Review

Vol. 116: Issue 6 - December 2024

The oral microbiome and its role in oral squamous cell carcinoma: a systematic review of microbial alterations and potential biomarkers

Authors

Key words: oral cancer, microbiome, oral microbiota, OSCC, systematic review
Publication Date: 2024-12-18

Abstract

Background. Oral squamous cell carcinoma (OSCC) is one of the most common cancers worldwide. Despite advances in diagnosis and treatment, the incidence of OSCC is increasing, and the mortality rate remains high. This systematic review aims to examine the potential association between the composition of the oral microbiota and OSCC. 

Materials and methods. This study’s protocol was developed according to the PRISMA guidelines. Several search engines, including Medline-PubMed, Scopus (via Elsevier), and Google Scholar, were used to identify original studies that analyzed differences in the oral microbiome between OSCC patients and controls. Twenty-seven studies were identified that reported significant differences in microbial abundance between OSCC and controls.

Results. The systematic review highlights a complex relationship between the oral microbiome and the pathogenesis of OSCC. Significant changes in the microbial composition were identified, with a predominance of phyla such as Bacteroidetes and Fusobacteria, which are associated with inflammatory mechanisms facilitating tumor progression. A remarkable variability in microbial profiles emerged based on the different stages of the disease and the types of samples analyzed, demonstrating the complexity of the oral microbial ecosystem.

Conclusion. Although alterations in the oral cavity microbiome composition are evident in patients with OSCC, identifying a specific pattern remains challenging. However, the integration of advanced analytical techniques, such as artificial intelligence, could overcome this problem, allowing the identification of crucial biomarkers and improving the understanding of the role of the microbiome in carcinogenesis. This approach could transform microbiome analysis into a useful tool for screening and monitoring patients with OSCC.

Introduction

Globally, head and neck cancers rank sixth in terms of malignancy 1. They are extremely aggressive tumors despite their rarity. As a result, the tumor is thoroughly examined by the scientific community, looking at any factor that might affect its course and be relevant to diagnosis, prognosis, and treatment. The primary issue with HNSCC is its delayed diagnosis, which contributes to low quality of life (QOL), significant impairments, and poor 5-year survival 2,3. Due to field cancerization, the latter frequently experience recurrence or develop second primary tumors 4.

HNSCC not only puts physical health at risk but also mental health, being the second cancer with the highest risk of suicide. Survivors of this cancer face comorbidities related to survival and lifestyle. It causes disfigurements and functional disorders, including swallowing, speech, and taste, which substantially impact patients’ quality of life. Psychological distress and impaired QOL are probably the main factors underlying suicide 5. Among cancer sites associated with suicide, HNSCC is ranked in the top four 6.

The fight against HNSCC has become increasingly urgent in recent years, as the Global Cancer Observatory (GCO) revealed a worryingly increasing trend. In 2020, the number of reported cases of HNSCC reached 377,713 worldwide, with a significant concentration in Asia 7. However, future predictions are even more disturbing: the GCO indicates that the incidence of HNSCC could increase by 40% by 2040, resulting in increased mortality rates 7. These data reflect a growing challenge for the medical and scientific community to find new diagnostic and therapeutic strategies to address this serious public health threat.

A crucial aspect emerges more clearly in the panorama of oncology research: the decisive role of the oral microbiota in carcinogenesis and the development of different forms of cancer. This awareness has roots in the last two decades, during which the scientific community has identified and analyzed the distinctive characteristics of cancer, “The Hallmarks of Cancer”, outlining fundamental traits that guide its development 8,9. In parallel, oral microbiota studies have revealed a close correlation between oral microbiota-mediated carcinogenesis, which satisfies or induces most hallmarks 10. Interestingly, these tumors are not limited to the oral cavity alone, but oral microbiota-associated primary tumors have been observed in the esophagus, stomach, pancreas, and colon/rectum 11.

The oral cavity hosts a huge collection of microbes, including bacteria, fungi, viruses, and bacteriophages, and is considered one of the largest microbiological reservoirs in the human body 12. While the microbiome provides an important homeostatic mechanism, the alteration of its balance, or dysbiosis, compromised by factors such as tobacco smoking, psychological stress, poor dietary habits, and chronic periodontitis, promotes carcinogenesis through several mechanisms, including damage to host DNA and chronic inflammation 13.

Studies suggest that alterations in the microbiota may also contribute to the development of tumors, including HNSCC. In particular P. gingivalis and F. nucleatum have both been shown to be able to induce the production of inflammatory cytokines (IL-1, IL-6, IL-17, IL-23, TNF-α and MMP-8, -9, -13), as well as cell proliferation and cellular invasion, in OSCC with various mechanisms 14,15,16,17 Further studies conducted on OSCC have demonstrated that microbes other than bacteria present in the microbiota can contribute to the development of cancer. In the oral cavity, persistent fungal infections (mainly Candida spp.) and human papillomavirus (HPV) infection and virus infection (Epstein-Barr EBV) may be involved in the formation of oncogenic mutations, leading to the development of carcinoma 18. Despite the various evidence present in the literature, the relationship between oral microbiome and OSCC remains complex and not fully explained. The lack of standardized analytical protocols makes establishing specific microbial models for OSCC difficult, limiting the early identification of oral cancer. Thus, this study aims to conduct an up-to-date systematic review of available oral microbiome composition studies in OSCC, identify current news of correlation with carcinoma, and outline directions for future research. In particular, the effect of the presence or absence of key bacteria and fungi such as Candida on carcinoma development will be compared, while outlining future research directions better to understand these interactions and their potential therapeutic implications.

Materials and methods

SEARCH STRATEGY

A systematic review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) criteria 19. We conducted a comprehensive search of the following databases: Medline-PubMed and Scopus (via Elsevier), to find original full-text research publications published between 2019 and 2024. Boolean operators (AND, OR, NOT) and different combinations of phrases (Tab. I) were used in the search according to the specifications of each database. In addition, the reference lists of the included studies were manually searched for further relevant publications. The search was completed in September 2024.

ELIGIBILITY CRITERIA

For our review, we selected studies published in English as inclusion criteria to investigate the relationship between oral squamous cell carcinoma (OSCC) and the oral microbiome. Only clinical studies conducted in humans, in which the microbiological content of all samples was assessed, were considered. In addition, studies that used healthy tissue samples from the same OSCC patient as a control group were accepted for inclusion. Exclusion criteria included articles presented at conferences or congresses, systematic reviews, and meeting abstracts, as well as studies based on experiments conducted on animals, research focused on tumors other than OSCC, and in vitro studies. The study design was performed according to the PECO (Population, Exposure, Control, and Outcome) method, in which:

  • P (population): represents patients with OSCC.
  • E (exposure): indicates alterations in the composition of the microbiota in the oral cavity.
  • C (control): includes both healthy tissue samples from the same patient with OSCC and healthy tissue samples from individuals without pathology.
  • O (result): concerns the microbial diversity and relative abundance of various oral bacteria, including Candida spp., which could be considered a risk factor for OSCC.

STUDY SELECTION AND DATA COLLECTION PROCESS

Once the eligibility criteria had been established, two reviewers proceeded with the independent selection of articles. To improve the efficiency of data collection, Rayyan software 20 was used, a tool designed to facilitate systematic reviews by removing duplicates and allowing a rapid initial assessment. All articles identified through the search strategy were collated in a central Rayyan database. The reviewers conducted a preliminary examination of the titles and abstracts to identify potentially relevant papers, excluding those not meeting the inclusion criteria. Subsequently, the full version of the selected articles was examined to assess their final eligibility. Each reviewer independently evaluated each article.

STUDY RISK OF BIAS ASSESSMENT

The quality of the included studies was assessed using the Newcastle-Ottawa Scale (NOS) 21. The NOS is a scoring system that assesses eight elements grouped into three categories: selection of participants (maximum 4 points), comparability of groups (maximum 2 points), and ascertainment of the outcome (for cohort studies) or exposure (for case-control studies) of interest (maximum 3 points). Two reviewers independently assessed the risk of bias, and disagreements were resolved by discussion with a third reviewer. Studies with a score above 7 were considered good.

DATA ANALYSIS

Data were collected from selected studies and recorded using a pre-designed Excel sheet for extraction. The following parameters were noted:

  • Study characteristics: publication year, first author name, name of the country where the study was conducted.
  • Study population characteristics: number of cases and controls, age and sex of the study population.
  • Methodologies used: type of samples, sample collection methods, DNA extraction methods, DNA amplification, sequencing platforms, and database.
  • Results: microbial abundance, genera, species and phyla detected.

The collected data provided an overall picture of the correlations between the oral microbiome and OSCC.

Results

 

Study selection

The initial search strategy identified a total of 4,025 studies. We identified and removed 3,017 duplicate studies using Rayyan. After this process, 1.008 articles were assessed through title and abstract analysis. Following this screening phase, 236 articles were excluded because they did not meet the inclusion criteria. Subsequently, 772 studies were examined in detail by applying the eligibility criteria. Of these, 745 studies were excluded for the following reasons: non-English language (n = 46), conference or congress abstracts and systematic reviews (n = 434), in vitro experiments (n = 132), and animal studies (n = 133). Qualitative analysis was mainly conducted through descriptive analysis using Microsoft Excel. Finally, 27 studies were subjected to a qualitative risk of bias assessment; of these, 27 passed the analysis and were included in our review. The PRISMA flowchart in Figure 1 illustrates the detailed study selection process.

STUDY CHARACTERISTICS

The characteristics of the selected studies are detailed in Table II. Overall, the studies came from various countries, with significant representation from the United States, China, and India. Of the 27 included studies, 3 were from the United States of America (USA) 22,23,24 ; 10 from China 25,26,27,29,30,31,32,33,34,35,36; six from India 29,37,38,39,40; 1 from Poland 41; 1 from Colombia 42; 1 from Spain 43; 1 from Turkey 44; 2 from Finland 45,46; one from Egypt 47 and 1 from Japan 48.

