Skip to main content
Erschienen in: Current Oral Health Reports 2/2019

Open Access 18.04.2019 | Microbiology (C Genco, Section Editor)

The Microbiome of Oral Squamous Cell Carcinomas: a Functional Perspective

verfasst von: Nezar N. Al-Hebshi, Wenche S. Borgnakke, Newell W. Johnson

Erschienen in: Current Oral Health Reports | Ausgabe 2/2019

Abstract

Purpose of Review

This decade has witnessed increasing interest in the potential role of the oral microbiome in head and neck cancers, particularly oral squamous cell carcinoma (OSCC). Most studies have focused on the bacterial component of the microbiome (bacteriome), but the fungal component (mycobiome) is also receiving attention. In this review, we provide an overview of mechanisms by which the microbiome can contribute to oral carcinogenesis, and summarize results from clinical studies, especially focusing on those reporting functional microbiome analysis. Synthesizing and illustrating the evidence, we also suggest a new “passenger-turning-driver” functional model for the role of the microbiome in oral cancer.

Recent Findings

In vitro studies provide convincing evidence for the carcinogenicity of the periodontal bacteria Fusobacterium nucleatum and Porphyromonas gingivalis. However, results from clinical studies are inconsistent, with significant variations in composition of the microbiome associated with oral cancer. Methodological differences may partially explain the differing conclusion. However, variations observed may also reflect functional redundancy: the phenomenon that different species may be enriched in different samples, but still serve the same functions. Indeed, functional analyses of the bacteriome associated with oral cancer have revealed more consistent results, namely enrichment of a virulent, inflammatory bacteriome in the tumors.

Summary

Apart from oncoviruses associated with a special entity of oral cancer, no consistent evidence implicates specific microbial species in OSCC etiology. Instead, the disturbed function of an initially “passenger” microbiome within the tumor microenvironment likely contributes to tumor progression by sustaining chronic inflammation.
Hinweise
This article is part of the Topical Collection on Microbiology

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Oral cancer (predominantly squamous cell carcinoma) is a subset of head and neck cancers (HNCs) affecting the oral cavity proper, i.e., mouth anterior to the palatine tonsils, also referred to as intra-oral. In the international databases, oral cancer is classified as “lip and oral cavity”: ICD-10 sites C00-C06. These constitute the 16th most prevalent malignancy worldwide, accounting for an estimated 247,563 new cases and 177,384 deaths annually [1]. There is, however, marked geographical and cultural variation. In much of South Asia, for example, oral cancer is the most common cancer among males, perhaps sixth among women, and second overall [2]. In the USA, the corresponding numbers are ~ 35,130 and 7410, respectively, although the anatomical sub-sites do not precisely match those used in the worldwide data [3]. There is a male predilection and the tongue is the most commonly affected sub-site [3]. In the West, nearly 74% of oral squamous cell carcinoma (OSCC) cases are attributed to tobacco smoking and heavy alcohol consumption [4], while in South Asia and the Pacific, smokeless tobacco and areca (betel) nut chewing are the major risk factors [5]. Unlike cancers of the oropharynx, only a small fraction of OSCC cases (2–6%) are attributed to human papilloma virus (HPV) infection [6, 7]—although greater proportions are reported from the Asia-Pacific Region—but there are questions about definitions of head and neck sub-sites and laboratory methods [8]. Around 15% of all OSCC cases remain unexplained by any of the known risk factors. In addition, and despite advances in cancer treatment modalities, OSCC continues to have poor prognosis with 5-year survival rates less than 50% in much of the world [9, 10]. These challenges have triggered scientists to search for novel risk factors and prognosis modifiers that eventually could present targets for preventive or therapeutic interventions. Particularly, there has been increasing interest in the role of the oral microbiome in oral carcinogenesis [11].
Apart from the debate on its origin [12], the term “microbiome” is currently used to refer to “all micro-organisms in a particular habitat and their collective genomes” [13]. The microbiomes associated with the human body, including the oral microbiome, and their role in health and disease have been studied extensively [14]. Due to differences in key ecological factors, such as redox potential, attachment legends, moisture level, acidity, etc., there are significant variations in the composition of the microbiome from one body site to another, and even between sub-sites in close proximity, e.g., sub- and supra-gingival plaque [15]. The human microbiomes form complex, but balanced (homeostatic), communities that are compatible with health, a status increasingly referred to as normobiosis. Under certain circumstances, however, this balance may be disturbed, leading to perturbations in the composition and function of the microbiome (dysbiosis) that are associated with transition from health to disease [16]. In the oral cavity, dental caries and periodontitis are typical examples of diseases associated with microbial dysbiosis. Bacteria are the predominant microorganisms in the oral cavity and therefore most research so far has focused on the bacterial component of the oral microbiome—the oral bacteriome. However, there is increasing interest in less abundant microbial communities, such as those involving fungi, the oral mycobiome [17, 18], and viruses, the oral virome [19], respectively, and their potential role in oral diseases. The advent of high-throughput molecular technologies, especially next-generation sequencing (NGS), has revolutionized the study of microbial communities. An overview of nucleic acid sequencing-based methods currently used to study the oral microbiome is provided in Fig. 1.
Evidence is emerging for the oral microbiome playing a role in oral cancer, a topic we reviewed comprehensively in 2016 [11]. Nonetheless, there has been a virtual explosion in research in this area (at least 18 additional studies since that report), with evolving concepts regarding the nature of contributions by the oral microbiome to oral carcinogenesis, which consequently warrants revisiting the topic. In this concise review, we summarize results from clinical studies on the bacteriome and mycobiome associated with oral cancer, and provide an overview of carcinogenic properties of some oral microbes. We also propose a new functional model for the role of the microbiome in oral carcinogenesis.

