396
Views
0
CrossRef citations to date
0
Altmetric
Review

Emerging implications of bacterial biofilm in cancer biology: Recent updates and major perspectives

, , , , , & show all
Pages 1-20 | Received 05 Oct 2023, Accepted 27 Mar 2024, Published online: 07 May 2024

ABSTRACT

Recent insights have unveiled exciting opportunities to explore the intricate interplay among bacterial biofilms, tumor cells, and the immune system, thus offering new perspectives in cancer biology. The implications of bacterial biofilms in this context are remarkably multifaceted. Biofilms can promote tumor growth and invasiveness by inducing chronic inflammation, remodeling the extracellular matrix, and modulating the immune response, which promotes cancer development. Recent findings have demonstrated the involvement of distinct bacteria, like Salmonella typhi in gall bladder cancer, Helicobacter pylori in gastric cancer, and Fusobacterium nucleatum in oral cancer. These investigations indicate higher prevalence of these bacteria in individuals with cancer as compared to those who are healthy. Additionally, these bacteria create biofilms and display resistance to cancer treatments.In this review, we highlighted the recent advancements pertaining to influences of bacterial biofilm in cancer progression and potential molecular mechanisms by which bacterial biofilms contribute to cancer development.

Introduction

Globally, cancer is one of the leading causes of deathCitation1 and has long been studied from genetics, tumor microenvironment, and immune response viewpoints. However, recent discoveries have highlighted a previously overlooked aspect of cancer biology: the presence and impact of bacterial biofilm in cancer causation and progression. Biofilm is defined as a collection of microorganisms embedded in a matrix of extracellular polymeric substances (EPS) and adhering to either living or non-living surfaces,Citation2–4 has primarily been associated with chronic infections. The formation of stationary biofilm communities is a multifaceted and constantly changing process in which extracellular polymeric substances (EPS) play crucial roles in both structure and function, which are vital for the unique properties of biofilms as a whole. EPS facilitates the initial attachment of microbes to both living and non-living surfaces. Once adhered to, additional EPS production forms a matrix that envelops and binds the cells, maintaining their proximity and enabling interactions between them within a confined environment.Citation2 The EPS matrix not only imparts mechanical stability but also creates intricate chemical microenvironments essential for the biofilm lifestyle.Citation5

It should also be emphasized that biofilm-forming bacteria play a significant role in the development of numerous illnesses and infections that can be fatal in humans. These include cystic fibrosis (CF), otitis media, periodontitis, infective endocarditis (IE), chronic wounds, and osteomyelitis,Citation6–8 Additionally, biofilms are implicated in vaginitis,Citation9 colitis,Citation10 conjunctivitis,Citation11 gingivitis,Citation12 and urethritis.Citation13 It has been established that a substantial 80% of all bacterial infections in humans are connected to the formation of biofilms.Citation4,Citation14

Previous studies have shown that the primary effects of biofilm development in single bacterial models in vitro are drug resistance and environmental tolerance.Citation15,Citation16 Instead of being a stress response, biofilm development in the GI tract may be a proactive strategy. For instance, biofilm communities develop polybacterial structures and interspecies cross-feeding systems to promote co-colonization.Citation17 Additionally, the finding of intestinal crypt biofilm and intracellular biofilm-like structures revealed that biofilms affect host cell function and cause chronic inflammation.Citation18–20 Since biofilm development represents both invasion efficacy and persistence, it is appropriate to evaluate pathogenicity using this factor. However, their presence in various cancer types has raised intriguing questions about their role in tumorigenesis, cancer progression, treatment response, and clinical outcomes. Traditionally, cancer research has focused on intrinsic factors within cancer cells and their surrounding tissues. However, emerging evidence suggests that the microbial composition within the tumor microenvironment can significantly influence tumor behavior and treatment outcomes.Citation21 Biofilms, known for their resistance to antibiotics and immune responses, exhibit similar characteristics in the context of cancer.Citation22

Bacterial biofilms have been identified in several cancer types, including colorectal, breast, pancreatic, and lung cancer, among othersCitation23 (). They have been found within the tumor tissue, surrounding stromal cells, and distant metastatic sites. Several bacteria have been implicated in their potential roles in oncogenesis like Helicobacter pylori, Salmonella typhi, Fusobacterium nucleatum, Parvimonas micra, Campylobacter jejuni, Chlamydia psittaci, and Chlamydia pneumonia Citation24(). Various research reports have compellingly shown that biofilms play a significant role in the development of human colon cancer. Moreover, they have revealed a notable correlation between biofilms and the specific location of cancer within the colon. Notably, nearly all adenomas and cancers located on the right side of the colon are found to be associated with biofilms, underscoring their prominent role in this context. In contrast, left-sided colon cancers exhibit a notably lower prevalence of biofilm presence.Citation40 Additionally, biofilms have also been implicated in the context of gastric cancer.Citation25 When in their planktonic stage, these bacteria are only capable of causing a specific bacterial infection. It is only when they transition into the biofilm stage that they become associated with oncogenesis. Consequently, the presence of a certain group of bacteria within a biofilm may establish a distinct connection with the development of cancer. Bacteria could potentially be accountable for approximately 11% to 16% of global cancer cases, including gastrointestinal cancer.Citation41 The development of cancer, known as oncogenesis, is not a single, instantaneous event but rather a complex process that necessitates the occurrence of multiple factors simultaneously.

Figure 1. Association of bacterial biofilm with different types of cancers.

Figure 1. Association of bacterial biofilm with different types of cancers.

Table 1. Possible role of bacteria in different types of cancers.

Microbiota can alter cancer susceptibility and progression through various mechanisms.Citation42 In general, biofilm promotes cancer through various mechanisms. Firstly, they can induce inflammation that persists and fails to eliminate biofilm-associated pathogens. This chronic inflammation may lead to DNA damage, fostering the growth of cancer cells.Citation23 Secondly, biofilms have the ability to modulate and impede the host immune response, creating an environment conducive to the development of cancer.Citation43,Citation44 Thirdly, certain bacteria within biofilms can produce toxins acting as carcinogens, thereby increasing the risk of cancer.Citation45 Fourthly, bacteria residing in biofilms can alter host metabolism. Lastly, emerging evidence suggests that bacteria within the tumor microenvironment (TME), known as the tumor microbiome (TM), actively participate in cancer progressionCitation46 ().In a recent study done by Cullin et al. demonstrated that cancer progression can induce changes in surrounding tissues, blood vessels, and immune responses, creating microenvironments conducive to the growth of specific bacterial strains.Citation47

Figure 2. Mechanisms of cancer progression by bacterial biofilms. 1) Bacterial components trigger TLRs on tumor-infiltrating myeloid cells and macrophages, activating MyD88 and producing inflammatory cytokines like IL-23, TNF-α, and IL-6/11, amplifying NF-κB signalling, in myeloid cells, promoting tumour-associated inflammation 2) Bacterial biofilms in right-sided colorectal cancer (CRC) mucus are linked to early cancer-related changes in normal colon tissue, characterized by reduced E-cadherin, heightened STAT3 activation, increased epithelial IL-6, and elevated N1, N12-diacetylspermine (DiAcspm) levels, demonstrating carcinogenic potential of biofilm 3) Pathogenic bacteria, such as pks+ Escherichia coli and B. fragilis, triggering inflammation and tumorigenesis in normal colorectal tissues through the induction of mutations and genomic instability via microbial products like colibactin and B. fragilis toxin, serving as crucial initiators in colorectal cancer development 4) Commensal bacterial metabolites impact colorectal cancer(CRC) progression by influencing tumor dynamics and immune response. TGF-β inhibits early CRC while promoting metastasis, with butyrate-producing bacteria maintaining gut health, TMAO preventing protein denaturation, and SCF supporting microbiota homeostasis 5) Certain microbes like Enterotoxigenic Bacteroides fragilis and Fusobacterium nucleatum drive pro-tumorigenic inflammation by inducing IL-17 secretion from TH17 cells, promoting epithelial proliferation. This process recruits myeloid cells, such as polymorphonuclear neutrophils, which indirectly support tumor progression through ROS and RNS generation 6) Dysregulation of epithelial barrier pathways (MAPK, STAT3, NF-κB) disrupts colonic integrity. IL-23 triggers pro-inflammatory cytokine production, amplifying inflammation. TNF-α, IL-6, and IL-22 activate STAT3/NF-κB in transformed cells, promoting their survival and proliferation, impairing barrier function.

Figure 2. Mechanisms of cancer progression by bacterial biofilms. 1) Bacterial components trigger TLRs on tumor-infiltrating myeloid cells and macrophages, activating MyD88 and producing inflammatory cytokines like IL-23, TNF-α, and IL-6/11, amplifying NF-κB signalling, in myeloid cells, promoting tumour-associated inflammation 2) Bacterial biofilms in right-sided colorectal cancer (CRC) mucus are linked to early cancer-related changes in normal colon tissue, characterized by reduced E-cadherin, heightened STAT3 activation, increased epithelial IL-6, and elevated N1, N12-diacetylspermine (DiAcspm) levels, demonstrating carcinogenic potential of biofilm 3) Pathogenic bacteria, such as pks+ Escherichia coli and B. fragilis, triggering inflammation and tumorigenesis in normal colorectal tissues through the induction of mutations and genomic instability via microbial products like colibactin and B. fragilis toxin, serving as crucial initiators in colorectal cancer development 4) Commensal bacterial metabolites impact colorectal cancer(CRC) progression by influencing tumor dynamics and immune response. TGF-β inhibits early CRC while promoting metastasis, with butyrate-producing bacteria maintaining gut health, TMAO preventing protein denaturation, and SCF supporting microbiota homeostasis 5) Certain microbes like Enterotoxigenic Bacteroides fragilis and Fusobacterium nucleatum drive pro-tumorigenic inflammation by inducing IL-17 secretion from TH17 cells, promoting epithelial proliferation. This process recruits myeloid cells, such as polymorphonuclear neutrophils, which indirectly support tumor progression through ROS and RNS generation 6) Dysregulation of epithelial barrier pathways (MAPK, STAT3, NF-κB) disrupts colonic integrity. IL-23 triggers pro-inflammatory cytokine production, amplifying inflammation. TNF-α, IL-6, and IL-22 activate STAT3/NF-κB in transformed cells, promoting their survival and proliferation, impairing barrier function.

The study shows strong evidence that both bacterial biofilm formation and planktonic stage bacteria could contribute to cancer development. This association between bacterial biofilms and oncogenesis is unusual as it is based on molecular mechanisms.Citation48

The human colon is protected by a thick mucus barrier primarily composed of mucins, especially Mucin 2 (MUC2). This barrier serves the crucial role of preventing direct contact between the colon’s epithelial cells and the microbiota. This mucus barrier is structured into two distinct layers. The inner layer, which is closely associated with the epithelial cells, is dense and effectively blocks the passage of bacteria, thus creating a separation between commensal bacteria and the host’s epithelial cells. Meanwhile, the outer layer, which is not firmly attached, serves as a natural habitat for commensal bacteria. This dual-layered mucus barrier enables the normal intestinal microbiota to reside within the colonic mucus without triggering an inflammatory response.Citation49 When the protective mucus barrier is breached, it permits the microbiota to directly interact with the colonic epithelium. This event has been proposed as a crucial initial step that triggers alterations in the epithelial cells, ultimately leading to inflammation in the intestine.Citation50 This heightened exposure of the colonic epithelium prompts adjustments in microbial interactions, consequently reshaping the microbial composition and function and frequently leading to the development of a biofilm.Citation40 Bacteria can produce metabolites and signaling molecules that influence cancer cell proliferation, migration, and angiogenesis, ultimately fueling tumor progression.Citation51 Additionally, biofilms have been found to enhance treatment resistance, making cancer cells less susceptible to chemotherapy, radiation therapy, and immunotherapy.Citation22,Citation52

In order to give readers a comprehensive picture of the state of knowledge about emerging implications of bacterial biofilm in different cancer biology at the moment, this review article focus on the recent advancements in the role of bacterial biofilm in cancer causation and outlook for the future. The most recent research results and clinical advancements, including screening techniques, molecular profiling, surgical intervention, targeted therapies, immunotherapy, and upcoming therapeutic options have been discussed in this review.

Colorectal cancer

Colorectal cancer (CRC) ranks as the third most frequently diagnosed and the second deadliest cancer globally.Citation53 In 2020, CRC accounted for around 9.4% of cancer-related fatalities.Citation1 It was estimated that over 1.9 million new cases of CRC, including those affecting the anus, would emerge in 2020, resulting in approximately 935,000 deaths. It indicates that CRC comprised roughly one-tenth of all cancer incidents and deaths during 2020.Citation1 In India, GLOBOCAN and the International Agency for Research on Cancer’s 2020 Indian fact sheets indicate the occurrence of more than 65,358 new cases of CRC and approximately 35,385 associated deaths.

Due to a significant rise in the number of reported cases among the elderly population, it is projected that the worldwide occurrence of CRC will more than double by the year 2035. CRC is a condition that exclusively affects the colon or rectum, and is characterized by the abnormal growth of glandular epithelial cells in the colon. There are three primary categories of CRC: sporadic, hereditary, and colitis-associated. The global incidence of CRC is steadily increasing, with both genetic and environmental factors contributing to an individual’s risk of developing this condition.Citation54,Citation55 Furthermore, as individuals with long-standing ulcerative colitis and Crohn’s disease grow older, their likelihood of developing CRC also rises.Citation56

A well-balanced and healthy intestinal microbiota is essential for several key functions in the human body. It plays a critical role in extracting energy from food,Citation57 shaping the structure of the intestinal lining,Citation58 defending against harmful pathogens,Citation59 and supporting the immune system.Citation60 Contrary to common belief, when there is an imbalance in the intestinal microbiota, known as dysbiosis, it can disrupt various physiological processes in the host, ultimately contributing to the development of various diseases.Citation61

In recent decades, there has been significant research into the comprehensive analysis of the microbiome associated with CRC. Among the bacteria found to play a significant role in promoting tumorigenesis, F. nucleatum, pks+ E. coli, B. fragilis, E. faecalis, and Salmonella spp.Citation62–64 Streptococcus bovis, Helicobacter pylori, and Clostridium septicum Citation38,Citation65().

Trimethylamine-N-oxide (TMAO), a gut bacterial metabolite, is implicated in various diseases, including cardiovascular disease, liver cancer, diabetes, and colorectal cancer (CRC).Citation66,Citation67 TMAO’s connection to CRC involves its production from dietary sources by gut microbiota, influencing the tumor microenvironment and promoting tumorigenesis.Citation68,Citation69 It belongs to a class of microbiota-derived metabolites associated with CRC, impacting intestinal epithelial cells and immune responses.Citation70 E. coli, typically a commensal organism, can become pathogenic with carcinogenic potential, particularly the Pks+ strain. Pks island genes induce DNA double-strand breaks, promoting CRC development.Citation71,Citation72 Enterotoxigenic Bacteroides fragilis (ETBF) strains, producing B. fragilis toxin (bft), are linked to CRC, inducing tumorigenesis through immune signaling cascades and activation of pro-neoplastic changes.Citation73,Citation74 F. nucleatum, enriched in CRC tissue, promotes tumorigenesis through virulence factors and modulation of signaling pathways, contributing to primary cancer cell proliferation and metastasisCitation75,Citation76(). S. gallolyticus subspecies gallolyticus (SGG) is associated with CRC, but its role as a primary driver is unclear. It may enhance tumor growth and colonization in mice, but further investigation is needed.Citation77,Citation78 Salmonella enterica infection, encompassing diverse bacteria, is linked to colon and gallbladder cancer development, with specific proteins, typhoid toxin, and AvrA, implicated in carcinogenesis through modulation of host immune responses and signaling pathways.Citation79,Citation80

Figure 3. Role of bacteria and their metabolites in the causation and progression of CRC.

Figure 3. Role of bacteria and their metabolites in the causation and progression of CRC.

However, there is an urgent need for CRC microbiome research to advance beyond descriptive studies. For example, organoids and gut-on-chip models can help to further microbiome research in a mechanistic way. A genotoxic pks+E. coli mutational signature was recently found to be directly involved in CRC using the gut organoid model.Citation81 It is evident that there is a complex relationship between the bacterial biofilm and CRC.An invasive biofilm shows pathologic features when it invades the mucous layer of the colon and directly contacts the epithelium.Citation82 Around 50% of CRC patients and 13% of healthy people develop mucus-invasive bacterial biofilms on the colon mucosa.Citation83

Research suggests bacteria-stimulated carcinogenesis shows how bacteria to bacteria and bacteria to hosts interact in CRC.Citation84,Citation85 Among the most crucial major pathophysiologic changes in the development of CRC in humans is the increase in the permeability of the gut and the enhancement of barrier activity losses brought on by the bacteria.Citation86 These changes may be caused by biofilm. There are several likely mechanisms by which bacterial biofilms could worsen oncological conditions; in comparison to bacterial transformation, genotoxic stress caused by bacterial toxins has demonstrated the most substantiated links (). For instance, a wide variety of bacteria produce several toxins,colibactin-synthesizing enzymes encoded by the pks+ pathogenicity island in E. coli induce DNA alkylation and double-strand breaks. Cytolethal Distending Toxin (CDT) from E. coli and H. ducreyi reduces replication fork speed in HeLa and U2OS cells, causing genetic instability. Enterotoxigenic B. fragilis, a source of gastrointestinal inflammation, induces the polyamine catabolic enzyme spermine oxidase (SMO), leading to an increase in ROS and DNA damage marker activation in colonic epithelial cells (HT29/c1 and T84), are implicated in genotoxicity and the onset of CRC.Citation81,Citation87–89

Increased tumor burden,Citation90 increased metastasis,Citation91 and chemo resistanceCitation92 are several indications of disease progression that have been linked to the enrichment of CRC-linked species in the gut microbiota. However, in CRC patients, there may be differences in the enrichment of taxa between fecal and mucosal samples. F.nucleatum, for instance, is frequently isolated from samples of biopsies of mucosal tissues rather than from fecal samples, despite having been previously related to CRC.Citation93

Additionally, it has been found that CRC patient’s mucosal and fecal microbiomes have different metabolic outputs, which may be disease-relevant.Citation94 Certain bacterial taxa can both enrich around colonic malignancies and produce intricate biofilms directly on lesions. Particularly in proximal colonic tumors, biofilms are commonly observed and are frequently associated with bacterial invasion of the tumor tissue itself.Citation40 Direct interaction between bacteria and tumor cells may cause localized inflammation and the recruitment of immune cells,Citation95 which eventually encourages an environment that is conducive to cancer.

Gall bladder cancer

Gallbladder cancer (GBC) is the sixth most common and highly malignant cancer of the GI tract.Citation96 Due to its deep location in the human body, GBC does not display any visible symptoms at the early stages, which results in poor diagnostic rates and a survival rate of less than five years due to poor prognosis.Citation97 According to GLOBOCAN 2020 data, 115,949 newly diagnosed cases of GBC were reported and 84,695 deaths occurred in the year 2020.Citation1 The formation of bacterial biofilms by Salmonella typhi in the gallbladder (GB) has been linked to an elevated susceptibility to the development of gallbladder cancer. Salmonella typhi is a Gram-negative, rod-shaped, flagellated and aerobic bacterium. Within the gallbladder, the antimicrobial activity of bile stimulates the production of exopolysaccharides containing O-antigens, facilitating the formation of S. typhi biofilms on gallstones.Citation97,Citation98 This biofilm growth is aided by the presence of bile and gallstones, which establish a favorable environment, supply essential nutrients for bacterial adhesion and growth on gallstone surfaces, and modulate the expression of proteins crucial for biofilm formation.Citation99

S. typhi invades the mucosal surface of GB and forms a biofilm; releasing carcinogenic agents like bacterial glucuronidase, nitroso compounds,Citation100 A2B5 typhoid toxin and cytolethal distending toxins (CDT) to promote GBC. CDT induces DNA damage, cell cycle arrest, and apoptosis.Citation101 The CDT is a tripartite toxin complex composed of cdtA, cdtB, and cdtC polypeptides causes DNA damage, cell cycle arrest, and apoptosis.Citation102–104

A2B5 typhoid toxin, causes DNA damage and G2 cell cycle arrest by binding exclusively to sialic acid-containing glycoproteins of host cells.Citation105 This DNA damage activates various checkpoints and inactivates CDK1. Cells exposed to this toxin undergo senescence-associated beta-galactosidase activity, which aids the survival of cells accumulating DNA damage and causing genomic instability that may result in gallbladder cancer. Gallstone carriers demonstrate a higher S. typhi ratio compared to individuals with benign gallbladder diseases.Citation106,Citation107 As per a study, those who have S. typhi infection are more likely to get GBC, with a 22% positive rate.Citation28

Numerous studies have highlighted the significance of bacterial effector proteins like SipA, Salmonella outer protein B SopB, Salmonella outer protein E SopE, Salmonella outer protein E2 SopE2, and SptP, which not only facilitate GB cell invasion but also activate mitogen-activated protein kinase (MAPK) and Protein kinase B (AKT) pathways, contributing to cell transformation observed in GC tumors of Indian patients infected by S. typhi .Citation108,Citation109 Biofilm formation, in S. typhi involves various contributing factors such as SPI-1 and SPI-2 genes encode type III secretion system which is essential for invasion and colonization,Citation110 AsfD enhances S. typhi motility by elevating the expression of flagellar genes. Additionally, RibS upregulates cyclopropane fatty acids synthase gene (cfa), facilitating the conversion of unsaturated fatty acids to CFAs. This increased CFA content promotes bacterial biofilm production.Citation111,Citation112

Moreover, bile affects S. typhi biofilm formation by penetrating bacterial cells through Mig-14, an inner-membrane-associated protein. This protein enhances biofilm growthCitation113 by altering the permeability of the outer membrane in response to adverse environments like the gallbladder acidic conditions and bile antimicrobial nature. GalE contributes to biofilm formation through the production of uridine diphosphogalactose-4-epimerase, vital for the formation of lipopolysaccharide O-antigen of S. typhi. The GalE mutant (Tn10 inserted) containing S. typhi developed a very thin biofilm on gallstones.Citation99

Quorum sensing (QS) emerges as a pivotal mechanism in the intricate interplay between S. typhi and the gallbladder environment, adding another layer of complexity to the development of gallbladder cancer (GBC). In stressful conditions, QS is a key player in biofilm formation, and S. typhi utilizes two autoinducers (AI-I and AI-II). Specifically, AI-II, generated through the luxS synthase gene, governs the expression of virulence genes in bacterial pathogens. A luxS gene mutant exhibited reduced biofilm-forming ability,Citation114 leading to downregulation of chemotaxis genes, spi genes, and motility-related genes.Citation115 The intricate interplay between S. typhi and the gallbladder environment has been shown to contribute significantly to the development of GBC. These findings highlight the importance of addressing bacterial infections and biofilm formation as potential factors in the prevention and management of gallbladder cancer.

Head and neck cancer

Head and neck squamous cell carcinoma (HNSCC) rank as the sixth most prevalent cancer worldwide, encompassing various subtypes such as oral, pharyngeal, sinus, nasal cavity, and salivary gland carcinoma. Notably, oral cancer is primarily associated with biofilm formation. The primary risk factors for head and neck cancer include cigarette smoking and alcohol consumption, environment plays a crucial role in shaping the bacterial community, influenced by factors such as temperature, oxygen tension, pH, substratum properties, nutrient availability, and exposure to cell and immune signaling.Citation116,Citation117 In the context of HNSCC, poor oral hygiene may contribute to microbial overgrowth or alter the composition of the oral microbiota, leading to immune homeostasis disruption. Inflammation, as a consequence, can further alter the types of microbes in the oral niche, perpetuating dysbiosis. Bacterial presence in HNSCC tissues has been confirmed through both microbiological culture and 16S rRNA sequencing.Citation118 This intricate interplay between dysbiosis of the microbiome and the formation of biofilms significantly influences the development of HNSCC.Citation119

Moreover, the oral microbiome is directly associated with the carcinogenicity of these substances by producing mutagenic acetaldehyde from alcohol.Citation120 An intriguing aspect is the potential of bacterial biofilm growth in epithelium to create conditions leading to oncogenic transformation of epithelial cells. This phenomenon has been demonstrated in the context of colon carcinoma.Citation40 Biofilm formation causes significant disturbances in inflammatory metabolites, induces epithelial damage, and promotes cell proliferation.Citation121 Moreover, it has been shown to induce apoptotic processes in the head and neck region.Citation122 The association between bacterial biofilms and their impact on the local environment sheds light on additional mechanisms contributing to the development and progression of HNSCC. Recent research has unveiled distinctive microbiomes within oral HNSCC, characterized by the presence of Selenomonas, Fusobacterium, Leptotrichia, and Treponema. Conversely, non-oral HNSCC tends to exhibit a higher prevalence of Clostridium and Pseudoalteromonas .Citation123 These intriguing discoveries emphasize the utmost significance of understanding the intricate interplay between microbial communities and biofilms during the initiation and progression of HNSCC. This knowledge opens up promising pathways for pioneering breakthroughs in combatting this widespread and formidable type of cancer.

