, Bhattacharyya I, Cha S. Presence of Porphyromonas gingivalis in gingival squamous cell carcinoma. Int J Oral Sci. 2011;3:209‐215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Al‐Hebshi NN, Nasher AT, Maryoud MY, et al. Inflammatory bacteriome featuring Fusobacterium nucleatum and Pseudomonas aeruginosa identified in association with oral squamous cell carcinoma. Sci Rep. 2017;7:1834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Zhang L, Liu Y, Zheng HJ, Zhang CP. The oral microbiota may have influence on oral cancer. Front Cell Infect Microbiol. 2019;9:476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Peters BA, Wu J, Pei Z, et al. Oral microbiome composition reflects prospective risk for esophageal cancers. Cancer Res. 2017;77:6777‐6787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. How KY, Song KP, Chan KG. Porphyromonas gingivalis: an overview of periodontopathic pathogen below the gum line. Front Microbiol. 2016;7:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Lin J, Huang D, Xu H, Zhan F, Tan X. Macrophages: a communication network linking Porphyromonas gingivalis infection and associated systemic diseases. Front Immunol. 2022;13:952040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Atanasova KR, Yilmaz O. Looking in the Porphyromonas gingivalis cabinet of curiosities: the microbium, the host and cancer association. Mol Oral Microbiol. 2014;29:55‐66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Olsen I, Yilmaz Ö. Possible role of Porphyromonas gingivalis in orodigestive cancers. J Oral Microbiol. 2019;11:1563410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Amano A, Nakagawa I, Okahashi N, Hamada N. Variations of Porphyromonas gingivalis fimbriae in relation to microbial pathogenesis. J Periodontal Res. 2004;39:136‐142. [DOI] [PubMed] [Google Scholar]
- 71. Moreno S, Contreras A. Functional differences of Porphyromonas gingivalis Fimbriae in determining periodontal disease pathogenesis: a literature review. Colomb Med. 2013;44:48‐56. [PMC free article] [PubMed] [Google Scholar]
- 72. Atanasova KR, Yilmaz O. Prelude to oral microbes and chronic diseases: past, present and future. Microbes Infect. 2015;17:473‐483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Holden JA, Attard TJ, Laughton KM, Mansell A, O'Brien‐Simpson NM, Reynolds EC. Porphyromonas gingivalis lipopolysaccharide weakly activates M1 and M2 polarized mouse macrophages but induces inflammatory cytokines. Infect Immun. 2014;82:4190‐4203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Xu W, Zhou W, Wang H, Liang S. Roles of Porphyromonas gingivalis and its virulence factors in periodontitis. Adv Protein Chem Struct Biol. 2020;120:45‐84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Potempa JAN, Banbula A, Travis JIM. Role of bacterial proteinases in matrix destruction and modulation of host responses. Periodontol 2000. 2003;24:153‐192. [DOI] [PubMed] [Google Scholar]
- 76. Bao K, Belibasakis GN, Thurnheer T, Aduse‐Opoku J, Curtis MA, Bostanci N. Role of Porphyromonas gingivalis gingipains in multi‐species biofilm formation. BMC Microbiol. 2014;14:258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Kitamura Y, Matono S, Aida Y, Hirofuji T, Maeda K. Gingipains in the culture supernatant of Porphyromonas gingivalis cleave CD4 and CD8 on human T cells. J Periodontal Res. 2002;37:464‐468. [DOI] [PubMed] [Google Scholar]
- 78. Lourbakos A, Potempa J, Travis J, et al. Arginine‐specific protease from Porphyromonas gingivalis activates protease‐activated receptors on human oral epithelial cells and induces interleukin‐6 secretion. Infect Immun. 2001;69:5121‐5130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Oido‐Mori M, Rezzonico R, Wang PL, et al. Porphyromonas gingivalis gingipain‐R enhances interleukin‐8 but decreases gamma interferon‐inducible protein 10 production by human gingival fibroblasts in response to T‐cell contact. Infect Immun. 2001;69:4493‐4501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Tam V, O'Brien‐Simpson NM, Chen YY, Sanderson CJ, Kinnear B, Reynolds EC. The RgpA‐Kgp proteinase‐adhesin complexes of Porphyromonas gingivalis inactivate the Th2 cytokines interleukin‐4 and interleukin‐5. Infect Immun. 2009;77:1451‐1458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Yun PL, Decarlo AA, Collyer C, Hunter N. Hydrolysis of interleukin‐12 by Porphyromonas gingivalis major cysteine proteinases may affect local gamma interferon accumulation and the Th1 or Th2 T‐cell phenotype in periodontitis. Infect Immun. 2001;69:5650‐5660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Wang T, Ishikawa T, Sasaki M, Chiba T. Oral and gut microbial dysbiosis and non‐alcoholic fatty liver disease: the central role of Porphyromonas gingivalis . Front Med. 2022;9:822190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Gui MJ, Dashper SG, Slakeski N, Chen YY, Reynolds EC. Spheres of influence: Porphyromonas gingivalis outer membrane vesicles. Mol Oral Microbiol. 2016;31:365‐378. [DOI] [PubMed] [Google Scholar]
- 84. Ho MH, Chen CH, Goodwin JS, Wang BY, Xie H. Functional advantages of Porphyromonas gingivalis vesicles. PLoS One. 2015;10:e0123448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Zhang Z, Liu D, Liu S, Zhang S, Pan Y. The role of Porphyromonas gingivalis outer membrane vesicles in periodontal disease and related systemic diseases. Front Cell Infect Microbiol. 2020;10:585917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Groeger S, Domann E, Gonzales JR, Chakraborty T, Meyle J. B7‐H1 andB7‐DC receptors of oral squamous carcinoma cells are upregulated by Porphyromonas gingivalis . Immunobiology. 2011;216:1302‐1310. [DOI] [PubMed] [Google Scholar]
- 87. Liu S, Zhou X, Peng X, et al. Porphyromonas gingivalis promotes immunoevasion of oral cancer by protecting cancer from macrophage attack. J Immunol. 2020;205:282‐289. [DOI] [PubMed] [Google Scholar]
- 88. Wen L, Mu W, Lu H, et al. Porphyromonas gingivalis promotes oral squamous cell carcinoma progression in an immune microenvironment. J Dent Res. 2020;99:666‐675. [DOI] [PubMed] [Google Scholar]
