Next Article in Journal
Biopatinas on Peperino Stone: Three Eco-Friendly Methods for Their Control and Multi-Technique Approach to Evaluate Their Efficacy
Next Article in Special Issue
The Oral–Gut Microbiome–Brain Axis in Cognition
Previous Article in Journal
Metagenomic and Metabolomic Analyses Reveal the Role of a Bacteriocin-Producing Strain of Enterococcus faecalis DH9003 in Regulating Gut Microbiota in Mice
Previous Article in Special Issue
Association of Longitudinal Oral Microbiome Activity and Pediatric Concussion Recovery
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Global Trends and Advances in Oral Microbiome Research on Oral Squamous Cell Carcinoma: A Systematic Review

by
Ramona Dumitrescu
1,2,
Vanessa Bolchis
1,2,*,
Aurora Doris Fratila
3,
Daniela Jumanca
1,2,
Berivan Laura Rebeca Buzatu
1,2,
Ruxandra Sava-Rosianu
1,2,
Vlad Tiberiu Alexa
1,2,
Atena Galuscan
1,2 and
Octavia Balean
1,2
1
Translational and Experimental Clinical Research Centre in Oral Health, University of Medicine and Pharmacy “Victor Babes”, 300040 Timisoara, Romania
2
Clinic of Preventive, Community Dentistry and Oral Health, University of Medicine and Pharmacy “Victor Babes”, Eftimie Murgu Sq. no 2, 300041 Timisoara, Romania
3
Faculty of Dental Medicine, Ludwig Maximilian University of Munich, Goethestrasse 70, 80336 Munich, Germany
*
Author to whom correspondence should be addressed.
Microorganisms 2025, 13(2), 373; https://doi.org/10.3390/microorganisms13020373
Submission received: 17 January 2025 / Revised: 3 February 2025 / Accepted: 6 February 2025 / Published: 8 February 2025
(This article belongs to the Special Issue Oral Microbes and Human Health)

Abstract

:
The oral microbiome is increasingly recognized as a key factor in the development and progression of oral squamous cell carcinoma (OSCC). Dysbiosis has been associated with inflammation and tumorigenesis, highlighting the potential of microbial alterations and salivary biomarkers as tools for early, non-invasive diagnosis. This review examines recent advancements in understanding the oral microbiome’s role in OSCC. A comprehensive synthesis of studies from 2016 to 2024 was conducted to identify emerging themes and significant findings in the field. Key topics included the interplay between microbiome-driven mechanisms and cancer development, with a focus on microbial communities and their metabolic byproducts. The findings emphasize the importance of specific microbial alterations in modulating immune responses and tumor microenvironments, as well as the promise of biomarkers such as interleukins and miRNA signatures in improving diagnostic accuracy. Recent research trends indicate growing interest in the therapeutic potential of targeting the oral microbiome in OSCC management. Despite significant advancements, gaps remain in the understanding of the precise mechanisms linking dysbiosis to cancer progression. This review underscores the need for continued research to develop personalized diagnostic and therapeutic strategies based on the oral microbiome, with the potential to transform OSCC management.

1. Introduction

The term “microbiome” was coined by Joshua Lederberg, a Nobel Prize laureate, to denote the diverse community of symbiotic, commensal, and pathogenic microorganisms residing within our bodies [1]. These microorganisms collectively inhabit our body space, contributing to an intricate ecological system. Remarkably, the abundance of microbes in our bodies rivals, if not exceeds, that of our own cells [2]. Among these microbiomes, the oral microbiome specifically refers to the array of microorganisms inhabiting the oral cavity of humans.
First recognized in 1674 by Antony van Leeuwenhoek, the oral microbiome has since become a key focus in microbial ecology, with research significantly advancing to reveal its crucial impact on both local and systemic health [3,4].
Recent advancements in salivary metabolomics have highlighted the potential of salivary biomarkers for the early detection and monitoring of various diseases. Salivary metabolites, derived from both oral microbes and host sources, provide valuable insights into physiological and pathological processes [5]. Saliva is a valuable biological fluid containing a diverse array of biomarkers derived from serum, gingival crevicular fluid, and oral microorganisms and their products, making it an attractive option for laboratory tests due to its non-invasive collection, ease of transportation and storage, cost-effectiveness, and efficiency [6,7,8]. Various molecular techniques are employed to detect and determine biomarkers, including DNA microarrays, the polymerase chain reaction (PCR), liquid chromatography, mass spectrometry, and nuclear magnetic resonance, among others. Oral squamous cell carcinoma (OSCC), a prevalent form of oral cancer, is characterized by significant diagnostic challenges due to its asymptomatic nature in early stages and poor 5-year survival rates, which remain unimproved despite advanced combination treatments such as surgery, chemotherapy, and radiotherapy. OSCC development has been closely linked to the oral microbiome, which triggers inflammatory responses through cytokines and chemokines, stimulating tumor cell proliferation and survival, emphasizing the importance of identifying salivary biomarkers for early diagnosis and improved prognosis [9].
Traditional diagnostic methods often fail to detect OSCC until it reaches advanced stages, underscoring the need for non-invasive and reliable biomarkers. Our study investigates the salivary metabolome of OSCC patients, identifying key metabolites that distinguish cancerous from non-cancerous states and exploring their potential as diagnostic and prognostic indicators. Salivary biomarkers have gained significance in the screening and early detection of oral squamous cell carcinoma (OSCC), with over 100 potential biomarkers reported in the literature. These biomarkers can be classified based on disease state, biomolecules, or other criteria, with diagnostic markers such as EFNB2 gene expression, interleukins (IL-6 and IL-8), and 8-oxoguanine DNA glycosylase being of particular interest for OSCC screening and prognosis evaluation. For IL-6, Rani et al. (2023) [10] reported a sensitivity of 85% and specificity of 78% (AUC = 0.82), demonstrating its diagnostic utility for OSCC. Similarly, salivary transferrin has shown an AUC of 0.89 in early-stage OSCC detection (Tavakoli et al., 2024 [11]). These findings highlight the potential of these biomarkers for non-invasive diagnostic applications. Recent efforts have focused on non-invasive methods for understanding OSCC genomic architecture, utilizing proteomic, transcriptomic, and metabolomic biomarkers extracted from human saliva samples [12].
Despite ongoing efforts in early screening and diagnosis, the burden of oral cancer treatment is expected to increase significantly due to factors such as its asymptomatic nature in early stages, which often leads to late diagnosis, and the limited improvements in survival rates despite advances in surgery, chemotherapy, and radiotherapy [13,14].
According to the latest data from the Institute for Health Metrics and Evaluation (IHME) on the global burden of disease (GBD), oral cancer poses a significant global health concern, with an estimated 373,000 incident cases, 199,000 deaths, and 5.51 million disability-adjusted life years (DALYs) in 2019 alone [15]. Research efforts towards oral cancer have been continuously rising, covering various aspects including diagnostic, prognostic, and therapeutic modalities, as well as qualitative and health system analyses.
The complex etiology of cancer involves both environmental and heritable risk factors, with increasing evidence suggesting that the microbiome plays a key role in modulating the carcinogenic process through distinct mechanisms. These include microbiome-driven dysbiosis leading to immune evasion, the production of pro-inflammatory cytokines such as IL-6 and TNF-α that contribute to chronic inflammation, and microbial metabolites influencing oncogenic pathways. While the microbiota harbors beneficial effects including active communication with host cells and maintenance of an environment rich in nutrients, it also poses risks by producing toxins that can induce mutations, alter signaling pathways, and promote cancer development in both oral and systemic contexts [16].
Given the accessibility of the oral cavity for non-invasive biological sampling, salivary-based diagnostic tools are increasingly being explored for their potential in early OSCC detection and personalized risk assessment. Future research should focus on refining these biomarkers to improve sensitivity and specificity, ultimately contributing to precision medicine approaches in oncology [17].
Bibliometric analysis [18] objectively evaluates extensive data from the past to quantify a study’s impact within its specific scientific field. Bibliometric analysis plays a crucial role in assessing, evaluating, and visualizing research trends and evidence within a specific field, thereby enabling future researchers and stakeholders to make informed decisions regarding research priorities [19,20].
A thorough bibliometric analysis should encompass both performance analysis, which quantifies citation and impact counts, and science mapping, which examines the key trends, topics, and collaboration networks among authors, affiliations, and countries involved [21].
Although many studies have explored the link between bacteria and oral cancer, no bibliometric analysis has been conducted on this topic. The oral microbiome’s impact on systemic health is gaining attention, as microbial metabolites influence both local and systemic conditions. Salivary metabolic profiling provides insights into disease mechanisms and potential therapies. By examining the salivary metabolome’s role in OSCC and its systemic implications, our research enhances the understanding of salivary metabolomics and its clinical applications. In this study, we conducted a comprehensive bibliometric analysis focusing on the literature published in the last eight years. Our analysis encompassed article citations, countries of origin, publishing journals, authors and their affiliations, highly cited studies, and keywords. Additionally, we explored research trends and identified hotspots. The aim of this analysis is to develop a set of salivary biomarkers for the oral microbiome and cytokines, with the aim of evaluating OSCC patients based on variations in these parameters, in the meantime offering researchers an unbiased comprehension of the research landscape in this domain, and to serve as a guide for future in-depth inquiries [12,22,23,24].

2. Materials and Methods

2.1. Data Sources, Collection, and Processing

The methodology employed in the present study’s analysis is derived from the bibliometric handbook on conducting bibliometric analyses by Rehn et al., published in 2014 [25]. The data under analysis were extracted from the Scopus database, a comprehensive and highly regarded platform that indexes a wide array of scholarly literature across multiple disciplines. Known for its extensive coverage, Scopus includes more than 82 million records from over 25,000 peer-reviewed journals, conference proceedings, and patents, encompassing scientific, technical, medical, and social sciences fields. From this reliable source, we gathered data focusing on recent advancements in our understanding of the human microbiome, particularly the oral microbiome and its connections to human health.
The Scopus database was selected due to its extensive multidisciplinary coverage and its capability to perform bibliometric analyses. While other databases, such as PubMed and Web of Science, also provide valuable studies, the decision to use Scopus ensures data consistency and minimizes redundancy in the literature analyzed. This approach aligns with previous bibliometric studies that have also relied on a single database for methodological rigor and analytical depth, such as Zyoud et al. (2022) [26] and Yuan et al. (2021) [27], both of which utilized Scopus to analyze microbiome-related research trends.
This research, focused on studies conducted on oral cancer and the oral microbiome spanning from January 2016 to April 2024, was carried out in April 2024 to mitigate database renewal bias. The Scopus database was selected for its robust collection of medical literature and comprehensive citation analysis capabilities. The search strategy, depicted in Figure 1, incorporated specific terms related to cancer (e.g., carcinoma, tumor) and the oral region (e.g., oral, mouth), along with terms associated with bacteria and the microbiota, while excluding viral, fungal, and mycotic terms. This meticulous approach ensured the inclusion of relevant and high-quality data pertinent to our study.
Several oral and systemic diseases associated with the oral microbiome were identified, including caries, periodontal diseases, and oral cancer, which have garnered significant attention within the scientific community. A combination of search terms, including “oral squamous cell carcinoma”, “oral microbiome”, “dysbiosis”, and “salivary biomarkers”, was employed using appropriate logical operators to yield relevant results. Initially, a sample of 826 publications spanning from 2016 to 2024 was obtained, which was subsequently narrowed down to 82 documents pertinent to dentistry, excluding other fields and publications not in English.
This bibliometric analysis was conducted in accordance with the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) 2020 guidelines. PRISMA provides a clear set of directives for systematic and meta-analytic reporting, thereby facilitating the evaluation and reproducibility of studies. In this research, we followed the PRISMA-recommended steps, including the formulation of a comprehensive search strategy, rigorous assessment of bias risk, and clear presentation of results via the PRISMA flow diagram. By adhering to these standards, we ensured a robust and transparent methodological approach in exploring changes in the oral microbiome in the context of oral squamous cell carcinoma and identifying relevant salivary biomarkers [28]. This systematic review, conducted retrospectively and focused on bibliometric analysis, adhered to the PRISMA 2020 guidelines to ensure transparency and reproducibility, although it was not pre-registered in a review database (Figure 2) (Supplementary Materials).

2.2. Risk of Bias

To systematically evaluate the methodological quality of the included studies, we utilized the ROB2 tool (Cochrane Risk of Bias Tool 2.0). This tool is a widely recognized and validated approach for assessing the risk of bias in randomized controlled trials, focusing on five critical domains: the randomization process, adherence to interventions, completeness of outcome data, measurement of outcomes, and selection of reported results. Each domain was assessed systematically to identify potential sources of bias that could influence the reliability or validity of the study findings. Domain-level evaluations were rated as “Low Risk”, “Some Concerns”, or “High Risk”, with clear criteria guiding each judgment. For example, in the domain of randomization, we assessed whether allocation concealment and sequence generation were appropriately implemented, while for adherence to interventions, we examined deviations from the intended protocol. The assessments were conducted independently by two trained reviewers to minimize subjective interpretation and ensure consistency. In cases of disagreement, discussions were facilitated to achieve consensus, and a third reviewer was consulted to resolve persistent conflicts. This multi-step approach reduced individual bias and strengthened the validity of the evaluation process. To ensure transparency, all judgments were documented with detailed justifications, allowing reproducibility of the evaluation process. Additionally, results were synthesized to provide an overall risk of bias for each study, highlighting areas of methodological strength and weakness. By employing this rigorous and standardized assessment framework, we enhanced the transparency, consistency, and reliability of the included studies’ evaluation, thereby providing a solid foundation for interpreting the findings of this systematic review [29].

2.3. Bibliometric Analysis

For data analysis and visualization, we utilized VOSviewer (version 1.6.20), enabling us to identify and explore research trends and clusters of interest in the field of the oral microbiome and associated diseases.
VOSviewer, a freely available software tool, is utilized to construct and visualize bibliometric networks, offering insights into research clusters, current interests, and emerging trends. Such networks encompass scientific publications or journals, keywords, researchers, countries, research organizations, or other specified inclusion criteria terms. These networks are established through various types of links, including citations, scientific co-authorship, co-occurrence, co-citations, and bibliographic connections. As outlined in the VOSviewer Manual, a link signifies a relation or connection between two items, with each link possessing a strength denoted by different positive numerical values corresponding to the type of analyzed items. For instance, the strength of a link in co-authorship networks could represent the number of documents co-authored by two researchers, while in bibliographic coupling networks, it could indicate the number of cited references shared by two documents [30].
Various analyses, including co-authorship, keyword co-occurrence, and co-citation, were utilized to explore interactions and relationships among publications, keywords, and authors in the field of study. These methods deepened our understanding of the evolution and research directions in the oral microbiome and related areas. Descriptive bibliometric analysis captured both objective indexes (e.g., number of publications, authors, affiliations, citation counts, and country/region) and subjective indexes (e.g., keywords). Version 1.6.20 of VOSviewer was employed to generate bibliometric network maps for co-authorship by country and for keywords [30]. To assess research trends and impact in the field of the oral microbiome and OSCC, a Spearman’s correlation analysis was conducted between the number of publications per year and their citation metrics. The analysis revealed a statistically significant positive correlation (ρ = 0.85, p < 0.01), indicating a steady increase in research interest and impact. This trend highlights the growing attention toward microbiome-based biomarkers and the integration of artificial intelligence in OSCC diagnostics.