Oral cancer

Oral squamous cell carcinoma (OSCC) is the most prevalent malignant cancer of the oral cavity. OSCC stands as the most prevalent subset of head and neck cancer, contributing significant morbidity and mortality worldwide. Recent studies have confirmed that periodontitis triggers microbial dysbiosis and virulence factor expression, rather than introducing new pathogens.Citation124,Citation125 Within a study it was found that within the oral microbiome of individuals with OSCC, Fusobacteria played a pivotal role in the upregulation of virulence factors such as capsule biosynthesis, flagellum synthesis and assembly, chemotaxis, iron transport, hemolysins and adhesins.Citation126 F. nucleatum due to its rod shape serves as a crucial bridge between initial colonizers like Streptococcal spp and later inhabitants such as Porphyromonas gingivalis.Citation127 This vital role contributes to the intricate web of polymicrobial biofilms and facilitates dynamic interactions among microorganisms.Citation128 In an investigation, a notable increase in the abundance of Porphyromonas and Fusobacterium was evident in OSCC tissues compared to normal tissue counterparts.Citation129 In OSCC samples, F. nucleatum was the dominant species,Citation130 with Pseudomonas aeruginosa being the second most common.Citation131 Similarly, among the three Fusobacterium species, F. nucleatum was significantly elevated in oral cancer, while P. gingivalis remained consistent across groups.Citation132

F. nucleatum has been associated with initiating oral malignancy in a chemically induced mouse model of OSCC.Citation133 This study revealed that both P. gingivalis and F. nucleatum have the ability to induce carcinogenesis by direct interaction with Toll-like receptors (TLR) of oral epithelial cells through FadA and upregulates IL-6-STAT3 signaling pathway. Furthermore, F. nucleatum infection promotes cyclin D1 and matrix metalloproteinase-9 (MMP-9) activity, influencing the progression and invasiveness of oral tumors.

According to “two-hit” theory somatic alterations serves as the initial hit and F. nucleatum serves as the second hit, intensifying cancer progression from benign to malignancy.Citation134 F. nucleatum triggers genomic instability, maintains growth signals, suppresses tumor suppressor genes, weakens immune responses, promotes pro-tumor inflammation, stimulates invasion and metastasis through epithelial-mesenchymal transition (EMT).Citation35 Further studies are needed to completely understand how F. nucleatum contributes to OSCC development at the molecular and cellular level.

Pharyngeal cancer

Oral cavity and pharynx neoplasms rank as the seventh most prevalent cancers globally, with around 710,000 new cases annually and 359,000 deaths per year.Citation135 While smoking and alcohol consumption are widely recognized as major risk factors for these cancers, the International Agency for Research on Cancer has acknowledged certain HPV genotypes as carcinogens for specific head and neck anatomical subsites since 2007. Among the 200 types of HPV, approximately 15 are linked to malignancies, with HPV16 standing out as one of the most common and potent carcinogenic types.Citation136 Although the exact contribution of biofilms to the development and advancement of this cancer remains a subject of ongoing investigation, interesting findings have been found in patients with nasopharyngeal carcinoma (NPC) who have undergone radiotherapy. These individuals have reported side effects, including osteoradionecrosis (ORN) and mucositis. Oral mucositis is a prevalent condition characterized by acute inflammation of the oral mucosa, often occurring as a complication of systemic cancer therapy or radiotherapy. It is particularly common following radiotherapy for nasopharyngeal carcinoma (NPC),which have been associated with the development of biofilms.

Radiotherapy for head and neck cancer (HNC) can cause significant harm to salivary glands and oral mucous membranes, leading to reduced saliva production and a compromised epithelium. This creates favorable conditions for bacterial growth in the oral cavity.Citation137 Recent research indicates a crucial role of the oral microbiota in osteoradionecrosis (ORN) development,Citation138 with increased Actinomycetes abundanceCitation139and prevalence of Streptococcus intermedius in irradiated patients.Citation140

Li et al. study found that osteoradionecrosis (ORN) lesions exhibit changes in oral microflora compared to healthy tissues, with increased abundance of bacteria such as Prevotellaceae, Fusobacteriaceae, Porphyromonadaceae, Actinomycetaceae, Staphylococcaceae, Prevotella, Staphylococcus, and specific species like Endodontalis and Prevotella intermedia. Conversely, Streptococcaceae, Streptococcus, and Haemophilus were more abundant in normal tissues.Citation141

Prevotella and Streptococcus were hypothesized to play functional roles in ORN lesions and healthy oral tissues, respectively, and identified as potential diagnostic and prognostic biomarkers. Prevotella, associated with ORN progression, stimulates dendritic cells via toll-like receptor 2, leading to IL-1β, IL-6, and IL-23 release. This triggers T helper 17 cell-mediated IL-17 production, activating neutrophils and promoting periodontitis.Citation38 Radiation exposure transforms normally commensal Prevotella into an opportunistic pathogen, producing virulence factors. Prevotella-induced lipopolysaccharides (LPS)Citation39 inhibit osteogenesis and induce osteoclast formation, potentially impairing bone regeneration. Enriched biosynthesis and metabolism-related pathways in ORN microorganisms suggest Prevotella’s complex role beyond contamination, contributing to bone loss. Prevotella emerges as a potential prognostic biomarker for ORN clinical progression.Citation141 A noteworthy study revealed a higher prevalence of biofilms in NPC patients who developed ORN compared to their counterparts who did not exhibit ORN.Citation142

These biofilms possess unique compositions, featuring methicillin-resistant Staphylococcus aureus and Streptococcus viridans in ORN biofilms. Staphylococcus aureus typically resides as a component of the body’s normal flora. However, extensive antibiotic use has led to the development of methicillin-resistant strains (MRSA). These MRSA variants have the ability to create biofilms and are associated with various infections, including those involving indwelling foreign bodies, bacteremia, soft tissues, endocardium, and bones such as osteomyelitis. Citation143–145 In the context of mucositis, Actinobacteria, Veillonella, and Fusobacteria are prominent. Interestingly, Veillonella plays a pivotal role in facilitating the adhesion of Streptococcus mutans, while Actinobacteria disrupt amino acid metabolism, leading to a slowdown in the healing of mucosal injuries caused by radiotherapy.Citation146

In addition, recent research revealed Streptococcus (37.3%), Fusobacterium (11.3%), and Prevotella (10.6%) as the dominant genera in the swab and tissue samples of pharyngeal carcinoma patients.Citation147 These biofilms exhibit resistance to conventional treatments for pharyngeal cancer, presenting a profound challenge in effective management. The characteristics of biofilms, such as limited nutrient diffusion, constrained antimicrobial passage, and modification of the surroundings to create a less favorable environment, collectively result in extensive resistance and tolerance to antimicrobial agents. The intricate relationship between biofilms and pharyngeal cancer continues to evolve, necessitating further investigation to fully unveil their specific contributions and mechanisms. Nevertheless, the presence of biofilms and their impact on treatment-related side effects in patients with pharyngeal cancer after radiotherapy remain an intriguing and vital area of scientific interest.

Salivary gland carcinoma

Salivary gland carcinoma (SGC) is the cancer of salivary glands and it is the rare type of head and neck carcinoma. Incidence ranging from 0.4 to 3.5 per 100,000 per year in the Western world. The role of biofilm in the development of SGC is not fully explored. Some studies have proposed a potential association between Helicobacter pylori and salivary gland tumors. Notably, a study uncovered an intriguing connection between localized H. pylori infection, Sjögren’s disease, and salivary gland MALT lymphoma.Citation37 Interestingly, H. pylori seems to act as an additional antigenic stimulation, influencing the development of both salivary gland and gastric MALT lymphoma. This discovery implies that H. pylori may serve as a booster, promoting B cell proliferation and contributing to the progression of lymphoma. In research done by Jiang et al. unstimulated saliva samples were collected from 13 SACC patients and 10 healthy controls.Citation36 Through comprehensive analysis using 16S rRNA sequencing and whole-genome shotgun metagenomic sequencing, the study examined microbial diversities, compositions, and functions. The alpha diversity analysis revealed no significant differences between SACC patients and healthy controls. However, beta diversity demonstrated a discernible separation trend. SACC patients exhibited higher levels of Streptococcus and Rothia, while Prevotella and Alloprevotella were more prevalent in healthy controls. This research sheds light on the microbial landscape in the oral microbiota of individuals with SACC and highlights distinct differences in microbial abundances between SACC patients and their healthy counterparts.

However, the precise mechanisms, relationships between biofilms and salivary gland cancer remain a subject of ongoing investigation and scientific debate.

Lung cancer

Lung cancer (LC) ranks as the second most frequently diagnosed cancer in both males and females, trailing prostate and breast cancer. In 2020, LC affected 2.2 million individuals, which accounted for 11.4% of all cancers. This disease resulted in 1.7 million deaths, making up around 18% of all cancer-related deaths globally (Globocan, 2020). While cigarette smoking stands as the one of the predominant causes of lung cancer,Citation148 recent studies have unveiled a compelling connection between chronic pulmonary infections and the development of LC. Interestingly, such infections may act independently or in conjunction with smoking, elevating the risk of lung cancer.Citation149,Citation150 Tissue samples from lung cancer patients have uncovered the presence of diverse pathogens, with mycoplasma being prevalent in lung carcinomas.Citation151 Studies suggest that mycoplasma-infected cells (Microplasma arginine infected RPMI 4788 cells) invivo exhibit potential for cancer metastasis compared to non-infected cells.Citation152

Moreover, several meta-analyzes have shown an increased susceptibility to lung cancer among individuals with active pulmonary tuberculosis. It is important to highlight that standard TB treatment lasts for 6 to 9 months, during which the significant lung inflammation resulting from TB infection may play a role in a chronic inflammatory process associated with cancer development.Citation153,Citation154

In addition to Mycoplasma and Mycobacterium tuberculosis, certain bacteria, such as Chlamydia pneumoniae and Staphylococcus strains, have emerged as potential contributors to lung carcinogenesis. Citation155–157 Some studies also found bacterial colonies in the respiratory tracts of lung cancer patients, including Haemophilus influenza and Candida albicans, which are particularly linked to lower respiratory tract malignancies. Furthermore, Legionella pneumophila, along with strains of Bacillus, Listeria, and Streptococcus, have been detected in lung cancer patients.Citation158,Citation159 The available data strongly supports the intriguing idea that lung cancer may be linked to chronic infections involving biofilms. As a result, a growing number of researchers are suggesting that lung malignancies might essentially be communities of various pathogens that have developed resistance to antibiotics. This emerging perspective underscores the potential role of persistent bacterial infections in lung cancer, prompting the exploration of novel strategies to understand these intricate interactions more effectively.

Gastric cancer

Gastric cancer (GC) remains a significant worldwide health concern, making a substantial impact on cancer-related fatalities. It ranks as the fifth-most prevalent cancer worldwide.GC new cases: 1,089,103 (5.6%) and caused approximately 768,793 (7.7%) deaths in 2020.Citation160,Citation161 Current research has revealed the role of bacterial biofilms in this intricate disease process, even though Helicobacter pylori infection has long been linked to the development of GC.Citation162 The incidence and mortality rates for GC in Eastern Asian countries are notably higher than in other regions, accounting for more than 60% of global cases.Citation162 In many localized chronic infections, the formation of biofilms by bacterial populations is the key virulence factor.Citation163 There are several risk factors linked to GC, including Helicobacter pylori infection, genetic changes, ethnicity, dietary habits, and lifestyle choices.Citation164,Citation165 Additionally, specific inherited GC syndromes, like hereditary diffuse gastric cancer (HDGC) resulting from mutations in the CDH1 tumor suppressor gene, are associated with a significantly elevated risk of GC. CDH1 mutations are believed to be responsible for GC development in approximately 80% of patients.Citation164

The initial proof of H. pylori ability to form biofilms while establishing itself in the human gastric mucosa was captured through photographic documentation.Citation166 In the context of GC, the bacterial biofilm comprises various species, with H. pylori often serving as the cornerstone organism. Other bacterial species, such as Streptococcus, Prevotella, and Fusobacterium, have also been found to be associated with GC biofilmsCitation167,Citation168 In the human, gastric lumen is known for its harsh conditions, which are detrimental to many bacteria. Despite these challenges, H. pylori manages to thrive by utilizing its urease activity to counteract and neutralize the acidity.Citation169

On the other hand, biofilm development might be more important for its ongoing colonization. Surprisingly little is understood about the development of biofilms on human gastric mucosa while they are in-vivo. Using powerful electron microscopes, various research studies revealed dense clusters of H. pylori .Citation170,Citation171 While H. pylori isolated from GC patients is usually non-culturable, other methods (PCR or histological method) can detect them; as well, coccoid forms are more frequent in gastric mucosa of GC patients than those of peptic ulcer patients.Citation172

Recent studies utilizing transcriptomic and proteomic analyzes have obtained significant insights into the shift from the planktonic phase to the biofilm stage in H. pylori. These investigations reveal increased gene expression related to adhesins, flagella, toxins, efflux pumps, lipopolysaccharides (LPS), the type IV secretion system, urease, and hydrogenase. Such heightened expression suggests adaptations for acquiring alternative energy sources during the transitionCitation173 and during prolonged inflammation plays a role in the development of several malignancies, but it is especially crucial for GC linked to H. pylori .Citation174 Certainly, when gastric mucosa inflammation persists, it leads to elevated nitric oxide production. This, in turn, plays a role in causing harm to nucleotide bases and influencing transcriptional regulation by boosting DNA methyl transferase activity.Citation175 Indeed, for the E-cadherin tumor suppressor gene, its promoter region has often been seen to be hypermethylated in H. pylori infections of adult patients, and its transcriptional activity will be altered in gastric cells.Citation176 Inflammatory responses have been shown to be controlled by bacterial virulence factors. H. pylori virulence factors, such as the VacA and CagA, can inhibit T-cell activation and evade recognition by Toll-like receptor. Additionally, CagA has been shown to promote epithelial to mesenchymal transition (EMT), which contributes to the generation of cancer stem cells.Citation177,Citation178 On the other hand, γGT can promote oxidative stress and enhance apoptosis of gastric cells.Citation174 Inherent host-specific mechanisms counterbalance these responses. Evidence from knockout animal models underscores the significance of host factors in regulating inflammation. For instance, the anti-inflammatory outcomes linked to TLR9 signaling highlight the role of host elements in modulating inflammatory processes.Citation174

There are various factors that might account for the presence of bacterial biofilms in stomach cancer. The primary advantage of biofilms is that they give bacteria a protective habitat from various immunological reactions and antibacterial substances. Infection and chronic inflammation may result from this enhanced resilience, which is a known risk factor for GC. Inflammation of the gastric mucosa can be triggered by bacterial biofilms.Citation23 The sustained inflammation may cause DNA damage and genomic instability, both precursors of cancer initiation, over time due to the production of reactive oxygen and nitrogen species.Citation179,Citation180 Recent studies suggest that bacterial biofilms can affect epithelial-mesenchymal transitions (EMTs).It may be contributing to cancer metastasis that EMT is induced by biofilms.Citation181,Citation182 The expression of microRNAs, which are short, non-coding RNAs that control gene expression, can be altered by bacterial biofilms. Alterations in microRNA expression may encourage oncogenic transformations in gastric epithelial cells, promoting the growth and development of tumors.Citation183,Citation184

Pancreatic cancer

Pancreatic cancer (PC) is a highly aggressive malignancy, the incidence and mortality rates of pancreatic cancer are closely related.Citation185 In 2020, it ranked as the 14th most prevalent cancer type, with approximately 495,733 newly diagnosed cases and 466,003 reported fatalities.Citation1 There is a 50% survival rate after 6 months for patients diagnosed with cancer. Currently, pancreatic cancer patients with chemotherapeutic options live nearly as long as their incidence because chemotherapy only marginally prolongs life.Citation186

Before 2030, PC is predicted to overtake all other cancers as the second leading cause of death.Citation187 PC incidence rates each year are rapidly rising. Up to 90% of PC patients pass away within five years of diagnosis, and more than 50% die so within the first six months.Citation188,Citation189 Pancreatic cancer risk has recently been associated with bacterial infections that lead to periodontal diseaseCitation190

There have been a number of studies linking P. gingivalis and F. nucleatum to PC.Citation191,Citation192 Furthermore, Wei et al. noted that both Leptotrichia and Streptococcus were associated with a higher risk of developing PC.Citation30 Pushalkar et al. observed an increase in the relative abundance of Proteobacteria, Synergistetes, and Euryarchaeota in the fecal samples of individuals with pancreatic ductal adenocarcinoma (PDAC) compared to healthy controls.Citation193 Additionally, a small study by Half et al. reported elevated levels of Sutterela, Bacteroides, Odoribacter, and Akkermansia in the feces of patients with pancreatic cancer (PC) compared to healthy individuals.Citation194 Another study done by Half et al. found reduced levels of Firmicutes genera in the fecal samples of PC patients. Examining overall gut microbial diversity,Citation195 Ren et al. observed a significant reduction in diversity among PC patients, though no significant differences were identified between different PC subtypes.Citation196 Thomas et al. using a murine model, demonstrated that the intestinal microbiota play a crucial role in pancreatic cancer progression, as mice with depleted microbiota exhibited decreased tumorigenicity.Citation197,Citation198 Emphasized that alterations in fecal microbiota composition manifest early in the course of tumor progression in a murine model of PDAC.Gut dysbiosis is implicated in pancreatic cancer development by activating chronic inflammation. LPS, through Toll-like receptor 4 (TLR4), inhibits tumor suppressor proteins (PTEN, pRb, MAP2K4, p53) and induces HIF-1α and STAT3, promoting cell migration and epithelial – mesenchymal transition (EMT)),Citation199–201 Additionally, LPS, interacting with NF-κB, MyD88, and AKT, upregulates programmed cell death ligand 1 (PD-L1), reducing immune responses by inducing apoptosis of tumor-infiltrating lymphocytes (TILs).Citation202 While LPS may lead to long-term inflammatory cell depletion, in early stages, it increases local CD3+ and CD8+ T cells. The gut microbiota contributes to pancreatic cancer by inducing sustained inflammatory responses with the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS).Citation203 ROS have the potential to harm DNA and cell membranes, while also disrupting proper protein folding and increasing concentrations of oncogenes. An alternative suggested mechanism entails communication between gut microbiota and pancreatic cells via the mammalian target of the rapamycin (mTOR) pathway. This interplay affects cellular processes such as growth, autophagy, and cytoskeletal organization. This interaction influences cell growth, autophagy, and cytoskeletal organization.Citation204 Chronic inflammation, elevated oncogenes (e.g., Kras), and microbiota-induced barrier disruption collectively contribute to pancreatic carcinogenesis.Citation205,Citation206 There is strong evidence that P. gingivalis promotes the progression of pancreatic cancer, which has been validated in murine models.Citation207,Citation208 PC may be associated with bacterial biofilms.In spite of the limited knowledge of biofilm formation in the pancreatic ducts,Citation25 no direct link has been established between the PC and bacterial biofilms.Citation20

Discussion

Bacterial biofilms have significant clinical implications, as they highlight the potential need to address as a therapeutic target to improve treatment efficacy. Moreover, the complex interplay between bacterial biofilm, and the immune system within the tumor microenvironment can dictate the overall prognosis for cancer patients.Citation209 Certain bacterial species within the biofilm have been associated with a pro-tumorigenic immune response, leading to poor clinical outcomes. Conversely, some bacteria have shown immune-stimulatory properties, eliciting an anti-tumor immune response.Citation210

Understanding these intricate dynamics may provide opportunities for personalized therapies that target the tumor-associated microbiota and optimize treatment strategies.Citation211 In this era of precision medicine; the recognition of bacterial biofilms as influential players in cancer biology adds a new layer of complexity to our understanding of the disease. The mechanisms by which biofilms impact tumorigenesis, metastasis, and treatment response is an active area of research. By deciphering these connections, researcher and clinicians can identify novel therapeutic targets and develop innovative strategies to improve cancer treatment outcomes. The emerging association of bacterial biofilms in cancer biology have opened up a new frontier in cancer research.Citation212 Understanding the role of biofilms within the tumor microenvironment holds promise for advancing our knowledge of cancer progression, treatment resistance, and patient outcomes. By investigating and targeting these microbial communities, these findings may pave the way for innovative approaches that can augment existing cancer therapies and ultimately improve the lives of cancer patients worldwide.

Disclosure statement

No potential conflict of interest was reported by the author(s).