- 89. Aral K, Milward MR, Gupta D, Cooper PR. Effects of Porphyromonas gingivalis and Fusobacterium nucleatum on inflammasomes and their regulators in H400 cells. Mol Oral Microbiol. 2020;35:158‐167. [DOI] [PubMed] [Google Scholar]
- 90. Hoppe T, Kraus D, Novak N, et al. Oral pathogens change proliferation properties of oral tumor cells by affecting gene expression of human defensins. Tumour Biol. 2016;37:13789‐13798. [DOI] [PubMed] [Google Scholar]
- 91. Groeger S, Jarzina F, Domann E, Meyle J. Porphyromonas gingivalis activates NFκB and MAPK pathways in human oral epithelial cells. BMC Immunol. 2017;18:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Chang C, Wang H, Liu J, et al. Porphyromonas gingivalis infection promoted the proliferation of Oral squamous cell carcinoma cells through the miR‐21/PDCD4/AP‐1 negative signaling pathway. ACS Infect Dis. 2019;5:1336‐1347. [DOI] [PubMed] [Google Scholar]
- 93. Binder Gallimidi A, Fischman S, Revach B, et al. Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral‐specific chemical carcinogenesis model. Oncotarget. 2015;6:22613‐22623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Inaba H, Sugita H, Kuboniwa M, et al. Porphyromonas gingivalis promotes invasion of oral squamous cell carcinoma through induction of proMMP9 and its activation. Cell Microbiol. 2014;16:131‐145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Inaba H, Amano A, Lamont RJ, Murakami Y. Involvement of protease‐activated receptor 4 in over‐expression of matrix metalloproteinase 9 induced by Porphyromonas gingivalis . Med Microbiol Immunol. 2015;204:605‐612. [DOI] [PubMed] [Google Scholar]
- 96. Abdulkareem AA, Shelton RM, Landini G, Cooper PR, Milward MR. Periodontal pathogens promote epithelial‐mesenchymal transition in oral squamous carcinoma cells in vitro. Cell Adh Migr. 2018;12:127‐137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Ha NH, Woo BH, Kim DJ, et al. Prolonged and repetitive exposure to Porphyromonas gingivalis increases aggressiveness of oral cancer cells by promoting acquisition of cancer stem cell properties. Tumour Biol. 2015;36:9947‐9960. [DOI] [PubMed] [Google Scholar]
- 98. Ikehata N, Takanashi M, Satomi T, et al. Toll‐like receptor 2 activation implicated in oral squamous cell carcinoma development. Biochem Biophys Res Commun. 2018;495:2227‐2234. [DOI] [PubMed] [Google Scholar]
- 99. Guo ZC, Jing SL, Jumatai S, Gong ZC. Porphyromonas gingivalis promotes the progression of oral squamous cell carcinoma by activating the neutrophil chemotaxis in the tumour microenvironment. Cancer Immunol Immunother. 2023;72:1523‐1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Yuan X, Liu Y, Li G, et al. Blockade of immune‐checkpoint B7‐H4 and lysine demethylase 5B in esophageal squamous cell carcinoma confers protective immunity against P. gingivalis infection. Cancer Immunol Res. 2019;7:1440‐1456. [DOI] [PubMed] [Google Scholar]
- 101. Li J, Lee Y, Li Y, et al. Co‐inhibitory molecule B7 superfamily member 1 expressed by tumor‐infiltrating myeloid cells induces dysfunction of anti‐tumor CD8(+) T cells. Immunity. 2018;48:773‐786.e5. [DOI] [PubMed] [Google Scholar]
- 102. Chen MF, Lu MS, Hsieh CC, Chen WC. Porphyromonas gingivalis promotes tumor progression in esophageal squamous cell carcinoma. Cell Oncol (Dordr). 2021;44:373‐384. [DOI] [PubMed] [Google Scholar]
- 103. Meng F, Li R, Ma L, et al. Porphyromonas gingivalis promotes the motility of esophageal squamous cell carcinoma by activating NF‐κB signaling pathway. Microbes Infect. 2019;21:296‐304. [DOI] [PubMed] [Google Scholar]
- 104. Liang G, Wang H, Shi H, et al. Porphyromonas gingivalis promotes the proliferation and migration of esophageal squamous cell carcinoma through the miR‐194/GRHL3/PTEN/Akt axis. ACS Infect Dis. 2020;6:871‐881. [DOI] [PubMed] [Google Scholar]
- 105. Gao S, Liu Y, Duan X, et al. Porphyromonas gingivalis infection exacerbates oesophageal cancer and promotes resistance to neoadjuvant chemotherapy. Br J Cancer. 2021;125:433‐444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Gao S, Liu K, Jiao Y, et al. Selective activation of TGFβ signaling by P. gingivalis‐mediated upregulation of GARP aggravates esophageal squamous cell carcinoma. Am J Cancer Res. 2023;13:2013‐2029. [PMC free article] [PubMed] [Google Scholar]
- 107. Adel‐Khattab D, Groeger S, Domann E, Chakraborty T, Lochnit G, Meyle J. Porphyromonas gingivalis induced up‐regulation of PD‐L1 in colon carcinoma cells. Mol Oral Microbiol. 2021;36:172‐181. [DOI] [PubMed] [Google Scholar]
- 108. Mu W, Jia Y, Chen X, Li H, Wang Z, Cheng B. Intracellular Porphyromonas gingivalis promotes the proliferation of colorectal cancer cells via the MAPK/ERK signaling pathway. Front Cell Infect Microbiol. 2020;10:584798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Wang X, Jia Y, Wen L, et al. Porphyromonas gingivalis promotes colorectal carcinoma by activating the hematopoietic NLRP3 Inflammasome. Cancer Res. 2021;81:2745‐2759. [DOI] [PubMed] [Google Scholar]
- 110. Hiraki D, Uehara O, Kuramitsu Y, et al. P. gingivalis lipopolysaccharide stimulates the upregulated expression of the pancreatic cancer‐related genes regenerating islet‐derived 3 A/G in mouse pancreas. Int J Mol Sci. 2020;21:7351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Gnanasekaran J, Binder Gallimidi A, Saba E, et al. Intracellular Porphyromonas gingivalis promotes the tumorigenic behavior of pancreatic carcinoma cells. Cancers. 2020;12:2331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Tan Q, Ma X, Yang B, et al. Periodontitis pathogen Porphyromonas gingivalis promotes pancreatic tumorigenesis via neutrophil elastase from tumor‐associated neutrophils. Gut Microbes. 2022;14:2073785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Saba E, Farhat M, Daoud A, et al. Oral bacteria accelerate pancreatic cancer development in mice. Gut. 2024;73(5):770‐786. [DOI] [PubMed] [Google Scholar]