3. Results

3.1. Study Selection, Data Extraction, and Risk of Bias

The data collection process was conducted by two independent reviewers. Data collection was performed manually using a predefined data extraction form to ensure consistency and transparency. The collected variables included study characteristics, population demographics, interventions, biomarkers analyzed, outcomes, and information on funding sources. Missing or unclear information was managed by excluding studies with incomplete data.
The statistical analysis further supports the observed research growth. The Spearman’s correlation analysis demonstrated a strong positive correlation (ρ = 0.85, p < 0.01) between publication counts and citation metrics, confirming an increasing trend in research activity. This suggests that microbiome-based OSCC studies have gained substantial academic and clinical relevance, with a significant rise in interest over the past eight years.
Effect measures, such as mean differences and risk ratios, were used to interpret the results depending on the type of biomarker. Studies were synthesized through tabulation and visual representation, and the methodological heterogeneity observed among the included studies was qualitatively assessed. This heterogeneity was primarily attributed to differences in study designs and biomarker measurement techniques. Reporting bias was not formally evaluated; however, studies excluded due to incomplete data were documented to ensure transparency in the selection process.
The certainty of the evidence was not assessed using formal frameworks, such as GRADE. Nevertheless, the methodological limitations of the included studies were discussed to adjust the overall confidence in the findings. These limitations, including the prevalence of high-risk domains, were considered in the interpretation of results and conclusions.
Both reviewers systematically extracted relevant information from the included studies, using the predefined data extraction form. Any discrepancies or disagreements between the reviewers were resolved through mutual discussion to ensure consistency and accuracy in the collected data. No third-party arbitration was required as all disagreements were resolved collaboratively. No automation tools were used in the data collection process, and all steps were conducted manually to ensure accuracy and consistency.
Figure 3 presents the distribution of the risk of bias across the included studies. The most common sources of bias were identified in the domains of “Bias due to missing outcome data” and “Bias in selection of the reported result”, where a significant number of studies were classified as high-risk (red). These findings highlight potential limitations in data completeness and reporting practices, which could impact the overall reliability and validity of the conclusions drawn from these studies. Conversely, the majority of studies demonstrated a low risk of bias (green) in the domains of “Bias arising from the randomization process” and “Bias in measurement of the outcome”, reflecting adequate methodologies in these areas.

3.2. Keyword Analysis of Research Themes in Oral Microbiome and Oral Cancer

Between 2016 and 2024, a total of 82 articles on the topic of the oral microbiome, salivary biomarkers, and oral cancer met the search criteria and were included in the evaluation.
These results underscore the need for improved reporting and data management practices in future research. Addressing issues related to missing data and selective reporting could significantly enhance the overall quality of evidence in this field. Moreover, by systematically identifying and visualizing these risks, this assessment provides a foundation for prioritizing methodological improvements in future studies.
A keyword analysis was conducted using VOSviewer (Center for Science and Technology Studies, Leiden University, The Netherlands) to discern the predominant research themes in the literature concerning the role of the microbiome in oral cancer development. This method highlights the most relevant keywords based on their frequency, indicated by the number of articles in which a keyword appears at least six times. This means that out of 2408 terms, only 82 meet the threshold, and we chose just the 60% most relevant of them. For our dataset, we chose to generate a map of the network of keywords with a minimum of six occurrences in the analyzed articles, such as oral microbiome (nine occurrences), OSCC (seventy-four occurrences), salivary biomarkers (sixteen occurrences), and early detection (eleven occurrences), in VOSviewer. The most important keywords and the links between them are presented in Figure 4. A larger node (keyword) indicates a greater weight (a higher number of occurrences); a smaller distance between nodes indicates a stronger relationship between them; the same color indicates a series of related keywords or a group of related keywords. Additionally, Table 1 provides a detailed description of these keyword groups, highlighting their occurrences and total link strength within the analyzed dataset.

3.3. Analysis of “Co-Authorship” in Terms of Number of Citations

Research interest in the topic of metabolic pathways can be identified by the number of authors addressing this subject, more concretely through the co-authorship relationships established during scientific research. The analysis in this section focuses on identifying the strongest co-authorship relationships on the analyzed topic, thereby visualizing the most relevant author groups. Out of a total of 553 authors identified in the analyzed articles, 77 of these are cited at least 50 times. Additionally, two authors have met the threshold of at least 50 citations per published document, while four authors have met the threshold of 556 citations per published document. However, their level of collaboration differs, which is why a graphical representation of all 553 authors is not relevant. Individual researchers who do not form clusters with multiple authors are specifically mentioned, while 77 authors have managed to form research teams with significant importance for the analyzed topic. Figure 5 distinguishes the clusters composed of authors collaborating on this topic. Furthermore, the analysis has concentrated on the research domain of the main authors’ network, specifically targeting the primary objective: alterations in the oral microbiome associated with oral and systemic pathology. From the total, 82 articles were selected based on their relevance and citation thresholds to ensure a focused and detailed examination of the collaborative research efforts in this field.
Table 2 organizes the primary authors based on color-coded groups, highlighting the number of citations per document and the total link strength (TLS) for each group. Each cell under the “Main Authors” column lists the authors associated with the respective color group, reflecting their collaborative network and research impact within the domain of oral microbiome alterations related to oral and systemic pathologies. The “Citations/Document” column indicates the minimum citations each author has received per published document, while the “TLS” column shows the cumulative strength of their collaborative links, illustrating the intensity and number of collaborations among these researchers.

3.4. Analysis of International Co-Authorship Networks in Scientific Research

To better understand the dynamics of international research collaborations, this bibliometric map provides a detailed visualization of co-authorship networks based on data obtained from Scopus.
This bibliometric map was generated using VOSviewer software and is based on bibliographic data. The analysis was conducted using the “co-authorship” type and the “full counting” method. The parameters of the analysis included a maximum of 25 countries per document, a minimum of one document per country, and a minimum of 50 citations per country. Of the 36 countries that met the citation threshold, 21 were interconnected, resulting in this map.
Figure 6 illustrates the collaboration networks between various countries in scientific publications indexed in Scopus. The United States is shown as a central node with numerous connections to other countries, highlighting its significant role in the global co-authorship network. Other notable countries, such as the United Kingdom, Japan, and Australia, also show multiple international connections. In contrast, India and China, although very active, are depicted with fewer direct connections to other countries, reflecting more limited international collaboration.

3.5. Visualizing Collaborative Networks in Dental Research: A Co-Authorship Bibliometric Analysis of Key Organizations

This bibliometric map, created using VOSviewer, visualizes the co-authorship networks among various organizations that have published articles in the field of dentistry, based on data from Scopus. The analysis used the full counting method with a maximum of 25 organizations per document and set thresholds of at least one document and 100 citations per organization. Out of twelve organizations meeting these criteria, nine were interconnected. Key organizations include the School of Dentistry and Oral Health at Griffith University, the Preventive Oral Health Unit at The National Dental Hospital, the School of Medical Science at Griffith University, the Dental Institute at King’s College London, and the Maurice H. Kornberg School of Dentistry at Temple University. The nodes represent organizations, while the links between them indicate the frequency and intensity of their collaborative efforts, highlighting the influential research networks in the field of dental science (Figure 7).

3.6. Exploring Co-Occurrence Relationships: Insights from Oral Microbiome Research

In this study, we conducted a detailed analysis of the relationships between key terms in the literature concerning the oral microbiome and associated diseases, using VOSviewer software. For mapping purposes, we utilized bibliographic data downloaded from Scopus and applied various analytical techniques, including co-occurrences, author keywords, and full counting.
After applying a minimum threshold of four co-occurrences, we identified 17 keywords that met this criterion. Among these, “OSCC” and “saliva” were the most frequently encountered, with 38 occurrences. “Biomarkers” were mentioned ten times, while “or cancer” was mentioned twelve times, and the “oral microbiome” was mentioned five times.
These statistics provide a clear picture of researchers’ concerns and interests in the field, highlighting the significance of the oral microbiome in the context of oral and systemic diseases. The resulting map enables us to visualize the networks of co-occurrences among these key terms, providing a comprehensive perspective on the interactions within the literature. A detailed analysis of these relationships can contribute to a deeper understanding of the complexity of the oral microbiome and its impact on human health (Figure 8).

4. Discussion

Recent insights into the human microbiome have shed light on the intricate role this microbial community plays in our health, especially focusing on the oral microbiome. This deeper understanding has unveiled a wide array of diseases linked to the oral microbiome, encompassing both oral and systemic conditions such as caries, periodontal diseases, oral cancer, colorectal cancer, pancreatic cancer, and irritable bowel syndrome. Moreover, advancements in our comprehension of the human microbiome, with a particular emphasis on the oral microbiome, have highlighted its vital impact on health and disease. This study utilized bibliometric methods to thoroughly review the literature concerning the oral microbiome’s role in oral squamous cell carcinoma (OSCC). The findings showed a notable rise in publications and citations, reflecting the growing interest and acknowledgment of the oral microbiome’s importance in OSCC [17].
The initial focus of oral cancer research primarily centered around epidemiology and the characteristics of oral cancer, likely as part of a population health strategy to identify high-prevalence groups and implement preventive measures. However, there has been a noticeable shift towards investigating cellular/molecular pathways, the tumor microenvironment, the microbiome, therapeutic targets, and biomarkers. These research areas have the potential to empower clinicians to intervene in the pathology at an earlier stage and develop treatment strategies based on specific therapeutic targets, facilitating more precise and minimally invasive treatment approaches. The publication landscape reflects a global interest in oral cancer, with prominent affiliations and countries frequently collaborating on similar research endeavors.
This study offers a thorough assessment of academic publications examining the link between oral cancer and bacteria from 2016 to 2024. The analysis highlights a steady rise in publication numbers over time, indicating a growing interest in this research domain. Furthermore, the citations garnered by these articles exhibit a positive trend, emphasizing their impact and importance in the scientific community. This mirrors the increasing global severity of oral cancer [31] and the pressing necessity to comprehend the role of bacteria in its development [32].
A total of 82 relevant articles on the oral microbiome and oral squamous cell carcinoma (OSCC) from 2016 to 2024 were included, with an average annual publication count of 10.25. The field has shown a steady increase in research interest, reflecting the growing recognition of the oral microbiome’s role in oral cancer. However, during the COVID-19 pandemic, particularly in 2020, there was a noticeable stagnation in publication growth, likely due to research disruptions and the reallocation of resources to pandemic-related studies. Despite the pandemic’s impact, the publication count resumed its growth post 2020, underscoring the field’s resilience and the sustained interest in exploring the oral microbiome’s implications in cancer research. Notably, the 2022 publication count did not reach expected levels, attributed to ongoing pandemic-related challenges and potential publication delays (Figure 9).
Several key studies have contributed to the understanding of the oral microbiome’s role in OSCC. Among the authors investigated, Ahmed et al. in 2024 demonstrated in their study that saliva DNA is a viable medium for detecting somatic mutations in patients with OSCC, with a detection rate of 82% regardless of the tumor stage or primary site, though sensitivity may be limited by low variant allele frequencies (VAFs); while traditional methods such as droplet digital PCR (ddPCR) offer high sensitivity, advancements in panel sequencing and bioinformatics, such as the Integrated Variant Analysis (INVAR) pipeline, may enhance detection capabilities, suggesting a promising avenue for the non-invasive monitoring of OSCC recurrence and treatment response [33]. Rapado-González’s study demonstrates that advanced DNA extraction and analysis techniques, including the use of specialized kits for Formalin-Fixed Paraffin-Embedded (FFPE) tissues and saliva samples, enable the precise identification of genome-wide DNA methylation changes. The methodologies employed, such as bisulfite conversion and methylation panel sequencing, along with sophisticated bioinformatic tools, facilitate detailed DNA methylation profiling, ensuring rigorous quality control and data normalization. These techniques revealed significant differences in DNA methylation and gene expression between OSCC samples and healthy controls, highlighting their potential for disease diagnosis and monitoring [34]. At the same time, Scheurer et al. show the potential of specific miRNA signatures from saliva to distinguish between healthy individuals and OSCC patients, suggesting that standardized protocols for miRNA analysis could be developed to facilitate the early detection of malignant changes, although further validation in larger and more homogeneous studies is necessary to confirm these findings [35]. Zhu et al. indicate that Capnocytophaga gingivalis, highly abundant in OSCC tissues, plays a crucial role in promoting OSCC invasion and metastasis by inducing the epithelial–mesenchymal transition (EMT), suggesting a novel direction for OSCC research focused on the microbial influence on cancer progression [36]. Riccardi et al. in their recent study highlight the potential of salivary transferrin as a biomarker for the early diagnosis of OSCC, emphasizing its role in iron transport and cell proliferation processes; despite promising results indicating suitable sensitivity and specificity, further research is needed to validate these findings and improve early detection strategies for better prognosis in OSCC patients [37,38].
The microbiome profile of OSF-related malignancy showed increased microbial stochastic fluctuation and species co-occurrence network collapse. Artificial intelligence algorithms identified five key species in the OSCC-OSF group, Porphyromonas catoniae, Prevotella multisaccharivorax, Prevotella sp. HMT-300, Mitsuokella sp. HMT-131, and Treponema sp. HMT-927, with robust accuracy in predicting oral carcinogenesis. Functional analysis indicated differences in microbial metabolite potential, suggesting roles in modulating metabolites during oral carcinogenesis. Overall, these findings provide new insights into salivary microbiome alterations during the malignant transformation of OSF [39].
Our analysis unveiled an expanding interest in the oral microbiome within oral cancer research. Studies indicate that dysbiosis in the oral and periodontal microbiome may heighten the risk of oral cancer [40,41].
At the same time, Scheurer et al. highlight in their study the significant role of specific microbial species in oral carcinogenesis, particularly in the transition from normal oral mucosa to OSCC. The genus Gemella, along with species such as Streptococcus, S. agalactiae, and G. haemolysans, were found to be enriched in OSCC samples. These findings suggest that these microbial species may be involved in the metabolic processes associated with cancer development. The study also notes that metabolic pathways, including those related to cysteine and methionine metabolism, are significantly altered in OSCC, pointing to the potential involvement of these microbes in creating a pro-carcinogenic environment. Further research with larger, diverse populations is necessary to confirm these associations and explore their potential as early diagnostic biomarkers for oral cancer [9].
Rani et al. demonstrate the potential of salivary IL-6 as a diagnostic biomarker for oral cancer, particularly OSCC, OPMDs, and CP. Elevated levels of salivary IL-6 were consistently observed in OSCC and moderately in OPMDs, indicating its potential as a reliable indicator of oral malignancy. These findings align with previous research highlighting the diagnostic role of IL-6 in oral cancer, reinforcing its significance as a non-invasive diagnostic tool. Moreover, our study underscores the importance of salivary IL-6 in differentiating between healthy individuals and those with oral lesions, emphasizing its utility in early detection and disease monitoring. While further research is warranted to validate these findings across diverse populations and refine diagnostic criteria, the evidence presented by Rani et al. supports the promising role of salivary IL-6 as a valuable biomarker for oral cancer detection and management [10].
Hashimoto et al.’s study demonstrated significant differences in microbiome profiles in saliva, with an increase in Fusobacteria and Fusobacterium and a decrease in Firmicutes and Streptococcus observed in the OSCC group compared to non-OSCC groups. These findings suggest the potential of these bacterial taxa as novel biomarkers for OSCC detection. Additionally, our analysis of patients with early recurrence suggests the prognostic value of the oral microbiome. Although this study could not elucidate the precise mechanisms by which these bacterial biomarkers influence carcinogenesis and tumor progression, they hold promise for identifying high-risk cases of oral leukoplakia or recurrence in postoperative OSCC patients. Further prospective studies are warranted to validate the clinical utility of oral microbiome profiling in oral diseases [42].
Recent advancements, such as AI-driven models for microbiome analysis, have demonstrated their ability to predict OSCC with high accuracy. For example, Chen et al. (2023) utilized machine learning algorithms to identify microbial patterns associated with early-stage OSCC, achieving a diagnostic accuracy of 92% [43].
By employing keyword analysis with VOSviewer, we identified four main research themes in oral cancer and the microbiome. The first theme emphasized the importance of sensitivity and specificity, focusing on reliable biomarkers like IL-6 and miRNA for the early detection of oral cancer. The second theme explored the interplay between periodontitis, the oral microbiome, and cancer, highlighting how dysbiosis may increase oral cancer risk. The third theme concentrated on malignant disorders and inflammation, examining specific bacteria’s roles in carcinogenesis and chronic inflammation. The final theme addressed evaluation and prognosis, analyzing risk factors and potential markers to develop preventive and diagnostic strategies, ultimately aiming for early intervention and better patient outcomes.
Salivary biomarkers have gained increasing attention for OSCC screening due to their non-invasive nature and potential diagnostic accuracy. For example, Fusobacterium nucleatum, associated with OSCC, can be incorporated into diagnostic panels through targeted qPCR assays, enabling the early detection and monitoring of tumor progression. Similarly, Capnocytophaga gingivalis has shown potential as a microbial marker for distinguishing cancerous from non-cancerous states. These practical applications bridge the gap between theoretical findings and their implementation in clinical diagnostics.
The co-authorship and co-occurrence analyses highlighted significant international and interdisciplinary collaborations. The United States emerged as a central node with numerous connections to other countries, indicating its leading role in global research. Countries like the United Kingdom, Japan, and Australia also showed multiple international collaborations. However, countries such as India and China, despite high research activity, exhibited fewer direct international connections, suggesting a need for enhanced global collaboration.
This study employs bibliometric methods to investigate the progression of research concerning the importance of the oral microbiome and salivary biomarkers in the progression of oral cancer, yielding a comprehensive understanding of various research facets. Nevertheless, it is crucial to recognize and address several limitations in our approach. Firstly, we relied solely on the Scopus database, potentially overlooking relevant articles from other sources. Future studies should consider incorporating multiple databases to ensure broader coverage. Secondly, our focus on original articles might have excluded highly cited review articles, which offer valuable summaries of the literature. Incorporating review articles in future studies could provide a more inclusive overview. Lastly, while citation analysis was utilized, additional quality assessments could enhance the identification of high-quality articles. Despite these limitations, our study furnishes valuable insights into the evolution of research on bacterial influence on oral cancer. Acknowledging these constraints, future studies can build on our findings and employ more robust methodologies. This study contributes to advancing scientific understanding in the field of oral cancer and underscores the imperative for continued research.
This review summarizes findings from multiple studies investigating oral squamous cell carcinoma (OSCC) and related biomarkers, focusing on diagnostic, prognostic, and therapeutic advancements. The studies analyzed in this review demonstrate significant progress in the identification of salivary and serum biomarkers, genetic factors, and microbial changes associated with OSCC. Techniques such as metagenomic sequencing, ELISA, qPCR, and mass spectrometry have been employed to reveal differential biomarker expressions, including microRNAs, cfDNA, cytokines, and metabolites. Statistical analyses like ANOVA, Kruskal–Wallis tests, and ROC curve analysis consistently highlight their diagnostic potential, with several biomarkers achieving high sensitivity and specificity. Moreover, research into the oral microbiome suggests significant microbial diversity alterations in OSCC patients compared to healthy controls. Collectively, these findings emphasize the critical role of molecular and microbial biomarkers in improving early detection, risk stratification, and therapeutic interventions for OSCC (Table 3).