The number of participants varied significantly from study to study, with samples ranging from a minimum of 10 to a maximum of 228 subjects. The mean ages of participants were recorded in different ranges, highlighting the prevalence of OSCC in an older age group, with values ranging from 26 to over 80 years. The studies show a predominance of male subjects, even if there is significant female participation. In addition to OSCC patients, 18 studies included healthy control groups (HC) 26,29,31,36,37,38,40,41,42,44,45,46,47,48,50,51,54,55. In comparison, 6 studies used normal tissue adjacent to the tumor to compare the microbiota composition of tumor and non-tumor areas of the same patient 24,31,36,37,41,51. Six studies analyzed samples from premalignant or potentially malignant lesions, such as oral leukoplakia (OLK), oral premalignant lesions (OPMD), and dysplasia. Other comparison groups included patients with buccal mucosal cancer or other head and neck cancer subtypes (2 studies), areca chewers without OSCC (1 study), and tobacco users (1 study).

DNA EXTRACTION TOOLS AND SEQUENCING STRATEGIES IN MICROBIOMES

The samples ranged from tumor tissues to saliva, oral rinses, bacterial plaque, and oral swabs. As described in Table III, 10 articles use saliva as the main matrix for microbiome analysis, while tumor tissue and adjacent tissue samples are present in 13 studies. In addition, 5 articles use oral swabs for sample collection. Only a few studies used combinations of multiple sample types, such as tumor tissues and saliva or bacterial plaque and saliva, highlighting various approaches in oral microbiome analysis.

Different methods were used for DNA extraction in the various studies in this review, listed in Table IV. The most commonly used commercial kits were the DNeasy Blood and Tissue Kit (Qiagen), which was used in 4 studies 23,29,37,38 and the DNeasy PowerSoil Kit (Qiagen), which was adopted in 2 studies 27,51. Other kits that were used include the QIAampFast DNA Stool Mini Kit 30, the Qiamp DNA Blood Mini Kit (Qiagen) 25, the modified QIAGEN DNA Extraction Method 22, and the TIANamp Swab DNA Kit 26. Other approaches for DNA extraction include the use of the E.Z.N.A.® soil DNA Kit 36, the FastDNA SPIN Kit 31, the HiPure tissue and blood DNA kit 32, the AllPrep DNA/RNA Mini Kit 24 and the DNA Master Pure TM kit (Epicentre Biotechnologies® Madison, WI) 42. Other approaches for DNA extraction include the use of the MagMini DNA Isolation Kit 51, the Oragene® DNA kit 48, and the DNAeasy kit (Qiagen, Barcelona, Spain) 43. Furthermore, the use of a cetyltrimethylammonium bromide (CTAB) extraction method was reported by Lan et al. (2023) 34. Finally, it is essential to note that extraction by the traditional phenol-chloroform method was adopted in 2 studies 33,41.

In these studies, DNA amplification was performed by targeting different hypervariable regions of bacterial 16S rRNA genes. In most studies, sequencing of the V3-V4 hypervariable regions of 16S rRNA genes was performed. Five studies used only the V4 region 25,31,41,45,48 and only one study used only the V3 region 23. After the amplification step, the sequencing step was performed using different platforms. Although the Illumina MiSeq system is the predominant choice, less common techniques were also employed, such as 454 pyroxeqing, used by Ganly et al. (2019) 22. Other platforms include the Illumina NovaSeq 6000 and the HiSeq 2500, used by Heng et al. (2022) 32 and Singh et al. (2023) 51, respectively. Furthermore, the DNBSEQ-T7 system was adopted in some research, as highlighted by Jain et al. (2023) 24. Multiple reference databases were employed for sequencing alignment, including Greengenes 22,29,37,38,51, SILVA 32,33,35,40,48,54, Human Oral Microbiome Database (HOMD) 26,41,45,48, NCBI 33,35, and Ribosomal Database Project (RDP) 37. In addition, the functional composition of the metagenomes was inferred from 16S rRNA data using PICRUSt in 5 studies 10,23,27,29,31, and with PICRUSt2 software in 3 studies 22,26,32.

ASSESSMENT OF RISK OF BIAS IN INCLUDED STUDIES

We used the Newcastle-Ottawa Scale (NOS) to assess the methodological quality of all included studies. Table IV summarizes all the scores assigned to each of the studies. Case selection was generally well-defined in almost all studies, scoring 1 for most articles. Furthermore, case representativeness was considered good in most studies, scoring 1. Regarding comparability, most studies matched comparison groups for at least one confounder, such as age or sex, resulting in a score of 1. Studies with the highest scores on the NOS scale (8 or 9 points), such as Ganly et al. (2019) 22 and Ye et al. (2021) 27, demonstrated transparent and replicable ‘exposure’ with well-defined methods and well-documented matching for multiple confounders. Finally, we only considered studies with scores higher than 7 for our analysis. This choice was made to ensure the inclusion of studies with robust methodology and to reduce the risk of bias, thus improving the reliability and validity of our results.

MICROBIOTA DIVERSITYMICROBIAL DIVERSITY AND ABUNDANCE

Alpha-diversity and beta-diversity were not evaluated in 14 of the 27 studies (Tab. V). Alpha diversity measures, which describe the variety of species within a single sample, were analyzed using various indices, including Shannon, Simpson, and Chao1. In contrast, beta diversity, which measures the difference between bacterial communities of distinct samples, was calculated mainly via UniFrac and Bray-Curtis distances and visualized through principal coordinate analysis (PCoA). In particular, two investigations that just looked at alpha diversity were those by Heng et al. (2022) 32 and Ye et al. (2021) 27. According to Shannon and Simpson indices, Ye et al. (2021) 27 found that tumor samples had a much higher alpha diversity than controls. However, no significant differences were detected with the Chao1 index or the observed OTUs. Heng et al. (2022) 32 determined the differences in alpha-diversity indices using the Wilcoxon test.

In the remaining 11 studies included in the review, both alpha and beta diversity of the oral microbiome were assessed. In the work of Ganly et al. (2019) 22, no significant difference in alpha diversity was observed between OSCC, pre-malignant lesions (PML), and control groups, while significant separation in beta diversity was observed, suggesting that oral microbiome composition varies substantially by disease. Similarly, Rai et al. (2020) 29 found no significant difference in alpha diversity between OSCC cases and controls. Notwithstanding, a beta diversity analysis revealed a significant separation of the groups, highlighting a change in microbial composition associated with OSCC. In contrast, Sarkar et al. (2021) 37 observed a reduction in alpha diversity in OSCC samples compared to controls, with significant differences in indices such as Chao1, Shannon and Simpson, confirming an alteration in microbial composition in cancerous lesions. Beta diversity analysis also revealed significant differences between groups, suggesting a correlation between the oral microbiome and OSCC. Makinen et al. (2023) 45 found a significant reduction in diversity in patients at follow-up compared to baseline, using the Shannon index, Bray-Curtis distance. Zhong et al. (2021) 30 noted a reduction in alpha diversity in OSF (oral submucosal fibrosis) samples. However, they did not observe significant differences between normal oral mucosa and OSCC lesions, indicating that external factors, such as alcohol or tobacco consumption, may influence microbial diversity.

Studies such as Saxena et al. (2022) 23 found reduced alpha diversity in OSCC samples, particularly concerning the Chao1 index, with a clear separation in beta diversity analyses between the OSCC and control samples. In contrast, Zeng et al. (2022) 31 observed a significant reduction in alpha diversity in OSCC samples. Nonetheless, they did not detect significant differences in beta diversity, suggesting that the oral microbiome composition may not undergo obvious changes at the beta level despite the reduction in microbial richness. However, Oyeyemi et al. (2023) 38 did not identify any significant differences in alpha and beta diversity between the groups, indicating that oral microbiome diversity may remain unaltered in certain instances, even in oral disease. Herreros-Pomares et al. (2023) 43 showed a reduction in microbial richness in OSCC patients. Still, beta diversity analysis revealed greater homogeneity in the microbiomes of OSCC and PVL-OSCC patients, indicating less variability between individuals. The Lan et al.(2023) 34 study showed significant differences in alpha diversity between healthy control and precancerous groups, with a clear separation in the PCoA plots of beta diversity, suggesting a change in microbiome composition during disease progression, but with minimal effects of external factors such as smoking and alcohol. Finally, Singh et al. (2023) 51 observed greater richness in pre-cancer samples compared to patients with advanced stage (T4) cancer and less microbial variability in stage T4 samples, with a significant shift in microbial composition between pre-cancer and cancer microbiomes but no differences between adjacent tumor tissue samples and actual tumors. These findings illustrate the variability in methodology and conclusions between studies, emphasizing the necessity for further research to understand better the complex interactions between the oral microbiome and oral disease.

The main data regarding microbial abundance are described in detail in Table VI. Supplementary I (Table VI).

BACTERIOME CHARACTERIZATION

Taxonomical level

Most of the studies concentrated on the most abundant 5 phyla in the oral cavity: Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, Fusobacteria, and Prevotella.

Bacteroidetes emerged as a key phylum, with thirteen of the 27 studies identifying its predominance in cancer lesions 23,24,25,26,27,29,31,32,33,35,37,38,43,48. Studies such as those by Erira et al. (2021) 42 confirmed these findings using next-generation sequencing on dental plaque, reporting high levels of Bacteroidetes in cancerous tissues. Chang et al. (2019) and Heng et al. (2022) similarly observed elevated levels of Bacteroidetes not only in OSCC lesions, but also in precancerous lesions, indicating that this phylum may be involved in early carcinogenesis. Zhong et al. (2021) 30 also observed an increase in Bacteroidetes in areca consumers, suggesting that this phylum could promote inflammation and tumor progression. Notably, Hashimoto et al. (2022) found elevated levels of Bacteroidetes in the saliva of OSCC patients, indicating a potential role in the disease’s diagnostic specificity and progression 48.

Conversely, Zhou et al. (2021) 36 highlighted reduced levels of Bacteroidetes in tumor lesions.

Fusobacteria was frequently associated with tumor samples, with 10 studies reporting significant enrichment compared to healthy controls. Its presence was strongly linked to dysbiosis and inflammation, both of which are factors in OSCC progression 23,25,26,29,31,33,36,37,38,48.

