Preprint
Review

Attempts to Understand Oral Mucositis in Head and Neck Cancer Patients Through Omics Studies

Altmetrics

Downloads

278

Views

149

Comments

0

Submitted:

21 April 2023

Posted:

23 April 2023

You are already at the latest version

Alerts
Abstract
Oral mucositis (OM) is inflammation of the mouth caused by damage to the mucous membranes that line the mouth and throat. It is a side effect of cancer treatment, particularly in patients with head and neck squamous cell carcinoma (HNSCC) who undergo radiotherapy, chemotherapy, and/or immunotherapy with immune checkpoint inhibitors. The etiology and pathogenic mechanisms of OM is complex and multifaceted, involving cytotoxicity (cell death), inflammation, infection, change in microbiome, and immune-mediated cytotoxicity. We summarize the literature about attempts to use various omics methodologies (genomics, transcriptomics, microbiomics and metabolomics) to elucidate the biological pathways associated with the development or the severity of OM. Integrating different omics into multi-omics approaches carries the potential to discover links among host factors (genomics), host responses (transcriptomics, metabolomics), and local environment (microbiomics).
Keywords: 
Subject: Biology and Life Sciences  -   Biochemistry and Molecular Biology

1. Introduction

Head and neck squamous cell carcinoma (HNSCC) is the seventh [1] most common type of cancer worldwide. There were an estimated 54,000 new cases in the United States in 2022 [2]. The treatment of HNSCC is based largely on the primary tumor location and stage of the disease. Early-stage disease is treated with single-modality treatment, and advanced-stage disease is treated with multi-modal therapy. The majority of HNSCC patients present with loco-regionally advanced disease. The treatment modalities for HNSCC with curative intent are surgery and/or radiotherapy with or without concurrent systemic therapy. However, treatment-related toxicities are a significant concern. In 2016, the American Cancer Society guidelines [3] for HNSCC underscored the need to recognize the potential late and long-term complications or toxicities of cancer treatment, as well as its undertreatment and management.
Cancer immunotherapy using immune checkpoint inhibitors (ICIs) is a recent advance in HNSCC treatment. In recent years, there have been several combination treatments approved by the FDA for recurrent or metastatic HNSCC that include ICIs [4]: pembrolizumab in combination with platinum and fluorouracil chemotherapy, nivolumab plus ipilimumab, cetuximab plus pembrolizumab, and durvalumab plus tremelimumab. ICI monotherapy (pembrolizumab or nivolumab) is approved by the FDA for the treatment of recurrent or metastatic HNSCC that have progressed on or after platinum-based chemotherapy. Pembrolizumab is also approved as monotherapy for the first-line treatment of patients with recurrent or metastatic HNSCC whose tumors express PD-L1 and who are not candidates for platinum-containing chemotherapy. Nivolumab is also approved as monotherapy for the treatment of patients with HNSCC who have metastatic disease and are intolerant to platinum-based chemotherapy.
OM is a painful and debilitating treatment-related toxicity in HNSCC patients. OM is a common side effect of cancer treatment, particularly in patients with HNSCC who undergo radiation therapy and/or chemotherapy. The mucous membranes of the mouth and throat are highly sensitive to radiation and chemotherapy, which can damage the cells and cause inflammation. OM is clinically characterized by erythematous lesions, ulcers, and edema in the oral mucosa. This can lead to pain, swelling, difficulty eating, drinking, and speaking, and an increased risk of infection. The incidence of OM caused by chemotherapy or radiotherapy can be very high, ranging from 40% to 90% [5,6].
OM is observed to appear around a week after chemotherapy and resolve a week after the cessation of chemotherapy. Whereas among those receiving radiotherapy, a dose-response relationship is observed, with OM appearing after a cumulative dose of 30 Gy. Patients with OM complain of painful mouth ulcers and difficulty eating or swallowing. OM-related complaints lead to dose reduction or interruption of treatment, increased opioid consumption, visits to the emergency department (ED), and hospitalization. While most patients develop OM, studies show individual variability in the severity and persistence of OM despite receipt of similar cancer treatments for HNSCC. In a recent study, up to 90% of patients developed OM, but only 36% developed severe OM [7]. Longitudinal patterns of OM also vary, with some patients experiencing early resolution [8,9,10] whereas others develop chronic [11] OM. Moreover, for some HNSCC patients, visits to the ED for OM-related complaints, including severe pain, occur within 7 days of the initiation of cancer treatment and may persist with pain as a chief complaint across multiple ED visits for up to 23% of HNSCC patients [12].
The above profiles of OM in HNSCC patients were based on studies prior to the common use of ICIs for HNSCC treatment. More recently, OM was also observed as treatment-related toxicity from ICI therapy [13,14,15]. Immune-mediated OM is caused by the immune system attacking the lining of the mouth and throat. The incidence rate of OM after ICI therapy appears to be much lower than that caused by chemotherapy or radiotherapy. The incidence rates of OM can vary depending on the specific treatment regimen, patient population, and other factors. For example, the incidence of grade 1-2 OM induced by pembrolizumab in solid tumor patients was 12.2%, and the incidence of grade 3-4 oral mucositis was 0.5%. Clinical features that may suggest immune-mediated mucositis include the timing of onset (typically within weeks to months of initiating immunotherapy), the presence of other immune-related adverse events, and the lack of response to conventional mucositis treatments such as pain relievers and topical anesthetics. Histological features that may suggest an immune-mediated cause include the presence of dense lymphocytic infiltrates and the absence of chemotherapy-related changes such as atrophy and fibrosis. If immune-mediated mucositis is suspected, a corticosteroid trial may be considered to determine if the symptoms improve with treatment. A positive response to corticosteroids would support an immune-mediated etiology and suggest that continued immunosuppressive therapy may be beneficial.
In patients with HNSCC, OM is a significant problem because it can interfere with treatment and impact their quality of life. Patients may require dose reductions or delays in their cancer treatment, which can decrease the effectiveness of the antineoplastic therapy. Additionally, OM can lead to malnutrition and dehydration, further compromising the patient's health and well-being. Therefore, effective management of OM is an important part of cancer treatment and may involve a range of supportive care measures to manage symptoms and prevent complications, such as pain management, nutritional support, oral hygiene measures, anti-infectives, and glucocorticoids. Studies continue to explore interventions [16,17,18,19] and potential treatments [20,21,22] for OM, showing mixed results. In 2020, the Multinational Association of Supportive Care in Cancer and International Society of Oral Oncology (MASCC/ISOO) updated their Clinical Practice Guidelines for the management of mucositis [23]. Among HNSCC patients, the use of benzydamine (a non-steroidal drug with local anti-inflammatory, analgesic, antipyretic, and anti-edema effects) was added as a new suggestion for the prevention of OM when receiving radiotherapy and chemotherapy and a recommendation for the prevention of OM with intraoral photobiomodulation therapy among those receiving radiotherapy with or without concurrent chemotherapy.
Clinician-rated and patient-rated scales are used to characterize OM. Table 1 shows a list of clinician-rated and patient-rated OM scales. Self-report measures of OM severity and patient-reported outcome measures of the impact of OM on bodily function and quality of life, have also been developed and validated for use in different languages. The scales developed by the World Health Organization [24], Radiation Therapy Oncology Group [25], and US National Institutes of Health (i.e., Common Terminology Criteria for Adverse Events) [26] are the most frequently used OM scales, especially in large clinical studies. Importantly, a recent study by Villa and colleagues [27] found discrepancies in concordance between these 3 scales. However, they noted that scale selection is less critical in studies where severe mucositis is the outcome of interest, but that scale selection is particularly important if the focus is to describe the clinical trajectory of OM and its impact.
Epidemiological, behavioral, and clinical variables explain some of the variation observed in OM. For some patients, OM may be very painful, leading to treatment interruption, increased opioid consumption, and increased healthcare utilization. With advances in molecular technology, studies have explored biomarkers for identifying patients at risk for severe OM and assessing potential biological mechanisms of this complex trait. Although there have been studies attempting to identify markers of risk and potential therapies for OM aiming to improve quality of life, similar studies will need to be conducted to reflect the addition of immune-mediated OM into the mix of OM in HNSCC patients in this new era of cancer immunotherapy.
Figure 1 shows factors associated with OM, i.e., older age, male gender, oral hygiene, total radiation dose, smoking, systemic diseases, radiotherapy technique, combined chemoradiation, ICI, malnutrition or cachexia, and lack of antibiotic use at the early stage of OM. However, evidence suggests these factors only explain some of the variation observed in OM.
OM may be a classic example of gene-environment interaction, requiring an initiating event (chemotherapy, radiation, or ICI therapy) and host genetic susceptibility. As a complex human trait, it is expected that multiple genes underlie the development of OM. Assessing the interaction of these genes with epidemiological, behavioral, and clinical variables will provide a better understanding of OM and allow early identification of patients at risk. With advances in high throughput molecular laboratory techniques, it is now possible to assess biomarkers that measure events at the physiological, cellular, and molecular levels. We reviewed studies conducted from 2016–2022 to identify putative biomarkers for OM (Figure 2), for predicting the severity of OM and identifying novel therapeutic targets. The goal is to highlight the knowledge gap that is required to improve patient outcomes by enabling personalized treatment strategies and reducing the burden of OM.