Carcinogenicity of Specific Oral Microbes

The Classical Microbial Suspects: Viruses

The vast majority of microbes designated as Class 1 carcinogens by the International Agency for Research on Cancer (IARC) are viruses. Therefore, viruses come to mind first when raising a question regarding the role of microorganisms in cancer. As far as HNCs are concerned, two families of oncogenic viruses are particularly important: Human Papillomaviruses (HPVs) and Human Herpesviruses (HHVs). There is currently incontrovertible evidence that a small number of so-called high-risk (hr) HPVs are responsible for a global epidemic of oropharyngeal cancer. By now, cancer of the oropharynx has replaced cancer of the uterine cervix as the most common HPV-related cancers in the USA [20]. A smaller proportion of intra-oral cancers are also caused by hrHPVs. Although these HPVs are epitheliotropic, cancerous lesions mostly arise in the mucosa associated with lymphoid tissue, due to interactions between lymphocytes and keratinocytes, namely in the posterior third of the tongue (such lesions located in the palatine tonsils and elsewhere in Waldeyer’s tonsillar ring are not classified as intra-oral). The underlying cellular mechanisms are well understood: HPV E6 and HPV E7 oncogenes interfere with p53 and retinoblastoma gene proteins, respectively, and thereby block their tumor suppressor actions [21]. Consequently, there are encouraging possibilities to block the oncogenic pathways with interfering ribonucleic acid (RNA) or by gene editing [22].
Among the HHVs, the Epstein-Barr virus (HHV-4) is the major cause of nasopharyngeal cancers, a distinct biological entity [23]. However, there is limited evidence that HHV-4 may play a role in oral cancer [21, 24]. HHV-8 is the etiologic agent for Kaposi’s sarcoma located in the mouth and elsewhere that is prevalent in immunosuppressed individuals, particularly in patients with acquired immune deficiency syndrome (AIDS) [21, 25]. The virus is carried in the oropharynx and can be recovered from saliva/oral fluid samples [21, 26]. There is circumstantial evidence that Herpes Simplex Viruses (HSV), both HSV-1 and HSV-2, are associated with oral (and cervical) squamous cell carcinoma in humans, as well as supportive animal studies [21], but a direct oncogenic role remains unsubstantiated [26].
Apart from human viruses, the oral cavity is also home to a complex community of bacteriophages that are thought to influence the ecology and pathogenicity of the oral bacterial community [27]. Whether or not this phage community plays a role in oral carcinogenesis has not been explored.

Emerging Role of Porphyromonas gingivalis and Fusobacterium nucleatum

In the 1990s, the relationship between bacteria and carcinogenesis was first established by demonstrating the causative role of Helicobacter pylori in gastric cancer [28], and since then, tremendous efforts have been invested in exploring the relationship between bacteria and cancer in sites elsewhere in the body. This has led to uncovering additional associations, such as that of Chlamydia trachomatis with cervical cancer [29], Salmonella typhi with gallbladder cancer [30], and Bacteroides fragilis with colon cancer [31]. As far as oral cancer is concerned, evidence is emerging, primarily from in vitro and animal studies, for the carcinogenicity of the periodontal bacteria Porphyromonas gingivalis (P. gingivalis) and Fusobacterium nucleatum (F. nucleatum). Interest in these two bacteria from the oral microbiome as potential carcinogens has been fueled by studies that implicated them in pancreatic and colorectal cancers (CRC), respectively [3235]. The mechanisms by which they are thought to contribute to oral carcinogenesis are shown conceptually in Fig. 2 [11].
P. gingivalis has been shown to inhibit apoptosis at different levels, including activation of JAK1/STAT3 and PI3K/Akt signaling pathways [36, 37], suppression of proapoptotic BCL-2-associated death promoter [38], blocking activity of caspase-3 and caspase-9 [37, 38], upregulation of microRNA-203 [39], and prevention of ATP-dependent P2X7-mediated apoptosis [40]. Both P. gingivalis and F. nucleatum activate cell proliferation through upregulation of kinases and cyclins [4143], activation of the β-catenin signaling pathway [44, 45], and downregulating level of the p53 tumor suppressor [41]. They also have been found to enhance cellular invasion, primarily through upregulation/activation of matrix metalloproteinases, including MMP-1, MMP-9, MMP-10, and MMP-13, and inducing stemness and epithelial to mesenchymal transition (EMT) [43, 4648]. In addition, P. gingivalis and F. nucleatum are believed to contribute to progression of oral cancer by inducing chronic inflammation via increasing production of pro-inflammatory cytokines [4952]. Production of carcinogenic substances, such as acetaldehyde (from ethanol), may also play an important role, but has not been documented for these two species. Nonetheless, there is evidence for such production by other oral microorganism species, such as Streptococci [53], Neisseria [54], and Candida [55, 56].
Despite the convincing evidence from in vitro studies on the carcinogenicity of P. gingivalis and F. nucleatum just summarized, evidence for a direct carcinogenic role in oral cancer based on clinical studies is still lacking, as described in the following section.