- 114. Han YW. Fusobacterium nucleatum: a commensal‐turned pathogen. Curr Opin Microbiol. 2015;23:141‐147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Brennan CA, Garrett WS. Fusobacterium nucleatum—symbiont, opportunist and oncobacterium. Nat Rev Microbiol. 2019;17:156‐166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Zhang WL, Wang SS, Wang HF, Tang YJ, Tang YL, Liang XH. Who is who in oral cancer? Exp Cell Res. 2019;384:111634. [DOI] [PubMed] [Google Scholar]
- 117. Allen‐Vercoe E, Strauss J, Chadee K. Fusobacterium nucleatum: an emerging gut pathogen? Gut Microbes. 2011;2:294‐298. [DOI] [PubMed] [Google Scholar]
- 118. Edwards AM, Grossman TJ, Rudney JD. Fusobacterium nucleatum transports noninvasive Streptococcus cristatus into human epithelial cells. Infect Immun. 2006;74:654‐662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Groeger S, Zhou Y, Ruf S, Meyle J. Pathogenic mechanisms of Fusobacterium nucleatum on oral epithelial cells. Front Oral Health. 2022;3:831607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Kaplan A, Kaplan CW, He X, McHardy I, Shi W, Lux R. Characterization of aid1, a novel gene involved in Fusobacterium nucleatum interspecies interactions. Microb Ecol. 2014;68:379‐387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Lima BP, Shi W, Lux R. Identification and characterization of a novel Fusobacterium nucleatum adhesin involved in physical interaction and biofilm formation with Streptococcus gordonii . Microbiology. 2017;6:e00444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Han YW, Ikegami A, Rajanna C, et al. Identification and characterization of a novel adhesin unique to oral fusobacteria. J Bacteriol. 2005;187:5330‐5340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Hussan H, Clinton SK, Roberts K, Bailey MT. Fusobacterium's link to colorectal neoplasia sequenced: a systematic review and future insights. World J Gastroenterol. 2017;23:8626‐8650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Gur C, Ibrahim Y, Isaacson B, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Galaski J, Shhadeh A, Umaña A, et al. Fusobacterium nucleatum CbpF mediates inhibition of T cell function through CEACAM1 activation. Front Cell Infect Microbiol. 2021;11:692544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Gur C, Maalouf N, Shhadeh A, et al. Fusobacterium nucleatum supresses anti‐tumor immunity by activating CEACAM1. Onco Targets Ther. 2019;8:e1581531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Gu X, Song LJ, Li LX, et al. Fusobacterium nucleatum causes microbial dysbiosis and exacerbates visceral hypersensitivity in a colonization‐independent manner. Front Microbiol. 2020;11:1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. McIlvanna E, Linden GJ, Craig SG, Lundy FT, James JA. Fusobacterium nucleatum and oral cancer: a critical review. BMC Cancer. 2021;21:1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Sun J, Tang Q, Yu S, et al. F. nucleatum facilitates oral squamous cell carcinoma progression via GLUT1‐driven lactate production. EBioMedicine. 2023;88:104444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Zhang S, Li C, Liu J, et al. Fusobacterium nucleatum promotes epithelial‐mesenchymal transiton through regulation of the lncRNA MIR4435‐2HG/miR‐296‐5p/Akt2/SNAI1 signaling pathway. FEBS J. 2020;287:4032‐4047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Harrandah AM, Chukkapalli SS, Bhattacharyya I, Progulske‐Fox A, Chan EKL. Fusobacteria modulate oral carcinogenesis and promote cancer progression. J Oral Microbiol. 2020;13:1849493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Geng F, Zhang Y, Lu Z, Zhang S, Pan Y. Fusobacterium nucleatum caused DNA damage and promoted cell proliferation by the Ku70/p53 pathway in oral cancer cells. DNA Cell Biol. 2020;39:144‐151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Da J, Wang X, Li L, Xu Y. Fusobacterium nucleatum promotes cisplatin‐resistance and migration of oral squamous carcinoma cells by up‐regulating Wnt5a‐mediated NFATc3 expression. Tohoku J Exp Med. 2021;253:249‐259. [DOI] [PubMed] [Google Scholar]
- 134. Kamarajan P, Ateia I, Shin JM, et al. Periodontal pathogens promote cancer aggressivity via TLR/MyD88 triggered activation of Integrin/FAK signaling that is therapeutically reversible by a probiotic bacteriocin. PLoS Pathog. 2020;16:e1008881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Shin JM, Gwak JW, Kamarajan P, Fenno JC, Rickard AH, Kapila YL. Biomedical applications of nisin. J Appl Microbiol. 2016;120:1449‐1465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Nomoto D, Baba Y, Liu Y, et al. Fusobacterium nucleatum promotes esophageal squamous cell carcinoma progression via the NOD1/RIPK2/NF‐κB pathway. Cancer Lett. 2022;530:59‐67. [DOI] [PubMed] [Google Scholar]
- 137. Liang M, Liu Y, Zhang Z, et al. Fusobacterium nucleatum induces MDSCs enrichment via activation the NLRP3 inflammosome in ESCC cells, leading to cisplatin resistance. Ann Med. 2022;54:989‐1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Wang X, Liu Y, Lu Y, et al. Clinical impact of Fn‐induced high expression of KIR2DL1 in CD8 T lymphocytes in oesophageal squamous cell carcinoma. Ann Med. 2022;54:51‐62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Abed J, Maalouf N, Manson AL, et al. Colon cancer‐associated Fusobacterium nucleatum may originate from the oral cavity and reach colon tumors via the circulatory system. Front Cell Infect Microbiol. 2020;10:400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E‐cadherin/β‐catenin signaling via its FadA adhesin. Cell Host Microbe. 2013;14:195‐206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Rubinstein MR, Baik JE, Lagana SM, et al. Fusobacterium nucleatum promotes colorectal cancer by inducing Wnt/β‐catenin modulator Annexin A1. EMBO Rep. 2019;20:e47638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Abed J, Emgård JE, Zamir G, et al. Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor‐expressed gal‐GalNAc. Cell Host Microbe. 2016;20:215‐225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Chen Y, Peng Y, Yu J, et al. Invasive Fusobacterium nucleatum activates beta‐catenin signaling in colorectal cancer via a TLR4/P‐PAK1 cascade. Oncotarget. 