2. "Omics" approaches may help in risk assessment and developing personalized and more effective therapies OM

2.1. Genomics

The Human Genome Project and the HapMap Project gave a rough sketch of how people's genes differ [37]. The Telomere-to-Telomere Consortium completed sequencing the last 8% of DNA of the human genome, i.e., the part of the human genome not covered by the Human Genome Project, in 2022 [38]. Now the complete blueprint of the human body can accelerate the advance in the study of human biology and medicine. Single nucleotide polymorphisms (SNPs), which happen about once every 1000 bases in a genome with 3 billion base pairs, are the most common type of DNA variation. SNPs that change how the gene product works are usually further examined because they may make individuals more likely to get sick or change how they react to drugs. Also, SNPs with a minor allele frequency of more than 5% are chosen because they have a higher chance of being linked to complex diseases. Both hypothesis-driven (candidate-gene, pathway-based) and hypothesis-generating (i.e., genome-wide association) approaches have been used to identify genes for OM.

2.1.1. Hypothesis-driven Approach

In a hypothesis-driven approach, researchers use knowledge of polymorphisms and gene functions in the candidate gene, investigating one or a few selected polymorphisms at a time.

2.1.2. Candidate-Gene Studies

Studies have demonstrated associations between gene polymorphism, DNA repair function, and sensitivity to radiotherapy. Using the search terms 'SNPs and mucositis" and "gene(s) and mucositis", Reyes-Gibby et al. [39] conducted a literature search of human studies of OM published before 2016. Their review of the literature identified 27 genes across the various studies, with the most commonly cited genes involved in methylation, DNA synthesis, and DNA repair mechanisms, i.e., X-ray repair cross-complementing gene 1 (XRCC1), and excision repair cross complementation group 1 (ERCC1).
A subsequent review of the literature for the period 2015–2022 showed studies focusing on HNSCC patients who received radiation therapy. Five studies were conducted on a small number of HNSCC patients receiving radiation therapy in the oncology department at the Medical University of Lublin (Lublin, Poland). They showed that polymorphisms in GHRL (rs1629816) [40], TNFRS1A [41] (rs4149570, rs767455) [42]), TNFA (rs1799964) [43], and APEH (rs4855883) [44] were significantly associated with OM. In a larger sample (n = 114) of Chinese patients with HNSCC receiving radiotherapy, Chen et al. [45] found the XRCC1 variant (rs25487) to be associated with an increased risk of OM.

2.1.3. Pathway-Based

Although comprehensive, the pathway-based approach relies on a priori knowledge of SNPs and gene functions and biological plausibility. Reyes-Gibby et al. [39] used a pathway-based approach for OM. They first reviewed literature before 2015 on genetic association studies of OM. They found 27 genes from 28 published studies. Using the 27 genes, they generated gene networks for OM using Ingenuity Pathway Analysis (IPA), which is a bioinformatics tool using the Ingenuity Knowledge Base comprised of information on biomolecules and their relationships. They found TP53, CTNNB1, MYC, RB1, P38 MAPK, and EP300 as the most biologically significant molecules and the uracil degradation II (reductive) and thymine degradation pathways as the most significant biological pathways. They then conducted a genetic association study for OM in 885 HNSCC patients (with OM = 186; without OM = 699) utilizing 66 SNPs within the 8 most connected IPA-derived candidate molecules. The top-ranked gene identified through this association analysis was RNA-binding proteins (RBP) (rs2227311, p-value = 0.034, odds ratio = 0.67). To date, there has been a limited application of the pathway-based approach in the study of OM.
Table 2. Summary of identified genes and corresponding SNPs that were found to be implicated in oral mucositis in head and neck cancer patients.
Table 2. Summary of identified genes and corresponding SNPs that were found to be implicated in oral mucositis in head and neck cancer patients.
Year First Author Sample Size Sample Phenotype Genes SNPs
2022 Schack [46] Discovery=1183
Danish Cohort
Replication=597 Danish Cohort
Validation=235 Asian Cohort
Buffy coats Mucositis
0: no
1: erythema
2: patchy
3: confluent 4:ulceration
STING1 rs1131769
2020 Mlak [42] 60 Peripheral blood Mucositis
RTOG/EORTC
TNFRS1 A rs767455
2020 Mlak [43] 62 Peripheral blood Mucositis
RTOG/EORTC
TNF alpha rs1799964
2020 Yang [47] 960
560
Blood RTOG/EORTC TNKS rs117157809
2018 Brzozowska [44] 62 Peripheral
blood
Mucositis
RTOG/EORTC
APEH rs4855883
2018 Brzozowska [41] 58 Peripheral
blood
Mucositis
RTOG/EORTC
TNFRS1 A rs4149570
2018 Brzozowska [40] 65 Peripheral blood Mucositis
RTOG/EORTC
GHRL Rs1629816
2017 Chen [45] 114 Peripheral blood Mucositis
RTOG/VRS
XRCC1 rs25487
2017 Reyes-Gibby [39] 885 Peripheral blood Oral Mucositis (ICD) RB1 rs2227311
RTOG-Radiation Therapy Oncology Group; WHO-World Health Organization; EORTC-European Organization for Research and Treatment of Cancer; ICD-International Classification of Diseases.

2.1.4. Genome-wide Approach

Hypothesis-free, whole genome-wide association studies are effective for identifying genetic factors contributing to complex diseases. However, there has been little use of this approach in research because of the cost and the need for very large sample sizes for replicable results. A study by Schack et al. [46] utilized 3 cohorts (discovery phase: 1183 Danes, replication phase: 597 Danes, and validation phase: 235 Asians) and found the rs1131769 of the STimulator of Interferon Response cGAMP Interactor 1 gene (STING1) to be significantly associated with OM. STING1 (also known as transmembrane protein 173) has been associated with infection, inflammation, immunity, autophagy, and cell death [48,49,50,51]. An earlier genome-wide study by Wang et al. [47] found SNP rs117157809 in the protein-coding gene TNKS (Tankyrase) associated with more than three-fold OM risk in patients with nasopharyngeal cancer. TNKS plays a role in radiation-induced damage. Depletion of TNKS is associated with increased sensitivity to ionizing radiation-induced mutagenesis, chromosone abberation, telemore fusion and cell-killing [52].

2.2. Microbiomics of OM

The application of microbiomics has been used in OM research in HNSCC patients since the microbiome (the collective community of microorganisms) in the oral cavity is implicated in the development of OM. It is hypothesized that changes in the composition of the oral microbiome can increase inflammation and tissue damage in the oral mucosa.
Reyes-Gibby et al. [7] identified different features associated with the risk of OM at baseline (Cardiobacterium, Granulicatella), immediately before the development of OM (Prevotella, Fusobacterium, Streptococcus), and immediately before the development of severe OM (Megasphera, Cardiobacterium). Interestingly, Prevotella and Fusobacterium have also been identified in the study of Hou et al. [53], where these features showed dynamic synchronous variations in abundance throughout the course of radiation therapy and frequently coincided with the onset of severe OM.
The combination of microbiomic and genomic data may also be a powerful approach to identifying key features for therapy. For example, in 24 HNSCC patients who received radiotherapy and concomitant chemoradiotherapy, microbial species (Staphylococcus aureus, S. epidermis, Pseudomonas aeruginosa, E. coli, and Klebsiella pneumoniae) from saliva samples expressed higher levels of antibiotic-resistance genes (VIM2, MCR-1, TET(K), blaKPC) after receiving cancer therapy [54].
Microbiomics has emerged as an important field of research in the study of OM, despite several challenges. For example, the interpretation of microbiome data can be challenging with the utilization of complex tools, a lack of standardization, and highly variable data that are easily influenced by factors such as diet, medications, and even oral hygiene. Moreover, observed changes in the oral microbiome associated with OM are not adequate to establish their causal or modifier roles in OM. Treatment-induced dental caries, hyposalivation, and xerostomia may change the environment in the upper orodigestive tract to affect the composition of the microbiome.

2.3. Metabolomics

Metabolomics involves the comprehensive characterization of low-molecular-weight molecules, metabolites, and metabolism in biological systems. Studies have collected saliva, serum, and volatile organic compounds to reflect various pathological conditions, making it an attractive source for the diagnosis of systemic diseases and potential biomarkers of pathological conditions. Analytical platforms such as liquid chromatography-mass spectrometry (LC-MS), gas chromatography-mass spectrometry (GC-MS), capillary electrophoresis time-of-flight mass spectrometry (CE-TOF-MS), and nuclear magnetic resonance (NMR) spectroscopy are commonly used to measure and profile small molecule metabolites [55]. In OM research, metabolomics provides valuable insights into the biochemical changes that occur in the oral mucosa as a result of radiation or chemotherapy treatment.
Yatsuoka et al. [56] (2021) analyzed the time course of salivary metabolic profiles using CE-TOF-MS in nine male patients with HNSCC who received radiation therapy of at least 50 Gy. Partial least squares regression-discriminant analyses showed that histidine and tyrosine highly discriminated between high-grade OM and low-grade OM at baseline. Moreover, γ-aminobutyric acid (GABA) and 2-aminobutyric acid (2AB) concentrations were higher in the high-grade OM group than in the low-grade OM group. While GABA is known to be correlated with stress levels, 2AB is known to increase under high oxidative stress conditions, which can be induced by radiation.
The metabolomic changes brought about by maxillofacial and oral massage (MOM) attempting to attenuate severe radiotherapy-induced OM in patients with nasopharyngeal carcinoma were also explored by Yang et al. [57]. They identified enhanced levels of the metabolites 9S-HEPE and 15-HETE among patients receiving MOM after radiotherapy, both of which are known to play a role in inhibiting inflammatory responses through different pathways [58].
Animal OM models have also been useful in understanding the pre-clinical potential of pharmacological targets. Two independent studies involving the use of Chinese herbal medicine, Shuanghua baihe tablet (SBT) and Kouyanqing granules (KQG), have used OM rat models to determine the effects in alleviating OM symptoms and elucidate the potential metabolic pathways involved. Geng et al. (2021) [59] showed the role of SBT in metabolic-related pathways such as linoleic acid and cholic acid metabolism to alleviate the inflammatory symptoms of OM. KQG, on the other hand, shows promising effects by attenuating symptoms of oral ulcers through regulation of the neuroimmunoendocrine system oxidative stress, and tryptophan metabolism [60].

2.4. Proteomics and transcriptomics

Transcriptome analysis typically investigates differential gene expressions that occur in different (e.g., normal vs. abnormal) states. Transcriptomic tools make use of high-throughput technologies such as microarrays, RNA sequencing, and, more recently, single-cell transcriptomics, wherein an individual cell is profiled.
Table 3. Summary of omics approaches and identified putative biomarkers for therapy-induced oral mucositis in head and neck cancer.
Table 3. Summary of omics approaches and identified putative biomarkers for therapy-induced oral mucositis in head and neck cancer.
Year First Author Phenotype Samples Sample size Methods Targets Results
Metabolomics.
Animal Models
2021 Geng [59] Mucositis (0-5) Serum from OM rat model,
induced with 5-FU and 10% acetic acid
30 rats UHPLC Cholic acid, linoleic acid, 4-pyridoxic acid, LysoPC Shuanghua Baihe tablets improve inflammatory symptoms of oral mucositis.
2020 Chen Induced oral ulcers and degree of healing Serum from OM rat model, induced with
15% chloral hydrate
42
rats
LC-QTOF/MS 5-HT, GABA Kouyanqing granules attenuate the symptoms of oral ulcers worsened by sleep deprivation through regulation of the neuroimmunoendocrine system, oxidative stress levels, and tryptophan metabolism.
Clinical Samples
2021 Yatsuoka [56] NCI CTCAE Saliva 9
HNC
CE-TOF-MS tryptophan, D-glucose, D-glutamate, GABA, 2-AB Pre-treatment concentrations of gamma-aminobutyric acid and 2-aminobutyric acids were higher in the high-grade OM group.
2021 Yang [57] NRS 0-10 Peripheral blood 10
NPC
UHPLC-MS/MS 9-HEPE, 15-HETE MOM promotes the release of anti-inflammatory lipids to reduce tissue damage; enhancement of 9S-HEPE and 15-HETE in all radiation doses.
Microbiomics
2020 Reyes-Gibby [7] NCI CTCAE buccal mucosal 66
Locoregional HNSCC
16S rRNA Cardiobacterium, Granulicatella, Prevotella,
Fusobacterium,
Streptococcus , Megasphaera,
Cardiobacterium
Genera abundance was associated with the hazard for the onset of severe OM.
2020 Vesty [61] WHO saliva and oral swabs 19
HNC
NGS Fusobacterium, Haemophilus, Tannerella, Porphyromonas and Eikenella, Candida Gram-negative bacteria on the buccal mucosa may influence susceptibility to developing OM.
2019 Subramaniam and Muthukrishnan [54] WHO unstimulated whole saliva 24
HNSCC
16S rRNA Staphylococcus aureus, Staphylococcus epidermidis, Pseudomonas aeruginosa, Escherichia coli, Klebsiella pneumoniae The bacterial isolates obtained during and at the end of therapy appeared to express a higher level of antibiotic-resistance genes (VIM2, MCR-1, TET[K], blaKPC) than those isolated at the onset of therapy.
2018 Hou [53] RTOG oral swabs 19
NPC
16S rRNA Prevotella, Fusobacterium, Treponema, Porphyromonas Prevotella, Fusobacterium, Treponema and Porphyromonas showed dynamic synchronous variations in abundance throughout the course of radiation therapy, frequently coinciding with the onset of severe mucositis.
2017 Zhu [62] RTOG oral or retropharyngeal mucosa swabs 41
NPC
16S rRNA Firmicutes, Proteobacteria, Bacteroidetes, Fusobacteria, Actinobacteria, Spirochaetes, Cyanobacteria, Verrucomicrobia, Acidobacteria, TM7, Deinococcus-Thermus and SR1 Oral microbiota changes correlate with the progression and aggravation of radiotherapy-induced mucositis in patients with nasopharyngeal carcinoma.
Microbiota
2018 Almstahl [63] WHO Swab culture 33
HNC
Culture Neisseria, Fusobacterium, Prevotella, Candida Levels of Neisseria decreased and mucosal pathogens increased during RT; 2 years post-treatment, Fusobacterium and Prevotella decreased; growth of Candida increased
2018 Gaetti-Jardim [64] NCI CTCAE Supra and subgingival biofilms 28
HNC
Culture Candida, Enterobacteriaceae Candida and family Enterobacteriaceae showed increased prevalence with RT, and were associated with the occurrence of mucositis and xerostomia
Transcriptomics
Animal Models
2021 Geng [59] Mucositis (0-5) Serum from OM rat model,
induced with 5-FU and 10% acetic acid
30
rats
Whole genome sequencing ALOX15, CYP2J2, CYP1A1, ALOX15, GATM, ALAS2, PLA2G5 Shuanghua Baihe tablets improve inflammatory symptoms of oral mucositis.
2021 Saul-McBeth [65] Induced oral ulcers, % damage OM mice model;
Induced with head and neck irradiation
3
mice
RNA Seq IL-17RA IL-17RA provides protection during HNI-induced OM by preventing excess inflammation during ulceration phase of OM.
Clinical Samples
2018 Mlak [66] RTOG/EORTC Plasma 60
HNC
Microarray RRM1 RRM1 gene expression in cfRNA allows for estimating risk of severe OM.
Proteomics
2015 Jehmlich [67] NCI CTC v3 Unstimulated whole saliva 50
HNC
MS RPL18A, C6orf115, PRTN3, RPS20, FGB, ARPC1B, PLBD1, GGH, ANXA6, FGG, ANP32E, CTSG, PTGR1, SERPINA1, MDH2, CORO1A, HSPE1, BAHCC1 CP, MMP9, GCA, PLYRP1, SCGB2A1, GPI, PPIC, QRDL, HIST1H4A, HNRNPA2B1, ATP5B, LTA4H, TIMP1, TKT, RPL10A, AZU1, MMP8, RPLP2, ARPC4, CAT, S100A8, B2M, SERPING1, CYBB, ELANE, C3, CALML5, ITIHRPS15A, ACTR2 48 proteins differed significantly between OM group and non-OM group. 17 proteins displayed increased levels and 31 proteins decreased in level in OM.
NCI CTCAE: National Cancer Institute Common Terminology Criteria for Adverse Events, NRS: numerical rating scale; WHO: World Health Organization; RTOG: Radiation Therapy Oncology Group; EORTC: European Organization for Research and Treatment of Cancer; 5-FU: 5-fluoruracil; HNC: head and neck cancer; NPC: nasopharyngeal cancer; HNSCC: head and neck squamous cell carcinoma; UHPLC: ultra-high-performance liquid chromatography; LC-QTOF/MS: liquid chromatography- quadrupole time-of-flight (TOF)/mass spectrometer (MS); CE-TOF-MS: capillary electrophoresis-TOF-MS; NGS: next generation sequencing; 5-hydroxytryptamine; GABA: γ-aminobutyric acid; 2-AB: 2-aminobutyric acid; 9-HEPE: 9-hydroxyeicosapentanoic acid; 15-HETE: 15-hydroxyeicosatetraenoic acid; RT: radiotherapy; OM: Oral mucositis; MOM: maxillofacial and oral massage.
Transcriptomic data aid in the elucidation of the mechanisms involved in OM. By analyzing patterns of gene expression, dysregulated key pathways and biomarkers can be identified. For example, microRNAs (miRNA) are small non-coding RNAs that play an important role in post-transcriptional gene regulation by binding to specific mRNA molecules and initiating degradation or translational inhibition, thereby affecting gene expression patterns. In the context of OM, the miR-1206 variant has been associated with a 3-fold increase in the risk of developing methotrexate-induced OM [68], whereas miR-200c showed promising results in reducing ROS production and repressing proinflammatory cytokines in animal models [69].
While transcriptomics and proteomics provide different types of data, there is a significant overlap between the two approaches, since changes in gene expression may lead to changes in protein expression, and changes in protein expression can also regulate other downstream gene expression and protein levels. By integrating data from both approaches, we can gain a more complete understanding of the biological processes, networks, and pathways involved in OM and identify targets for the treatment or prevention of therapy-induced OM.
The proteomic approach for OM research has focused on biomarker development, particularly on inflammatory proteins observed after treatment-induced OM. Kiyomi et al. [70] utilized a bead array and an enzyme-linked immunosorbent assay (ELISA) to identify potential biomarkers of OM from saliva or oral swab samples were taken from 20 leukemia or head and neck cancer patients undergoing treatment. Their study suggests that salivary IL-6, IL-10, and TNF-α may serve as predictors of OM occurrence and grade. Additionally, Jehmlich et al. [71] were able to identify 48 (see Table 3) unique proteins that differ significantly between OM and non-OM groups with saliva and/or oral swabs obtained from a pool of 50 head and neck cancer patients. Among those identified is proteinase 3 (PRTN 3), a secretory multifunctional serine protease that can degrade elastin, fibronectin, and collagen. PRTN3 is released upon neutrophil activation and degranulation during tissue injury inflammation. This has also been implicated in the serum proteomic profile of HNSCC patients. While different protein profiles were obtained from the patients due to variations in tumor status and collection time points, most of the 48 proteins are extracellular and have important roles in inflammatory and innate immune responses, and complement activation cascade [71].

3. Conclusions

These different types of “omics” studies provide preliminary evidence that high-throughput methodologies applied to study different aspects of the host (host biomarkers), response to cancer treatment (chemotherapy and/or radiotherapy), and microbiological factors (microbiome and infection) are feasible. There is a paucity of these studies, and there is a complete lack of studies that integrate and analyze multi-omics data to examine the importance of genomic variations, gene-environment interactions, and mechanisms of the host response to a chemical or radiational cytotoxicity to the development of OM. Moreover, the addition of ICI therapy to the mix of antineoplastic treatment for HNSCC adds immune-mediated etiology into the pathogenic mechanism of OM, and this mechanism has not been studied in HNSCC patients.

4. Future Directions

The basic principle of molecular epidemiology [72] is that neither genetics nor environment alone is responsible for individual variation in disease presentation and severity. Whereas traditional field-based epidemiological approaches have identified subgroups at higher risk for OM (older age, body mass index, radiotherapy dose), the development of high throughput molecular laboratory techniques has allowed for the use of biological markers in disease prevention and risk prediction. These biomarkers measure events at the physiological, cellular, and molecular levels, thus improving our understanding of the epidemiology of diseases. Integrating the use of molecular epidemiology methods allows for a better understanding of biological mechanisms and improves assessments of individual risks by providing person-specific information (a genetic profile, etc.) along with clinical information. Such tools could identify subgroups who might benefit most from intervention and contribute to developing personalized and more effective therapies while reducing toxic side effects.
Applying omics approaches may potentially identify subgroups of patients who will benefit from a specific intervention or treatment. However, a common limitation of the studies was the heterogeneity of the population sample, the small sample size, the use of different OM measures, and the retrospective study design.
Challenges include the heterogeneity of the underlying pathogenic mechanisms of OM given that there are now three main types of cancer treatment for HNSCC (chemotherapy, radiotherapy, and ICIs) that can cause OM. Prospective studies that will allow assessment of the timing of OM onset and the response to conventional mucositis treatments are needed to identify pathogenic mechanisms. In addition, the clinical features of OM can help identify the involvement of infection (viral, fungal, or bacterial) or immune reaction, ie. lesion appearance, location, redness, swelling, ulceration, pain severity, etc. The use of statistical approaches such as hierarchical clustering to assess which clinical features of OM will correlate with different multi-omics patterns will provide insight into the pathogenic mechanisms and potential treatment targets.
Importantly, omic studies are resource intensive and therefore, it is important to note that funding announcements by the US National Institute of Dental and Craniofacial Research (NIDCR) and the US National Cancer Institute (NCI) may provide the opportunities to pursue multi-center studies applying omics approaches to OM. In particular, a reissue by NIDCR of PAR-17-154 [73] calls for prospective observational designs and biomarker validation studies. Considered appropriate would be epidemiologic studies of disease prevalence or incidence, cohort studies prospectively ascertaining risk factors for disease development, cohort studies that provide longitudinal follow-up of treatment outcomes, case-control studies with longitudinal follow-up, and large cross-sectional studies or case-control studies evaluating genomic changes, gene-environment interactions, or disease/treatment mechanisms through -omics, cellular, and imaging analyses. For the biomarker validation studies, the reissue of PAR-17-154 will [73] promote advanced analytic and/or clinical validation of strong candidate biomarkers and endpoints for the diagnostic or prognostic utility to demonstrate that biomarker or endpoint change is reliably correlated with pathophysiology, clinical outcome, therapeutic target engagement or treatment response.

Author Contributions

Conceptualization, C.C.R.G.; writing—original draft preparation, C.C.R.G., E.M.D.S.V., and S.J.Y.; writing—review and editing, C.C.R.G., E.M.D.S.V., and S.J.Y.; visualization, E.M.D.S.V.; funding acquisition, C.C.R.G., S.J.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by grants from the National Institutes of Health (NIH) to Drs. Reyes-Gibby (R01CA267856; R21DE026837) and Yeung (R01CA267856).

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Mody, M.D.; Rocco, J.W.; Yom, S.S.; Haddad, R.I.; Saba, N.F. Head and neck cancer. Lancet 2021, 398, 2289–2299. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J Clin 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  3. Cohen, E.E.; LaMonte, S.J.; Erb, N.L.; Beckman, K.L.; Sadeghi, N.; Hutcheson, K.A.; Stubblefield, M.D.; Abbott, D.M.; Fisher, P.S.; Stein, K.D.; et al. American Cancer Society Head and Neck Cancer Survivorship Care Guideline. CA Cancer J Clin 2016, 66, 203–239. [Google Scholar] [CrossRef] [PubMed]
  4. Devices@Fda, A.U. Available online: https://www.accessdata.fda.gov/scripts/cdrh/devicesatfda/index.cfm (accessed on 1 April 2023).
  5. Maria, O.M.; Eliopoulos, N.; Muanza, T. Radiation-Induced Oral Mucositis. Front Oncol 2017, 7, 89. [Google Scholar] [CrossRef] [PubMed]
  6. Pulito, C.; Cristaudo, A.; Porta, C.; Zapperi, S.; Blandino, G.; Morrone, A.; Strano, S. Oral mucositis: the hidden side of cancer therapy. J Exp Clin Cancer Res 2020, 39, 210. [Google Scholar] [CrossRef] [PubMed]
  7. Reyes-Gibby, C.C.; Wang, J.; Zhang, L.; Peterson, C.B.; Do, K.A.; Jenq, R.R.; Shelburne, S.; Shah, D.P.; Chambers, M.S.; Hanna, E.Y.; et al. Oral microbiome and onset of oral mucositis in patients with squamous cell carcinoma of the head and neck. Cancer 2020, 126, 5124–5136. [Google Scholar] [CrossRef]
  8. Sunaga, T.; Nagatani, A.; Fujii, N.; Hashimoto, T.; Watanabe, T.; Sasaki, T. The association between cumulative radiation dose and the incidence of severe oral mucositis in head and neck cancers during radiotherapy. Cancer Rep (Hoboken) 2021, 4, e1317. [Google Scholar] [CrossRef] [PubMed]
  9. de Pauli Paglioni, M.; Faria, K.M.; Palmier, N.R.; Prado-Ribeiro, A.C.; RB, E.D.; da Graça Pinto, H.; Treister, N.S.; Epstein, J.B.; Migliorati, C.A.; Santos-Silva, A.R.; et al. Patterns of oral mucositis in advanced oral squamous cell carcinoma patients managed with prophylactic photobiomodulation therapy-insights for future protocol development. Lasers Med Sci 2021, 36, 429–436. [Google Scholar] [CrossRef]
  10. Chen, S.C.; Lai, Y.H.; Huang, B.S.; Lin, C.Y.; Fan, K.H.; Chang, J.T. Changes and predictors of radiation-induced oral mucositis in patients with oral cavity cancer during active treatment. Eur J Oncol Nurs 2015, 19, 214–219. [Google Scholar] [CrossRef]
  11. Elad, S.; Zadik, Y. Chronic oral mucositis after radiotherapy to the head and neck: a new insight. Support Care Cancer 2016, 24, 4825–4830. [Google Scholar] [CrossRef]
  12. Reyes-Gibby, C.C.; Melkonian, S.C.; Hanna, E.Y.; Yeung, S.J.; Lu, C.; Chambers, M.S.; Banala, S.R.; Gunn, G.B.; Shete, S.S. Cohort study of oncologic emergencies in patients with head and neck cancer. Head Neck 2017, 39, 1195–1204. [Google Scholar] [CrossRef] [PubMed]
  13. Jacob, J.S.; Dutra, B.E.; Garcia-Rodriguez, V.; Panneerselvam, K.; Abraham, F.O.; Zou, F.; Ma, W.; Grivas, P.; Thompson, J.A.; Altan, M.; et al. Clinical Characteristics and Outcomes of Oral Mucositis Associated With Immune Checkpoint Inhibitors in Patients With Cancer. J Natl Compr Canc Netw 2021, 19, 1415–1424. [Google Scholar] [CrossRef] [PubMed]
  14. Elad, S.; Epstein, J.B.; Raber-Durlacher, J.; Donnelly, P.; Strahilevitz, J. The antimicrobial effect of Iseganan HCl oral solution in patients receiving stomatotoxic chemotherapy: analysis from a multicenter, double-blind, placebo-controlled, randomized, phase III clinical trial. J Oral Pathol Med 2012, 41, 229–234. [Google Scholar] [CrossRef] [PubMed]
  15. Xu, Y.; Wen, N.; Sonis, S.T.; Villa, A. Oral side effects of immune checkpoint inhibitor therapy (ICIT): An analysis of 4683 patients receiving ICIT for malignancies at Massachusetts General Hospital, Brigham & Women's Hospital, and the Dana-Farber Cancer Institute, 2011 to 2019. Cancer 2021, 127, 1796–1804. [Google Scholar] [CrossRef] [PubMed]
  16. Davy, C.; Heathcote, S. A systematic review of interventions to mitigate radiotherapy-induced oral mucositis in head and neck cancer patients. Support Care Cancer 2021, 29, 2187–2202. [Google Scholar] [CrossRef] [PubMed]
  17. Alsubaie, H.M.; Alsini, A.Y.; Alsubaie, K.M.; Abu-Zaid, A.; Alzahrani, F.R.; Sayed, S.; Pathak, A.K.; Alqahtani, K.H. Glutamine for prevention and alleviation of radiation-induced oral mucositis in patients with head and neck squamous cell cancer: Systematic review and meta-analysis of controlled trials. Head Neck 2021, 43, 3199–3213. [Google Scholar] [CrossRef]
  18. Al-Rudayni, A.H.M.; Gopinath, D.; Maharajan, M.K.; Veettil, S.K.; Menon, R.K. Efficacy of Oral Cryotherapy in the Prevention of Oral Mucositis Associated with Cancer Chemotherapy: Systematic Review with Meta-Analysis and Trial Sequential Analysis. Curr Oncol 2021, 28, 2852–2867. [Google Scholar] [CrossRef]
  19. Di Fede, O.; Canepa, F.; Maniscalco, L.; Tozzo, P.; Matranga, D.; Giuliana, G. Prevention and the treatment of oral mucositis: the efficacy of sodium bicarbonate vs other agents: a systematic review. BMC Oral Health 2023, 23, 4. [Google Scholar] [CrossRef]
  20. Cronshaw, M.; Parker, S.; Anagnostaki, E.; Mylona, V.; Lynch, E.; Grootveld, M. Photobiomodulation and Oral Mucositis: A Systematic Review. Dent J (Basel) 2020, 8. [Google Scholar] [CrossRef]
  21. Zadik, Y.; Arany, P.R.; Fregnani, E.R.; Bossi, P.; Antunes, H.S.; Bensadoun, R.J.; Gueiros, L.A.; Majorana, A.; Nair, R.G.; Ranna, V.; et al. Systematic review of photobiomodulation for the management of oral mucositis in cancer patients and clinical practice guidelines. Support Care Cancer 2019, 27, 3969–3983. [Google Scholar] [CrossRef]