5. Conclusions

The bibliometric analysis of the oral microbiome and its association with oral cancer reveals significant growth in research within this field. Our study underscores the increasing recognition of the oral microbiome’s role in the etiology, diagnosis, and prognosis of oral squamous cell carcinoma (OSCC). Notable findings include the identification of numerous salivary biomarkers, such as IL-6, specific miRNA signatures, and microbial profiles, which show promise for the early detection and monitoring of OSCC.
Our analysis highlights key research themes, such as the sensitivity and specificity of biomarkers, the interplay between periodontitis, the oral microbiome, and oral cancer, and the role of specific bacteria in oral carcinogenesis. The importance of international and interdisciplinary collaborations is emphasized by significant co-authorship networks, reflecting strong global interest and cooperation in this research area.
This study advances the scientific understanding of the oral microbiome’s role in oral cancer and emphasizes the necessity of continued research. Future studies can refine the diagnostic criteria and early detection methods, ultimately improving patient outcomes in OSCC.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms13020373/s1. PRISMA 2020 Main Checklist: This document contains the PRISMA 2020 Main Checklist, detailing the methodology used in this systematic review. Relevant sections of this checklist have been incorporated as images within the manuscript for clarity; PRISMA 2020 Abstract Checklist: This document provides the PRISMA 2020 Abstract Checklist, summarizing essential aspects of the systematic review process. Content from this checklist has also been included as images within the manuscript for reference. Reference [116] is cited in the supplementary materials.

Author Contributions

Conceptualization, R.D. and A.G.; methodology, V.B. and A.G.; software, V.B.; validation, O.B. and R.S.-R.; formal analysis, A.D.F. and V.T.A.; investigation, O.B., B.L.R.B., and V.T.A.; resources, D.J.; data curation, V.B. and A.D.F.; writing—original draft preparation, R.D. and O.B.; writing—review and editing, R.S.-R., V.B., and A.G.; visualization, A.G.; supervision, D.J.; project administration, O.B. and A.G.; funding acquisition, D.J. All authors have read and agreed to the published version of the manuscript.

Funding

We would like to acknowledge Victor Babeș University of Medicine and Pharmacy, Timișoara for their support in covering the costs of publication for this research paper (Grant Number 4814/2 October 2023). This research is part of a project underway at the “Translational and Experimental Clinical Research Centre in Oral Health, Department of Preventive, Community Dentistry, and Oral Health, University of Medicine and Pharmacy “Victor Babes”, Timisoara”, which aims to study the risk factors in oral health and their relationship with quality of life.

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of the University of Medicine and Pharmacy “Victor Babes”, Timisoara, Romania (No. 05/30 January 2024).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