Similarly, Firmicutes was identified as abundant in cancerous lesions by 7 studies 24,27,29,31,37,38,41.

This phylum has been correlated with both inflammatory states 36 and areca nut consumption 30, potentially exacerbating existing inflammation and fostering neoplastic progression. For example, Singh et al. (2023) 51 observed an abundance of Firmicutes of 40.90% in precancerous lesions and 28.38% in cancerous samples.

In contrast, Heng et al. (2022) 32 noted a reduction in Firmicutes in OSCC patients, with percentages of 25% in buccal swab and plaque samples and 19% in saliva, compared to healthy controls (46% in buccal swab and 42% in saliva) and leukoplakia lesions (32% in buccal swab and plaque).

Proteobacteria was less frequently mentioned but still appeared relevant, with 4 studies reporting its abundance in OSCC samples 33,34,37,43. Notably, Yang et al. (2021) 26 observed a high abundance in salivary samples from OSCC patients. Singh et al. (2023) 51 reported that this phylum was abundant in both precancerous (22.09%) and cancerous samples (29.39%). In contrast, Herreros-Pomares et al. (2023) 43 found low levels of Proteobacteria in both OSCC and proliferative verrucous leukoplakia (PVL)-associated samples.

Lastly, Actinobacteria was highlighted in both precancerous and cancerous samples. Lan et al. (2023) 34 and Singh et al. (2023) 51 observed the enrichment of Actinobacteria in patients with precancerous lesions, suggesting a potential role in malignant transformation. Jain et al. (2023) 24 and Rai et al. (2020) 29 found it high in the presence of the cancerous lesion OSCC. However, Heng et al. (2022) 32 reported a significant reduction of Actinobacteria in saliva samples from OSCC patients compared to healthy controls. Liu et al. (2022) 33 found that it was markedly reduced in patients with deep invasion. Saxena et al.(2022) 23 found an increased abundance of Actinobacteria in the contralateral healthy sites.

In several studies, Fusobacterium has emerged as one of the most abundant genera in tumor samples. Eleven out of 27 studies found that Fusobacterium is significantly more abundant in tumor tissues than in normal tissues 22,23,25,26,29,30,31,33,36,43,48. Similarly, Torralba et al. (2021) 41, using a Random Forest analysis, identified Fusobacterium as a predictive genus of virulence factors associated with inflammatory processes and immunoevasion mechanisms. Like Fusobacterium, Prevotella is also frequently reported to be prevalent in tumor samples. Five out of 27 studies found an increase in Prevotella in cancerous lesions compared to healthy controls 22,25,29,31,37 associated Prevotella with inflammatory processes. Torralba et al. (2021) 41 highlighted its connection to growth factors and inflammatory metabolites, suggesting its potential role in promoting tumor progression. Porphyromonas is another genus frequently found in tumor samples. Zeng et al. (2022) 31 and Rai et al. (2020) 29 identified it as abundant in tumor tissues, highlighting its potential involvement in the pathogenesis of oral cancer. On the other hand, it is interesting to note that nine of the analyzed studies reported Streptococcus spp. release in carcinoma lesions. These studies observed a higher abundance of this genus in healthy sites, suggesting that Streptococcus may serve as a marker of a healthy oral microbiome 22,23,25,26,27,31,32,36,43 48. While in precancerous lesions, Singh et al. (2023) 51 found a significant abundance of Streptococcus, indicating a potential diagnostic role in the early stages of the disease. However, some studies present conflicting results: five of the 27 articles analyzed highlighted significant amounts of Streptococcus in patients with OSCC 24,29,34,42,45. Furthermore, Herreros-Pomares et al. (2023) 43 documented high levels of Streptococcus not only in healthy subjects but also in those affected by proliferative verrucous leukoplakia (PVL), suggesting a possible variability in the microbial composition depending on the clinical context. Finally, three of the 27 studies analyzed reported a high abundance of Neisseria in tumor samples, suggesting a potential diagnostic role in the evaluation of lesions 23,29,31,32,33,38,42. However, Ye et al. (2021) 27 and Chang et al., 2019 35 found low levels of Neisseria in OSCC samples compared to healthy controls, highlighting some variability in the results. Rothia has been identified with high abundance in tumor samples 29,42. Singh et al. (2023) 51 highlighted a higher abundance of Rothia in precancerous samples, suggesting a potential role in disease progression.

Fusobacterium nucleatum is one of the most abundant species in OSCC patients. Eight of the 27 studies analyzed highlight a significant correlation with the pathogenesis of oral cancer 23,29,33,36,41,42,45,51. Torralba et al. (2021) 41 confirmed its role in tumor progression through virulence factors. Prevotella spp. is another significant genus, particularly Prevotella intermedia, whose presence has been documented in several studies of OSCC patients. Liu et al., 2022 33, Heng et al., 2022 32, and Rai et al., 2020 29 reported its presence in tumor samples. Furthermore, Saxena et al.(2022) 23 and Zhou et al.(2021) 36 confirmed the abundance of Prevotella intermedia in tumor and paracancerous tissues. Herreros-Pomares et al. (2023) 43 detected an enrichment of Prevotella nanceiensis in OSCC samples associated with proliferative verrucous leukoplakia (PVL). Finally, Torralba et al. (2021) 41, through their metagenomic analysis, documented a high presence of several Prevotella spp. in samples. Rai et al.(2020) 29 reported a high presence of Prevotella spp. and Porphyromonas spp. in tumor tissues. Rai et al. (2021) 29 highlighted the association of Streptococcus anginosus with the early stages of oral cancer development. Furthermore, Erira et al. (2021) 42 documented a significant prevalence of several Streptococcus spp. in the saliva of OSCC patients. Also, among the genus Rothia (Rothia in grassetto), Singh et al. (2023) 51 identified high levels in the pre-cancerous lesions. Finally, Herreros-Pomares et al., 2023 43 found a notably higher presence of Campylobacter jejuni in the microbiomes of patients with proliferative verrucous leukoplakia.

METHODS USED FOR CANDIDA SPP. IDENTIFICATION

Unlike the bacteriome, which has been characterized mainly by sequencing techniques, the importance of the role of Candida spp. in the context of OSCC has led some authors to use a polyphasic approach. The techniques used to identify Candida spp. in the reviewed articles include phenotypic and genotypic methods (Tab. VII). CHROMagar Agar was used among the phenotypic methods, which allows rapid identification of Candida spp. through specific chromogenic reactions 39. Periodic Acid-Schiff (PAS) staining was employed by Hafed et al. (2019) 47 to visualize Candida hyphae in tissue samples, allowing for histological identification of the fungus. Colony counting was also performed by Rusanen et al. (2024) 46, providing quantitative data on fungal presence. DNA extraction was performed by boiling lysis for cell samples 37, while tissue samples were deparaffinized and assessed with a specific kit 44. Real-time PCR was employed to identify and quantify species using specific primers and standard curves 44. Hafed et al. (2019) 47 performed RNA extraction with an RNeasy® FFPE kit from tissue samples and analyzed it by RT-PCR (Reverse Transcription PCR) to study gene expression. Furthermore, Sankari et al.(2020) 40 applied the restriction fragment length polymorphism (RFLP) technique to distinguish species through amplification of the ITS1-5.8SrDNA-ITS2 region.

INCIDENCE OF CANDIDA SPP. IN OSCC PATIENTS

We examined 5 articles that were specifically about Candida spp. in the current literature evaluation. The results of the five studies included in this review indicated a significantly higher prevalence of Candida spp. in patients with OSCC and oral potentially malignant disorders (OPMD) compared to healthy subjects. Arya et al. (2021) 39 found an increasing incidence of Candida albicans, Candida krusei, Candida tropicalis, and Candida parapsilosis in healthy subjects, patients with OPMD, and patients with OSCC. Similarly, Sankari et al. (2020) 40 reported oral Candida prevalence of 72.2%, 58%, and 20.5% in patients with OSCC, OPMD, and healthy controls, respectively. Differences in the distribution of Candida spp. emerged between the groups, predominating Candida krusei, Candida tropicalis, and Pichia anomala (formerly known as Candida pelliculosa). In contrast, Candida and other pathogenic fungi such as Acremonium exuviarum and Aspergillus fumigatus were significantly increased in OSCC patients compared to healthy controls and OPMD patients 32. During follow-up, a significant reduction in salivary flow and an increase in Candida spp. in saliva post-treatment was observed, suggesting that the treatment may have impaired salivary function and facilitated Candida spp. proliferation 45.

Discussion

In this systematic review, we examined 27 studies on the role of different microorganisms, including bacteria and fungi, concerning OSCC, highlighting the geographical variables, DNA extraction methods, amplification, and sequencing strategies used.

Our analysis confirms that DNA extraction is crucial to high-quality 16S rDNA sequences for reliable microbiome profiling.

Most studies included in this review indicated a preference for commercial kits due to their consistency and reduced risk of contamination. This finding supports the conclusions of Zhang et al. (2022) 52, who emphasize the reproducibility offered by standardized kits. Only a minority employed traditional extraction methods, such as CTAB or phenol-chloroform. These methods, although convenient, present greater variability and risk of contamination 53. However, there is a preference for commercial kits despite their limitations due to potential PCR inhibitors and cost 54. Selection of the appropriate extraction technique is crucial; recent guidelines emphasize the need for standardization to ensure reproducibility 55.

The selection of hypervariable regions for 16S rRNA amplification significantly impacts taxonomic resolution, as demonstrated by Gopinath et al.(2019) 56.

Our results showed that the V3-V4 regions were selected the most, followed by the V4 region. Indeed, studies like that by Johnson et al. 57 have highlighted the potential to produce an accurate, high-resolution taxonomy of organisms via full-length 16S sequencing 57. In contrast, the V4 region has been employed less frequently, although it is often chosen for compatibility with platforms that support shorter reads 58. This choice depends on the sequencing platform used; for example, the 250-bp paired-end reads of the Illumina MiSeq system fit the V3-V4 region well 59, ensuring complete coverage. Although the Illumina MiSeq is a prevalent platform, while some studies, such as that by Ganly et al. (2019) 22, have used less common platforms, including the NovaSeq 6000 and 454 pyrosequencing.