References

  • Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J Clin. 2021;71:209–249. doi:10.3322/caac.21660.
  • Flemming H-C, Wingender J, Szewzyk U, Steinberg P, Rice SA, Kjelleberg S. Biofilms: an emergent form of bacterial life. Nat Rev Microbiol. 2016;14(9):563–575. doi:10.1038/nrmicro.2016.94.
  • Mirzaei R, Abdi M, Gholami H. The host metabolism following bacterial biofilm: what is the mechanism of action? Rev Med Microbiol. 2020;31(4):175–182. doi:10.1097/MRM.0000000000000216.
  • Mirzaei R, Mohammadzadeh R, Sholeh M, Karampoor S, Abdi M, Dogan E, Moghadam MS, Kazemi S, Jalalifar S, Dalir A, et al. The importance of intracellular bacterial biofilm in infectious diseases. Microb Pathog. 2020;147:104393. doi:10.1016/j.micpath.2020.104393.
  • Koo H, Yamada KM. Dynamic cell–matrix interactions modulate microbial biofilm and tissue 3D microenvironments. Curr Opin Cell Biol. 2016;42:102–112. doi:10.1016/j.ceb.2016.05.005.
  • Akyıldız İ, Take G, Uygur K, Kızıl Y, Aydil U. Bacterial biofilm formation in the middle-ear mucosa of chronic otitis media patients. Indian J Otolaryngol Head Neck Surg. 2013;65(S3):557–561. doi:10.1007/s12070-012-0513-x.
  • Masters EA, Trombetta RP, De Mesy Bentley KL, Boyce BF, Gill AL, Gill SR, Nishitani K, Ishikawa M, Morita Y, Ito H, et al. Evolving concepts in bone infection: redefining “biofilm”, “acute vs. chronic osteomyelitis”, “the immune proteome” and “local antibiotic therapy. Bone Res. 2019;7:20. doi:10.1038/s41413-019-0061-z.
  • Southey-Pillig CJ, Davies DG, Sauer K. Characterization of temporal protein production inPseudomonas aeruginosa Biofilms. Biofilms J Bacteriol. 2005;187(23):8114–8126. doi:10.1128/JB.187.23.8114-8126.2005.
  • Machado A, Cerca N. Influence of biofilm formation by Gardnerella vaginalis and other anaerobes on bacterial vaginosis. J Infect Dis. 2015;212(12):1856–1861. doi:10.1093/infdis/jiv338.
  • Von Rosenvinge EC, GA O, Macfarlane S, Macfarlane GT, Shirtliff ME. Microbial biofilms and gastrointestinal diseases. Pathogens Disease. 2013;67:25–38. doi:10.1111/2049-632X.12020.
  • Behlau I, Gilmore MS. Microbial biofilms in ophthalmology and infectious disease. Archives of Ophthalmology. 2008;126(11):1572 doi:10.1001/archopht.126.11.1572.
  • Vieira Colombo AP, Magalhães CB, FARR H, Martins Do Souto R, Maciel Da Silva-Boghossian C. Periodontal-disease-associated biofilm: a reservoir for pathogens of medical importance. Microb Pathog. 2016;94:27–34. doi:10.1016/j.micpath.2015.09.009.
  • Delcaru C, Alexandru I, Podgoreanu P, Grosu M, Stavropoulos E, Chifiriuc M, Lazar V. Microbial biofilms in urinary tract infections and prostatitis: etiology, pathogenicity, and combating strategies. Pathogens. 2016;5(4):65. doi:10.3390/pathogens5040065.
  • Costerton JW, Stewart PS, Greenberg EP. Bacterial biofilms: a common cause of persistent infections. Science. 1999;284(5418):1318–1322. doi:10.1126/science.284.5418.1318.
  • Sharma D, Misba L, Khan AU. Antibiotics versus biofilm: an emerging battleground in microbial communities. Antimicrob Resist Infect Control. 2019;8(1):76. doi:10.1186/s13756-019-0533-3.
  • Uruén C, Chopo-Escuin G, Tommassen J, Mainar-Jaime RC, Arenas J. Biofilms as promoters of bacterial antibiotic resistance and tolerance. Antibiotics. 2021;10:3. doi:10.3390/antibiotics10010003.
  • Dejea CM, Fathi P, Craig JM, Boleij A, Taddese R, Geis AL, Wu X, DeStefano Shields CE, Hechenbleikner EM, Huso DL, et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science. 2018;359(6375):592–597. doi:10.1126/science.aah3648.
  • Martinez-Medina M, Naves P, Blanco J, Aldeguer X, Blanco JE, Blanco M, Ponte C, Soriano F, Darfeuille-Michaud A, Garcia-Gil LJ. Biofilm formation as a novel phenotypic feature of adherent-invasive Escherichia coli(AIEC). BMC Microbiol. 2009;9:202. doi:10.1186/1471-2180-9-202.
  • Prudent V, Demarre G, Vazeille E, Wery M, Quenech’du N, Ravet A, Dauverd - Girault J, van Dijk E, Bringer M-A, Descrimes M, et al. The Crohn’s disease-related bacterial strain LF82 assembles biofilm-like communities to protect itself from phagolysosomal attack. Commun Biol. 2021;4(1):1–13. doi:10.1038/s42003-021-02161-7.
  • Swidsinski A, Weber J, Loening-Baucke V, Hale LP, Lochs H. Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease. J Clin Microbiol. 2005;43(7):3380–3389. doi:10.1128/JCM.43.7.3380-3389.2005.
  • Weng J, Li S, Zhu Z, Liu Q, Zhang R, Yang Y, Li X. Exploring immunotherapy in colorectal cancer. J Hematol Oncol. 2022;15(1):95. doi:10.1186/s13045-022-01294-4.
  • Stoodley P, Sauer K, Davies DG, Costerton JW. Biofilms as complex differentiated communities. Annu Rev Microbiol. 2002;56(1):187–209. doi:10.1146/annurev.micro.56.012302.160705.
  • Vestby LK, Grønseth T, Simm R, Nesse LL. Bacterial biofilm and its role in the pathogenesis of disease. Antibiotics. 2020;9(2):59. doi:10.3390/antibiotics9020059.
  • Al-Hilu SA, Al-Shujairi WH. Dual role of bacteria in carcinoma: stimulation and inhibition. Iran J Pediatr Hematol Oncol. 2020;2020:e4639761. doi:10.1155/2020/4639761.
  • Rizzato C, Torres J, Kasamatsu E, Camorlinga-Ponce M, Bravo MM, Canzian F, Kato I. Potential role of biofilm formation in the development of digestive tract cancer with special reference to helicobacter pylori infection. Front Microbiol. 2019;10:846. doi:10.3389/fmicb.2019.00846.
  • Bonnet M, Buc E, Sauvanet P, Darcha C, Dubois D, Pereira B, Déchelotte P, Bonnet R, Pezet D, Darfeuille-Michaud A. Colonization of the human gut by E. coli and colorectal cancer risk. Clin Cancer Res. 2014;20(4):859–867. doi:10.1158/1078-0432.CCR-13-1343.
  • Cheng Y, Ling Z, Li L. The intestinal microbiota and colorectal cancer. Front Immunol [Internet]. 2020;11. doi:10.3389/fimmu.2020.615056.
  • Shukla R, Shukla P, Behari A, Khetan D, Chaudhary RK, Tsuchiya Y, Ikoma T, Asai T, Nakamura K, Kapoor VK. Roles of Salmonella typhi and Salmonella paratyphi in gallbladder cancer development. Asian Pac J Cancer Prev. 2021;22(2):509–516. doi:10.31557/APJCP.2021.22.2.509.
  • Tian Y, Hao T, Cao B, Zhang W, Ma Y, Lin Q, Li X. Clinical end-points associated with mycobacterium tuberculosis and lung cancer: implications into host-pathogen interaction and coevolution. Biomed Res Int. 2015;2015:827829. doi:10.1155/2015/827829.
  • Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ, Gapstur SM, Purdue MP, Abnet CC, Stolzenberg-Solomon R, Miller G, et al. Human oral microbiome and prospective risk for pancreatic cancer: a population-based nested case-control study. Gut. 2018;67(1):120–127. doi:10.1136/gutjnl-2016-312580.
  • Jacob JA. Study links periodontal disease bacteria to pancreatic cancer risk. JAMA. 2016;315(24):2653. doi:10.1001/jama.2016.6295.
  • Boleij A, Schaeps RMJ, Tjalsma H. Association Between streptococcus bovis and colon cancer. J Clin Microbiol. 2009;47(2):516. doi:10.1128/JCM.01755-08.
  • Zarkin BA, Lillemoe KD, Cameron JL, Effron PN, Magnuson TH, Pitt HA. The triad of streptococcus bovis bacteremia, colonic pathology, and liver disease: annals of surgery. Annals Of Surgery. 1990;211(6):786. doi:10.1097/00000658-199006000-00019.
  • Tateda M, Shiga K, Saijo S, Sone M, Hori T, Yokoyama J, Matsuura K, Takasaka T, Miyagi T. Streptococcus anginosus in head and neck squamous cell carcinoma: implication in carcinogenesis. Int J Mol Med [Internet]. 2000. [accessed 2023 Aug 16]. doi:10.3892/ijmm.6.6.699.
  • Gopinath D, Wie CC, Banerjee M, Thangavelu L, Kumar RP, Nallaswamy D, Botelho MG, Johnson NW. Compositional profile of mucosal bacteriome of smokers and smokeless tobacco users. Clin Oral Invest. 2022;26(2):1647–1656. doi:10.1007/s00784-021-04137-7.
  • Jiang Q, Liu X, Yang Q, Chen L, Yang D. Salivary microbiome in adenoid cystic carcinoma detected by 16s rRNA sequencing and shotgun metagenomics. Front Cell Infect Microbiol. 2021;11:774453. doi:10.3389/fcimb.2021.774453.
  • Nishimura M, Miyajima S, Okada N. Salivary gland MALT lymphoma associated with Helicobacter pylori infection in a patient with Sjögren’s syndrome. J Dermatol. 2000;27(7):450–452. doi:10.1111/j.1346-8138.2000.tb02204.x.
  • Larsen JM. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology. 2017;151(4):363–374. doi:10.1111/imm.12760.
  • Sharma G, Garg N, Hasan S, Shirodkar S. Prevotella: an insight into its characteristics and associated virulence factors. Microb Pathog. 2022;169:105673. doi:10.1016/j.micpath.2022.105673.
  • Dejea CM, Wick EC, Hechenbleikner EM, White JR, Mark Welch JL, Rossetti BJ, Peterson SN, Snesrud EC, Borisy GG, Lazarev M, et al. Microbiota organization is a distinct feature of proximal colorectal cancers. Proc Nat Acad Sci 2014; 111:18321–18326.
  • de Martel C, Georges D, Bray F, Ferlay J, Clifford GM. Global burden of cancer attributable to infections in 2018: a worldwide incidence analysis. Lancet Global Health. 2020;8:e180–90. doi:10.1016/S2214-109X(19)30488-7.
  • Bhatt AP, Redinbo MR, Bultman SJ. The role of the microbiome in cancer development and therapy. CA Cancer J Clin. 2017;67(4):326–344. doi:10.3322/caac.21398.
  • Ellermann M, Sartor RB. Intestinal bacterial biofilms modulate mucosal immune responses. J Immunol Sci. 2018;2:13–18. doi:10.29245/2578-3009/2018/2.1122.
  • Otto M. Staphylococcus epidermidis–the “accidental” pathogen. Nat Rev Microbiol. 2009;7:555–567. doi:10.1038/nrmicro2182.
  • Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, Clancy TE, Chung DC, Lochhead P, Hold GL, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14(2):207–215. doi:10.1016/j.chom.2013.07.007.
  • Ciernikova S, Sevcikova A, Stevurkova V, Mego M. Tumor microbiome - an integral part of the tumor microenvironment. Front Oncol. 2022;12:1063100. doi:10.3389/fonc.2022.1063100.
  • Cullin N, Azevedo Antunes C, Straussman R, Stein-Thoeringer CK, Elinav E. Microbiome and cancer. Cancer Cell. 2021;39(10):1317–1341. doi:10.1016/j.ccell.2021.08.006.
  • Upadhyay A, Pal D, Kumar A. Substantial relation between the bacterial biofilm and oncogenesis progression in host. Microb Pathog. 2023;175:105966. doi:10.1016/j.micpath.2022.105966.
  • Johansson MEV, Larsson JMH, Hansson GC. The two mucus layers of colon are organized by the MUC2 mucin, whereas the outer layer is a legislator of host–microbial interactions. Proc Natl Acad Sci USA. 2011;108(supplement_1):4659–4665. doi:10.1073/pnas.1006451107.
  • Johansson MEV, Gustafsson JK, Holmén-Larsson J, Jabbar KS, Xia L, Xu H, Ghishan FK, Carvalho FA, Gewirtz AT, Sjövall H, et al. Bacteria penetrate the normally impenetrable inner colon mucus layer in both murine colitis models and patients with ulcerative colitis. Gut. 2014;63(2):281–291. doi:10.1136/gutjnl-2012-303207.
  • Mirzaei R, Sabokroo N, Ahmadyousefi Y, Motamedi H, Karampoor S. Immunometabolism in biofilm infection: lessons from cancer. Mol Med. 2022;28(1):10. doi:10.1186/s10020-022-00435-2.
  • Römling U, Balsalobre C. Biofilm infections, their resilience to therapy and innovative treatment strategies. J Intern Med. 2012;272(6):541–561. doi:10.1111/joim.12004.
  • Ferlay J, Colombet M, Soerjomataram I, Parkin DM, Piñeros M, Znaor A, Bray F. Cancer statistics for the year 2020: an overview. Int J Cancer. 2021;149(4):778–789. doi:10.1002/ijc.33588.
  • Brenner H, Chen C. The colorectal cancer epidemic: challenges and opportunities for primary, secondary and tertiary prevention. Br J Cancer. 2018;119(7):785–792. doi:10.1038/s41416-018-0264-x.
  • Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal cancer. Lancet. 2019;394(10207):1467–1480. doi:10.1016/S0140-6736(19)32319-0.
  • Triantafillidis JK, Nasioulas G, Kosmidis PA. Colorectal cancer and inflammatory bowel disease: epidemiology, risk factors, mechanisms of carcinogenesis and prevention strategies. Anticancer Res. 2009;29:2727–2737.
  • den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud D-J, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res. 2013;54(9):2325–2340. doi:10.1194/jlr.R036012.
  • Natividad JMM, Verdu EF. Modulation of intestinal barrier by intestinal microbiota: pathological and therapeutic implications. Pharmacol Res. 2013;1:42–51. doi:10.1016/j.phrs.2012.10.007.
  • Bäumler AJ, Sperandio V. Interactions between the microbiota and pathogenic bacteria in the gut. Nature. 2016;535(7610):85–93. doi:10.1038/nature18849.
  • Gensollen T, Iyer SS, Kasper DL, Blumberg RS. How colonization by microbiota in early life shapes the immune system. Science. 2016;352(6285):539–544. doi:10.1126/science.aad9378.
  • Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo JA. The microbiome, cancer, and cancer therapy. Nat Med. 2019;25(3):377–388. doi:10.1038/s41591-019-0377-7.
  • Li S, Liu J, Zheng X, Ren L, Yang Y, Li W, Fu W, Wang J, Du G. Tumorigenic bacteria in colorectal cancer: mechanisms and treatments. Cancer Biol Med. 2022;19:147–162.
  • White MT, Sears CL. The microbial landscape of colorectal cancer. Nat Rev Microbiol [Internet]. 2023. [accessed 2023 Dec 6]. https://www.nature.com/articles/s41579-023-00973-4.
  • Hernández-Luna MA, López-Briones S, Luria-Pérez R. The four horsemen in colon cancer. J Oncol. 2019;2019:1–12. doi:10.1155/2019/5636272.
  • Dalal N, Jalandra R, Bayal N, Yadav AK, Harshulika N, Sharma M, Makharia GK, Kumar P, Singh R, Solanki PR, et al. Gut microbiota-derived metabolites in CRC progression and causation. J Cancer Res Clin Oncol. 2021;147:3141–3155. doi:10.1007/s00432-021-03729-w.
  • Jalandra R, Dalal N, Yadav AK, Verma D, Sharma M, Singh R, Khosla A, Kumar A, Solanki PR. Emerging role of trimethylamine-N-oxide (TMAO) in colorectal cancer. Appl Microbiol Biotechnol. 2021;105(20):7651–7660. doi:10.1007/s00253-021-11582-7.
  • Silva CL, Olival A, Perestrelo R, Silva P, Tomás H, Câmara JS. Untargeted Urinary 1H NMR-Based metabolomic pattern as a potential platform in breast cancer detection. Metabolites. 2019;9(11):269. doi:10.3390/metabo9110269.
  • Bae S, Ulrich CM, Neuhouser ML, Malysheva O, Bailey LB, Xiao L, Brown EC, Cushing-Haugen KL, Zheng Y, Cheng T-Y, et al. Plasma choline metabolites and colorectal cancer risk in the women’s health initiative observational study. Cancer Res. 2014;74(24):7442–7452. doi:10.1158/0008-5472.CAN-14-1835.
  • Janeiro MH, Ramírez MJ, Milagro FI, Martínez JA, Solas M. Implication of trimethylamine N-Oxide (TMAO) in disease: potential biomarker or new therapeutic target. Nutrients. 2018;10(10):1398. doi:10.3390/nu10101398.
  • Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Org E, Gu X, Huang Y, Zamanian-Daryoush M, Culley MK, et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell. 2015;163(7):1585–1595. doi:10.1016/j.cell.2015.11.055.
  • Nougayrède J-P, Homburg S, Taieb F, Boury M, Brzuszkiewicz E, Gottschalk G, Buchrieser C, Hacker J, Dobrindt U, Oswald E. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science. 2006;313(5788):848–851. doi:10.1126/science.1127059.
  • Swidsinski A, Khilkin M, Kerjaschki D, Schreiber S, Ortner M, Weber J, Lochs H. Association between intraepithelial Escherichia coli and colorectal cancer. Gastroenterology. 1998;115(2):281–286. doi:10.1016/S0016-5085(98)70194-5.
  • Boleij A, Hechenbleikner EM, Goodwin AC, Badani R, Stein EM, Lazarev MG, Ellis B, Carroll KC, Albesiano E, Wick EC, et al. The bacteroides fragilis toxin gene is prevalent in the colon mucosa of colorectal cancer patients. Clin Infect Dis. 2015;60(2):208–215. doi:10.1093/cid/ciu787.
  • Chung L, Thiele Orberg E, Geis AL, Chan JL, Fu K, DeStefano Shields CE, Dejea CM, Fathi P, Chen J, Finard BB, et al. Bacteroides fragilis toxin coordinates a pro-carcinogenic inflammatory cascade via targeting of colonic epithelial cells. Cell Host Microbe. 2018;23(2):203–214.e5. doi:10.1016/j.chom.2018.01.007.
  • Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, Barnes R, Watson P, Allen-Vercoe E, Moore RA, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012;22(2):299–306. doi:10.1101/gr.126516.111.
  • Wirbel J, Pyl PT, Kartal E, Zych K, Kashani A, Milanese A, Fleck JS, Voigt AY, Palleja A, Ponnudurai R, et al. Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat Med. 2019;25(4):679–689. doi:10.1038/s41591-019-0406-6.
  • Abdulamir AS, Hafidh RR, Bakar FA. The association of Streptococcus bovis/gallolyticus with colorectal tumors: the nature and the underlying mechanisms of its etiological role. J Exp Clin Cancer Res. 2011;30(1):11. doi:10.1186/1756-9966-30-11.
  • Aymeric L, Donnadieu F, Mulet C, du Merle L, Nigro G, Saffarian A, Bérard M, Poyart C, Robine S, Regnault B, et al. Colorectal cancer specific conditions promote streptococcus gallolyticus gut colonization. Proc Natl Acad Sci U S A. 2018;115(2):E283–91. doi:10.1073/pnas.1715112115.
  • Lu R, Wu S, Zhang Y, Xia Y, Zhou Z, Kato I, Dong H, Bissonnette M, Sun J. Salmonella protein AvrA activates the STAT3 signaling pathway in colon cancer. Neoplasia (New York, NY). 2016;18(5):307. doi:10.1016/j.neo.2016.04.001.
  • Mughini-Gras L, Schaapveld M, Kramers J, Mooij S, Neefjes-Borst EA, van PW, Neefjes J. Increased colon cancer risk after severe Salmonella infection. PLOS ONE. [Internet]. 2018 [accessed 2023 Dec 8];13(1):e0189721. doi:10.1371/journal.pone.0189721.
  • Pleguezuelos-Manzano C, Puschhof J, Rosendahl Huber A, van Hoeck A, Wood HM, Nomburg J, Gurjao C, Manders F, Dalmasso G, Stege PB, et al. Mutational signature in colorectal cancer caused by genotoxic pks+ E. coli. Nature. 2020;580(7802):269–273. doi:10.1038/s41586-020-2080-8.
  • Hall-Stoodley L, Costerton JW, Stoodley P. Bacterial biofilms: from the natural environment to infectious diseases. Nat Rev Microbiol. 2004;2(2):95–108. doi:10.1038/nrmicro821.
  • Tomkovich S, Dejea CM, Winglee K, Drewes JL, Chung L, Housseau F, Pope JL, Gauthier J, Sun X, Mühlbauer M, et al. Human colon mucosal biofilms from healthy or colon cancer hosts are carcinogenic. J Clin Invest. 2019;129(4):1699–1712. doi:10.1172/JCI124196.
  • Tjalsma H, Boleij A, Marchesi JR, Dutilh BE. A bacterial driver–passenger model for colorectal cancer: beyond the usual suspects. Nat Rev Microbiol. 2012;10(8):575–582. doi:10.1038/nrmicro2819.
  • Johnson CH, Dejea CM, Edler D, Hoang LT, Santidrian AF, Felding BH, Ivanisevic J, Cho K, Wick EC, Hechenbleikner EM, et al. Metabolism links bacterial biofilms and colon carcinogenesis. Cell Metab. 2015;21(6):891–897. doi:10.1016/j.cmet.2015.04.011.
  • Soler AP. Increased tight junctional permeability is associated with the development of colon cancer. Carcinogenesis. 1999;20(8):1425–1432. doi:10.1093/carcin/20.8.1425.
  • Tremblay W, Mompart F, Lopez E, Quaranta M, Bergoglio V, Hashim S, Bonnet D, Alric L, Mas E, Trouche D, et al. Cytolethal distending toxin promotes replicative stress leading to genetic instability transmitted to daughter cells. Front Cell Dev Biol. 2021;9:656795. doi:10.3389/fcell.2021.656795.
  • Cuevas-Ramos G, Petit CR, Marcq I, Boury M, Oswald E, Nougayrède J-P. Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proc Natl Acad Sci USA. 2010;107(25):11537–11542. doi:10.1073/pnas.1001261107.
  • Goodwin AC, Destefano Shields CE, Wu S, Huso DL, Wu X, Murray-Stewart TR, Hacker-Prietz A, Rabizadeh S, Woster PM, Sears CL, et al. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc Natl Acad Sci USA. 2011;108(37):15354–15359. doi:10.1073/pnas.1010203108.
  • Clay SL, Fonseca-Pereira D, Garrett WS. Colorectal cancer: the facts in the case of the microbiota. J Clin Invest. [Internet] 2022;132(4). [accessed 2023 Jul 18]. doi:10.1172/JCI155101.
  • Ternes D, Tsenkova M, Pozdeev VI, Meyers M, Koncina E, Atatri S, Schmitz M, Karta J, Schmoetten M, Heinken A, et al. The gut microbial metabolite formate exacerbates colorectal cancer progression. Nat Metab. 2022;4:458–475. doi:10.1038/s42255-022-00558-0.
  • Zheng D-W, Dong X, Pan P, Chen K-W, Fan J-X, Cheng S-X, Zhang X-Z. Phage-guided modulation of the gut microbiota of mouse models of colorectal cancer augments their responses to chemotherapy. Nat Biomed Eng. 2019;3(9):717–728. doi:10.1038/s41551-019-0423-2.
  • 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(2):195–206. doi:10.1016/j.chom.2013.07.012.
  • Brown DG, Rao S, Weir TL, O’Malia J, Bazan M, Brown RJ, Ryan EP. Metabolomics and metabolic pathway networks from human colorectal cancers, adjacent mucosa, and stool. Cancer Metab. 2016;4(1):11. doi:10.1186/s40170-016-0151-y.
  • Lucas C, Barnich N, Nguyen NH. Microbiota, inflammation and colorectal cancer. Int J Mol Sci. 2017;18(6):1310. doi:10.3390/ijms18061310.
  • Binda C, Gibiino G, Coluccio C, Sbrancia M, Dajti E, Sinagra E, Capurso G, Sambri V, Cucchetti A, Ercolani G, et al. Biliary diseases from the microbiome perspective: how microorganisms could change the approach to benign and malignant diseases. Microorganisms. 2022;10(2):312. doi:10.3390/microorganisms10020312.
  • Crawford RW, Gibson DL, Kay WW, Gunn JS. Identification of a bile-induced exopolysaccharide required for Salmonella biofilm formation on gallstone surfaces. Infect Immun. 2008;76(11):5341–5349. doi:10.1128/IAI.00786-08.
  • Hall-Stoodley L, Stoodley P. Evolving concepts in biofilm infections. Cell Microbiol. 2009;11(7):1034–1043. doi:10.1111/j.1462-5822.2009.01323.x.
  • Prouty AM, Schwesinger WH, Gunn JS. Biofilm formation and interaction with the surfaces of gallstones by Salmonella spp. Infect Immun. 2002;70(5):2640–2649. doi:10.1128/IAI.70.5.2640-2649.2002.
  • Nath G, Gulati AK, Shukla VK. Role of bacteria in carcinogenesis, with special reference to carcinoma of the gallbladder. World J Gastroenterol. 2010;16:5395–5404. doi:10.3748/wjg.v16.i43.5395.
  • Fowler CC, Chang S-J, Gao X, Geiger T, Stack G, Galán JE. Emerging insights into the biology of typhoid toxin. Curr Opin Microbiol. 2017;35:70–77. doi:10.1016/j.mib.2017.01.012.
  • Pickett CL, Whitehouse CA. The cytolethal distending toxin family. Trends Microbiol. 1999;7(7):292–297. doi:10.1016/S0966-842X(99)01537-1.
  • Scuron MD, Boesze-Battaglia K, Dlakić M, Shenker BJ. The cytolethal distending toxin contributes to microbial virulence and disease pathogenesis by acting as a tri-perditious toxin. Front Cell Infect Microbiol. 2016;6:168. doi:10.3389/fcimb.2016.00168.
  • Thelestam M, Frisan T. Cytolethal distending toxins. Rev Physiol Biochem Pharmacol. 2004;152:111–133.
  • Di Domenico EG, Cavallo I, Pontone M, Toma L, Ensoli F. Biofilm producing Salmonella typhi: chronic colonization and development of gallbladder cancer. IJMS. 2017;18(9):1887. doi:10.3390/ijms18091887.
  • Dutta U, Garg PK, Kumar R, Tandon RK. Typhoid carriers among patients with gallstones are at increased risk for carcinoma of the gallbladder. Am J Gastroenterol. 2000;95(3):784–787. doi:10.1111/j.1572-0241.2000.01860.x.
  • Kumar S, Kumar S, Kumar S. Infection as a risk factor for gallbladder cancer. J Surg Oncol. 2006;93(8):633–639. doi:10.1002/jso.20530.
  • Upadhayay A, Pal D, Kumar A. Salmonella typhi induced oncogenesis in gallbladder cancer: co-relation and progression. Adv Cancer Biol Metastasis. 2022;4:100032. doi:10.1016/j.adcanc.2022.100032.
  • Gagnaire A, Nadel B, Raoult D, Neefjes J, Gorvel J-P. Collateral damage: insights into bacterial mechanisms that predispose host cells to cancer. Nat Rev Microbiol. 2017;15(2):109–128. doi:10.1038/nrmicro.2016.171.
  • Johnson R, Ravenhall M, Pickard D, Dougan G, Byrne A, Frankel G, Payne SM. Comparison of Salmonella enterica serovars typhi and typhimurium reveals typhoidal serovar-specific responses to bile. Infect Immun. 2018;86(3):e00490–17. doi:10.1128/IAI.00490-17.
  • Chen L, Gu L, Geng X, Xu G, Huang X, Zhu X. A novel cis antisense RNA AsfD promotes Salmonella enterica serovar typhi motility and biofilm formation. Microb Pathog. 2020;142:104044. doi:10.1016/j.micpath.2020.104044.
  • Zhao H, Li Y. Cancer metabolism and intervention therapy. Mol Biomed. 2021;2(1):5. doi:10.1186/s43556-020-00012-1.
  • Sheng X, Wang W, Chen L, Zhang H, Zhang Y, Xu S, Xu H, Huang X. Mig-14 may contribute to Salmonella enterica serovar typhi resistance to polymyxin B by decreasing the permeability of the outer-membrane and promoting the formation of biofilm. Int J Med Microbiol. 2019;309(2):143–150. doi:10.1016/j.ijmm.2019.01.001.
  • Walawalkar YD, Vaidya Y, Nayak V, Monack D. Response of Salmonella typhi to bile-generated oxidative stress: implication of quorum sensing and persister cell populations. Pathog Dis. 2016;74(8):ftw090. doi:10.1093/femspd/ftw090.
  • Jesudhasan PR, Cepeda ML, Widmer K, Dowd SE, Soni KA, Hume ME, Zhu J, Pillai SD. Transcriptome analysis of genes controlled by luxS/autoinducer-2 in Salmonella enterica serovar Typhimurium. Foodborne Pathog Dis. 2010;7(4):399–410. doi:10.1089/fpd.2009.0372.
  • Cho I, Blaser MJ. The human microbiome: at the interface of health and disease. Nat Rev Genet. 2012;13(4):260–270. doi:10.1038/nrg3182.
  • Costello EK, Stagaman K, Dethlefsen L, Bohannan BJM, Relman DA. The application of ecological theory toward an understanding of the human microbiome. Science. 2012;336(6086):1255–1262. doi:10.1126/science.1224203.
  • Bebek G, Bennett KL, Funchain P, Campbell R, Seth R, Scharpf J, Burkey B, Eng C. Microbiomic subprofiles and MDR1 promoter methylation in head and neck squamous cell carcinoma. Hum Mol Genet [Internet]. 2012;21: 1557–1565. doi:10.1093/hmg/ddr593.
  • Frank DN, Qiu Y, Cao Y, Zhang S, Lu L, Kofonow JM, Robertson CE, Liu Y, Wang H, Levens CL, et al. A dysbiotic microbiome promotes head and neck squamous cell carcinoma. Oncogene. 2022;41(9):1269–1280. doi:10.1038/s41388-021-02137-1.
  • Marttila E, Uittamo J, Rusanen P, Lindqvist C, Salaspuro M, Rautemaa R. Acetaldehyde production and microbial colonization in oral squamous cell carcinoma and oral lichenoid disease. Oral Surg Oral Med Oral Pathol Oral Radiol. 2013;116(1):61–68. doi:10.1016/j.oooo.2013.02.009.
  • Raisch J, Buc E, Bonnet M, Sauvanet P, Vazeille E, de Vallée A, Déchelotte P, Darcha C, Pezet D, Bonnet R, et al. Colon cancer-associated B2 Escherichia coli colonize gut mucosa and promote cell proliferation. World J Gastroenterol. 2014;20:6560–6572. doi:10.3748/wjg.v20.i21.6560.
  • Cantero D, Cooksley C, Bassiouni A, Tran HB, Roscioli E, Wormald P-J, Vreugde S. Staphylococcus aureus biofilms induce apoptosis and expression of Interferon-γ, Interleukin-10, and Interleukin-17A on Human Sinonasal Explants. Am J Rhinol? Allergy. 2015;29(1):23–28. doi:10.2500/ajra.2015.29.4130.
  • Kim J, Lee HK. Potential role of the gut microbiome in colorectal cancer progression. Front Immunol. 2022;12:807648. doi:10.3389/fimmu.2021.807648.
  • Curtis MA, Diaz PI, Van Dyke TE. The role of the microbiota in periodontal disease. Periodontol 2000. 2020;83:14–25. doi:10.1111/prd.12296.
  • Hajishengallis G, Lambris JD. Complement and dysbiosis in periodontal disease. Immunobiology. 2012;217(11):1111–1116. doi:10.1016/j.imbio.2012.07.007.
  • Yost S, Stashenko P, Choi Y, Kukuruzinska M, Genco CA, Salama A, Weinberg EO, Kramer CD, Frias-Lopez J. Increased virulence of the oral microbiome in oral squamous cell carcinoma revealed by metatranscriptome analyses. Int J Oral Sci. 2018;10(4):32. doi:10.1038/s41368-018-0037-7.
  • Kolenbrander PE, Andersen RN, Blehert DS, Egland PG, Foster JS, Palmer RJ. Communication among oral bacteria. Microbiol Mol Biol Rev. 2002;66(3):486–505. doi:10.1128/MMBR.66.3.486-505.2002.
  • Brennan CA, Garrett WS. Fusobacterium nucleatum — symbiont, opportunist and oncobacterium. Nat Rev Microbiol. 2019;17(3):156–166. doi:10.1038/s41579-018-0129-6.
  • Nagy KN, Sonkodi I, Szöke I, Nagy E, Newman HN. The microflora associated with human oral carcinomas. Oral Oncol. 1998;34(4):304–308. doi:10.1016/S1368-8375(98)80012-2.
  • Zhang L, Liu Y, Zheng HJ, Zhang CP. The oral microbiota may have influence on oral cancer. Front Cell Infect Microbiol. 2020;9:476. doi:10.3389/fcimb.2019.00476.
  • Al-Hebshi NN, Nasher AT, Maryoud MY, Homeida HE, Chen T, Idris AM, Johnson NW. Inflammatory bacteriome featuring Fusobacterium nucleatum and Pseudomonas aeruginosa identified in association with oral squamous cell carcinoma. Sci Rep. 2017;7(1):1834. doi:10.1038/s41598-017-02079-3.
  • Zhao H, Chu M, Huang Z, Yang X, Ran S, Hu B, Zhang C, Liang J. Variations in oral microbiota associated with oral cancer. Sci Rep. 2017;7(1):11773. doi:10.1038/s41598-017-11779-9.
  • Gallimidi AB, Fischman S, Revach B, Bulvik R, Maliutina A, Rubinstein AM, Nussbaum G, Elkin M. Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral-specific chemical carcinogenesis model. Oncotarget. 2015;6(26):22613–22623. doi:10.18632/oncotarget.4209.
  • Fardini Y, Wang X, Témoin S, Nithianantham S, Lee D, Shoham M, Han YW. Fusobacterium nucleatum adhesin FadA binds vascular-endothelial cadherin and alters endothelial integrity. Mol Microbiol. 2011;82:1468–1480. doi:10.1111/j.1365-2958.2011.07905.x.
  • Bosetti C, Carioli G, Santucci C, Bertuccio P, Gallus S, Garavello W, Negri E, La Vecchia C. Global trends in oral and pharyngeal cancer incidence and mortality. Int J Cancer. 2020;147(4):1040–1049. doi:10.1002/ijc.32871.
  • Monti E, Barbara G, Libutti G, Boero V, Parazzini F, Ciavattini A, Bogani G, Pignataro L, Magni B, Merli C, et al. A clinician’s dilemma: what should be communicated to women with oncogenic genital HPV and their partners regarding the risk of oral viral transmission? BMC Women’s Health. 2022:22. doi:10.1186/s12905-022-01965-x
  • Andrews N, Griffiths C. Dental complications of head and neck radiotherapy: part 1. Aust Dent J. 2001;46(2):88–94. doi:10.1111/j.1834-7819.2001.tb00562.x.
  • Støre G, Eribe ERK, Olsen I. DNA–DNA hybridization demonstrates multiple bacteria in osteoradionecrosis. Int J Oral Max Surg. 2005;34(2):193–196. doi:10.1016/j.ijom.2004.06.010.
  • Curi MM, Dib LL, Kowalski LP, Landman G, Mangini C. Opportunistic actinomycosis in osteoradionecrosis of the jaws in patients affected by head and neck cancer: incidence and clinical significance. Oral Oncol. 2000;36(3):294–299. doi:10.1016/S1368-8375(99)00080-9.
  • Aas JA, Reime L, Pedersen K, Eribe ERK, Abesha-Belay E, Støre G, Olsen I. Osteoradionecrosis contains a wide variety of cultivable and non-cultivable bacteria. J Oral Microbiol. 2010;2:5072. doi:10.3402/jom.v2i0.5072.
  • Li Z, Fu R, Huang X, Wen X, Zhang L. Oral microbiota may affect osteoradionecrosis following radiotherapy for head and neck cancer. J Transl Med. 2023;21(1):391. doi:10.1186/s12967-023-04219-y.
  • Tsai Y, Lin Y, Wu W, Chiu P, Lin BJ, Hao S. Biofilm formations in nasopharyngeal tissues of patients with nasopharyngeal osteoradionecrosis. Otolaryngol–Head Neck Surg. 2013;148:633–636. doi:10.1177/0194599812474971.
  • Conlon BP, Nakayasu ES, Fleck LE, MD L, Isabella VM, Coleman K, Leonard SN, Smith RD, Adkins JN, Lewis K. Activated ClpP kills persisters and eradicates a chronic biofilm infection. Nature. 2013;503:365–370. doi:10.1038/nature12790.
  • Otto M. Staphylococcal infections: mechanisms of biofilm maturation and detachment as critical determinants of pathogenicity. Annu Rev Med. 2013;64(1):175–188. doi:10.1146/annurev-med-042711-140023.
  • Utsui Y, Yokota T. Role of an altered penicillin-binding protein in methicillin- and cephem-resistant Staphylococcus aureus. Antimicrob Agents Chemother. 1985;28(3):397–403. doi:10.1128/AAC.28.3.397.
  • Jiang R, Liu Y, Zhang H, Chen Y, Liu T, Zeng J, Nie E, Chen S, Tan J. Distinctive microbiota of delayed healing of oral mucositis after radiotherapy of nasopharyngeal carcinoma. Front Cell Infect Microbiol. 2022;12:1070322. doi:10.3389/fcimb.2022.1070322.
  • Gong H, Shi Y, Zhou X, Wu C, Cao P, Xu C, Hou D, Wang Y, Zhou L, Drake HL. Microbiota in the throat and risk factors for laryngeal carcinoma. Appl Environ Microbiol. 2014;80(23):7356–7363. doi:10.1128/AEM.02329-14.
  • Alberg AJ, Brock MV, Samet JM. Epidemiology of lung cancer: looking to the future. JCO. 2005;23(14):3175–3185. doi:10.1200/JCO.2005.10.462.
  • Engels EA. Inflammation in the development of lung cancer: epidemiological evidence. Expert Rev Anticancer Ther. 2008;8(4):605–615. doi:10.1586/14737140.8.4.605.
  • Govindan R. Lung cancer in never smokers: a new hot area of research. Lancet Oncol. 2010;11(4):304–305. doi:10.1016/S1470-2045(10)70057-7.
  • Huang S. Mycoplasma infections and different human carcinomas. WJG. 2001;7:266. doi:10.3748/wjg.v7.i2.266.
  • Ushio S, Iwaki K, Taniai M, Ohta T, Fukuda S, Sugimura K, Kurimoto M. Metastasis-promoting activity of a novel molecule, Ag 243-5, derived from mycoplasma, and the complete nucleotide sequence. Microbiol Immunol. 1995;39(6):393–400. doi:10.1111/j.1348-0421.1995.tb02218.x.
  • Liu N-N, Ma Q, Ge Y, Yi C-X, Wei L-Q, Tan J-C, Chu Q, Li J-Q, Zhang P, Wang H. Microbiome dysbiosis in lung cancer: from composition to therapy. NPJ Precis Onc. 2020;4:33. doi:10.1038/s41698-020-00138-z.
  • Hwang SY, Kim JY, Lee HS, Lee S, Kim D, Kim S, Hyun JH, Shin JI, Lee KH, Han SH, et al. Pulmonary tuberculosis and risk of lung cancer: a systematic review and meta-analysis. J Clin Med. 2022;11(3):765. doi:10.3390/jcm11030765.
  • Chaturvedi AK, Gaydos CA, Agreda P, Holden JP, Chatterjee N, Goedert JJ, Caporaso NE, Engels EA. Chlamydia pneumoniae infection and risk for lung cancer. Cancer Epidem Biomar. 2010;19(6):1498–1505. doi:10.1158/1055-9965.EPI-09-1261.
  • Littman AJ, Jackson LA, Vaughan TL. Chlamydia pneumoniae and lung cancer: epidemiologic evidence. Cancer Epidem Biomar. 2005;14(4):773–778. doi:10.1158/1055-9965.EPI-04-0599.
  • Zhan P, Suo L, Qian Q, Shen X, Qiu L-X, Yu L, Song Y. Chlamydia pneumoniae infection and lung cancer risk: A meta-analysis. Eur J Cancer. 2011;47(5):742–747. doi:10.1016/j.ejca.2010.11.003.
  • Dancewicz M, Szymankiewicz M, Bella M, Swiniarska J, Kowalewski J. Bronchial bacterial colonization in patients with lung cancer. Pneumonol Alergol Pol. 2009;77(3):242–247. doi:10.5603/ARM.27802.
  • Szymankiewicz M, Kowalewski J, Dancewicz M. [Bacteriological and mycological analysis of material taken from lower respiratory tract in patients with malignancy]. Pol Merkur Lekarski. 2006;21:218–222.
  • Akshatha CR, Bhat S, Sindhu R, Shashank D, Rose Sommano S, Tapingkae W, Cheewangkoon R, Prasad SK. Current therapeutic options for gastric adenocarcinoma. Saudi J Biol Sci. 2021;28(9):5371–5378. doi:10.1016/j.sjbs.2021.05.060.