The Bacteriome Associated with Oral Cancer: Clinical Studies

Variations in Composition of the Bacteriome Associated with Tumors

Clinical studies that assessed the association between bacteria and cancers of the oral cavity are summarized in Table 1. Most of these studies restricted inclusion to OSCC samples, but a number of them also included HNCs located at sites other than the oral cavity (parts of the pharynx, larynx, or esophagus) as well as lesions that were only potentially malignant. Only two decades ago, namely in 1998, Nagy and collaborators first demonstrated differences in composition of the microbial community colonizing the surface of OSCC and adjacent normal tissue, using culture techniques [57•]. Between 2000 and 2005, Streptococcus anginosus (S. anginosus) became implicated in the etiology of HNC, including OSCC, by research groups from Japan [58, 59, 61]. However, these studies suffered from lack of proper control samples: the high detection rates of the bacterium observed in HNC samples were simply because it is a normal colonizer of the oropharynx. Interest in S. anginosus thus faded quickly.
Table 1
Clinical studies assessing the association between bacteria and oral cancer (in chronologic order) (an expansion of Table 1 in the 2016 review by Perera et al. [11], with permission)
Study
Sample size (tumor/control)*
Technology used
Case sample
Control sample
Bacterial taxa associated with the tumors
Bacterial functions associated with the tumors
Nagy et al. 1998 [57•], Hungary
21/21 (self)
Cultivation; biochemical identification
Surface swabs: tumors
Surface swabs: contagious normal mucosa
Fusobacterium, Porphyromonas, Actinomyces, and Propionibacterium spp.
N/A
Tateda et al. 2000 [58], Japan
205/10 (self)
OSCC 68
Others 137
Detection of Streptococcus anginosus by PCR and Southern-blot
Tumor tissue, gingival smears, and oropharyngeal swabs
Fresh contagious normal tissue
Authors concluded S. sanginosus was associated, but conclusion is not supported by the results (lack of proper control)
N/A
Morita et al. 2003 [59], Japan
OSCC 38/7
Esophageal 18/6
Detection of S. anginosus by q-PCR
Fresh tumor tissue
Fresh non-cancerous tissue
S. anginosus with esophageal, but not oral, cancer
N/A
Mager et al. 2005 [60], USA
45/229
Checkboard DNA-DNA hybridization
Unstimulated saliva
Unstimulated saliva
Capnocytophaga gingivalis, Prevotella melaninogenica, Streptococcus mitis
N/A
Sasaki et al. 2005 [61] Japan
42/7
Detection of S. anginosus by PCR
Fresh tumor tissue, dental plaque, and saliva
Fresh tissue: leukoplakia, lymphoma, and rhabdomyosarcoma
Authors concluded S. anginosus was associated, but conclusion is not supported by the results (lack of proper control)
N/A
Hooper et al. 2006 [62•], UK
20/12 (self)
Cultivation; isolates identified with 16S rRNA gene sequencing (Sanger)
Fresh tumor tissue: deep and superficial
Fresh contagious normal tissue
Micrococcus luteus, Prevotella melaninogenica, Exiguo-bacterium oxidotolerans, Fusobacterium naviforme, Staphylococcus aureus, Veillonella parvula
N/A
Hooper et al. 2007 [63], UK
10/10 (self)
16S rRNA clone sequencing (Sanger)
Fresh tumor tissue
Fresh contagious normal tissue
Ralstonia insidiosa, Fusobacterium naviforme, Peptostreptococcus micros, Clavibacter michiganensis subsp. tessellarius, Capocytophaga sp. oral strain S3, Prevotella sp. oral clone BE073
N/A
Katz et al. 2011 [64], USA
10/5
Immunohistochemical staining
FFPE gingival carcinoma tissue
FFPE normal gingival tissue
Porphyromonas gingivalis
N/A
Pushalkar et al. 2011 [65], USA
3/2
16S rRNA (V4–5) amplicon sequencing (454); DGGE
Stimulated saliva
Stimulated saliva
Genera Streptococcus, Rothia, Gemella, Peptostreptococcus, Lactobacillus, Micromonas, and Porphyromonas
N/A
Pushalkar et al. 2012 [66], USA
10/10 (self)
16S rRNA clone sequencing (Sanger); DGGE
Fresh tumor tissue
Fresh contagious normal tissue
Parvimonas sp. oral taxon 110, Eubacterium infirmum, Eubacterium brachy, Gemella haemolysans, Gemella morbillorum, Gemella sanguinis, Johnsonella ignava, Peptostreptococcus stomatis, Streptococcus gordonii, and Streptococcus parasanguinis I
N/A
Schmidt et al. 2014 [67], USA
16/8/6
(OSCC/“pre-cancer”/
healthy)
16S rRNA (V4) amplicon sequencing (454 and Miseq)
Surface swabs: tumors and “pre-cancer” lesions
Surface swabs: contralateral normal; normal mucosa from healthy subjects
Genera Fusobacterium and Prevotella (Streptococcus and Rothia showed inverse association)
N/A
Al-Hebshi et al. 2015 [68], Yemen
3/0
16S rRNA (V1–3) amplicon sequencing (454)
Fresh tumor tissue
None
Bacteroides fragilis?
N/A
Guerrero-Preston et al. 2016 [69], USA
17/25
OSCC 6
Oropharyngeal 11
16S rRNA (V3-V5) amplicon sequencing (454)
Oral rinse
Oral rinse
Genera Streptococcus, Lactobacillus, Staphylococcus, and Parvimonas—also Fusobacterium by G-test (Haemophilus and Neisseria among others showed inverse association)
N/A
Hu et al. 2016 [70], China
16/10/19 (OSCC/“pre-cancer”/healthy)
16S rRNA amplicon sequencing (Miseq)
Unstimulated saliva
Unstimulated saliva
Genera Bacillus (Streptococcus and Abiotrophia showed inverse association)
N/A
Al-Hebshi et al. 2017 [71•], Yemen/Saudi Arabia
20/20
16S rRNA (V1-V3) amplicon sequencing (Miseq)
Fresh tumor tissue
Deep epithelium swabs
Fusobacterium nucleatum, Pseudomonas aeruginosa, Campylobacter oral taxon 44, Haemophilus influenza, Campylobacter showae, and Parvimonas micra (Streptococcus mitis, Rothia mucilaginosa, and Haemophilus parainfluenzae among others showed inverse association)
As predicted with PICRUSt: bacterial mobility, flagellar assembly, bacterial chemotaxis, and LPS biosynthesis (DNA repair, glycolysis/gluconeogenesis and biosynthesis of amino acids were enriched in health)
Amer et al. 2017 [72], Ireland
36/32 (Leukoplakia/healthy)
No OSCC
16S rRNA (V1–V2) amplicon sequencing (Miseq)
Surface swabs: leukoplakia patients
Surface swabs: contralateral normal; normal mucosa from healthy subjects
Genera Fusobacterium, Leptotrichia, and Campylobacter; Rothia mucilaginosa
N/A
Börnigen et al. 2017 [73], USA
112/242
OSCC 43
Oropharyngeal 64
Unknown 5
16S rRNA (V4) amplicon sequencing (Miseq)
Oral rinse
Oral rinse
Genus Dialister only (Streptococcus and Rothia showed inverse association)
Results ambiguous: discrepancies between the figure and text
Guerrero-Preston et al. 2017 [74], USA
As above + 154 subjects from the HMP
Same dataset above re-analyzed, along with HMP data, with Resphera Insight
Oral rinse
Oral rinse
Fusobacterium nucleatum, Streptococcus mutans, Parvimonas micra, and Lactobacillus spp.
N/A
Lee et al. 2017 [75], Taiwan
125/124/127
(OSCC/“pre-cancer”/healthy)
16S rRNA (V4) amplicon sequencing (Miseq)
Unstimulated saliva
Unstimulated saliva
Genera Bacillus, Enterococcus, Parvimonas, Peptostreptococcs, and Slackia
N/A
Mukherjee et al. 2017 [76], USA
39/39 (self) All mobile tongue cancer
16S rRNA (V4) amplicon sequencing (Ion Torrent)
Fresh-frozen tumor tissue
Fresh-frozen adjacent normal tissue
Genera Streptococcus, Rothia, Actinomyces, Enterococcus, and Microococcus (Fusobacterium, Prevotella, Parvimonas, Campylobacter, and Porphyromonas showed inverse association)
N/A
Shin et al. 2017 [77], USA