The studies analyzed in this review also employed newer platforms like DNBSEQ-T7 to explore deeper coverage. The databases used for sequence alignment exhibited notable variability, with Greengenes, SILVA, and the Human Oral Microbiome Database (HOMD) the most frequently utilized. This diversity reflects an ongoing effort to align databases with specific research requirements, affecting downstream bioinformatics analysis and taxonomic accuracy.

The different approaches highlight the challenges in drawing uniform conclusions about OSCC-related microbial communities. Our results are consistent with those of Bars-Cortina et al. (2024) 60, who compared 16S rRNA sequencing to shotgun metagenomics and found that targeted 16S regions remain superior for identifying disease-associated microbiota changes. These insights are critical because they imply that, although shotgun metagenomics offers broader functional insights, 16S sequencing retains its value for taxonomic investigations in targeted disease studies.

The analysis of the studies revealed a need for more consistency in the results about the microbial diversity associated with OSCC. Some studies have reported significant differences in alpha and beta diversity between OSCC samples and controls, thereby underscoring alterations in the microbial composition associated with the disease. For instance, several studies have documented a reduction in alpha diversity in tumor samples, as indicated by indices such as Shannon and Simpson. However, other studies have not identified any significant differences. While alpha diversity does not appear to be consistently affected by oral diseases, beta diversity frequently demonstrates a notable distinction between OSCC groups and controls, suggesting that the composition of the oral microbiome may vary considerably concerning the pathology. However, the variability of the results indicates that there may be discrepancies in the analytical methods, samples, and confounding factors that influence the outcomes.

The oral microbiome composition in OSCC patients differs markedly from that of healthy subjects, with significant alterations in several bacterial phyla and genera. The collected data show a predominance of some phyla, including Bacteroidetes and Fusobacteria, which are frequently associated with tumor lesions. For example, the phylum Bacteroidetes was reported to be enriched in 13 out of 27 studies, confirming a close association with cancerous lesions 23,24,25,26,27,29,31,32,33,37,38,43,49. This phylum has been associated with inflammatory processes and tumor progression in studies of areca consumers 30. In contrast, other studies, such as that by Zhou et al. (2021) 36, reported a reduction of Bacteroidetes in tumor samples. Fusobacteria emerges closely associated with dysbiosis and inflammatory states that promote carcinogenesis 25,36. The role of Firmicutes is more controversial: while some studies report it as abundant in tumor samples 41, others, such as Heng et al. (2022) 32, observed a significant reduction in OSCC patients compared to healthy controls. In particular, Streptococcus spp., a key genus within the Firmicutes, shows variable behavior. Some studies, such as those by Ganly et al. (2019) 22 and Heng et al. (2022) 32, report a reduction in Streptococcus spp. in OSCC patients, suggesting a possible protective role. However, other research indicates an increased presence of Streptococcus spp. in precancerous stages, associating it with chronic inflammation 42,49. For example, a significantly higher abundance of Streptococcus infantis was found in smokeless tobacco nonusers than in smokeless tobacco users and the contralateral buccal site of OSCC specimens than in the OSCC tumor site 23.

Furthermore, Mäkinen et al. (2023) 45 documented elevated levels of multiple Streptococcus spp. in smoking patients with oral squamous cell carcinoma (OSCC). Furthermore, a reduction in Streptococcus spp. and Rothia spp. levels have been documented during the development of OSCC in patients with oral potentially malignant disorders (OPMD) 51. This duality may reflect the differing roles of Streptococcus spp. in the various stages of the disease. Fusobacterium nucleatum has been identified as one of the most strongly associated genera with tumor occurrence. Studies such those by Ganly et al. (2019) 22, Zhou et al. (2021) 36 and Zeng et al. (2022) 31 have shown a marked increase in F. nucleatum in tumor samples compared to controls, with direct implications for tumor progression through mechanisms including chronic inflammation and immunoevasion. Its association with virulence factors and inflammatory processes further supports the dysbiosis theory as a pathogenic mechanism in OSCC 41. Porphyromonas gingivalis has been linked to a more aggressive malignant profile in patients with OSCC, with the bacterium modulating the local immune response and promoting tumor growth by activating inflammatory pathways 28. However, Chen et al. (2021) 28 also revealed a lower incidence of relapse in patients with high P. gingivalis load, raising interesting questions regarding the complexity of the role of this bacterium in different stages of the disease. Chang et al. 2019 35 demonstrated that Fusobacterium nucleatum and Porphyromonas gingivalis activate Toll-like receptors (TLRs), triggering chronic inflammatory responses that facilitate tumor progression. The authors identified an elevation in the levels of inflammatory cytokines, including interleukin-6 (IL-6), IL-8, and tumor necrosis factor alpha (TNF-α). This correlation between alterations in microbial composition and elevated levels of inflammatory cytokines has also been observed by Singh et al. (2023) 51, further supporting the link between dysbiosis and inflammation in the context of OSCC. In addition, Ganly et al. 22 found significant increases in the expression of the HSP90 gene and TLR ligands 1, 2, and 4 as oral tissues progressed from health to OSCC. Specifically, genes related to heat shock protein 90 and TLR ligands for various microbial components were progressively enriched. Prevotella was identified as the primary contributor to this enrichment of pro-inflammatory genes, with notable additional contributions from Alloprevotella, Fusobacterium, Veillonella, and Porphyromonas 22. Liu et al. 33 further linked lipopolysaccharides (LPS) to specific bacteria in the oral microbiome, identifying Fusobacterium nucleatum as a key pathogenic bacterium responsible for increased LPS levels. This suggests that F. nucleatum plays a significant role in inflammation and the progression of OSCC, highlighting its impact on the microbial ecosystem. These findings have also been found for fungi such as Candida. For example, Rusanen et al. (2024) confirmed that Candida albicans induces chronic inflammation by activating Toll-like receptors (TRL 1-10) and the transcription factor NF-κB in OSCC patients. Candida albicans also produce acetaldehyde, which increases the risk of carcinogenic mutations. Its ability to metabolize ethanol into acetaldehyde in the oral environment, thanks to the expression of alcohol dehydrogenase 1, amplifies the carcinogenic effect linked to alcohol consumption 47. In recent years, the role of interactions between members of the oral microbiota in oral tumorigenesis, progression, and metastasis has gained widespread attention. For example, acetaldehyde produced by C. albicans facilitates the production of phenazines by Pseudomonas aeruginosa, which further induces the production of acetaldehyde by C. albicans, and the two jointly promote oral carcinogenesis 61. Also, Heng et al. (2022) 32, using both traditional and metagenomic methods, found a significant increase in OSCC patients. Specifically, Candida spp. levels were higher in buccal swabs, plaque samples, and saliva from the OSCC group compared to healthy controls (HC) (5% vs. 1%, P = 0.002; 5% vs. 1%, P = 0.007; 5% vs. 1%, P = 0.012). Additionally, levels were elevated in plaque swabs of the OSCC group compared to the oral potentially malignant lesion (OPL) group (5% vs. 1%, P = 0.008) 32.

Several studies have shown that the composition of the oral microbiome varies with the stage OSCC. Singh et al., (2023) 51 showed that specific bacterial genera, such as Fusobacterium, are more abundant in the early stages of the disease, while others, such as Capnocytophaga, prevail in the advanced stages. Yang et al. (2021) 26 analyzed the relationship between microbiota and clinical stages (I–IV), finding that Treponema spp. and Leptotrichia spp. were found to be more abundant in early stages (I/II), while Prevotella spp. and Capnocytophaga spp. were enriched in tumor samples. In advanced stages (III/IV), Solobacterium moorei and Slackia exigua were predominant in tumor tissues. Zeng et al. (2022) 31 correlated the clinicopathological data with the microbiome, noting that higher T-stage was associated with a higher N-stage. They suggested that Fusobacterium may have an oncogenic role, more frequent in T3/T4 stages and lymphatic metastasis. However, contradictions emerged, such as a higher proportion of Porphyromonas in the T1/T2 groups. These observations highlight the complexity of the interactions between oral microbiota and OSCC progression, suggesting a significant impact of specific bacteria on the disease.

Our review also highlighted significant differences in oral microbiome composition among different sample types analyzed. Heng et al. (2022) 32 showed an increase in diversity in OSCC swab samples, while saliva samples from both OSCC and OED showed a significant decrease in diversity. In particular, Yang et al. (2021) 26 reported that the phyla Bacteroidetes and Proteobacteria were significantly enriched in tumor samples (tumor-Saliva). Four genera, such as Filifactor and Peptostreptococcus, showed high abundance in tumor tissues (TT), while 9 species, including Streptococcus oralis and Neisseria macacae, predominated in TS samples. Similarly, Erira et al.(2021) 42 demonstrated that dental plaque, saliva, and tumor tissue had significantly different microbiome compositions, highlighting how the oral microenvironment affects the distribution of bacterial species. In tumor tissue, bacteria such as Fusobacterium nucleatum, Porphyromonas gingivalis, Peptostreptococcus, Prevotella, and Parvimonas micra are abundant and are associated with inflammatory processes and carcinoma progression.

In contrast, dental plaque shows a higher microbial diversity, predominating anaerobic bacteria such as Streptococcus, Veillonella, and Actinomyces, which are not identified as key species in tumor tissues. Finally, saliva has a less diverse bacterial composition than dental plaque, but more heterogeneous than tumor tissue. The detected species, such as Streptococcus and Haemophilus, reflect the recirculation of bacteria in oral biofilm, with a lower representation of those directly associated with tumor progression. These results highlight the importance of the analyzed sample in understanding the dynamics of the oral microbiome concerning OSCC.

The reliability of microbiome studies depends largely on the molecular biology techniques used downstream. Most studies sequenced the V3-V4 regions, although some chose the V4 and V3 regions. Experimental studies have concluded that the choice of 16S rRNA region type for amplification can significantly influence the size of distinct taxa 56.