  22. Hong, C.H.L.; Gueiros, L.A.; Fulton, J.S.; Cheng, K.K.F.; Kandwal, A.; Galiti, D.; Fall-Dickson, J.M.; Johansen, J.; Ameringer, S.; Kataoka, T.; et al. Systematic review of basic oral care for the management of oral mucositis in cancer patients and clinical practice guidelines. Support Care Cancer 2019, 27, 3949–3967. [Google Scholar] [CrossRef] [PubMed]
  23. Elad, S.; Cheng, K.K.F.; Lalla, R.V.; Yarom, N.; Hong, C.; Logan, R.M.; Bowen, J.; Gibson, R.; Saunders, D.P.; Zadik, Y.; et al. MASCC/ISOO clinical practice guidelines for the management of mucositis secondary to cancer therapy. Cancer 2020, 126, 4423–4431. [Google Scholar] [CrossRef] [PubMed]
  24. World Health, O. WHO handbook for reporting results of cancer treatment. 1979.
  25. Cox, J.D.; Stetz, J.; Pajak, T.F. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European Organization for Research and Treatment of Cancer (EORTC). Int J Radiat Oncol Biol Phys 1995, 31, 1341–1346. [Google Scholar] [CrossRef] [PubMed]
  26. Diagnosis, N.C.I.D.D.o.C.T. CTEP Cancer Therapy Evaluation Program. Available online: https://ctep.cancer.gov/protocoldevelopment/electronic_applications/ctc.htm (accessed on 1 April 2023).
  27. Villa, A.; Vollemans, M.; De Moraes, A.; Sonis, S. Concordance of the WHO, RTOG, and CTCAE v4.0 grading scales for the evaluation of oral mucositis associated with chemoradiation therapy for the treatment of oral and oropharyngeal cancers. Support Care Cancer 2021, 29, 6061–6068. [Google Scholar] [CrossRef] [PubMed]
  28. Trotti, A.; Byhardt, R.; Stetz, J.; Gwede, C.; Corn, B.; Fu, K.; Gunderson, L.; McCormick, B.; Morrisintegral, M.; Rich, T.; et al. Common toxicity criteria: version 2.0. an improved reference for grading the acute effects of cancer treatment: impact on radiotherapy. Int J Radiat Oncol Biol Phys 2000, 47, 13–47. [Google Scholar] [CrossRef] [PubMed]
  29. Development of a staging system for chemotherapy-induced stomatitis. Western Consortium for Cancer Nursing Research. Cancer Nurs 1991, 14, 6–12.
  30. Eilers, J.; Berger, A.M.; Petersen, M.C. Development, testing, and application of the oral assessment guide. Oncol Nurs Forum 1988, 15, 325–330. [Google Scholar] [PubMed]
  31. Sonis, S.T.; Eilers, J.P.; Epstein, J.B.; LeVeque, F.G.; Liggett, W.H., Jr.; Mulagha, M.T.; Peterson, D.E.; Rose, A.H.; Schubert, M.M.; Spijkervet, F.K.; et al. Validation of a new scoring system for the assessment of clinical trial research of oral mucositis induced by radiation or chemotherapy. Mucositis Study Group. Cancer 1999, 85, 2103–2113. [Google Scholar] [CrossRef]
  32. Stiff, P.J.; Emmanouilides, C.; Bensinger, W.I.; Gentile, T.; Blazar, B.; Shea, T.C.; Lu, J.; Isitt, J.; Cesano, A.; Spielberger, R. Palifermin reduces patient-reported mouth and throat soreness and improves patient functioning in the hematopoietic stem-cell transplantation setting. J Clin Oncol 2006, 24, 5186–5193. [Google Scholar] [CrossRef]
  33. Stiff, P.J.; Erder, H.; Bensinger, W.I.; Emmanouilides, C.; Gentile, T.; Isitt, J.; Lu, Z.J.; Spielberger, R. Reliability and validity of a patient self-administered daily questionnaire to assess impact of oral mucositis (OM) on pain and daily functioning in patients undergoing autologous hematopoietic stem cell transplantation (HSCT). Bone Marrow Transplant 2006, 37, 393–401. [Google Scholar] [CrossRef]
  34. Slade, G.D. Derivation and validation of a short-form oral health impact profile. Community Dent Oral Epidemiol 1997, 25, 284–290. [Google Scholar] [CrossRef] [PubMed]
  35. Rosenthal, D.I.; Mendoza, T.R.; Chambers, M.S.; Burkett, V.S.; Garden, A.S.; Hessell, A.C.; Lewin, J.S.; Ang, K.K.; Kies, M.S.; Gning, I.; et al. The M. D. Anderson symptom inventory-head and neck module, a patient-reported outcome instrument, accurately predicts the severity of radiation-induced mucositis. Int J Radiat Oncol Biol Phys 2008, 72, 1355–1361. [Google Scholar] [CrossRef] [PubMed]
  36. Kushner, J.A.; Lawrence, H.P.; Shoval, I.; Kiss, T.L.; Devins, G.M.; Lee, L.; Tenenbaum, H.C. Development and validation of a Patient-Reported Oral Mucositis Symptom (PROMS) scale. J Can Dent Assoc 2008, 74, 59. [Google Scholar] [PubMed]
  37. Institute, N.H.G.R. The Human Genome Project. Available online: https://www.genome.gov/human-genome-project (accessed on 1 April 2023).
  38. Bates, S.A. Telomere-to-Telomere (T2T) consortium reasearchers to discuss completion of a human genome sequence. Available online: https://www.genome.gov/news/media-adivsory/telomere-to-telomere-t2t-consortium-researchers-to-discuss-completion-of-human-genome-sequence (accessed on 1 April 2023).
  39. Reyes-Gibby, C.C.; Melkonian, S.C.; Wang, J.; Yu, R.K.; Shelburne, S.A.; Lu, C.; Gunn, G.B.; Chambers, M.S.; Hanna, E.Y.; Yeung, S.J.; et al. Identifying novel genes and biological processes relevant to the development of cancer therapy-induced mucositis: An informative gene network analysis. PLoS ONE 2017, 12, e0180396. [Google Scholar] [CrossRef] [PubMed]
  40. Brzozowska, A.; Homa-Mlak, I.; Mlak, R.; Golebiowski, P.; Mazurek, M.; Ciesielka, M.; Malecka-Massalska, T. Polymorphism of regulatory region of GHRL gene (-2531C>T) as a promising predictive factor for radiotherapy-induced oral mucositis in patients with head neck cancer. Head Neck 2018, 40, 1799–1811. [Google Scholar] [CrossRef] [PubMed]
  41. Brzozowska, A.; Powrozek, T.; Homa-Mlak, I.; Mlak, R.; Ciesielka, M.; Golebiowski, P.; Malecka-Massalska, T. Polymorphism of Promoter Region of TNFRSF1A Gene (-610 T > G) as a Novel Predictive Factor for Radiotherapy Induced Oral Mucositis in HNC Patients. Pathol Oncol Res 2018, 24, 135–143. [Google Scholar] [CrossRef] [PubMed]
  42. Mlak, R.; Powrozek, T.; Brzozowska, A.; Homa-Mlak, I.; Mazurek, M.; Golebiowski, P.; Korzeb, D.; Rahnama-Hezavah, M.; Malecka-Massalska, T. Polymorphism of TNFRSF1 A may act as a predictor of severe radiation-induced oral mucositis and a prognosis factor in patients with head and neck cancer. Oral Surg Oral Med Oral Pathol Oral Radiol 2020, 130, 283–291.e282. [Google Scholar] [CrossRef] [PubMed]