34/24 (self)
OSCC 4
Others 30
16S rRNA (V4) amplicon sequencing (Ion Torrent)
Fresh-frozen tumor tissue: primary tumor or/and metastatic
Fresh-frozen adjacent normal tissue
No significant difference for OSCC analyzed separately.
When all tumors pooled: an increase in Fusobacterium and Parvimonas; a decrease in Streptococcus, Veillonella, Actinomyces, and Rothia
N/A
Wang et al. 2017 [78], USA
121/121 (self)
OSCC 59
Others 62
16S rRNA clone sequencing (Sanger)
Fresh-frozen tumor tissue
Fresh-frozen adjacent normal tissue
Genus Parvimonas only (Actinomyces was inversely associated)
N/A
Zhao et al. 2017 [79], China
40/40 (self)
16S rRNA (V4–V5) amplicon sequencing (Miseq)
Surface swabs: tumors
Surface swabs: self- matched normal mucosa
Genera Fusobacterium, Capnocytophaga, Alloprevotella, Treponema, Campylobacter, Parvimonas, and Dialister (Streptococcus, Veillonenlla, Lautropia, and Rothia showed inverse association)
As predicted with PICRUSt: Translation, metabolism of cofactors and vitamins; metabolism of terpenoids and polyketides; replication and repair
Hayes et al. 2018 [80•], USA
129/254
OSCC 41
Oropharyngeal 30
Larynx 58
Nested case-control study in two larger cohort studiesa
16S rRNA (V3–V4) amplicon sequencing (454)
Oral mouthwash samples (collected at baseline)
Oral mouthwash sample (collected at baseline)
Pooling all cases and controls: genera Kingella and Corynebacterium and species Prevotella nanceiensis, Capnocytophaga leadbetteri and Selenomonas sputigena were associated with decreased risk of HNSCC (Parvimonas micra and Neisseria sicca were associated with reduced risk of OSCC)
N/A
Lim et al. 2018 [81], Australia
52/21
OSCC 15
Oropharyngeal 37
Healthy subdivided into low and high risk
16S rRNA (V6–8) amplicon sequencing (Miseq)
Oral rinse
Oral rinse
Pooling all cases: genus Oribacterium only (Rothia, Haemophilus, Corynebacterium, Paludibacter, Porphyromonas, and Capnocytophaga were inversely associated)
N/A
Perera et al. 2018 [82], Sri Lanka
25/27
16S rRNA (V1–V3) amplicon sequencing (Miseq)
Fresh tumor tissue
Fresh tissue of fibroepithelial polyp
Campylobacter concisus, Prevotella salivae, Prevotella loeschii, and Fusobacterium oral taxon 204 (Streptococcus mitis, 6 other Streptococcus spp., Rothia spp. and Lautropia mirabilis among others showed inverse association)
As predicted with PICRUSt: LPS biosynthesis, peptidases and carbon fixation in photosynthetic organisms among others (Base excision repair, glycolysis/gluconeogenesis, and biosynthesis of amino acids were enriched in health)
Yang C et al. 2018 [83•], Taiwan
197/51
(41 stage 1 + 66
stage 2/3 + 90 stage 4)b
16S rRNA (V3–V4) amplicon sequencing (Miseq)
Oral rinse
Oral rinse
Fusobacterium periodonticum, Parvimonas micra, Streptococcus constellatus, Haemophilus influenza, and Filifactor alocis (in contrast to decrease of Streptococcus mitis, Haemophilus parainfluenzae, Porphyromonas pasteri, Veillonella parvula)—strength of association with staging
As predicted with PICRUSt (in stage 4 vs. health): cytoskeleton proteins, methane metabolism, carbon fixation in photosynthetic organisms, restriction enzymes, others (amino acid synthesis and metabolism were enriched in health)
Yang S et al. 2018 [84], Taiwan
39/0
Cases clustered into 3 groups by mutational signature:
MSC1 (n = 9)
MSC2(n = 10)
MSC3 (n = 11)
16S rRNA (V4) amplicon sequencing (Miseq)
Unstimulated saliva
None
N/A (no control group to compare with: comparisons were made between the three patient clusters)
N/A (no control group to compare with: comparisons were made between the three patient clusters)
Yost et al. 2018 [85••], USA
4/4
Metatranscriptome (mRNA) sequencing (NextSeq 500)
Surface swabs: tumors
Surface swabs: adjacent normal mucosa from the cancer patient; tumor-matching and buccal sites from the healthy subjects
Genera Fusobacteria, Selenomonas, Capnocytophaga, Dialister, and Johnsonella (genus Bacillus; species Porphyromonas catoniae, Kingella denitricans, Capnocytophaga gingivalis, among others, were associated with healthy, tumor-matching sites)
Iron ion transport; tryptophanase activity; peptidases; superoxide dismutase; capsule biosynthesis; flagellum synthesis and assembly; chemotaxis; hemolysins and adhesins (base excision repair only was enriched in health)
*OSCC/healthy, unless otherwise indicated
DGGE denaturing gradient gel electrophoresis, FFPE formalin-fixed paraffin-embedded, HMP Human Microbiome Project, N/A not available, HNSCC head and neck squamous cell carcinoma, LPS lipopolysaccharide, MSC mutational signature cluster, OSCC oral squamous cell carcinoma, PCR polymerase chain reaction, PICRUSt phylogenetic investigation of communities by reconstruction of unobserved states, pt(s) patient(s), rRNA ribosomal ribonucleic acid, sp(p) species
aThe American Cancer Society Cancer Prevention Study II Nutrition Cohort (CPS-II) (n = 58) + the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO) (n = 71)
bAmerican Joint Committee on Cancer. AJCC cancer staging manual [86]
In 2006, Hooper et al. demonstrated, for the first time, the presence of a viable complex bacterial community within the OSCC tissues that is compositionally different from that found in the tumors’ healthy margins [62•]. Since then, the vast majority of investigations have employed sequencing of the 16S rRNA (ribosomal RNA) gene to characterize the microbiomes in study samples, initially with the Sanger method and more recently with NGS chemistries [63, 6570, 71•, 7279, 80•, 81, 82, 83•, 84]. Nevertheless, there are significant methodological differences between these studies in terms of (1) the type of samples analyzed (saliva (stimulated or unstimulated), surface swabs, or biopsies); (2) the nature of control or comparison samples used (tumor-adjacent, clinically normal; contralateral to tumor; healthy subject; or benign lesions); (3) the hypervariable region of the 16S rRNA gene selected for sequencing (e.g., V1–V3 or V4–V5); and (4) the bioinformatic analysis method used for analysis of sequencing data (sequence quality control, reference database, and taxonomy assignment algorithm). The latter particularly affects taxonomic resolution, with only a few studies reporting species-level profiles and the rest limited to the genus level.
As displayed in Table 1, a number of bacterial taxa, particularly Fusobacterium spp., and to a lesser extent, Campylobacter, Parvimonas, and Prevotella spp., were repeatedly found to be significantly enriched in OSCC samples (Table 1), whereas Streptococci frequently were found in association with health. Nevertheless, a more careful examination of the results from these studies reveals there are significant variations (and sometimes contradictions) in the composition of the bacteriome associated with tumors from one cohort to another, and even from one subject to another within the same cohort, especially at the species level. Overall, there is, therefore, not sufficient evidence to implicate specific bacterial species or any consortium thereof in the etiology of OSCC. Interestingly, only one of these studies identified P. gingivalis in association with oral cancer [64].