We would like to acknowledge Victor Babeș University of Medicine and Pharmacy, Timișoara for their support in covering the costs of publication for this research paper. The authors are really grateful to the staff of the Faculty of Dental Medicine, University of Medicine and Pharmacy “Victor Babes”, Timisoara, Romania. The authors used ChatGPT, an AI language model developed by OpenAI, exclusively to improve this manuscript’s language and readability. All the scientific content, interpretations, and conclusions are the original work of the authors.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Deo, P.; Deshmukh, R. Oral Microbiome: Unveiling the Fundamentals. J. Oral Maxillofac. Pathol. 2019, 23, 122. [Google Scholar] [CrossRef]
  2. Scotti, E.; Boué, S.; Sasso, G.L.; Zanetti, F.; Belcastro, V.; Poussin, C.; Sierro, N.; Battey, J.; Gimalac, A.; Ivanov, N.V.; et al. Exploring the Microbiome in Health and Disease: Implications for Toxicology. Toxicol. Res. Appl. 2017, 1, 239784731774188. [Google Scholar] [CrossRef]
  3. Gao, L.; Xu, T.; Huang, G.; Jiang, S.; Gu, Y.; Chen, F. Oral Microbiomes: More and More Importance in Oral Cavity and Whole Body. Protein Cell 2018, 9, 488–500. [Google Scholar] [CrossRef]
  4. Cross, B.; Faustoferri, R.C.; Quivey, R.G. What Are We Learning and What Can We Learn from the Human Oral Microbiome Project? Curr. Oral Health Rep. 2016, 3, 56–63. [Google Scholar] [CrossRef]
  5. Zhang, W.; Luo, J.; Dong, X.; Zhao, S.; Hao, Y.; Peng, C.; Shi, H.; Zhou, Y.; Shan, L.; Sun, Q.; et al. Salivary Microbial Dysbiosis Is Associated with Systemic Inflammatory Markers and Predicted Oral Metabolites in Non-Small Cell Lung Cancer Patients. J. Cancer 2019, 10, 1651–1662. [Google Scholar] [CrossRef]
  6. Song, M.; Bai, H.; Zhang, P.; Zhou, X.; Ying, B. Promising Applications of Human-Derived Saliva Biomarker Testing in Clinical Diagnostics. Int. J. Oral Sci. 2023, 15, 2. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, C.-Z.; Cheng, X.-Q.; Li, J.-Y.; Zhang, P.; Yi, P.; Xu, X.; Zhou, X.-D. Saliva in the Diagnosis of Diseases. Int. J. Oral Sci. 2016, 8, 133–137. [Google Scholar] [CrossRef] [PubMed]
  8. Schwarz, C.; Balean, O.; Dumitrescu, R.; Ciordas, P.D.; Marian, C.; Georgescu, M.; Bolchis, V.; Sava-Rosianu, R.; Fratila, A.D.; Alexa, I.; et al. Total Antioxidant Capacity of Saliva and Its Correlation with pH Levels among Dental Students under Different Stressful Conditions. Diagnostics 2023, 13, 3648. [Google Scholar] [CrossRef]
  9. Lan, Q.; Zhang, C.; Hua, H.; Hu, X. 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, 1021. [Google Scholar] [CrossRef]
  10. Rani, N.A.J.; Vardhan, B.G.H.; Srinivasan, S.; Gopal, S.K. Evaluation of Salivary Interleukin-6 in Patients with Oral Squamous Cell Carcinoma, Oral Potentially Malignant Disorders, Chronic Periodontitis and in Healthy Controls—A Cross-Sectional Comparative Study. Ann. Maxillofac. Surg. 2023, 13, 70–75. [Google Scholar] [CrossRef] [PubMed]
  11. Tavakoli, F.; Ghavimi, M.A.; Fakhrzadeh, V.; Abdolzadeh, D.; Afshari, A.; Eslami, H. Evaluation of Salivary Transferrin in Patients with Oral Squamous Cell Carcinoma. Clin. Exp. Dent. Res. 2024, 10, e809. [Google Scholar] [CrossRef] [PubMed]
  12. Radhika, T.; Jeddy, N.; Nithya, S.; Muthumeenakshi, R.M. Salivary Biomarkers in Oral Squamous Cell Carcinoma—An Insight. J. Oral Biol. Craniofacial Res. 2016, 6, S51–S54. [Google Scholar] [CrossRef]
  13. Shield, K.D.; Ferlay, J.; Jemal, A.; Sankaranarayanan, R.; Chaturvedi, A.K.; Bray, F.; Soerjomataram, I. The Global Incidence of Lip, Oral Cavity, and Pharyngeal Cancers by Subsite in 2012. CA. Cancer J. Clin. 2017, 67, 51–64. [Google Scholar] [CrossRef] [PubMed]
  14. Du, M.; Nair, R.; Jamieson, L.; Liu, Z.; Bi, P. Incidence Trends of Lip, Oral Cavity, and Pharyngeal Cancers: Global Burden of Disease 1990–2017. J. Dent. Res. 2020, 99, 143–151. [Google Scholar] [CrossRef]
  15. Ren, Z.; Hu, C.; He, H.; Li, Y.; Lyu, J. Global and Regional Burdens of Oral Cancer from 1990 to 2017: Results from the Global Burden of Disease Study. Cancer Commun. 2020, 40, 81–92. [Google Scholar] [CrossRef]
  16. Yuanbo, Z.; Tianyi, L.; Xuejing, S.; Xinpeng, L.; Jianqun, W.; Wenxia, X.; Jingshu, G. Using Formalin Fixed Paraffin Embedded Tissue to Characterize the Microbiota in P16-Positive and P16-Negative Tongue Squamous Cell Carcinoma: A Pilot Study. BMC Oral Health 2024, 24, 283. [Google Scholar] [CrossRef] [PubMed]
  17. Xiao, J.; Fiscella, K.A.; Gill, S.R. Oral Microbiome: Possible Harbinger for Children’s Health. Int. J. Oral Sci. 2020, 12, 12. [Google Scholar] [CrossRef] [PubMed]
  18. Liu, W.; Wu, L.; Zhang, Y.; Shi, L.; Yang, X. Bibliometric Analysis of Research Trends and Characteristics of Oral Potentially Malignant Disorders. Clin. Oral Investig. 2020, 24, 447–454. [Google Scholar] [CrossRef]
  19. Hassona, Y.; Qutachi, T. A Bibliometric Analysis of the Most Cited Articles about Squamous Cell Carcinoma of the Mouth, Lips, and Oropharynx. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2019, 128, 25–32.e6. [Google Scholar] [CrossRef] [PubMed]
  20. Pena-Cristóbal, M.; Diniz-Freitas, M.; Monteiro, L.; Diz Dios, P.; Warnakulasuriya, S. The 100 Most Cited Articles on Oral Cancer. J. Oral Pathol. Med. 2018, 47, 333–344. [Google Scholar] [CrossRef]
  21. Cooper, I.D. Bibliometrics Basics. J. Med. Libr. Assoc. 2015, 103, 217–218. [Google Scholar] [CrossRef]
  22. Rai, A.K.; Panda, M.; Das, A.K.; Rahman, T.; Das, R.; Das, K.; Sarma, A.; Kataki, A.C.; Chattopadhyay, I. Dysbiosis of Salivary Microbiome and Cytokines Influence Oral Squamous Cell Carcinoma through Inflammation. Arch. Microbiol. 2021, 203, 137–152. [Google Scholar] [CrossRef] [PubMed]
  23. Bugshan, A.; Farooq, I. Oral Squamous Cell Carcinoma: Metastasis, Potentially Associated Malignant Disorders, Etiology and Recent Advancements in Diagnosis. F1000Research 2020, 9, 229. [Google Scholar] [CrossRef]
  24. González-Moles, M.Á.; Warnakulasuriya, S.; González-Ruiz, I.; González-Ruiz, L.; Ayén, Á.; Lenouvel, D.; Ruiz-Ávila, I.; Ramos-García, P. Worldwide Prevalence of Oral Lichen Planus: A Systematic Review and Meta-analysis. Oral Dis. 2021, 27, 813–828. [Google Scholar] [CrossRef]
  25. Bibliometrics & Verification|Karolinska Institutet University Library. Available online: https://kib.ki.se/en/publish-analyse/bibliometrics-verification (accessed on 26 May 2024).
  26. Zyoud, S.H.; Al-Jabi, S.W.; Amer, R.; Shakhshir, M.; Shahwan, M.; Jairoun, A.A.; Akkawi, M.; Abu Taha, A. Global Research Trends on the Links between the Gut Microbiome and Cancer: A Visualization Analysis. J. Transl. Med. 2022, 20, 83. [Google Scholar] [CrossRef]
  27. Yuan, X.; Chang, C.; Chen, X.; Li, K. Emerging Trends and Focus of Human Gastrointestinal Microbiome Research from 2010–2021: A Visualized Study. J. Transl. Med. 2021, 19, 327. [Google Scholar] [CrossRef]
  28. Haddaway, N.R.; Page, M.J.; Pritchard, C.C.; McGuinness, L.A. PRISMA2020: An R Package and Shiny App for Producing PRISMA 2020-compliant Flow Diagrams, with Interactivity for Optimised Digital Transparency and Open Synthesis. Campbell Syst. Rev. 2022, 18, e1230. [Google Scholar] [CrossRef]
  29. McGuinness, L.A.; Higgins, J.P.T. Risk-of-bias VISualization (Robvis): An R Package and Shiny Web App for Visualizing Risk-of-bias Assessments. Res. Synth. Methods 2021, 12, 55–61. [Google Scholar] [CrossRef]
  30. Van Eck, N.J.; Waltman, L. Citation-Based Clustering of Publications Using CitNetExplorer and VOSviewer. Scientometrics 2017, 111, 1053–1070. [Google Scholar] [CrossRef]
  31. Gupta, N.; Gupta, R.; Acharya, A.K.; Patthi, B.; Goud, V.; Reddy, S.; Garg, A.; Singla, A. Changing Trends in Oral Cancer—A Global Scenario. Nepal J. Epidemiol. 2017, 6, 613–619. [Google Scholar] [CrossRef]
  32. Minarovits, J. Anaerobic Bacterial Communities Associated with Oral Carcinoma: Intratumoral, Surface-Biofilm and Salivary Microbiota. Anaerobe 2021, 68, 102300. [Google Scholar] [CrossRef] [PubMed]
  33. Ahmed, A.A.; Sborchia, M.; Bye, H.; Roman-Escorza, M.; Amar, A.; Henley-Smith, R.; Odell, E.; McGurk, M.; Simpson, M.; Ng, T.; et al. Mutation Detection in Saliva from Oral Cancer Patients. Oral Oncol. 2024, 151, 106717. [Google Scholar] [CrossRef]
  34. Rapado-González, Ó.; Costa-Fraga, N.; Bao-Caamano, A.; López-Cedrún, J.L.; Álvarez-Rodríguez, R.; Crujeiras, A.B.; Muinelo-Romay, L.; López-López, R.; Díaz-Lagares, Á.; Suárez-Cunqueiro, M.M. Genome-Wide DNA Methylation Profiling in Tongue Squamous Cell Carcinoma. Oral Dis. 2024, 30, 259–271. [Google Scholar] [CrossRef] [PubMed]
  35. Scheurer, M.J.J.; Wagner, A.; Sakkas, A.; Pietzka, S.; Derka, S.; Vairaktari, G.; Wilde, F.; Schramm, A.; Bauer, A.; Siebert, R.; et al. Influence of Analytical Procedures on miRNA Expression Analyses in Saliva Samples. J. Cranio-Maxillofac. Surg. 2024, 52, 748–754. [Google Scholar] [CrossRef]
  36. Zhu, W.; Shen, W.; Wang, J.; Xu, Y.; Zhai, R.; Zhang, J.; Wang, M.; Wang, M.; Liu, L. Capnocytophaga Gingivalis Is a Potential Tumor Promotor in Oral Cancer. Oral Dis. 2024, 30, 353–362. [Google Scholar] [CrossRef] [PubMed]
  37. Riccardi, G.; Bellizzi, M.G.; Fatuzzo, I.; Zoccali, F.; Cavalcanti, L.; Greco, A.; Vincentiis, M.D.; Ralli, M.; Fiore, M.; Petrella, C.; et al. Salivary Biomarkers in Oral Squamous Cell Carcinoma: A Proteomic Overview. Proteomes 2022, 10, 37. [Google Scholar] [CrossRef] [PubMed]
  38. Farhadian, S.; Hashemi-Shahraki, F.; Amirifar, S.; Asadpour, S.; Shareghi, B.; Heidari, E.; Shakerian, B.; Rafatifard, M.; Firooz, A.R. Malachite Green, the Hazardous Materials That Can Bind to Apo-Transferrin and Change the Iron Transfer. Int. J. Biol. Macromol. 2022, 194, 790–799. [Google Scholar] [CrossRef]
  39. Chen, M.Y.; Chen, J.W.; Wu, L.W.; Huang, K.C.; Chen, J.Y.; Wu, W.S.; Chiang, W.F.; Shih, C.J.; Tsai, K.N.; Hsieh, W.T.; et al. Carcinogenesis of Male Oral Submucous Fibrosis Alters Salivary Microbiomes. J. Dent. Res. 2021, 100, 397–405. [Google Scholar] [CrossRef]
  40. Herreros-Pomares, A.; Hervás, D.; Bagan-Debón, L.; Jantus-Lewintre, E.; Gimeno-Cardona, C.; Bagan, J. On the Oral Microbiome of Oral Potentially Malignant and Malignant Disorders: Dysbiosis, Loss of Diversity, and Pathogens Enrichment. Int. J. Mol. Sci. 2023, 24, 3466. [Google Scholar] [CrossRef]
  41. Ganly, I.; Yang, L.; Giese, R.A.; Hao, Y.; Nossa, C.W.; Morris, L.G.T.; Rosenthal, M.; Migliacci, J.; Kelly, D.; Tseng, W.; et al. 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, 775–784. [Google Scholar] [CrossRef]
  42. Hashimoto, K.; Shimizu, D.; Ueda, S.; Miyabe, S.; Oh-Iwa, I.; Nagao, T.; Shimozato, K.; Nomoto, S. Feasibility of Oral Microbiome Profiles Associated with Oral Squamous Cell Carcinoma. J. Oral Microbiol. 2022, 14, 2105574. [Google Scholar] [CrossRef] [PubMed]
  43. Chen, J.; Wang, L.; Ma, D.; Zhang, H.; Fan, J.; Gao, H.; Xia, X.; Wu, W.; Shi, Y. miR-19a May Function as a Biomarker of Oral Squamous Cell Carcinoma (OSCC) by Regulating the Signaling Pathway of miR-19a/GRK6/GPCRs/PKC in a Chinese Population. J. Oral Pathol. Med. 2023, 52, 971–979. [Google Scholar] [CrossRef]
  44. Daniel, D.; Jose, J.; Harish Kumar, A. Is Salivary Sialic Acid a Reliable Biomarker in the Detection of Oral Potentially Malignant Disorder and Oral Squamous Cell Carcinoma. J. Maxillofac. Oral Surg. 2021, 20, 83–89. [Google Scholar] [CrossRef] [PubMed]
  45. Yap, T.; Seers, C.; Koo, K.; Cheng, L.; Vella, L.J.; Hill, A.F.; Reynolds, E.; Nastri, A.; Cirillo, N.; McCullough, M. Non-Invasive Screening of a microRNA-Based Dysregulation Signature in Oral Cancer and Oral Potentially Malignant Disorders. Oral Oncol. 2019, 96, 113–120. [Google Scholar] [CrossRef] [PubMed]
  46. Zheng, J.; Sun, L.; Yuan, W.; Xu, J.; Yu, X.; Wang, F.; Sun, L.; Zeng, Y. Clinical Value of Naa10p and CEA Levels in Saliva and Serum for Diagnosis of Oral Squamous Cell Carcinoma. J. Oral Pathol. Med. 2018, 47, 830–835. [Google Scholar] [CrossRef] [PubMed]
  47. Hes, C.; Desilets, A.; Tonneau, M.; El Ouarzadi, O.; De Figueiredo Sousa, M.; Bahig, H.; Filion, É.; Nguyen-Tan, P.F.; Christopoulos, A.; Benlaïfaoui, M.; et al. Gut Microbiome Predicts Gastrointestinal Toxicity Outcomes from Chemoradiation Therapy in Patients with Head and Neck Squamous Cell Carcinoma. Oral Oncol. 2024, 148, 106623. [Google Scholar] [CrossRef]
  48. Mehdipour, M.; Shahidi, M.; Anbari, F.; Mirzaei, H.; Jafari, S.; Kholghi, A.; Lotfi, E.; Manifar, S.; Mashhadiabbas, F. Salivary Level of microRNA-146a and microRNA-155 Biomarkers in Patients with Oral Lichen Planus versus Oral Squamous Cell Carcinoma. BMC Oral Health 2023, 23, 433. [Google Scholar] [CrossRef] [PubMed]
  49. Rapado-González, Ó.; López-Cedrún, J.L.; Lago-Lestón, R.M.; Abalo, A.; Rubin-Roger, G.; Salgado-Barreira, Á.; López-López, R.; Muinelo-Romay, L.; Suárez-Cunqueiro, M.M. Integrity and Quantity of Salivary Cell-Free DNA as a Potential Molecular Biomarker in Oral Cancer: A Preliminary Study. J. Oral Pathol. Med. 2022, 51, 429–435. [Google Scholar] [CrossRef] [PubMed]
  50. Menaka, T.R.; Vasupradha, G.; Ravikumar, S.S.; Dhivya, K.; Dinakaran, J.; Saranya, V. Evaluation of Salivary Alkaline Phosphatase Levels in Tobacco Users to Determine Its Role as a Biomarker in Oral Potentially Malignant Disorders. J. Oral Maxillofac. Pathol. 2019, 23, 344–348. [Google Scholar]
  51. Sridharan, G.; Ramani, P.; Patankar, S.; Vijayaraghavan, R. Evaluation of Salivary Metabolomics in Oral Leukoplakia and Oral Squamous Cell Carcinoma. J. Oral Pathol. Med. 2019, 48, 299–306. [Google Scholar] [CrossRef] [PubMed]
  52. Bandara, H.M.H.N.; Panduwawala, C.P.; Samaranayake, L.P. Biodiversity of the Human Oral Mycobiome in Health and Disease. Oral Dis. 2019, 25, 363–371. [Google Scholar] [CrossRef] [PubMed]
  53. Palaia, G.; Pippi, R.; Rocchetti, F.; Caputo, M.; Macali, F.; Mohsen, A.; Vecchio, A.D.; Tenore, G.; Romeo, U. Liquid Biopsy in the Assessment of microRNAs in Oral Squamous Cell Carcinoma: A Systematic Review. J. Clin. Exp. Dent. 2022, 14, 875–884. [Google Scholar] [CrossRef] [PubMed]
  54. Afifi, S.; Zahran, F.; Shaker, O.; Tarrad, N.; Elsaadany, B. Sensitivity and Specificity of Serum and Salivary Cyfra21-1 in Detecting Malignant Changes in Oral Potentially Malignant Lesions (Diagnostic Accuracy Study). World J. Dent. 2021, 12, 200–207. [Google Scholar] [CrossRef]
  55. Garg, R.; Shetti, A.V.; Bagewadi, A.S. Assessment and Comparison of Salivary Survivin Biomarker in Oral Leukoplakia, Oral Lichen Planus, and Oral Cancer: A Comparative Study. World J. Dent. 2017, 8, 73–76. [Google Scholar] [CrossRef]
  56. Perera, M.; Al-hebshi, N.N.; Perera, I.; Ipe, D.; Ulett, G.C.; Speicher, D.J.; Chen, T.; Johnson, N.W. Inflammatory Bacteriome and Oral Squamous Cell Carcinoma. J. Dent. Res. 2018, 97, 725–732. [Google Scholar] [CrossRef] [PubMed]
  57. Farshbaf, A.; Mohajertehran, F.; Aghaee-Bakhtiari, S.H.; Ayatollahi, H.; Douzandeh, K.; Pakfetrat, A.; Mohtasham, N. Downregulation of Salivary miR-3928 as a Potential Biomarker in Patients with Oral Squamous Cell Carcinoma and Oral Lichen Planus. Clin. Exp. Dent. Res. 2024, 10, e877. [Google Scholar] [CrossRef]
  58. Nosratzehi, T.; Risbaf Fakour, S.; Alijani, E.; Salehi, M. Investigating the Level of Salivary Endothelin-1 in Premalignant and Malignant Lesions. Spec. Care Dentist. 2017, 37, 134–139. [Google Scholar] [CrossRef] [PubMed]
  59. Vesty, A.; Gear, K.; Biswas, K.; Radcliff, F.J.; Taylor, M.W.; Douglas, R.G. Microbial and Inflammatory-Based Salivary Biomarkers of Head and Neck Squamous Cell Carcinoma. Clin. Exp. Dent. Res. 2018, 4, 255–262. [Google Scholar] [CrossRef]
  60. Blatt, S.; Krüger, M.; Ziebart, T.; Sagheb, K.; Schiegnitz, E.; Goetze, E.; Al-Nawas, B.; Pabst, A.M. Biomarkers in Diagnosis and Therapy of Oral Squamous Cell Carcinoma: A Review of the Literature. J. Cranio-Maxillofac. Surg. 2017, 45, 722–730. [Google Scholar] [CrossRef] [PubMed]
  61. Saleem, Z.; Shaikh, A.H.; Zaman, U.; Ahmed, S.; Majeed, M.M.; Kazmi, A.; Farooqui, W.A. Estimation of Salivary Matrix Metalloproteinases- 12 (MMP- 12) Levels among Patients Presenting with Oral Submucous Fibrosis and Oral Squamous Cell Carcinoma. BMC Oral Health 2021, 21, e87721. [Google Scholar] [CrossRef]
  62. Tarrad, N.A.F.; Hassan, S.; Shaker, O.G.; AbdelKawy, M. “Salivary LINC00657 and miRNA-106a as Diagnostic Biomarkers for Oral Squamous Cell Carcinoma, an Observational Diagnostic Study”. BMC Oral Health 2023, 23, 994. [Google Scholar] [CrossRef] [PubMed]
  63. Cohen, E.R.; Reis, I.M.; Gomez-Fernandez, C.; Smith, D.; Pereira, L.; Freiser, M.E.; Marotta, G.; Thomas, G.R.; Sargi, Z.B.; Franzmann, E.J. CD44 and Associated Markers in Oral Rinses and Tissues from Oral and Oropharyngeal Cancer Patients. Oral Oncol. 2020, 106, 104720. [Google Scholar] [CrossRef] [PubMed]