  43. Mlak, R.; Powrozek, T.; Brzozowska, A.; Homa-Mlak, I.; Mazurek, M.; Golebiowski, P.; Sobieszek, G.; Malecka-Massalska, T. The relationship between TNF-alpha gene promoter polymorphism (- 1211 T > C), the plasma concentration of TNF-alpha, and risk of oral mucositis and shortening of overall survival in patients subjected to intensity-modulated radiation therapy due to head and neck cancer. Support Care Cancer 2020, 28, 531–540. [Google Scholar] [CrossRef]
  44. Brzozowska, A.; Mlak, R.; Homa-Mlak, I.; Golebiowski, P.; Mazurek, M.; Ciesielka, M.; Malecka-Massalska, T. Polymorphism of regulatory region of APEH gene (c.-521G>C, rs4855883) as a relevant predictive factor for radiotherapy induced oral mucositis and overall survival in head neck cancer patients. Oncotarget 2018, 9, 29644–29653. [Google Scholar] [CrossRef]
  45. Chen, H.; Wu, M.; Li, G.; Hua, L.; Chen, S.; Huang, H. Association between XRCC1 single-nucleotide polymorphism and acute radiation reaction in patients with nasopharyngeal carcinoma: A cohort study. Medicine (Baltimore) 2017, 96, e8202. [Google Scholar] [CrossRef]
  46. Schack, L.M.H.; Naderi, E.; Fachal, L.; Dorling, L.; Luccarini, C.; Dunning, A.M.; Head; Neck Group of the Radiogenomics, C.; Danish, H.; Neck Cancer, G.; et al. A genome-wide association study of radiotherapy induced toxicity in head and neck cancer patients identifies a susceptibility locus associated with mucositis. Br J Cancer 2022, 126, 1082–1090. [CrossRef]
  47. Yang, D.W.; Wang, T.M.; Zhang, J.B.; Li, X.Z.; He, Y.Q.; Xiao, R.; Xue, W.Q.; Zheng, X.H.; Zhang, P.F.; Zhang, S.D.; et al. Genome-wide association study identifies genetic susceptibility loci and pathways of radiation-induced acute oral mucositis. J 2020, 18, 224. [Google Scholar] [CrossRef] [PubMed]
  48. Zhang, R.; Kang, R.; Tang, D. The STING1 network regulates autophagy and cell death. Signal Transduct Target Ther 2021, 6, 208. [Google Scholar] [CrossRef] [PubMed]
  49. Patel, S.; Jin, L. TMEM173 variants and potential importance to human biology and disease. Genes Immun 2019, 20, 82–89. [Google Scholar] [CrossRef] [PubMed]
  50. Motwani, M.; Pesiridis, S.; Fitzgerald, K.A. DNA sensing by the cGAS-STING pathway in health and disease. Nat Rev Genet 2019, 20, 657–674. [Google Scholar] [CrossRef] [PubMed]
  51. Barber, G.N. STING: infection, inflammation and cancer. Nat Rev Immunol 2015, 15, 760–770. [Google Scholar] [CrossRef] [PubMed]
  52. Dregalla, R.C.; Zhou, J.; Idate, R.R.; Battaglia, C.L.; Liber, H.L.; Bailey, S.M. Regulatory roles of tankyrase 1 at telomeres and in DNA repair: suppression of T-SCE and stabilization of DNA-PKcs. Aging (Albany NY) 2010, 2, 691–708. [Google Scholar] [CrossRef]
  53. Hou, J.; Zheng, H.; Li, P.; Liu, H.; Zhou, H.; Yang, X. Distinct shifts in the oral microbiota are associated with the progression and aggravation of mucositis during radiotherapy. Radiother Oncol 2018, 129, 44–51. [Google Scholar] [CrossRef]
  54. Subramaniam, N.; Muthukrishnan, A. Oral mucositis and microbial colonization in oral cancer patients undergoing radiotherapy and chemotherapy: A prospective analysis in a tertiary care dental hospital. J 2019, 10, e12454. [Google Scholar] [CrossRef]
  55. Segers, K.; Declerck, S.; Mangelings, D.; Heyden, Y.V.; Eeckhaut, A.V. Analytical techniques for metabolomic studies: a review. Bioanalysis 2019, 11, 2297–2318. [Google Scholar] [CrossRef]
  56. Yatsuoka, W.; Ueno, T.; Miyano, K.; Enomoto, A.; Ota, S.; Sugimoto, M.; Uezono, Y. Time-Course of Salivary Metabolomic Profiles during Radiation Therapy for Head and Neck Cancer. J 2021, 10, 15. [Google Scholar] [CrossRef] [PubMed]
  57. Yang, G.; Feng, D.; Li, F.; Luo, B.; Zhu, J.; Yang, Q.; Zheng, L.; Dong, Q.; Chen, M.; Xu, Z.; et al. A randomized, controlled phase II trial of maxillofacial and oral massage in attenuating severe radiotherapy-induced oral mucositis and lipid metabolite changes in nasopharyngeal carcinoma. Radiother Oncol 2021, 163, 76–82. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, C.; Liu, W.; Yao, L.; Zhang, X.; Zhang, X.; Ye, C.; Jiang, H.; He, J.; Zhu, Y.; Ai, D. Hydroxyeicosapentaenoic acids and epoxyeicosatetraenoic acids attenuate early occurrence of nonalcoholic fatty liver disease. Br J Pharmacol 2017, 174, 2358–2372. [Google Scholar] [CrossRef] [PubMed]
  59. Geng, Q.S.; Liu, R.J.; Shen, Z.B.; Wei, Q.; Zheng, Y.Y.; Jia, L.Q.; Wang, L.H.; Li, L.F.; Li, J.; Xue, W.H. Transcriptome sequencing and metabolome analysis reveal the mechanism of Shuanghua Baihe Tablet in the treatment of oral mucositis. Chin J Nat Med 2021, 19, 930–943. [Google Scholar] [CrossRef] [PubMed]
  60. Chen, W.; Li, C.; Jin, D.; Shi, Y.; Zhang, M.; Bo, M.; Qian, D.; Wang, M.; Li, G. Metabolomics Combined with Network Pharmacology-Based Strategy to Reveal the Underlying Mechanism of Zhenhuang Submicron Emulsion in Treating Oropharyngeal Mucositis Complications of Radiation Therapy for Head and Neck Cancer. Drug Des Devel Ther 2022, 16, 3169–3182. [Google Scholar] [CrossRef] [PubMed]
  61. Vesty, A.; Gear, K.; Biswas, K.; Mackenzie, B.W.; Taylor, M.W.; Douglas, R.G. Oral microbial influences on oral mucositis during radiotherapy treatment of head and neck cancer. Support Care Cancer 2020, 28, 2683–2691. [Google Scholar] [CrossRef] [PubMed]
  62. Zhu, X.X.; Yang, X.J.; Chao, Y.L.; Zheng, H.M.; Sheng, H.F.; Liu, H.Y.; He, Y.; Zhou, H.W. The Potential Effect of Oral Microbiota in the Prediction of Mucositis During Radiotherapy for Nasopharyngeal Carcinoma. EBioMedicine 2017, 18, 23–31. [Google Scholar] [CrossRef] [PubMed]
  63. Almstahl, A.; Finizia, C.; Carlen, A.; Fagerberg-Mohlin, B.; Alstad, T. Mucosal microflora in head and neck cancer patients. Int 2018, 16, 459–466. [Google Scholar] [CrossRef]
  64. Gaetti-Jardim, E., Jr.; Jardim, E.C.G.; Schweitzer, C.M.; da Silva, J.C.L.; Oliveira, M.M.; Masocatto, D.C.; Dos Santos, C.M. Supragingival and subgingival microbiota from patients with poor oral hygiene submitted to radiotherapy for head and neck cancer treatment. Arch Oral Biol 2018, 90, 45–52. [Google Scholar] [CrossRef]
  65. Saul-McBeth, J.; Dillon, J.; Lee, A.; Launder, D.; Kratch, J.M.; Abutaha, E.; Williamson, A.A.; Schroering, A.G.; Michalski, G.; Biswas, P.; et al. Tissue Damage in Radiation-Induced Oral Mucositis Is Mitigated by IL-17 Receptor Signaling. Front 2021, 12, 687627. [Google Scholar] [CrossRef]
  66. Mlak, R.; Powrozek, T.; Brzozowska, A.; Homa-Mlak, I.; Mazurek, M.; Malecka-Massalska, T. RRM1 gene expression evaluated in the liquid biopsy (blood cfRNA) as a non-invasive, predictive factor for radiotherapy-induced oral mucositis and potential prognostic biomarker in head and neck cancer patients. Cancer Biomark 2018, 22, 657–667. [Google Scholar] [CrossRef] [PubMed]
  67. Jehmlich, N.; Stegmaier, P.; Golatowski, C.; Salazar, M.G.; Rischke, C.; Henke, M.; Volker, U. Differences in the whole saliva baseline proteome profile associated with development of oral mucositis in head and neck cancer patients undergoing radiotherapy. J Proteomics 2015, 125, 98–103. [Google Scholar] [CrossRef] [PubMed]
  68. Gutierrez-Camino, A.; Oosterom, N.; den Hoed, M.A.H.; Lopez-Lopez, E.; Martin-Guerrero, I.; Pluijm, S.M.F.; Pieters, R.; de Jonge, R.; Tissing, W.J.E.; Heil, S.G.; et al. The miR-1206 microRNA variant is associated with methotrexate-induced oral mucositis in pediatric acute lymphoblastic leukemia. Pharmacogenet Genomics 2017, 27, 303–306. [Google Scholar] [CrossRef] [PubMed]
  69. Tao, J.; Fan, M.; Zhou, D.; Hong, Y.; Zhang, J.; Liu, H.; Sharma, S.; Wang, G.; Dong, Q. miR-200c Modulates the Pathogenesis of Radiation-Induced Oral Mucositis. Oxid Med Cell Longev 2019, 2019, 2352079. [Google Scholar] [CrossRef] [PubMed]
  70. Kiyomi, A.; Yoshida, K.; Arai, C.; Usuki, R.; Yamazaki, K.; Hoshino, N.; Kurokawa, A.; Imai, S.; Suzuki, N.; Toyama, A.; et al. Salivary inflammatory mediators as biomarkers for oral mucositis and oral mucosal dryness in cancer patients: A pilot study. PLoS ONE 2022, 17, e0267092. [Google Scholar] [CrossRef]
  71. Jehmlich, N.; Stegmaier, P.; Golatowski, C.; Salazar, M.G.; Rischke, C.; Henke, M.; Volker, U. Proteome data of whole saliva which are associated with development of oral mucositis in head and neck cancer patients undergoing radiotherapy. Data Brief 2016, 8, 501–505. [Google Scholar] [CrossRef]
  72. Reyes-Gibby, C.C.; Wu, X.; Spitz, M.; Kurzrock, R.; Fisch, M.; Bruera, E.; Shete, S. Molecular epidemiology, cancer-related symptoms, and cytokines pathway. Lancet Oncol 2008, 9, 777–785. [Google Scholar] [CrossRef]
  73. Health, N.I.O. NIDCR Prospective Observational or Biomarker Validation Study Cooperative Agreement. Available online: https://grants.nih.gov/grants/guide/pa-files/PAR-20-060.html (accessed on 13 April 2023).
Figure 1. Oral mucositis phenotype is heterogenous and known to vary by epidemiological clinical, and behavioral factors. Word cloud showing frequency of appearance of terms related to oral mucositis based on the abstract of publications for oral mucositis.
Figure 1. Oral mucositis phenotype is heterogenous and known to vary by epidemiological clinical, and behavioral factors. Word cloud showing frequency of appearance of terms related to oral mucositis based on the abstract of publications for oral mucositis.
Preprints 71569 g001
Figure 2. Putative biomarkers identified using the different omics approaches.
Figure 2. Putative biomarkers identified using the different omics approaches.
Preprints 71569 g002
Table 1. List of clinician-rated and patient-rated OM scales.
Table 1. List of clinician-rated and patient-rated OM scales.
Scale Items
World Health Organization* [24] 0=No changes; 1= Soreness/erythema; 2= Soreness/erythema; ulceration ability to eat solid food; 3= Soreness/erythema + ulceration + ability to use a liquid diet only; 4= Soreness/erythema + ulceration + no possible oral alimentation
Radiation Therapy Oncology Group [25,28] Grade I: Erythema; Grade II: Patchy reaction (<1.5 cm, non-contiguous); Grade III: Confluent mucositis (>1.5 cm, contiguous); Grade IV: Ulceration, necrosis, bleeding.
National Cancer Institute [26] Grade 0 (None) None; Grade 1 (Mild) Painless ulcers, erythema, or mild soreness in the absence of lesions; Grade 2 (Moderate) Painful erythema, oedema, or ulcers but eating or swallowing possible; Grade 3 (Severe) Painful erythema, oedema, or ulcers requiring IV hydration; Grade 4 (Life-threatening) Severe ulceration or requiring parenteral or enteral nutritional support or prophylactic intubation; Grade 5 (Death) Death related to toxicity
Western Consortium for Cancer Nursing Research [29] Lesions: none; Color: pink; Bleeding: none; 1= Lesions: 1–4; Color: slight red; Bleeding: N/A; 2= Lesions: >4; Color: moderate red; Bleeding: with eating and oral hygiene; 3= Lesions: coalescing; Color: very red; Bleeding: spontaneous;
Oral Assessment Guide [30] Eight categories of oral health (lips, tongue, gums and tissues, saliva, natural teeth, dentures, oral cleanliness and dental pain) are assessed as healthy, changes or unhealthy.
Oral Mucositis Assessment Scale (OMAS) [31] Graded 0–3 for ulceration and 0–2 for erythema at nine sites within the oral cavity.
The Oral Mucositis Weekly Questionnaire 8 items; Likert scale: Soreness/Pain, Activity limitation, Quality of Life
Oral mucositis daily questionnaire [32,33] 10 items; Likert scale: Overall health, Severity , Functional limitations
Oral Health Impact Profile-14 (OHIP-14) [34] 14 items; Likert scale : Functional limitation, Physical pain, Psychological discomfort, Psychological disability, Physical disability, Social disability, Handicap
MD Anderson Symptom Inventory- Head and Neck (MDASI-HN) [35] 28 items: 0–10 scale
Along with the core MDASI’s 13 symptom items and 6 interference items, the MDASI-HN also assesses 9 symptoms relevant to head and neck cancer
Patient-Reported Oral Mucositis Symptoms (PROMS) [36] 10 items: Visual analogue scale : Pain, Oral functions
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.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

© 2024 MDPI (Basel, Switzerland) unless otherwise stated