More Consistent Results Obtained with Functional Analysis

While the observed variation in composition of the bacteriome associated with oral cancer may, at least in part, be attributed to the methodological differences among studies described above, it may also be explained by the fact that different species frequently can serve the same functions in their communities and thus substitute for each other, a phenomenon called functional redundancy [87••]. That is, a microbial community with a certain subset of species enriched may perform the same function as another community with a different subset of overabundant species. This concept can be likened to player substitution in team sports like soccer. Indeed, functional analyses of bacteriomes associated with oral cancer produced more consistent results than those obtained with compositional profiling reflecting only the abundance/presence of specific individual members of the bacteriome. Some of the studies listed in Table 1 have used Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) to infer metagenomes from the 16S rRNA profiles obtained (functional prediction) [71•, 79, 82, 83•]. Furthermore, in 2018 and for the first time, Yost and team used metatranscriptome sequencing to explore the transcriptional activity (gene expression) of the microbiome associated with OSCC [85••]. In these studies, the tumor-associated bacteriomes possessed similar functional signatures despite variation in their compositional profiles. For example, enrichment of primarily pro-inflammatory features, such as LPS biosynthesis, flagella assembly, bacterial chemotaxis, and production of peptidases, were enriched in the tumors, while activities like glycolysis and gluconeogenesis, amino acid synthesis and metabolism, and DNA repair were enhanced in the healthy samples. The study by Yost and colleagues identified additional virulence factors associated with OSCC, including increased expression of tryptophanase, superoxide dismutase, hemolysins, and adhesins [85••]. More details about the microbiome functional features associated with OSCC identified in this study are shown in Fig. 3 [85••].

The Mycobiome Associated with Oral Cancer

The role of members of the oral microbiome other than bacteria, namely fungi, has been studied in conjunction with OSCC as well as potentially malignant oral lesions (Table 2). Three decades ago, Krogh’s group assessed the nitrosation potential of Candida strains isolated from oral leukoplakia lesions, and found strains with high potential for nitrosation to be associated with greater levels of dysplasia [88]. A series of studies published between 1994 and 2013 used periodic Acid-Schiff (PAS) staining and cultivation techniques to detect/isolate yeasts in mucosal swabs or biopsies of oral dysplasia or OSCC [8993]. Two studies also performed culturing and counting of colony-forming unit (CFU) in oral rinse samples [91, 93]. Most of these studies, however, did not include healthy controls, but instead used for comparison other mucosal lesions, such as hyperkeratosis, benign fibrous growths, and lichen planus. Overall, these studies found the detection frequency and counts of yeasts (predominantly Candida) to be significantly greater in dysplasia and OSCC, and such elevation correlated with the severity of dysplasia.
Table 2
Epidemiological studies assessing the association between fungi and oral cancer/potentially malignant lesions
Study
Sample size (tumor/control)*
Technology used
Case samples
Control samples
Key findings
Krogh et al. 1987 [88], Denmark
12/12 (self) Leukoplakia only
Cultivation for isolation
HPLC for measuring nitrosation potential
Surface swabs: lesion surface
Surface swabs: adjacent normal mucosa
Candida strains with high nitrosation potential found in lesions with advanced “precancerous” changes
Rindum et al. 1994 [89], Denmark
53/100
  Leukoplakia 32
  CEOC 21
  No OSCC
PAS stain for cytological examination
Cultivation and biochemical assays for isolation, identification, and typing
Smears and swabs: lesion surface
Smears and surface swabs: adjacent normal mucosa and normal mucosa (cheek, tongue, and palate) from healthy subjects
Greater detection rates (predominantly C. albicans) in pathological lesions than in normal mucosa
Rare types of C. albicans tended to be associated with dysplasia and non-homogenous leukoplakia
Barrett et al. 1998 [90], UK
4724 mucosal biopsies:
  OSCC 424
  Dysplasia 597
  Other lesions 3703
PAS stain for detection of fungal hyphae
Mucosal biopsy
N/A
Greater detection rates in moderate and severe epithelial dysplasia, median rhomboid glossitis, squamous papillomas, and progressive dysplasia
No association with OSCC
Nagy et al. 1998 [57], Hungary
21/21 (self)
Cultivation; biochemical identification
Surface swabs: tumors
Surface swabs: contagious normal mucosa
C. albicans
McCullough et al. 2002 [91], UK
223 subjects:
  OSCC and dysplasia combined 103
  Other mucosal lesions 120
PAS stain for detection of fungal hyphae; cultivation for isolation and CFU count
Mucosal biopsy; mouth rinse
N/A
Significantly higher prevalence and counts in subjects with OSCC and dysplasia
Counts correlated with severity of dysplasia and presence of OSCC
Spolidorio et al. 2003 [92], Brazil
832 mucosal biopsies:
  OSCC 109
  Dysplasia 315
  Other lesions 408
PAS stain for detection of fungal hyphae
Mucosal biopsy
N/A
Greater detection rates in epithelial dysplasia, OSCC, and hyperkeratosis
Hebbar et al. 2013 [93], India
50 subjects:
  OSCC 9
  Dysplasia 20
  Other lesions 21
PAS stain for detection of fungal hyphae
Cultivation for isolation and CFU count
Mucosal biopsy; mouth rinse
N/A
C. albicans prevalence (in both biopsies and mouth rinse) and counts correlated with severity of dysplasia and OSCC (lowest for mild dysplasia, highest for OSCC)
Berkovits et al. 2016 [94], Hungary
20/40
Cultivation for isolation and CFU count
MALDI-TOF-MS for identification
Surface swabs: tumor
Surface swabs: adjacent normal mucosa and normal mucosa from healthy subjects
Greater average fungal burden (predominantly Candida) in OSCC vs. healthy and on tumor surface compared to the normal epithelium in the OSCC patients
More diverse fungal community in association with OSCC: Candida, Rhodotorula, Saccharomyces, Kloeckera, and others
Mukherjee et al. 2017 [76], USA
39/39 (self) All mobile tongue cancer
ITS1 amplicon sequencing (Ion Torrent)
Fresh-frozen tumor tissue
Fresh-frozen adjacent normal tissue
Genera Ophiocordyceps and Guignardia significantly enriched in tumors (Lysurus, Leptosphaeria, Antrodiella, and Boletus more abundant in control samples)
Perera et al. 2018 [95•], Sri Lanka
25/27
ITS2 amplicon sequencing (Miseq)
Fresh tumor tissue
Fresh tissue from fibroepithelial polyp
C. albicans, C. etchellsii, and Hannaella luteola–like species enriched in OSCC (A Hanseniaspora uvarum–like species, Malassezia spp., Aspergillus tamarii, Cladosporium halotolerans, and Alternaria alternata more abundant in control samples)
*OSCC/healthy unless otherwise indicated
C. albicans Candida albicans, CEOC chronic erythematous oral candidosis, CFU colony-forming unit(s), HPLC high-performance liquid chromatography, high-pressure liquid chromatography, ITS internal transcribed spacer, N/A not available, OSCC oral squamous cell carcinoma, PAS periodic Acid-Schiff, MALDI-TOF-MS matrix-assisted laser desorption/ionization–time-of-flight mass spectrometry, spp. species, vs. versus
Because recent studies that used NGS have demonstrated that the oral fungal community (mycobiome) is far more complex than previously thought [17, 18], a few studies have recently re-attempted to profile the mycobiome associated with OSCC, aided by novel technology. Using cultivation techniques coupled with matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS), Berkovits and team identified a more diverse OSCC-associated fungal community, comprising Rhodotorula, Saccharomyces, and Kloeckera in addition to Candida species [94]. Consistent with earlier studies, the fungal load was also greater in OSCC compared to healthy controls. In another study, Perera and colleagues used sequencing of the Internal Transcribed Spacer (ITS) to characterize the mycobiome within OSCC tissues compared to that in fibroepithelial polyps, and found a dysbiotic mycobiome dominated by C. albicans to be associated with OSCC [96]. Interestingly, Mukherjee’s team also employed ITS sequencing to profile the mycobiome in tongue cancer tissues compared to adjacent normal tissues [76]. Surprisingly, however, despite identification of significant differences, Candida was not found to be differentially abundant, neither was it the most abundant genus.

“Passenger-Turning-Driver” Microbiome Model for Oral Cancer

Synthesizing all results from the studies presented, we developed a novel “passenger-turning-driver” conceptual model of the potential role of the microbiome in oral cancer as illustrated in Fig. 4. Unlike the bacterial “driver-passenger” model for colon cancer, in which tumorigenesis is initiated by driver species that subsequently are replaced in the tumor microenvironment by passenger species that can then either suppress or promote tumor progression [97], our model assumes the oral microbiome is not involved in initiation of oral cancer. Instead, we propose that the initial intra-tumor microbiome represents a passenger event that results from selection by the tumor microenvironment of a subset of species with competitive advantage, e.g., Fusobacteria, Gram-negative anaerobic rods (Prevotella, Camphylobacter, Selenomonas, etc.), anaerobic cocci (e.g., Parvimonas), and C. albicans. The particular species enriched can vary from one subject to another. As it matures, the intra-tumor microbiome expresses pro-inflammatory microbial features and virulence factors (LPS biosynthesis, flagella assembly, bacterial chemotaxis, peptidases, etc.), turning into a functionally dysbiotic, “driver” microbiome that enhances progression of oral cancer by sustaining chronic inflammation.
This conceptual model parallels the model for periodontitis pathogenesis proposed by Bartold and Van Dyke, which also is based on no particular, individual, specific, “pathogenic” bacterium being responsible for initiating the disease process [98, 99]. Rather, certain bacteria become overabundant due to the changes in the microenvironment caused by the initial inflammation and swelling of the gingival tissues that subsequently favor the bacteria that thrive in environments with less oxygen.

Conclusions

Based on existing evidence, we conclude there are significant variations in the composition of the microbiome associated with OSCC, and there are no specific species to implicate in its etiology—of course excluding oncoviruses that are associated with a special entity of oral cancer. Rather, it is the disturbed function of an initially “passenger” microbiome within the tumor microenvironment that is likely to contribute to progression of the tumor by overexpression of virulence factors and pro-inflammatory features. These attributes are common to many taxa that can substitute for each other in different subjects. Moving forward, therefore, a functional approach, particularly metatranscriptomics, is the right way to assess the role of the microbiome in oral cancer. In addition to characterizing the transcriptional activity of the microbiome within the tumor, metatranscriptomics also provides an opportunity to explore all microbial kingdoms (viruses, phages bacteria, archaea, fungi, and protozoa) present in the tumor simultaneously, and to study the interaction between the host and microbiome if the host transcriptome is sequenced in parallel. Further research in this direction will improve our understanding of the mechanisms by which the tumor’s microbial community function contributes to oral carcinogenesis and influences the behavior of the neoplasm: this may open new avenues for the diagnosis, prevention and treatment of oral cancer.

Compliance with Ethical Standards

Conflict of Interest

The authors declare that they have no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Dent – Das Online-Abo der Zahnmedizin

Online-Abonnement

Mit e.Dent erhalten Sie Zugang zu allen zahnmedizinischen Fortbildungen und unseren zahnmedizinischen und ausgesuchten medizinischen Zeitschriften.

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Ferlay J, Ervik M, Lam F, et al. Global Cancer Observatory: Cancer Today. Lyon, France: International Agency for Research on. Cancer. 2018; https://gco.iarc.fr/today. Accessed 19 February, 2019. Ferlay J, Ervik M, Lam F, et al. Global Cancer Observatory: Cancer Today. Lyon, France: International Agency for Research on. Cancer. 2018; https://​gco.​iarc.​fr/​today. Accessed 19 February, 2019.
3.
Zurück zum Zitat American Cancer Society. Cancer facts & figures 2017. Atlanta: American Cancer Society; 2017. American Cancer Society. Cancer facts & figures 2017. Atlanta: American Cancer Society; 2017.
21.
Zurück zum Zitat Johnson NW, Gupta B, Speicher DJ, et al. Chapter 2. Etiology and risk factors. In: Shah JP, Johnson NW, editors. Oral and oropharyngeal cancer. 2nd ed. Boca Raton: CRC Press, Taylor & Francis Group; 2018. p. 19–94. Johnson NW, Gupta B, Speicher DJ, et al. Chapter 2. Etiology and risk factors. In: Shah JP, Johnson NW, editors. Oral and oropharyngeal cancer. 2nd ed. Boca Raton: CRC Press, Taylor & Francis Group; 2018. p. 19–94.
29.
Zurück zum Zitat Markowska J, Fischer N, Markowski M, et al. The role of Chlamydia trachomatis infection in the development of cervical neoplasia and carcinoma. Med Wieku Rozwoj. 2005;9(1):83–6.PubMed Markowska J, Fischer N, Markowski M, et al. The role of Chlamydia trachomatis infection in the development of cervical neoplasia and carcinoma. Med Wieku Rozwoj. 2005;9(1):83–6.PubMed
57.
Zurück zum Zitat • Nagy KN, Sonkodi I, Szoke I, et al. The microflora associated with human oral carcinomas. Oral Oncol. 1998;34(4):304–8. https://doi.org/10.1016/S1368-8375(98)80012-2 Based on the available culturing techniques, this study was the first to identify compositional differences in the microbial community colonizing the surface of OSCC vs adjacent clinically healthy tissue. CrossRefPubMed • Nagy KN, Sonkodi I, Szoke I, et al. The microflora associated with human oral carcinomas. Oral Oncol. 1998;34(4):304–8. https://​doi.​org/​10.​1016/​S1368-8375(98)80012-2 Based on the available culturing techniques, this study was the first to identify compositional differences in the microbial community colonizing the surface of OSCC vs adjacent clinically healthy tissue. CrossRefPubMed
71.
Zurück zum Zitat • Al-Hebshi NN, Nasher AT, Maryoud MY, et al. Inflammatory bacteriome featuring Fusobacterium nucleatum and Pseudomonas aeruginosa identified in association with oral squamous cell carcinoma. Sci Rep. 2017;7(1):1834. https://doi.org/10.1038/s41598-017-02079-3 This is the first attempt to perform functional analysis of the microbiome associated with OSCC and to describe it as being inflammatory. CrossRefPubMedPubMedCentral • Al-Hebshi NN, Nasher AT, Maryoud MY, et al. Inflammatory bacteriome featuring Fusobacterium nucleatum and Pseudomonas aeruginosa identified in association with oral squamous cell carcinoma. Sci Rep. 2017;7(1):1834. https://​doi.​org/​10.​1038/​s41598-017-02079-3 This is the first attempt to perform functional analysis of the microbiome associated with OSCC and to describe it as being inflammatory. CrossRefPubMedPubMedCentral
74.
Zurück zum Zitat Guerrero-Preston R, White JR, Godoy-Vitorino F, et al. High-resolution microbiome profiling uncovers Fusobacterium nucleatum, Lactobacillus gasseri/johnsonii, and Lactobacillus vaginalis associated to oral and oropharyngeal cancer in saliva from HPV positive and HPV negative patients treated with surgery and chemo-radiation. Oncotarget. 2017;8(67):110931–48. https://doi.org/10.18632/oncotarget.20677.CrossRefPubMedPubMedCentral Guerrero-Preston R, White JR, Godoy-Vitorino F, et al. High-resolution microbiome profiling uncovers Fusobacterium nucleatum, Lactobacillus gasseri/johnsonii, and Lactobacillus vaginalis associated to oral and oropharyngeal cancer in saliva from HPV positive and HPV negative patients treated with surgery and chemo-radiation. Oncotarget. 2017;8(67):110931–48. https://​doi.​org/​10.​18632/​oncotarget.​20677.CrossRefPubMedPubMedCentral
85.
Zurück zum Zitat •• Yost S, Stashenko P, Choi Y, et al. Increased virulence of the oral microbiome in oral squamous cell carcinoma revealed by metatranscriptome analyses. Int J Oral Sci. 2018;10(4):32. https://doi.org/10.1038/s41368-018-0037-7 This is the only study so far that used metatranscriptome sequencing to assess the function/gene expression of the microbiome associated with OSCC. Transcripts of virulence factors and pro-inflammatory features were found to be enriched in the tumor samples. CrossRefPubMedPubMedCentral •• Yost S, Stashenko P, Choi Y, et al. Increased virulence of the oral microbiome in oral squamous cell carcinoma revealed by metatranscriptome analyses. Int J Oral Sci. 2018;10(4):32. https://​doi.​org/​10.​1038/​s41368-018-0037-7 This is the only study so far that used metatranscriptome sequencing to assess the function/gene expression of the microbiome associated with OSCC. Transcripts of virulence factors and pro-inflammatory features were found to be enriched in the tumor samples. CrossRefPubMedPubMedCentral
87.
Zurück zum Zitat •• Tian L, Wu A-K, Friedman J, et al. Deciphering functional redundancy in the human microbiome. bioRxiv. 2017;(n/a):176313. https://doi.org/10.1101/176313 This is a key article that provides insight and mathematical modeling of the concept of functional redundancy. It highlights the importance of a function-based approach to studying the human-associated microbiomes and not relying on only microbial profiles based on their abundance. •• Tian L, Wu A-K, Friedman J, et al. Deciphering functional redundancy in the human microbiome. bioRxiv. 2017;(n/a):176313. https://​doi.​org/​10.​1101/​176313 This is a key article that provides insight and mathematical modeling of the concept of functional redundancy. It highlights the importance of a function-based approach to studying the human-associated microbiomes and not relying on only microbial profiles based on their abundance.
96.
Zurück zum Zitat Perera I, Ekanayake L. Prevalence of oral impacts in a Sinhala-speaking older population in urban Sri Lanka. Community Dent Health. 2003;20(4):236–40.PubMed Perera I, Ekanayake L. Prevalence of oral impacts in a Sinhala-speaking older population in urban Sri Lanka. Community Dent Health. 2003;20(4):236–40.PubMed
Metadaten
Titel
The Microbiome of Oral Squamous Cell Carcinomas: a Functional Perspective
verfasst von
Nezar N. Al-Hebshi
Wenche S. Borgnakke
Newell W. Johnson
Publikationsdatum
18.04.2019
Verlag
Springer International Publishing
Erschienen in
Current Oral Health Reports / Ausgabe 2/2019
Elektronische ISSN: 2196-3002
DOI
https://doi.org/10.1007/s40496-019-0215-5

Weitere Artikel der Ausgabe 2/2019

Current Oral Health Reports 2/2019 Zur Ausgabe

Oral Disease and Nutrition (F Nishimura, Section Editor)

Vitamin Status and Mineralized Tissue Formation

Microbiology (C Genco, Section Editor)

Oral Health and Cancer

Oral Disease and Nutrition (F Nishimura, Section Editor)

Tooth Loss and Alzheimer’s Disease

Newsletter

Bestellen Sie unseren kostenlosen Newsletter Update Zahnmedizin und bleiben Sie gut informiert – ganz bequem per eMail.