2017;8:31802‐31814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Yang Y, Weng W, Peng J, et al. Fusobacterium nucleatum increases proliferation of colorectal cancer cells and tumor development in mice by activating toll‐like receptor 4 signaling to nuclear factor‐κB, and up‐regulating expression of MicroRNA‐21. Gastroenterology. 2017;152:851‐866.e24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Chen T, Li Q, Wu J, et al. Fusobacterium nucleatum promotes M2 polarization of macrophages in the microenvironment of colorectal tumours via a TLR4‐dependent mechanism. Cancer Immunol Immunother. 2018;67:1635‐1646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Kostic AD, Chun E, Robertson L, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor‐immune microenvironment. Cell Host Microbe. 2013;14:207‐215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Coussens LM, Pollard JW. Leukocytes in mammary development and cancer. Cold Spring Harb Perspect Biol. 2011;3:a003285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Toor SM, Syed Khaja AS, El Salhat H, et al. Increased levels of circulating and tumor‐infiltrating granulocytic myeloid cells in colorectal cancer patients. Front Immunol. 2016;7:560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Gabrilovich DI, Ostrand‐Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12:253‐268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Chen T, Li Q, Zhang X, et al. TOX expression decreases with progression of colorectal cancers and is associated with CD4 T‐cell density and Fusobacterium nucleatum infection. Hum Pathol. 2018;79:93‐101. [DOI] [PubMed] [Google Scholar]
- 151. Mima K, Sukawa Y, Nishihara R, et al. Fusobacterium nucleatum and T cells in colorectal carcinoma. JAMA Oncol. 2015;1:653‐661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Li G, Sun Y, Huang Y, et al. Fusobacterium nucleatum‐derived small extracellular vesicles facilitate tumor growth and metastasis via TLR4 in breast cancer. BMC Cancer. 2023;23:473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Liu C, Zhang H, Li T, Jiang Z, Yuan Y, Chen X. Fusobacterium nucleatum promotes megakaryocyte maturation in patients with gastric cancer via inducing the production of extracellular vesicles containing 14‐3‐3ε. Infect Immun. 2023;91:e0010223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Meng X, Ma G, Zhang X, Yin H, Miao Y, He F. Extracellular vesicles from Fusobacterium nucleatum: roles in the malignant phenotypes of gastric cancer. Cell Cycle. 2024;23(3):294‐307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Möller B, Kollert F, Sculean A, Villiger PM. Infectious triggers in periodontitis and the gut in rheumatoid arthritis (RA): a complex story about association and causality. Front Immunol. 2020;11:1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Khaledi M, Sameni F, Afkhami H, et al. Infective endocarditis by HACEK: a review. J Cardiothorac Surg. 2022;17:185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Krueger E, Brown AC. Aggregatibacter actinomycetemcomitans leukotoxin: from mechanism to targeted anti‐toxin therapeutics. Mol Oral Microbiol. 2020;35:85‐105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Gholizadeh P, Pormohammad A, Eslami H, Shokouhi B, Fakhrzadeh V, Kafil HS. Oral pathogenesis of Aggregatibacter actinomycetemcomitans . Microb Pathog. 2017;113:303‐311. [DOI] [PubMed] [Google Scholar]
- 159. Balashova N, Dhingra A, Boesze‐Battaglia K, Lally ET. Aggregatibacter actinomycetemcomitans leukotoxin induces cytosol acidification in LFA‐1 expressing immune cells. Mol Oral Microbiol. 2016;31:106‐114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. DiFranco KM, Gupta A, Galusha LE, et al. Leukotoxin (Leukothera®) targets active leukocyte function antigen‐1 (LFA‐1) protein and triggers a lysosomal mediated cell death pathway. J Biol Chem. 2012;287:17618‐17627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Kieba IR, Fong KP, Tang HY, et al. Aggregatibacter actinomycetemcomitans leukotoxin requires beta‐sheets 1 and 2 of the human CD11a beta‐propeller for cytotoxicity. Cell Microbiol. 2007;9:2689‐2699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Vega BA, Schober LT, Kim T, Belinka BA Jr, Kachlany SC. Aggregatibacter actinomycetemcomitans leukotoxin (LtxA) requires death receptor Fas, in addition to LFA‐1, to trigger cell death in T lymphocytes. Infect Immun. 2019;87:e00309‐19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Belibasakis GN, Maula T, Bao K, et al. Virulence and pathogenicity properties of Aggregatibacter actinomycetemcomitans . Pathogens. 2019;8:222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Kelk P, Johansson A, Claesson R, Hänström L, Kalfas S. Caspase 1 involvement in human monocyte lysis induced by Actinobacillus actinomycetemcomitans leukotoxin. Infect Immun. 2003;71:4448‐4455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Kelk P, Claesson R, Hänström L, Lerner UH, Kalfas S, Johansson A. Abundant secretion of bioactive interleukin‐1beta by human macrophages induced by Actinobacillus actinomycetemcomitans leukotoxin. Infect Immun. 2005;73:453‐458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Lara‐Tejero M, Galán JE. A bacterial toxin that controls cell cycle progression as a deoxyribonuclease I‐like protein. Science. 2000;290:354‐357. [DOI] [PubMed] [Google Scholar]
- 167. Ando‐Suguimoto ES, da Silva MP, Kawamoto D, Chen C, DiRienzo JM, Mayer MP. The cytolethal distending toxin of Aggregatibacter actinomycetemcomitans inhibits macrophage phagocytosis and subverts cytokine production. Cytokine. 2014;66:46‐53. [DOI] [PubMed] [Google Scholar]
- 168. Belibasakis GN, Bostanci N. Inflammatory and bone remodeling responses to the cytolethal distending toxins. Cells. 2014;3:236‐246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Kawamoto D, Ando‐Suguimoto ES, Bueno‐Silva B, DiRienzo JM, Mayer MP. Alteration of homeostasis in pre‐osteoclasts induced by Aggregatibacter actinomycetemcomitans CDT. Front Cell Infect Microbiol. 2016;6:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Bodet C, Chandad F, Grenier D. Anti‐inflammatory activity of a high‐molecular‐weight cranberry fraction on macrophages stimulated by lipopolysaccharides from periodontopathogens. J Dent Res. 2006;85:235‐239. [DOI] [PubMed] [Google Scholar]