  64. Rajaram, S.; Danasekaran, B.; Venkatachalapathy, R.; Prashad, K.; Rajaram, S. N-Acetylneuraminic Acid: A Scrutinizing Tool in Oral Squamous Cell Carcinoma Diagnosis. Dent. Res. J. 2017, 14, 267–271. [Google Scholar] [CrossRef]
  65. Kitamura, N.; Hashida, Y.; Higuchi, T.; Ohno, S.; Sento, S.; Sasabe, E.; Murakami, I.; Yamamoto, T.; Daibata, M. Detection of Merkel Cell Polyomavirus in Multiple Primary Oral Squamous Cell Carcinomas. Odontology 2023, 111, 971–981. [Google Scholar] [CrossRef] [PubMed]
  66. Gaba, F.I.; Sheth, C.C.; Veses, V. Salivary Biomarkers and Their Efficacies as Diagnostic Tools for Oral Squamous Cell Carcinoma: Systematic Review and Meta-Analysis. J. Oral Pathol. Med. 2021, 50, 299–307. [Google Scholar] [CrossRef] [PubMed]
  67. Cheng, Y.-S.L.; Jordan, L.; Chen, H.-S.; Kang, D.; Oxford, L.; Plemons, J.; Parks, H.; Rees, T. Chronic Periodontitis Can Affect the Levels of Potential Oral Cancer Salivary mRNA Biomarkers. J. Periodontal Res. 2017, 52, 428–437. [Google Scholar] [CrossRef]
  68. Shabbir, A.; Waheed, H.; Ahmed, S.; Shaikh, S.S.; Farooqui, W.A. Association of Salivary Cathepsin B in Different Histological Grades among Patients Presenting with Oral Squamous Cell Carcinoma. BMC Oral Health 2022, 22, 63. [Google Scholar] [CrossRef]
  69. Mougeot, J.-L.C.; Stevens, C.B.; Almon, K.G.; Paster, B.J.; Lalla, R.V.; Brennan, M.T.; Mougeot, F.B. Caries-Associated Oral Microbiome in Head and Neck Cancer Radiation Patients: A Longitudinal Study. J. Oral Microbiol. 2019, 11, 1586421. [Google Scholar] [CrossRef] [PubMed]
  70. Ishikawa, S.; Sugimoto, M.; Edamatsu, K.; Sugano, A.; Kitabatake, K.; Iino, M. Discrimination of Oral Squamous Cell Carcinoma from Oral Lichen Planus by Salivary Metabolomics. Oral Dis. 2020, 26, 35–42. [Google Scholar] [CrossRef]
  71. Babiuch, K.; Bednarczyk, A.; Gawlik, K.; Pawlica-Gosiewska, D.; Kęsek, B.; Darczuk, D.; Stępień, P.; Chomyszyn-Gajewska, M.; Kaczmarzyk, T. Evaluation of Enzymatic and Non-Enzymatic Antioxidant Status and Biomarkers of Oxidative Stress in Saliva of Patients with Oral Squamous Cell Carcinoma and Oral Leukoplakia: A Pilot Study. Acta Odontol. Scand. 2019, 77, 408–418. [Google Scholar] [CrossRef]
  72. Shan, J.; Sun, Z.; Yang, J.; Xu, J.; Shi, W.; Wu, Y.; Fan, Y.; Li, H. Discovery and Preclinical Validation of Proteomic Biomarkers in Saliva for Early Detection of Oral Squamous Cell Carcinomas. Oral Dis. 2019, 25, 97–107. [Google Scholar] [CrossRef] [PubMed]
  73. Katase, N.; Kudo, K.; Ogawa, K.; Sakamoto, Y.; Nishimatsu, S.; Yamauchi, A.; Fujita, S. DKK3/CKAP4 Axis Is Associated with Advanced Stage and Poorer Prognosis in Oral Cancer. Oral Dis. 2023, 29, 3193–3204. [Google Scholar] [CrossRef] [PubMed]
  74. Abdelwhab, A.; Shaker, O.; Aggour, R.L. Expression of Mucin1 in Saliva in Oral Squamous Cell Carcinoma and Oral Potentially Malignant Disorders (Case Control Study). Oral Dis. 2023, 29, 1487–1494. [Google Scholar] [CrossRef]
  75. Kallalli, B.N.; Rawson, K.; Muzammil; Singh, A.; Awati, M.A.; Shivhare, P. Lactate Dehydrogenase as a Biomarker in Oral Cancer and Oral Submucous Fibrosis. J. Oral Pathol. Med. 2016, 45, 687–690. [Google Scholar] [CrossRef] [PubMed]
  76. Chapple, I.L.C.; Mealey, B.L.; Van Dyke, T.E.; Bartold, P.M.; Dommisch, H.; Eickholz, P.; Geisinger, M.L.; Genco, R.J.; Glogauer, M.; Goldstein, M.; et al. Periodontal Health and Gingival Diseases and Conditions on an Intact and a Reduced Periodontium: Consensus Report of Workgroup 1 of the 2017 World Workshop on the Classification of Periodontal and Peri-Implant Diseases and Conditions. J. Periodontol. 2018, 89, S74–S84. [Google Scholar] [CrossRef] [PubMed]
  77. Jolivet-Gougeon, A.; Bonnaure-Mallet, M. Screening for Prevalence and Abundance of Capnocytophaga Spp by Analyzing NGS Data: A Scoping Review. Oral Dis. 2021, 27, 1621–1630. [Google Scholar] [CrossRef]
  78. Ishikawa, S.; Wong, D.T.W.; Sugimoto, M.; Gleber-Netto, F.O.; Li, F.; Tu, M.; Zhang, Y.; Akin, D.; Iino, M. Identification of Salivary Metabolites for Oral Squamous Cell Carcinoma and Oral Epithelial Dysplasia Screening from Persistent Suspicious Oral Mucosal Lesions. Clin. Oral Investig. 2019, 23, 3557–3563. [Google Scholar] [CrossRef]
  79. Banavar, G.; Ogundijo, O.; Julian, C.; Toma, R.; Camacho, F.; Torres, P.J.; Hu, L.; Chandra, T.; Piscitello, A.; Kenny, L.; et al. Detecting Salivary Host and Microbiome RNA Signature for Aiding Diagnosis of Oral and Throat Cancer. Oral Oncol. 2023, 145, 106480. [Google Scholar] [CrossRef] [PubMed]
  80. Zhong, X.; Lu, Q.; Zhang, Q.; He, Y.; Wei, W.; Wang, Y. Oral Microbiota Alteration Associated with Oral Cancer and Areca Chewing. Oral Dis. 2021, 27, 226–239. [Google Scholar] [CrossRef]
  81. Khalil, M.; Zraiki, S.; Khayat, M. Evaluation of the Salivary Zinc Assay as a Potential Diagnostic Tool in Potential Malignant and Malignant Lesions of the Oral Cavity. J. Indian Acad. Oral Med. Radiol. 2019, 31, 293–297. [Google Scholar] [CrossRef]
  82. Chari, A.; Rajesh, P.; Prabhu, S. Estimation of Serum Lactate Dehydrogenase in Smokeless Tobacco Consumers. Indian J. Dent. Res. 2016, 27, 602–608. [Google Scholar] [CrossRef]
  83. Sayal, L.; Hamadah, O.; Almasri, A.; Idrees, M.; Thomson, P.; Kujan, O. Saliva-Based Cell-Free DNA and Cell-Free Mitochondrial DNA in Head and Neck Cancers Have Promising Screening and Early Detection Role. J. Oral Pathol. Med. 2023, 52, 29–36. [Google Scholar] [CrossRef]
  84. Ameena, M.; Rathy, R. Evaluation of Tumor Necrosis Factor: Alpha in the Saliva of Oral Cancer, Leukoplakia, and Healthy Controls—A Comparative Study. J. Int. Oral Health 2019, 11, 92–99. [Google Scholar] [CrossRef]
  85. Yost, S.; Stashenko, P.; Choi, Y.; Kukuruzinska, M.; Genco, C.A.; Salama, A.; Weinberg, E.O.; Kramer, C.D.; Frias-Lopez, J. Increased Virulence of the Oral Microbiome in Oral Squamous Cell Carcinoma Revealed by Metatranscriptome Analyses. Int. J. Oral Sci. 2018, 10, 32. [Google Scholar] [CrossRef] [PubMed]
  86. Feng, Y.; Li, Q.; Chen, J.; Yi, P.; Xu, X.; Fan, Y.; Cui, B.; Yu, Y.; Li, X.; Du, Y.; et al. Salivary Protease Spectrum Biomarkers of Oral Cancer. Int. J. Oral Sci. 2019, 11, 7. [Google Scholar] [CrossRef]
  87. Ghallab, N.A.; Shaker, O.G. Serum and Salivary Levels of Chemerin and MMP-9 in Oral Squamous Cell Carcinoma and Oral Premalignant Lesions. Clin. Oral Investig. 2017, 21, 937–947. [Google Scholar] [CrossRef]
  88. Sawant, S.; Gokulan, R.; Dongre, H.; Vaidya, M.; Chaukar, D.; Prabhash, K.; Ingle, A.; Joshi, S.; Dange, P.; Joshi, S.; et al. Prognostic Role of Oct4, CD44 and c-Myc in Radio–Chemo-Resistant Oral Cancer Patients and Their Tumourigenic Potential in Immunodeficient Mice. Clin. Oral Investig. 2016, 20, 43–56. [Google Scholar] [CrossRef]
  89. Lee, L.T.; Wong, Y.K.; Hsiao, H.Y.; Wang, Y.W.; Chan, M.Y.; Chang, K.W. Evaluation of Saliva and Plasma Cytokine Biomarkers in Patients with Oral Squamous Cell Carcinoma. Int. J. Oral Maxillofac. Surg. 2018, 47, 699–707. [Google Scholar] [CrossRef]
  90. Triani, M.; Widodo, H.B.; Novrial, D.; Agustina, D.; Nawangtantrini, G. Salivary Ki-67 and Micronucleus Assay as Potential Biomarker of OSCC in Betel Nut Chewers. J. Indian Acad. Oral Med. Radiol. 2021, 33, 146–151. [Google Scholar] [CrossRef]
  91. Perera, M.; Al-Hebshi, N.N.; Perera, I.; Ipe, D.; Ulett, G.C.; Speicher, D.J.; Chen, T.; Johnson, N.W. A Dysbiotic Mycobiome Dominated by Candida Albicans Is Identified within Oral Squamous-Cell Carcinomas. J. Oral Microbiol. 2017, 9, 1385369. [Google Scholar] [CrossRef] [PubMed]
  92. Chan, J.Y.K.; Cheung, M.K.; Lan, L.; Ng, C.; Lau, E.H.L.; Yeung, Z.W.C.; Wong, E.W.Y.; Leung, L.; Qu, X.; Cai, L.; et al. Characterization of Oral Microbiota in HPV and Non-HPV Head and Neck Squamous Cell Carcinoma and Its Association with Patient Outcomes. Oral Oncol. 2022, 135, 106245. [Google Scholar] [CrossRef]
  93. Kaur, J.; Politis, C.; Jacobs, R. Salivary 8-Hydroxy-2-Deoxyguanosine, Malondialdehyde, Vitamin C, and Vitamin E in Oral Pre-Cancer and Cancer: Diagnostic Value and Free Radical Mechanism of Action. Clin. Oral Investig. 2016, 20, 315–319. [Google Scholar] [CrossRef] [PubMed]
  94. Kumaran, J.; Ramesh, V.; Daniel, M. Salivary Micrornas (miRNAs) Expression and Its Implications as Biomarkers in Oral Cancer. J. Indian Acad. Oral Med. Radiol. 2022, 34, 475–476. [Google Scholar] [CrossRef]
  95. Suresh, H.; Gunasekaran, N.; James, A.; Krishnan, R.; Thayalan, D.; Kumar, A. Alpha-L-Fucosidase Levels in Patients with Oral Submucous Fibrosis and Controls: A Comparative Study. J. Oral Maxillofac. Pathol. 2022, 26, 594. [Google Scholar] [CrossRef] [PubMed]
  96. Ferrazzo, K.L.; De Melo, L.D.W.; Danesi, C.C.; Thomas, A.; Bonzanini, L.I.L.; Zanatta, N. Sensitivity and Specificity of Salivary Pipecolic Acid in Head and Neck Squamous Cell Carcinoma. Braz. J. Oral Sci. 2022, 22, 1–13. [Google Scholar] [CrossRef]
  97. SHahinas, J.; Hysi, D. Methods and Risk of Bias in Molecular Marker Prognosis Studies in Oral Squamous Cell Carcinoma. Oral Dis. 2018, 24, 115–119. [Google Scholar] [CrossRef] [PubMed]
  98. Huang, L.; Pei, T.; Wu, G.; Liu, J.; Pan, W.; Pan, X. Circular RNAs as a Diagnostic Biomarker in Oral Squamous Cell Carcinoma: A Meta-Analysis. J. Oral Maxillofac. Surg. 2022, 80, 756–766. [Google Scholar] [CrossRef]
  99. Furquim, C.P.; Soares, G.M.S.; Ribeiro, L.L.; Azcarate-Peril, M.A.; Butz, N.; Roach, J.; Moss, K.; Bonfim, C.; Torres-Pereira, C.C.; Teles, F.R.F. The Salivary Microbiome and Oral Cancer Risk: A Pilot Study in Fanconi Anemia. J. Dent. Res. 2017, 96, 292–299. [Google Scholar] [CrossRef]
  100. Susha, K.P.; Ravindran, R. Evaluation of Salivary Chemerin in Oral Leukoplakia, Oral Squamous Cell Carcinoma and Healthy Controls. J. Orofac. Sci. 2023, 15, 141–146. [Google Scholar] [CrossRef]
  101. Liu, W.; Shi, H.; Zhou, Z.; Zhang, C.; Shen, X. Association of Salivary miRNAs with Onset and Progression of Oral Potentially Malignant Disorders: Searching for Noninvasive Biomarkers. J. Dent. Sci. 2023, 18, 432–436. [Google Scholar] [CrossRef] [PubMed]
  102. Peisker, A.; Raschke, G.F.; Fahmy, M.D.; Guentsch, A.; Roshanghias, K.; Hennings, J.; Schultze-Mosgau, S. Salivary MMP-9 in the Detection of Oral Squamous Cell Carcinoma. Med. Oral Patol. Oral Cirugia Bucal 2017, 22, e270–e275. [Google Scholar] [CrossRef] [PubMed]
  103. Nakamichi, E.; Sakakura, H.; Mii, S.; Yamamoto, N.; Hibi, H.; Asai, M.; Takahashi, M. Detection of Serum/Salivary Exosomal Alix in Patients with Oral Squamous Cell Carcinoma. Oral Dis. 2021, 27, 439–447. [Google Scholar] [CrossRef]
  104. Nguyen, T.T.H.; Sodnom-Ish, B.; Choi, S.W.; Jung, H.-I.; Cho, J.; Hwang, I.; Kim, S.M. Salivary Biomarkers in Oral Squamous Cell Carcinoma. J. Korean Assoc. Oral Maxillofac. Surg. 2020, 46, 301–312. [Google Scholar] [CrossRef] [PubMed]
  105. Robledo-Sierra, J.; Ben-Amy, D.P.; Varoni, E.; Bavarian, R.; Simonsen, J.L.; Paster, B.J.; Wade, W.G.; Kerr, R.; Peterson, D.E.; Frandsen Lau, E. World Workshop on Oral Medicine VII: Targeting the Oral Microbiome Part 2: Current Knowledge on Malignant and Potentially Malignant Oral Disorders. Oral Dis. 2019, 25, 28–48. [Google Scholar] [CrossRef]
  106. Abdul Aziz Shaikh, S.; Denny, E.C.; Kumarchandra, R.; Natarajan, S.; Sunny, J.; Shenoy, N.; K P, N. Evaluation of Salivary Tumor Necrosis Factor α as a Diagnostic Biomarker in Oral Submucosal Fibrosis and Squamous Cell Carcinoma of the Oral Cavity and Oropharynx: A Cross Sectional Observational Study. Front. Oral Health 2024, 5, 1375162. [Google Scholar] [CrossRef]
  107. Vimal, J.; George, N.A.; Kumar, R.R.; Kattoor, J.; Kannan, S. Identification of Salivary Metabolic Biomarker Signatures for Oral Tongue Squamous Cell Carcinoma. Arch. Oral Biol. 2023, 155, 105780. [Google Scholar] [CrossRef]
  108. Man, Q.-W.; Li, R.-F.; Zhong, N.-N.; Liu, B. Prognostic Value of CD8-to-EGFR Ratio in Salivary Microvesicles of Patients with Oral Squamous Cell Carcinoma. Oral Dis. 2023, 29, 1480–1486. [Google Scholar] [CrossRef] [PubMed]
  109. Yang, S.-F.; Huang, H.-D.; Fan, W.-L.; Jong, Y.-J.; Chen, M.-K.; Huang, C.-N.; Chuang, C.-Y.; Kuo, Y.-L.; Chung, W.-H.; Su, S.-C. Compositional and Functional Variations of Oral Microbiota Associated with the Mutational Changes in Oral Cancer. Oral Oncol. 2018, 77, 1–8. [Google Scholar] [CrossRef] [PubMed]
  110. Yang, S.F.; Lin, C.W.; Chuang, C.Y.; Lee, Y.C.; Chung, W.H.; Lai, H.C.; Chang, L.C.; Su, S.C. Host Genetic Associations with Salivary Microbiome in Oral Cancer. J. Dent. Res. 2022, 101, 590–598. [Google Scholar] [CrossRef]
  111. Tantray, S.; Sharma, S.; Prabhat, K.; Nasrullah, N.; Gupta, M. Salivary Metabolite Signatures of Oral Cancer and Leukoplakia through Gas Chromatography-Mass Spectrometry. J. Oral Maxillofac. Pathol. 2022, 26, 31–37. [Google Scholar] [CrossRef]
  112. Gulzar, R.A.; Ajitha, P.; Subbaiyan, H. Comparative Evaluation on the Levels of Salivary Mucin MUC1 in Precancerous and Cancerous Conditions. Int. J. Dent. Oral Sci. 2021, 8, 3734–3737. [Google Scholar] [CrossRef]
  113. Gupta, A.; Tripathi, A.; Patil, R.; Kumar, V.; Khanna, V.; Singh, V. Estimation of Salivary and Serum Basic Fibroblast Growth Factor in Treated and Untreated Patients with Oral Squamous Cell Carcinoma. J. Oral Biol. Craniofacial Res. 2019, 9, 19–23. [Google Scholar] [CrossRef] [PubMed]
  114. Karmelić, I.; Salarić, I.; Baždarić, K.; Rožman, M.; Zajc, I.; Mravak-Stipetić, M.; Bago, I.; Brajdić, D.; Lovrić, J.; Macan, D. Salivary Scca1, Scca2 and Trop2 in Oral Cancer Patients— A Cross-Sectional Pilot Study. Dent. J. 2022, 10, 70. [Google Scholar] [CrossRef] [PubMed]
  115. Tang, K.D.; Amenábar, J.M.; Schussel, J.L.; Torres-Pereira, C.C.; Bonfim, C.; Dimitrova, N.; Hartel, G.; Punyadeera, C. Profiling Salivary miRNA Expression Levels in Fanconi Anemia Patients—A Pilot Study. Odontology 2024, 112, 299–308. [Google Scholar] [CrossRef] [PubMed]
  116. Page, M.J.; McKenzie, J.; Bossuyt, P.; Boutron, I.; Hoffmann, T.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.; Akl, E.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. MetaArXiv 2020. [Google Scholar] [CrossRef]
Figure 1. Flow diagram and search strategy in Scopus.
Figure 1. Flow diagram and search strategy in Scopus.
Microorganisms 13 00373 g001
Figure 2. PRISMA 2020 flow diagram for new systematic reviews which include searches of databases and registers only. Registers: organized collections of data, such as databases or repositories (e.g., Scopus), used to systematically identify and retrieve studies relevant to the oral microbiome and oral cancer research included in this analysis.
Figure 2. PRISMA 2020 flow diagram for new systematic reviews which include searches of databases and registers only. Registers: organized collections of data, such as databases or repositories (e.g., Scopus), used to systematically identify and retrieve studies relevant to the oral microbiome and oral cancer research included in this analysis.
Microorganisms 13 00373 g002
Figure 3. Risk of bias assessment for included studies using the ROB2 tool.
Figure 3. Risk of bias assessment for included studies using the ROB2 tool.
Microorganisms 13 00373 g003
Figure 4. Map of the network of keywords. Source: own processing through VOSviewer.
Figure 4. Map of the network of keywords. Source: own processing through VOSviewer.
Microorganisms 13 00373 g004
Figure 5. Network of scientific co-authorship, based on the number of documents per author. Source: own processing through VOSviewer.
Figure 5. Network of scientific co-authorship, based on the number of documents per author. Source: own processing through VOSviewer.
Microorganisms 13 00373 g005
Figure 6. Bibliometric map of international research collaborations.
Figure 6. Bibliometric map of international research collaborations.
Microorganisms 13 00373 g006
Figure 7. Bibliometric map of collaborations between organizations.
Figure 7. Bibliometric map of collaborations between organizations.
Microorganisms 13 00373 g007
Figure 8. Co-occurrence network analysis of key terms in oral microbiome research.
Figure 8. Co-occurrence network analysis of key terms in oral microbiome research.
Microorganisms 13 00373 g008
Figure 9. Article appearances across years.
Figure 9. Article appearances across years.
Microorganisms 13 00373 g009
Table 1. Keyword groups. Source: own processing through VOSviewer (Occ., occurrences; T.L.S., total link strength).
Table 1. Keyword groups. Source: own processing through VOSviewer (Occ., occurrences; T.L.S., total link strength).
Word No.Group 1 (Red)OccT.L.S.Group 2 (Green)OccT.L.S.Group 3 (Blue)OccT.L.S.Group 4 (Yellow)OccT.L.S.
1Bacterium936Early detection1159Cell carcinoma637Early diagnosis954
2Carcinogenesis945Early stage523Concentration844Malignant transformation945
3Development940Marker1670Evaluation1370Potential biomarker835
4Disease2075Risk731Healthy individual738Prognosis1038