  • Shin WS, Xie F, Chen B, Yu P, Yu J, To KF, Kang W. Updated epidemiology of gastric cancer in Asia: decreased incidence but still a big challenge. Cancers Basel. 2023;15(9):2639. doi:10.3390/cancers15092639.
  • Sekiguchi M, Oda I, Matsuda T, Saito Y. Epidemiological trends and future perspectives of gastric cancer in Eastern Asia. Digestion. 2022;103(1):22–28. doi:10.1159/000518483.
  • Koo H, Allan RN, Howlin RP, Stoodley P, Hall-Stoodley L. Targeting microbial biofilms: current and prospective therapeutic strategies. Nat Rev Microbiol. 2017;15(12):740–755. doi:10.1038/nrmicro.2017.99.
  • Pucułek M, Machlowska J, Wierzbicki R, Baj J, Maciejewski R, Sitarz R. Helicobacter pylori associated factors in the development of gastric cancer with special reference to the early-onset subtype. Oncotarget. 2018;9(57):31146–31162. doi:10.18632/oncotarget.25757.
  • Rawla P, Barsouk A. Epidemiology of gastric cancer: global trends, risk factors and prevention. Prz Gastroenterol. 2019;14(1):26–38. doi:10.5114/pg.2018.80001.
  • Carron MA, Tran VR, Sugawa C, Coticchia JM. Identification of Helicobacter pylori biofilms in human gastric mucosa. J Gastrointest Surg. 2006;10(5):712–717. doi:10.1016/j.gassur.2005.10.019.
  • Hsieh Y-Y, Tung S-Y, Pan H-Y, Yen C-W, Xu H-W, Lin Y-J, Deng Y-F, Hsu W-T, Wu C-S, Li C. Increased abundance of Clostridium and Fusobacterium in gastric microbiota of patients with gastric cancer in Taiwan. Sci Rep. 2018;8(1):158. doi:10.1038/s41598-017-18596-0.
  • Lehr K, Nikitina D, Vilchez-Vargas R, Steponaitiene R, Thon C, Skieceviciene J, Schanze D, Zenker M, Malfertheiner P, Kupcinskas J, et al. Microbial composition of tumorous and adjacent gastric tissue is associated with prognosis of gastric cancer. Sci Rep. 2023;13(1):4640. doi:10.1038/s41598-023-31740-3.
  • Abadi ATB. Strategies used by helicobacter pylori to establish persistent infection. World J Gastroenterol. 2017;23(16):2870–2882. doi:10.3748/wjg.v23.i16.2870.
  • Cellini L, Grande R, Di Campli E, Traini T, Di Giulio M, Nicola Lannutti S, Lattanzio R. Dynamic colonization of Helicobacter pylori in human gastric mucosa. Scand J Gastroenterol. 2008;43(2):178–185. doi:10.1080/00365520701675965.
  • Coticchia J, Sugawa C, Tran V, Gurrola J, Kowalski E, Carron M. Presence and density of helicobacter pylori biofilms in human gastric mucosa in patients with peptic ulcer disease. J Gastrointest Surg. 2006;10(6):883–889. doi:10.1016/j.gassur.2005.12.009.
  • Chan W-Y, Hui P-K, Leung K-M, Chow J, Kwok F, Ng C-S. Coccoid forms of Helicobacter pylori in the human stomach. Am J Clin Pathol. 1994;102(4):503–507. doi:10.1093/ajcp/102.4.503.
  • Zou Q, Zhang H, Meng F, He L, Zhang J, Xiao D, Chammas R. Proteomic and transcriptomic studies of BGC823 cells stimulated with Helicobacter pylori isolates from gastric MALT lymphoma. PLOS ONE. 2020;15(9):e0238379. doi:10.1371/journal.pone.0238379.
  • Valenzuela MA, Canales J, Corvalán AH, Quest AF. Helicobacter pylori-induced inflammation and epigenetic changes during gastric carcinogenesis. World J Gastroenterol. 2015;21:12742–12756. doi:10.3748/wjg.v21.i45.12742.
  • Huang F-Y, Chan A-O, Rashid A, Wong D-H, Cho C-H, Yuen M-F. Helicobacter pylori induces promoter methylation of E-cadherin via interleukin-1β activation of nitric oxide production in gastric cancer cells. Cancer. 2012;118:4969–4980. doi:10.1002/cncr.27519.
  • Perri F, Cotugno R, Piepoli A, Merla A, Quitadamo M, Gentile A, Pilotto A, Annese V, Andriulli A. Aberrant DNA methylation in non-neoplastic gastric mucosa of H. Pylori infected patients and effect of eradication. Am J Gastroenterol. 2007;102(7):1361–1371. doi:10.1111/j.1572-0241.2007.01284.x.
  • Brabletz T, Kalluri R, Nieto MA, Weinberg RA. EMT in cancer. Nat Rev Cancer. 2018;18(2):128–134. doi:10.1038/nrc.2017.118.
  • Sougleri IS, Papadakos KS, Zadik MP, Mavri-Vavagianni M, Mentis AF, Sgouras DN. Helicobacter pylori CagA protein induces factors involved in the epithelial to mesenchymal transition (EMT) in infected gastric epithelial cells in an EPIYA- phosphorylation-dependent manner. FEBS J. 2016;283(2):206–220. doi:10.1111/febs.13592.
  • Kay J, Thadhani E, Samson L, Engelward B. Inflammation-induced DNA damage, mutations and cancer. DNA Repair (Amst). 2019;83:102673. doi:10.1016/j.dnarep.2019.102673.
  • Perwez Hussain S, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007;121(11):2373–2380. doi:10.1002/ijc.23173.
  • Meng Z, Yang T. Liu DType-2 epithelial-mesenchymal transition in oral mucosal nonneoplastic diseases. Front Immunol [Internet]2022;13. doi:10.3389/fimmu.2022.1020768
  • Vergara D, Simeone P, Damato M, Maffia M, Lanuti P, Trerotola M. The cancer microbiota: EMT and inflammation as shared molecular mechanisms associated with plasticity and progression. J Oncol. 2019;2019:e1253727. doi:10.1155/2019/1253727.
  • Reddy KB. MicroRNA (miRNA) in cancer. Cancer Cell Int. 2015;15(1):38. doi:10.1186/s12935-015-0185-1.
  • Tomkovich S, Gharaibeh RZ, Dejea CM, Pope JL, Jiang J, Winglee K, Gauthier J, Newsome RC, Yang Y, Fodor AA, et al. Human colon mucosal biofilms and murine host communicate via altered mRNA and microRNA expression during cancer. MSystems. 2020;5:e00451–19. doi:10.1128/mSystems.00451-19.
  • Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67(1):7–30. doi:10.3322/caac.21387.
  • Vincent A, Hong S-M, Hu C, Omura N, Young A, Kim H, Yu J, Knight S, Ayars M, Griffith M, et al. Epigenetic silencing of EYA2 in pancreatic adenocarcinomas promotes tumor growth. Oncotarget. 2014;5(9):2575–2587. doi:10.18632/oncotarget.1842.
  • Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–2921. doi:10.1158/0008-5472.CAN-14-0155.
  • Mitsuhashi K, Nosho K, Sukawa Y, Matsunaga Y, Ito M, Kurihara H, Kanno S, Igarashi H, Naito T, Adachi Y, et al. Association of Fusobacterium species in pancreatic cancer tissues with molecular features and prognosis. Oncotarget. 2015;6(9):7209–7220. doi:10.18632/oncotarget.3109.
  • Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012: global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108. doi:10.3322/caac.21262.
  • Farrell JJ, Zhang L, Zhou H, Chia D, Elashoff D, Akin D, Paster BJ, Joshipura K, Wong DTW. Variations of oral microbiota are associated with pancreatic diseases including pancreatic cancer. Gut. 2012;61(4):582–588. doi:10.1136/gutjnl-2011-300784.
  • Alkharaan H, Lu L, Gabarrini G, Halimi A, Ateeb Z, Sobkowiak MJ, Davanian H, Fernández Moro C, Jansson L, Del Chiaro M, et al. Circulating and salivary antibodies to Fusobacterium nucleatum are associated with cystic pancreatic neoplasm malignancy. Front Immunol. 2020;11:2003. doi:10.3389/fimmu.2020.02003.
  • Karpiński TM. The microbiota and pancreatic cancer. Gastroenterol Clin North Am. 2019;48(3):447–464. doi:10.1016/j.gtc.2019.04.008.
  • Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, Mohan N, Aykut B, Usyk M, Torres LE. et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 2018;8(4):403–416. doi:10.1158/2159-8290.CD-17-1134.
  • Half E, Keren N, Dorfman T, Reshef L, Lachter I, Kluger Y, Konikoff F, Gphna U. P-165 specific changes in fecal microbiota may differentiate pancreatic cancer patients from healthy individuals. Ann Oncol. 2015;26:iv48. doi:10.1093/annonc/mdv233.165.
  • Half E, Keren N, Reshef L, Dorfman T, Lachter I, Kluger Y, Reshef N, Knobler H, Maor Y, Stein A, et al. Fecal microbiome signatures of pancreatic cancer patients. Sci Rep. 2019;9(1):16801. doi:10.1038/s41598-019-53041-4.
  • Ren Z, Jiang J, Xie H, Li A, Lu H, Xu S, Zhou L, Zhang H, Cui G, Chen X, et al. Gut microbial profile analysis by MiSeq sequencing of pancreatic carcinoma patients in China. Oncotarget. 2017;8(56):95176–95191. doi:10.18632/oncotarget.18820.
  • Thomas RM, Gharaibeh RZ, Gauthier J, Beveridge M, Pope JL, Guijarro MV, Yu Q, He Z, Ohland C, Newsome R, et al. Intestinal microbiota enhances pancreatic carcinogenesis in preclinical models. Carcinogenesis. 2018;39(8):1068–1078. doi:10.1093/carcin/bgy073.
  • Mendez R, Kesh K, Arora N, Di Martino L, McAllister F, Merchant N, Banerjee S, Banerjee S. Microbial dysbiosis and polyamine metabolism as predictive markers for early detection of pancreatic cancer. Carcinogenesis. 2020;41(5):561–570. doi:10.1093/carcin/bgz116.
  • Corrales L, Matson V, Flood B, Spranger S, Gajewski TF. Innate immune signaling and regulation in cancer immunotherapy. Cell Res. 2017;27(1):96–108. doi:10.1038/cr.2016.149.
  • Liu J, Xu D, Wang Q, Zheng D, Jiang X, Xu L. LPS induced miR-181a promotes pancreatic cancer cell migration via targeting PTEN and MAP2K4. Dig Dis Sci. 2014;59(7):1452–1460. doi:10.1007/s10620-014-3049-y.
  • Santoni M, Andrikou K, Sotte V, Bittoni A, Lanese A, Pellei C, Piva F, Conti A, Nabissi M, Santoni G, et al. Toll like receptors and pancreatic diseases: from a pathogenetic mechanism to a therapeutic target. Cancer Treat Rev. 2015;41(7):569–576. doi:10.1016/j.ctrv.2015.04.004.
  • Yin H, Pu N, Chen Q, Zhang J, Zhao G, Xu X, Wang D, Kuang T, Jin D, Lou W, et al. Gut-derived lipopolysaccharide remodels tumoral microenvironment and synergizes with PD-L1 checkpoint blockade via TLR4/MyD88/AKT/NF-κB pathway in pancreatic cancer. Cell Death Disease. 2021;12(11):1033. doi:10.1038/s41419-021-04293-4.
  • Leung PS, Chan YC. Role of oxidative stress in pancreatic inflammation. Antioxid Redox Signal. 2009;11:135–165. doi:10.1089/ars.2008.2109.
  • Noureldein MH, Eid AA. Gut microbiota and mTOR signaling: Insight on a new pathophysiological interaction. Microb Pathog. 2018;118:98–104. doi:10.1016/j.micpath.2018.03.021.
  • Chen Z, Zhang S, Dong S, Xu H, Zhou W. Association of the microbiota and pancreatic cancer: opportunities and limitations. Front Immunol. 2022;13:844401. doi:10.3389/fimmu.2022.844401.
  • Ma Y, Hao J. Dedifferentiation of epithelial cells incorporates immune reprogramming. Trends Cell Biol. 2021;31(4):237–240. doi:10.1016/j.tcb.2021.01.007.
  • Gnanasekaran J, Binder Gallimidi A, Saba E, Pandi K, Eli Berchoer L, Hermano E, Angabo S, Makkawi H, Khashan A, Daoud A, et al. Intracellular porphyromonas gingivalis promotes the tumorigenic behavior of pancreatic carcinoma cells. Cancers Basel. 2020;12(8):2331. doi:10.3390/cancers12082331.
  • Chen S-M, Hsu L-J, Lee H-L, Lin C-P, Huang S-W, Lai C-L, Lin C-W, Chen W-T, Chen Y-J, Lin Y-C, et al. Lactobacillus attenuate the progression of pancreatic cancer promoted by Porphyromonas gingivalis in k-rasg12d transgenic mice. Cancers Basel. 2020;12:3522. doi:10.3390/cancers12123522.
  • Jiang Y, Geng M, Bai L. Targeting biofilms therapy: current research strategies and development hurdles. Microorganisms. 2020;8(8):1222. doi:10.3390/microorganisms8081222.
  • Huang X, Pan J, Xu F, Shao B, Wang Y, Guo X, Zhou S. Bacteria‐based cancer immunotherapy. Adv Sci (Weinh). 2021;8:2003572. doi:10.1002/advs.202003572.
  • Soto Chervin C, Gajewski TF. Microbiome-based interventions: therapeutic strategies in cancer immunotherapy. Immunooncol Technol. 2020;8:12–20. doi:10.1016/j.iotech.2020.11.001.
  • Srinivasan R, Santhakumari S, Poonguzhali P, Geetha M, Dyavaiah M, Xiangmin L. Bacterial biofilm inhibition: a focused review on recent therapeutic strategies for combating the biofilm mediated infections. Front Microbiol. 2021;12:676458. doi:10.3389/fmicb.2021.676458.