- 171. Stuehr DJ. Mammalian nitric oxide synthases. Biochim Biophys Acta. 1999;1411:217‐230. [DOI] [PubMed] [Google Scholar]
- 172. Kieselbach T, Zijnge V, Granström E, Oscarsson J. Proteomics of Aggregatibacter actinomycetemcomitans outer membrane vesicles. PLoS One. 2015;10:e0138591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Choi JW, Kim SC, Hong SH, Lee HJ. Secretable small RNAs via outer membrane vesicles in periodontal pathogens. J Dent Res. 2017;96:458‐466. [DOI] [PubMed] [Google Scholar]
- 174. Teshima R, Hanada K, Akada J, Kawano K, Yamaoka Y. Aggregatibacter actinomycetemcomitans infection causes DNA double‐strand breaks in host cells. Genes Cells. 2018;23:264‐273. [DOI] [PubMed] [Google Scholar]
- 175. Söder B, Källmén H, Yucel‐Lindberg T, Meurman JH. Periodontal microorganisms and diagnosis of malignancy: a cross‐sectional study. Tumour Biol. 2021;43:1‐9. [DOI] [PubMed] [Google Scholar]
- 176. Yamamoto K, Tominaga K, Sukedai M, et al. Delivery of cytolethal distending toxin B induces cell cycle arrest and apoptosis in gingival squamous cell carcinoma in vitro. Eur J Oral Sci. 2004;112:445‐451. [DOI] [PubMed] [Google Scholar]
- 177. Listyarifah D, Nieminen MT, Mäkinen LK, et al. Treponema denticola chymotrypsin‐like proteinase is present in early‐stage mobile tongue squamous cell carcinoma and related to the clinicopathological features. J Oral Pathol Med. 2018;47:764‐772. [DOI] [PubMed] [Google Scholar]
- 178. Ksiazek M, Mizgalska D, Enghild JJ, Scavenius C, Thogersen IB, Potempa J. Miropin, a novel bacterial serpin from the periodontopathogen Tannerella forsythia, inhibits a broad range of proteases by using different peptide bonds within the reactive center loop. J Biol Chem. 2015;290:658‐670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Friedrich V, Gruber C, Nimeth I, et al. Outer membrane vesicles of Tannerella forsythia: biogenesis, composition, and virulence. Mol Oral Microbiol. 2015;30:451‐473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Sharma A. Virulence mechanisms of Tannerella forsythia . Periodontol 2000. 2010;54:106‐116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Yoneda M, Hirofuji T, Anan H, et al. Mixed infection of Porphyromonas gingivalis and Bacteroides forsythus in a murine abscess model: involvement of gingipains in a synergistic effect. J Periodontal Res. 2001;36:237‐243. [DOI] [PubMed] [Google Scholar]
- 182. Kesavalu L, Sathishkumar S, Bakthavatchalu V, et al. Rat model of polymicrobial infection, immunity, and alveolar bone resorption in periodontal disease. Infect Immun. 2007;75:1704‐1712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Shin JM, Luo T, Kamarajan P, Fenno JC, Rickard AH, Kapila YL. Microbial communities associated with primary and metastatic head and neck squamous cell carcinoma—a high fusobacterial and low streptococcal signature. Sci Rep. 2017;7:9934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Peng RT, Sun Y, Zhou XD, et al. Treponema denticola promotes OSCC development via the TGF‐β signaling pathway. J Dent Res. 2022;101:704‐713. [DOI] [PubMed] [Google Scholar]
- 185. Nieminen MT, Listyarifah D, Hagström J, et al. Treponema denticola chymotrypsin‐like proteinase may contribute to orodigestive carcinogenesis through immunomodulation. Br J Cancer. 2018;118:428‐434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Meng C, Bai C, Brown TD, Hood LE, Tian Q. Human gut microbiota and gastrointestinal cancer. Genomics Proteomics Bioinformatics. 2018;16:33‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Bakhti SZ, Latifi‐Navid S. Oral microbiota and Helicobacter pylori in gastric carcinogenesis: what do we know and where next? BMC Microbiol. 2021;21:71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Karpinski TM. Role of oral microbiota in cancer development. Microorganisms. 2019;7:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Szkaradkiewicz AK, Karpiński TM. Microbiology of chronic periodontitis. J Biol Earth Sci. 2013;3:14‐20. [Google Scholar]
- 190. Metzner B, Hofmann C, Heinemann C, et al. Overexpression of CXC‐chemokines and CXC‐chemokine receptor type II constitute an autocrine growth mechanism in the epidermoid carcinoma cells KB and A431. Oncol Rep. 1999;6:1405‐1410. [DOI] [PubMed] [Google Scholar]
- 191. Christofakis EP, Miyazaki H, Rubink DS, Yeudall WA. Roles of CXCL8 in squamous cell carcinoma proliferation and migration. Oral Oncol. 2008;44:920‐926. [DOI] [PubMed] [Google Scholar]
- 192. Park D‐G, Woo BH, Lee B‐J, et al. Serum levels of interleukin‐6 and titers of antibodies against Porphyromonas gingivalis could be potential biomarkers for the diagnosis of oral squamous cell carcinoma. Int J Mol Sci. 2019;20:2749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. St John MA, Li Y, Zhou X, et al. Interleukin 6 and interleukin 8 as potential biomarkers for oral cavity and oropharyngeal squamous cell carcinoma. Arch Otolaryngol Head Neck Surg. 2004;130:929‐935. [DOI] [PubMed] [Google Scholar]
- 194. Vogelmann R, Amieva MR. The role of bacterial pathogens in cancer. Curr Opin Microbiol. 2007;10:76‐81. [DOI] [PubMed] [Google Scholar]
- 195. Zhang G, Ghosh S. Molecular mechanisms of NF‐kappaB activation induced by bacterial lipopolysaccharide through Toll‐like receptors. J Endotoxin Res. 2000;6:453‐457. [DOI] [PubMed] [Google Scholar]
- 196. Wang H, Zhou H, Duan X, et al. Porphyromonas gingivalis‐induced reactive oxygen species activate JAK2 and regulate production of inflammatory cytokines through c‐Jun. Infect Immun. 2014;82:4118‐4126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Fujita Y, Nakayama M, Naito M, et al. Hemoglobin receptor protein from Porphyromonas gingivalis induces interleukin‐8 production in human gingival epithelial cells through stimulation of the mitogen‐activated protein kinase and NF‐κB signal transduction pathways. Infect Immun. 2014;82:202‐211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Wang PL, Azuma Y, Shinohara M, Ohura K. Toll‐like receptor 4‐mediated signal pathway induced by Porphyromonas gingivalis lipopolysaccharide in human gingival fibroblasts. Biochem Biophys Res Commun. 2000;273:1161‐1167. [DOI] [PubMed] [Google Scholar]
- 199. Savitri IJ, Ouhara K, Fujita T, et al. Irsogladine maleate inhibits Porphyromonas gingivalis‐mediated expression of toll‐like receptor 2 and interleukin‐8 in human gingival epithelial cells. J Periodontal Res. 2015;50:486‐493. [DOI] [PubMed] [Google Scholar]
- 200. Andrukhov O, Ertlschweiger S, Moritz A, Bantleon HP, Rausch‐Fan X. Different effects of P. gingivalis LPS and E. coli LPS on the expression of interleukin‐6 in human gingival fibroblasts. Acta Odontol Scand. 2014;72:337‐345. [DOI] [PubMed] [Google Scholar]
- 201. Olsen I, Singhrao SK, Osmundsen H. Periodontitis, pathogenesis and progression: miRNA‐mediated cellular responses to Porphyromonas gingivalis . J Oral Microbiol. 2017;9:1333396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Li J, Li L, Wang X, Xiao L. Porphyromonas gingivalis inhibition of microRNA‐205‐5p expression modulates proinflammatory cytokines in gingival epithelial cells. Biochem Genet. 2020;58:566‐579. [DOI] [PubMed] [Google Scholar]
- 203. Bui FQ, Johnson L, Roberts J, et al. Fusobacterium nucleatum infection of gingival epithelial cells leads to NLRP3 inflammasome‐dependent secretion of IL‐1beta and the danger signals ASC and HMGB1. Cell Microbiol. 2016;18:970‐981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Gholizadeh P, Eslami H, Kafil HS. Carcinogenesis mechanisms of Fusobacterium nucleatum . Biomed Pharmacother. 2017;89:918‐925. [DOI] [PubMed] [Google Scholar]
- 205. Peyyala R, Kirakodu SS, Novak KF, Ebersole JL. Oral microbial biofilm stimulation of epithelial cell responses. Cytokine. 2012;58:65‐72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Dickinson BC, Moffatt CE, Hagerty D, et al. Interaction of oral bacteria with gingival epithelial cell multilayers. Mol Oral Microbiol. 2011;26:210‐220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Zhang G, Chen R, Rudney JD. Streptococcus cristatus modulates the Fusobacterium nucleatum‐induced epithelial interleukin‐8 response through the nuclear factor‐kappa B pathway. J Periodontal Res. 2011;46:558‐567. [DOI] [PubMed] [Google Scholar]
- 208. Gholizadeh P, Eslami H, Yousefi M, Asgharzadeh M, Aghazadeh M, Kafil HS. Role of oral microbiome on oral cancers, a review. Biomed Pharmacother. 2016;84:552‐558. [DOI] [PubMed] [Google Scholar]
- 209. Lee SJ, Choi BK. Involvement of NLRP10 in IL‐1alpha induction of oral epithelial cells by periodontal pathogens. Innate Immun. 2017;23:569‐577. [DOI] [PubMed] [Google Scholar]
- 210. Krisanaprakornkit S, Kimball JR, Weinberg A, Darveau RP, Bainbridge BW, Dale BA. Inducible expression of human beta‐defensin 2 by Fusobacterium nucleatum in oral epithelial cells: multiple signaling pathways and role of commensal bacteria in innate immunity and the epithelial barrier. Infect Immun. 2000;68:2907‐2915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Ji S, Kim Y, Min BM, Han SH, Choi Y. Innate immune responses of gingival epithelial cells to nonperiodontopathic and periodontopathic bacteria. J Periodontal Res. 2007;42:503‐510. [DOI] [PubMed] [Google Scholar]
- 212. Hung SC, Huang PR, Almeida‐da‐Silva CLC, Atanasova KR, Yilmaz O, Ojcius DM. NLRX1 modulates differentially NLRP3 inflammasome activation and NF‐kappaB signaling during Fusobacterium nucleatum infection. Microbes Infect. 2018;20:615‐625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010;28:367‐388. [DOI] [PubMed] [Google Scholar]
- 214. Jo AR, Baek KJ, Shin JE, Choi Y. Mechanisms of IL‐8 suppression by Treponema denticola in gingival epithelial cells. Immunol Cell Biol. 2014;92:139‐147. [DOI] [PubMed] [Google Scholar]
- 215. Shin JE, Baek KJ, Choi YS, Choi Y. A periodontal pathogen Treponema denticola hijacks the Fusobacterium nucleatum‐driven host response. Immunol Cell Biol. 2013;91:503‐510. [DOI] [PubMed] [Google Scholar]
- 216. Pahumunto N, Chotjumlong P, Makeudom A, Krisanaprakornkit S, Dahlen G, Teanpaisan R. Pro‐inflammatory cytokine responses in human gingival epithelial cells after stimulation with cell wall extract of Aggregatibacter actinomycetemcomitans subtypes. Anaerobe. 2017;48:103‐109. [DOI] [PubMed] [Google Scholar]
- 217. Ko YK, An SJ, Han NY, Lee H, Choi BK. Regulation of IL‐24 in human oral keratinocytes stimulated with Tannerella forsythia . Mol Oral Microbiol. 2019;34:209‐218. [DOI] [PubMed] [Google Scholar]
- 218. Issrani R, Reddy RJ, El‐Metwally TH, Prabhu N. Periodontitis as a risk factor for breast cancer—what we know till date? Asian Pac J Cancer Prev. 2021;22:3109‐3114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Garrett WS. Cancer and the microbiota. Science. 2015;348:80‐86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. da Silva MP, Silva VO, Pasetto S, et al. Aggregatibacter actinomycetemcomitans outer membrane proteins 29 and 29 paralogue induce evasion of immune response. Front Oral Health. 2022;3:835902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Teng MW, Galon J, Fridman WH, Smyth MJ. From mice to humans: developments in cancer immunoediting. J Clin Invest. 2015;125:3338‐3346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Groeger S, Denter F, Lochnit G, Schmitz ML, Meyle J. Porphyromonas gingivalis cell wall components induce programmed death ligand 1 (PD‐L1) expression on human oral carcinoma cells by a receptor‐interacting protein kinase 2 (RIP2)‐dependent mechanism. Infect Immun. 2020;88:e00051‐20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Groeger S, Howaldt HP, Raifer H, Gattenloehner S, Chakraborty T, Meyle J. Oral squamous carcinoma cells express B7‐H1 and B7‐DC receptors in vivo. Pathol Oncol Res. 2017;23:99‐110. [DOI] [PubMed] [Google Scholar]
- 224. Groeger S, Jarzina F, Mamat U, Meyle J. Induction of B7‐H1 receptor by bacterial cells fractions of Porphyromonas gingivalis on human oral epithelial cells: B7‐H1 induction by Porphyromonas gingivalis fractions. Immunobiology. 2017;222:137‐147. [DOI] [PubMed] [Google Scholar]
- 225. Fankhauser SC, Starnbach MN. PD‐L1 limits the mucosal CD8+ T cell response to Chlamydia trachomatis . J Immunol. 2014;192:1079‐1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Jauregui CE, Wang Q, Wright CJ, Takeuchi H, Uriarte SM, Lamont RJ. Suppression of T‐cell chemokines by Porphyromonas gingivalis . Infect Immun. 2013;81:2288‐2295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Morandini AC, Ramos‐Junior ES, Potempa J, et al. Porphyromonas gingivalis fimbriae dampen P2X7‐dependent interleukin‐1β secretion. J Innate Immun. 2014;6:831‐845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Zhou Y, Luo GH. Porphyromonas gingivalis and digestive system cancers. World J Clin Cases. 2019;7:819‐829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Arjunan P, Meghil MM, Pi W, et al. Oral pathobiont activates anti‐apoptotic pathway, promoting both immune suppression and oncogenic cell proliferation. Sci Rep. 2018;8:16607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230. Ryder MI. Comparison of neutrophil functions in aggressive and chronic periodontitis. Periodontol 2000. 2010;53:124‐137. [DOI] [PubMed] [Google Scholar]
- 231. Segal AW. How neutrophils kill microbes. Annu Rev Immunol. 2005;23:197‐223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Zheng S, Yu S, Fan X, et al. Porphyromonas gingivalis survival skills: immune evasion. J Periodontal Res. 2021;56:1007‐1018. [DOI] [PubMed] [Google Scholar]
- 233. Makkawi H, Hoch S, Burns E, et al. Porphyromonas gingivalis stimulates TLR2‐PI3K signaling to escape immune clearance and induce bone resorption independently of MyD88. Front Cell Infect Microbiol. 2017;7:359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Wingrove JA, DiScipio RG, Chen Z, Potempa J, Travis J, Hugli TE. Activation of complement components C3 and C5 by a cysteine proteinase (gingipain‐1) from Porphyromonas (Bacteroides) gingivalis . J Biol Chem. 1992;267:18902‐18907. [PubMed] [Google Scholar]
- 235. Bryzek D, Ciaston I, Dobosz E, et al. Triggering NETosis via protease‐activated receptor (PAR)‐2 signaling as a mechanism of hijacking neutrophils function for pathogen benefits. PLoS Pathog. 2019;15:e1007773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Mikolai C, Branitzki‐Heinemann K, Ingendoh‐Tsakmakidis A, Stiesch M, von Köckritz‐Blickwede M, Winkel A. Neutrophils exhibit an individual response to different oral bacterial biofilms. J Oral Microbiol. 2020;13:1856565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Slocum C, Coats SR, Hua N, et al. Distinct lipid a moieties contribute to pathogen‐induced site‐specific vascular inflammation. PLoS Pathog. 2014;10:e1004215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Castro SA, Collighan R, Lambert PA, et al. Porphyromonas gingivalis gingipains cause defective macrophage migration towards apoptotic cells and inhibit phagocytosis of primary apoptotic neutrophils. Cell Death Dis. 2017;8:e2644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Wilensky A, Tzach‐Nahman R, Potempa J, Shapira L, Nussbaum G. Porphyromonas gingivalis gingipains selectively reduce CD14 expression, leading to macrophage hyporesponsiveness to bacterial infection. J Innate Immun. 2015;7:127‐135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Hajishengallis G, McIntosh ML, Nishiyama SI, Yoshimura F. Mechanism and implications of CXCR4‐mediated integrin activation by Porphyromonas gingivalis . Mol Oral Microbiol. 2013;28:239‐249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Yang X, Pan Y, Xu X, et al. Sialidase deficiency in Porphyromonas gingivalis increases IL‐12 secretion in stimulated macrophages through regulation of CR3, IncRNA GAS5 and miR‐21. Front Cell Infect Microbiol. 2018;8:100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Hajishengallis G, Wang M, Liang S, Triantafilou M, Triantafilou K. Pathogen induction of CXCR4/TLR2 cross‐talk impairs host defense function. Proc Natl Acad Sci U S A. 2008;105:13532‐13537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Boor PPC, Bosma BM, Tran KTC, et al. Characterization of antigen‐presenting cell subsets in human liver‐draining lymph nodes. Front Immunol. 2019;10:441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Khalaf H, Bengtsson T. Altered T‐cell responses by the periodontal pathogen Porphyromonas gingivalis . PLoS One. 2012;7:e45192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Gaddis DE, Maynard CL, Weaver CT, Michalek SM, Katz J. Role of TLR2‐dependent IL‐10 production in the inhibition of the initial IFN‐γ T cell response to Porphyromonas gingivalis . J Leukoc Biol. 2013;93:21‐31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Fitzsimonds ZR, Rodriguez‐Hernandez CJ, Bagaitkar J, Lamont RJ. From beyond the pale to the pale riders: the emerging association of bacteria with oral cancer. J Dent Res. 2020;99:604‐612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Kuboniwa M, Hasegawa Y, Mao S, et al. P. gingivalis accelerates gingival epithelial cell progression through the cell cycle. Microbes Infect. 2008;10:122‐128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Zhou L, Dörfer CE, Chen L, Fawzy El‐Sayed KM. Porphyromonas gingivalis lipopolysaccharides affect gingival stem/progenitor cells attributes through NF‐κB, but not Wnt/β‐catenin, pathway. J Clin Periodontol. 2017;44:1112‐1122. [DOI] [PubMed] [Google Scholar]
- 249. Hoppe T, Kraus D, Probstmeier R, Jepsen S, Winter J. Stimulation with Porphyromonas gingivalis enhances malignancy and initiates anoikis resistance in immortalized oral keratinocytes. J Cell Physiol. 2019;234:21903‐21914. [DOI] [PubMed] [Google Scholar]
- 250. Seoane JM, Varela‐Centelles PI, Ramirez JR, Cameselle‐Teijeiro J, Romero MA, Aguirre JM. Heat shock proteins (HSP70 and HSP27) as markers of epithelial dysplasia in oral leukoplakia. Am J Dermatopathol. 2006;28:417‐422. [DOI] [PubMed] [Google Scholar]
- 251. Uitto VJ, Baillie D, Wu Q, et al. Fusobacterium nucleatum increases collagenase 3 production and migration of epithelial cells. Infect Immun. 2005;73:1171‐1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Perera M, Al‐hebshi NN, Speicher DJ, Perera I, Johnson NW. Emerging role of bacteria in oral carcinogenesis: a review with special reference to perio‐pathogenic bacteria. J Oral Microbiol. 2016;8:32762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Mahtout H, Chandad F, Rojo JM, Grenier D. Fusobacterium nucleatum binding to complement regulatory protein CD46 modulates the expression and secretion of cytokines and matrix metalloproteinases by oral epithelial cells. J Periodontol. 2011;82:311‐319. [DOI] [PubMed] [Google Scholar]
- 254. Jia X, Liu J, He Y, Huang X. Porphyromonas gingivalis secretion leads to dysplasia of normal esophageal epithelial cells via the Sonic hedgehog pathway. Front Cell Infect Microbiol. 2022;12:982636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007;35:495‐516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Yilmaz O, Yao L, Maeda K, et al. ATP scavenging by the intracellular pathogen Porphyromonas gingivalis inhibits P2X7‐mediated host‐cell apoptosis. Cell Microbiol. 2008;10:863‐875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Lee J, Roberts JS, Atanasova KR, Chowdhury N, Yilmaz Ö. A novel kinase function of a nucleoside‐diphosphate‐kinase homologue in Porphyromonas gingivalis is critical in subversion of host cell apoptosis by targeting heat‐shock protein 27. Cell Microbiol. 2018;20:e12825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Mao S, Park Y, Hasegawa Y, et al. Intrinsic apoptotic pathways of gingival epithelial cells modulated by Porphyromonas gingivalis . Cell Microbiol. 2007;9:1997‐2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Yilmaz O, Jungas T, Verbeke P, Ojcius DM. Activation of the phosphatidylinositol 3‐kinase/Akt pathway contributes to survival of primary epithelial cells infected with the periodontal pathogen Porphyromonas gingivalis . Infect Immun. 2004;72:3743‐3751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260. Jewett A, Hume WR, Le H, et al. Induction of apoptotic cell death in peripheral blood mononuclear and polymorphonuclear cells by an oral bacterium, Fusobacterium nucleatum . Infect Immun. 2000;68:1893‐1898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Li TJ, Hao YH, Tang YL, Liang XH. Periodontal pathogens: a crucial link between periodontal diseases and oral cancer. Front Microbiol. 2022;13:919633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Murata T, Yaegaki K, Qian W, et al. Hydrogen sulfide induces apoptosis in epithelial cells derived from human gingiva. J Breath Res. 2008;2:017007. [DOI] [PubMed] [Google Scholar]
- 263. Calenic B, Yaegaki K, Murata T, et al. Oral malodorous compound triggers mitochondrial‐dependent apoptosis and causes genomic DNA damage in human gingival epithelial cells. J Periodontal Res. 2010;45:31‐37. [DOI] [PubMed] [Google Scholar]
- 264. Ratkay LG, Waterfield JD, Tonzetich J. Stimulation of enzyme and cytokine production by methyl mercaptan in human gingival fibroblast and monocyte cell cultures. Arch Oral Biol. 1995;40:337‐344. [DOI] [PubMed] [Google Scholar]
- 265. Milella L. The negative effects of volatile sulphur compounds. J Vet Dent. 2015;32:99‐102. [DOI] [PubMed] [Google Scholar]
- 266. Zorov DB, Juhaszova M, Sollott SJ. Mitochondrial reactive oxygen species (ROS) and ROS‐induced ROS release. Physiol Rev. 2014;94:909‐950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. Yang HL, Thiyagarajan V, Shen PC, et al. Anti‐EMT properties of CoQ0 attributed to PI3K/AKT/NFKB/MMP‐9 signaling pathway through ROS‐mediated apoptosis. J Exp Clin Cancer Res. 2019;38:186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. Kuznetsov JN, Leclerc GJ, Leclerc GM, Barredo JC. AMPK and Akt determine apoptotic cell death following perturbations of one‐carbon metabolism by regulating ER stress in acute lymphoblastic leukemia. Mol Cancer Ther. 2011;10:437‐447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269. Yang J‐h, Yu K, Si X‐k, et al. Liensinine inhibited gastric cancer cell growth through ROS generation and the PI3K/AKT pathway. J Cancer. 2019;10:6431‐6438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270. Aggarwal V, Tuli HS, Varol A, et al. Role of reactive oxygen species in cancer progression: molecular mechanisms and recent advancements. Biomolecules. 2019;9:735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Choi CH, Spooner R, DeGuzman J, Koutouzis T, Ojcius DM, Yilmaz Ö. Porphyromonas gingivalis‐nucleoside‐diphosphate‐kinase inhibits ATP‐induced reactive‐oxygen‐species via P2X7 receptor/NADPH‐oxidase signalling and contributes to persistence. Cell Microbiol. 2013;15:961‐976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Guo W, Zhao Y, Li H, Lei L. NCOA4‐mediated ferritinophagy promoted inflammatory responses in periodontitis. J Periodontal Res. 2021;56:523‐534. [DOI] [PubMed] [Google Scholar]
- 273. Kang W, Jia Z, Tang D, et al. Fusobacterium nucleatum facilitates apoptosis, ROS generation, and inflammatory cytokine production by activating AKT/MAPK and NF‐kappaB signaling pathways in human gingival fibroblasts. Oxid Med Cell Longev. 2019;2019:1681972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274. Fan T, Chen Y, He Z, et al. Inhibition of ROS/NUPR1‐dependent autophagy antagonises repeated cadmium exposure‐induced oral squamous cell carcinoma cell migration and invasion. Toxicol Lett. 2019;314:142‐152. [DOI] [PubMed] [Google Scholar]
- 275. Okinaga T, Ariyoshi W, Nishihara T. Aggregatibacter actinomycetemcomitans invasion induces interleukin‐1β production through reactive oxygen species and cathepsin B. J Interferon Cytokine Res. 2015;35:431‐440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Wu J, Li Q, Fu X. Fusobacterium nucleatum contributes to the carcinogenesis of colorectal cancer by inducing inflammation and suppressing host immunity. Transl Oncol. 2019;12:846‐851. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no new data were created or analyzed in the current study.