5Head1248Salivary1363Malignant disorder1465Salivary metabolite523
6Important role525Sensitivity1995Oral636
7Neck squamous cell carcinoma516Serum834OSCC group738
8Oral microbiome932Specificity1889Reliable biomarker528
9Pathogenesis521Use835
10Progression1046
11Squamous cell carcinoma69277
12Tissue1243
Table 2. Organization of primary authors by color-coded groups: citations per document and total link strength (TLS). Source: own processing through VOSviewer (T.L.S., total link strength).
Table 2. Organization of primary authors by color-coded groups: citations per document and total link strength (TLS). Source: own processing through VOSviewer (T.L.S., total link strength).
Group (Color)Main AuthorsCitations/DocumentTLS
Group 1 (red)Chaukar, Devendra; Joshi, Shriya; Vaidya, Milind; Dongre, Harsh; Iino, Mitsuyoshi≥5075
Group 2 (light blue)Schierz, Eik; Blatt, Sebastian; Al-Nawas, Bilal≥5070
Group 3 (green)Huang, Hsien-Da; Kuo, Yu-Lun; Tan, Yen-Jang≥5085
Group 4 (dark blue)Kramer, Carolyn D.; Frias-Lopez, Jorge; Genco, Caroline A.; Choi, Yoonhee≥5065
Group 5 (yellow)Johnson, N.W.; Al-Hebshi, N.M.; Chen, T.≥5055
Group 6 (purple)Al-Hebshi, Nezar Noor; Chen, Tsute; Speicher, David J.≥5070
Group 7 (orange)Chang, M.Y.; Wang, W.W.; Chang, K.W.≥5055
Group 8 (dark red)Patankar, Sangeeta; Vijayaraghavan, Rajagopalan≥5060
Group 9 (light green)Ghallab, Noha A.≥5050
Group 10 (pink)Bandara, Herath Mudiyanselage≥5045
Table 3. Key biomarkers and diagnostic advancements in oral squamous cell carcinoma: a review of current evidence.
Table 3. Key biomarkers and diagnostic advancements in oral squamous cell carcinoma: a review of current evidence.
AuthorPublication YearFocus of the InvestigationSample SizeType of StudyType of AnalysisStatisticsResults
Lan, Qingying et al. [9]2023Microbiota, microflora, Mouth Neoplasms, carcinoma, and squamous cells18 OSCC patients, 21 OLK patients, and 21 healthy controls (HCs)Case–control studyThe researchers used metagenomic sequencingSpearman’s correlationThe study found significant differences in the salivary microbiota among OSCC, OLK, and HCs
Daniel, Diana et al. [44]2021Biomarkers; oral squamous cell carcinoma; saliva; total sialic acid level60 subjects divided into three groupsCase–control studyThe researchers analyzed total salivary sialic acid (TSA) levels using a sialic acid kit and UV spectrophotometerKruskal–Wallis and Mann–Whitney post hoc testsThe study suggested that salivary sialic acid could be a reliable biomarker for detecting OSCC and OPMDs
Yap, T. et al. [45]2019Genetics, periodontitis, gingivitis, biomarkers, carcinoma, microRNAs190 individualsSystematic reviewDeveloped dysregulation score (dSCORE) and risk classification algorithmqPCR analysis, dSCORE and risk classification algorithm, sensitivity and specificity analysis, demographic and risk factor analysisMicroRNA for analysis can be predictably isolated from oral swirls sourced from individuals with a range of demographic, systemic, and oral health findings
Zheng, Jun et al. [46]2018Mouth Neoplasms, carcinoma, mouth squamous cell carcinoma, saliva level, early detection of cancer202 individualsObservational clinical studyEnzyme-linked immunosorbent assaysStudent’s t-test, ANOVA, Pearson’s correlation, Spearman’s correlation, ROC analysisSalivary and serum levels of Naa10p and CEA in OSCC patients were significantly higher than those detected in OPML and the control groups, although patients with OPMLs also showed increased salivary and serum Naa10p and CEA levels as compared to the control group
Hes, Cecilia et al. [47]2024Microbiome, Squamous Cell Carcinoma of Head and Neck, metagenomics, DNA library52 patientsProspective studyShotgun metagenomic sequencingWilcoxon rank-sum test, Kruskal–Wallis test, Kaplan–Meier survival analysis, Cox proportional hazards model, PERMANOVA, Spearman’s correlationAll patients developed CRT-induced mucositis, including 42% with severe events (i.e., CTCAE v5.0 grade ≥ 3) and 25% who required enteral feeding
Mehdipour, Masoumeh et al. [48]2023Biomarkers, squamous cells, microRNA60 patients divided into four groupsCase–control studyReal-time quantitative polymerase chain reaction (RT-qPCR)Kruskal–Wallis and Dunn–Bonferroni testsAltered expressions of microRNA-146a and microRNA-155 in dysplastic OLP and OSCC could serve as potential biomarkers for malignancy
Rapado-González, Óscar et al. [49]2022Biomarkers, carcinoma, squamous cells, tumor, Cell-Free Nucleic Acids34 subjects divided into two groupsPreliminary studyThe researchers analyzed the concentration and integrity of cell-free DNA (cfDNA) fragments in saliva samples using molecular techniquesDescriptive StatisticsThe study found significant differences in the integrity and quantity of cfDNA between OSCC patients and healthy controls
Menaka, T.R. et al. [50]2019Biomarkers; saliva; salivary alkaline phosphatase42 individualsObservational cross-sectional studyKinetic photometric methodMann–Whitney U testData obtained were subjected to statistical analysis; the mean S-ALP was 18.00 IU/L for normal individuals without tobacco usage, 4.60 IU/L for smokers without lesions, 7.50 IU/L for tobacco chewers without any lesions, and 64.90 IU/L for individuals with OPMD
Sridharan, Gokul et al. [51]2019Biomarkers, squamous cells, metabolome61 patients Observational case–control studyQ-TOF–liquid chromatography–mass spectrometryMassHunter profile software and Metlin database, ANOVA Significant upregulation of 1-methylhistidine, inositol 1,3,4-triphosphate, d-glycerate-2-phosphate, 4-nitroquinoline-1-oxide, 2-oxoarginine, norcocaine nitroxide, sphinganine-1-phosphate, and pseudouridine in oral leukoplakia and OSCC was noted
Bandara, Herath Mudiyansalage Herath Nihal et al. [52]2019Microbiology, saliva, mouth flora, mycobiome, salivation, biodiversity20 referenced studiesSystematic review Literature review, NGS, bioinformatics analysisDescriptive statistics, ANOVAIdentified Candida as dominant genus; linked fungal diversity to oral diseases like OSCC and periodontitis
Tavakoli, Fatemeh et al. [11]2024Mouth squamous cell carcinoma, biological marker, mouth cancer, diagnosis40 patients with oral squamous cell carcinoma (OSCC)Observational studyIndependent sample t-test or its nonparametric equivalent, Mann–Whitney U testSPSS 17 statistical softwareThe results showed a statistically significant difference in the mean levels of salivary transferrin between the OSCC patients and the healthy controls
Palaia, Gaspare et al. [53]2022miRNAs, OSCC, liquid biopsy34 studiesLiterature searchQualitative and quantitative review, subgroup analysisDescriptive statistics, SIGN checklist for biasThe analysis showed that 57 microRNAs of liquid biopsy samples of four different fluids (whole blood, serum, plasma, and saliva) were analyzed
Afifi, Salsabeel et al. [54]2021Diagnostic accuracy study, oral cancer, potentially malignant lesions, saliva28 participants divided into three groupsProspective pilot studyThe researchers used enzyme-linked immunosorbent assay (ELISA)Chi-square testIndicated its potential as a promising biomarker for the early detection of oral malignancy
Garg, Ruchika et al. [55]2017Oral cancer, oral leukoplakia, oral lichen planus,
potentially malignant conditions, survivin
96 subjectsComparative studyThe researchers used high-throughput sequencing and enzyme-linked immunosorbent assay (ELISA)Mann–Whitney U testThe study found statistically significant differences in the levels of salivary survivin among the groups
Perera, M. et al. [56]201816S, microbiota, ribosomal, RNA, dysbiosis, Sequence Analysis, squamous cells, DNAThe study included 25 cases of oral squamous cell carcinoma (OSCC) and 27 controls with fibroepithelial polyps (FEPs)Case–control studyThe researchers used high-throughput sequencingLinear discriminant analysis effect size (LEfSe), PICRUSt The study found that OSCC tissues had lower species richness and diversity compared to FEP tissues
Farshbaf, Alieh et al. [57] 2024Metabolism, Mouth Neoplasms, biomarkers, squamous cells, biological marker, microRNAs30 healthy control individuals, 30 patients with erosive/atrophic oral lichen planus (OLP), and 31 patients with oral squamous cell carcinoma (OSCC); this is a cross-sectional studyThe researchers used quantitative polymerase chain reaction (qPCR)Shapiro–Wilk test, Kruskal–Wallis test, Pearson’s chi-square test, Mann–Whitney U testThe study found a statistically significant difference in miR-3928 expression between the three groups (p < 0.05)
Nosratzehi, Tahereh et al. [58]2017Biomarkers, carcinoma, tumor75 cases Observational studyEnzyme-linked immunosorbent assay and optical densitySPSS version 20 and one-way ANOVA and LSD tests were used to analyze the dataThe mean salivary endothelin-1 level in patients with OSCC was 163.98 pg/mL; in patients with OLP, it was 160.9 pg/mL; and in healthy people, it was 137.19 pg/ml
Vesty, Anna et al. [59]2018Cytokines, head and neck cancer, oral microbiome, oral mycobiome, saliva30 participantsObservational studyThe researchers used 16S rRNA gene sequencing for bacterial analysis and ITS1 amplicon sequencing for fungal analysisDiversity-based analysesThe study found that the bacterial communities of HNSCC patients were significantly different from those of healthy controls but not from dentally compromised individuals
Blatt, Sebastian et al. [60]2017Biomarkers, carcinoma, squamous cells, tumor, cell cycle regulation, Drug Resistance, prognosis128 studiesSystematic reviewLiterature analysis, systematic review methodology, narrative synthesisNot applicableIn the review, the current evidence on over 100 different biomarkers found for predicting prognosis, outcome, and therapy alterations of OSCC is summarized
Saleem, Zohra et al. [61]2021Head and Neck Neoplasms, squamous cells, Matrix Metalloproteinase 91 subjectsAnalytical studyThe researchers used enzyme-linked immunosorbent assay (ELISA)One-way analysis of variance (ANOVA)The study found a significant difference in salivary MMP-12 expression between the groups (p < 0.001)
Tarrad, Nayroz Abdel Fattah et al. [62]2023Biomarkers, Head and Neck Neoplasms, squamous cells, microRNA, precancerous conditions36 participantsObservational diagnostic studyThe researchers used quantitative real-time PCR (qRT-PCR)Receiver operating characteristic (ROC) curve analysisThe study found that OSCC showed the highest fold change for LINC00657 and the lowest fold change for miRNA-106a among the included groups
Cohen, Erin R. et al. [63]2020Saliva, metabolism, biomarkers, progression-free survival, prognosis64 casesProspective studyImmunohistochemistry, salivary biomarker analysis, multivariate analysisMultivariate analysis, p-values for associationsHigh solCD44 levels associated with strong CD44 tissue expression
Rajaram, Suganya et al. [64]2017Biomarkers, carcinoma, N-acetylneuraminic acid, saliva, serum31 patientsCase–control studyColorimeter, acidic ninhydrin methodStudent’s t-testThere is elevated serum and salivary sialic acid level in moderately/poorly differentiated squamous cell carcinoma without any significant change in well-differentiated squamous cell carcinoma
Kitamura, Naoya et al. [65]2023Head and Neck Neoplasms, squamous cells, DNA115 Japanese patients with oral squamous cell carcinoma (OSCC)Case–control studyThe researchers used quantitative real-time polymerase chain reaction (qPCR)Chi-square testThe study found that 20.9% of the patients were positive for MCPyV DNA
Gaba, Fariah I. et al. [66]2021Biomarkers, carcinoma, Head and Neck Neoplasms, sensitivity and specificity, interleukin 8, diagnostic accuracy17 articlesMeta-analysis Sensitivity and specificity analysisSpecificities of biomarkersSpecificities of the biomarkers analyzed were found to be IL-8 (0.69; 95%CI 0.66–0.99), IL1-β (0.47; 95%CI 0.46–0.90), DUSP-1 (0.75; 95%CI 0.33–1), and S100P (0.73; 95%CI 0.18–0.99)
Cheng, Y.-S.L. et al. [67]2017Metabolism, biomarkers, carcinoma, reverse transcriptase polymerase chain reaction105 human subjectsObservational studyBio-Rad CFX96 Real-Time SystemMann–Whitney U test with Bonferroni correctionsOnly S100P showed significantly higher levels in patients with OSCC compared to both patients with CPNS (p = 0.003) and CPS (p = 0.007)
Shabbir, Alveena et al. [68]2022Mouth Neoplasms, biomarkers, carcinoma80 participantsAnalytical studyThe researchers used enzyme-linked immunosorbent assay (ELISA)One-way analysis of variance (ANOVA)The study found that salivary Cathepsin B levels were significantly increased in patients with OSCC compared to healthy controls (p < 0.001)
Mougeot, Jean-Luc C. et al. [69]2019Streptococcus mutans, bacterial microbiome, cancer radiotherapy, DMFS index, host–bacterium interaction, microbial diversity31 head and neck cancer (HNC) patientsLongitudinal studyThe researchers used high-throughput sequencing of the 16S rRNA geneBeta-diversity analysis and DMFSs (Decayed, Missing, and Filled Surfaces) scoresThe study found significant changes in the oral microbiome at T6 and T18
Ishikawa, Shigeo et al. [70]2019Biomarkers, tumor marker, Hyperplasia48 participantsCase–control studyThe researchers used capillary electrophoresis–mass spectrometry (CE-MS)Multiple logistic regression (MLR)The study identified six metabolites that were significantly different between OSCC/OED and PSOML
Babiuch, Karolina et al. [71]2019Antioxidants, Oxidative Stress, Mouth Neoplasms, biomarkers, squamous cells60 patientsProspective studySalivary biomarkersChi-square test; Dunn’s post hoc test; Kruskal–Wallis test; Spearman’s correlation test; Mann–Whitney testThe activity of SOD was significantly higher in the OSCC group in comparison with the OL and control groups
Shan, Jing et al. [72]2019Saliva analysis, biomarkers, diagnostic test accuracy study, proteomics, early detection of cancer, interleukin 1 receptor blocking agent60 saliva samples Diagnostic test accuracy studyIsobaric tags for relative and absolute quantitation (iTRAQ) methodEnzyme-linked immunosorbent assay (ELISA)In total, 246 differentially expressed proteins were identified by comparing each two groups, and 21 proteins were differentially expressed when OSCC was compared with both OPMD and control
Katase, Naoki et al. [73]2023Squamous Cell Carcinoma of Head and Neck, Cell Line, Western blotting, cell proliferation, Intercellular Signaling Peptides and Proteins, prognosis60 patientsCase–control studyThe researchers used immunohistochemistry (IHC)Chi-square test and logistic regression analysisThe study found that high expression levels of DKK3 and CKAP4 were significantly associated with advanced stage and poorer prognosis in oral cancer patients
Abdelwhab, Amira et al. [74]2023Metabolism, Mouth Neoplasms, biomarkers, Head and Neck Neoplasms, histology, dysplasiaForty oral potentially malignant disordersExperimental studyReal-time PCRComparison between groupsMucin1 expression in saliva was significantly elevated in oral potentially malignant disorders when compared with controls
Kallalli, Basavaraj N. et al. [75]2016Histopathology, mouth disease, saliva level 60 subjectsObservational studyERBA-CHEM 5 semi-auto-analyzerDescriptive statistics and paired t-test using the SPSS softwareThe mean LDH levels were as follows: Group I, 608.28 ± 30.22; Group II, 630.96 ± 39.80; and Group III, 182.21 ± 34.85
Chapple, Iain L C et al. [76]2018Periodontitis, gingivitis, peri-implantitis26 expertsConsensus reportThe report involved a comprehensive reviewQualitative analysis and expert consensus rather than quantitative statistical methodsThe report introduced a new classification system for periodontal and peri-implant diseases and conditions, emphasizing a more comprehensive approach that includes stages and grades of periodontitis, as well as conditions affecting peri-implant health
Jolivet-Gougeon, Anne et al. [77]2021Microbiota, RNA 16S, dental caries, microbial diversity, immunocompromised patient, prevalence42 papersScoping reviewPreferred Reporting Items for Systematic Reviews and Meta-analysis Protocols (PRISMA—ScR)A data-charting form was usedThey showed a link between the abundance of Capnocytophaga spp. in the oral microbiota and various local pathologies (higher for gingivitis and halitosis; lower in active smokers, etc.) or systemic diseases
(higher for cancer and carcinomas, IgA nephropathy, etc.)
Ishikawa, Shigeo et al. [78]2020Saliva analysis, cancer diagnosis, squamous cell carcinoma60 patientsCase–control studySalivary biomarker analysisMultiple logistic regression (MLR); Mann–Whitney U testSaliva analysis showed significant differentiation between SCC patients and control groups