Analysis of DNA extraction methodologies and sequencing techniques has revealed a variety of approaches, each with its advantages and disadvantages. The predominant use of commercial kits such as the DNeasy® Blood and Tissue Kit and the QIAampFast® DNA Stool Mini Kit suggests a necessary standardization to facilitate comparison between studies. Additionally, the type of 16S rRNA region chosen for amplification can significantly influence the results 25. Metagenomics represents a more comprehensive approach than 16S rRNA alone as it allows for identifying fungi and viruses, reducing the bias due to genetic amplification. Recent studies, such as that by Torralba et al.(2021) 41, have used metagenomics and metaproteomic analyses to analyze saliva from patients with OSCC. This combination of methodologies offers a more integrated view of the microbial composition and functional dynamics within the oral microenvironment.

Despite promising results, most of the included studies have significant limitations. As they are mainly case-control studies, there is a high risk of bias due to confounding factors. However, variables such as age, smoking habits, alcohol consumption, and chewing were considered, and only a few studies adequately adjusted for these factors in the design or analysis phases.

Another methodological issue that emerged in this review is the need for more standardization in microbiome analysis techniques, which made it difficult to compare results between studies. Differences in DNA extraction protocols, sequencing methods, and bioinformatics analysis contributed to this heterogeneity. This highlights the need to integrate more precise and standardized techniques.

Artificial intelligence (AI) can overcome many challenges of analyzing and managing large volumes of complex data, offering advanced solutions beyond the limits of traditional techniques Thanks to its ability to learn from data, AI can detect hidden patterns, improve predictions, and support evidence-based decisions. In our review, we explored studies such as those by Ganly et al., 2019 22 who used Support Vector Machine (SVM) and Random Forest models, and Sarkar et al., 2021 37 with Random Forest to analyze complex datasets, obtaining excellent predictive results. Zhou et al. (2021) 36, for example, achieved a diagnostic accuracy of 95.7% in gene sequencing of oral tissues and predicted OSCC with 100% accuracy using Random Forest. Torralba et al. (2021) 42 also took an innovative approach, combining metagenomics and metaproteomics to identify the presence of bacteria and characterize the proteins expressed in the samples. Next, they applied Random Forest analysis to distinguish distinctive markers between tumor and non-tumor tissues.

This is a promising first approach. An alternative methodology would be to utilize AI in the context of Digital Pathology (DP), specifically in analyzing digital slides. A recent example of bacterial identification using AI is the detection of Helicobacter pylori (HP) in intestinal biopsies 62. The study was conducted with the assistance of DP, utilizing deep learning algorithms to analyze digitized histological images (WSI), which resulted in a notable enhancement in diagnostic efficiency compared to traditional methods. The researchers demonstrated that AI, when applied to low-resolution images (20X), can identify HP infection in intestinal tissue samples with remarkable accuracy while simultaneously reducing the number of false positives and negatives. This demonstrated that the integration of DP and AI accelerates the examination of intricate specimens and yields outcomes analogous to those obtained through conventional microscopy. This validates the efficacy of AI in authentic clinical contexts. While there are currently no studies exploring the use of this approach in the context of oral biofilm, preliminary evidence suggests that similar techniques can be used to identify bacterial species in the intestine. This perspective opens the way to possible future applications of AI in analyzing the OSCC-associated microbiome, offering a unique opportunity to improve diagnostic accuracy and understand microbial interactions in the tumor context.

Conclusion

The present systematic review elucidates the complex relationship between the oral microbiome and the pathogenesis of OSCC. Our findings indicate that OSCC is associated with specific alterations in microbial composition, characterized by a predominance of specific phyla, such as Bacteroidetes and Fusobacteria, which appear to facilitate tumor progression through inflammatory mechanisms. The diversity of microbial profiles observed across different stages of OSCC and between various types of samples underscores the intricate nature of the oral microbial ecosystem. Moreover, incorporating sophisticated analytical techniques, including AI, presents promising avenues for future research, enabling the identification of crucial biomarkers and enhancing our comprehension of the microbiome’s role in cancerogenesis. Addressing the methodological limitations identified in this review is crucial for establishing a robust framework for future studies. In conclusion, these insights could pave the way for innovative diagnostic and therapeutic strategies targeting the oral microbiome in the context of OSCC.

CONFLICTS OF INTEREST STATEMENT

Any current or prospective financial, personal, or other conflicts of interest that might influence or skew their study are completely disclosed by the authors.

FUNDING

Rare cancers of the head and neck: a comprehensive approach combining genomic, immunophenotypic and computational aspects to improve patient prognosis and establish innovative preclinical models – RENASCENCE “(project code PNRR-TR1-2023-12377661)”.

AUTHORS’ CONTRIBUTIONS

AC: Performed the analysis, Wrote the paper; SV: Performed the analysis; AE: Collected the data; AM: Collected the data; DC: Collected the data; AP: Revised the manuscript; GPP: Revised the manuscript; SS: Conceived and designed the analysis; FM: Conceived and designed the analysis, revised the manuscript; GI: Performed the analysis, revised the manuscript.

History

Received: December 1, 2024

Accepted: December 12, 2024

Figures and tables

Figure 1. The flowchart summarizes the selection process for the articles included.

Figure 2. Summary image of typically used samples and workflow to identify the microbiome in OSCC (Created with BioRender.com).

Figure 3. Healthy oral cavity and OSCC in bacteriome results.

Query: ((microbiota) AND (oral cavity)); ((microbiome)AND (oral squamous cell carcinoma OR OSCC ));
((oral microflora OR oral microbes OR oral bacteria OR oral microorganism) AND (OSCC));( (oral pathogen) AND (OSCC));
((oral dysbiosis AND (OSCC OR oral squamous cell carcinoma)); ((dysbiosis) AND (Inflammation) AND (OSCC OR oral squamous Cell Carcinoma )).
Table I. The search strategy used.
Author, year Country Sample Size Group Average/Range of Age (Years) Sex
Chang et al., 2019 [ 35 ] China OSCC 61 57.4 ± 10.4 22 F 39M
HC 30 55.4 ± 10.2 12 F 18M
Ganly et al., 2019 [ 22 ] USA OSCC 18 59.8±10.9 7F 11M
PML 8 66.1±17.9 5F 3M
HC 12 44.4±15.6 9F 3M
Su et al., 2021 [ 25 ] China OSCC 74 53.96±11.07 Only males
Cancers of the buccal mucosa 42 56.31±10.05 n.d
Sarkar et al., 2021 [ 37 ] India OSCC 50 52.6 n.d.
Torralba et al., 2021 [ 41 ] Poland OSCC 18 n.d. n.d.
Yang et al., 2021 [ 26 ] China OSCC 23 61.9 ± 12.3 12 F 11M
19 62.7 ± 12.1 9F 10M
Ye et al., 2021 [ 27 ] China OSCC 23 63.0 ± 9.6 5 F 18 M
Rai et al., 2020 [ 29 ] India OSCC 25 55.32 8F 16M
HC 24 50.375 9F 16M
Zhou et al., 2021 [ 36 ] China OSCC 47 n.d. n.d.
HC 46
Zhong et al., 2021 [ 30 ] China OSCC 45 49.62 ± 9.28 2F 43M
OSF 42 32.62 ± 9.22 2F 40M
HC 46 30.58 ± 10.34 33F 12M
Healthy areca chewer 29 26.16 ± 6.53 2F 27M
Erira et al., 2021 [ 42 ] Colombia OSCC 10 56.5 ± 15.59 90% Female
HC 20 56.4 ± 18.36
Saxena et al., 2022 [ 23 ] USA OSCC 34 48.61 ± 12.76 n.d.
HC 32 32.15 ± 9.19
Zeng et al., 2022 [ 31 ] China OSCC 228 52.4 n.d.
Heng et al., 2022 [ 32 ] China OSCC 29 61.97±10.11 13F 16M
OPL 32 56.00±13.67 19F 13M
HC 30 56.63±11.12 15F 15M
Liu et al., 2022 [ 33 ] China OSCC 40 59.25 ± 16.6 13F 27M
HC 10 60.3 ± 8.4 3F 7M
Hashimoto et al., 2022 [ 48 ] Japan OSCC 41 67.7(28–92) 111F 30M
Postoperative 20 68.2 (29–85) 6F 14M
OLK 25 64.4 (29–91) 9F 16M
Oyeyemi et al., 2023 [ 38 ] India OSCC 10 55.30 n.d.
TA (tabacco abuse) 10 34.30
HC 10 23.60
Mäkinen et al., 2023 [ 45 ] Finland OSCC 99 68.0 (10.3) n.d.
HC 101 66.4 (14.3)
Lan et al., 2023 [ 34 ] China OSCC 18 59.24 ± 12.30 10F 8M
OLK 21 54.11 ± 15.43 17F 4M
HC 21 48.81 ± 12.38 14F 7M
Jain et al., 2023 [ 24 ] USA OSCC 20 n.d. n.d.
Herreros-Pomares et al., 2023 [ 43 ] Spain OSCC 10 72.80 ± 9.05 4F 6M
OSCC-PVL 8 80.25 ± 12.42 2F 6M
HL 9 66.33 ± 8.12 5F 4M
PVL 12 66.83 ± 12.16 9F 3M
HC 11 67.91 ± 12.00 6F 5M
Singh et al., 2023 [ 51 ] India OSCC 69 48 ± 12 24F 71M
Pre-cancer 15
HC 11
Hafed et al., 2019 [ 47 ] Egypt OSCC without LNM 16 53,44 6 F 9 M
OSCC with LNM 15 54,87 7 F 9 M
Oral dysplasia 16 46,56 6 F 10 M
HC 7 44,43 3 F 4 M
Sankari. et al., 2020 [ 40 ] India OSCC 97 n.d. n.d.
OPMD 200
HC 200
Arya et al., 2021 [ 39 ] India OSCC 20 54 3 F 17M
OPMD 20 45 6 F 14 M
HC 20 n.d. n.d.
İlhan et al., 2023 [44] Turkey Benign 20 46.45 ± 8.16 11 F 9 M
Mild/moderate dysplasia 20 58.7 ± 6.90 11 F 9 M
Carcinoma in-situ 6 F 14M
OSCC 20 69.05 ± 5.02
20 61.07 ± 11.54 8 F 12M
Rusanen et al; 2024 [ 46 ] Finland OSCC 30 64 (31–85) 12 F 18M
HC 26 30.8 (18–56) 17 F 9M
*HC = Healthy control; AP = Adjacent paracancerous tissues; F = Female; M = Male; OPMD = Potentially malignant disorders; LNM = Lymph node metastasis; OLK = oral leukoplakia.
Table II. Analysis of case characteristics.
Author, year Sample Type Extraction DNA DNA Amplification Amplification Sequencing Reference database
Chang et al., 2019 [ 35 ] Cancer tissues QIAampFast DNA Stool Mini Ki V3-V4 Illumina MiSeq NCBI
AP tissue
Subgingival plaque
Normal oral tissues
Ganly et al., 2019 [ 22 ] Oral rinse samples between patients and normal people as a control Modified QIAGEN DNA Extraction Method V3-V4 454 FLX platform Greengenes (v13.5)
Su et al., 2021 [ 25 ] Oral swab QIAamp® DNA Blood Mini Kit V4 Illumina MiSeq SILVA
Sarkar et al., 2021 [37] Tissue cancerous lesions DNeasy Blood and Tissue Kit (Qiagen, Hilden, Germany) V3-V4 Illumina MiSeq Greengenes (v13.8) RDP
Adjoining normal area
Torralba et al., 2021 [ 41 ] Paired tumor and adjacent normal tissue samples Phenol-chloroform based DNA extraction V4 Illumina MiSeq HOMD
Yang et al., 2021 [26] Tissue TIANamp Swab DNA Kit V3-V4 Illumina MiSeq HOMD
Saliva
Ye et al., 2021 [ 27 ] Tissue cancer lesions DNeasy PowerSoil Kit (Qiagen, Hilden, Germany) V3-V4 Illumina MiSeq SILVA
Paracancerous normal tissues
Rai et al., 2020 [ 29 ] Saliva Qiagen DNeasy Blood and Tissue Kit V3-V4 Illumina MiSeq Greengenes (v13.8)
Zhou et al., 2021 [36] Saliva, tumor tissue, Plaque, control side of healthy mucosa E.Z.N.A.® soil DNA Kit V3-V4 Illumina MiSeq SILVA
Zhong et al., 2021 [ 30 ] non-stimulating oral swabs QIAamp® DNA Stool Mini Kit (Qiagen) V3–V4 Illumina MiSeq n.d.
Erira et al., 2021 [ 42 ] Tumor tissues, DNA Master Pure TM kit (EpicentreBiotechnologies® Madison, WI) n.d. Illumina MiSeq n.d.
Saliva
Bacterial plaque
Saliva
Bacterial plaque
Saxena et al., 2022 [ 23 ] Swab samples Qiagen DNeasy Blood and Tissue Kit V3 Illumina Nextera XT Greengenes (v13.5) SILVA
Saliva
Zeng et al., 2022 [ 31 ] Saliva, Swab, Adjacent Normal Tissues, Tumor tissue, Lymph Nodes FastDNA SPIN Kit V4 HiSeq 2500 (Illumina Inc). SILVA
Heng et al., 2022 [32] Saliva, plaque swabs, buccal swabs HiPure tissue and blood DNA kit (Magen Biotechnology Co. Ltd., Guangzhou, China). V3-V4 Illumina NovaSeq 6000 platform SILVA
Liu et al., 2022 [ 33 ] Swab samples Phenol-chloroform based DNA extraction n.d. Illumina NovaSeq 6000 platform NCBI
Hashimoto et al., 2022 [ 48 ] Saliva Oragene® DNA kit V4 Illumina MiSeq HOMD
Oyeyemi et al., 2023 [ 38 ] Saliva Qiagen DNeasy Blood & Tissue Kit) V3–V4 Illumina MiSeq Greengenes (v13.8)
Mäkinen et al., 2023 [ 45 ] Saliva MagMini DNA Isolation Kit V4 Illumina MiSeq HOMD
Lan et al., 2023 [ 34 ] Saliva Extraction method with cetyltrimethylammonium bromide (CTAB) n.d. NovaSeq platform n.d.
Jain et al., 2023 [ 24 ] Tissue Tumor, adjacent normal tissue AllPrep DNA/RNA Micro kit (Qiagen) nd Illumina, DNBSEQ-T7 SILVA
Herreros-Pomares et al., 2023 [ 43 ] Tissue DNAeasy kit (QiaGen, Barcelona, Spain) V3–V4 Illumina MiSeq SILVA
Singh et al., 2023 [ 51 ] Peripheral blood,Tumor tissues,Adjacent normal tissues (AT),Precancerous lesion tissues DNeasy power soil kit (Qiagen, USA) V3–V4 Illumina HiSeq 2500 i Greengenes (v13.8)
*HOMD: Human Oral Microbiome Database, NCBI: National Center for Biotechnology Information, NA: not available, SILVA: Silva ribosomal RNA Gene Database Project.
Table III. A summary of DNA extraction, amplification, sequencing techniques, and reference databases.
Author, year Selection Comparability Exposure Total
Definition of cases Representativeness of the cases Selection of Controls Definition of Controls Comparability of cases and controls Ascertainment of exposure Same method of ascertainment for cases and controls Non-Response rate
Chang et al., 2019 [ 35 ] 1 1 1 1 1 1 1 0 7
Ganly et al., 2019 [ 22 ] 1 1 1 1 2 1 1 0 8
Hafed et al., 2019 [ 47 ] 1 1 1 1 1 1 1 0 7
Sankari et al., 2020 [ 40 ] 1 1 1 1 1 1 1 1 8
Su et al., 2021 [ 25 ] 1 1 1 1 1 1 1 1 8
Sarkar et al., 2021 [ 37 ] 1 1 1 1 1 1 1 0 7
Torralba et al., 2021 [ 41 ] 1 1 1 1 1 1 1 1 8
Yang et al., 2021 [ 26 ] 1 1 1 1 1 1 1 1 8
Ye et al., 2021 [ 27 ] 1 1 1 1 2 1 1 1 9
Rai et al., 2020 [ 29 ] 1 1 1 1 1 1 1 1 8
Zhou et al., 2021 [ 36 ] 1 1 1 1 1 1 1 0 8
Zhong et al., 2021 [ 30 ] 1 1 1 1 2 1 1 1 9
Erira et al., 2021 [ 42 ] 1 1 1 1 1 1 1 0 7
Arya et al., 2021 [ 39 ] 1 1 1 1 1 1 1 0 7
Saxena et al., 2022 [ 23 ] 1 1 1 1 2 1 1 1 9
Zeng et al., 2022 [ 31 ] 1 1 1 1 2 1 0 1 8
Heng et al., 2022 [ 32 ] 1 1 1 1 1 1 1 0 8
Liu et al., 2022 [ 33 ] 1 1 1 1 1 1 1 1 9
Hashimoto et al., 2022 [ 48 ] 1 1 1 1 1 1 1 1 8
Oyeyemi et al., 2023 [ 38 ] 1 1 1 1 2 1 1 0 8
Mäkinen et al., 2023 [ 45 ] 1 1 1 1 1 1 1 1 8
Lan et al., 2023 [ 34 ] 1 1 1 1 1 1 1 1 8
Jain et al., 2023 [ 24 ] 1 1 1 1 1 1 1 0 7
Herreros-Pomares et al., 2023 [ 43 ] 1 1 1 1 1 1 1 0 7
Singh et al., 2023 [ 51 ] 1 1 1 1 1 1 1 1 8
Jain et al., 2023 [ 24 ] 1 1 1 1 1 1 1 0 7
İlhan B. et al., 2023 [ 44 ] 1 1 1 1 1 1 1 1 8
Rusanen et al., 2024 [ 46 ] 1 1 1 1 1 1 1 1 8
Table IV. Newcastle-Ottawa scale showing the quality evaluation of studies included.
Study Indices of α-Diversity Microbiota α-Diversity Indices of β-Diversity Microbiota β-Diversity
Ganly et al. 2019 [22] Shannon index, Chao1 index, ANOVA No differences were observed UniFrac analysis(weighted and unweighted analyses) Significant difference
Rai et al. 2020 [29] Shannon index, Simpson index,Chao1 index No differences were observed UniFrac analysis (weighted and unweighted analyses), PCoA Significant difference
Sarkar et al. 2021 [37] Shannon index, Simpson index,Chao1 index Lower diversity in OSCC UniFrac analysis(weighted and unweighted analyses), analysis (PCoA) was performed using Bray-Curtis dissimilarity, PERMANOVA Significant difference
Ye et al. 2021 [27] Shannon index, Simpson index, Chao1 index, number of observed OTUs Significant increase in tumor samples (Shannon and Simpson); no difference with Chao1 and OTU N.D. N.D
Zhong et al. 2021 [30] Shannon index, Simpson index, number of observed OTUs Lower diversity in OSF UniFrac analysis(weighted and unweighted analyses) No differences were observed OSCC vs normal mucosa
Saxena et al. 2022 [23] Shannon index, Simpson index, Chao1 index, Lower diversity in OSCC UniFrac analysis(weighted and unweighted analyses),analysis (PCoA) was performed, using Bray-Curtis dissimilarity,PERMANOVA Significant difference
Zeng et al. 2022 [31] Shannon index Lower diversity in OSCC Bray-Curtis dissimilarity No differences were observed
Oyeyemi et al. 2023 [38] Shannon index, Simpson index, Chao1 index, Fisher No differences were observed Analysis (PCoA) was performed using Bray-Curtis dissimilarity No differences were observed
Heng et al., 2022 [32] Test of Wilcoxon. No differences were observed n.d n.d
Herreros-Pomares et al. 2023 [43] Shannon index, Simpson index,Chao1 index,Inverse Simpson Lower diversity in OSCC n.d. Greater homogeneity between OSCC and PVL-OSCC
Makinen et al. 2023 [45] Shannon index,Wilcoxon signed-rank test Lower diversity follow up vs basal PCA, permutational analysis of variance, Bray–Curtis distance,PERMANOVA Significant difference
Lan et al., 2023 [34] Shannon index, Simpson index, Chao1 index,ACE index,Wilcox test Significant differences between healthy and precancerous lesions PCA, Bray-Curtis distance, PERMANOVA Significant difference
Singh et al. 2023 [51] Shannon index, Chao1 index Higher in precancerous samples PCA based on the Bray-Curtis dissimilarity distance, UniFrac analysis Significant change between pre-cancer and cancer
ACE: abundance-based coverage estimator; OTUs: operational taxonomic units; PCoA: principal component analysis.
Table V. Alpha- and beta-diversity of oral microbiome in OSCC patients compared with healthy controls.
Author, year Sampling methods Detection methods
Hafed et al., 2019 [47] Tissue biopsy - PAS staining and Real-time PCR
Sankari et al., 2020 [40] Saliva collecting -DNA extraction by boiling lysis method -PCR-ITS1-5.8SrDNA-ITS2 region -Sequenced using ABI PRISM® BigDye™ Terminator e ABI 3730XL sequencer -NCBI GenBank
Arya et al., 2021 [39] Saliva swab Colony morphology on CHROM-agar Candida medium
Ilhan B. et al., 2023 [44] Tissue biopsy -Colony count Fungal/bacterial DNA isolation kit (Zymo Research, Irvine, CA, USA) -Tissue Samples High Pure PCR Template Preparation Kit
Rusanen et al., 2024 [46] Tissue biopsy -Colony count -Immunohistochemistry
Table VII. Sampling and polyphasic approach used in Candida spp. identification.
Author, year OSCC↑:
Hafed et al., 2019 [ 47 ] Candida spp.↑ n.d. HC:
Candida spp. ↑
Sankari et al., 2020 [40] C. krusei: 21% C. tropicalis: 21% Pichia anomala (ex C. pelliculosa): 21% C. famata: 17% C. rugosa: 6% OPMD (Potentially malignant oral diseases) Pichia anomala (ex C. pelliculosa): 33% C. krusei: 27% C. tropicalis: 10% C. famata: 9% C. rugosa: 7% HC: Pichia anomala (ex C. pelliculosa): 40% C. tropicalis: 24% C. krusei: 17%
Arya et al., 2021 [ 39 ] C. albicans ↑ C. krusei ↑ C. tropicalis ↑ C. parapsilosis ↑ C. albicans ↑ C. krusei ↑ C. tropicalis ↑ C. parapsilosis C. albicans ↑ C. krusei ↑ C. tropicalis ↑ C. parapsilosis ↑
Ilhan B. et al., 2023 [ 44 ] C. albicans ↑ C. glabrata ↑ C. sarcofago ↑ C. tropicalis ↑ C. parapsilosi ↑ Mild/moderate dysplasia C. albicans ↑ C. glabrata ↑ C. sarcofago ↑ C. tropicalis ↑ C. parapsilosi ↑ Carcinoma in situ C. albicans ↑ C. glabrata ↑ C. sarcofago ↑ C. tropicalis ↑ Benign C. albicans ↑ C. glabrata ↑ C. sarcofago ↑ C. parapsilosi ↑
Rusanen et al., 2024 [ 46 ] Candida spp. HC↑: Candida spp.
Table VIII. Summary of Candida spp. and their associations with oral conditions.

References

  1. Johnson D, Burtness B, Leemans C. Head and neck squamous cell carcinoma. Nat Rev Dis Primers. 2020;6:1-22. doi:https://doi.org/10.1038/s41572-020-00224-3
  2. Gondivkar S, Gadbail A, Sarode S. Prevalence of Trismus and Its Impact on Oral Health-Related Quality of Life in Patients Treated for Oral Squamous Cell Carcinoma. Asian Pac J Cancer Prev. 2021;22(8):2437-2444. doi:https://doi.org/10.31557/APJCP.2021.22.8.2437
  3. Kjeldsted E, Oksbjerg Dalton S, Frederiksen K. Association between human papillomavirus status and health-related quality of life in oropharyngeal and oral cavity cancer survivors. Oral Oncol. 2020;109. doi:https://doi.org/10.1016/j.oraloncology.2020.104918
  4. Petersen L, Jensen J, . Second primary cancer following primary oral squamous cell carcinoma: a population-based, retrospective study. Acta Oncol. 2022;61(8):916-921. doi:https://doi.org/10.1080/0284186X.2022.2079958
  5. Osazuwa-Peters N, Simpson M, Zhao L. Suicide risk among cancer survivors: Head and neck versus other cancers. Cancer. 2018;124(20):4072-4079. doi:https://doi.org/10.1002/cncr.31675
  6. Osazuwa-Peters N, Arnold L, Loux T. Factors associated with increased risk of suicide among survivors of head and neck cancer: A population-based analysis. Oral Oncol. 2018;81:29-34. doi:https://doi.org/10.1016/j.oraloncology.2018.03.017
  7. Sung H, Ferlay J, Siegel R. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209-249. doi:https://doi.org/10.3322/caac.21660
  8. Hanahan D, Weinberg R. The Hallmarks of Cancer. Cell. 2000;100(1):57-70. doi:https://doi.org/10.1016/S0092-8674(00)81683-9
  9. Hanahan D, Weinberg R. Hallmarks of Cancer: The Next Generation. Cell. 2011;144(5):646-674. doi:https://doi.org/10.1016/j.cell.2011.02.013
  10. Sun J, Tang Q, Yu S. Role of the oral microbiota in cancer evolution and progression. Cancer Med. 2020;9(17):6306-6321. doi:https://doi.org/10.1002/cam4.3206
  11. Mascitti M, Togni L, Troiano G. Beyond Head and Neck Cancer: The Relationship Between Oral Microbiota and Tumour Development in Distant Organs. Front Cell Infect Microbiol. 2019;9. doi:https://doi.org/10.3389/fcimb.2019.00232
  12. Lu M, Xuan S, Wang Z. Oral microbiota: A new view of body health. Food Sci Hum Wellness. 2019;8(1):8-15. doi:https://doi.org/10.1016/j.fshw.2018.12.001
  13. La Rosa G, Gattuso G, Pedullà E. Association of oral dysbiosis with oral cancer development. Oncol Lett. 2020;19(4):3045-3058. doi:https://doi.org/10.3892/ol.2020.11441
  14. Tuominen H, Rautava J. Oral Microbiota and Cancer Development. Pathobiology. 2020;88(2):116-126. doi:https://doi.org/10.1159/000510979
  15. Wei W, Li J, Shen X. Oral Microbiota from Periodontitis Promote Oral Squamous Cell Carcinoma Development via γδ T Cell Activation. mSystems. 2022;7(5):e00469-22. doi:https://doi.org/10.1128/msystems.00469-22
  16. Whitmore S, Lamont R. Oral Bacteria and Cancer. PLOS Pathog. 2014;10(3). doi:https://doi.org/10.1371/journal.ppat.1003933
  17. Zhang L, Liu Y, Zheng H. The Oral Microbiota May Have Influence on Oral Cancer. Front Cell Infect Microbiol. 2020;9. doi:https://doi.org/10.3389/fcimb.2019.00476
  18. Yang Z, Zhang S, Ji N. The evil companion of OSCC: Candida albicans. Oral Dis. 2024;30(4):1873-1886. doi:https://doi.org/10.1111/odi.14700
  19. Page M, McKenzie J, Bossuyt P. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372. doi:https://doi.org/10.1136/bmj.n71
  20. Ouzzani M, Hammady H, Fedorowicz Z. Rayyan—a web and mobile app for systematic reviews. Syst Rev. 2016;5(1). doi:https://doi.org/10.1186/s13643-016-0384-4
  21. Wells G, Shea B, O’Connell D. The Newcastle-Ottawa Scale (NOS) for Assessing the Quality of Nonrandomised Studies in Meta-Analyses. Ottawa Hospital Research Institute; 2014.
  22. Ganly I, Yang L, Giese R. Periodontal pathogens are a risk factor of oral cavity squamous cell carcinoma, independent of tobacco and alcohol and human papillomavirus. Int J Cancer. 2019;145(3). doi:https://doi.org/10.1002/ijc.32152
  23. Saxena R, Prasoodanan PKV, Vidushi Gupta S. Assessing the Effect of Smokeless Tobacco Consumption on Oral Microbiome in Healthy and Oral Cancer Patients. Front Cell Infect Microbiol. 2022;12. doi:https://doi.org/10.3389/fcimb.2022.841465
  24. Jain V, Baraniya D, El-Hadedy D. Integrative Metatranscriptomic Analysis Reveals Disease-specific Microbiome–host Interactions in Oral Squamous Cell Carcinoma. Cancer Res Commun. 2023;3(5):807-820. doi:https://doi.org/10.1158/2767-9764.CRC-22-0349
  25. Su S, Chang L, Huang H. Oral microbial dysbiosis and its performance in predicting oral cancer. Carcinogenesis. 2021;42(1):127-135. doi:https://doi.org/10.1093/carcin/bgaa062
  26. Yang K, Wang Y, Zhang S. Oral Microbiota Analysis of Tissue Pairs and Saliva Samples From Patients With Oral Squamous Cell Carcinoma – A Pilot Study. Front Microbiol. 2021;12. doi:https://doi.org/10.3389/fmicb.2021.719601
  27. Ye P, Liu Y, Cai Y. Microbial community alteration in tongue squamous cell carcinoma. Appl Microbiol Biotechnol. 2021;105(21):8457-8467. doi:https://doi.org/10.1007/s00253-021-11593-4
  28. Chen Q, Shao Z, Liu K. Salivary Porphyromonas gingivalis predicts outcome in oral squamous cell carcinomas: a cohort study. BMC Oral Health. 2021;21(1). doi:https://doi.org/10.1186/s12903-021-01580-6
  29. Rai A, Panda M, Das A. Dysbiosis of salivary microbiome and cytokines influence oral squamous cell carcinoma through inflammation. Arch Microbiol. 2021;203(1):137-152. doi:https://doi.org/10.1007/s00203-020-02011-w
  30. Zhong X, Lu Q, Zhang Q. Oral microbiota alteration associated with oral cancer and areca chewing. Oral Dis. 2021;27(2):226-239. doi:https://doi.org/10.1111/odi.13545
  31. Zeng B, Tan J, Guo G. The oral cancer microbiome contains tumor space–specific and clinicopathology-specific bacteria. Front Cell Infect Microbiol. 2022;12. doi:https://doi.org/10.3389/fcimb.2022.942328
  32. Heng W, Wang W, Dai T. Oral Bacteriome and Mycobiome across Stages of Oral Carcinogenesis. Microbiol Spectr. 2022;10(6):e02737-22. doi:https://doi.org/10.1128/spectrum.02737-22
  33. Liu Y, Li Z, Yanxu Q. Metagenomic Analysis Reveals a Changing Microbiome Associated With the Depth of Invasion of Oral Squamous Cell Carcinoma. Front Microbiol. 2022;13. doi:https://doi.org/10.3389/fmicb.2022.795777
  34. Lan Q, Zhang C, Hua H. Compositional and functional changes in the salivary microbiota related to oral leukoplakia and oral squamous cell carcinoma: a case control study. BMC Oral Health. 2023;23(1). doi:https://doi.org/10.1186/s12903-023-03760-y
  35. Chang C, Geng F, Shi X. The prevalence rate of periodontal pathogens and its association with oral squamous cell carcinoma. Appl Microbiol Biotechnol. 2019;103(3):1393-1404. doi:https://doi.org/10.1007/s00253-018-9475-6
  36. Zhou X, Hao Y, Peng X. The Clinical Potential of Oral Microbiota as a Screening Tool for Oral Squamous Cell Carcinomas. Front Cell Infect Microbiol. 2021;11. doi:https://doi.org/10.3389/fcimb.2021.728933
  37. Sarkar P, Malik S, Sayantan L. Dysbiosis of Oral Microbiota During Oral Squamous Cell Carcinoma Development. Front Oncol. 2021;11. doi:https://doi.org/10.3389/fonc.2021.614448
  38. Oyeyemi B, Kaur U, Paramraj A. Microbiome analysis of saliva from oral squamous cell carcinoma (OSCC) patients and tobacco abusers with potential biomarkers for oral cancer screening. Heliyon. 2023;9(11). doi:https://doi.org/10.1016/j.heliyon.2023.e21773
  39. Arya C, Jaiswal R, Tandon A. Isolation and identification of oral Candida species in potentially malignant disorder and oral squamous cell carcinoma. Natl J Maxillofac Surg. 2021;12(3):387-391. doi:https://doi.org/10.4103/njms.NJMS_80_19
  40. Sankari S, Mahalakshmi K, Kumar V. A comparative study of Candida species diversity among patients with oral squamous cell carcinoma and oral potentially malignant disorders. BMC Res Notes. 2020;13(1). doi:https://doi.org/10.1186/s13104-020-05336-3
  41. Torralba M, Aleti G, Li W. Oral Microbial Species and Virulence Factors Associated with Oral Squamous Cell Carcinoma. Microb Ecol. 2021;82(4):1030-1046. doi:https://doi.org/10.1007/s00248-020-01596-5
  42. Erira A, García Robayo D, Chalá A. Bacteriome identified by next-generation sequencing in saliva, dental plaque, and tumor tissue of patients with oral squamous cell carcinoma. Published online 2021. doi:https://doi.org/10.2174/1874285802115010098
  43. Herreros-Pomares A, Hervás D, Bagan-Debón L. On the Oral Microbiome of Oral Potentially Malignant and Malignant Disorders: Dysbiosis, Loss of Diversity, and Pathogens Enrichment. Int J Mol Sci. 2023;24(4). doi:https://doi.org/10.3390/ijms24043466
  44. İlhan B, Vural C, Gürhan C. Real-Time PCR Detection of Candida Species in Biopsy Samples from Non-Smokers with Oral Dysplasia and Oral Squamous Cell Cancer: A Retrospective Archive Study. Cancers (Basel). 2023;15(21). doi:https://doi.org/10.3390/cancers15215251
  45. Mäkinen A, Pappalardo V, Buijs M. Salivary microbiome profiles of oral cancer patients analyzed before and after treatment. Microbiome. 2023;11(1). doi:https://doi.org/10.1186/s40168-023-01613-y
  46. Rusanen P, Marttila E, Amatyaet S. Expression of Toll-like receptors in oral squamous cell carcinoma. PLoS One. 2024;19(4). doi:https://doi.org/10.1371/journal.pone.0300437
  47. Hafed L, Farag H, El-Roubyet D. Candida Albicans Alcohol Dehydrogenase 1 gene in oral dysplasia and oral squamous cell carcinoma. Pol J Pathol. 2019;70(3):210-216. doi:https://doi.org/10.5114/pjp.2019.90398
  48. Hashimoto K, Shimizu D, Ueda S. Feasibility of oral microbiome profiles associated with oral squamous cell carcinoma. J Oral Microbiol. 2022;14(1). doi:https://doi.org/10.1080/20002297.2022.2105574
  49. Unlu O, Demirci M, Paksoy T. Oral microbial dysbiosis in patients with oral cavity cancers. Clin Oral Investig. 2024;28(7). doi:https://doi.org/10.1007/s00784-024-05770-8
  50. Matsui K, Tani R, Yamasaki S. Analysis of Oral and Gut Microbiome Composition and Its Impact in Patients with Oral Squamous Cell Carcinoma. Int J Mol Sci. 2024;25(11). doi:https://doi.org/10.3390/ijms25116077
  51. Pratap Singh R, Kumari N, Gupta S. Intratumoral Microbiota Changes with Tumor Stage and Influences the Immune Signature of Oral Squamous Cell Carcinoma. Microbiol Spectr. 2022;11(4):e04596-22. doi:https://doi.org/10.1128/spectrum.04596-22
  52. Zhang L, Chen T, Wang Y. Comparison Analysis of Different DNA Extraction Methods on Suitability for Long-Read Metagenomic Nanopore Sequencing. Front Cell Infect Microbiol. 2022;12. doi:https://doi.org/10.3389/fcimb.2022.919903
  53. Prins T, Broothaerts W, Burns M. Guidance on the selection and use of DNA extraction methods. Publications Office of the European Union. Published online 2024.
  54. Marotz C, Sanders J, Zuniga C. Improving saliva shotgun metagenomics by chemical host DNA depletion. Microbiome. 2018;6(1). doi:https://doi.org/10.1186/s40168-018-0426-3
  55. Greathouse K, Sinha R, Vogtmann E. DNA extraction for human microbiome studies: the issue of standardization. Genome Biology. 2019;20(1). doi:https://doi.org/10.1186/s13059-019-1843-8
  56. Gopinath D, Menon R, Banerjee M. Culture-independent studies on bacterial dysbiosis in oral and oropharyngeal squamous cell carcinoma: A systematic review. Crit Rev Oncol Hematol. 2019;139:31-40. doi:https://doi.org/10.1016/j.critrevonc.2019.04.018
  57. Johnson J, Spakowicz D, Hong B. Evaluation of 16S rRNA gene sequencing for species and strain-level microbiome analysis. Nat Commun. 2019;10(1). doi:https://doi.org/10.1038/s41467-019-13036-1
  58. Liu P, Wu W, Chen C. Evaluation of Compatibility of 16S rRNA V3V4 and V4 Amplicon Libraries for Clinical Microbiome Profiling. bioRxiv. 2020;2020. doi:https://doi.org/10.1101/2020.08.18.256818
  59. Loman N, Misra R, Dallman T. Performance comparison of benchtop high-throughput sequencing platforms. Nat Biotechnol. 2012;30(5):434-439. doi:https://doi.org/10.1038/nbt.2198
  60. Bars-Cortina D, Ramon E, Rius-Sansalvador B. Comparison between 16S rRNA and shotgun sequencing in colorectal cancer, advanced colorectal lesions, and healthy human gut microbiota. BMC Genomics. 2024;25(1). doi:https://doi.org/10.1186/s12864-024-10621-7
  61. d’Enfert C, Kaune A, Alabanet L. The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: current knowledge and new perspectives. FEMS Microbiol Rev. 2021;45(3). doi:https://doi.org/10.1093/femsre/fuaa060
  62. Liscia D, D’Andrea M, Bilettaet E. Use of digital pathology and artificial intelligence for the diagnosis of Helicobacter pylori in gastric biopsies. Pathologica - Journal of the Italian Society of Anatomic Pathology and Diagnostic Cytopathology. 2022;114:295-303. doi:https://doi.org/10.32074/1591-951X-751
Authors

Angela Crispino - Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples “Federico II”, Naples, Italy https://orcid.org/0009-0003-6344-7942

Silvia Varricchio - Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples “Federico II”, Naples, Italy

Aurora Esposito - Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy

Alessandra Marfella - Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples “Federico II”, Naples, Italy

Dora Cerbone - Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples “Federico II”, Naples, Italy

Angelica Perna - Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy https://orcid.org/0000-0002-7853-9159

Giulio Petronio Petronio - Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy

Stefania Staibano - Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples “Federico II”, Naples, Italy

Francesco Merolla - Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy https://orcid.org/0000-0003-0711-6738

Gennaro Ilardi - Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples “Federico II”, Naples, Italy https://orcid.org/0000-0002-9354-4087

How to Cite
Crispino, A., Varricchio, S., Esposito, A., Marfella, A., Cerbone, D., Perna, A., Petronio Petronio, G., Staibano, S., Merolla, F. ., & Ilardi, G. (2024). The oral microbiome and its role in oral squamous cell carcinoma: a systematic review of microbial alterations and potential biomarkers. Pathologica - Journal of the Italian Society of Anatomic Pathology and Diagnostic Cytopathology, 116(6). https://doi.org/10.32074/1591-951X-N867
  • Abstract viewed - 392 times
  • PDF downloaded - 185 times
  • SUPPL. FILE downloaded - 1 times