Abstract
There is growing evidence that altered microbiota abundance of a range of specific anaerobic bacteria are associated with cancer, including Peptoniphilus spp., Porphyromonas spp., Fusobacterium spp., Fenollaria spp., Prevotella spp., Sneathia spp., Veillonella spp. and Anaerococcus spp. linked to multiple cancer types. In this review we explore these pathogenic associations. The mechanisms by which bacteria are known or predicted to interact with human cells are reviewed and we present an overview of the interlinked mechanisms and hypotheses of how multiple intracellular anaerobic bacterial pathogens may act together to cause host cell and tissue microenvironment changes associated with carcinogenesis and cancer cell invasion. These include combined effects on changes in cell signalling, DNA damage, cellular metabolism and immune evasion. Strategies for early detection and eradication of anaerobic cancer-associated bacterial pathogens that may prevent cancer progression are proposed.
Keywords: aggressive cancer, anaerobic bacteria, bacterial invasion, cancer, infection, intracellular bacteria, mechanisms, metastases, pathogenic, polymicrobial
Infectious agents and anaerobic bacteria associated with cancer
There are currently ten well-established carcinogenic infectious agents associated with cancer development, including Helicobacter pylori causing stomach cancer, hepatitis B and C virus causing liver cancer, human papilloma virus causing cervix uteri cancer and Schistosoma haematobium causing bladder cancer [1], producing an estimated 2 million cancer cases each year [1]. There is growing evidence to indicate that many other pathogenic and clinically significant bacterial species may also play a role in cancer development and/or progression at multiple cancer sites, perhaps most strikingly for specific anaerobic bacteria as detailed in Table 1 (see also Table S1, available in the online version of this article). These are the topic of this review. Specifically, we present new observations and hypotheses involving interlinked mechanisms of polymicrobial infections (complex mixtures of multiple) anaerobic bacteria and cancer, focusing on how multiple anaerobic bacteria may work together to promote cancer development and cancer progression.
Table 1. Anaerobic bacteria taxa (phyla, genera and species), identified in clinical samples reported to be associated with cancer.
| Bacteria phyla | Bacteria genera | Bacterial species | Association with cancer type | Summary cancer types |
| Actinobacteria / Actinomycetota | Propionimicrobium | P. lymphophilum | P. lymphophilum more abundant in urine samples from prostate cancer patients compared with controls [66]. | Prostate |
| Varibaculum | V. cambriense, Varibaculum sp. nov./ Varibaculum prostatecancerukia | V. cambriense more abundant in urine samples from patients with prostate cancer compared with controls [66,146]. Varibaculum one of several taxa enriched in urine samples from bladder cancer cases compared with controls [147]. | ProstateBladder | |
| Bacteroidetes / Bacteroidota | Prevotella | P. timonensis, P. bivia P. buccalis, P. corporis, P. disiens, P. melaninogenica, P. nigrescens, P. pallens, P. nanceiensis, P. copri | Altered abundance of Prevotella sp. with other microbiota associated with laryngeal squamous cell carcinoma [148], gingival and oral squamous cell carcinomas [149,151], intestinal metaplasia [152], colorectal cancer [153,157], breast [158], cervical, endometrial [159], gastric [128,160,162], lung [163, 164] and bladder cancer [165]. Prevotella species associated with cancer include: P. copri plus other bacteria associated with gastric cancer [161,166], P. bivia and cervical cancer [167,168], P. timonensis and high-grade cervical intraepithelial lesions [130]. P. melaninogenica, P. pallens, P. nigrescens, P. nanceiensis associated with oral squamous cell carcinoma [169]. P. buccalis one of several anaerobes detected in melanoma metastases [170]. P. nanceiensis associated with higher risk oesophageal squamous cell carcinoma [171]. | LaryngealOralGingivalColorectalBreastCervicalEndometrialBladderGastricLungMelanomaOesophageal |
| Porphyromonas* | P. bennonis, P. somerae, P. asaccharolytica, P. gingivalis, Porphyromonas sp. nov./ Porphyromonas bobii, P. uenonis, P. endodontalis | Several Porphyromonas sp. [67] including P. asaccharolytica [172] and other anaerobic bacteria including Fusobacterium and Prevotella species detected with increased abundance in gut and cancer tissue associated with colorectal cancer [67,172]. Associated with high- grade/advanced prostate cancer in combination with other anaerobes [10], see Fig. 1. P. gingivalis associated with oral squamous cell carcinoma [173], higher risk oesophageal squamous cell carcinoma [137,171] and pancreatic cancer [174]. Novel uncultured Porphyromonas sp. together with other bacteria species associated with the presence of endometrial cancer [175]. P. uenonis one of several bacteria associated with cervical cancer [130]. P. bennonis one of several anaerobes detected in melanoma metastases [170]. Higher abundance of P. endodontalis associated with oral squamous cell carcinoma [169]. | ColorectalProstateOralOesophagealPancreaticEndometrialCervicalMelanoma | |
| Bacteroides | B. coagulansB. ovatusB. uniformisB. fragilisB. vulgatusB. dorei | Bacteroides sp. isolates frequently isolated from clinical samples including cancer patients [176,177]. Bacteroides sp. and Streptococcus sp. enriched in rectal swab samples of prostate cancer patients compared with non-cancer controls [90]. B. ovatus and B. uniformis two of several anaerobes associated with increased risk of colorectal cancer diagnosis [178]. B. fragilis and Prevotella increased in abundance in faecal samples from colorectal cancer patients compared with control patients [157]. B. vulgatus, B. ovatus, B. dorei detected as intratumoural bacteria in melanoma metastases [170]. Bacteroides spp. and other faecal microbiota taxa associated with pancreatic ductal adenocarcinoma [179]. Bacteroides and other taxa enriched in gastric cancer tissue compared with control tissue [162] and one of several dominant genera in colorectal cancer [76]. | Multiple cancer typesProstateColorectalMelanomaPancreaticGastric | |
| Firmicutes/ Bacillota | Fenollaria* / Ezakiella/ [Sporobacterium] | F. sporofastidiosus | Fenollaria/F. sporofastidiosus associated with high-grade prostate cancer in combination with other anaerobes [10], see Fig. 1. ‘[Sporobacterium]’ decreased together with other microbiota in stool samples from colorectal cancer patients after curative surgery [180]. Ezakiella was identified as one of several anaerobic taxa enriched in vaginal microbiome of patients with cervical or endometrial cancer [158]. Also, Ezakiella one of several anaerobic bacteria genera detected in relatively high abundance in urine of male and female bladder cancer patients [165]. | ProstateColorectalCervicalEndometrialBladder |
| Peptoniphilus* | P. harei, P. coxii, Peptoniphilus sp. nov./ Peptoniphilus rachelemmaiella | Peptoniphilus spp. associated with high-grade advanced prostate cancer in combination with other anaerobes [10], see Fig. 1. Involved in polymicrobial infections, enriched abundance associated with endometrial cancer [175]. P. harei infection in lymphocele in prostate cancer patient post-prostatectomy [181] and one of several anaerobes frequently isolated from clinical samples from cancer patients [176]. Peptoniphilus together with other bacteria enriched in breast cancer tissue compared to healthy control tissue samples [158], also detected in relatively high abundance in urine of bladder cancer patients [165]. Peptoniphilus, and other anaerobic genera dominant in oral squamous cell carcinoma [76]. Peptoniphilus enriched in vaginal microbiota of endometrial and cervical cancer patients compared to controls with no cancer [159]. | ProstateEndometrialBreastBladderOralCervical | |
| Anaerococcus* | A. lactolyticus, A. prevotii, A. tetradius | Anaerococcus sp. associated with high-grade advanced prostate cancer in combination with other anaerobes [10] see Fig. 1. A. lactolyticus more abundant in urine samples from prostate cancer patients compared with controls [66] Anaerococcus sp. and other anaerobes associated with high-grade cervical cancer [131] and enriched in vaginal microbiome of cervical and endometrial cancer patients compared to control group [159]. Anaerococcus one of several anaerobic bacteria genera in relatively high abundance in urine of bladder cancer patients [165] and identified as related to progression of cervical intraepithelial neoplasia [130]. Anaerococcus sp. and Fusobacterium sp. were two of several urinary microbiome bacteria associated with bladder cancer [182]. | ProstateCervicalEndometrialBladder | |
| Veillonella | V. atypicaV. parvulaV. dispar | Microbiota in lower airway, including Veillonella, predictive for lung cancer [163] and lung cancer progression [138]. Veillonella was one of several anaerobic bacteria genera in relatively high abundance in urine of bladder cancer patients compared to controls [147,165]. Veillonella atypica, Veillonella spp. and other taxa enriched in faeces from patients with pancreatic ductal adenocarcinoma [179]. V. dispar and V. parvula detected in melanoma metastases [170]. Gastrointestinal microbiota including Veillonella significantly increased in acute lymphoblastic leukaemia in children compared with controls [183]. Veillonella detected in cancer tissue samples was one of eight taxa with altered abundance associated with gastric cancer [128]. | LungBladderPancreaticMelanomaAcute lymphoblastic leukaemiaGastric | |
| Fusobacteria / Fusobacteriota | Fusobacterium* | F. nucleatumF. hwasookii | F. nucleatum together with other bacteria associated with colorectal cancer [13,14, 178, 184,187], cervical cancer [188], prostate cancer [10] see Fig. 1. Enriched in breast cancer tissue [158,189]. Fusobacterium one of several dominant genera in colorectal cancer and oral squamous cell carcinoma [76]. Fusobacterium sp. with other microbiota in altered abundance associated with laryngeal and oral squamous cell carcinoma [148,190, 191], bladder cancer [182], gastric [128,162], plus cervical and endometrial cancer [130,159]. Fusobacterium and several other anaerobes including Prevotella and Porphyromonas in increased abundance in oral squamous cell carcinoma tumour compared with control healthy mucosa [150,151]. F. nucleatum, F. hwasookii, A. omnicolens, Veillonella atypica, Veillonella spp. and Bacteroides spp. enriched in faeces from patients with pancreatic ductal adenocarcinoma [179]. F. nucleatum and other bacteria associated with breast and pancreatic cancer [12] and detected in melanoma metastases [170]. F. nucleatum enrichment in colorectal tissue associated with poor prognosis colon cancer [192]. In a meta-analysis of several studies Fusobacterium plus several other genera in increased abundance in gastric cancer tissue associated with gastric cancer progression [128]. | ColorectalCervicalEndometrialProstateBreastLaryngealOralBladderMelanomaGastricPancreatic |
| Sneathia | S. sanguinegens | Increased abundance of Sneathia sanguinegens and other anaerobes (Anaerococcus sp.) associated with high-grade cervical cancer [131]. Increased abundance of Sneathia and other anaerobes associated with cervical and endometrial cancer [130,159]. | CervicalEndometrial | |
| Multiple anaerobes including those belonging to phyla Bacteroidetes, Firmicutes and Fusobacteria | Multiple genera including Porphyromonas*, Prevotella, Bacteroides,Veillonella, Fenollaria*, Ezakiella/[Sporobacterium], Peptoniphilus*, Anaerococcus*Fusobacterium*Sneathia | Multiple species, polymicrobial infection | See above, most studies [10,66, 67, 76, 128, 130, 147, 148, 150, 151, 158,160, 162, 169] report altered abundance of several anaerobes, often co-occurring as polymicrobial infection, that are associated with multiple cancer types. Increased number of anaerobes cultured from prostate cancer tissue compared with benign tissue (bacteria not fully characterized) [17]. Multiple bacteria in tissue, including Fusobacterium, associated with many types of cancer [11], analyses of data [11] indicated other anaerobes present in cancer tissue. Intracellular bacteria associated with multiple cancers including ovary, lung, bone, colorectal, breast, taxa including anaerobe Bacteroidia, Prevotellaceae, in addition, Fusobacterium sp. associated with breast and pancreatic cancer [12]. Clostridia class of bacteria and Bacteroidia two anaerobic taxa enriched in pancreatic cancer tissue associated with pancreatic cancer patient shorter term survival [129]. | ProstateColorectalCervicalBreastEndometrialLaryngealOralGastricPancreaticBladderLungOesophagealMelanoma |
♦*Anaerobic bacteria biomarkers set (ABBS) associated with prostate cancer progression and also associated with several other types of cancer as detailed above.
Other infectious agents and their links to cancer have been reviewed elsewhere [2,8] including Mycoplasma spp. [3,4], microaerophilic Campylobacter sp. [5], facultative Salmonella [8] and microaerophilic/capnophilic H. pylori [2]; where relevant we mention the known oncomicrobe H. pylori for comparison. Associations of bacteria to anticancer drug response are also presented elsewhere [9].
Recently, data have linked specific pathogenic anaerobic bacteria to aggressive disease. The bacteria Anaerococcus spp., Fusobacterium spp., Peptoniphilus spp., Porphyromonas spp. and Fenollaria spp. called the Anaerobic Bacteria Biomarkers Set (ABBS) [10] are associated with high-grade prostate cancer, cancer progression and increased risk of metastases. Notably, many of these bacteria are also associated with other cancer types (Tables 1 and S1). Thus, specific bacteria can potentially have roles in the development of multiple cancers, contrasting with the recent view that bacteria may be specific to and used in the diagnosis of individual cancer types [11]. Cancer types associated with anaerobic pathogenic bacteria and/or microbiota dysbiosis additionally include prostate, oral/head and neck, oesophageal, gastric, pancreatic, colorectal, breast, endometrial, cervical, acute lymphoblastic leukaemia, melanoma, bladder and lung cancer (detailed in Tables 1 and S1). In our own analysis of public datasets (unpublished), we found evidence that ABBS bacteria were present in cancer tissues from stomach, colorectal, oesophageal, head and neck, breast, cervical, bladder, and prostate cancer in agreement with Table 1, but also in ovarian, and kidney cancer.
Importantly bacteria associated with cancer are commonly present intracellularly, inside the human cells [10,12], and invasive within cancer tissue [12,15]. The terms used to describe the bacteria were ‘intracellular bacterial pathogens’ and ‘intratumour(al) bacteria’ [12,13, 15]. A review of specific cancer-associated bacteria and the mechanisms used to invade and persist within human host cells is included below. H. pylori a known carcinogenic bacterial pathogen, can also be invasive and intracellular (reviewed in [16]). Publications from the 1970s and 1980s [17,18] link anaerobic bacteria and cancer but the exact identities of the bacteria could not, at the time, be easily determined [17,18]. Recent technologies have allowed the identification of specific bacteria associated with cancer types (Tables 1, and S1) leading, through experimentation and genome analysis, to hypotheses and discoveries of how the bacteria, including recently isolated species (Table 1), may contribute to cancer progression. We hypothesise that there may be common inter-linked mechanisms for the effect of invasive cancer-associated anaerobic bacterial pathogens on cancer progression relevant to several cancer types.
The proposed mechanisms of action of polymicrobial anaerobic bacteria combined to cause cancer development and cancer invasion are presented in steps 1 to 7 in this review. We provide a review of the seven steps of how multiple anaerobic bacteria may contribute to both the early events in cancer development steps 1 to 3 causing several changes in normal cells to cancer development. In addition also to the proposed later events of the effects and mechanisms of action of cancer-associated anaerobic bacteria discussed from steps 1 through 7 to cause cancer progression and invasion. The seven steps could be considered as hallmarks of cancer associated with anaerobic cancer pathogens.
Step 1 Bacteria eliciting microenvironment changes
One of the early events of bacterial effects on the human host includes bacterial secretion of proteases, including metalloproteases, and activation of host cell matrix metalloproteases (MMPs), leading to degradation of the extracellular matrix (collagen, laminin, fibronectin). For reviews on the impact of bacterial pathogens and effects on host extracellular matrix in other models see [19] and [20]. Anaerobic cancer-associated bacteria proposed to be involved in this process (Fig. 1, step 1), include F. sporofastidiosus, F. nucleatum, Porphyromonas spp., Varibaculum spp., Anaerococcus spp. and Peptoniphilus spp. that encode predicted HtrA/DegQ family proteins, FtsH metalloprotease zincins, PrsW protease, serine protease and/or predicted collagenase activity [10]. Collagenase activity, predicted in F. sporofastidiosus, Porphyromonas spp., Peptoniphilus spp. and Anaerococcus spp., could enable the bacteria to cause extracellular matrix remodelling. In addition, bacterial secreted HtrA can open cell-to-cell junctions and break down the epithelial barrier [21,23], allowing bacteria to access the basement membrane. This would in turn enable bacterial pathogen interaction with integrins leading to subsequent signalling pathways plus E-cadherin-β-catenin signalling, see step 2. Anaerobic bacteria can also decrease pH in the microenvironment, with the low pH leading to the high affinity extended open state integrin headpiece [24] allowing bacterial binding, integrin activation and signalling mechanisms, as outlined in step 2A.
Fig. 1. The seven steps for how human cell invasive pathogenic bacterial species may cause cancer and invasion. The key bacteria involved are included in the legend, other bacteria may be involved at specific steps as discussed in the text and detailed in Table 1, including Sneathia spp., Prevotella spp. and Veillonella spp. Early events of effects of cancer associated bacteria steps 1 to 3, proposed later events of cancer-associated bacteria steps 4 to 7 (also including step 2). The detailed mechanisms of action of the anaerobic bacteria causing host cell and tissue changes associated with carcinogenesis and cancer cell invasion and the proposed order of events are described, several of the mechanisms in the steps are strongly supported by evidence and some are hypotheses, as described in further detail below in the review main text, steps 1 to 7.
Mechanisms Step 1: multiple anaerobic cancer-associated bacteria cause altered MMPs proteases, E-cadherin cleavage, allowing the bacteria to access the basement membrane and cause other microenvironment changes in human host tissue associated with cancer.
Several of the cancer anaerobic bacteria effects on the microenvironment have also been associated with later stages of cancer progression and invasion. F. nucleatum and Prevotella sp. have been documented to increase secretion of host MMPs, including MMP7, MMP13, MMP9, activated MMP9 [19,25], involved in degradation of extracellular matrix, contributing both to increased invasion of the bacteria into the tissue and to cancer invasion [26,27]. Porphyromonas spp. P. bobii and Fusobacterium nucleatum encode predicted components of the Tol-Pal system (including Tol R, Q, A, C, A, Ybg) and may secrete outer membrane vesicles (OMVs) and extracellular metalloproteases into the extracellular environment [20,28,30]. HtrA can also be secreted in bacterial OMVs [21] contributing to breakdown of epithelial barrier via cleavage of E-cadherin [21,23], enabling further bacterial invasion and microenvironment changes associated with cancer. Altered E-cadherin can have significant effects on cancer formation and progression [31,33]. Bacterial effects on the microenvironment, Mechanisms Step 1 (box 1), are also linked with later mechanisms, see steps 2 and 7, and Fig. 1.
Step 2 Bacterial activation of host cell signalling events and modulation of pathways linked with cancer
Step 2A Bacteria interaction with integrin α5β1 and signalling pathway
Integrins are located on the basement membrane [34] and may be accessible to bacterial pathogen binding following changes in the microenvironment (step 1). Multiple bacterial species have been shown to interact with host cell integrins [20,35, 36], particularly species belonging to Porphyromonas genera [20] (Table 1). Bacteria may also bind to CD44 and/or ITGα5β1, other integrins such as αvβ3 and αvβ5 [20,35, 37], plus may interact with integrins directly or using vitronectin as a crosslinker between bacteria and host [20,35, 37].
Integrin binding and activation of host cell-signalling events is one mechanism by which bacteria can stimulate their own uptake and invasion into the human cell: for review articles see [20,35, 38]. The summary of steps involved in bacteria and integrin binding plus the subsequent activation of host cell signalling pathways is described below and is compiled from multiple sources [19,20, 35,42] to reach a consensus. What is striking is that these same cell signalling cascades and proteins involved in response to the bacteria are also frequently aberrant in multiple types of cancer [33,43] and thus lead to the Mechanisms Step 2 in box 2 below plus mechanisms detailed in Fig. 1.
Bacteria including Porphyromonas sp. bind to integrins via fimbriae, (e.g. FimA; other bacteria may use adhesins IpaA/Ipa complex, ApaH [20] or bind via vitronectin [35]), resulting in formation of focal adhesion complexes [20,35] and recruitment, within the human cell, of focal adhesion proteins, talin, vinculin, activated focal adhesion kinase (FAK), Src, paxillin (PXN), and integrin-linked kinase (ILK) [20,35, 37]. In turn this leads to activation of Ras, PI3K, GTPases Cdc42, Rho and Rac1 [20,35, 36, 38, 41] (See Fig. 1). Filopodia/invadopodia and membrane ruffle formation then ensues via invadopodia protein complex formation including α-actinin, Rac1 activation and formation of complex with IRSp53, WAVE complex, cofilin and the Arp2/3 complex at the site [20,35]. Cofilin and Arp2/3 are involved in F-actin polymerization [20]. Other proteins implicated as involved in bacterial invasion invadopodia complex, actin nucleation and polymerization (depending on cell type) include N-WASP (Cdc42 activates N-WASP), profilin, Nck and cortactin (cortactin and vinculin have F-actin binding sites) [20,35, 38, 39, 41, 43]. Cdc42 also regulates MMPs at the protruding tip of the invadopodia including membrane type 1 matrix metalloproteinase (MT1-MMP/MMP14), with subsequent extracellular matrix degradation and formation of filopodia/ invadopodium, membrane ruffles and bacteria uptake into cell [19,20, 35,43]. All of the proposed interconnected signalling steps are summarized in Fig. 1. Bacterial activation of host cell signalling and modulation of pathways is also associated with host cell changes in gene expression/cell proliferation (Fig. 1, step 2 summary). These bacterial effects on cell signalling and in particular on Cdc42 are also critical for later steps in cancer progression including cancer invasion (Fig. 1, step 7 [20,43]).
Step 2B Bacteria interaction with E-cadherin receptor and cell signalling events
In an established mechanism, F. nucleatum binds E-cadherin receptor via bacterial FadA adhesin stimulating activation of the host β-catenin signalling pathways and downstream effects on the human host cell [6,44]. Host changes include increased gene expression of NFKβ, IL8, IL6 and Wnt, alteration in oncogenes Myc and Cyclin D1, and Rac1 and Cyclin D1 activation, overall causing increased growth stimulation of the host cell and facilitating bacteria uptake [6,44, 45]. Additionally, there is cross talk between the E-cadherin-β-catenin signalling pathway and integrin pathway as illustrated by the combined effects on PI3K, Cdc42 and Rac1 activation [34]. Activation of the Wnt/β-catenin pathway in human cells results in nuclear accumulation of β-catenin and aberrant accumulation of β-catenin in the nucleus of cancer cells has frequently been reported [46]. Bacteria can also directly mediate nuclear accumulation of β-catenin, for example, via protease mediated interaction with E-cadherin, causing dissociation of β-catenin from E-cadherin allowing β-catenin to be translocated to the nucleus where the effects subsequently result in altered gene expression and cell proliferation [46,47].
Steps 2A and 2B above facilitate the uptake of bacteria into the host cell together with modulation of host signalling pathways, including activation of Ras signalling pathways and small GTPases Cdc42, Rac1, stimulation of MAPKs, JNK [20], PI3K, Akt, NFKβ [20,35, 36, 38, 41, 42]. In turn, JNK, Ras and MAPK, ERK are known ETS modulators [48] (Fig. 1) potentially inducing an aggressive/invasive phenotype in multiple cancer types [49]. The combined consequences of bacteria on the human host cell pathways include: (i) changes in human host cell gene expression; (ii) modulation of inflammation regulators including IL8; (iii) increased cell proliferation; (iv) suppression of apoptosis/cell death and (v) dysregulated cell growth [6,20, 34,39, 41, 42, 44] (Fig. 1).
Overall, the pathways of effects of bacteria upon host cell invasion and cell signalling (Mechanisms Step 2) are frequently detected as aberrant pathways in carcinogenesis [33,40, 43] therefore may indicate a potential polymicrobial pathogen bacterial driven process.
Mechanisms Step 2: multiple bacterial species interact with human cells facilitating bacterial uptake and cause aberrant cell signalling (incl. GTPases, Ras, MAPK, PI3K, Cdc42 pathways), promoting cancer progression.
Step 3 Invasive pathogenic bacteria causing host cell DNA damage and genomic instability
Infected host cells are often unable to eradicate bacteria, which then grow intracellularly in the nutrient rich cytoplasm and may locate to the cells perinuclear region [20], close to the nuclear membrane [12]. There they are well placed to cause chromosome damage, including single strand and double-strand breaks, through secreting nucleases and impairing DNA repair, similar to the mechanisms observed for H. pylori [50,52]. In lung cancer the presence of Porphyromonas, Fusobacterium and Bacteroides genera were significantly associated with chromosomal aberrations [53]. Fusobacterium spp. are known to encode and secrete extracellular deoxyribonucleases [10,54], and elevated levels of F. nucleatum DNA in colorectal cancer tissue sections were (i) detected close to the marker γH2AX for DNA double-strand breaks, and (ii) found to be associated with increased number of somatic mutations and microsatellite instability [55,56].
Other bacterial infections have been associated with host cell DNA damage via known mechanisms, for example, specific strains of E. coli, may have pathogenicity island pks that encodes enzymes for synthesis of the DNA damaging agent colibactin [57,58]. pks+ E.coli are associated with human host cell DNA damage and oncogenic gene fusions in prostate cancer [58] and with an oncogenic mutational signature in colorectal cancer [59]. Known toxic bacterial metabolites including nitrosamines may also cause host cell DNA damage [2] and bile acid degradation products produced by bacteria can result in increased reactive oxygen and nitrogen species leading to increased DNA damage (reviewed in [2]).
With respect to the role of the anaerobic bacteria in step 3, several bacteria listed in Table 1 (F. sporofastidiosus, Porphyromonas sp., P. bobii, Peptoniphilus sp., Peptoniphilus rachelemmaiella, Anaerococcus prevotii, Fusobacterium nucleatum) encode predicted MutS homologs (MutS, MutS2) but not MutH (extended data ref [10]), and thus are similar to H. pylori in lacking the MutH protein required for the methyl-directed MutSLH DNA mismatch system [60]. Chronic persistent bacterial pathogen infection may then alter the human host cell mismatch repair system as does H. pylori [50,51, 60]. Several strains with homologs MutS and MutS2 (F. sporofastidiosus, P. bobii, Peptoniphilus harei, Peptoniphilus rachelemmaiella, Anaerococcus prevotii) also lack MutY and MutM, a genotype reported to be more common in clinical bacteria associated with long term infection persistence [61], and with enhanced G:C to T:A transversions [50,60,62]. F. sporofastidiosus, P. bobii, F. nucleatum, Peptoniphilus harei and Peptoniphilus rachelemmaiella have both MutS and RuvC endonuclease that support genetic DNA recombination to provide oxidative DNA damage repair in the bacterial cells [60] to the benefit of bacterial growth. Varibaculum prostatecancerukia identified in prostate tissue [10] encodes purine deoxyribonucleosides salvage pathway components, similar to H. pylori [52], enzymes and extracellular nuclease capable of scavenging purine nucleotides from host DNA [52] causing DNA damage.
Overall the anaerobic bacteria associated with cancer are predicted to be involved in host cell DNA damage, increase in mutations, potential gene rearrangements/fusions and impaired DNA repair (another one of the features of cancer [50,51, 63,65]) and lead to Mechanisms Step 3 (box 3), Fig. 1. There are also additional effects of anaerobic bacteria on histone acetylation and epigenetic effects detailed in step 5.
Mechanisms Step 3: multiple anaerobic bacteria species cause host cell genome instability, increased DNA damage and impaired DNA repair contributing to carcinogenesis.
Step 4 Immune modulation by bacteria and bacterial evasion of the immune system
Bacterial infection can lead to inflammation in the tissue microenvironment [44,66,68] and bacteria can be detected intracellularly in immune cells within cancers [12]. Certain Gram-negative bacteria, including Porphyromonas sp. and F. nucleatum (Table 1), interact with immune cells such as macrophages through the Toll-like receptor 4 (TLR4) via recognition of bacterial endotoxin/ lipopolysaccharide (LPS) leading to macrophage activation with increases in MyD88, IL1β, IL6, IRAK, IL12, IL8 TNFα, STAT3 and IL10 production [3,13, 20, 69, 70] (Fig. 1). Over time, chronic infection may ensue with multiple intracellular invasive bacterial species infecting macrophages in the tissue environment [12,68, 71] with the bacteria surviving and multiplying intracellularly [68,71] leading to bacterial immune cell evasion and increased polarization of macrophages from the M1 to the M2 tumour associated macrophages (TAMs) [69,71]. The consequence is increased cytokine release: activated macrophages produce proteases, secrete cytokines including IL1β, TNFα, TGFβ, IL10, and IL6, and secrete osteopontin/secreted phosphoprotein 1, SPP1 [70,72] (Fig. 1). Contrasting to the initial induction of inflammation (see above), these changes cascade to anti-inflammation effects (including reduction in T lymphocytes), tissue remodelling, fibrosis and angiogenesis [70,72] in turn promoting tumour growth and reducing immune response to pathogens [68,70].
Fusobacterium spp. are correlated with markers of immune cells including dendritic cells and TAMs in human colorectal cancer tissue [13]. The change in the balance between increased levels of TAMs and reduced tumour infiltrating lymphocytes (TILs) leads to immune cell evasion for the bacteria, increased growth and spread/invasion into tissue. This is due to the reduction in CD8+, CD3+ and CD4+ T cells, TILs, in the environment which would typically contribute to the main defences against intracellular bacterial pathogens [68], but are significantly decreased by the effects of the bacteria, thus perpetuating further bacteria growth and survival.
The overall bacterial effects result in chronic cancer-promoting inflammation, with presence of certain sub-types of macrophages (including M2 TAMs), myeloid-derived suppressor cells (MDSCs), dendritic cells and other types of T lymphocytes (regulatory T cells) in cancer tissue also associated with cancer and progression/metastases [73,75]. In human colorectal cancer F. nucleatum was associated with recruitment of myeloid cells to the infected tissue and promoted changes associated with invasion [76]. In mouse models, more rapid intestinal and colon tumorigenesis induced by F. nucleatum [13] resulted in increased MDSCs, including monocytic and granulocytic MDSCs, which have strong immune suppressive effects, in particular suppression of CD4+ T cells [13]. The overall effects on immune cells (↑MDSCs, ↑M2 TAMs, ↓CD8+ CD4+ T cells) provide tumour enhancing activity and contribute to cancer progression [13,68, 70]. Indeed, removal of bacterial pathogens using an antibiotic cocktail (vancomycin, neomycin, metronidazole, amphotericin) in a mouse model reduced the MDSCs and increased M1 macrophage cells, promoting activation of CD4+ and CD8+ T cells, and protecting against invasive pancreatic ductal adenocarcinoma [77,78]. In several mouse model studies, microbiota have been associated with modulation of the tumour immune response [13,77, 78].
Downstream effects of F. nucleatum infection are in some cases linked to IL6 secretion by host cells [13,44] and activation of macrophages [13,70, 71]. IL6 can impact on neighbouring tumour cells via the IL6 cytokine receptor, causing activation of JAK and STAT3 signalling [79,81] (Fig. 1) leading to changes in gene expression and cellular proliferation [79,80]. Alteration of the STAT3 pathway is one of the aberrant signalling pathways contributing to the hallmarks of cancer [33]. Importantly, bacterial effects linked in with the downstream effects on STAT3 activation may also increase the expression of programmed death-ligand 1 (PD-L1) (Fig. 1). The effect on STAT3 and increased PD-L1 has been shown in several cancer types [82,83], leading to T cell suppression and tumour cell immune evasion [82,83] plus further contribution to bacteria immune evasion. TNFα and IL10 secreted by activated macrophages (Fig. 1) can also increase cancer cell and TAM expression of PD-L1 [70,83]. Indeed, F. nucleatum infection of cultured macrophages increased expression of TNFα and PD-L1 [84]. Bacterial OMVs secreted by Porphyromonas sp. and F. nucleatum have also been linked with effects on immune cells and modulation of immune response to pathogens, increasing the pathogenic effects of bacteria and enabling immune cell evasion [28,85,87]. Microbiota may in addition affect anticancer immunotherapy treatment, including anti-PD-L1 therapy [78], and other immunotherapies reviewed in [88,89].
Overall, the effects of the intracellular anaerobic bacterial infection on immune cells and signalling pathways are predicted to be substantial, allow the bacteria to grow undetected and to perpetuate, impacting cancer progression, leading to Mechanisms Step 4 (box 4).
Mechanisms Step 4: anaerobic cancer-associated bacteria may cause polarization of macrophages to M2 TAMs, and increases in MDSCs, leading to downstream effects on cytokines and modulation of signalling pathways resulting in increased angiogenesis, bacteria and cancer cell immune evasion and cancer progression /invasion.
Mechanisms Step 4 provides a potential route for treatment options for the cancer-associated anaerobic bacteria associated with immunosuppressive effects (see section below ‘Potential options for treatment of cancer associated with anaerobic bacterial pathogens’).
Step 5 Widespread effects of invasive intracellular bacteria on host cell metabolism pathways
It is established that bacteria associated with cancer can perturb metabolism in host cells and tissue. This is illustrated by the gut microbiota modulation of short-chain fatty acids and downstream anti-inflammatory effects, altering host gene expression and cell proliferation [6]. Bacterial metabolites including nitrosamines and bile acid degradation products can result in host cell effects including increased DNA damage [2] (step 3). Gut microbiota enriched in patients with prostate cancer (including Bacteroides, see Tables 1, and S1) were associated with encoded folate, biotin and arginine metabolism pathways [90]. Microbiota have also been associated with metabolism of oestrogen the ‘estrobolome’ (reviewed in [91,92]). We have been able to postulate several interlinked effects of the intracellular anaerobic bacteria on host cell metabolism (Fig. 1, steps 5A to E and Fig. 2, step 5A with expanded details), relevant to several types of cancer as follows.
Fig. 2. Step 5A, potential mechanisms of action of human cell invasive intracellular pathogenic bacterial species on host cell glycine metabolism and associated pathways. hCYS: homocysteine; MET: methionine; meTHF: 5,10-methylenetetrahydrofolate; MS: bacterial methionine synthase; mTHF: 5-methyltetrahydrofolate; MTHFR: bacterial methylenetetrahydrofolate reductase; SAH: S-adenosylhomocysteine; SAM: S-adenosylmethionine; THF: tetrahydrofolate; ThyX: bacterial thymidylate synthase. Impacts of specific bacteria on the host cell glycine metabolism and associated pathways are as indicated with the symbolized images of the bacteria displayed in the Fig. 2 key legend. Bacterial enzyme activities and enzyme complexes are detailed (i) to (v) above and in the review main text.
Step 5A Effects on glycine metabolism and associated pathways: folate, one-carbon and methylation
Bacterial glycine cleavage complex – several of the anaerobic bacteria, Porphyromonas spp. (including P. bobii), Fusobacterium spp. (including F. nucleatum) and F. sporofastidiosus, encode predicted bacterial multienzyme glycine cleavage complex [10] (including glycine decarboxylase/glycine dehydrogenase, glycine reductase, aminomethyltransferase). Via the glycine cleavage complex intracellular bacteria would metabolize glycine to ammonia, CO2, NADPH, with formation of 5,10-methylenetetrahydrofolate (meTHF); the latter used for nucleotide synthesis [93,94]. These alterations would modulate host cell glycine levels, increase meTHF (See Fig. 2, step 5A), and alter folate and one-carbon metabolism potentially impacting on DNA methylation. These pathways have also been identified as aberrant in cancer progression [95,97]. Increased activation of glycine and serine pathways and one-carbon metabolism have been linked to drivers of oncogenesis with associated epigenetic effects and cancer progression/development [95,97]. Increased glycine consumption in cancer is one of the main features identified in cancer metabolic reprogramming [95,96]. With flux regulation glycine can also be metabolized to form sarcosine, and choline and betaine can be metabolized to glycine (step 5, Fig. 1); this is relevant because glycine and sarcosine uptake and metabolism are linked to cancer invasiveness [96,98].
Via bacterial thymidylate synthase – several of the anaerobic bacteria (F. sporofastidiosus, Anaerococcus prevotii and Porphyromonas spp.) also encode ThyX [10], thymidylate synthase, that catalyses meTHF to THF, for thymidylate synthesis, contributing to growth benefits and resistance to certain antifolate treatment [99] (with the bacterial glycine cleavage (i) described above affecting this metabolism via production of meTHF [94,99]), see Fig. 2.
Other features of the bacteria modulating host metabolite levels are via bacterial methylenetetrahydrofolate reductase [NAD(P)H] activity, (MTHFR) (iii) and via bacterial methionine synthase, (MS) (iv) see Fig. 2. Both MTHFR and MS are encoded in the genomes of anaerobic bacteria Peptoniphilus (including P. rachelemmaiella [10] and P. coxii) and F. nucleatum. The latter encodes a vitamin B12 dependent MS activation domain. Bacterial MTHFR activity catalyses the reaction meTHF to mTHF and MS catalyses the reaction of l-homocysteine and 5-methyltetrahydrofolate to methionine and tetrahydrofolate, with predicted impact on host cell metabolism (Fig. 2, steps 5A iii and iv).
Bacterial S-adenosylmethionine decarboxylase (v) (Fig. 2, 5A v) encoded by Porphyromonas sp. (including P. bobii) and F. nucleatum may impact on host S-adenosylmethionine (SAM), histone DNA and RNA methylation, methionine metabolism and polyamines [96,97, 100, 101]. An overview of the predicted effects of invasive intracellular bacteria on glycine and one-carbon host cell metabolism pathways are shown in Fig. 2.
In addition to the mechanisms shown in Fig. 2, changes in glycine metabolism may be linked to further downstream effects on de novo serine biosynthesis, impaired pyruvate metabolism, γ-glutamyl transferase (GGT) and glutathione (GSH) metabolism step 5B (see Fig. 1). Several of the anaerobic cancer-associated bacteria have relatively advanced antioxidant enzyme protection systems protecting against damage from the host cell and enabling growth of the bacteria intracellularly. For example, F. sporofastidiosus and Peptoniphilus sp. encode both protein–glutamine gamma glutamyl transferase (GGT) and glutathione-s-transferase (GST) (the latter also predicted in Varibaculum sp. nov.) [10]. Bacterial GGT and GST activities are quite rare [102,103], and provide protection against bacterial cell damage during growth and persistence of infection in human host cells [102,103]. Several of the anaerobic bacteria (F. sporofastidiosus, and Peptoniphilus sp.) also encode l-seryl tRNASec selenium transferase activity plus selenocysteine incorporation linked in with the importance of selenium in the glycine cleavage pathway in anaerobes (reviewed by Andreesen [94]).
Overall, the effects of the bacteria on glycine and one-carbon metabolism link in with other effects of other bacterial intracellular species on host cell citrate metabolism pathways and further alterations in metabolism pathways (Fig. 1, step 5).
Step 5C and D Effects on citrate metabolism
Several of the cancer-associated anaerobic bacteria: Peptoniphilus sp., (including P. harei), F. sporofastidiosus and F. nucleatum, encode a predicted citrate lyase complex [10] involved in degradation of citrate and in citrate metabolism pathways. Citrate is metabolized via the bacterial citrate lyase complex to oxaloacetate (that in turn may be converted to pyruvate), and acetyl CoA for increased lipid synthesis [93,104,106]. The bacterial ability to metabolize citrate from the surrounding environment, reviewed in [107], is quite a rare feature. Invasive intracellular bacterial species may alter host cell citrate metabolism for increased lipid synthesis and growth [104,105]. Notably aberrant citrate lyase activity is detected in many cancer types [106,108] with an increase in acetyl CoA and downstream metabolism associated with increased acetylation of histones affecting host global chromatin architecture and altering gene transcription [106,108, 109]. The effects of invasive intracellular bacteria on citrate metabolism together with the bacterial glycine cleavage complex (see above) may lead to epigenetic changes, DNA methylation and alteration of apoptosis [95,96, 106, 108] in the host cell.
This also links in with the effects of bacteria alterations on cell signalling pathways and oncoproteins (step 2B for example) with Ras activation, β-catenin and NFkB impacting on glutamate and glucose metabolism [110] (Fig. 1): glucose feeds into serine and glycine pathways, as well as pyruvate [96,108, 110]. In addition, F. sporofastidiosus, F. nucleatum and Porphyromonas sp. have predicted components for catalysing l-glutamate degradation VI to pyruvate step 5D (Fig. 1) [10], which combined with the other bacterial effects on pyruvate lead to altered pyruvate (Fig. 1).
Step 5E Cholesterol metabolism to androstenedione and androstenedione degradation
Several anaerobic bacteria (Peptoniphilus spp., including P. harei, P. rachelemmaiella, F. nucleatum and Anaerococcus prevotii) are predicted to encode components of superpathways ‘cholesterol to androstenedione I (cholesterol oxidase)’ and ‘cholesterol to androstenedione II (cholesterol dehydrogenase)’ [10]. Only a few other bacterial species (including Mycobacterium sp., Rhodococcus sp. and R. gnavus) possess pathways needed for steroid synthesis [93,111, 112] and metabolites known to be produced by these few specific bacteria species include androstadienedione, testosterone and 1-dehydrotestosterone [111,114]. During bacterial cholesterol metabolism acetyl CoA is also produced [93,115] which may have potential epigenetic effects as described above in step 5C [106,108, 109] (Fig. 1). Peptoniphilus spp. (including P. rachelemmaiella, P. coxii, P. harei) and Anaerococcus prevotii also encode components of the androstenedione degradation pathway [10]. Steroid metabolism in bacteria [10,93, 111,113] has the potential to feed host steroid hormones production, impacting on hormone sensitive cancers (e.g. breast and prostate cancer) driving cancer progression and invasion [114,116, 117].
The wider contribution of metabolites from microbiota to the estrobolome (microbiota gene products that are capable of metabolizing oestrogens) is well established and reviewed in [92], as are its impact on oestrogen driven cancers, including endometrial and breast [92]. Metabolites may also impact on ovarian, cervical, gastric, prostate, bone and lung cancers [92,118]. Bacterial encoded enzymes are capable of producing oestrogen from cholesterol impacting on human host circulated and excreted oestrogen levels [92]. Several known anaerobes have this activity, including [92] Clostridium perfringens, Bacteroides fragilis, species belonging to Firmicutes (in particular Clostridia taxa), Actinobacteria and Bacteroidetes [118,119]. The activity is also predicted in anaerobic bacteria including Varibaculum prostatecancerukia and Anaerococcus prevotii [10].
Overall, the intracellular anaerobic cancer-associated bacteria have the potential to incur widespread effects on human host cell metabolic pathways (steps 5A to E, Figs1 2) relevant to cancer development and progression leading to Mechanisms Step 5 (box 5).
Mechanisms Step 5: intracellular anaerobic bacteria are driving the changes in human host cell metabolism (including glycine, citrate, cholesterol to androstenedione, anaerobic respiration) and methylation impacting through multiple mechanisms on cancer aggression and progression.
Step 6 Bacteria driving hypoxia, cell-cell communication and signalling pathways crosstalk
There is support for a general link between the presence of bacteria and hypoxia. For example, Mycobacterium tuberculosis is associated with hypoxia in human tissue TB lesions [120]. Fusobacterium and Porphyromonas infected cell clusters in colorectal cancer, when compared with bacteria negative cell clusters, showed upregulation of pathways including those related to hypoxia [76]. In lung cancer, intratumoural bacterial infection burden was significantly correlated with HIF1A gene expression and hypoxia pathways [121]. HIF-1 is the key master regulator of hypoxia status and hypoxia-inducible factor 1-alpha (HIF-1α) has been shown to be overexpressed in many cancer types and is associated with cancer progression [122].
We propose that the anaerobic bacteria associated with cancer may induce hypoxia and promote cancer development. There are several observations that we are now drawing together that are consistent with this idea. Particularly several of the pathways expected to be activated by bacterial contact with human cells (step 2) have the potential to increase activity and/or stability of HIF-1α and contribute to cancer progression. These include Akt activation [110] (Fig. 1) and the Ras-MAPK pathways [27]. Additionally, SPP1 (also known as osteopontin/secreted phosphoprotein 1) [72,123] secreted by M2 macrophages (step 5) can modulate HIF-1α [70,123] (step 6, Fig. 1), and is associated with hypoxia and cancer metastases [123]. Secreted SPP1 may bind to CD44 and integrins [72,123] altering cell signalling pathways favouring cancer progression [123]. There are a number of additional complexities and supporting observations (see Fig. 1). Hypoxia and HIF-1α increase expression of integrins ITGA5, ITGβ1 [124], which could potentially create a positive feedback for enhancing further bacterial infection. In addition, the cellular effects of anaerobic bacterial infection on signalling pathways including, PI3K/Akt, NFKβ and Ras (step 2) may also impact on Notch signalling, Notch receptors and ligands, with direct crosstalk between signalling pathways [81,125]. Ras can activate Notch signalling and upregulate Notch ligands including Delta-like 1 (DLL1) [81]. Other cellular effects of bacteria on TGFβ, STAT3 and IL6, (step 4) may also upregulate/stimulate the Notch receptor and Notch ligands including JAG1 [81] and DLL1 [126] (see steps 4 and 6, Fig. 1) with engagement of Notch and Notch-ligand (JAG/DLL1) on neighbouring cell facilitating cell-cell communication and signalling [81,126, 127] (Fig. 1). This is relevant as Notch also has critical roles in hypoxia induced epithelial-mesenchymal transformation and cancer invasiveness [81,125]. In addition, Notch expression and ligands are found to be upregulated in many cancers including cervical, head and neck, renal, colon, pancreatic, breast and prostate [81,127].
These considerations lead to Mechanisms Step 6 (box 6).
Mechanisms Step 6: intracellular anaerobic bacteria are driving the changes in cell signalling causing hypoxia plus an increase in cell-to-cell communication and signalling pathway crosstalk increasing cancer progression.
Step 7 Bacteria causing cancer cell invasion and metastases
We have proposed that mechanisms from steps 1 through 6 impact, either directly or indirectly, on step 7 leading to cancer invasion and metastases (Fig. 3).
Fig. 3. Summary overview of steps 1 to 7 and the interlinked mechanisms of how multiple invasive anaerobic bacteria may cause cancer and cancer progression. The key bacteria involved are included in the legend, other bacteria may be involved at specific steps as discussed in the text and detailed in Table 1, including Sneathia spp., Prevotella spp. and Veillonella spp.
Evidence for the association of anaerobic bacteria with cancer progression exists, including genera Fusobacterium and Prevotella, associated with gastric cancer progression [128], Clostridia and Bacteroidia associated with shorter survival time in pancreatic cancer [129], plus a group of five bacteria genera (Fusobacterium, Anaerococcus, Fenollaria, Porphyromonas and Peptoniphilus, ABBS) associated with prostate cancer progression [10]. Anaerobic bacteria associated with high grade cervical intraepithelial neoplasia were Sneathia sp., Prevotella, Fusobacterium, Veillonella, Anaerococcus and Porphyromonas species [130,131], with cervical cancer progression associated with higher levels of Sneathia sanguinegens [131] and Anaerococcus sp. [130].
Detailed mechanisms of how bacteria may be associated with cancer invasion are included in the steps above except for the following as a highlighted example. The effects of bacteria on cell signalling including Ras and Cdc42 [20,35, 36, 38, 41], MT1-MMP and invadopodia (detailed in step 2) associated with membrane ruffles and uptake of bacteria; incredibly similar pathways are detailed as associated with cancer invasion into the bloodstream and metastases [27,43, 132]. Cdc42 and MT1-MMP, increased by the downstream effects of intracellular pathogens on human host cells (see above steps), are critical for invasion of many cancer types [27] but also cdc42 may be linked with formation of tunnelling nanotubes which may enable further cell to cell spread of invasive intracellular bacteria (Fig. 1) [20,133,135]. Other detailed mechanisms are included in the steps above.
The mechanisms are interlinked as shown in Fig. 3 with several anaerobic bacteria (as listed in Table 1) associated with cancer progression in various types of cancer. The combined and interlinked effects of multiple invasive bacteria driving changes in cellular signalling pathways, gene expression changes, DNA damage, host cell metabolism, immune cell modulation and macrophage polarization, leading to widespread changes in human cells and the tumour microenvironment as described in the steps above (steps 1–6; Figs1 3). This may lead to promotion of cancer, increased growth and spread, increased tissue invasion, disruption of surrounding tissue structure and architecture, increased angiogenesis, and potential for increased cancer invasion to blood, nerves and lymphatic tissue. Known oncomicrobe H. pylori infection is significantly associated with advanced lymphatic metastases [136]. Porphyromonas gingivalis an anaerobic pathogen detected in oesophageal tissue was positively associated with lymph node metastases and poor overall survival rate in oesophageal squamous cell carcinoma patients [137]. In addition, airway microbiota enriched with Veillonella parvula in lung cancer patients was significantly associated with poor prognosis and cancer metastases [138]. Taken together these observations lead to Mechanisms Step 7, Fig. 1, summarised in box 7.
Mechanisms Step 7: long-term chronic infection with intracellular anaerobic bacteria in human host cells and cancer tissue drives cellular changes linked with cancer invasion, with interlinked ‘vicious circle’ effects of multiple invasive pathogenic bacteria leading to cancer progression and metastases.
Potential options for treatment of cancer associated with anaerobic bacterial pathogens
The framework of mechanisms of action by which specific anaerobic bacteria may cause cancer (Fig. 1) provides treatment options to halt cancer development and progression. The aim would be to detect and eradicate anaerobic pathogens associated with cancer (Tables 1 and S1) to treat the cancer at the ‘top of the chain’, eradicate the anaerobic invasive pathogens and prevent effects of the intracellular pathogens on cancer progression.
Specific treatment options to eradicate the anaerobic invasive bacteria include modulation of macrophage polarization, specific targeted antimicrobial therapy and if late chronic stage infection in cancer tissue surgery to reduce high burden infected cancer tissue. The association of the anaerobic bacteria with immunosuppressive effects, particularly on polarization of macrophage to M2 TAMs (as discussed in step 4) presents options for immune modulation to increase success for the removal of the anaerobic bacteria. It is possible to reverse macrophage polarization of M2 TAMs back to M1 phenotype [70,71] resulting in recruitment of T cells into the tissue. Reversal of M2 to M1 macrophage may include via IFNα or CD40 activation [70], or via bacterial eradication – good antimicrobial options for emerging anaerobes include ertapenem, imipenem, meropenem, metronidazole, tigecycline, plus other antimicrobials [139]. An additional provision of immune modulation to increase healthy active CD4+, total CD3+, cytotoxic CD8+ T cells to the tissue to kill the remaining intracellular pathogens and cancer cells, based on mechanism of effects of the bacteria, would prevent cancer development and cancer progression.
Other routes of treatment for bacterial infection in cancer in the future may include phage-based therapy and engineered bacteria with care required for use of antibiotics as reviewed in [140].
We are proposing, based on steps 1–7 and the overarching mechanisms of action of the anaerobic bacteria linked to many cancer types as discussed in this review, that specific targeted antimicrobial therapy to remove the specific anaerobic bacteria (+/- surgery/immune therapy in chronic cases) would be an option for the prevention of cancer progression and metastases. A similar approach has been used for the oncomicrobe H. pylori with antimicrobial treatment options, protecting against H. pylori infection and gastric cancer (reviewed in [2,141, 142]). Eradication therapy to target H. pylori infection significantly reduced risk of gastric adenocarcinoma [143] and reduced cancer mortality [141,142]. Doxycycline antimicrobial treatment of infection in ophthalmic MALT lymphoma [144] and antimicrobial clarithromycin in advanced non-small cell lung cancer improved survival in patients with advanced non-small cell lung cancer [145]. There are a number of critical issues to consider when considering if antimicrobial treatment is appropriate. First, are all associations of bacteria with cancer development causal or is there a common underlying reason for observing an association of the presence of specific bacteria and aggressive disease? For example, a defect in the immune system promoting both cancer and bacterial growth – mechanisms that underpin each of the steps 1 to 7 would argue for the former. Secondly, where multiple bacteria are working together can a targeted treatment regime be developed that will penetrate the target tissue and eradicate the target anaerobic pathogens while sparing the commensal bacteria species.
Concluding remarks and future perspectives
We have presented hypotheses of how multiple anaerobic pathogens may act together to cause cancer development and progression providing potential mechanisms detailed in the above steps 1 to 7 and summarized in Figs13. Cancer types associated with anaerobic pathogenic bacteria include oral/head and neck, oesophageal, gastric, pancreatic, colorectal, breast, ovarian, endometrial, cervical, prostate, bladder, kidney, acute lymphoblastic leukaemia, bone, melanoma and lung cancer. Mechanistically, the anaerobic cancer-associated bacteria possess highly specialized homologs, unique bacterial proteins and machinery to alter the human host cell to obtain growth advantage for themselves intracellularly, while causing multiple changes in the human cell promoting carcinogenesis and cancer spread. The actions of different bacteria may synergize with each other to cause cancer development. With regard to prevention, it will be important to determine the routes of infection of the multiple cancer-associated anaerobic pathogens. Future studies need to be directed towards screening and treatment of specific anaerobic bacterial infections, assessing the value of antimicrobial therapies in preventing cancer progression.
supplementary material
Footnotes
Funding: This work was funded by the Bob Champion Cancer Trust, Prostate Cancer UK (RIA15-ST2-029), and the Alan Boswell Group Cancer Research Fellowship Funding.
Contributor Information
Rachel Hurst, Email: R.hurst1@uea.ac.uk.
Daniel S. Brewer, Email: D.Brewer@uea.ac.uk.
Abraham Gihawi, Email: A.Gihawi@uea.ac.uk.
John Wain, Email: john.wain@quadram.ac.uk.
Colin S. Cooper, Email: Colin.Cooper@uea.ac.uk.
References
- 1.Plummer M, de Martel C, Vignat J, Ferlay J, Bray F, et al. Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Glob Health. 2016;4:e609–16. doi: 10.1016/S2214-109X(16)30143-7. [DOI] [PubMed] [Google Scholar]
- 2.Chang AH, Parsonnet J. Role of bacteria in oncogenesis. Clin Microbiol Rev. 2010;23:837–857. doi: 10.1128/CMR.00012-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sinkovics JG. Molecular biology of oncogenic inflammatory processes. I. Non-oncogenic and oncogenic pathogens, intrinsic inflammatory reactions without pathogens, and microRNA/DNA interactions (Review) Int J Oncol. 2012;40:305–349. doi: 10.3892/ijo.2011.1248. [DOI] [PubMed] [Google Scholar]
- 4.Tsai S, Wear DJ, Shih JW, Lo SC. Mycoplasmas and oncogenesis: persistent infection and multistage malignant transformation. Proc Natl Acad Sci USA. 1995;92:10197–10201. doi: 10.1073/pnas.92.22.10197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kaakoush NO, Castaño-Rodríguez N, Man SM, Mitchell HM. Is Campylobacter to esophageal adenocarcinoma as Helicobacter is to gastric adenocarcinoma? Trends Microbiol. 2015;23:455–462. doi: 10.1016/j.tim.2015.03.009. [DOI] [PubMed] [Google Scholar]
- 6.Garrett WS. Cancer and the microbiota. Science. 2015;348:80–86. doi: 10.1126/science.aaa4972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lax AJ, Thomas W. How bacteria could cause cancer: one step at a time. Trends Microbiol. 2002;10:293–299. doi: 10.1016/s0966-842x(02)02360-0. [DOI] [PubMed] [Google Scholar]
- 8.Khan AA, Bano Y. Salmonella enterica subsp. enterica host-pathogen interactions and their implications in gallbladder cancer. Microb Pathog. 2021;157:105011. doi: 10.1016/j.micpath.2021.105011. [DOI] [PubMed] [Google Scholar]
- 9.Fan JY, Huang Y, Li Y, Muluh TA, Fu SZ, et al. Bacteria in cancer therapy: a new generation of weapons. Cancer Med. 2022;11:4457–4468. doi: 10.1002/cam4.4799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hurst R, Meader E, Gihawi A, Rallapalli G, Clark J, et al. Microbiomes of urine and the prostate are linked to human prostate cancer risk groups. Eur Urol Oncol. 2022;5:412–419. doi: 10.1016/j.euo.2022.03.006. [DOI] [PubMed] [Google Scholar]
- 11.Poore GD, Kopylova E, Zhu Q, Carpenter C, Fraraccio S, et al. Microbiome analyses of blood and tissues suggest cancer diagnostic approach. Nature. 2020;579:567–574. doi: 10.1038/s41586-020-2095-1. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 12.Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science. 2020;368:973–980. doi: 10.1126/science.aay9189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14:207–215. doi: 10.1016/j.chom.2013.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012;22:299–306. doi: 10.1101/gr.126516.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Geller LT, Barzily-Rokni M, Danino T, Jonas OH, Shental N, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357:1156–1160. doi: 10.1126/science.aah5043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Dubois A, Borén T. Helicobacter pylori is invasive and it may be a facultative intracellular organism. Cell Microbiol. 2007;9:1108–1116. doi: 10.1111/j.1462-5822.2007.00921.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Cooper PN, Millar MR, Godwin PGR. Anaerobes and carcinoma of the prostate. BMJ. 1988;296:466–467. doi: 10.1136/bmj.296.6620.466-a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Livingston VW, Alexander-Jackson E. A specific type of organism cultivated from malignancy: bacteriology and proposed classification. Ann N Y Acad Sci. 1970;174:636–654. doi: 10.1111/j.1749-6632.1970.tb45588.x. [DOI] [PubMed] [Google Scholar]
- 19.Singh B, Fleury C, Jalalvand F, Riesbeck K. Human pathogens utilize host extracellular matrix proteins laminin and collagen for adhesion and invasion of the host. FEMS Microbiol Rev. 2012;36:1122–1180. doi: 10.1111/j.1574-6976.2012.00340.x. [DOI] [PubMed] [Google Scholar]
- 20.Bacterial Invasion of Host Cells, Chapter 1, 2, 3, 4, 5, 6, 7, 8, 9, 10. Cambridge University Press, USA: Cambridge University Press; 2004. [Google Scholar]
- 21.Wessler S, Schneider G, Backert S. Bacterial serine protease HtrA as a promising new target for antimicrobial therapy? Cell Commun Signal. 2017;15 doi: 10.1186/s12964-017-0162-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hoy B, Löwer M, Weydig C, Carra G, Tegtmeyer N, et al. Helicobacter pylori HtrA is a new secreted virulence factor that cleaves E-cadherin to disrupt intercellular adhesion. EMBO Rep. 2010;11:798–804. doi: 10.1038/embor.2010.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Boehm M, Hoy B, Rohde M, Tegtmeyer N, Bæk KT, et al. Rapid paracellular transmigration of Campylobacter jejuni across polarized epithelial cells without affecting TER: role of proteolytic-active HtrA cleaving E-cadherin but not fibronectin. Gut Pathog. 2012;4:3. doi: 10.1186/1757-4749-4-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Paradise RK, Lauffenburger DA, Van Vliet KJ. Acidic extracellular pH promotes activation of integrin α(V)β(3) PLoS One. 2011;6:e15746. doi: 10.1371/journal.pone.0015746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Vanlaere I, Libert C. Matrix metalloproteinases as drug targets in infections caused by gram-negative bacteria and in septic shock. Clin Microbiol Rev. 2009;22:224–239. doi: 10.1128/CMR.00047-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Quintero-Fabián S, Arreola R, Becerril-Villanueva E, Torres-Romero JC, Arana-Argáez V, et al. Role of matrix metalloproteinases in angiogenesis and cancer. Front Oncol. 2019;9:1370. doi: 10.3389/fonc.2019.01370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Alexander S, Friedl P. Cancer invasion and resistance: interconnected processes of disease progression and therapy failure. Trends Mol Med. 2012;18:13–26. doi: 10.1016/j.molmed.2011.11.003. [DOI] [PubMed] [Google Scholar]
- 28.Kaparakis-Liaskos M, Ferrero RL. Immune modulation by bacterial outer membrane vesicles. Nat Rev Immunol. 2015;15:375–387. doi: 10.1038/nri3837. [DOI] [PubMed] [Google Scholar]
- 29.Gui MJ, Dashper SG, Slakeski N, Chen YY, Reynolds EC. Spheres of influence: Porphyromonas gingivalis outer membrane vesicles. Mol Oral Microbiol. 2016;31:365–378. doi: 10.1111/omi.12134. [DOI] [PubMed] [Google Scholar]
- 30.Cecil JD, O’Brien-Simpson NM, Lenzo JC, Holden JA, Singleton W, et al. Outer membrane vesicles prime and activate macrophage inflammasomes and cytokine secretion In Vitro and In Vivo. Front Immunol. 2017;8:1017. doi: 10.3389/fimmu.2017.01017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yu W, Yang L, Li T, Zhang Y. Cadherin signaling in cancer: its functions and role as a therapeutic target. Front Oncol. 2019;9:989. doi: 10.3389/fonc.2019.00989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.David JM, Rajasekaran AK. Dishonorable discharge: the oncogenic roles of cleaved E-cadherin fragments. Cancer Res. 2012;72:2917–2923. doi: 10.1158/0008-5472.CAN-11-3498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/S0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
- 34.Weber GF, Bjerke MA, DeSimone DW. Integrins and cadherins join forces to form adhesive networks. J Cell Sci. 2011;124:1183–1193. doi: 10.1242/jcs.064618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Singh B, Su YC, Riesbeck K. Vitronectin in bacterial pathogenesis: a host protein used in complement escape and cellular invasion. Mol Microbiol. 2010;78:545–560. doi: 10.1111/j.1365-2958.2010.07373.x. [DOI] [PubMed] [Google Scholar]
- 36.Eitel J, Heise T, Thiesen U, Dersch P. Cell invasion and IL-8 production pathways initiated by YadA of Yersinia pseudotuberculosis require common signalling molecules (FAK, c-Src, Ras) and distinct cell factors. Cell Microbiol. 2005;7:63–77. doi: 10.1111/j.1462-5822.2004.00434.x. [DOI] [PubMed] [Google Scholar]
- 37.Bergmann S, Lang A, Rohde M, Agarwal V, Rennemeier C, et al. Integrin-linked kinase is required for vitronectin-mediated internalization of Streptococcus pneumoniae by host cells. J Cell Sci. 2009;122:256–267. doi: 10.1242/jcs.035600. [DOI] [PubMed] [Google Scholar]
- 38.Lewis AJ, Richards AC, Mulvey MA. Invasion of host cells and tissues by uropathogenic bacteria. Microbiol Spectr. 2016;4 doi: 10.1128/microbiolspec.UTI-0026-2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Pizarro-Cerdá J, Cossart P. Bacterial adhesion and entry into host cells. Cell. 2006;124:715–727. doi: 10.1016/j.cell.2006.02.012. [DOI] [PubMed] [Google Scholar]
- 40.Yau CYF, Wheeler JJ, Sutton KL, Hedley DW. Inhibition of integrin-linked kinase by a selective small molecule inhibitor, QLT0254, inhibits the PI3K/PKB/mTOR, Stat3, and FKHR pathways and tumor growth, and enhances gemcitabine-induced apoptosis in human orthotopic primary pancreatic cancer xenografts. Cancer Res. 2005;65:1497–1504. doi: 10.1158/0008-5472.CAN-04-2940. [DOI] [PubMed] [Google Scholar]
- 41.Principles of Bacterial Pathogenesis, Chapter 6, 7, 8, 11, 12, 16. Academic Press; 2001. [Google Scholar]
- 42.Bacterial Pathogenesis Molecular and Cellular Mechanisms, Chapter 4, 5, 9, 11, 14, 15. Caister, Norfolk, UK: Caister Academic Press; 2012. [Google Scholar]
- 43.Stengel K, Zheng Y. Cdc42 in oncogenic transformation, invasion, and tumorigenesis. Cell Signal. 2011;23:1415–1423. doi: 10.1016/j.cellsig.2011.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Rubinstein MR, Wang XW, Liu WD, Hao YJ, Cai GF, et al. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013;14:195–206. doi: 10.1016/j.chom.2013.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Shang FM, Liu HL. Fusobacterium nucleatum and colorectal cancer: a review. World J Gastrointest Oncol. 2018;10:71–81. doi: 10.4251/wjgo.v10.i3.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.van Schie EH, van Amerongen R. Aberrant WNT/CTNNB1 signaling as a therapeutic target in human breast cancer: weighing the evidence. Front Cell Dev Biol. 2020;8:25. doi: 10.3389/fcell.2020.00025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Silva-García O, Valdez-Alarcón JJ, Baizabal-Aguirre VM. Wnt/β-catenin signaling as a molecular target by pathogenic bacteria. Front Immunol. 2019;10:2135. doi: 10.3389/fimmu.2019.02135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hsu T, Trojanowska M, Watson DK. Ets proteins in biological control and cancer. J of Cell Biochem. 2004;91:896–903. doi: 10.1002/jcb.20012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sizemore GM, Pitarresi JR, Balakrishnan S, Ostrowski MC. The ETS family of oncogenic transcription factors in solid tumours. Nat Rev Cancer. 2017;17:337–351. doi: 10.1038/nrc.2017.20. [DOI] [PubMed] [Google Scholar]
- 50.Toller IM, Neelsen KJ, Steger M, Hartung ML, Hottiger MO, et al. Carcinogenic bacterial pathogen Helicobacter pylori triggers DNA double-strand breaks and a DNA damage response in its host cells. Proc Natl Acad Sci U S A. 2011;108:14944–14949. doi: 10.1073/pnas.1100959108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kim JJ, Tao H, Carloni E, Leung WK, Graham DY, et al. Helicobacter pylori impairs DNA mismatch repair in gastric epithelial cells. Gastroenterology. 2002;123:542–553. doi: 10.1053/gast.2002.34751. [DOI] [PubMed] [Google Scholar]
- 52.Liechti GW, Goldberg JB. Helicobacter pylori salvages purines from extracellular host cell DNA utilizing the outer membrane-associated nuclease NucT. J Bacteriol. 2013;195:4387–4398. doi: 10.1128/JB.00388-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Druzhinin VG, Matskova LV, Demenkov PS, Baranova ED, Volobaev VP, et al. Genetic damage in lymphocytes of lung cancer patients is correlated to the composition of the respiratory tract microbiome. Mutagenesis. 2021;36:143–153. doi: 10.1093/mutage/geab004. [DOI] [PubMed] [Google Scholar]
- 54.Porschen RK, Sonntag S. Extracellular deoxyribonuclease production by anaerobic bacteria. Appl Microbiol. 1974;27:1031–1033. doi: 10.1128/am.27.6.1031-1033.1974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Okita Y, Koi M, Takeda K, Ross R, Mukherjee B, et al. Fusobacterium nucleatum infection correlates with two types of microsatellite alterations in colorectal cancer and triggers DNA damage. Gut Pathog. 2020;12:46. doi: 10.1186/s13099-020-00384-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Tahara T, Yamamoto E, Suzuki H, Maruyama R, Chung W, et al. Fusobacterium in colonic flora and molecular features of colorectal carcinoma. Cancer Res. 2014;74:1311–1318. doi: 10.1158/0008-5472.CAN-13-1865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Nougayrède J-P, Homburg S, Taieb F, Boury M, Brzuszkiewicz E, et al. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science. 2006;313:848–851. doi: 10.1126/science.1127059. [DOI] [PubMed] [Google Scholar]
- 58.Shrestha E, Coulter JB, Guzman W, Ozbek B, Hess MM, et al. Oncogenic gene fusions in nonneoplastic precursors as evidence that bacterial infection can initiate prostate cancer. Proc Natl Acad Sci U S A. 2021;118:32. doi: 10.1073/pnas.2018976118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Pleguezuelos-Manzano C, Puschhof J, Rosendahl Huber A, van Hoeck A, Wood HM, et al. Mutational signature in colorectal cancer caused by genotoxic pks+ E. coli. Nature. 2020;580:269–273. doi: 10.1038/s41586-020-2080-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wang G, Alamuri P, Maier RJ. The diverse antioxidant systems of Helicobacter pylori. Mol Microbiol. 2006;61:847–860. doi: 10.1111/j.1365-2958.2006.05302.x. [DOI] [PubMed] [Google Scholar]
- 61.Jolivet-Gougeon A, Kovacs B, Le Gall-David S, Le Bars H, Bousarghin L, et al. Bacterial hypermutation: clinical implications. J Med Microbiol. 2011;60:563–573. doi: 10.1099/jmm.0.024083-0. [DOI] [PubMed] [Google Scholar]
- 62.Shimizu T, Marusawa H, Matsumoto Y, Inuzuka T, Ikeda A, et al. Accumulation of somatic mutations in TP53 in gastric epithelium with Helicobacter pylori infection. Gastroenterology. 2014;147:407–417. doi: 10.1053/j.gastro.2014.04.036. [DOI] [PubMed] [Google Scholar]
- 63.Zhang M, Yang D, Gold B. The Adenomatous Polyposis Coli (APC) mutation spectra in different anatomical regions of the large intestine in colorectal cancer. Mutat Res. 2018;810:1–5. doi: 10.1016/j.mrfmmm.2018.04.003. [DOI] [PubMed] [Google Scholar]
- 64.Mateo J, Boysen G, Barbieri CE, Bryant HE, Castro E, et al. DNA repair in prostate cancer: biology and clinical implications. Eur Urol. 2017;71:417–425. doi: 10.1016/j.eururo.2016.08.037. [DOI] [PubMed] [Google Scholar]
- 65.Lim B, Mun J, Kim YS, Kim SY. Variability in chromatin architecture and associated DNA repair at genomic positions containing somatic mutations. Cancer Res. 2017;77:2822–2833. doi: 10.1158/0008-5472.CAN-16-3033. [DOI] [PubMed] [Google Scholar]
- 66.Shrestha E, White JR, Yu S-H, Kulac I, Ertunc O, et al. Profiling the urinary microbiome in men with positive versus negative biopsies for prostate cancer. J Urol. 2018;199:161–171. doi: 10.1016/j.juro.2017.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ahn J, Sinha R, Pei Z, Dominianni C, Wu J, et al. Human gut microbiome and risk for colorectal cancer. J Natl Cancer Inst. 2013;105:1907–1911. doi: 10.1093/jnci/djt300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Medical Microbiology, A guide to Microbial Infections: Pathogenesis, immunity, laboratory investigation and control. Churchill Livingstone Elsevier. Chapter 12. 2012. [DOI] [Google Scholar]
- 69.Mai CW, Kang YB, Pichika MR. Should a Toll-like receptor 4 (TLR-4) agonist or antagonist be designed to treat cancer? TLR-4: its expression and effects in the ten most common cancers. Onco Targets Ther. 2013;6:1573–1587. doi: 10.2147/OTT.S50838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Poh AR, Ernst M. Targeting macrophages in cancer: from bench to bedside. Front Oncol. 2018;8:49. doi: 10.3389/fonc.2018.00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Benoit M, Desnues B, Mege JL. Macrophage polarization in bacterial infections. J Immunol. 2008;181:3733–3739. doi: 10.4049/jimmunol.181.6.3733. [DOI] [PubMed] [Google Scholar]
- 72.Castello LM, Raineri D, Salmi L, Clemente N, Vaschetto R, et al. Osteopontin at the crossroads of inflammation and tumor progression. Mediators Inflamm. 2017;2017:4049098. doi: 10.1155/2017/4049098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Coussens LM, Zitvogel L, Palucka AK. Neutralizing tumor-promoting chronic inflammation: a magic bullet? Science. 2013;339:286–291. doi: 10.1126/science.1232227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–444. doi: 10.1038/nature07205. [DOI] [PubMed] [Google Scholar]
- 75.Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8:618–631. doi: 10.1038/nrc2444. [DOI] [PubMed] [Google Scholar]
- 76.Galeano Niño JL, Wu H, LaCourse KD, Kempchinsky AG, Baryiames A, et al. Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer. Nature. 2022;611:810–817. doi: 10.1038/s41586-022-05435-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 2018;8:403–416. doi: 10.1158/2159-8290.CD-17-1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084–1089. doi: 10.1126/science.aac4255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Ihle JN, Witthuhn BA, Quelle FW, Yamamoto K, Thierfelder WE, et al. Signaling by the cytokine receptor superfamily: JAKs and STATs. Trends Biochem Sci. 1994;19:222–227. doi: 10.1016/0968-0004(94)90026-4. [DOI] [PubMed] [Google Scholar]
- 80.Fu X-L, Duan W, Su C-Y, Mao F-Y, Lv Y-P, et al. Interleukin 6 induces M2 macrophage differentiation by STAT3 activation that correlates with gastric cancer progression. Cancer Immunol Immunother. 2017;66:1597–1608. doi: 10.1007/s00262-017-2052-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Guo S, Liu M, Gonzalez-Perez RR. Role of Notch and its oncogenic signaling crosstalk in breast cancer. Biochim Biophys Acta. 2011;1815:197–213. doi: 10.1016/j.bbcan.2010.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Alsaab HO, Sau S, Alzhrani R, Tatiparti K, Bhise K, et al. PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: mechanism, combinations, and clinical outcome. Front Pharmacol. 2017;8:561. doi: 10.3389/fphar.2017.00561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Lamprinaki D, Garcia-Vello P, Marchetti R, Hellmich C, McCord KA, et al. Siglec-7 mediates immunomodulation by colorectal cancer-associated Fusobacterium nucleatum ssp. animalis. Front Immunol. 2021;12:744184. doi: 10.3389/fimmu.2021.744184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Yang C, Chalasani G, Ng YH, Robbins PD. Exosomes released from mycoplasma infected tumor cells activate inhibitory B cells. PLoS One. 2012;7:e36138. doi: 10.1371/journal.pone.0036138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hoshino D, Kirkbride KC, Costello K, Clark ES, Sinha S, et al. Exosome secretion is enhanced by invadopodia and drives invasive behavior. Cell Rep. 2013;5:1159–1168. doi: 10.1016/j.celrep.2013.10.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Schwab A, Meyering SS, Lepene B, Iordanskiy S, van Hoek ML, et al. Extracellular vesicles from infected cells: potential for direct pathogenesis. Front Microbiol. 2015;6:1132. doi: 10.3389/fmicb.2015.01132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Gharaibeh RZ, Jobin C. Microbiota and cancer immunotherapy: in search of microbial signals. Gut. 2019;68:385–388. doi: 10.1136/gutjnl-2018-317220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.He Y, Huang J, Li Q, Xia W, Zhang C, et al. Gut microbiota and tumor immune escape: a new perspective for improving tumor immunotherapy. Cancers. 2022;14:5317. doi: 10.3390/cancers14215317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Liss MA, White JR, Goros M, Gelfond J, Leach R, et al. Metabolic biosynthesis pathways identified from fecal microbiome associated with prostate cancer. Eur Urol. 2018;74:575–582. doi: 10.1016/j.eururo.2018.06.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sha S, Ni L, Stefil M, Dixon M, Mouraviev V. The human gastrointestinal microbiota and prostate cancer development and treatment. Investig Clin Urol. 2020;61:S43–S50. doi: 10.4111/icu.2020.61.S1.S43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Plottel CS, Blaser MJ. Microbiome and malignancy. Cell Host Microbe. 2011;10:324–335. doi: 10.1016/j.chom.2011.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Caspi R, Billington R, Fulcher CA, Keseler IM, Kothari A, et al. The MetaCyc database of metabolic pathways and enzymes. Nucleic Acids Res. 2018;46:D633–D639. doi: 10.1093/nar/gkx935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Andreesen JR. Glycine metabolism in anaerobes. Antonie van Leeuwenhoek. 1994;66:223–237. doi: 10.1007/BF00871641. [DOI] [PubMed] [Google Scholar]
- 95.Jain M, Nilsson R, Sharma S, Madhusudhan N, Kitami T, et al. Metabolite profiling identifies a key role for glycine in rapid cancer cell proliferation. Science. 2012;336:1040–1044. doi: 10.1126/science.1218595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Locasale JW. Serine, glycine and one-carbon units: cancer metabolism in full circle. Nat Rev Cancer. 2013;13:572–583. doi: 10.1038/nrc3557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Strmiska V, Michalek P, Eckschlager T, Stiborova M, Adam V, et al. Prostate cancer-specific hallmarks of amino acids metabolism: towards a paradigm of precision medicine. Biochim Biophys Acta Rev Cancer. 2019;1871:248–258. doi: 10.1016/j.bbcan.2019.01.001. [DOI] [PubMed] [Google Scholar]
- 98.Sreekumar A, Poisson LM, Rajendiran TM, Khan AP, Cao Q, et al. Metabolomic profiles delineate potential role for sarcosine in prostate cancer progression. Nature. 2009;457:910–914. doi: 10.1038/nature07762. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
- 99.Leduc D, Escartin F, Nijhout HF, Reed MC, Liebl U, et al. Flavin-dependent thymidylate synthase ThyX activity: implications for the folate cycle in bacteria. J Bacteriol. 2007;189:8537–8545. doi: 10.1128/JB.01380-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Pegg AE. Mammalian polyamine metabolism and function. IUBMB Life. 2009;61:880–894. doi: 10.1002/iub.230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Casero RA, Murray Stewart T, Pegg AE. Polyamine metabolism and cancer: treatments, challenges and opportunities. Nat Rev Cancer. 2018;18:681–695. doi: 10.1038/s41568-018-0050-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Vuilleumier S. Bacterial glutathione S-transferases: what are they good for? J Bacteriol. 1997;179:1431–1441. doi: 10.1128/jb.179.5.1431-1441.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ricci V, Giannouli M, Romano M, Zarrilli R. Helicobacter pylori gamma-glutamyl transpeptidase and its pathogenic role. World J Gastroenterol. 2014;20:630–638. doi: 10.3748/wjg.v20.i3.630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Srinivasan V, Morowitz HJ. Ancient genes in contemporary persistent microbial pathogens. Biol Bull. 2006;210:1–9. doi: 10.2307/4134531. [DOI] [PubMed] [Google Scholar]
- 105.Antranikian G, Giffhorn F. Citrate metabolism in anaerobic bacteria. Fems Microbiol Lett. 1987;46:175–198. doi: 10.1111/j.1574-6968.1987.tb02458.x. [DOI] [Google Scholar]
- 106.Zaidi N, Swinnen JV, Smans K. ATP-citrate lyase: a key player in cancer metabolism. Cancer Res. 2012;72:3709–3714. doi: 10.1158/0008-5472.CAN-11-4112. [DOI] [PubMed] [Google Scholar]
- 107.Subramanian S, Sivaraman C. Bacterial citrate lyase. J Biosci. 1984;6:379–401. doi: 10.1007/BF02703895. [DOI] [Google Scholar]
- 108.Icard P, Lincet H. The reduced concentration of citrate in cancer cells: an indicator of cancer aggressiveness and a possible therapeutic target (vol 29, pg 47, 2016) Drug Resist Updat. 2017;30:63. doi: 10.1016/j.drup.2017.01.003. [DOI] [PubMed] [Google Scholar]
- 109.Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, et al. ATP-citrate lyase links cellular metabolism to histone acetylation. Science. 2009;324:1076–1080. doi: 10.1126/science.1164097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Koppenol WH, Bounds PL, Dang CV. Otto Warburg’s contributions to current concepts of cancer metabolism. Nat Rev Cancer. 2011;11:325–337. doi: 10.1038/nrc3038. [DOI] [PubMed] [Google Scholar]
- 111.Smith M, Zahnley J, Pfeifer D, Goff D. Growth and cholesterol oxidation by Mycobacterium species in Tween 80 medium. Appl Environ Microbiol. 1993;59:1425–1429. doi: 10.1128/aem.59.5.1425-1429.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Nesbitt NM, Yang X, Fontán P, Kolesnikova I, Smith I, et al. A thiolase of Mycobacterium tuberculosis is required for virulence and production of androstenedione and androstadienedione from cholesterol. Infect Immun. 2010;78:275–282. doi: 10.1128/IAI.00893-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Hesselink PGM, van Vliet S, de Vries H, Witholt B. Optimization of steroid side chain cleavage by Mycobacterium sp. in the presence of cyclodextrins. Enzyme Microb Technol. 1989;11:398–404. doi: 10.1016/0141-0229(89)90133-6. [DOI] [Google Scholar]
- 114.Pernigoni N, Zagato E, Calcinotto A, Troiani M, Mestre RP, et al. Commensal bacteria promote endocrine resistance in prostate cancer through androgen biosynthesis. Science. 2021;374:216–224. doi: 10.1126/science.abf8403. [DOI] [PubMed] [Google Scholar]
- 115.Kreit J, Sampson NS. Cholesterol oxidase: physiological functions. FEBS J. 2009;276:6844–6856. doi: 10.1111/j.1742-4658.2009.07378.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Cai C, Balk SP. Intratumoral androgen biosynthesis in prostate cancer pathogenesis and response to therapy. Endocr Relat Cancer. 2011;18:R175–82. doi: 10.1530/ERC-10-0339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Snaterse G, Visser JA, Arlt W, Hofland J. Circulating steroid hormone variations throughout different stages of prostate cancer. Endocr Relat Cancer. 2017;24:R403–R420. doi: 10.1530/ERC-17-0155. [DOI] [PubMed] [Google Scholar]
- 118.Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: physiological and clinical implications. Maturitas. 2017;103:45–53. doi: 10.1016/j.maturitas.2017.06.025. [DOI] [PubMed] [Google Scholar]
- 119.Dashnyam P, Mudududdla R, Hsieh T-J, Lin T-C, Lin H-Y, et al. β-Glucuronidases of opportunistic bacteria are the major contributors to xenobiotic-induced toxicity in the gut. Sci Rep. 2018;8:16372. doi: 10.1038/s41598-018-34678-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Belton M, Brilha S, Manavaki R, Mauri F, Nijran K, et al. Hypoxia and tissue destruction in pulmonary TB. Thorax. 2016;71:1145–1153. doi: 10.1136/thoraxjnl-2015-207402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Wong-Rolle A, Dong Q, Zhu Y, Divakar P, Hor JL, et al. Spatial meta-transcriptomics reveal associations of intratumor bacteria burden with lung cancer cells showing a distinct oncogenic signature. J Immunother Cancer. 2022;10:e004698. doi: 10.1136/jitc-2022-004698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wicks EE, Semenza GL. Hypoxia-inducible factors: cancer progression and clinical translation. J Clin Invest. 2022;132:11. doi: 10.1172/JCI159839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Wei R, Wong JPC, Kwok HF. Osteopontin -- a promising biomarker for cancer therapy. J Cancer. 2017;8:2173–2183. doi: 10.7150/jca.20480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Ju JA, Godet I, Ye IC, Byun J, Jayatilaka H, et al. Hypoxia selectively enhances integrin α5β1 receptor expression in breast cancer to promote metastasis. Mol Cancer Res. 2017;15:723–734. doi: 10.1158/1541-7786.MCR-16-0338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Sahlgren C, Gustafsson MV, Jin S, Poellinger L, Lendahl U. Notch signaling mediates hypoxia-induced tumor cell migration and invasion. Proc Natl Acad Sci USA. 2008;105:6392–6397. doi: 10.1073/pnas.0802047105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hildebrand D, Uhle F, Sahin D, Krauser U, Weigand MA, et al. The interplay of notch signaling and STAT3 in TLR-activated human primary monocytes. Front Cell Infect Microbiol. 2018;8:241. doi: 10.3389/fcimb.2018.00241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Aster JC, Pear WS, Blacklow SC. The varied roles of notch in cancer. Annu Rev Pathol. 2017;12:245–275. doi: 10.1146/annurev-pathol-052016-100127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Liu C, Ng S-K, Ding Y, Lin Y, Liu W, et al. Meta-analysis of mucosal microbiota reveals universal microbial signatures and dysbiosis in gastric carcinogenesis. Oncogene. 2022;41:3599–3610. doi: 10.1038/s41388-022-02377-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Riquelme E, Zhang Y, Zhang L, Montiel M, Zoltan M, et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell. 2019;178:795–806. doi: 10.1016/j.cell.2019.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Chen Y, Qiu X, Wang W, Li D, Wu A, et al. Human papillomavirus infection and cervical intraepithelial neoplasia progression are associated with increased vaginal microbiome diversity in a Chinese cohort. BMC Infect Dis. 2020;20 doi: 10.1186/s12879-020-05324-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Mitra A, MacIntyre DA, Lee YS, Smith A, Marchesi JR, et al. Cervical intraepithelial neoplasia disease progression is associated with increased vaginal microbiome diversity. Sci Rep. 2015;5:16865. doi: 10.1038/srep16865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Eddy RJ, Weidmann MD, Sharma VP, Condeelis JS. Tumor cell invadopodia: invasive protrusions that orchestrate metastasis. Trends Cell Biol. 2017;27:595–607. doi: 10.1016/j.tcb.2017.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.McCoy-Simandle K, Hanna SJ, Cox D. Exosomes and nanotubes: control of immune cell communication. Int J Biochem Cell Biol. 2016;71:44–54. doi: 10.1016/j.biocel.2015.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Onfelt B, Nedvetzki S, Benninger RKP, Purbhoo MA, Sowinski S, et al. Structurally distinct membrane nanotubes between human macrophages support long-distance vesicular traffic or surfing of bacteria. J Immunol. 2006;177:8476–8483. doi: 10.4049/jimmunol.177.12.8476. [DOI] [PubMed] [Google Scholar]
- 135.Hanna SJ, McCoy-Simandle K, Miskolci V, Guo P, Cammer M, et al. The Role of Rho-GTPases and actin polymerization during macrophage tunneling nanotube biogenesis. Sci Rep. 2017;7:8547. doi: 10.1038/s41598-017-08950-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Chen C, Chen S, Han Z, Xie W, Zhang T, et al. Patients with Helicobacter pylori-positive gastric cancer with human cytomegalovirus infection have a low tendency of advanced lymphatic metastasis in a Chinese population. Oncol Lett. 2021;21:402. doi: 10.3892/ol.2021.12663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Gao S, Li S, Ma Z, Liang S, Shan T, et al. Presence of Porphyromonas gingivalis in esophagus and its association with the clinicopathological characteristics and survival in patients with esophageal cancer. Infect Agents Cancer. 2016;11:3. doi: 10.1186/s13027-016-0049-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Tsay J-CJ, Wu BG, Sulaiman I, Gershner K, Schluger R, et al. Lower airway dysbiosis affects lung cancer progression. Cancer Discov. 2021;11:293–307. doi: 10.1158/2159-8290.CD-20-0263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Brook I. Treatment of anaerobic infection. Expert Rev Anti Infect Ther. 2007;5:991–1006. doi: 10.1586/14787210.5.6.991. [DOI] [PubMed] [Google Scholar]
- 140.El Tekle G, Garrett WS. Bacteria in cancer initiation, promotion and progression. Nat Rev Cancer. 2023;23:600–618. doi: 10.1038/s41568-023-00594-2. [DOI] [PubMed] [Google Scholar]
- 141.Ford AC, Yuan Y, Moayyedi P. Helicobacter pylori eradication therapy to prevent gastric cancer: systematic review and meta-analysis. Gut. 2020;69:2113–2121. doi: 10.1136/gutjnl-2020-320839. [DOI] [PubMed] [Google Scholar]
- 142.Ford AC, Yuan Y, Moayyedi P. Long-term impact of Helicobacter pylori eradication therapy on gastric cancer incidence and mortality in healthy infected individuals: a meta-analysis beyond 10 years of follow-up. Gastroenterology. 2022;163:754–756. doi: 10.1053/j.gastro.2022.05.027. [DOI] [PubMed] [Google Scholar]
- 143.Li D, Jiang SF, Lei NY, Shah SC, Corley DA. Effect of Helicobacter pylori eradication therapy on the incidence of Noncardia gastric adenocarcinoma in a large diverse population in the United States. Gastroenterology. 2023;165:391–401. doi: 10.1053/j.gastro.2023.04.026. [DOI] [PubMed] [Google Scholar]
- 144.Ferreri AJM, Ponzoni M, Guidoboni M, Resti AG, Politi LS, et al. Bacteria-eradicating therapy with doxycycline in ocular adnexal MALT lymphoma: a multicenter prospective trial. J Natl Cancer Inst. 2006;98:1375–1382. doi: 10.1093/jnci/djj373. [DOI] [PubMed] [Google Scholar]
- 145.Mikasa K, Sawaki M, Kita E, Hamada K, Teramoto S, et al. Significant survival benefit to patients with advanced non-small-cell lung cancer from treatment with clarithromycin. Chemotherapy. 1997;43:288–296. doi: 10.1159/000239580. [DOI] [PubMed] [Google Scholar]
- 146.Brahimi SE-H, Khelaifia S, Raoult D, Moal V. “Varibaculum massiliense” sp. nov., a new bacterial species isolated from human urine. New Microbes New Infect. 2016;13:75–76. doi: 10.1016/j.nmni.2016.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Hrbáček J, Tláskal V, Čermák P, Hanáček V, Zachoval R. Bladder cancer is associated with decreased urinary microbiota diversity and alterations in microbial community composition. Urol Oncol. 2023;41:107. doi: 10.1016/j.urolonc.2022.09.018. [DOI] [PubMed] [Google Scholar]
- 148.Gong H, Shi Y, Zhou X, Wu C, Cao P, et al. Microbiota in the throat and risk factors for laryngeal carcinoma. Appl Environ Microbiol. 2014;80:7356–7363. doi: 10.1128/AEM.02329-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Li Y, Tan X, Zhao X, Xu Z, Dai W, et al. Composition and function of oral microbiota between gingival squamous cell carcinoma and periodontitis. Oral Oncol. 2020;107:104710. doi: 10.1016/j.oraloncology.2020.104710. [DOI] [PubMed] [Google Scholar]
- 150.Nagy KN, Sonkodi I, Szöke I, Nagy E, Newman HN. The microflora associated with human oral carcinomas. Oral Oncol. 1998;34:304–308. [PubMed] [Google Scholar]
- 151.Bolz J, Dosá E, Schubert J, Eckert AW. Bacterial colonization of microbial biofilms in oral squamous cell carcinoma. Clin Oral Investig. 2014;18:409–414. doi: 10.1007/s00784-013-1007-2. [DOI] [PubMed] [Google Scholar]
- 152.Yang L, Lu X, Nossa CW, Francois F, Peek RM, et al. Inflammation and intestinal metaplasia of the distal esophagus are associated with alterations in the microbiome. Gastroenterology. 2009;137:588–597. doi: 10.1053/j.gastro.2009.04.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, et al. Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS One. 2011;6:e16393. doi: 10.1371/journal.pone.0016393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Prizment AE, Staley C, Onyeaghala GC, Vivek S, Thyagarajan B, et al. Randomised clinical study: oral aspirin 325 mg daily vs placebo alters gut microbial composition and bacterial taxa associated with colorectal cancer risk. Aliment Pharmacol Ther. 2020;52:976–987. doi: 10.1111/apt.16013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Wang Q, Ye J, Fang D, Lv L, Wu W, et al. Multi-omic profiling reveals associations between the gut mucosal microbiome, the metabolome, and host DNA methylation associated gene expression in patients with colorectal cancer. BMC Microbiol. 2020;20:83. doi: 10.1186/s12866-020-01762-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Zorron Cheng Tao Pu L, Yamamoto K, Honda T, Nakamura M, Yamamura T, et al. Microbiota profile is different for early and invasive colorectal cancer and is consistent throughout the colon. J Gastroenterol Hepatol. 2020;35:433–437. doi: 10.1111/jgh.14868. [DOI] [PubMed] [Google Scholar]
- 157.Du X, Li Q, Tang Z, Yan L, Zhang L, et al. Alterations of the gut microbiome and fecal metabolome in colorectal cancer: implication of intestinal metabolism for tumorigenesis. Front Physiol. 2022;13:854545. doi: 10.3389/fphys.2022.854545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Banerjee S, Tian T, Wei Z, Shih N, Feldman MD, et al. Distinct microbial signatures associated with different breast cancer types. Front Microbiol. 2018;9:951. doi: 10.3389/fmicb.2018.00951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Tsementzi D, Pena-Gonzalez A, Bai J, Hu Y-J, Patel P, et al. Comparison of vaginal microbiota in gynecologic cancer patients pre- and post-radiation therapy and healthy women. Cancer Med. 2020;9:3714–3724. doi: 10.1002/cam4.3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Coker OO, Dai Z, Nie Y, Zhao G, Cao L, et al. Mucosal microbiome dysbiosis in gastric carcinogenesis. Gut. 2018;67:1024–1032. doi: 10.1136/gutjnl-2017-314281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Gunathilake MN, Lee J, Choi IJ, Kim Y-I, Ahn Y, et al. Association between the relative abundance of gastric microbiota and the risk of gastric cancer: a case-control study. Sci Rep. 2019;9:13589. doi: 10.1038/s41598-019-50054-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Dai D, Yang Y, Yu J, Dang T, Qin W, et al. Interactions between gastric microbiota and metabolites in gastric cancer. Cell Death Dis. 2021;12:1104. doi: 10.1038/s41419-021-04396-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Tsay J-CJ, Wu BG, Badri MH, Clemente JC, Shen N, et al. Airway microbiota is associated with upregulation of the PI3K pathway in lung cancer. Am J Respir Crit Care Med. 2018;198:1188–1198. doi: 10.1164/rccm.201710-2118OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Kim O-H, Choi B-Y, Kim DK, Kim NH, Rho JK, et al. The microbiome of lung cancer tissue and its association with pathological and clinical parameters. Am J Cancer Res. 2022;12:2350–2362. [PMC free article] [PubMed] [Google Scholar]
- 165.Hussein AA, Elsayed AS, Durrani M, Jing Z, Iqbal U, et al. Investigating the association between the urinary microbiome and bladder cancer: an exploratory study. Urol Oncol. 2021;39:370. doi: 10.1016/j.urolonc.2020.12.011. [DOI] [PubMed] [Google Scholar]
- 166.Liu X, Shao L, Liu X, Ji F, Mei Y, et al. Alterations of gastric mucosal microbiota across different stomach microhabitats in a cohort of 276 patients with gastric cancer. EBioMedicine. 2019;40:336–348. doi: 10.1016/j.ebiom.2018.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Lam KC, Vyshenska D, Hu J, Rodrigues RR, Nilsen A, et al. Transkingdom network reveals bacterial players associated with cervical cancer gene expression program. PeerJ. 2018;6:e5590. doi: 10.7717/peerj.5590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Mikamo H, Izumi K, Ito K, Watanabe K, Ueno K, et al. Internal bacterial flora of solid uterine cervical cancer. Kansenshogaku Zasshi. 1993;67:1057–1061. doi: 10.11150/kansenshogakuzasshi1970.67.1057. [DOI] [PubMed] [Google Scholar]
- 169.Rai AK, Panda M, Das AK, Rahman T, Das R, et al. Dysbiosis of salivary microbiome and cytokines influence oral squamous cell carcinoma through inflammation. Arch Microbiol. 2021;203:137–152. doi: 10.1007/s00203-020-02011-w. [DOI] [PubMed] [Google Scholar]
- 170.Kalaora S, Nagler A, Nejman D, Alon M, Barbolin C, et al. Identification of bacteria-derived HLA-bound peptides in melanoma. Nature. 2021;592:138–143. doi: 10.1038/s41586-021-03368-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Peters BA, Wu J, Pei Z, Yang L, Purdue MP, et al. Oral microbiome composition reflects prospective risk for esophageal cancers. Cancer Res. 2017;77:6777–6787. doi: 10.1158/0008-5472.CAN-17-1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Ai D, Pan H, Han R, Li X, Liu G, et al. Using decision tree aggregation with random forest model to identify gut microbes associated with colorectal cancer. Genes. 2019;10:112. doi: 10.3390/genes10020112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Katz J, Onate MD, Pauley KM, Bhattacharyya I, Cha S. Presence of Porphyromonas gingivalis in gingival squamous cell carcinoma. Int J Oral Sci. 2011;3:209–215. doi: 10.4248/IJOS11075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ, et al. Human oral microbiome and prospective risk for pancreatic cancer: a population-based nested case-control study. Gut. 2018;67:120–127. doi: 10.1136/gutjnl-2016-312580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Walther-António MRS, Chen J, Multinu F, Hokenstad A, Distad TJ, et al. Potential contribution of the uterine microbiome in the development of endometrial cancer. Genome Med. 2016;8:122. doi: 10.1186/s13073-016-0368-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Shilnikova II, Dmitrieva NV. Evaluation of antibiotic susceptibility of bacteroides, prevotella and Fusobacterium species isolated from patients of the N. N. Blokhin Cancer Research Center, Moscow, Russia. Anaerobe. 2015;31:15–18. doi: 10.1016/j.anaerobe.2014.08.003. [DOI] [PubMed] [Google Scholar]
- 177.Sfanos KS, Sauvageot J, Fedor HL, Dick JD, De Marzo AM, et al. A molecular analysis of prokaryotic and viral DNA sequences in prostate tissue from patients with prostate cancer indicates the presence of multiple and diverse microorganisms. Prostate. 2008;68:306–320. doi: 10.1002/pros.20680. [DOI] [PubMed] [Google Scholar]
- 178.Justesen US, Nielsen SL, Jensen TG, Dessau RB, Møller JK, et al. Bacteremia with anaerobic bacteria and association with colorectal cancer: a population-based cohort study. Clin Infect Dis. 2022;75:1747–1753. doi: 10.1093/cid/ciac259. [DOI] [PubMed] [Google Scholar]
- 179.Kartal E, Schmidt TSB, Molina-Montes E, Rodríguez-Perales S, Wirbel J, et al. A faecal microbiota signature with high specificity for pancreatic cancer. Gut. 2022;71:1359–1372. doi: 10.1136/gutjnl-2021-324755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Park SS, Kim B, Kim MJ, Roh SJ, Park SC, et al. The effect of curative resection on fecal microbiota in patients with colorectal cancer: a prospective pilot study. Ann Surg Treat Res. 2020;99:44–51. doi: 10.4174/astr.2020.99.1.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Cobo F. Lymphocele infection due to Peptoniphilus harei after radical prostatectomy. Med Mal Infect. 2018;48:154–155. doi: 10.1016/j.medmal.2017.10.003. [DOI] [PubMed] [Google Scholar]
- 182.Wu P, Zhang G, Zhao J, Chen J, Chen Y, et al. Profiling the urinary microbiota in male patients with bladder cancer in China. Front Cell Infect Microbiol. 2018;8:167. doi: 10.3389/fcimb.2018.00167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Gao X, Miao R, Zhu Y, Lin C, Yang X, et al. A new insight into acute lymphoblastic leukemia in children: influences of changed intestinal microfloras. BMC Pediatr. 2020;20:290. doi: 10.1186/s12887-020-02192-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Mira-Pascual L, Cabrera-Rubio R, Ocon S, Costales P, Parra A, et al. Microbial mucosal colonic shifts associated with the development of colorectal cancer reveal the presence of different bacterial and archaeal biomarkers. J Gastroenterol. 2015;50:167–179. doi: 10.1007/s00535-014-0963-x. [DOI] [PubMed] [Google Scholar]
- 185.Mima K, Nishihara R, Qian ZR, Cao Y, Sukawa Y, et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut. 2016;65:1973–1980. doi: 10.1136/gutjnl-2015-310101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Flanagan L, Schmid J, Ebert M, Soucek P, Kunicka T, et al. Fusobacterium nucleatum associates with stages of colorectal neoplasia development, colorectal cancer and disease outcome. Eur J Clin Microbiol Infect Dis. 2014;33:1381–1390. doi: 10.1007/s10096-014-2081-3. [DOI] [PubMed] [Google Scholar]
- 187.Warren RL, Freeman DJ, Pleasance S, Watson P, Moore RA, et al. Co-occurrence of anaerobic bacteria in colorectal carcinomas. Microbiome. 2013;1:16. doi: 10.1186/2049-2618-1-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Audirac-Chalifour A, Torres-Poveda K, Bahena-Román M, Téllez-Sosa J, Martínez-Barnetche J, et al. Cervical microbiome and cytokine profile at various stages of cervical cancer: a pilot study. PLoS One. 2016;11:e0153274. doi: 10.1371/journal.pone.0153274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Hieken TJ, Chen J, Hoskin TL, Walther-Antonio M, Johnson S, et al. The microbiome of aseptically collected human breast tissue in benign and malignant disease. Sci Rep. 2016;6:30751. doi: 10.1038/srep30751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Al-Hebshi NN, Nasher AT, Maryoud MY, Homeida HE, Chen T, et al. Inflammatory bacteriome featuring Fusobacterium nucleatum and Pseudomonas aeruginosa identified in association with oral squamous cell carcinoma. Sci Rep. 2017;7:1834. doi: 10.1038/s41598-017-02079-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Zhao H, Chu M, Huang Z, Yang X, Ran S, et al. Variations in oral microbiota associated with oral cancer. Sci Rep. 2017;7 doi: 10.1038/s41598-017-11779-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Lee JB, Kim K-A, Cho HY, Kim D, Kim WK, et al. Association between Fusobacterium nucleatum and patient prognosis in metastatic colon cancer. Sci Rep. 2021;11:20263. doi: 10.1038/s41598-021-98941-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