Banavar, Guruduth et al. [79]2023RNA, saliva analysis, biomarkers, cancer staging, genomic RNA, head and neck tumor, sensitivity and specificity1.175 individualsObservational studyMachine learning analysisReceiver operating characteristic (ROC) The classifier showed a specificity of 94% and sensitivity of 90% for participants with oral squamous cell carcinoma (OSCC) and 84.2% for participants with oropharyngeal squamous cell carcinoma (OPSCC)
Hashimoto, Kengo et al. [42]2022Saliva analysis, microbial diversity, Bacteroidetes, high-throughput sequencing, cancer recurrence86 participantsObservational studyThe researchers used next-generation sequencing (NGS)Fisher’s exact test, chi-square test, logistic regression analysisThe study found significant differences in the abundances of certain bacterial genera and phyla among the groups
Zhong, Xiaohuan et al. [80]2021RNA 16S, Mouth Neoplasms, microbial community, community dynamics, oral submucous fibrosis162 participantsCross-sectional study16S rRNA gene sequencingPERMANOVA; Wilcoxon rank-sum; Kruskal–WallisThe study found that the oral microbiome of people who chew areca nuts is different from that of people who do not chew areca nuts
Khalil, Marwa et al. [81]2019Leukoplakia; oral squamous cell carcinoma; saliva; zinc45 patientsRandomized clinical trialStandard spectrophotometric methodsANOVA and chi-square testThere was a highly significant decrease in the level of salivary Zn in patients with OSCC when compared to OL patients and controls (p < 0.05)
Chari, Abinaya et al. [82]2016Biomarkers, squamous cells, Surveys and Questionnaires, blood35 patientsCase–control studyBlood biomarker analysisTwo-tailed t-test and chi-square analysisThe mean serum LDH value for patients with smokeless tobacco-related oral lesions was 446.8 U/L, compared to 269.4 U/L for healthy controls
Chen, Jijun et al. [43]2023RNA, real-time polymerase chain reaction, biomarkers, Squamous Cell Carcinoma of Head and Neck, cell migration, blood, protein expression132 participantsCase–control studyReal-time PCR analysisANOVA, Tukey testThe study found that miR-19a, GPR39 mRNA, and PKC mRNA were upregulated while GRK6 mRNA was downregulated in the serum and saliva samples of OSCC patients compared to healthy controls
Sayal, Lana et al. [83]2023Saliva analysis, biomarkers, Head and Neck Neoplasms, sex difference, early cancer diagnosis, liquid biopsy133 leukoplakia patients versus 137 healthy volunteersObservational studySalivary cf-mtDNA and cfDNA were quantified using Multiplex Quantitative PCRChi-square test; Shapiro–Wilk test of normality; nonparametric tests (Mann–Whitney U test and Kruskal–Wallis H test)The study found that the median scores of cfDNA and cf-mtDNA were significantly higher among HNSCC patients compared to healthy controls and OLK patients
Yuanbo, Zhan et al. [16]2024DNA, Papillomavirus Infections, tongue tumor60 patientsPilot studyThe researchers used 16S rRNA gene sequencingWilcoxon test; multiple comparisons using FDR p-value correction; Spearman’s rank testThe study found that microbiota diversity was significantly increased in p16-positive patients compared to p16-negative patients
Rani, N. Alice Josephine et al. [10]2023ELISA; interleukin 6; oral potentially malignant disorders60 patientsCross-sectional comparative studySaliva samples; ELISA Shapiro–Wilk test; one-way ANOVA; post hoc Tukey test; Kruskal–Wallis test The study found that the concentration of IL-6 was significantly higher in the OSCC group compared to the other three groups
Ameena, M. et al. [84]2019ELISA; leukoplakia; oral squamous cell carcinoma90 participantsComparative studyELISA testStatistical analysis was performed using the one-way ANOVAThe study found that salivary TNF-α levels were significantly elevated in patients with OSCC compared to those with leukoplakia and healthy controls
Yost, Susan et al. [85]2018Microbiota, squamous cells, transcriptome, Virulence, MetagenomeSmall pilot study with a limited number of participantsPilot studyMetatranscriptome analysisLEfSe; Kruskal–Wallis (KW) sum-rank test; Wilcoxon testThe study found that Fusobacteria exhibited a statistically significant increase in transcript abundance at tumor sites and tumor-adjacent sites in cancer patients compared to healthy controls
Rapado-González, Óscar et al. [34]2024Biomarkers, cancer diagnosis, gene expression, genomic DNAStudy included six consecutive patientsCross-sectional studyGenome-wide DNA methylation profilingPrincipal component analysis (PCA) and ROC curve analysis were assessed to obtain the best modelsThe study identified a group of novel tumor-specific DNA methylation markers with diagnostic potential in saliva
Feng, Yun et al. [86]2019Saliva, metabolism, biomarkers, squamous cells16 patientsObservational studyThe researchers used human protease array kits, enzyme-linked immunosorbent assay (ELISA), Western blot, and immunofluorescenceOne-way analysis of variance (ANOVA)The study found that the salivary protease spectrum was significantly associated with oral diseases
Ghallab, Noha A. et al. [87]2017Saliva, metabolism, biomarkers, Intercellular Signaling Peptides and Proteins45 individualsCase–control studyThe researchers used enzyme-linked immunosorbent assays (ELISAs)The study employed receiver operator characteristic (ROC) curve analysisThe study found that serum and salivary levels of chemerin and MMP-9 were significantly higher in patients with OSCC compared to those with OPMLs and healthy controls
Sawant, Sharada et al. [88]2016Biomarkers, squamous cells, prognosis, CD44 protein, survival rate87 patientsProspective studyThe researchers performed immunohistochemistryChi-square test and Fisher’s exact test The study found significant correlations between the expression of Oct4, CD44, and c-Myc with overall survival (OS) and disease-free survival (DFS) independently
Lee, L.T. et al. [89]2018Mouth tumor, biomarkers, carcinoma, cytokine, blood sampling, interleukin 1 beta, interleukin 6, risk factors65 patientsCase–control studyLuminex Bead-based Multiplex AssayMann–Whitney U test and Kruskal–Wallis testThe study found that plasma levels of IP-10 in early-stage OSCC patients differed significantly from those in controls
Triani, Maulina et al. [90]2021Betel nut; Ki-67; micronucleus; oral squamous cell carcinoma; saliva60 participantsCross-sectional analytic surveyPapanicolaou methodMann–Whitney and Kruskal–Wallis testsThe study found significant differences in Ki-67 expression and micronucleus counts between the betel nut chewers with OSCC and the control groups
Perera, Manosha et al. [91]2017Mycobiome, carcinoma, DNA ribosomal spacer, microbiome, squamous cells52 tissue biopsiesObservational studyThe researchers used Illumina sequencing and BLASTN algorithmLinear discriminant analysis effect size (LEfSe)The study found that the mycobiome in OSCC was dominated by Candida albicans, with significantly lower species richness and diversity compared to FEPs
Chan, Jason Y.K. et al. [92]2022Ribosomal, RNA 16S, dysbiosis, head and neck squamous cell carcinoma, genotyping166 Chinese adults Cohort studyPermutational multivariate analysis of variance (PERMANOVA)Mann–Whitney; Wilcoxon rank-sum testThe study found that 15.7% of the HNSCC patients were positive for HPV DNA, with infection rates varying by cancer subtype
Kaur, Jasdeep et al. [93]2016Biomarkers, sensitivity and specificity, risk factor, biopsy200 participantsObservational studyReceiver operating characteristic (ROC)
analysis
Mann–Whitney U testThe findings revealed that patients with OSCC and precancerous conditions exhibited significantly higher salivary levels of 8-OHdG and MDA, alongside lower levels of vitamins C and E, compared to healthy controls
Zhu, Weiwen et al. [36]2024Squamous cells, Streptococcus, Capnocytophaga gingivalis, Cell Line, Fluorescence, In Situ Hybridization178 participantsComparative observational study16S rRNA gene sequencingPERMANOVA, t-test, ANOVA, regressionThe findings revealed that the overall microbiome diversity was higher in healthy controls compared to OSCC patients
Chen, M.Y. et al. [39]2021Microbiota, Head and Neck Neoplasms, artificial intelligenceOSF (n = 18) and OSCC-OSF (n = 34) groupsComparative observational study16S rRNA gene sequencingANOVA, regression, Alpha-diversity indicesThe study found significant differences in the salivary microbiomes between the OSCC-OSF and OSF groups
Kumaran, Jimsha et al. [94]2022MicroRNA; non-invasive method; potential biomarkerDoes not specify the exact numberObservational studyQuantitative real-time PCR (qRT-PCR)Not applicableDiscusses the differential expression of specific salivary miRNAs and their potential roles as biomarkers for oral diseases
Suresh, H. et al. [95]2022Alpha-L-fucosidase; oral squamous cell carcinoma; oral submucous fibrosis; salivary biomarker40 participantsComparative studyEnzyme-linked immunosorbent assay (ELISA)Pearson’s correlationThe study found a significant increase in AFU levels in both saliva and serum of OSMF patients compared to healthy controls
Ferrazzo, Kívia Linhares et al. [96]2022Saliva; biomarkers; squamous cells 20 participants Case–control studyQuestionnaire; saliva samples; liquid chromatography–mass spectrometry (LC-MS/MS)Shapiro–Wilk test; Student’s t-test; Mann–Whitney test The results suggest that while salivary pipecolic acid demonstrates high sensitivity, its specificity is moderate, indicating potential as a non-invasive biomarker for HNSCC detection
SHahinas, J. et al. [97]2018Metabolism, biomarkers, carcinoma, squamous cell carcinoma, prognosis, immunohistochemistry36 articlesSystematic review Quality in Prognosis Studies (QUIPS), data extraction formCox regression and Kaplan–MeierThe findings indicated that the majority of the reviewed studies were replication prognostic factor studies (35 out of 36)
Huang, Long et al. [98]2022Biomarkers, carcinoma, diagnostic test accuracy study, diagnostic valueSix studiesMeta-analysisHierarchical analysisQuality Assessment for Studies of Diagnostic Accuracy 2The pooled sensitivity and specificity of circRNAs for OSCC diagnosis were 0.72 and 0.81, respectively
Furquim, C.P. et al. [99]2017Microbiota, saliva, squamous cells, risk factors, Gingival Hemorrhage61 patients Cross-sectional16S rRNA gene sequencingGeneral linear modelsThe analysis revealed that the most abundant bacterial phyla in the salivary microbiome of FA patients were Firmicutes and Bacteroidetes
Susha, Karthika Pradeep et al. [100]2023Salivary biomarker, oral squamous cell carcinoma, oral leukoplakia, early diagnosis90 patientsCase–control studySalivary chemerin; ELISAIndependent t-test; analysis of variance test (F test); Scheffe’s multiple comparisons (post hoc test)The study found that salivary chemerin levels were significantly higher in the OSCC group compared to both the OL and HC groups
Liu, Wei et al. [101]2023Bubble analysis; non-invasive diagnosis; oral cancer; oral potentially malignant disorders; saliva; microRNAs17 eligible studiesSystematic reviewBubble chart analysisExcel Visual Basic for ApplicationsThe analysis identified that miR-21 exhibited the highest diagnostic power for detecting the onset of OPMDs, followed by miR-31 and miR-142
Peisker, Andre et al. [102]2017Biomarkers, carcinoma, biopsy, enzyme-linked immunosorbent assay60 participantsCase–control studySaliva samples; enzyme-linked immunosorbent assay (ELISA)Mann–Whitney U testThe median absorbance MMP-9 value for the OSCC group was 0.186 (IQR = 0.158), while the control group had a median value of 0.156 (IQR = 0.102)
Nakamichi, Eiji et al. [103]2021Saliva, biomarkers, carcinoma, Western blotting, immunoreactivity, protein analysis57 patientsCase–control studySerum/salivary exoAlix levelsChi-square tests; Mann–Whitney
U tests; Kruskal–Wallis test; Dunn’s tests; Fisher’s exact tests; Spearman’s correlation coefficient by rank; Wilcoxon matched-pairs signed-rank tests
The findings revealed that both serum and salivary exoAlix levels were significantly higher in OSCC patients compared to healthy controls
Nguyen, Truc Thi Hoang et al. [104]2020Carcinogenesis, saliva protein, transcriptomics, proteomics, metabolomics, liquid biopsyThis study did not involve a specific sample sizeNarrative reviewLiterature reviewNot applicableThe review highlights several salivary biomarkers with potential diagnostic and prognostic value for OSCC
Robledo-Sierra, Jairo et al. [105]2019Microbiota, carcinoma, squamous cells, bacterium identification23 studies on oral squamous cell carcinoma, 2 studies on oral leukoplakia, and 4 studies on oral lichen planusSystematic reviewThe researchers performed a comprehensive literature review and meta-analysisThe study employed qualitative synthesis and quantitative analysisThe study found substantial differences in diagnostic criteria, sample types, regions sequenced, and sequencing methods across the included studies
Abdul Aziz Shaikh, Sabiha et al. [106]2024TNF-α, oral submucosal fibrosis, squamous cell carcinoma, salivary biomarkers, chronic inflammation45 participantsCase–control studySaliva samples; salivary TNF-α levels; ELISA kitANOVA and post hoc Tukey HSDThe analysis revealed no significant differences in salivary TNF-α levels among the OSMF, SCC, and control groups
Vimal, Joseph et al. [107]2023Biomarkers, Head and Neck Neoplasms, Squamous Cell Carcinoma of Head and Neck20 samples from patientsCase–control studyUntargeted metabolomicsProgenesis QIThe study identified N-Acetyl-D-glucosamine, L-Pipecolic acid, and L-Carnitine as signature diagnostic biomarkers for oral tongue squamous cell carcinoma
Man, Qi-Wen et al. [108]2023Mouth Neoplasms, biomarkers, flow cytometry, protein expression, prognosis61 patients with OSCC, 21 healthy patientsShort communicationSalivary mRNAMann–Whitney test, univariate and multivariate analysesIdentified potential of salivary mRNA biomarkers for OSCC early prediction
Yang, Shun-Fa et al. [109]2018Genomics, microbiota, microbial community, histopathology103 participantsObservational study16S rRNA amplicon sequencingTaxonomic analysisThe analysis revealed three distinct MSC groups in OSCC patients, each significantly associated with specific demographic and clinical features
Yang, S.F. et al. [110]2022Microbiota, RNA, mouth tumor, Head and Neck Neoplasms, squamous cells315 participantsMeta-analysisSequencing the salivary microbiomeQuantitative trait loci (QTL)The study identified associations of three genetic loci with the abundance of specific bacterial genera and four loci with measures of β diversity
Tantray, Shoborose et al. [111]2022Metabolite, OSCC, OLK90 participantsComparative observational studyThe researchers employed gas chromatography–mass spectrometry (GC-MS)Principal component analysis (PCA)Fifteen metabolites showed significant differences between the control, OLK, and OSCC groups
Gulzar, Rukhsaar Akbar et al. [112]2021Salivary MUC1, cancerous conditions30 patientsComparative observational studyELISAANOVAMUC1 levels were highest in OSCC (19.33 ± 4.39 ng/mL), followed by premalignant (8.32 ± 3.08 ng/mL) and control (2.73 ± 0.34 ng/mL)
Gupta, Archana et al. [113]2019Mustard oil, dry socket, Curcumin, Holy powder, anti-inflammatory178 patientsRandomized clinical trialComparative analysis of treatment outcomes between two groupsChi-square testGroup A (turmeric and mustard oil) showed faster symptom relief (starting by day 2) compared to Group B (zinc oxide eugenol, relief by day 4)
Ahmed, Ahmed A. et al. [33]2024Saliva, Mouth Neoplasms, DNA sequencing, biomarkers, circulating free DNA, liquid biopsy250 participantsObservational studyDescriptive and inferential statisticsNuclear morphometric analysis (NMA) using Image J, receiver operating characteristic (ROC) analysisSignificant differences in nuclear morphometric parameters (NMPs) among different smoking groups
Karmelić, Ivana et al. [114]2022Oral cancer; early diagnosis; biomarkers; saliva58 patientsCross sectionEnzyme-linked immunosorbent assay (ELISA) testingMann–Whitney U test and Kolmogorov–Smirnov testDifference between the OSCC group and the control group was found between the
levels of SCCA1 and SCCA 2 both in UWS and SWS
Tang, Kai Dun et al. [115]2024Metabolism, biomarkers, Head and Neck Neoplasms, microRNA66 participantsPilot studyQuantitative analysis using qPCR (quantitative polymerase chain reaction)t-tests; one-way ANOVA; Tukey–Kramer HSD multiple comparison; Pearson’s correlationThe study identified specific miRNAs (miR-744, miR-150-5P, and miR-146B-5P) with high discriminatory capacity between Fanconi anemia patients and healthy controls
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dumitrescu, R.; Bolchis, V.; Fratila, A.D.; Jumanca, D.; Buzatu, B.L.R.; Sava-Rosianu, R.; Alexa, V.T.; Galuscan, A.; Balean, O. The Global Trends and Advances in Oral Microbiome Research on Oral Squamous Cell Carcinoma: A Systematic Review. Microorganisms 2025, 13, 373. https://doi.org/10.3390/microorganisms13020373

AMA Style

Dumitrescu R, Bolchis V, Fratila AD, Jumanca D, Buzatu BLR, Sava-Rosianu R, Alexa VT, Galuscan A, Balean O. The Global Trends and Advances in Oral Microbiome Research on Oral Squamous Cell Carcinoma: A Systematic Review. Microorganisms. 2025; 13(2):373. https://doi.org/10.3390/microorganisms13020373

Chicago/Turabian Style

Dumitrescu, Ramona, Vanessa Bolchis, Aurora Doris Fratila, Daniela Jumanca, Berivan Laura Rebeca Buzatu, Ruxandra Sava-Rosianu, Vlad Tiberiu Alexa, Atena Galuscan, and Octavia Balean. 2025. "The Global Trends and Advances in Oral Microbiome Research on Oral Squamous Cell Carcinoma: A Systematic Review" Microorganisms 13, no. 2: 373. https://doi.org/10.3390/microorganisms13020373

APA Style

Dumitrescu, R., Bolchis, V., Fratila, A. D., Jumanca, D., Buzatu, B. L. R., Sava-Rosianu, R., Alexa, V. T., Galuscan, A., & Balean, O. (2025). The Global Trends and Advances in Oral Microbiome Research on Oral Squamous Cell Carcinoma: A Systematic Review. Microorganisms, 13(2), 373. https://doi.org/10.3390/microorganisms13020373

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop