Skip to content
BY 4.0 license Open Access Published by De Gruyter October 27, 2022

Saliva – a new opportunity for fluid biopsy

  • Zhijie Huang ORCID logo , Xiaoxia Yang , Yisheng Huang , Zhengming Tang , Yuanxin Chen , Hongyu Liu , Mingshu Huang , Ling Qing , Li Li , Qin Wang , Zhuye Jie , Xin Jin EMAIL logo and Bo Jia EMAIL logo

Abstract

Saliva is a complex biological fluid with a variety of biomolecules, such as DNA, RNA, proteins, metabolites and microbiota, which can be used for the screening and diagnosis of many diseases. In addition, saliva has the characteristics of simple collection, non-invasive and convenient storage, which gives it the potential to replace blood as a new main body of fluid biopsy, and it is an excellent biological diagnostic fluid. This review integrates recent studies and summarizes the research contents of salivaomics and the research progress of saliva in early diagnosis of oral and systemic diseases. This review aims to explore the value and prospect of saliva diagnosis in clinical application.

Introduction

Traditionally, doctors usually need to issue a large number of tests, such as blood, urine and fecal examinations, to assist in the diagnosing and determining the type of disease. For some more complex diseases, the diagnosis needs to be identified by pathologists through the diseased tissues obtained by surgery, puncture or extraction, which is also the gold standard for diagnosing most tumor diseases [1, 2]. However, tissue biopsy also has many disadvantages. First of all, it needs to cut or extract part of the tissue from the patient, which will cause some trauma to the patient. In addition, it may be necessary for some diseases to dynamically detect the progress and evolution, while repeated tissue biopsies are unrealistic. Finally, the differential diagnosis of pathological sections is a great challenge to the ability of pathologists, which usually requires numerous and rigorous training to achieve a higher level [3], [4], [5] (Tables 13).

Table 1:

Part of salivary biomarkers for oral disease.

Disease Methodology Biomarkers Sensitivity, % Specificity, % AUC Ref
Periodontitis Genome Global 5-methylcytosine methylation 1.00 [6]
Transcriptome miRNA-155 86.7 78.6 0.861 [7]
hsa-miR-125a-3p 1.00 [8]
Proteome MMP-9, S100A8 85.0 76.0 0.860 [9]
IL-1β, MMP-8, ICTP 90.3 84.0 0.929 [10]
IL-1β, IL-1ra, MMP-9 73.3 88.9 0.853 [11]
Del-1, IL-17, LFA-1 0.893 [12]
MUC4, MMP-7 0.931 [13]
C3 0.910 [14]
C3c 0.840
IL-1β 78.0 100.0 0.960 [15]
PGE2 78.0 91.0 0.920
IL-6 78.0 75.9 0.830 [16]
MMP-8 65.3 66.7 0.728
MIP-1α 66.3 67.6 0.723
ARPC5, CLUS 0.970 [17]
DMTB1, RAP1A 0.950
DMTB1, MMP-9 0.940
Metabolome Cadaverine, 5-oxoproline, histidine 0.881 [18]
Microbiome Tannerella forsythia, Eikenella corrodens, Porphyromonas gingivalis, Peptostreptococcus anaerobius, Treponema denticola 93.0 79.0 0.940 [19]
Prevotella intermedia, Catonella morbi, Porphyromonas pasteri, Prevotella nanceiensis, Haemophilus parainfluenzae 0.973 [20]
Sjögren’s syndrome Transcriptome miR-17-5p, let-7i-5p 0.969 [21]
MNDA, FCGR3BL, GBP-2 96.0 73.0 0.860 [22]
Proteome Neutrophil elastase, calreticulin, tripartite Motif-containing protein 29 0.974 [23]
α-Enolase, β2m, cathepsin D 92.0 88.0 0.940 [22]
Soluble siglec-5 69.0 70.0 0.774 [24]
TRIM29 0.880 [25]
Oral lichen planus Transcriptome miR-27b, miR-21, miR-125a [26, 27]
Proteome IFN-γ, IL-33, IL-4, CPR, soluble CD14, soluble TLR2 [2831]
Oral leukoplakia Proteome CD44 91.6 72.7 0.712 [32]
S100A7 81.8 72.7 0.744
S100P 81.8 54.5 0.760
IL-6 [33]
OSCC Transcriptome miR-106b-5p, miR-423-5p, miR-193b-3p 97.4 94.2 0.980 [34]
MAOB, NAB2 92.0 86.0 0.910 [35]
IL-1B, OAZ1, SAT, IL-8 91.0 91.0 0.950 [36]
CCL20 100.0 98.0 0.892 [37]
miR-30c-5p 86.0 74.0 0.820 [38]
miR-24-3p 64.4 80.0 0.738 [39]
miR-512-3p 0.847 [40]
miR-412-3p 0.871
miR-27b 85.7 100.0 0.964 [41]
miR-136 88.8 100.0 0.968
IL-6 94.5 81.9 0.937 [42]
Proteome Cathepsin V, kallikrein5、ADAM9 90.0 99.1 0.938 [43]
MMP-1, cathepsin V, kallikrein 5, ADAM9 85.0 93.3 0.963
AHSG, KRT6C, KLK1, AZGP1 78.0 73.5 0.824 [44]
KLK5, uPA 94.2 89.5 0.950 [45]
PRDX-2, ZAG 100.0 98.77 0.999 [46]
IL-6, IL-8, TNF-α, MCP-1, HCC-1, PF-4 95.4 88.0 0.980 [47]
M2BP, MRP14, CD59, catalase, profilin 90.0 83.0 0.930 [48]
IL8, IL1β 0.817 [49]
SLC3A2, S100A2, IL1RN 83.3 83.3 0.890 [50]
MMP1, KNG1, ANXA2, HSPA5 87.5 80.5 0.910 [51]
CFAH, AFAM, GELS, SAMP, VTDB 93.4 96.5 0.979 [52]
Naa10p, CEA 92.5 85.0 0.944 [53]
Transferrin 0.950 [54]
Chemerin 100.0 100.0 1.000 [55]
MMP-9 100.0 100.0 1.000
CFH 0.661 [56]
FGA 0.740
SERPINA1 0.740
MMP-1 78.0 88.1 0.898 [57]
THBS2 55.1 89.4 0.756 [58]
Metabolome Ornithine, o-hydroxybenzoate, R5F 0.869 [59]
627 common peaks 0.991 [60]
Indole-3-acetate, ethanolamine phosphate 0.856 [61]
Choline, betaine, pipecolinic acid, L-carnitine 100.0 96.7 0.997 [62]
L-phenylalanine, L-leucine 92.3 91.7 0.871 [63]
Propionylcholine, acetylphenylalanine, sphinganine, phytosphingosine, S-carboxymethyl-L-cysteine 100.0 96.7 0.997 [64]
Microbiome Actinobacteria, Fusobacterium, Moraxella, Bacillus, Veillonella 100.0 0.957 [65]
Table 2:

Part of salivary biomarkers for malignant disease.

Disease Methodology Biomarkers Sensitivity, % Specificity, % AUC Ref
Pancreatic cancer Transcriptome miR-3679-5p, miR-940 70.0 70.0 0.763 [66]
KRAS, MBD3L2, ACRV1, DPM1 90.0 95.0 0.971 [67]
has-miR-21 100.0 71.0 [68]
has-miR-23a 100.0 86.0
has-miR-23b 100.0 86.0
has-miR-29c 100.0 57.0
Proteome Cytokeratin-14, lactoperoxidase, cytokeratin-16, cytokeratin-17, peptidyl-prolylcis–transisomerase B 90.0 90.0 0.910 [69]
Metabolome Alanine, N1-cetylspermidine, 2-oxobutyrate, 2-hydroxybutyrate 0.887 [70]
Leucine, isoleucine, valine, tryptophan, glutamic Acid, phenylalanine, glutamine, aspartic acid 0.930 [71]
Microbiome Neisseria elongate, Streptococcus mitis 96.4 82.1 0.895 [72]
Gastric cancer Transcriptome SPINK7, PPL, SEMA4B, miR-140-5p, miR-301a 75.0 83.0 0.810 [73]
Proteome CSTB, TPI1, DMBT1 85.0 80.0 0.930 [74]
Breast cancer Transcriptome, Proteome CSTA, TPT1, IGF2BP1, GRM1, GRIK1, H6PD, MDM4, S100A8, CA6 83.0 97.0 0.920 [75]
Proteome CA125 62.5 59.4 0.686 [76]
sFAS 72.5 61.1 0.676
BS-I, NPA 73.3 74.2 0.755 [77]
VEGF, EGF 83.0 74.0 0.840 [78]
15 free amino acid profile 88.2 85.7 0.916 [79]
Sialic acid 80.0 93.0 0.950 [80]
Metabolome PG14:2 65.2 77.1 0.732 [81]
Spermine 0.766 [82]
LysoPC(18:1) 77.8 100.0 0.920 [83]
LysoPC(22:6) 81.5 91.7 0.920
MG(0:0/14:0/0:0) 92.6 91.7 0.929
Lung cancer Genome EGFR 19-del 0.940 [84]
EGFR 21-L858R 0.960
Transcriptome CCNI, EGFR, FGF19, FRS2, GREB1 93.7 82.8 0.925 [85]
Proteome Calprotectin, zinc α2-glycoprotein, haptoglobin hp2 88.5 92.3 0.900 [86]
IL-10, CXCL10 60.6 80.8 0.701 [87]
Metabolome Catalase, triene conjugates, schiff bases, pH, sialic acids, alkaline phosphatase, chlorides 69.5 87.5 0.803 [88]
Microbiome Capnocytophaga, Veillonella 84.6/78.6 86.7/80.0 0.860/0.800 [89]
Veillonella, Streptococcus [90]
Table 3:

Part of salivary biomarkers for systemic disease.

Disease Methodology Biomarkers Sensitivity, % Specificity, % AUC Ref
Acute myocardial infarction Proteome CPR [91]
CK-MB [92]
Tn1 [93]
Diabetes Proteome S100A9, PRP-1/PRP3 [94]
Metabolome 1,5-AG0 63.5 60.6 0.657 [95]
1,5-AG120 62.2 60.4 0.660
Rheumatoid arthritis Proteome Calgranulin A, calgranulin B, apolipoprotein A-1, 6-phosphogluconate dehydrogenase, peroxiredoxin 5, epidermal fatty acid-binding protein, GRP78/BiP, 14-3-3 proteins [96]
Alzheimer’s disease Proteome Lactoferrin, amyloid-β 42, P-tau [9799]
Microbiome Porphyromonas gingivalis, Veillonella parvula [100]
Parkinson’s disease Transcriptome miR-153 81.8 71.4 0.790 [101]
miR-223 72.7 71.4 0.770
Proteome α-Synuclein, DJ-1 [102]
Heme oxygenase-1 75.0 70.0 0.760 [103]
Stress Proteome α-Amylase, cortisol [104]
Depression Proteome Cortisol, s-IgA [105, 106]
Anxiety Proteome Cortisol, s-IgA [106, 107]

Fluid biopsy detects and analyses various biological body fluids instead of tissue biopsy. Currently, most of the fluid biopsies take blood as the main test object, and most diseases tested are tumors. High-throughput technology is used to detect circulating tumor cells, circulating free DNA, circulating tumor DNA and exosomes in peripheral blood, and to evaluate genomics, transcriptome and proteomics to provide an auxiliary diagnosis for diseases [108, 109]. However, blood samples are usually collected by venipuncture or other invasive methods, which will cause pain and inconvenience to patients [110]. Meanwhile, the operation difficulty and cost of the blood testing platform are on the high side. The storage and transportation of blood samples also need considerable conditions, limiting the promotion of blood-based fluid biopsy [111, 112]. Some studies have shown that other body fluids, such as saliva, cerebrospinal fluid, and pleural fluid, may contain more biological information than blood and contain many reliable markers, which can be used to detect various diseases [113, 114]. It suggests that non-blood derived fluid biopsy has considerable application value, broadening the concept of fluid biopsy. In the non-blood derived body fluids, saliva has the advantages of non-invasive and cost-effective, but also contains a wide range of proteins, DNA, RNA, various metabolites and microflora, which can be used as molecular biomarkers for early detection, monitoring of diseases, and guiding individual and precision therapy [115, 116].

In this article, we review the research related with salivary diagnostics, including the content of salivaomics, the techniques and processes related to saliva detection samples, and the studies of saliva detection in various diseases (Figures 1 and 2).

Figure 1: 
The source and composition of saliva. Saliva is mainly secreted by salivary glands. Blood, oral mucosa cells, gingival crevicular fluid and food are also the importance source of saliva, providing rich informative biomolecular. Saliva is a kind of biological fluid containing DNA, RNA, proteins, inorganic substance, and metabolite.
Figure 1:

The source and composition of saliva. Saliva is mainly secreted by salivary glands. Blood, oral mucosa cells, gingival crevicular fluid and food are also the importance source of saliva, providing rich informative biomolecular. Saliva is a kind of biological fluid containing DNA, RNA, proteins, inorganic substance, and metabolite.

Figure 2: 
Process and application of salivary diagnosis.
Figure 2:

Process and application of salivary diagnosis.

Saliva

Saliva is mainly secreted by three major glands in the oral cavity (parotid gland, submandibular gland and sublingual gland), and the minor salivary glands distributed in the oral mucosa also secreted. Additionally, gingival crevicular fluid, oral mucosal exudate, leukocytes, epithelial cells and many microorganisms are involved in the composition of saliva [110, 117]. The acini of the salivary glands are highly permeable and surrounded by rich capillaries. Small molecules in the blood circulation can infiltrate into the acini and eventually become a part of saliva. Therefore, the electrolyte composition of saliva is similar to that of plasma ultrafiltrate [118]. Saliva is a foamy, slightly cloudy, milky hypertonic solution, slightly acidic (pH=6.6–7.1), odorless, with a specific gravity of 1.004–1.009, 94–99% composed of water, and also contains a small portion of organic compound (such as mucin, globulin, salivary amylase, uric acid, lysozyme, lactic acid, lactoferrin, cortisol and cytokines) and some inorganic compound (such as sodium, potassium, calcium, chloride, thiocyanate, bicarbonate and phosphate) [110]. The saliva secreted by the gland is different from each other. The parotid gland secretes serous saliva, while the sublingual gland produces mucinous saliva. The submandibular gland is a mixed gland, but mainly secretes mucinous saliva. Healthy adults can produce 1.0–1.5 L saliva per day, with an average flow rate of 0.3–0.5 mL per min [119]. Saliva secretion is affected by many factors. Smell, taste, age, mental state, oral hygiene, drugs and body exercise can stimulate saliva secretion [120]. Saliva has many functions: 1. Saliva can wash away food residues and microbes in the mouth, thus helping to clean the mouth; 2. Various enzymes in saliva can decompose carbohydrates and lipids, thus promoting the digestive process; 3. In saliva, lysozyme, immunoglobulin and thiocyanate have antibacterial and bactericidal effects, which are important components of human non-specific immune system; 4. The inorganic compound in saliva play an essential role in preventing enamel demineralization and promoting remineralization, as same as maintaining the dynamic balance of enamel [121].

Saliva, as a part of the endocrine system, is closely related to the blood, and most of the components are permeated from the blood. Components in the blood can enter saliva by diffusion, active transport or extracellular ultrafiltration [122124]. Small molecules can be passively diffused from the blood to acinar cells, which are affected by the size and charge of these molecules. For example, steroids are made up of fatty acids and have a smaller volume, making it easier for them to spread to the acini [125]. On the other hand, proteins in the blood can be actively transported by ligand-receptor binding. For instance, IgA secreted by B cells can be released into saliva by binding to receptors on the acini [126127]. In addition, biomolecules in the blood can enter saliva by ultrafiltration. Steroids can migrate through the gap between acini and ductal cells, and molecules smaller than 1900 Da (such as water, catecholamines, ions) can be transferred through gap junctions between secretory units [128]. Some studies have shown that disease-related biomolecules in the blood can eventually enter saliva [110, 129, 130]. Therefore, saliva, like blood, can be used as a "mirror" to reflect the physiological and pathological state of the body. Moreover, unlike blood, saliva collection is a non-invasive operation, its transportation and preservation are also quite convenient, and the risk of cross-contamination is relatively low. All these are the advantage of saliva as a diagnostic fluid. However, compared with blood, the content of biomolecules in saliva is lower, and the microbial environment in saliva is more complex, which will have an unpredictable impact on the detection results. Nevertheless, the construction and development of a highly sensitive detection platform will gradually solve these problems, and saliva testing may also become a new opportunity for disease screening, detection and surveillance [131].

Salivaomics

Saliva contains various biological components, including DNA, RNA, proteins, microorganisms and metabolites, which are potential biomarkers. Biomarkers can reflect the physiological and pathological state of the body, which is an important basis of personalized medicine. Therefore, a comprehensive analysis and identification of various components in human saliva will greatly help us to develop biomarkers related to human health and disease status, early identification of diseases, assessment of disease prognosis and risk, and monitoring the effect of treatment [110]. Salivaomics is a broad collection of groups used to explore different types of components in saliva and their significance in health and disease states, which include genomics, epigenomics, transcriptomics, proteomics, metabolomics and microbiomics [132]. The research objects of genomics and epigenomics are the biochemical characteristics of DNA, genes and their methylation modification. Transcriptomics investigate RNA in cells and organs, including mRNA and non-coding RNA. Proteomics is the methods to detect the protein profile of the target. Metabolomics focuses on the overall metabolic profile of the samples. Microbiomics study the diversity of microbial flora and its relationship with the body during the development of diseases. Salivaomics will provide us with a complete and profound saliva profile, so we can better discover and develop more accurate biomarkers and contribute to precision medicine.

The genome and epigenome of saliva

The saliva genome contains human and microbial DNA, about 70% of which comes from humans, while the rest from microbiota in the mouth [133]. DNA in the saliva is of good quality and can be preserved for a long time with a low degradation rate. Abraham et al. [134] evaluated the quality of DNA in blood and saliva by measuring absorbance ratios at 260 and 280 nm (A260/A280). The results show that the average value in the saliva is 1.56, and blood is 1.71, indicating the DNA quality of saliva is comparable to the blood. In quantity, the total amount of DNA in the saliva is about 12 μg/mL, ranging from 0.1–26 μg/mL, nearly half of the total DNA in blood (6–73 μg/mL, with an average of about 26 μg/mL). The quantity of DNA in saliva can meet the requirements of chip genotyping but also be used for sequencing arrays and polymerase chain reaction analysis. Epigenetics means that when the DNA sequence of the gene does not change, the gene function changes heritable, and finally leads to the phenotypic change. DNA methylation is the covalent binding of a methyl group on the cytosine base in the genomic CpG island under the action of DNA methyltransferase. It is a common epigenetic process and the most well-studied epigenetic modification [135]. DNA methylation can cause changes in chromatin structure, DNA conformation, DNA stability and the interaction between DNA and protein, thus controlling gene expression, which is necessary to maintain cell growth and metabolism [136, 137]. With the passage of time, or environmental exposure, aberrant methylation of genes may appear, leading to the occurrence and development of the disease, especially in the tumor oncogenesis [138].

Recently, the detection and analysis techniques of saliva genome and epigenome are various and more accurate, such as DNA sequencing, methylation array, and quantitative PCR genotyping, showing excellent effects in detecting many diseases. Carvalho et al. [139] used the quantitative methylation-specific PCR to measure the tumor suppressor gene promoter in the saliva of patients with head and neck squamous cell carcinoma (HNSCC) before treatment. It showed that about 54% of patients occur methylation of more than one selected gene in saliva DNA. In addition, patients with hypermethylation in saliva DNA presented lower local disease control and overall survival. Schussel et al. [140] analyzed the methylation status of 9 genes related to HNSCC in saliva of 191 patients by quantitative methylation-specific PCR, compared with the risk assessment made by expert clinicians. It showed that the methylation of endothelin receptor type B (ENDRB) and deleted in colorectal cancer (DCC) in saliva was associated with premalignant or malignant oral cancer lessions. The sensitivity, specificity and area under the curve (AUC) of the ENDRB and DCC combined diagnosis were 46%, 72% and 0.60, respectively, while the clinical risk classification (CRC) evaluated by expert clinicians were 56%, 66%, and 0.61. Furthermore, the optimal AUC (0.67) occurred in the combination of ENDRB, DCC and CRC. The results suggested that the methylation levels of ENDRB and DCC in the saliva is comparable to the CRC of expert clinician, which is of considerable guiding significance. Obviously, the genome and epigenome of saliva can provide many vital information for diagnosis of disease. However, it cannot afford information about the up-regulation or down-regulation of gene expression, nor can it reflect the expression or deletion of specific genes and gene mutations.

The transcriptome of saliva

The mRNAs and non-coding RNAs (such as microRNAs, piRNAs and circular RNAs) produced by cells consist of the transcriptome of saliva and flow into the oral cavity from various sources, such as exfoliated oral epithelial cells, gingival crevicular fluid and salivary glands [129]. Abnormal expression of specific mRNAs or non-coding RNAs can be detected in many diseases, indicating that transcriptome analysis has a certain value in disease diagnosis.

Microarray analysis and high-throughput sequence analysis are the main technologies for studying the transcriptome. Li et al. [141] first discovered the human transcriptome of saliva through the microarray technology. They reported the salivary mRNA biomarkers detected in the malignant tumors and systemic disease, indicating high-throughput technology is promising in analysing salivary transcriptome. Gomes et al. [142] reported that the mRNA expression of interferon-γ (IFN-γ) was correlated with the severity of periodontitis in saliva samples of patients with chronic periodontitis and type 2 diabetes. Hu et al. [22] found that 3 mRNA biomarkers in saliva samples of patients with primary Sjogren syndrome were significantly higher than those of patients with systemic lupus erythematosus and healthy subjects. Some studies have found a simple and direct way to stabilize saliva mRNAs at room temperature without further processing [143, 144]. However, mRNA is still not a good candidate for salivary transcriptome analysis due to its highly fragmented under decomposition of RNA enzyme, and is easy to mix with bacterial RNA. Therefore, most salivary transcriptome studies are focused on the analysis of non-coding RNAs, which are the new regulatory factor with a variety of biological functions, and play an important role in cell development, differentiation, tumorigenesis and development. Compared with mRNA, the non-coding RNA (especially microRNA) in saliva is abundant, stable and shorter sequence, so it is not easy to be degraded by RNA enzyme and is more suitable for hybridization analysis with oligonucleotides on biosensors [145]. The microRNAs participate in various biological processes, regulating cell differentiation, proliferation and survival, especially in cancers [146148]. Park et al. [149] detected microRNAs in saliva of patients with oral squamous cell carcinoma and healthy controls by reverse transcription-pre-amplification-quantitative PCR. The results showed that the contents of miR-125a and miR-200a in saliva of patients with oral squamous cell carcinoma were significantly lower than those of healthy controls. Xie et al. [66] found the potential biomarkers of resectable pancreatic cancer by detecting the microRNA in saliva. The combination of miR-3679-5p and miR-940 presents well discriminatory ability to detect resectable pancreatic cancer with acceptable specificity and sensitivity. With the development of high-throughput RNA sequencing analysis technology, it is possible to analyze the whole RNA sequence in saliva, and explore more potential biomarkers in the disease. Wong et al. [73] investigated the saliva extracellular RNA biomarkers in evaluating the gastric cancer. Saliva samples from 63 patients with gastric cancer and 31 patients without gastric cancer were analyzed by transcriptome sequencing. The results showed that there were 30 mRNAs and 15 microRNAs expression patterns associated with gastric cancer. Among them, the biomarker group composed of 3 mRNAs (SPINK7, PPL and SEMA4B) and 2 microRNAs (miR140-5p and miR301a) had reliable performance, which was significantly down-regulated in gastric cancer, and the AUC reached 0.81.

The proteome of saliva

The salivary proteome mainly studies the protein profile in the oral cavity. These proteins participate in different biological functions in the oral cavity and are the most important components in saliva. Saliva contains more than 2000 proteins produced by salivary glands and microorganisms in mucous membrane and oral cavity [150]. The proteins secreted by acinar cells of salivary glands account for more than 85% of salivary proteins, while the proteins secreted by glandular cells, such as growth factors and immunoglobulins, have important biological functions [151]. The proteolytic enzymes in oral cavity can decompose salivary proteins into small molecular proteins or peptides, which account for 40–50% of salivary proteins [152, 153]. The molecular weights of various proteins in saliva vary greatly. The existence of high molecular weight proteins such as amylase and mucin will interfere with the detection and identification of relatively low abundance proteins, which may be potential biomarkers. Therefore, the ability of salivary proteome technology to detect and identify low abundance proteins is very important. The commonly used proteomic techniques include western blotting (WB), enzyme-linked immunosorbent assay (ELISA), two-dimensional gel electrophoresis (2D-GE), sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), quantitative mass spectrometry (qMS), surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF-MS) and Raman spectroscopy (RS). Mass spectrometry (MS) is a special technique for the identification of compounds, providing structural information with high specificity and accuracy. MS has an excellent ability to measure peptides and small molecular proteins, and has become the core technology of proteomics, which can greatly improve the research progress of salivary proteome, broadening our understanding of saliva proteins. Moreover, combined with advance separation techniques, such as high-performance liquid chromatography (HPLC), small proteins and peptides in saliva can be analyzed qualitatively and quantitatively. Pappa et al. [154] identified more than 2000 proteins in the whole saliva by HPLC-MS.

At present, four main salivary proteins have been identified on the research of human salivary proteome, which are proline-rich proteins (PRPs), cystatins, statherins and histatins [155]. These proteins play a vital role in maintaining the integrity of teeth by regulating the balance of enamel demineralization and remineralization [156]. It has been found that about 27% of plasma proteins are present in saliva, which reflects the contribution of vascular leakage or fluid from interstitial compartment to saliva components [154, 157]. The main components of human plasma protein are immunoglobulin and albumin, which represent 60–80% of the total weight [158]. The 22 most abundant proteins in plasma account for 99% of the total protein content in plasma, while the 20 most abundant proteins account for only 40% of the saliva protein content. For the abundant and wide dynamic range proteins in the plasma fluid, it is more difficult to identify the lower abundance proteins which are important in detecting disease. Comparatively, the proportion of high abundance proteins in saliva is moderate, which is more beneficial for detecting low abundance proteins. Furthermore, some of the most abundant proteins in plasma have moderate to high relative abundance in saliva, while only a small number of high abundance salivary proteins have an equally abundance in plasma [159]. These suggest that salivary proteins have certain potential as biomarkers. Xiao et al. [74] quantitatively identified more than 500 proteins by detecting the saliva protein components of gastric cancer patients and healthy controls, of which 48 were differentially expressed, 41 were down-regulated, and 7 were up-regulated in patients with gastric cancer. Cystatin B (CSTB), triosephosphate isomerase (TPI1) and deleted in malignant brain tumors 1 protein (DMBT1) can significantly distinguish gastric cancer patients from healthy controls. The sensitivity of the combination of these three protein markers for the detection of gastric cancer is 85%, the specificity is 80%, and the AUC is 0.93.

The metabolome of saliva

As the final product of gene expression, metabolites can reflect the actual biological status of the body. Metabolomics is the quantitative analysis of all metabolites in the body, such as nucleic acids, amino acids, peptides, lipids, carbohydrates and vitamins, so as to find the relative relationship between metabolites and physiological and pathological changes. It is helpful for us to monitor the state of the body and make correct treatment decisions [160, 161]. Especially in cancer, changes in metabolites can occur before the symptoms of the disease so that abnormalities in the body can be detected as early as possible [162]. The metabolites in saliva can reflect the health and disease state in the oral cavity, but also the state outside the oral cavity. Most of the metabolites in the blood can transport to saliva due to the close relation between salivary glands and capillaries, and the small molecular weight of metabolites. Additionally, the changes of metabolites in saliva are generally consistent with the blood so that they can reflect the effect of disease, nutrition, drugs and environment on the body status [130]. Nonetheless, the fact is that the concentration of metabolites in saliva is much lower than that in blood, which requires the measure technique with a high level of sensitivity. The emergence of MS makes up for the deficiency of the detection of low molecular weight compounds, and it is a common research method in proteomics and metabolomics. The metabolomic technologies are usually based on various separation techniques combined with MS, such as liquid chromatography mass spectrometry (LC-MS), high performance liquid chromatography-mass spectrometry (HPLC-MS), ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS), gas chromatography mass spectrometry (GC-MS), plasma mass spectrometry, proton-nuclear magnetic resonance (H NMR) spectroscopy, and capillary electrophoresis-based mass spectrometry (CE-MS) [71].

Most salivary metabolomic studies focus on whole-mouth saliva (WMS). The composition of WMS is complex, including saliva secreted by three major glands and hundreds of small glands in the mouth, but a large number of microorganisms and host cells (such as neutrophils). These microorganisms and host cells may have continuous metabolic activities in saliva, which challenges the metabolomic analysis of WMS [156, 163]. Saliva collected directly from the gland can eliminate the interference of microorganisms to the sample (commonly collected in the parotid gland). The secretions collected by this method do not flow into the oral cavity, without modification from oral microorganisms, which can simply reflect the metabolic activity of the host [164]. Research found that oral microorganisms importantly contribute to the WMS metabolomes, and some WMS metabolites (such as lactate, citrate and urea) are derived from the blood circulation, indicating that WMS may play a special role in helping to diagnose bacteria dysbiosis [165]. Saliva metabolites are a collection of metabolic activities of a complex system. Combining hosts and microorganisms to study salivary metabolomics is a potential direction which is helpful for us to further understand the role of salivary metabolites in health and disease. Sugimoto et al. [71] used capillary electrophoresis time-of-flight mass spectrometry to study metabolites in saliva samples from healthy controls and patients with periodontitis, oral, breast and pancreatic cancer, finding 57 metabolites that predict these diseases. Among them, three metabolites (taurine, piperidine and a peak at 120.0801 m/z) are specific markers of oral cancer with an AUC of 0.865, and eight specific markers of pancreatic cancer (leucine, isoleucine, valine, tryptophan, glutamic acid, phenylalanine, glutamine and aspartic acid) with an AUC of 0.993. Song et al. [60] collected saliva samples from healthy people, patients with precancerous lesions and oral squamous cell carcinoma (OSCC). The abnormal metabolites and metabolic pathways were found by conductive polymer spray ionization mass spectrometry (CPSI-MS) analysis. Then the reliability of metabolic markers in diagnosis was verified by desorption electrospray ionization MS imaging (DESI-MSI) at the tissue level of the primary tumor. Finally, combined with machine learning (ML), it can distinguish OSCC, precancerous lesions and healthy people in real time, and the accuracy is as high as 86.7%. This method is a direction with huge potential for developing salivary metabolomics in point-of-care technology (POCT).

The microbiome of saliva

The oral cavity is the second largest microbial reservoir in the human body after the intestinal tract. The diversity of the microbiome is also second only to the intestinal tract, with more than 700 bacterial species [166]. As we all know, oral microflora is closely related to oral health and diseases. Studies have shown the relationship between oral bacteria and dental caries, periodontitis, dry socket, pulpitis and odontogenic infection [167, 168]. In addition, oral microbes are potentially associated with systemic diseases, such as diabetes, heart disease and pancreatic cancer [72, 169, 170]. Most microbial species detected in different parts of the mouth overlap, but there are usually some specific species in some parts. Saliva microbiome is basically composed of a mixture of microorganisms from all parts of the mouth, which can include microorganisms at almost all sites. It is a relatively ideal specimen for microbial detection. The common research methods of salivary microbiome are bacterial microarrays and next-generation sequencing technology (NGS), of which the NGS is the most important research method. Wei et al. [171] detected the samples of pancreatic cancer patients and healthy people by 16S rRNA sequencing, and compared the difference of microbiome between them. The results showed that the carriage of Streptococcus and Leptotrichina had a higher risk of developing pancreatic cancer (PDAC), while those carrying Veillonella and Neisseria had a relatively lower risk of developing PDAC. These two microorganisms may be protective bacteria to prevent PDAC. Among PDAC patients, those with abdominal distension had a higher amount of Porphyromonas (p=0.039), Fusobacterium (p=0.024), and Alloprevotella (p=0.041), while patients with jaundice had a higher level of Prevotella (p=0.008), patients reporting diarrhea had a lower amount of Neisseria and Campylobacter (p=0.024 and p=0.034), and patients with vomit had a lower amount of Alloprevotella (p=0.036). These suggest that salivary microbiome was likely to distinguish patients with PDCA and healthy individuals. Furthermore, combining symptoms and microbiological assessment may be helpful for early diagnosis of PDCA. Jiang et al. [172] performed 16S rRNA sequencing and whole-genome shotgun metagenomic sequencing on saliva samples from 13 salivary adenoid cystic carcinoma (SACC) patients and 10 healthy controls to analyze microbial diversity, composition and function in both groups. The α diversity was not significantly different between SACC patients and healthy controls, while the β diversity showed a separation trend. And Streptococcus and Rothia were more abundant in SACC patients, while Prevotella and Alloprevotella were more abundant in the healthy controls. These results showed the potential of saliva to distinguish SACC patients from healthy controls.

The exosomes in saliva

Exosomes are a kind of nanoscale lipid bilayer vesicles (about 30–120 nm) secreted by nearly all cell types, mainly derived from multivesicular bodies (MVBs) formed by invagination of lysosomal particles. In forming MVBs, some small molecules in the cytoplasm, including mRNAs, microRNAs and proteins, are intake by budding inward to form small internal vesicles. Part of MVBs is fused by lysosomes and decomposes the contents. When the MVBs fuse with the cell membrane, the internal vesicles, as exosomes, explosively release out of the cell and act on the adjacent cells. Moreover, it can also spread to the distant tissue through the vascular system and promote the communication between cells, affecting the biological behavior of cells. Exosomes recognize target cells through surface-specific ligands and usually enter the target cells in two ways. One of them is that the target cells absorb it into the cytoplasm by endocytosis. The other is by fusing with the membrane of the target cell and releasing the contents directly into the cytoplasm [173]. Exosomes can be found in various biological body fluids, such as blood, saliva, urine, cerebrospinal fluid and breast milk [174176]. A large number of high-throughput exosomes studies have shown that exosomes contain a variety of small molecular substances, including mRNA, non-coding RNA, mitochondrial DNA, proteins, lipids and metabolites, and their size and cargo are heterogeneous [177179]. Exosomes exist widely in a variety of extracellular fluids, and transport cargo with diversity, which endows them with multiple and complex biological functions. Studies have shown that exosomes play an important role in cell development and differentiation, tumor growth and invasion. Sphingosine kinase 2 can be transferred into target hepatocytes to form sphingosine-1-phosphate in hepatocyte-derived exosomes, which leads to cell proliferation and liver regeneration [180]. The exosomes derived from breast cancer cells contain Dicer, AGO2 and TRBP. After being transported to the target cells, the cargo can process the precursor microRNA into mature miRNA, silence some tumor suppressor genes and induce non-carcinogenic epithelial cells to form tumors [181]. Additionally, the heterogeneity of exosome size and cargo can reflect the state and type of parental cells, which makes it a potential biomarker for disease diagnosis and prognosis. Some proteins in exosomes, such as CD9, tumor rejection antigen 1 (gp96) and caveolin 1 (CAV1), are valuable in the diagnosis of HNSCC, and can be used as potential biomarkers for diagnosis and prognosis [182]. The combined detection of exosome miR-21 and lncRNA HOTAIR as biomarkers for the diagnosis of laryngeal squamous cell carcinoma achieved a sensitivity and specificity of 94.2 and 73.5%, respectively [183]. For the study of exosomes, different methods can be adopted according to the research object, usually combined with proteomics, transcriptome or metabolomics to study the biomarkers. In the development of systemic cancers such as lung and pancreatic cancer, some specific biomarkers can be detected in saliva samples [67, 184]. This may all be due to the role of exosomes, which can carry substances from the source cells to drift through the vascular system, thus passing these goods into saliva and being detected.

The exosomes in saliva can be used as a new diagnostic platform for disease research. Saliva is easy to collect and non-invasive. Combined with the various and important biological functions of exosomes, it will become a new potential biomarker of oral and other systemic diseases and tumors. Machida et al. [185] found that miR-1246 and miR-4644 are highly expressed in plasma exosomes of patients with pancreatic cancer, and can also be identified in salivary exosomes, which can be used as potential biomarkers of pancreatobiliary tract cancer. Zhang et al. [186] identified cancer-enrich small tsRNA signature by RNA sequencing of salivary exosomes obtained from 3 patients with esophageal squamous cell carcinoma (ESCC) and 3 healthy controls, and further verified in the discovery cohort (66 individuals). The results showed that tsRNA (tRNA-GlyGCC-5) and previously unrecorded small RNAs were specifically enriched in salivary exosomes of ESCC patients, ESCC tissues and ESCC cells. The bi-signature composed of these small RNAs can discriminate ESCC patients from healthy controls with high sensitivity (90.50%) and specificity (94.20%). Furthermore, a bi-signature Risk Score for Prognosis (RSP) was set up to evaluate the risk. The patients with high RSP present shorter overall survival (OS) and progression-free survival (PFS) than those with low RSP. In addition, adjuvant therapy improved OS and PFS only in patients with high RSP. These results are consistent in the training and validation cohort. It suggested that tsRNA-based signature has diagnostic and prognostic potential, but also can be used as pre-operative biomarkers to screen patients more sensitive to adjuvant therapy. This study broadens the prospect of salivary exosomes research and has considerable guiding significance for the future study of salivary exosomes. The study of salivary exosomes has a broad prospect, but it is still in the early stage, and a lot of research is still needed to lay the foundation for this hot direction.

Saliva collection and standardization procedure

Among the body fluid collection methods, saliva collection is the most simple and fast. Notably, the accurate participant identification, sufficient sample size and appropriate container type are indispensable for each collection methods. Additionally, maintaining consistency in the marking and processing of samples is critical, which can ensure the accession to the high-quality data [187]. Due to the influence of saliva flow rate, type of salivary gland, circadian rhythm, saliva stimulation type, age, sex, physiological status, diet and collection methods, the detection results of saliva samples will be some difference. Therefore, we need to further standardize the methods of saliva collection. It is also necessary to adopt corresponding methods for different saliva detection contents, which can reduce the error as much as possible.

The composition of saliva is complex, and will change with age. Compared with the young, the saliva flow rate and calcium concentration of the elderly decreased, while the ion concentration increased significantly [188]. Similarly, the salivary mucin levels between the young and the old present significant differences [189]. Moreover, the protein profiles between newborns and adults have been reported to differ [190]. In the aspect of gender, the size of salivary glands in males is generally larger than that in females, so it also has a certain effect on saliva secretion. The unstimulated saliva secretion of healthy men is higher than that of women, presenting gender-dependence [191]. Hence, subjects of different ages and genders should be classified, tested and counted to order to reduce errors. In addition, the flow rate of saliva of the same person is different at different time points, so it is best to collect saliva at the same time. In collecting large samples, it should also be considered to eliminate individuals with extremely large and small amounts of saliva to minimize errors.

Salivary glands include three major salivary glands and many small salivary glands. Although most of the components secreted by these salivary glands are the same, there are differences in concentration. The parotid gland mainly produces serous type of saliva, which contain high levels of α-amylase and proline-rich proteins. The sublingual gland is mainly mucous acini, which mainly secretes mucin and lysozyme. As a mixed gland, the submandibular gland secretes both α-amylase and mucin. Other minor salivary glands mainly secrete mucin and lipase. The saliva collected from each salivary gland is generally sterile, and has a similar composition to that of plasma. It is generally used to measure the flow of each gland and to detect for salivary gland diseases. Whole mouth saliva (WMS) is a mixture of salivary gland secretions, gingival crevicular fluid, nasal and bronchial secretions, food residues, bacteria and their metabolites [192]. It can be divided into stimulated whole saliva (SWA) and unstimulated whole saliva (USWA). The production, composition and main sources of these two types of saliva are different. The WMS is rich in ingredients, which is suitable for the assessment of systemic diseases. And the interference factors of USWA collection are small, so it is a commonly used sample in the study. In collecting saliva samples, it is necessary to consider the gland sources and the state of stimulation. We should ensure that the samples collected are consistent, and different types of saliva should be selected according to the content of the study.

The common methods of collecting WMS include the draining method, the spitting method, the suction method, and the swab method [193]. And the collection of saliva from a single gland needs some special methods and equipment [194]. As mentioned above, WMS is more suitable for evaluating systemic diseases for containing more analyzable substances. Therefore, the methods and equipment for collecting WMS are more commonly used. Currently, there is still no generally accepted WMS sample collection technique, which will also result in poor reliability and reproducibility of the results, hindering the process of saliva research [195]. The UWS collected by swab method or suctioning will increase the variability of data, because the wiping action will cause certain stimulation [193]. The samples collected by spitting may contain more microbe than the draining method, which can be used to identify microorganisms, but can also affect the storage of saliva and the follow-up analysis of some compounds [196]. At present, passive drainage of UWS from the lower lip and spitting directly into the tube is considered the most promising and recommended method to minimize the error in the saliva collection process. This method has the following advantages: 1. collecting a large number of samples in a short time [197]; 2. containing a variety of substances in the samples, which can be used to detect a variety of biomarkers, and is suitable for most of the studies of omics; 3. can be preserved for a long time without affecting the detection. In addition, it is suggested that only one type of collection method and device should be used in a given study, and some guidance should be given to the sample provider. At present, many companies have produced special equipment for saliva collection, and even special versions for children, infants or for the detection of some specific compounds. These commercial salivary collectors include: (1) Salivette (Sarstedt AG & Co, Germany); (2) Oragene saliva kit (DNA Genotek, Canada); (3) Aware Messenger Device (Calypte Biomedical Corp, USA); (4) BBL Culture swab (BD, USA); (5) Intercept Oral Specimen Collection Device (OraSure Technologies Inc, USA); (6) UpLink saliva collector (OraSure Technologies Inc, USA); (7) Saliva Collection device (DNA Genotek, Canada) [198]. Although these devices are varied, they all have a similar protocol: subjects are told to avoid brushing for at least 30 min before collecting saliva, and not to eat food, drinking (except water) or chew gum, then gargle and spit into the collection tube. Saliva samples can universally be preserved at room temperature for 30–90 min. Low temperature can reduce the degradation rate of protein in saliva [196, 199]. In terms of storage, saliva samples should be frozen at −20 °C or below immediately after collection. Without this condition, the sample can also be stored at 4 °C to prevent bacterial growth and degradation of saliva components [196]. Generally, the sample can be stored at −80 °C for several years without degradation [187]. It is a routine process to add inhibitors before storage. However, for the RNA analysis, QIAzol method has been reported that could separate high yield RNA without additional RNA stabilizers. By using this method, saliva samples can be successfully stored at −80 °C for more than 2 years without adding RNase inhibitors [200].

Regardless of saliva collection methods, pre-analysis and analytical variables need to be standardized, such as collection and storage methods, circadian variation, sample recovery, sample contamination prevention and analytical procedures. The salivary flow rates, age, gingival health and physiological status of subjects can also affect the concentration of biomarkers. This significant range of variables raises concerns about the accuracy and repeatability of diagnosis using salivary biomarkers [195, 201]. How to overcome these challenges will be one of the keys to develop salivaomics research, which requires us to carry out a large number of samples to determine.

Clinical application of salivary biomarkers

Disease detection

Oral disease and cancer

Caries

Microorganisms are an important cause of dental caries. The colonization, proliferation and metabolism of cariogenic bacteria, especially Streptococcus mutans and Lactobacillus, have been widely used to identify individuals prone to caries. Samaranayake et al. [202] collected the salivary sample of the high and low caries activity population by paraffin wax stimulation method, and compared them after incubation. It suggested that the densities of Streptococcus mutans and Lactobacillus in people with high caries activity were much higher than those with low caries, and the difference was about an order of magnitude. With the application of high-throughput sequencing technology in microbiome, microorganisms in saliva are expected to become potential markers. Yang et al. [203] analyzed the salivary samples of 19 caries-active and 26 healthy control by 16S rRNA sequencing. The results showed that the Prevotella genus increased significantly in the caries active group, distinguishing the caries microbiota from the healthy ones. The results are also cross-verified by whole-genome-based deep-sequencing data. More importantly, it suggested that a single microbe change is not enough to identify caries, but it needs to be verified by the changing whole microbiome structure. Moreover, the changes of electrolytes in saliva can also be used as predictors of caries, and can be combined with microbiome to construct a diagnostic model [204].

Periodontal diseases

Periodontal diseases (PD) are serious infection of gingival tissue caused by many factors, which can lead to the destruction of periodontal ligament and alveolar bone. The microbes play a vital role in the occurrence and development of periodontal disease. The pathogenic bacteria can produce a variety of toxic factors to reduce the resistance of host, and directly destroy the periodontal tissue. In addition, they also stimulate the host’s immune response, and induce the release of a variety of inflammatory factors to further aggravate the destruction of periodontal tissue. Therefore, the pathogenic bacteria and inflammatory cytokines will be significant factors in predicting periodontal disease. Porphyromonas gingivalis has been proved to be closely associated with periodontitis. Currently, an ELISA kit has been developed for the special detection of P. gingivalis in saliva. Compared with qPCR, its detection speed is faster, sensitivity and specificity are as high as 92 and 96%, respectively. Hence, it is expected to become an easy and rapid chair-side diagnostic tool for the detection of P. gingivalis, and is of great potential in rapid screening of periodontitis [205]. Nonetheless, most studies have focused on the detection of inflammatory cytokines in periodontal disease as diagnostic markers. Kim et al. [9] used ELISA kit to detect matrix metalloproteinase-9 (MMP-9) and S100A8 (a molecular subgroup of acid calcium binding protein S100) in saliva of 99 patients with periodontitis and 50 healthy controls. In addition, patients with periodontitis were processed with non-surgical treatment, and saliva MMP-9 and S100A8 were detected again 3 months later. The results showed that MMP-9 and S100A8 were associated with periodontitis. Using saliva MMP-9 and S100A8 algorithm (combining age, sex, smoking, drinking and other factors), the screening ability of periodontitis can reach 0.86. Comparing saliva samples of patients with periodontitis before and after treatment, it was found that S100A8 and MMP9 decreased by 83.7% and 23.5%, respectively, suggesting that MMP-9 and S100A8 have the potential to be used as models for diagnosis and prognosis of periodontitis. Junior et al. [206] evaluated the level of myeloid-related proteins in saliva in relation to PD, and their potential screening ability. The results showed that the colony-stimulating factor-1 (CSF-1), S100A8/A9, S100A12, interleukin-1β (IL-1β), matrix metalloproteinase-8 (MMP-8) and hepatocyte growth factor (HGF) in saliva of patients with periodontitis and gingivitis increased significantly, while IL-34 of patients with periodontitis was significantly lower than that of healthy people and patients with gingivitis. After 3 months of treatment, IL-34 increased significantly, whereas IL-1β and MMP-8 decreased 1 month after treatment. In addition, patients with periodontitis clustered in high and low levels of S100A8/A9, and the patients with high levels had deeper pockets, more bleeding, and higher S100A12. It suggested that the salivary level of myeloid-related proteins changes in periodontitis and is partially regulated by periodontal treatment. The S100A8/A9 level in saliva may be helpful in identifying different groups of periodontitis patients. Sukriti et al. [207] used the revised Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool to assess the methodological quality of articles searching for periodontal disease and related saliva diagnostic markers, and seven studies were selected in the evaluation system. The results showed that macrophage inflammatory protein-1α (MIP-1α), IL-1β, IL-6 and MMP-8 were identified as acceptable biomarkers for the diagnosis of PD. Generally, the combination of MMP-8 and IL-6 showed the best diagnostic performance. Furthermore, the combination of IL-1β, IL-6, MMP-8 and MIP-1α presented promising results for distinguishing gingivitis from periodontitis, as well as periodontitis from gingival health. In addition to bacteria and inflammatory cytokines, microRNA and exosomes are also reported to be of certain value in the diagnosis of periodontitis [6, 7]. Overall, we need to expand the sample size and further implement structured research to verify these biomarkers.

Primary Sjögren’s syndrome

Primary Sjögren’s syndrome (pSS) is a chronic systemic autoimmune disease that damages the exocrine glands (mainly the salivary and lacrimal glands), characterized by dry mouth and keratoconjunctivitis [208]. The decrease of saliva flow rate and the change in saliva composition are important manifestations of the progression of pSS. These symptoms can lead to serious complications such as dental caries, periodontal disease, and Candida albicans infection. The B cell hyperactivity expressed as hypergammaglobulinemia, the existence of several serum autoantibodies and the activation of type I interferon (IFN-1) pathway are the immunopathogenic mechanisms of pSS, which lead to the excessive production of immunoglobulin [209, 210]. In addition, the secretory function of salivary glands is impaired, which makes the salivary protein profile of pSS patients change. Hu et al. [211] identified saliva autoantibodies of pSS using immune-response protoarrays. Of the 24 candidate autoantibody biomarkers, anti-histone, anti-transglutaminase, anti-SSA and anti-SSB were validated in a cohort of 534 pSS patients, 534 SLE patients and 534 HC. All four autoantibodies were overexpressed in pSS patients. The AUC of anti-histone, anti-transglutaminase, anti-SSA and anti-SSB to pSS (relative to HC) were 0.95, 0.87, 0.93 and 0.94, respectively. Lee et al. [24] found abnormally high expression of soluble sialic acid-binding immunoglobulin like lectin (siglec-5) in serum and saliva of patients with pSS. Then investigated the correlation between the level of siglec-5 in saliva of pSS patients and clinical parameter. The results showed that the level of siglec-5 was negatively correlated with saliva flow rate, but positively correlated with dry mouth scale (XI) score and ocular staining score (OSS). The ROC analysis showed that when the cutoff value was 400 pg/mL and the area under the siglec-5 curve was 0.774, the sensitivity and specificity were 0.69 and 0.70, respectively. In the verification cohort, the sensitivity and specificity of siglec-5 in distinguishing pSS patients from non-SS patients were 64.4 and 77.8%, respectively. It is possible to be used as a new saliva biomarker of pSS.

Oral lichen planus

Oral lichen planus (OLP) is a chronic inflammatory disease that often affects the tongue, gums and buccal mucosa. As a potential malignant disease, OLP has a certain carcinogenic rate, so early diagnosis and intervention treatment are particularly important [212]. Abnormal activation of the immune system is an important mechanism for the occurrence and development of OLP. Therefore, various signal molecules in the immune system pathway may be potential biomarkers for diagnosing OLP. Liu et al. [28] found that IL-4 was significantly increased, while IFN- γ decreased in salivary sample of OLP patients. IL-4 and IFN- γ are the principal cytokines of Th1 and Th2 cells, respectively. This result is consistent with the phenomenon that Th2 cells are predominant in saliva of patients with OLP. These also reflect the potential of IL-4 as a biomarker to detect the severity of OLP. Additionally, it has also been reported that other inflammatory cytokines such as IL-1, IL-6, IL-8 and TNF- α in saliva can be used as biomarkers for the diagnosis and prognosis of OLP [213, 214]. On the other hand, oxidative stress and impaired antioxidant defense are also possible causes of OLP. Many studies have shown that the levels of oxidative stress molecules such as lipid peroxide malondialdehyde, 8-Hydroxy-deoxy (8-OHdG) and nitric oxide (NO) in saliva of patients with OLP are significantly increased [215, 216]. Although no studies have shown the relationship between the concentration of these substances in saliva and the severity of OLP, they have a potential role in monitoring the progress of the disease. Recently, Mehdipour et al. [26] found that miR-21 was significantly increased in salivary samples of OLP and OSCC patients, while miR-125a levels were significantly decreased. These results may become the research direction of the salivary transcriptome in OLP.

Oral leukoplakia

Oral leukoplakia (OLK) is a white plaque on oral mucosa that cannot be removed by scratching. It is the most common precancerous lesion in oral cavity and has a high carcinogenic rate. Traditionally, resection and biopsy are the main diagnostic methods, which are more traumatic. Therefore, many diagnostic studies pay more and more attention to less invasive methods, such as the analysis of inflammatory cytokines in saliva. Previous studies have shown that the levels of IL-6 and TNF-α in saliva of patients with OLK are significantly higher than those of healthy people, which can be used as potential biomarkers of OLK [33, 217]. However, many reports show no significant difference between TNF- α and ILs in saliva of healthy individuals and patients with OLK, even decreased [218220]. In addition to cytokines, other salivary proteomic studies have reported possible markers. Sivadasan et al. [32] used liquid chromatography and tandem mass spectrometry (LC-MS/MS) to analyze protein components in saliva samples of healthy controls, patients with leukoplakia, OSCC patients with negative and positive lymph node metastasis. The results showed that the expression of S100A7, S100P, CD44 and COL5A1 increased in leukoplakia and tumor patients. Subsequently, through the detection of these proteins in the verification queue by ELISA, it was found that CD44, S100A7 and S100P could distinguish dysplastic leukoplakia from normal people with high sensitivity (CD44: 91.67%; S100A7: 81.82%; S100P: 81.82%), but relatively low specificity (S100A7, S100P: 72.73%; CD44: 54.55%). The result suggested that the saliva levels of these proteins can be used as potential markers for predicting patients with OLK, but further sample expansion and clinical verification are necessary. Precancerous lesions involve changes in some special genes and proteins, but also changes in the concentration of endogenous metabolites. Wei et al. [221] found that threonine, isoleucine, phenylalanine, n-tetradecanoic acid, homocysteine and 4-methoxyphenylacetic acid in saliva of patients with OLK were significantly higher than those of healthy people. Sridharan et al. [222] also reported the differential expression of 157compounds in patients with OLK compared with healthy people. All these provide a direction for applying salivary metabolomics in OLK, which is worthy of our in-depth study. There is no clear clinical data to support the effective diagnostic markers of OLK in saliva. Further research is needed to find and construct a biomolecule model of saliva diagnosis which can be used for rapid clinical detection.

Oral cancer

Oral cancer is a malignant tumor occurring in the oral cavity, and OSCC is the most common, accounting for about 95%. Most of the OSCC cases in the world are concentrated in Asia, and the prognosis is very poor. Despite the improvement in surgical and chemotherapy techniques, the 5-year overall survival rate of patients was only about 64.4% [223]. Hence, the early diagnosis and prevention of OSCC are particularly important, which is conducive to treatment and prognosis. Especially in densely populated Asia, simple and rapid techniques for extensive scale screening is necessary. OSCC can occur in many factors, including smoking, drinking and microbial infection. The DNA mutation, abnormal transcription, abnormal protein expression, metabolic disorder and microbial composition will change, and this abnormal information can be spread throughout the oral cavity. Long-term direct contact between saliva and oral tissue can better reflect the information on oral diseases, which is also the reason why early saliva screening and diagnosis have become a research hotspot in OSCC. At present, there are many high-quality saliva diagnostic markers in OSCC, including genome, transcriptome, proteome, metabolic group and microbiome.

Gene mutation is one of the markers of carcinogenic progression, and the mutation of cancer-specific gene can accurately distinguish all kinds of tumors. The mutated DNA can flow into various body fluids, including saliva, which is the basis of the saliva genome. The lesions of OSCC patients are immersed in saliva for a long time, and the released factors or exfoliated cells also become components of saliva. Therefore, salivary genomics has a natural advantage in the early screening and diagnosis of OSCC. Preston et al. [224] found that HOXA9 and NID2 in saliva samples can distinguish between OSCC patients and healthy controls, with a certain sensitivity (75 and 87%, respectively) and specificity (53 and 21%, respectively). The AUC is 0.75 and 0.73, respectively, which can be used for early detection and follow-up patients with OSCC. Nagata et al. [225] analyzed the methylation status of genes in salivary samples from patients with OSCC and healthy people. It was found that the DNA methylation level of 8 genes was significantly increased in the OSCC group. The methylation status of combination of ECAD, TMEFF2, RAR β and MGMT showed high sensitivity (100%) and specificity (87.5%) in detecting OSCC. Shanmugam et al. [226] designed a custom next-generation sequencing panel with a unique molecular identifier that covers the coding regions of seven frequently mutated genes (CASP8, PIK3CA, FAT1, CDKN2A, NOTCH1, HRAS and TP53) in OSCC. It was used to detect saliva samples from patients with OSCC, and the results showed a detection rate of 95.87%. This shows that the targeted next generation sequencing technique is feasible and has a good prospect in the early screening and diagnosis of OSCC. Notably, in these studies, salivary samples were collected by salt water to reduce the degradation of free DNA in saliva, which can increase the content of DNA in the samples as much as possible.

The microRNA is stable in biological fluids and differentially expressed in all kinds of cancers. It is a commonly used fluid biopsy marker in transcriptome. Previous studies have shown that the levels of miR-125a and miR-200a in saliva of patients with OSCC are significantly decreased and can be used to detect oral cancer [149]. It was also found that the miR-31 in saliva was more abundant than in plasma, suggesting miR-31 in saliva is more sensitive in detecting OSCC. Additionally, the miR-31 decreased significantly after focal resection. These results indicated that salivary miR-31 could be used as a biomarker for early detection and postoperative follow-up of OSCC [227]. Recently, Romani et al. [34] collected saliva from patients with untreated primary OSCC and healthy controls, screening the differentially expressed miRNA through microarray analysis, using RT-qPCR for signature validation. In the training set, the saliva of patients with OSCC present high levels of expression of miR-106b-5p, miR-423-5p and miR-193b-3p. The accuracy of miR-106b-5p, miR-423-5p and miR-193b-3p as diagnostic biomarkers was evaluated by logical model prediction. The results showed that the combination of the three had excellent sensitivity (0.974) and specificity (0.942), and AUC was 0.98. The results in the validation set also confirmed this result, indicating that the combined application of miR-106b-5p, miR-423-5p and miR-193b-3p can be used as biomarkers for the detection and diagnosis of OSCC. Furthermore, it was also found that the high expression of miR-423-5p was an independent predictor of poor disease-free survival (DFS) when positive lymph nodes (the number of positive lymph nodes was the only important clinical prognostic factor) were included in multivariate survival analysis. Finally, it was also found that miR-423-5p decreased significantly after tumor resection, which may be a specific prognostic biomarker of OSCC progression and metastasis. In addition to microRNA, other RNAs can be used as diagnostic markers. Li et al. [36] found and identified 7 transcripts (DUSP1, H3F3A, IL1B, IL8, OAZ1, S100P and SAT) up-regulated in saliva samples of OSCC patients, which could distinguish OSCC patients from healthy controls. Tang et al. [228] found that all OSCC saliva samples contained lncRNA MALAT-1, indicating that lncRNA in saliva could also be used as a potential marker for OSCC diagnosis.

Abnormal protein expression often occurs in the process of tumor occurrence and development. These proteins usually enter the bloodstream and can be detected in the early stage. Saliva contains more than 2000 kinds of proteins, similar in composition to those in plasma, most of which are blood sources. Tumor antigen CA15-3 and tumor protein markers CA-125, c-erbB2 and p53 in saliva present increased expression in oral cancer and other malignant tumors, which can be used as biomarkers [229]. Yu et al. [51] detected and analyzed saliva from healthy people, oral potentially-malignant disorders (OMPD) patients and OSCC patients by LC-MS combined with multiple reaction monitoring (MRM). The MMP1, KNG1, ANXA2 and HSPA5 were selected from the differentially expressed proteins to form the diagnostic model. The model shows high sensitivity (87.5%) and specificity (80.5%) in distinguishing OSCC from non-OSCC patients. Moreover, a risk scoring scheme is constructed based on this model. When the risk scores >0.4, 84% of OSCC patients and about 42% of high-risk OPMD can be detected. In addition, of the 88 high-risk OPMD patients followed up, 18 developed to OSCC within 5 years, and 14 of them had risk scores >0.4. Chu et al. [56] screened 6 proteins (APOA1, APOA4, CFH, FGA, SERPINA1 and SERPIND1) by targeted and non-targeted quantitative proteomic approaches, which can effectively distinguish OSCC from healthy controls (AUC ≤0.8). Among them, the levels of CFH, FGA and SERPINA1 in saliva of patients with advanced primary OSCC are much higher than those of patients with early primary OSCC. In addition, some studies have found that proteases are related to metastasis and translocation of cancer. Inhibition of these proteases can reduce the invasion and metastasis of cancer cells [230232]. Feng et al. [43] found that the salivary protease spectrum of OSCC patients is significantly different from that of health and patients with other oral diseases, such as jaw bone ossification fibroma (JBO) and mild chronic periodontitis (CPD). The saliva of patients with OSCC present more protease types, and the levels of MMP-1, MMP-2, MMP-10, MMP-12, A disintegrin and metalloprotease (ADAM) 9, A disintegrin and metalloprotease with thrombospondin type 13 motifs (ADAMST13) and cathepsin V and kallikrein 5 increased significantly. The combination of cathepsin V, kallikrein 5 and ADAM9 has good sensitivity (85.0%), specificity (93.3%), and the AUC was 0.963.

Metabolites can reflect the real state of the body. Changes in cell metabolism have been identified as a new marker of cancer [233]. In the process of tumorigenesis and development, it is usually accompanied by changes in metabolic pathways, resulting in some abnormal metabolites, forming a unique metabolic profile of tumors. Saliva contains a large number of metabolites, especially the metabolites from the primary site of oral cancer, which will be mixed directly into saliva. At present, more than 100 metabolites have been reported to change with the malignant progression of OSCC, such as lactate, choline, glutamate, histidine, sialic acid and trimethylamine N-oxide [221, 222, 234]. Song et al. [60] used conductive polymer spray ionization mass spectrometry (CPSI-MS) to detect the salivary metabolic profiling of healthy people, patients with premalignant lesions and patients with OSCC. The results showed that the metabolic profiles of the three groups could be distinguished from each other and had considerable stability (the similarity of the metabolic maps of saliva collected at different times was as high as 86%). Further analysis showed that 58 metabolites changed significantly between healthy group and premalignant lesions group, while 116 metabolites increased or decreased significantly between premalignant lesions group and OSCC group. These results are also similar to those in the validation set. In addition, the Lasso regression was introduced to construct a machine learning (ML) model for OSCC prediction. The model showed an accuracy of up to 86.7% when it was applied to validation set. These results indicate that the metabolomic has a broad prospect in the development of saliva diagnostic markers for OSCC. In particular, CPSI-MS has the following excellent: rapid and direct metabolic analysis of biological liquids; low cost; cleanliness; and wide coverage of metabolites, which will make the point-of-care test (POCT) of OSCC possible, and will be the hot direction of large-scale early screening in the future.

Oral microorganisms play an important role in the health and disease of the host. In recent years, some studies have shown that the change of oral microflora will destroy the balance between microorganisms and human body, coupled with the influence of risk factors, which will lead to the occurrence of OSCC [235, 236]. Oral microorganisms, epithelial barrier, immune system and chronic inflammation constitute the four important factors leading to oral cancer [237]. Therefore, oral microorganisms are also promising potential biomarkers for early diagnosis of OSCC. Single microorganism, such as P. gingivalis, Fusobacterium nucleatum and Staphylococcus aureus have all been reported to play an important role in the occurrence and development of OSCC [238240]. However, a single microbe is not enough for the diagnosis of OSCC. It is necessary to identify the core microbiome that causes OSCC for developing a clinically applicable and highly accurate diagnostic model. Zhou et al. [65] sequenced saliva samples from 47 OSCC patients and 46 healthy subjects based on 16SrRNA sequencing, and established a random forest model. The results show that the accuracy of the model is 95.70%, and the sensitivity is 100%. Additionally, they found that even in healthy controls, saliva samples from different regions showed different microbial compositions. Finally, they also put forward the strategy of developing the prediction model: on the basis of establishing small samples, and gradually adding new samples to update the model, so as to improve the accuracy. This method is also applicable to healthy individual groups. The results suggest that salivary microbiome combined with machine learning is a new direction for early detection of OSCC and can be used in extensive scale population screening in the future.

Systemic malignant tumor

Pancreatic cancer

The researches of saliva diagnostic markers of pancreatic cancer are still in preliminary stage. There have been some reports on transcriptome in pancreatic cancer. Zhang et al. [67] analyzed the transcriptome of saliva samples from pancreatic cancer patients, pancreatitis patients and healthy controls. They found that the combination of 4 mRNA (KRAS, MBD3L2, ACRV1 and DPM1) could be used as markers to distinguish pancreatic cancer patients from non-pancreatic cancer patients (sensitivity of 90.0%, specificity of 95.0%, and AUC of 0.971). In addition to mRNA, more than 10 kinds of saliva microRNA have been reported [66, 68, 185, 241]. Humeau et al. [68] found that hsa-miR-21, hsa-miR-23a, hsa-miR-23b, and miR-29c in saliva of patients with pancreatic cancer were significantly up-regulated compared with the control group with 100% specificity, but relatively low sensitivity. The concentration of endogenous metabolites in saliva of patients with pancreatic cancer often changes, which can be used as a target for detection. Asai et al. [70] found that the combination of alanine, N1-acetylspermidine, 2-oxobutyrate and 2-hydroxybutyrate had ability to distinguish pancreatic cancer patients from non-pancreatic cancer patients, with an AUC of 0.887. Sugimoto et al. [71] found that eight metabolites, leucine, isoleucine, valine, tryptophan, glutamic acid, phenylalanine, glutamine and aspartic acid, could distinguish healthy controls from pancreatic cancer patients, with an AUC of 0.993. Additionally, Farrell et al. [72] found significant differences in salivary microbiota between pancreatic cancer patients and healthy controls based on human oral microbe identification microarray (HOMIM) and verified by qPCR in an independent cohort. The results showed that the combination of Neisseria elongata and Streptococcus mitis had a sensitivity of 96.4% and a specificity of 82.1% in distinguishing pancreatic cancer patients from healthy subjects, with an AUC of 0.90. Torres et al. [242] reported similar results in saliva samples from pancreatic cancer and healthy people by 16SrRNA sequencing. These reports confirmed that salivary microbiota could be used as an important informative source for exploring biomarkers of systemic malignant tumors.

Gastric cancer

Compared with the traditional liquid biopsy (such as blood, gastric juice), saliva biopsy of gastric cancer still lacks the consensus of analytical methods and clinical verification, which is also a problem faced by most systemic malignant tumors. During the development of gastric cancer, salivary glands are stimulated by nerve growth factors released from distant tumors, which can lead to significant changes in salivary RNA profiles. These differentially expressed RNA can be used to detect gastric cancer [243]. Li et al. [73] identified 30 candidate mRNAs and 15 candidate microRNAs expression patterns associated with gastric cancer. Through another independent cohort verification, it was found that the biomarker combination of 3 mRNAs (SPINK7, PPL and SEMA4B) and 2 microRNAs (miR-140-5p and miR-301a) could be used to distinguish gastric cancer patients from non-gastric cancer patients, with a sensitivity of 75% and specificity of 83%. This study proved for the first time that saliva extracellular RNA biomarkers have potential utility in early detection and risk assessment of gastric cancer. Xiao et al. [74] identified and quantified more than 500 proteins from saliva. Among them, 48 were differentially expressed between patients with gastric cancer and the control group. And the combination of Cystatin B, triosephosphate isomerase and malignant brain tumors 1 protein can be used to distinguish between gastric cancer patients and normal controls. The sensitivity and specificity were 85 and 80%, respectively, and the AUC was 0.93. Huang et al. [244] compared salivary microflora profiles of patients with gastric cancer, superficial gastritis and atrophic gastritis based on 16SrRNA sequencing. Through the construction of random forest model, it can accurately distinguish gastric cancer patients from non-gastric cancer patients, with an AUC of 0.91.

Breast cancer

At present, the evidence for saliva biomarkers of breast cancer is limited, and there are many interference factors. The chronic gingival inflammation has been proved to be the main confounding factor, reducing the clinical effectiveness of saliva biomarkers of breast cancer [76]. Through the detection and analysis of transcriptome and proteome of saliva from patients with breast cancer and normal controls, 8 mRNAs and 1 protein were found as biomarkers. The sensitivity and specificity of the combination in distinguishing breast cancer patients from normal controls were 83 and 97%, respectively, and the AUC was 0.92 [67]. However, the results of most proteome studies are not ideal, such as CA15-3. The CA15-3 is a serum proteomic biomarker approved by FDA to monitor breast cancer metastasis, but its use in saliva is still immature [245, 246]. Recently, metabolomics is becoming more and more important in the study of salivary biomarkers of breast cancer. Zhong et al. [83] proposed for the first time a method based on UPLC-MS and multivariate data analysis for salivary metabolomics analysis of breast cancer, and 18 potential metabolites were identified. Among them, three upregulated metabolites, LysoPC (18:1), LysoPC (22:6) and MG (0:0/14:0/0:0), had higher curvilinear AUC values of 0.920, 0.920 and 0.929, respectively, presenting potential to be used as biomarkers. Assad et al. [81] performed non-targeted metabonomic analysis of saliva samples from breast cancer patients and healthy controls, and 31 compounds up-regulated in the breast cancer group were identified. Among them, the lipid PG14:2 has a good sensitivity (65.22%) and specificity (77.14%) in distinguishing breast cancer patients from healthy controls, and the AUC is 0.73, which has the potential to be used as a saliva biomarker of breast cancer.

Lung cancer

Compared with other systemic malignant tumors, there are relatively many salivary studies on lung cancer, and many biomarkers have been found. Nonetheless, how to make the most effective use of these biomarkers in clinic needs further research.

Zhang et al. [85] identified and verified 7 highly discriminatory salivary biomarkers in saliva transcriptomes of patients with lung cancer. The logistic regression model conbined with CCNI, EGFR, FGF19, FRS2 and GREB1 could distinguish lung cancer patients from normal controls, with a sensitivity of 93.75%, a specificity of 82.81%, and an AUC of 0.925. Epidermal growth factor receptor (EGFR) is a membrane receptor frequently expressed in non-small cell lung cancer (NSCLC), which is also one of the most frequently studied genetic markers in the diagnosis of lung cancer. It is really vital to identify the presence and type of EGFR mutation in NSCLC, which is important for the treatment, for the common mutations, exon 19 deletion and exon point mutation 21-L858R of EGFR are sensitive to the treatment of tyrosine kinase inhibitors [247, 248]. Wei et al. [84] developed electric field-induced release and measurement (EFIRM), an electrochemical based technology using specific mutation-detecting probes, which can be used for cell-free DNA analysis of samples. Then they established EFIRM for detecting the EGFR mutation, and applied it in saliva samples of 40 NSCLC patients. The ROC analysis suggested that EFIRM can detect the exon 19 deletion (AUC=0.94) and the L858R mutation (AUC=0.96). These results indicate that the EFIRM for detecting EGFR is rapid and simple, meeting many clinical requirements for successful and efficient detection, and may become a clinical method in the future, either alone or in conjunction with supplementary analysis of biopsies.

The proteomic researches of lung cancer are mostly used for blood analysis, while saliva samples are relatively seldom used. Xiao et al. [86] identified 16 proteins for salivary biomarkers in patients with lung cancer. The combination of haptoglobin, zinc-a-2-glycoprotein and calprotectin presented a sensitivity of 88.5% and a specificity of 92.3%, promising saliva biomarkers. In addition, the proteins in salivary exosome have some potential biomarkers that need to be further verification [249].

As well as other systemic malignant tumors, the study of salivary metabolomics in lung cancer is still in its infancy. Li et al. [184] used Surface-enhanced Raman spectroscopy (SERS) to identify biomarkers in saliva of patients with lung cancer. They found 9 differential peaks between patients with lung cancer and healthy controls, most of which were nucleic acid bases and amino acids. Through the analysis of SERS data by PCA-LDA algorithm, it is found that lung cancer patients can be well distinguished from healthy controls, with a sensitivity and specificity of 94 and 81%, respectively. Jiang et al. [250] established an ultralow noise tip-enhanced laser desorption/ionization (TELDI) platform, combined with MS, to study the metabolic profile of saliva samples from patients with early lung cancer and healthy volunteers. After multivariate analysis, 23 metabolites were selected. Combined with artificial neural networks, the prediction model was constructed and verified by another independent queue. The results show that the model can distinguish between patients with early lung cancer and normal controls, with a sensitivity and specificity of 97.2 and 92%, respectively. And the AUC is 0.986. This study provided a new platform for the study of salivary metabolomics in lung cancer, and can also be used for the analysis of other diseases.

There are also relatively few studies on salivary microbiome of lung cancer. Yan et al. [89] proved the relationship between salivary microflora and lung cancer for the first time. Saliva from patients with lung cancer and normal controls were studied by 16S rRNA sequencing. It was found that the abundance of Veillonella and Capnocytophaga in saliva of patients with lung cancer increased, and their enrichment characteristics could distinguish lung cancer patients from normal controls. The sensitivity and specificity of the combination of Veillonella and Capnocytophaga to distinguish lung squamous cell carcinoma from normal controls were 84.6 and 86.7%, respectively, and the sensitivity and specificity of distinguishing lung adenocarcinoma from normal controls were 78.6 and 80.0%, respectively. It suggested that Veillonella and Capnocytophaga may serve as potential biomarkers for the detection or classification of lung cancer.

Systemic diseases

Cardiovascular disease

The salivaomics study of cardiovascular disease is mainly focused on proteome. C-reactive protein (CRP), salivary creatinine kinase myocardial band (CK-MB), myoglobin (MYO), troponin-1 (Tn1), myeloperoxidase (MPO) and matrix metalloproteinase-9 (MMP-9) are the most studied salivary biomarkers of cardiovascular diseases. Among them, CRP, CK-MB, MYO and Tn1 are the most promising biomarkers, and the results of others are controversial. Miller et al. [251] studied the role of serum and saliva as diagnostic fluids for identifying biomarkers of acute myocardial infarction (AMI). Through logical regression analysis, it was found that CRP was the best biomarker for predicting AMI. Floriano et al. [252] showed that the combination of salivary biomarkers containing CRP and electrocardiogram (ECG) could effectively distinguish AMI from healthy controls (AUC=0.96), which was much higher than that of ECG alone. These reports indicate that salivary CRP has considerable potential in the diagnosing AMI. Other biomarkers such as CK-MB, MYO and Tn1 present consistent increases in blood and saliva in patients with AMI, significantly higher than in healthy controls [93, 252, 253]. However, the current research evidence is insufficient to support them as saliva biomarkers of AMI, which needs to further expand the sample for research.

Diabetes mellitus

In the past, many studies have investigated the salivary protein profile of patients with diabetes mellitus (DM), and identified a large number of differentially expressed proteins, such as S100 calcium-binding protein A7, alpha-2 macroglobulin, alpha-1 antitrypsin and complement component 3, which can change with the development of diabetes [94, 154, 254]. However, most of these studies have failed to find diagnostic markers or combinations. The 1,5-AG (C6H12O5) can reflect the average glycemic level in the past one week, and is more sensitive to postprandial hyperglycemia and blood glucose fluctuations. During long-term hyperglycemia, a large amount of urine glucose was filtered out continuously, which inhibited the reabsorption of 1,5-AG in the proximal renal tubules, resulting in the loss of 1,5-AG. As a result, the level of 1,5-AG in diabetes patients was lower. Jian et al. [95] detected the level of 1,5-AG in saliva of DM patients and subjects without DM by liquid chromatography-mass spectrometry (LC-MS). It was found that the level of 1,5-AG in patients with DM was lower, and it was positively correlated with serum 1,5-AG, and negatively correlated with blood glucose and glycosylated hemoglobin. Further analysis showed that the best cutoff points of saliva 1,5-AG0 and 1,5-AG120 for DM screening were 0.436 μg/mL (sensitivity: 63.58%, specificity: 60.61%, AUC: 0.657) and 0.438 μg/mL (sensitivity: 62.25%, specificity: 60.41%, AUC: 0.660), respectively. Compared with fasting blood glucose (FPG) alone, the combination of FPG and saliva 1,5-AG reduced the proportion of people who needed oral glucose tolerance test (OGTT) by 47.22%. It is suggested that saliva 1,5-AG is a convenient and non-invasive tool for screening DM.

Other diseases

Differential expression of 8 proteins (calgranulin A, calgranulin B, apolipoprotein A-1, 6-phosphogluconate dehydrogenase, peroxiredoxin 5, epidermal fatty acid-binding protein, 78 kDa glucose-regulated protein precursor (GRP78/BiP), and 14-3-3 proteins) can be detected in saliva of patients with rheumatoid arthritis, which may play a role in diagnosis [96]. The level of salivary lactoferrin in patients with Alzheimer’s disease (AD) is significantly decreased, which can distinguish prodromal AD/AD from healthy controls, and may have a good diagnostic performance for the detection of AD [255]. Twenty-five proteins were identified as overexpressed in saliva of children with autistic spectrum disorders (ASD). Eight have been identified as potential biomarkers of ASD [256]. Salivary cortisol and α-amylase levels have long been reported as routine biomarkers reflecting psychological stress [257].

Personalized medicine

Classifying the patients according to the changed salivaomics profile, and then using statistical analysis to develop targeted therapy for different types of patients can greatly improve the therapeutic effect. EGFR in lung cancer is a good example. Some common mutations, exon 19 deletion and exon point mutation 21-L858R of EGFR are sensitive to tyrosine kinase inhibitors, such as Osimertinib, gefitinib and erlotinib [247, 248]. The mutation of EGFR in saliva can be directly detected by EFIRM, and the treatment plan can be made according to the results. HER2 overexpression occurs in 15–20% of breast cancer patients, which is an indication of trastuzumab therapy [258]. Detecting the level of HER2 in saliva may help to classify breast cancer patients, and identify subgroups of patients who will benefit from trastuzumab therapy. In addition, studies have shown that salivary IL-6 and IL-8 levels can distinguish patients who are more sensitive to gingival inflammation [259].

Therapeutic effect monitoring

Saliva biomarkers can be used in the diagnosis of diseases, but also can be used to detect the therapeutic efficiency of some diseases. Sexton et al. [260] found that the salivary levels of IL-1β, MMP-8, OPG and MIP-1α can reflect the severity of periodontitis and its response to treatment, which has potential use in monitoring the status of periodontal disease. The miR-31 and miR-423-5p in saliva decreased significantly after OSCC resection, which can be used as biomarkers of therapeutic effect and postoperative follow-up monitoring of OSCC [34, 227].

COVID-19 screening

Since the end of 2019, coronavirus disease2019 (COVID-19) has swept the world, bringing the most serious crisis to global health. Over the past two years, COVID-19 continues to wreak havoc worldwide because of its high mutagenicity and strong infectious ability. How to quickly detect the virus, identify, isolate and control positive infected person as soon as possible is an important measure. Therefore, efficient, rapid and accurate screening is the cornerstone of successful public health response to COVID-19. At present, nasopharyngeal sampling for nucleic acid amplification testing (NATT) is a non-invasive standard test for the diagnosis of COVID-19. However, this method requires trained personnel and specially designed swabs. On the other hand, the process of collecting nasopharyngeal swabs is painful for the subjects, and some elderly people or children cannot easily cooperate with the collection. And there is also a risk of infection when taking samples from some patients who have been diagnosed. Furthermore, although RT-qPCR is the gold standard for the diagnosis of COVID-19, the time it takes to provide results (a few hours to 1 day) does not meet the requirements of public health and epidemic prevention to respond quickly to emergencies in the face of large-scale screening. Therefore, saliva samples instead of nasopharyngeal swab samples, looking for faster detection methods than RT-qPCR, and developing kits or tools convenient for people to detect by themselves will be the direction of diagnosis of COVID-19 in the future. A recent systematic review and meta-analysis comparing saliva samples with nasopharyngeal swab RT-qPCR for COVID-19 indicated that the sensitivity and specificity of saliva samples were 83.2 and 99.2%, respectively, while the NATT with a sensitivity of 84.8% and a specificity of 98.9%. The results showed that the diagnostic accuracy of saliva samples was similar to that of NATT, which supported the use of saliva samples as a substitute for nasopharyngeal swabs for extensive scale screening [261]. On the other side, finding a faster and more convenient detection method than RT-qPCR is also on the agenda. Huang et al. [262] developed reverse transcription loop-mediated isothermal amplification (RT-LAMP) to rapid screening of COVID-19 in saliva. It is confirmed that multiple RT-LAMP can directly detect COVID-19 particles as low as 1.5 copies/ul in saliva without isolating RNA. It has the same sensitivity and specificity as standard RT-qPCR. Wood et al. [263] used synchrotron-Fourier transform infrared (FTIR) and purified virus Raman spectra to study the characteristic spectral signals of COVID-19 biomarkers in saliva samples. Through the construction of the model to detect COVID-19 in saliva samples, they achieved 93% sensitivity and 82% specificity. The above researches shows that the using saliva samples, combined with new detection techniques for COVID-19 screening, will be the future research direction, which is of great significance for the global prevention of COVID-19.

The limitations and future prospects

With the in-depth study of salivomics, saliva plays an increasingly important role in fluid biopsy, considered a reliable biological diagnostic fluid. Simple collection, convenient storage, non-invasive and containing a large number of diagnostic biomolecules are the advantages of saliva. It makes the impression that saliva will replace blood as the main body of fluid biopsies. However, saliva diagnosis is still in the early stage of development, and further research is necessary to determine and consolidate its position. Generally, the current studies on saliva biomarkers also show a lot of limitations.

Firstly, saliva analysis does have its limitations. The volume of saliva in the body is much lower than that of blood, and the concentration of saliva biomarkers is very low, which poses a challenge to the analysis. And the overlap of specific saliva markers in some diseases makes it difficult to clearly associate the disease with salivary biomarkers. For example, the level of MMP has significant changes in chronic periodontal diseases, oral mucosal diseases, and even cardiovascular diseases. This makes it difficult to accurately determine whether changes in biomarkers are caused by one particular disease or another. If a specific link between these biomarker levels in serum and saliva can be established, clinicians will have another tool to improve patient diagnosis and care.

Secondly, a single biomarker is not enough to define the pathogenesis of the disease, and using a variety of biomarkers to build a diagnostic model seems to be the most likely method. Additionally, to identify specific salivary biomarkers for specific diseases, the biomarkers need to be studied in large cohorts and long-term follow-ups to verify their accuracy. The clinical-oriented research will fundamentally improve the level of existing supporting evidence. The best diagnostic model of saliva may come from the combination of biomarkers of each pathological condition, with clinical effectiveness and higher accuracy and specificity.

Thirdly, the composition of saliva may vary with the method and time of collection, thus affecting the concentration of salivary biomarkers. Standardized programmes should be developed to reduce heterogeneity between studies on a given disease. In addition, continuous measurements may be more valuable than single-point measurements, and are more capable of analyzing biomarkers in saliva.

Finally, it must be pointed out that although there have been tremendous studies related to saliva diagnosis, the development of related kits is still in the exploratory stage. On the one hand, disease-related salivary biomarkers require more research to be identified. On the other hand, saliva testing requires some nanoscale devices with high sensitivity and specificity for accurate detection. In addition, current gold standard detection methods, such as PCR, gel electrophoresis, chromatography, microarray, are time-consuming and require trained professionals for analysis. These are important factors that limit the commercialization of salivary kits for disease diagnosis. Therefore, the development of fast, convenient and inexpensive detection methods will also be the focus of future research on saliva diagnosis, which will make saliva diagnosis go further.

Conclusions

As mentioned above, saliva biopsy will bring new opportunities for fluid biopsies. It is expected that with the accumulation of salivomics research data, a salivary biomarker group will be established for most diseases, through which diagnosis will become more and more common. Furthermore, it will also lead to the development of real-time testing and screening tools, providing faster and more effective treatment and patient management. As the researches progress, we believe that evidence will emerge to determine the relationship between various diseases and salivary biomarkers. Most of the evidence may come from large-scale longitudinal studies, and will be applied in the clinic, such as diagnostic testing, therapy management and monitoring. This field is still in the infant. However, salivary biomarker research is receiving rapid attention, and significant progress is expected in the future.


Corresponding authors: Xin Jin, BGI Genomics, BGI-Shenzhen, Shenzhen, P.R. China; and School of Medicine, South China University of Technology, Guangzhou, P.R. China, E-mail: ; and Bo Jia, Department of Oral Surgery, Stomatological Hospital, Southern Medical University, South Jiangnan Road 366, Guangzhou, 510280, P.R. China, E-mail:
Zhijie Huang and Xiaoxia Yang contributed equally to this work.

Award Identifier / Grant number: 2019A1515010408

Funding source: Research and Cultivation Project of Stomatological Hospital, Southern Medical University

Award Identifier / Grant number: PY2020029

  1. Research funding: Bo Jia was supported by the Natural Science Foundation of Guangdong Province, Grant 2019A1515010408. Xiaoxia Yang was supported by the Research and Cultivation Project of Stomatological Hospital, Southern Medical University, Grant PY2020029.

  2. Author contributions: Zhijie Huang, Xiaoxia Yang, Xin Jin, and Bo Jia wrote the manuscript; Yisheng Huang, Zhengming Tang, Yuanxin Chen, Hongyu Liu and Mingshu Huang made the figures and edited the manuscript; Ling Qing, Li Li, Qin Wang and Zhuye Jie helped organize the references; Xin Jin and Bo Jia conceived of and supervised the work. All authors have read and agreed to the published version of the manuscript.

  3. Competing interests: Authors state no conflict of interest.

  4. Informed consent: Not applicable.

  5. Ethical approval: Not applicable.

  6. Data availability: All data in the review can be found in the references, and the associated references can be found in PubMed https://pubmed.ncbi.nlm.nih.gov/.

References

1. Vaidyanathan, R, Soon, RH, Zhang, P, Jiang, K, Lim, CT. Cancer diagnosis: from tumor to liquid biopsy and beyond. Lab Chip 2018;19:11–34. https://doi.org/10.1039/c8lc00684a.Search in Google Scholar PubMed

2. Ye, Q, Ling, S, Zheng, S, Xu, X. Liquid biopsy in hepatocellular carcinoma: circulating tumor cells and circulating tumor DNA. Mol Cancer 2019;18:114. https://doi.org/10.1186/s12943-019-1043-x.Search in Google Scholar PubMed PubMed Central

3. Remon, J, Majem, M. EGFR mutation heterogeneity and mixed response to EGFR tyrosine kinase inhibitors of non small cell lung cancer: a clue to overcoming resistance. Transl Lung Cancer Res 2013;2:445–8.Search in Google Scholar

4. Allott, EH, Geradts, J, Sun, X, Cohen, SM, Zirpoli, GR, Khoury, T, et al.. Intratumoral heterogeneity as a source of discordance in breast cancer biomarker classification. Breast Cancer Res 2016;18:68. https://doi.org/10.1186/s13058-016-0725-1.Search in Google Scholar PubMed PubMed Central

5. Poulet, G, Massias, J, Taly, V. Liquid biopsy: general concepts. Acta Cytol 2019;63:449–55. https://doi.org/10.1159/000499337.Search in Google Scholar PubMed

6. Han, P, Bartold, PM, Salomon, C, Ivanovski, S. Salivary outer membrane vesicles and DNA methylation of small extracellular vesicles as biomarkers for periodontal status: a pilot study. Int J Mol Sci 2021;22:2423. https://doi.org/10.3390/ijms22052423.Search in Google Scholar PubMed PubMed Central

7. Al-Rawi, NH, Al-Marzooq, F, Al-Nuaimi, AS, Hachim, MY, Hamoudi, R. Salivary microRNA 155, 146a/b and 203: a pilot study for potentially non-invasive diagnostic biomarkers of periodontitis and diabetes mellitus. PLoS One 2020;15:e237004. https://doi.org/10.1371/journal.pone.0237004.Search in Google Scholar PubMed PubMed Central

8. Nik, MKN, Awang, R, Mohamad, S, Shahidan, W. Plasma- and saliva exosome profile reveals a distinct microRNA signature in chronic periodontitis. Front Physiol 2020;11:587381. https://doi.org/10.3389/fphys.2020.587381.Search in Google Scholar PubMed PubMed Central

9. Kim, HD, Kim, S, Jeon, S, Kim, SJ, Cho, HJ, Choi, YN. Diagnostic and prognostic ability of salivary MMP-9 and S100A8 for periodontitis. J Clin Periodontol 2020;47:1191–200. https://doi.org/10.1111/jcpe.13349.Search in Google Scholar PubMed

10. Zhang, Y, Kang, N, Xue, F, Qiao, J, Duan, J, Chen, F, et al.. Evaluation of salivary biomarkers for the diagnosis of periodontitis. BMC Oral Health 2021;21:266. https://doi.org/10.1186/s12903-021-01600-5.Search in Google Scholar PubMed PubMed Central

11. Wu, YC, Ning, L, Tu, YK, Huang, CP, Huang, NT, Chen, YF, et al.. Salivary biomarker combination prediction model for the diagnosis of periodontitis in a Taiwanese population. J Formos Med Assoc 2018;117:841–8. https://doi.org/10.1016/j.jfma.2017.10.004.Search in Google Scholar PubMed

12. Inonu, E, Kayis, SA, Eskan, MA, Hakki, SS. Salivary Del-1, IL-17, and LFA-1 levels in periodontal health and disease. J Periodontal Res 2020;55:511–8. https://doi.org/10.1111/jre.12738.Search in Google Scholar PubMed

13. Lundmark, A, Johannsen, G, Eriksson, K, Kats, A, Jansson, L, Tervahartiala, T, et al.. Mucin 4 and matrix metalloproteinase 7 as novel salivary biomarkers for periodontitis. J Clin Periodontol 2017;44:247–54. https://doi.org/10.1111/jcpe.12670.Search in Google Scholar PubMed PubMed Central

14. Grande, MA, Belstrom, D, Damgaard, C, Holmstrup, P, Thangaraj, SS, Nielsen, CH, et al.. Complement split product C3c in saliva as biomarker for periodontitis and response to periodontal treatment. J Periodontal Res 2021;56:27–33. https://doi.org/10.1111/jre.12788.Search in Google Scholar PubMed PubMed Central

15. Sanchez, GA, Miozza, VA, Delgado, A, Busch, L. Salivary IL-1beta and PGE2 as biomarkers of periodontal status, before and after periodontal treatment. J Clin Periodontol 2013;40:1112–7. https://doi.org/10.1111/jcpe.12164.Search in Google Scholar PubMed

16. Ebersole, JL, Nagarajan, R, Akers, D, Miller, CS. Targeted salivary biomarkers for discrimination of periodontal health and disease(s). Front Cell Infect Microbiol 2015;5:62. https://doi.org/10.3389/fcimb.2015.00062.Search in Google Scholar PubMed PubMed Central

17. Bostanci, N, Selevsek, N, Wolski, W, Grossmann, J, Bao, K, Wahlander, A, et al.. Targeted proteomics guided by label-free quantitative proteome analysis in saliva reveal transition signatures from health to periodontal disease. Mol Cell Proteomics 2018;17:1392–409. https://doi.org/10.1074/mcp.ra118.000718.Search in Google Scholar

18. Kuboniwa, M, Sakanaka, A, Hashino, E, Bamba, T, Fukusaki, E, Amano, A. Prediction of periodontal inflammation via metabolic profiling of saliva. J Dent Res 2016;95:1381–6. https://doi.org/10.1177/0022034516661142.Search in Google Scholar PubMed

19. Kim, EH, Kim, S, Kim, HJ, Jeong, HO, Lee, J, Jang, J, et al.. Prediction of chronic periodontitis severity using machine learning models based on salivary bacterial copy number. Front Cell Infect Microbiol 2020;10:571515. https://doi.org/10.3389/fcimb.2020.571515.Search in Google Scholar PubMed PubMed Central

20. Diao, J, Yuan, C, Tong, P, Ma, Z, Sun, X, Zheng, S. Potential roles of the free salivary microbiome dysbiosis in periodontal diseases. Front Cell Infect Microbiol 2021;11:711282. https://doi.org/10.3389/fcimb.2021.711282.Search in Google Scholar PubMed PubMed Central

21. Sembler-Moller, ML, Belstrom, D, Locht, H, Pedersen, A. Distinct microRNA expression profiles in saliva and salivary gland tissue differentiate patients with primary Sjogren’s syndrome from non-Sjogren’s sicca patients. J Oral Pathol Med 2020;49:1044–52. https://doi.org/10.1111/jop.13099.Search in Google Scholar PubMed

22. Hu, S, Gao, K, Pollard, R, Arellano-Garcia, M, Zhou, H, Zhang, L, et al.. Preclinical validation of salivary biomarkers for primary Sjogren’s syndrome. Arthritis Care Res 2010;62:1633–8. https://doi.org/10.1002/acr.20289.Search in Google Scholar PubMed PubMed Central

23. Sembler-Moller, ML, Belstrom, D, Locht, H, Pedersen, A. Proteomics of saliva, plasma, and salivary gland tissue in Sjogren’s syndrome and non-Sjogren patients identify novel biomarker candidates. J Proteonomics 2020;225:103877. https://doi.org/10.1016/j.jprot.2020.103877.Search in Google Scholar PubMed

24. Lee, J, Lee, J, Baek, S, Koh, JH, Kim, JW, Kim, SY, et al.. Soluble siglec-5 is a novel salivary biomarker for primary Sjogren’s syndrome. J Autoimmun 2019;100:114–9. https://doi.org/10.1016/j.jaut.2019.03.008.Search in Google Scholar PubMed

25. Sembler-Moller, ML, Belstrom, D, Locht, H, Pedersen, A. Combined serum anti-SSA/Ro and salivary TRIM29 reveals promising high diagnostic accuracy in patients with primary Sjogren’s syndrome. PLoS One 2021;16:e258428. https://doi.org/10.1371/journal.pone.0258428.Search in Google Scholar PubMed PubMed Central

26. Mehdipour, M, Shahidi, M, Manifar, S, Jafari, S, Mashhadi, AF, Barati, M, et al.. Diagnostic and prognostic relevance of salivary microRNA-21, -125a, -31 and -200a levels in patients with oral lichen planus - a short report. Cell Oncol 2018;41:329–34. https://doi.org/10.1007/s13402-018-0372-x.Search in Google Scholar PubMed

27. Stasio, DD, Mosca, L, Lucchese, A, Cave, DD, Kawasaki, H, Lombardi, A, et al.. Salivary mir-27b expression in oral lichen planus patients: a series of cases and a narrative review of literature. Curr Top Med Chem 2019;19:2816–23. https://doi.org/10.2174/1568026619666191121144407.Search in Google Scholar PubMed

28. Liu, WZ, He, MJ, Long, L, Mu, DL, Xu, MS, Xing, X, et al.. Interferon-gamma and interleukin-4 detected in serum and saliva from patients with oral lichen planus. Int J Oral Sci 2014;6:22–6. https://doi.org/10.1038/ijos.2013.74.Search in Google Scholar PubMed PubMed Central

29. Carvalho, M, Cavalieri, D, Do, NS, Lourenco, T, Ramos, D, Pasqualin, D, et al.. Cytokines levels and salivary microbiome play a potential role in oral lichen planus diagnosis. Sci Rep 2019;9:18137. https://doi.org/10.1038/s41598-019-54615-y.Search in Google Scholar PubMed PubMed Central

30. Shahidi, M, Jafari, S, Barati, M, Mahdipour, M, Gholami, MS. Predictive value of salivary microRNA-320a, vascular endothelial growth factor receptor 2, CRP and IL-6 in oral lichen planus progression. Inflammopharmacology 2017;25:577–83. https://doi.org/10.1007/s10787-017-0352-1.Search in Google Scholar PubMed

31. Srinivasan, M, Kodumudi, KN, Zunt, SL. Soluble CD14 and toll-like receptor-2 are potential salivary biomarkers for oral lichen planus and burning mouth syndrome. Clin Immunol 2008;126:31–7. https://doi.org/10.1016/j.clim.2007.08.014.Search in Google Scholar PubMed

32. Sivadasan, P, Gupta, MK, Sathe, G, Sudheendra, HV, Sunny, SP, Renu, D, et al.. Salivary proteins from dysplastic leukoplakia and oral squamous cell carcinoma and their potential for early detection. J Proteonomics 2020;212:103574. https://doi.org/10.1016/j.jprot.2019.103574.Search in Google Scholar PubMed

33. Sharma, M, Bairy, I, Pai, K, Satyamoorthy, K, Prasad, S, Berkovitz, B, et al.. Salivary IL-6 levels in oral leukoplakia with dysplasia and its clinical relevance to tobacco habits and periodontitis. Clin Oral Invest 2011;15:705–14. https://doi.org/10.1007/s00784-010-0435-5.Search in Google Scholar PubMed

34. Romani, C, Salviato, E, Paderno, A, Zanotti, L, Ravaggi, A, Deganello, A, et al.. Genome-wide study of salivary miRNAs identifies miR-423-5p as promising diagnostic and prognostic biomarker in oral squamous cell carcinoma. Theranostics 2021;11:2987–99. https://doi.org/10.7150/thno.45157.Search in Google Scholar PubMed PubMed Central

35. Oh, SY, Kang, SM, Kang, SH, Lee, HJ, Kwon, TG, Kim, JW, et al.. Potential salivary mRNA biomarkers for early detection of oral cancer. J Clin Med 2020;9:243. https://doi.org/10.3390/jcm9010243.Search in Google Scholar PubMed PubMed Central

36. Li, Y, St, JM, Zhou, X, Kim, Y, Sinha, U, Jordan, RC, et al.. Salivary transcriptome diagnostics for oral cancer detection. Clin Cancer Res 2004;10:8442–50. https://doi.org/10.1158/1078-0432.ccr-04-1167.Search in Google Scholar

37. Ueda, S, Goto, M, Hashimoto, K, Hasegawa, S, Imazawa, M, Takahashi, M, et al.. Salivary CCL20 level as a biomarker for oral squamous cell carcinoma. Cancer Genomics Proteomics 2021;18:103–12. https://doi.org/10.21873/cgp.20245.Search in Google Scholar PubMed PubMed Central

38. Mehterov, N, Vladimirov, B, Sacconi, A, Pulito, C, Rucinski, M, Blandino, G, et al.. Salivary miR-30c-5p as potential biomarker for detection of oral squamous cell carcinoma. Biomedicines 2021;9:1079. https://doi.org/10.3390/biomedicines9091079.Search in Google Scholar PubMed PubMed Central

39. He, L, Ping, F, Fan, Z, Zhang, C, Deng, M, Cheng, B, et al.. Salivary exosomal miR-24-3p serves as a potential detective biomarker for oral squamous cell carcinoma screening. Biomed Pharmacother 2020;121:109553. https://doi.org/10.1016/j.biopha.2019.109553.Search in Google Scholar PubMed

40. Gai, C, Camussi, F, Broccoletti, R, Gambino, A, Cabras, M, Molinaro, L, et al.. Salivary extracellular vesicle-associated miRNAs as potential biomarkers in oral squamous cell carcinoma. BMC Cancer 2018;18:439. https://doi.org/10.1186/s12885-018-4364-z.Search in Google Scholar PubMed PubMed Central

41. Momen-Heravi, F, Trachtenberg, AJ, Kuo, WP, Cheng, YS. Genomewide study of salivary microRNAs for detection of oral cancer. J Dent Res 2014;93(7 Suppl):86S–93S. https://doi.org/10.1177/0022034514531018.Search in Google Scholar PubMed PubMed Central

42. Marton, IJ, Horvath, J, Labiscsak, P, Markus, B, Dezso, B, Szabo, A, et al.. Salivary IL-6 mRNA is a robust biomarker in oral squamous cell carcinoma. J Clin Med 2019;8:1958. https://doi.org/10.3390/jcm8111958.Search in Google Scholar PubMed PubMed Central

43. Feng, Y, Li, Q, Chen, J, Yi, P, Xu, X, Fan, Y, et al.. Salivary protease spectrum biomarkers of oral cancer. Int J Oral Sci 2019;11:7. https://doi.org/10.1038/s41368-018-0032-z.Search in Google Scholar PubMed PubMed Central

44. Jain, A, Kotimoole, CN, Ghoshal, S, Bakshi, J, Chatterjee, A, Prasad, T, et al.. Identification of potential salivary biomarker panels for oral squamous cell carcinoma. Sci Rep 2021;11:3365. https://doi.org/10.1038/s41598-021-82635-0.Search in Google Scholar PubMed PubMed Central

45. Kang, Y, Chen, J, Li, X, Luo, M, Chen, H, Cui, B, et al.. Salivary KLK5 and uPA are potential biomarkers for malignant transformation of OLK and OLP. Cancer Biomarkers 2021;31:317–28. https://doi.org/10.3233/cbm-203105.Search in Google Scholar PubMed

46. Heawchaiyaphum, C, Pientong, C, Phusingha, P, Vatanasapt, P, Promthet, S, Daduang, J, et al.. Peroxiredoxin-2 and zinc-alpha-2-glycoprotein as potentially combined novel salivary biomarkers for early detection of oral squamous cell carcinoma using proteomic approaches. J Proteonomics 2018;173:52–61. https://doi.org/10.1016/j.jprot.2017.11.022.Search in Google Scholar PubMed

47. Dikova, V, Jantus-Lewintre, E, Bagan, J. Potential non-invasive biomarkers for early diagnosis of oral squamous cell carcinoma. J Clin Med 2021;10:1658. https://doi.org/10.3390/jcm10081658.Search in Google Scholar PubMed PubMed Central

48. Hu, S, Arellano, M, Boontheung, P, Wang, J, Zhou, H, Jiang, J, et al.. Salivary proteomics for oral cancer biomarker discovery. Clin Cancer Res 2008;14:6246–52. https://doi.org/10.1158/1078-0432.ccr-07-5037.Search in Google Scholar

49. Gleber-Netto, FO, Yakob, M, Li, F, Feng, Z, Dai, J, Kao, HK, et al.. Salivary biomarkers for detection of oral squamous cell carcinoma in a Taiwanese population. Clin Cancer Res 2016;22:3340–7. https://doi.org/10.1158/1078-0432.ccr-15-1761.Search in Google Scholar PubMed PubMed Central

50. Shan, J, Sun, Z, Yang, J, Xu, J, Shi, W, Wu, Y, et al.. Discovery and preclinical validation of proteomic biomarkers in saliva for early detection of oral squamous cell carcinomas. Oral Dis 2019;25:97–107. https://doi.org/10.1111/odi.12971.Search in Google Scholar PubMed

51. Yu, JS, Chen, YT, Chiang, WF, Hsiao, YC, Chu, LJ, See, LC, et al.. Saliva protein biomarkers to detect oral squamous cell carcinoma in a high-risk population in Taiwan. Proc Natl Acad Sci USA 2016;113:11549–54. https://doi.org/10.1073/pnas.1612368113.Search in Google Scholar PubMed PubMed Central

52. Chen, YT, Chen, HW, Wu, CF, Chu, LJ, Chiang, WF, Wu, CC, et al.. Development of a multiplexed liquid chromatography multiple-reaction-monitoring mass spectrometry (LC-MRM/MS) method for evaluation of salivary proteins as oral cancer biomarkers. Mol Cell Proteomics 2017;16:799–811. https://doi.org/10.1074/mcp.m116.064758.Search in Google Scholar PubMed PubMed Central

53. Zheng, J, Sun, L, Yuan, W, Xu, J, Yu, X, Wang, F, et al.. Clinical value of Naa10p and CEA levels in saliva and serum for diagnosis of oral squamous cell carcinoma. J Oral Pathol Med 2018;47:830–5. https://doi.org/10.1111/jop.12767.Search in Google Scholar PubMed

54. Jou, YJ, Lin, CD, Lai, CH, Chen, CH, Kao, JY, Chen, SY, et al.. Proteomic identification of salivary transferrin as a biomarker for early detection of oral cancer. Anal Chim Acta 2010;681:41–8. https://doi.org/10.1016/j.aca.2010.09.030.Search in Google Scholar PubMed

55. Ghallab, NA, Shaker, OG. Serum and salivary levels of chemerin and MMP-9 in oral squamous cell carcinoma and oral premalignant lesions. Clin Oral Invest 2017;21:937–47. https://doi.org/10.1007/s00784-016-1846-8.Search in Google Scholar PubMed

56. Chu, HW, Chang, KP, Hsu, CW, Chang, IY, Liu, HP, Chen, YT, et al.. Identification of salivary biomarkers for oral cancer detection with untargeted and targeted quantitative proteomics approaches. Mol Cell Proteomics 2019;18:1796–806. https://doi.org/10.1074/mcp.ra119.001530.Search in Google Scholar PubMed PubMed Central

57. Chang, YT, Chu, LJ, Liu, YC, Chen, CJ, Wu, SF, Chen, CH, et al.. Verification of saliva matrix metalloproteinase-1 as a strong diagnostic marker of oral cavity cancer. Cancers 2020;12:2273. https://doi.org/10.3390/cancers12082273.Search in Google Scholar PubMed PubMed Central

58. Hsu, CW, Yu, JS, Peng, PH, Liu, SC, Chang, YS, Chang, KP, et al.. Secretome profiling of primary cells reveals that THBS2 is a salivary biomarker of oral cavity squamous cell carcinoma. J Proteome Res 2014;13:4796–807. https://doi.org/10.1021/pr500038k.Search in Google Scholar PubMed

59. Ishikawa, S, Wong, D, Sugimoto, M, Gleber-Netto, FO, Li, F, Tu, M, et al.. Identification of salivary metabolites for oral squamous cell carcinoma and oral epithelial dysplasia screening from persistent suspicious oral mucosal lesions. Clin Oral Invest 2019;23:3557–63. https://doi.org/10.1007/s00784-018-2777-3.Search in Google Scholar PubMed

60. Song, X, Yang, X, Narayanan, R, Shankar, V, Ethiraj, S, Wang, X, et al.. Oral squamous cell carcinoma diagnosed from saliva metabolic profiling. Proc Natl Acad Sci USA 2020;117:16167–73. https://doi.org/10.1073/pnas.2001395117.Search in Google Scholar PubMed PubMed Central

61. 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. https://doi.org/10.1111/odi.13209.Search in Google Scholar PubMed

62. Wang, Q, Gao, P, Wang, X, Duan, Y. Investigation and identification of potential biomarkers in human saliva for the early diagnosis of oral squamous cell carcinoma. Clin Chim Acta 2014;427:79–85. https://doi.org/10.1016/j.cca.2013.10.004.Search in Google Scholar PubMed

63. Wang, Q, Gao, P, Cheng, F, Wang, X, Duan, Y. Measurement of salivary metabolite biomarkers for early monitoring of oral cancer with ultra performance liquid chromatography-mass spectrometry. Talanta 2014;119:299–305. https://doi.org/10.1016/j.talanta.2013.11.008.Search in Google Scholar PubMed

64. Wang, Q, Gao, P, Wang, X, Duan, Y. The early diagnosis and monitoring of squamous cell carcinoma via saliva metabolomics. Sci Rep 2014;4:6802. https://doi.org/10.1038/srep06802.Search in Google Scholar PubMed PubMed Central

65. Zhou, X, Hao, Y, Peng, X, Li, B, Han, Q, Ren, B, et al.. The Clinical potential of oral microbiota as a screening tool for oral squamous cell carcinomas. Front Cell Infect Microbiol 2021;11:728933. https://doi.org/10.3389/fcimb.2021.728933.Search in Google Scholar PubMed PubMed Central

66. Xie, Z, Yin, X, Gong, B, Nie, W, Wu, B, Zhang, X, et al.. Salivary microRNAs show potential as a noninvasive biomarker for detecting resectable pancreatic cancer. Cancer Prev Res 2015;8:165–73. https://doi.org/10.1158/1940-6207.capr-14-0192.Search in Google Scholar PubMed

67. Zhang, L, Farrell, JJ, Zhou, H, Elashoff, D, Akin, D, Park, NH, et al.. Salivary transcriptomic biomarkers for detection of resectable pancreatic cancer. Gastroenterology 2010;138:949–57. https://doi.org/10.1053/j.gastro.2009.11.010.Search in Google Scholar PubMed PubMed Central

68. Humeau, M, Vignolle-Vidoni, A, Sicard, F, Martins, F, Bournet, B, Buscail, L, et al.. Salivary microRNA in pancreatic cancer patients. PLoS One 2015;10:e130996. https://doi.org/10.1371/journal.pone.0130996.Search in Google Scholar PubMed PubMed Central

69. Deutsch, O, Haviv, Y, Krief, G, Keshet, N, Westreich, R, Stemmer, SM, et al.. Possible proteomic biomarkers for the detection of pancreatic cancer in oral fluids. Sci Rep 2020;10:21995. https://doi.org/10.1038/s41598-020-78922-x.Search in Google Scholar PubMed PubMed Central

70. Asai, Y, Itoi, T, Sugimoto, M, Sofuni, A, Tsuchiya, T, Tanaka, R, et al.. Elevated polyamines in saliva of pancreatic cancer. Cancers 2018;10:43. https://doi.org/10.3390/cancers10020043.Search in Google Scholar PubMed PubMed Central

71. Sugimoto, M, Wong, DT, Hirayama, A, Soga, T, Tomita, M. Capillary electrophoresis mass spectrometry-based saliva metabolomics identified oral, breast and pancreatic cancer-specific profiles. Metabolomics 2010;6:78–95. https://doi.org/10.1007/s11306-009-0178-y.Search in Google Scholar PubMed PubMed Central

72. Farrell, JJ, Zhang, L, Zhou, H, Chia, D, Elashoff, D, Akin, D, et al.. Variations of oral microbiota are associated with pancreatic diseases including pancreatic cancer. Gut 2012;61:582–8. https://doi.org/10.1136/gutjnl-2011-300784.Search in Google Scholar PubMed PubMed Central

73. Li, F, Yoshizawa, JM, Kim, KM, Kanjanapangka, J, Grogan, TR, Wang, X, et al.. Discovery and validation of salivary extracellular RNA biomarkers for noninvasive detection of gastric cancer. Clin Chem 2018;64:1513–21. https://doi.org/10.1373/clinchem.2018.290569.Search in Google Scholar PubMed PubMed Central

74. Xiao, H, Zhang, Y, Kim, Y, Kim, S, Kim, JJ, Kim, KM, et al.. Differential proteomic analysis of human saliva using tandem mass tags quantification for gastric cancer detection. Sci Rep 2016;6:22165. https://doi.org/10.1038/srep22165.Search in Google Scholar PubMed PubMed Central

75. Zhang, L, Xiao, H, Karlan, S, Zhou, H, Gross, J, Elashoff, D, et al.. Discovery and preclinical validation of salivary transcriptomic and proteomic biomarkers for the non-invasive detection of breast cancer. PLoS One 2010;5:e15573. https://doi.org/10.1371/journal.pone.0015573.Search in Google Scholar PubMed PubMed Central

76. Lopez-Jornet, P, Aznar, C, Ceron, J, Asta, T. Salivary biomarkers in breast cancer: a cross-sectional study. Support Care Cancer 2021;29:889–96. https://doi.org/10.1007/s00520-020-05561-3.Search in Google Scholar PubMed

77. Liu, X, Yu, H, Qiao, Y, Yang, J, Shu, J, Zhang, J, et al.. Salivary glycopatterns as potential biomarkers for screening of early-stage breast cancer. EBioMedicine 2018;28:70–9. https://doi.org/10.1016/j.ebiom.2018.01.026.Search in Google Scholar PubMed PubMed Central

78. Brooks, MN, Wang, J, Li, Y, Zhang, R, Elashoff, D, Wong, DT. Salivary protein factors are elevated in breast cancer patients. Mol Med Rep 2008;1:375–8.10.3892/mmr.1.3.375Search in Google Scholar

79. Cheng, F, Wang, Z, Huang, Y, Duan, Y, Wang, X. Investigation of salivary free amino acid profile for early diagnosis of breast cancer with ultra performance liquid chromatography-mass spectrometry. Clin Chim Acta 2015;447:23–31. https://doi.org/10.1016/j.cca.2015.05.008.Search in Google Scholar PubMed

80. Hernandez-Arteaga, AC, de Jesus, ZJ, Martinez-Martinez, MU, Hernandez-Cedillo, A, Ojeda-Galvan, HJ, Jose-Yacaman, M, et al.. Determination of salivary sialic acid through nanotechnology: a useful biomarker for the screening of breast cancer. Arch Med Res 2019;50:105–10. https://doi.org/10.1016/j.arcmed.2019.05.013.Search in Google Scholar PubMed

81. Xavier, AD, Acevedo, AC, Cancado, PM, Costa, NA, Pichon, V, Chardin, H, et al.. Using an untargeted metabolomics approach to identify salivary metabolites in women with breast cancer. Metabolites 2020;10:506. https://doi.org/10.3390/metabo10120506.Search in Google Scholar PubMed PubMed Central

82. Murata, T, Yanagisawa, T, Kurihara, T, Kaneko, M, Ota, S, Enomoto, A, et al.. Salivary metabolomics with alternative decision tree-based machine learning methods for breast cancer discrimination. Breast Cancer Res Treat 2019;177:591–601. https://doi.org/10.1007/s10549-019-05330-9.Search in Google Scholar PubMed

83. Zhong, L, Cheng, F, Lu, X, Duan, Y, Wang, X. Untargeted saliva metabonomics study of breast cancer based on ultra performance liquid chromatography coupled to mass spectrometry with HILIC and RPLC separations. Talanta 2016;158:351–60. https://doi.org/10.1016/j.talanta.2016.04.049.Search in Google Scholar PubMed

84. Wei, F, Lin, CC, Joon, A, Feng, Z, Troche, G, Lira, ME, et al.. Noninvasive saliva-based EGFR gene mutation detection in patients with lung cancer. Am J Respir Crit Care Med 2014;190:1117–26. https://doi.org/10.1164/rccm.201406-1003oc.Search in Google Scholar PubMed PubMed Central

85. Zhang, L, Xiao, H, Zhou, H, Santiago, S, Lee, JM, Garon, EB, et al.. Development of transcriptomic biomarker signature in human saliva to detect lung cancer. Cell Mol Life Sci 2012;69:3341–50. https://doi.org/10.1007/s00018-012-1027-0.Search in Google Scholar PubMed PubMed Central

86. Xiao, H, Zhang, L, Zhou, H, Lee, JM, Garon, EB, Wong, DT. Proteomic analysis of human saliva from lung cancer patients using two-dimensional difference gel electrophoresis and mass spectrometry. Mol Cell Proteomics 2012;11:M111–12112. https://doi.org/10.1074/mcp.m111.012112.Search in Google Scholar

87. Koizumi, T, Shetty, V, Yamaguchi, M. Salivary cytokine panel indicative of non-small cell lung cancer. J Int Med Res 2018;46:3570–82. https://doi.org/10.1177/0300060518775563.Search in Google Scholar PubMed PubMed Central

88. Bel’Skaya, LV, Sarf, EA, Kosenok, VK, Gundyrev, IA. Biochemical markers of saliva in lung cancer: diagnostic and prognostic perspectives. Diagnostics 2020;10:186. https://doi.org/10.3390/diagnostics10040186.Search in Google Scholar PubMed PubMed Central

89. Yan, X, Yang, M, Liu, J, Gao, R, Hu, J, Li, J, et al.. Discovery and validation of potential bacterial biomarkers for lung cancer. Am J Cancer Res 2015;5:3111–22.Search in Google Scholar

90. Zhang, W, Luo, J, Dong, X, Zhao, S, Hao, Y, Peng, C, 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;10:1651–62. https://doi.org/10.7150/jca.28077.Search in Google Scholar PubMed PubMed Central

91. Ebersole, JL, Kryscio, RJ, Campbell, C, Kinane, DF, McDevitt, J, Christodoulides, N, et al.. Salivary and serum adiponectin and C-reactive protein levels in acute myocardial infarction related to body mass index and oral health. J Periodontal Res 2017;52:419–27. https://doi.org/10.1111/jre.12406.Search in Google Scholar PubMed PubMed Central

92. Mirzaii-Dizgah, I, Hejazi, SF, Riahi, E, Salehi, MM. Saliva-based creatine kinase MB measurement as a potential point-of-care testing for detection of myocardial infarction. Clin Oral Invest 2012;16:775–9. https://doi.org/10.1007/s00784-011-0578-z.Search in Google Scholar PubMed

93. Mirzaii-Dizgah, I, Riahi, E. Salivary troponin I as an indicator of myocardial infarction. Indian J Med Res 2013;138:861–5.Search in Google Scholar

94. Cabras, T, Pisano, E, Mastinu, A, Denotti, G, Pusceddu, PP, Inzitari, R, et al.. Alterations of the salivary secretory peptidome profile in children affected by type 1 diabetes. Mol Cell Proteomics 2010;9:2099–108. https://doi.org/10.1074/mcp.m110.001057.Search in Google Scholar

95. Jian, C, Zhao, A, Ma, X, Ge, K, Lu, W, Zhu, W, et al.. Diabetes screening: detection and application of saliva 1, 5-anhydroglucitol by liquid chromatography-mass spectrometry. J Clin Endocrinol Metab 2020;105:114. https://doi.org/10.1210/clinem/dgaa114.Search in Google Scholar PubMed

96. Giusti, L, Baldini, C, Ciregia, F, Giannaccini, G, Giacomelli, C, De Feo, F, et al.. Is GRP78/BiP a potential salivary biomarker in patients with rheumatoid arthritis? Proteonomics Clin Appl 2010;4:315–24. https://doi.org/10.1002/prca.200900082.Search in Google Scholar PubMed

97. Bermejo-Pareja, F, Del, ST, Valenti, M, de la Fuente, M, Bartolome, F, Carro, E. Salivary lactoferrin as biomarker for Alzheimer’s disease: brain-immunity interactions. Alzheimers Dement 2020;16:1196–204. https://doi.org/10.1002/alz.12107.Search in Google Scholar PubMed PubMed Central

98. Lee, M, Guo, JP, Kennedy, K, McGeer, EG, McGeer, PL. A method for diagnosing Alzheimer’s disease based on salivary amyloid-beta protein 42 levels. J Alzheimers Dis 2017;55:1175–82. https://doi.org/10.3233/jad-160748.Search in Google Scholar

99. Shi, M, Sui, YT, Peskind, ER, Li, G, Hwang, H, Devic, I, et al.. Salivary tau species are potential biomarkers of Alzheimer’s disease. J Alzheimers Dis 2011;27:299–305. https://doi.org/10.3233/jad-2011-110731.Search in Google Scholar

100. Guo, H, Li, B, Yao, H, Liu, D, Chen, R, Zhou, S, et al.. Profiling the oral microbiomes in patients with Alzheimer’s disease. Oral Dis 2021. https://doi.org/10.1111/odi.14110. In press.Search in Google Scholar PubMed

101. Cressatti, M, Juwara, L, Galindez, JM, Velly, AM, Nkurunziza, ES, Marier, S, et al.. Salivary microR-153 and microR-223 Levels as potential diagnostic biomarkers of idiopathic Parkinson’s disease. Mov Disord 2020;35:468–77. https://doi.org/10.1002/mds.27935.Search in Google Scholar PubMed

102. Devic, I, Hwang, H, Edgar, JS, Izutsu, K, Presland, R, Pan, C, et al.. Salivary alpha-synuclein and DJ-1: potential biomarkers for Parkinson’s disease. Brain 2011;134:e178. https://doi.org/10.1093/brain/awr015.Search in Google Scholar PubMed PubMed Central

103. Song, W, Kothari, V, Velly, AM, Cressatti, M, Liberman, A, Gornitsky, M, et al.. Evaluation of salivary heme oxygenase-1 as a potential biomarker of early Parkinson’s disease. Mov Disord 2018;33:583–91. https://doi.org/10.1002/mds.27328.Search in Google Scholar PubMed

104. Takai, N, Yamaguchi, M, Aragaki, T, Eto, K, Uchihashi, K, Nishikawa, Y. Effect of psychological stress on the salivary cortisol and amylase levels in healthy young adults. Arch Oral Biol 2004;49:963–8. https://doi.org/10.1016/j.archoralbio.2004.06.007.Search in Google Scholar PubMed

105. Yonekura, T, Takeda, K, Shetty, V, Yamaguchi, M. Relationship between salivary cortisol and depression in adolescent survivors of a major natural disaster. J Physiol Sci 2014;64:261–7. https://doi.org/10.1007/s12576-014-0315-x.Search in Google Scholar PubMed PubMed Central

106. Engeland, CG, Hugo, FN, Hilgert, JB, Nascimento, GG, Junges, R, Lim, HJ, et al.. Psychological distress and salivary secretory immunity. Brain Behav Immun 2016;52:11–7. https://doi.org/10.1016/j.bbi.2015.08.017.Search in Google Scholar PubMed PubMed Central

107. Hek, K, Direk, N, Newson, RS, Hofman, A, Hoogendijk, WJ, Mulder, CL, et al.. Anxiety disorders and salivary cortisol levels in older adults: a population-based study. Psychoneuroendocrinology 2013;38:300–5. https://doi.org/10.1016/j.psyneuen.2012.06.006.Search in Google Scholar PubMed

108. Mathai, RA, Vidya, R, Reddy, BS, Thomas, L, Udupa, K, Kolesar, J, et al.. Potential utility of liquid biopsy as a diagnostic and prognostic tool for the assessment of solid tumors: implications in the precision oncology. J Clin Med 2019;8:373. https://doi.org/10.3390/jcm8030373.Search in Google Scholar PubMed PubMed Central

109. Mader, S, Pantel, K. Liquid biopsy: current status and future perspectives. Oncol Res Treat 2017;40:404–8. https://doi.org/10.1159/000478018.Search in Google Scholar PubMed

110. Zhang, CZ, Cheng, XQ, Li, JY, Zhang, P, Yi, P, Xu, X, et al.. Saliva in the diagnosis of diseases. Int J Oral Sci 2016;8:133–7. https://doi.org/10.1038/ijos.2016.38.Search in Google Scholar PubMed PubMed Central

111. Nijakowski, K, Surdacka, A. Salivary biomarkers for diagnosis of inflammatory bowel diseases: a systematic review. Int J Oral Sci 2020;21:7477.10.3390/ijms21207477Search in Google Scholar PubMed PubMed Central

112. Segal, A, Wong, DT. Salivary diagnostics: enhancing disease detection and making medicine better. Eur J Dent Educ 2008;12(1 Suppl):22–9. https://doi.org/10.1111/j.1600-0579.2007.00477.x.Search in Google Scholar PubMed PubMed Central

113. Setti, G, Pezzi, ME, Viani, MV, Pertinhez, TA, Cassi, D, Magnoni, C, et al.. Salivary microRNA for diagnosis of cancer and systemic diseases: a systematic review. Int J Mol Sci 2020;21:907. https://doi.org/10.3390/ijms21030907.Search in Google Scholar PubMed PubMed Central

114. Ponti, G, Manfredini, M, Tomasi, A. Non-blood sources of cell-free DNA for cancer molecular profiling in clinical pathology and oncology. Crit Rev Oncol Hematol 2019;141:36–42. https://doi.org/10.1016/j.critrevonc.2019.06.005.Search in Google Scholar PubMed

115. Wong, DT. Towards a simple, saliva-based test for the detection of oral cancer ’oral fluid (saliva), which is the mirror of the body, is a perfect medium to be explored for health and disease surveillance. Expert Rev Mol Diagn 2006;6:267–72. https://doi.org/10.1586/14737159.6.3.267.Search in Google Scholar PubMed

116. Kaczor-Urbanowicz, KE, Wei, F, Rao, SL, Kim, J, Shin, H, Cheng, J, et al.. Clinical validity of saliva and novel technology for cancer detection. Biochim Biophys Acta Rev Cancer 2019;1872:49–59. https://doi.org/10.1016/j.bbcan.2019.05.007.Search in Google Scholar PubMed PubMed Central

117. Lamster, IB, Ahlo, JK. Analysis of gingival crevicular fluid as applied to the diagnosis of oral and systemic diseases. Ann N Y Acad Sci 2007;1098:216–29. https://doi.org/10.1196/annals.1384.027.Search in Google Scholar PubMed

118. Roussa, E. Channels and transporters in salivary glands. Cell Tissue Res 2011;343:263–87. https://doi.org/10.1007/s00441-010-1089-y.Search in Google Scholar PubMed

119. Mese, H, Matsuo, R. Salivary secretion, taste and hyposalivation. J Oral Rehabil 2007;34:711–23. https://doi.org/10.1111/j.1365-2842.2007.01794.x.Search in Google Scholar PubMed

120. Walsh, NP, Montague, JC, Callow, N, Rowlands, AV. Saliva flow rate, total protein concentration and osmolality as potential markers of whole body hydration status during progressive acute dehydration in humans. Arch Oral Biol 2004;49:149–54. https://doi.org/10.1016/j.archoralbio.2003.08.001.Search in Google Scholar PubMed

121. Stookey, GK. The effect of saliva on dental caries. J Am Dent Assoc 2008;139 Suppl:11S–17S. https://doi.org/10.14219/jada.archive.2008.0347.Search in Google Scholar PubMed

122. Vining, RF, McGinley, RA, Symons, RG. Hormones in saliva: mode of entry and consequent implications for clinical interpretation. Clin Chem 1983;29:1752–6. https://doi.org/10.1093/clinchem/29.10.1752.Search in Google Scholar

123. Cone, EJ, Huestis, MA. Interpretation of oral fluid tests for drugs of abuse. Ann N Y Acad Sci 2007;1098:51–103. https://doi.org/10.1196/annals.1384.037.Search in Google Scholar PubMed PubMed Central

124. Wang, J, Liang, Y, Wang, Y, Cui, J, Liu, M, Du, W, et al.. Computational prediction of human salivary proteins from blood circulation and application to diagnostic biomarker identification. PLoS One 2013;8:e80211. https://doi.org/10.1371/journal.pone.0080211.Search in Google Scholar PubMed PubMed Central

125. Konttinen, YT, Stegaev, V, Mackiewicz, Z, Porola, P, Hanninen, A, Szodoray, P. Salivary glands – ‘an unisex organ’? Oral Dis 2010;16:577–85. https://doi.org/10.1111/j.1601-0825.2010.01669.x.Search in Google Scholar PubMed

126. Kubagawa, H, Bertoli, LF, Barton, JC, Koopman, WJ, Mestecky, J, Cooper, MD. Analysis of paraprotein transport into the saliva by using anti-idiotype antibodies. J Immunol 1987;138:435–9.10.4049/jimmunol.138.2.435Search in Google Scholar

127. Carpenter, GH, Proctor, GB, Anderson, LC, Zhang, XS, Garrett, JR. Immunoglobulin A secretion into saliva during dual sympathetic and parasympathetic nerve stimulation of rat submandibular glands. Exp Physiol 2000;85:281–6. https://doi.org/10.1111/j.1469-445x.2000.01968.x.Search in Google Scholar

128. Pfaffe, T, Cooper-White, J, Beyerlein, P, Kostner, K, Punyadeera, C. Diagnostic potential of saliva: current state and future applications. Clin Chem 2011;57:675–87. https://doi.org/10.1373/clinchem.2010.153767.Search in Google Scholar PubMed

129. Park, NJ, Li, Y, Yu, T, Brinkman, BM, Wong, DT. Characterization of RNA in saliva. Clin Chem 2006;52:988–94. https://doi.org/10.1373/clinchem.2005.063206.Search in Google Scholar PubMed PubMed Central

130. Yoshizawa, JM, Schafer, CA, Schafer, JJ, Farrell, JJ, Paster, BJ, Wong, DT. Salivary biomarkers: toward future clinical and diagnostic utilities. Clin Microbiol Rev 2013;26:781–91. https://doi.org/10.1128/cmr.00021-13.Search in Google Scholar

131. Aro, K, Wei, F, Wong, DT, Tu, M. Saliva liquid biopsy for point-of-care applications. Front Public Health 2017;5:77. https://doi.org/10.3389/fpubh.2017.00077.Search in Google Scholar PubMed PubMed Central

132. Wang, X, Kaczor-Urbanowicz, KE, Wong, DT. Salivary biomarkers in cancer detection. Med Oncol 2017;34:7. https://doi.org/10.1007/s12032-016-0863-4.Search in Google Scholar PubMed PubMed Central

133. Rylander-Rudqvist, T, Hakansson, N, Tybring, G, Wolk, A. Quality and quantity of saliva DNA obtained from the self-administrated oragene method--a pilot study on the cohort of Swedish men. Cancer Epidemiol Biomarkers Prev 2006;15:1742–5. https://doi.org/10.1158/1055-9965.epi-05-0706.Search in Google Scholar

134. Abraham, JE, Maranian, MJ, Spiteri, I, Russell, R, Ingle, S, Luccarini, C, et al.. Saliva samples are a viable alternative to blood samples as a source of DNA for high throughput genotyping. BMC Med Genomics 2012;5:19. https://doi.org/10.1186/1755-8794-5-19.Search in Google Scholar PubMed PubMed Central

135. Paszkowski, J, Whitham, SA. Gene silencing and DNA methylation processes. Curr Opin Plant Biol 2001;4:123–9. https://doi.org/10.1016/s1369-5266(00)00147-3.Search in Google Scholar PubMed

136. Carvalho, AL, Jeronimo, C, Kim, MM, Henrique, R, Zhang, Z, Hoque, MO, et al.. Evaluation of promoter hypermethylation detection in body fluids as a screening/diagnosis tool for head and neck squamous cell carcinoma. Clin Cancer Res 2008;14:97–107. https://doi.org/10.1158/1078-0432.ccr-07-0722.Search in Google Scholar

137. Chang, X, Monitto, CL, Demokan, S, Kim, MS, Chang, SS, Zhong, X, et al.. Identification of hypermethylated genes associated with cisplatin resistance in human cancers. Cancer Res 2010;70:2870–9. https://doi.org/10.1158/0008-5472.can-09-3427.Search in Google Scholar PubMed PubMed Central

138. Bryan, AD, Magnan, RE, Hooper, AE, Harlaar, N, Hutchison, KE. Physical activity and differential methylation of breast cancer genes assayed from saliva: a preliminary investigation. Ann Behav Med 2013;45:89–98. https://doi.org/10.1007/s12160-012-9411-4.Search in Google Scholar PubMed PubMed Central

139. Carvalho, AL, Henrique, R, Jeronimo, C, Nayak, CS, Reddy, AN, Hoque, MO, et al.. Detection of promoter hypermethylation in salivary rinses as a biomarker for head and neck squamous cell carcinoma surveillance. Clin Cancer Res 2011;17:4782–9. https://doi.org/10.1158/1078-0432.ccr-11-0324.Search in Google Scholar PubMed PubMed Central

140. Schussel, J, Zhou, XC, Zhang, Z, Pattani, K, Bermudez, F, Jean-Charles, G, et al.. EDNRB and DCC salivary rinse hypermethylation has a similar performance as expert clinical examination in discrimination of oral cancer/dysplasia versus benign lesions. Clin Cancer Res 2013;19:3268–75. https://doi.org/10.1158/1078-0432.ccr-12-3496.Search in Google Scholar

141. Li, Y, Zhou, X, St, JM, Wong, DT. RNA profiling of cell-free saliva using microarray technology. J Dent Res 2004;83:199–203. https://doi.org/10.1177/154405910408300303.Search in Google Scholar PubMed

142. Gomes, MA, Rodrigues, FH, Afonso-Cardoso, SR, Buso, AM, Silva, AG, Favoreto, SJ, et al.. Levels of immunoglobulin A1 and messenger RNA for interferon gamma and tumor necrosis factor alpha in total saliva from patients with diabetes mellitus type 2 with chronic periodontal disease. J Periodontal Res 2006;41:177–83. https://doi.org/10.1111/j.1600-0765.2005.00851.x.Search in Google Scholar PubMed

143. Hu, Z, Zimmermann, BG, Zhou, H, Wang, J, Henson, BS, Yu, W, et al.. Exon-level expression profiling: a comprehensive transcriptome analysis of oral fluids. Clin Chem 2008;54:824–32. https://doi.org/10.1373/clinchem.2007.096164.Search in Google Scholar PubMed PubMed Central

144. Lee, YH, Zhou, H, Reiss, JK, Yan, X, Zhang, L, Chia, D, et al.. Direct saliva transcriptome analysis. Clin Chem 2011;57:1295–302. https://doi.org/10.1373/clinchem.2010.159210.Search in Google Scholar PubMed

145. Majem, B, Rigau, M, Reventos, J, Wong, DT. Non-coding RNAs in saliva: emerging biomarkers for molecular diagnostics. Int J Mol Sci 2015;16:8676–98. https://doi.org/10.3390/ijms16048676.Search in Google Scholar PubMed PubMed Central

146. Bartel, DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281–97. https://doi.org/10.1016/s0092-8674(04)00045-5.Search in Google Scholar PubMed

147. Ueda, T, Volinia, S, Okumura, H, Shimizu, M, Taccioli, C, Rossi, S, et al.. Relation between microRNA expression and progression and prognosis of gastric cancer: a microRNA expression analysis. Lancet Oncol 2010;11:136–46. https://doi.org/10.1016/s1470-2045(09)70343-2.Search in Google Scholar PubMed PubMed Central

148. Amin, M, Trevelyan, CJ, Turner, NA. MicroRNA-214 in health and disease. Cells 2021;10:3274. https://doi.org/10.3390/cells10123274.Search in Google Scholar PubMed PubMed Central

149. Park, NJ, Zhou, H, Elashoff, D, Henson, BS, Kastratovic, DA, Abemayor, E, et al.. Salivary microRNA: discovery, characterization, and clinical utility for oral cancer detection. Clin Cancer Res 2009;15:5473–7. https://doi.org/10.1158/1078-0432.ccr-09-0736.Search in Google Scholar PubMed PubMed Central

150. Dodds, MW, Johnson, DA, Yeh, CK. Health benefits of saliva: a review. J Dent 2005;33:223–33. https://doi.org/10.1016/j.jdent.2004.10.009.Search in Google Scholar PubMed

151. Vitorino, R, Lobo, MJ, Ferrer-Correira, AJ, Dubin, JR, Tomer, KB, Domingues, PM, et al.. Identification of human whole saliva protein components using proteomics. Proteomics 2004;4:1109–15. https://doi.org/10.1002/pmic.200300638.Search in Google Scholar PubMed

152. Amado, F, Lobo, MJ, Domingues, P, Duarte, JA, Vitorino, R. Salivary peptidomics. Expert Rev Proteomics 2010;7:709–21. https://doi.org/10.1586/epr.10.48.Search in Google Scholar PubMed

153. Vitorino, R, Alves, R, Barros, A, Caseiro, A, Ferreira, R, Lobo, MC, et al.. Finding new posttranslational modifications in salivary proline-rich proteins. Proteomics 2010;10:3732–42. https://doi.org/10.1002/pmic.201000261.Search in Google Scholar PubMed

154. Pappa, E, Vastardis, H, Mermelekas, G, Gerasimidi-Vazeou, A, Zoidakis, J, Vougas, K. Saliva proteomics analysis offers insights on type 1 diabetes pathology in a pediatric population. Front Physiol 2018;9:444. https://doi.org/10.3389/fphys.2018.00444.Search in Google Scholar PubMed PubMed Central

155. Dawes, C, Wong, D. Role of saliva and salivary diagnostics in the advancement of oral health. J Dent Res 2019;98:133–41. https://doi.org/10.1177/0022034518816961.Search in Google Scholar PubMed PubMed Central

156. Helmerhorst, EJ, Dawes, C, Oppenheim, FG. The complexity of oral physiology and its impact on salivary diagnostics. Oral Dis 2018;24:363–71. https://doi.org/10.1111/odi.12780.Search in Google Scholar PubMed PubMed Central

157. Miller, CS, Foley, JD, Bailey, AL, Campell, CL, Humphries, RL, Christodoulides, N, et al.. Current developments in salivary diagnostics. Biomarkers Med 2010;4:171–89. https://doi.org/10.2217/bmm.09.68.Search in Google Scholar PubMed PubMed Central

158. Bjorhall, K, Miliotis, T, Davidsson, P. Comparison of different depletion strategies for improved resolution in proteomic analysis of human serum samples. Proteomics 2005;5:307–17. https://doi.org/10.1002/pmic.200400900.Search in Google Scholar PubMed

159. Loo, JA, Yan, W, Ramachandran, P, Wong, DT. Comparative human salivary and plasma proteomes. J Dent Res 2010;89:1016–23. https://doi.org/10.1177/0022034510380414.Search in Google Scholar PubMed PubMed Central

160. Arakaki, AK, Skolnick, J, McDonald, JF. Marker metabolites can be therapeutic targets as well. Nature 2008;456:443. https://doi.org/10.1038/456443c.Search in Google Scholar PubMed

161. Zhang, A, Sun, H, Wang, P, Han, Y, Wang, X. Recent and potential developments of biofluid analyses in metabolomics. J Proteonomics 2012;75:1079–88. https://doi.org/10.1016/j.jprot.2011.10.027.Search in Google Scholar PubMed

162. Zhang, A, Sun, H, Wang, X. Saliva metabolomics opens door to biomarker discovery, disease diagnosis, and treatment. Appl Biochem Biotechnol 2012;168:1718–27. https://doi.org/10.1007/s12010-012-9891-5.Search in Google Scholar PubMed

163. Gardner, A, Parkes, HG, Carpenter, GH, So, PW. Developing and standardizing a protocol for quantitative proton nuclear magnetic resonance (1H NMR) spectroscopy of saliva. J Proteome Res 2018;17:1521–31. https://doi.org/10.1021/acs.jproteome.7b00847.Search in Google Scholar PubMed PubMed Central

164. Schroder, SA, Bardow, A, Eickhardt-Dalboge, S, Johansen, HK, Homoe, P. Is parotid saliva sterile on entry to the oral cavity? Acta Otolaryngol 2017;137:762–4. https://doi.org/10.1080/00016489.2016.1272002.Search in Google Scholar PubMed

165. Gardner, A, Parkes, HG, So, PW, Carpenter, GH. Determining bacterial and host contributions to the human salivary metabolome. J Oral Microbiol 2019;11:1617014. https://doi.org/10.1080/20002297.2019.1617014.Search in Google Scholar PubMed PubMed Central

166. Pathak, JL, Yan, Y, Zhang, Q, Wang, L, Ge, L. The role of oral microbiome in respiratory health and diseases. Respir Med 2021;185:106475. https://doi.org/10.1016/j.rmed.2021.106475.Search in Google Scholar PubMed

167. Wade, WG. The oral microbiome in health and disease. Pharmacol Res 2013;69:137–43. https://doi.org/10.1016/j.phrs.2012.11.006.Search in Google Scholar PubMed

168. Shen, LH, Xiao, E, Wang, EB, Zheng, H, Zhang, Y. High-throughput sequencing analysis of microbial profiles in the dry socket. J Oral Maxillofac Surg 2019;77:1548–56. https://doi.org/10.1016/j.joms.2019.02.041.Search in Google Scholar PubMed

169. Sampaio-Maia, B, Caldas, IM, Pereira, ML, Perez-Mongiovi, D, Araujo, R. The oral microbiome in health and its implication in oral and systemic diseases. Adv Appl Microbiol 2016;97:171–210.10.1016/bs.aambs.2016.08.002Search in Google Scholar PubMed

170. Matsha, TE, Prince, Y, Davids, S, Chikte, U, Erasmus, RT, Kengne, AP, et al.. Oral microbiome signatures in diabetes mellitus and periodontal disease. J Dent Res 2020;99:658–65. https://doi.org/10.1177/0022034520913818.Search in Google Scholar PubMed

171. Wei, AL, Li, M, Li, GQ, Wang, X, Hu, WM, Li, ZL, et al.. Oral microbiome and pancreatic cancer. World J Gastroenterol 2020;26:7679–92. https://doi.org/10.3748/wjg.v26.i48.7679.Search in Google Scholar PubMed PubMed Central

172. Jiang, Q, Liu, X, Yang, Q, Chen, L, Yang, D. Salivary microbiome in adenoid cystic carcinoma detected by 16S rRNA sequencing and shotgun metagenomics. Front Cell Infect Microbiol 2021;11:774453. https://doi.org/10.3389/fcimb.2021.774453.Search in Google Scholar PubMed PubMed Central

173. He, C, Zheng, S, Luo, Y, Wang, B. Exosome theranostics: biology and translational medicine. Theranostics 2018;8:237–55. https://doi.org/10.7150/thno.21945.Search in Google Scholar PubMed PubMed Central

174. Lasser, C, Alikhani, VS, Ekstrom, K, Eldh, M, Paredes, PT, Bossios, A, et al.. Human saliva, plasma and breast milk exosomes contain RNA: uptake by macrophages. J Transl Med 2011;9:9. https://doi.org/10.1186/1479-5876-9-9.Search in Google Scholar PubMed PubMed Central

175. Street, JM, Barran, PE, Mackay, CL, Weidt, S, Balmforth, C, Walsh, TS, et al.. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J Transl Med 2012;10:5. https://doi.org/10.1186/1479-5876-10-5.Search in Google Scholar PubMed PubMed Central

176. Jia, S, Zocco, D, Samuels, ML, Chou, MF, Chammas, R, Skog, J, et al.. Emerging technologies in extracellular vesicle-based molecular diagnostics. Expert Rev Mol Diagn 2014;14:307–21. https://doi.org/10.1586/14737159.2014.893828.Search in Google Scholar PubMed

177. Valadi, H, Ekstrom, K, Bossios, A, Sjostrand, M, Lee, JJ, Lotvall, JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007;9:654–9. https://doi.org/10.1038/ncb1596.Search in Google Scholar PubMed

178. Guescini, M, Genedani, S, Stocchi, V, Agnati, LF. Astrocytes and glioblastoma cells release exosomes carrying mtDNA. J Neural Transm 2010;117:1–4. https://doi.org/10.1007/s00702-009-0288-8.Search in Google Scholar PubMed

179. Ferguson, SW, Nguyen, J. Exosomes as therapeutics: the implications of molecular composition and exosomal heterogeneity. J Contr Release 2016;228:179–90. https://doi.org/10.1016/j.jconrel.2016.02.037.Search in Google Scholar PubMed

180. Nojima, H, Freeman, CM, Schuster, RM, Japtok, L, Kleuser, B, Edwards, MJ, et al.. Hepatocyte exosomes mediate liver repair and regeneration via sphingosine-1-phosphate. J Hepatol 2016;64:60–8. https://doi.org/10.1016/j.jhep.2015.07.030.Search in Google Scholar PubMed PubMed Central

181. Melo, SA, Sugimoto, H, O’Connell, JT, Kato, N, Villanueva, A, Vidal, A, et al.. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell 2014;26:707–21. https://doi.org/10.1016/j.ccell.2014.09.005.Search in Google Scholar PubMed PubMed Central

182. Wortzel, I, Dror, S, Kenific, CM, Lyden, D. Exosome-mediated metastasis: communication from a distance. Dev Cell 2019;49:347–60. https://doi.org/10.1016/j.devcel.2019.04.011.Search in Google Scholar PubMed

183. Wang, J, Zhou, Y, Lu, J, Sun, Y, Xiao, H, Liu, M, et al.. Combined detection of serum exosomal miR-21 and HOTAIR as diagnostic and prognostic biomarkers for laryngeal squamous cell carcinoma. Med Oncol 2014;31:148. https://doi.org/10.1007/s12032-014-0148-8.Search in Google Scholar PubMed

184. Li, X, Yang, T, Lin, J. Spectral analysis of human saliva for detection of lung cancer using surface-enhanced Raman spectroscopy. J Biomed Opt 2012;17:37003. https://doi.org/10.1117/1.jbo.17.3.037003.Search in Google Scholar PubMed

185. Machida, T, Tomofuji, T, Maruyama, T, Yoneda, T, Ekuni, D, Azuma, T, et al.. miR1246 and miR4644 in salivary exosome as potential biomarkers for pancreatobiliary tract cancer. Oncol Rep 2016;36:2375–81. https://doi.org/10.3892/or.2016.5021.Search in Google Scholar PubMed

186. Li, K, Lin, Y, Luo, Y, Xiong, X, Wang, L, Durante, K, et al.. A signature of saliva-derived exosomal small RNAs as predicting biomarker for esophageal carcinoma: a multicenter prospective study. Mol Cancer 2022;21:21. https://doi.org/10.1186/s12943-022-01499-8.Search in Google Scholar PubMed PubMed Central

187. Bhattarai, KR, Kim, HR, Chae, HJ. Compliance with saliva collection protocol in healthy volunteers: strategies for managing risk and errors. Int J Med Sci 2018;15:823–31. https://doi.org/10.7150/ijms.25146.Search in Google Scholar PubMed PubMed Central

188. Xu, F, Laguna, L, Sarkar, A. Aging-related changes in quantity and quality of saliva: where do we stand in our understanding? J Texture Stud 2019;50:27–35. https://doi.org/10.1111/jtxs.12356.Search in Google Scholar PubMed

189. Denny, PC, Denny, PA, Klauser, DK, Hong, SH, Navazesh, M, Tabak, LA. Age-related changes in mucins from human whole saliva. J Dent Res 1991;70:1320–7. https://doi.org/10.1177/00220345910700100201.Search in Google Scholar PubMed

190. Inzitari, R, Vento, G, Capoluongo, E, Boccacci, S, Fanali, C, Cabras, T, et al.. Proteomic analysis of salivary acidic proline-rich proteins in human preterm and at-term newborns. J Proteome Res 2007;6:1371–7. https://doi.org/10.1021/pr060520e.Search in Google Scholar PubMed

191. Fenoll-Palomares, C, Munoz, MJ, Sanchiz, V, Herreros, B, Hernandez, V, Minguez, M, et al.. Unstimulated salivary flow rate, pH and buffer capacity of saliva in healthy volunteers. Rev Esp Enferm Dig 2004;96:773–83. https://doi.org/10.4321/s1130-01082004001100005.Search in Google Scholar PubMed

192. Amerongen, AV, Veerman, EC. Saliva--the defender of the oral cavity. Oral Dis 2002;8:12–22. https://doi.org/10.1034/j.1601-0825.2002.1o816.x.Search in Google Scholar PubMed

193. Navazesh, M. Methods for collecting saliva. Ann N Y Acad Sci 1993;694:72–7. https://doi.org/10.1111/j.1749-6632.1993.tb18343.x.Search in Google Scholar PubMed

194. Khurshid, Z, Zohaib, S, Najeeb, S, Zafar, MS, Slowey, PD, Almas, K. Human saliva Collection devices for proteomics: an update. Int J Mol Sci 2016;17:846. https://doi.org/10.3390/ijms17060846.Search in Google Scholar PubMed PubMed Central

195. Pappa, E, Vougas, K, Zoidakis, J, Vastardis, H. Proteomic advances in salivary diagnostics. Biochim Biophys Acta Protein Proteonomics 2020;1868:140494. https://doi.org/10.1016/j.bbapap.2020.140494.Search in Google Scholar PubMed

196. Chiappin, S, Antonelli, G, Gatti, R, De Palo, EF. Saliva specimen: a new laboratory tool for diagnostic and basic investigation. Clin Chim Acta 2007;383:30–40. https://doi.org/10.1016/j.cca.2007.04.011.Search in Google Scholar PubMed

197. Granger, DA, Johnson, SB, Szanton, SL, Out, D, Schumann, LL. Incorporating salivary biomarkers into nursing research: an overview and review of best practices. Biol Res Nurs 2012;14:347–56. https://doi.org/10.1177/1099800412443892.Search in Google Scholar PubMed PubMed Central

198. Malamud, D. Saliva as a diagnostic fluid. Dent Clin 2011;55:159–78. https://doi.org/10.1016/j.cden.2010.08.004.Search in Google Scholar PubMed PubMed Central

199. Thomadaki, K, Helmerhorst, EJ, Tian, N, Sun, X, Siqueira, WL, Walt, DR, et al.. Whole-saliva proteolysis and its impact on salivary diagnostics. J Dent Res 2011;90:1325–30. https://doi.org/10.1177/0022034511420721.Search in Google Scholar PubMed PubMed Central

200. Pandit, P, Cooper-White, J, Punyadeera, C. High-yield RNA-extraction method for saliva. Clin Chem 2013;59:1118–22. https://doi.org/10.1373/clinchem.2012.197863.Search in Google Scholar PubMed

201. Novy, BB. Saliva and biofilm-based diagnostics: a critical review of the literature concerning sialochemistry. J Evid Base Dent Pract 2014;14 Suppl:27–32. https://doi.org/10.1016/j.jebdp.2014.04.004.Search in Google Scholar PubMed

202. Samaranayake, L. Saliva as a diagnostic fluid. Int Dent J 2007;57:295–9. https://doi.org/10.1111/j.1875-595x.2007.tb00135.x.Search in Google Scholar PubMed

203. Yang, F, Zeng, X, Ning, K, Liu, KL, Lo, CC, Wang, W, et al.. Saliva microbiomes distinguish caries-active from healthy human populations. ISME J 2012;6:1–10. https://doi.org/10.1038/ismej.2011.71.Search in Google Scholar PubMed PubMed Central

204. Zhang, Y, Huang, S, Jia, S, Sun, Z, Li, S, Li, F, et al.. The predictive power of saliva electrolytes exceeds that of saliva microbiomes in diagnosing early childhood caries. J Oral Microbiol 2021;13:1921486. https://doi.org/10.1080/20002297.2021.1921486.Search in Google Scholar PubMed PubMed Central

205. O’Brien-Simpson, NM, Burgess, K, Brammar, GC, Darby, IB, Reynolds, EC. Development and evaluation of a saliva-based chair-side diagnostic for the detection of porphyromonas gingivalis. J Oral Microbiol 2015;7:29129. https://doi.org/10.3402/jom.v7.29129.Search in Google Scholar PubMed PubMed Central

206. Lira-Junior, R, Bissett, SM, Preshaw, PM, Taylor, JJ, Bostrom, EA. Levels of myeloid-related proteins in saliva for screening and monitoring of periodontal disease. J Clin Periodontol 2021;48:1430–40. https://doi.org/10.1111/jcpe.13534.Search in Google Scholar PubMed

207. Kc, S, Wang, XZ, Gallagher, JE. Diagnostic sensitivity and specificity of host-derived salivary biomarkers in periodontal disease amongst adults: systematic review. J Clin Periodontol 2020;47:289–308. https://doi.org/10.1111/jcpe.13218.Search in Google Scholar PubMed

208. Brito-Zeron, P, Baldini, C, Bootsma, H, Bowman, SJ, Jonsson, R, Mariette, X, et al.. Sjogren syndrome. Nat Rev Dis Prim 2016;2:16047. https://doi.org/10.1038/nrdp.2016.47.Search in Google Scholar PubMed

209. Mavragani, CP, Crow, MK. Activation of the type I interferon pathway in primary Sjogren’s syndrome. J Autoimmun 2010;35:225–31. https://doi.org/10.1016/j.jaut.2010.06.012.Search in Google Scholar PubMed

210. Mavragani, CP, Moutsopoulos, HM. Sjogren’s syndrome. Annu Rev Pathol 2014;9:273–85. https://doi.org/10.1146/annurev-pathol-012513-104728.Search in Google Scholar PubMed

211. Hu, S, Vissink, A, Arellano, M, Roozendaal, C, Zhou, H, Kallenberg, CG, et al.. Identification of autoantibody biomarkers for primary Sjogren’s syndrome using protein microarrays. Proteomics 2011;11:1499–507. https://doi.org/10.1002/pmic.201000206.Search in Google Scholar PubMed PubMed Central

212. Gonzalez-Moles, MA, Ruiz-Avila, I, Gonzalez-Ruiz, L, Ayen, A, Gil-Montoya, JA, Ramos-Garcia, P. Malignant transformation risk of oral lichen planus: a systematic review and comprehensive meta-analysis. Oral Oncol 2019;96:121–30. https://doi.org/10.1016/j.oraloncology.2019.07.012.Search in Google Scholar PubMed

213. Lu, R, Zhang, J, Sun, W, Du, G, Zhou, G. Inflammation-related cytokines in oral lichen planus: an overview. J Oral Pathol Med 2015;44:1–14. https://doi.org/10.1111/jop.12142.Search in Google Scholar PubMed

214. Mozaffari, HR, Ramezani, M, Mahmoudiahmadabadi, M, Omidpanah, N, Sadeghi, M. Salivary and serum levels of tumor necrosis factor-alpha in oral lichen planus: a systematic review and meta-analysis study. Oral Surg Oral Med Oral Pathol Oral Radiol 2017;124:e183–9. https://doi.org/10.1016/j.oooo.2017.06.117.Search in Google Scholar PubMed

215. 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 Invest 2016;20:315–9. https://doi.org/10.1007/s00784-015-1506-4.Search in Google Scholar PubMed

216. Tvarijonaviciute, A, Aznar-Cayuela, C, Rubio, CP, Ceron, JJ, Lopez-Jornet, P. Evaluation of salivary oxidate stress biomarkers, nitric oxide and C-reactive protein in patients with oral lichen planus and burning mouth syndrome. J Oral Pathol Med 2017;46:387–92. https://doi.org/10.1111/jop.12522.Search in Google Scholar PubMed

217. Brailo, V, Vucicevic-Boras, V, Cekic-Arambasin, A, Alajbeg, IZ, Milenovic, A, Lukac, J. The significance of salivary interleukin 6 and tumor necrosis factor alpha in patients with oral leukoplakia. Oral Oncol 2006;42:370–3. https://doi.org/10.1016/j.oraloncology.2005.09.001.Search in Google Scholar PubMed

218. Brailo, V, Vucicevic-Boras, V, Lukac, J, Biocina-Lukenda, D, Zilic-Alajbeg, I, Milenovic, A, et al.. Salivary and serum interleukin 1 beta, interleukin 6 and tumor necrosis factor alpha in patients with leukoplakia and oral cancer. Med Oral Patol Oral Cir Bucal 2012;17:e10–5. https://doi.org/10.4317/medoral.17323.Search in Google Scholar PubMed PubMed Central

219. Punyani, SR, Sathawane, RS. Salivary level of interleukin-8 in oral precancer and oral squamous cell carcinoma. Clin Oral Invest 2013;17:517–24. https://doi.org/10.1007/s00784-012-0723-3.Search in Google Scholar PubMed

220. Hsu, HJ, Yang, YH, Shieh, TY, Chen, CH, Kao, YH, Yang, CF, et al.. Role of cytokine gene (interferon-gamma, transforming growth factor-beta1, tumor necrosis factor-alpha, interleukin-6, and interleukin-10) polymorphisms in the risk of oral precancerous lesions in Taiwanese. Kaohsiung J Med Sci 2014;30:551–8. https://doi.org/10.1016/j.kjms.2014.09.003.Search in Google Scholar PubMed

221. Wei, J, Xie, G, Zhou, Z, Shi, P, Qiu, Y, Zheng, X, et al.. Salivary metabolite signatures of oral cancer and leukoplakia. Int J Cancer 2011;129:2207–17. https://doi.org/10.1002/ijc.25881.Search in Google Scholar PubMed

222. 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. https://doi.org/10.1111/jop.12835.Search in Google Scholar PubMed

223. Zanoni, DK, Montero, PH, Migliacci, JC, Shah, JP, Wong, RJ, Ganly, I, et al.. Survival outcomes after treatment of cancer of the oral cavity (1985-2015). Oral Oncol 2019;90:115–21. https://doi.org/10.1016/j.oraloncology.2019.02.001.Search in Google Scholar PubMed PubMed Central

224. Guerrero-Preston, R, Soudry, E, Acero, J, Orera, M, Moreno-Lopez, L, Macia-Colon, G, et al.. NID2 and HOXA9 promoter hypermethylation as biomarkers for prevention and early detection in oral cavity squamous cell carcinoma tissues and saliva. Cancer Prev Res 2011;4:1061–72. https://doi.org/10.1158/1940-6207.capr-11-0006.Search in Google Scholar PubMed PubMed Central

225. Nagata, S, Hamada, T, Yamada, N, Yokoyama, S, Kitamoto, S, Kanmura, Y, et al.. Aberrant DNA methylation of tumor-related genes in oral rinse: a noninvasive method for detection of oral squamous cell carcinoma. Cancer Am Cancer Soc 2012;118:4298–308. https://doi.org/10.1002/cncr.27417.Search in Google Scholar

226. Shanmugam, A, Hariharan, AK, Hasina, R, Nair, JR, Katragadda, S, Irusappan, S, et al.. Ultrasensitive detection of tumor-specific mutations in saliva of patients with oral cavity squamous cell carcinoma. Cancer-Am Cancer Soc. 2021;127:1576–89. https://doi.org/10.1002/cncr.33393.Search in Google Scholar

227. Liu, CJ, Lin, SC, Yang, CC, Cheng, HW, Chang, KW. Exploiting salivary miR-31 as a clinical biomarker of oral squamous cell carcinoma. Head Neck 2012;34:219–24. https://doi.org/10.1002/hed.21713.Search in Google Scholar

228. Tang, H, Wu, Z, Zhang, J, Su, B. Salivary lncRNA as a potential marker for oral squamous cell carcinoma diagnosis. Mol Med Rep 2013;7:761–6. https://doi.org/10.3892/mmr.2012.1254.Search in Google Scholar

229. Wong, DT. Salivary diagnostics. Operat Dent 2012;37:562–70.10.2341/12-143-BLSearch in Google Scholar

230. Liotta, L. The role of cellular proteases and their inhibitors in invasion and metastasis. Introductionary overview. Cancer Metastasis Rev 1990;9:285–7. https://doi.org/10.1007/bf00049519.Search in Google Scholar

231. McCarthy, K, Maguire, T, McGreal, G, McDermott, E, O’Higgins, N, Duffy, MJ. High levels of tissue inhibitor of metalloproteinase-1 predict poor outcome in patients with breast cancer. Int J Cancer 1999;84:44–8. https://doi.org/10.1002/(sici)1097-0215(19990219)84:1<44::aid-ijc9>3.0.co;2-p.10.1002/(SICI)1097-0215(19990219)84:1<44::AID-IJC9>3.0.CO;2-PSearch in Google Scholar

232. Wu, CC, Chu, HW, Hsu, CW, Chang, KP, Liu, HP. Saliva proteome profiling reveals potential salivary biomarkers for detection of oral cavity squamous cell carcinoma. Proteomics 2015;15:3394–404. https://doi.org/10.1002/pmic.201500157.Search in Google Scholar

233. Hanahan, D, Weinberg, RA. Hallmarks of cancer: the next generation. Cell 2011;144:646–74. https://doi.org/10.1016/j.cell.2011.02.013.Search in Google Scholar

234. Yang, XH, Zhang, XX, Jing, Y, Ding, L, Fu, Y, Wang, S, et al.. Amino acids signatures of distance-related surgical margins of oral squamous cell carcinoma. EBioMedicine 2019;48:81–91. https://doi.org/10.1016/j.ebiom.2019.10.005.Search in Google Scholar

235. Pushalkar, S, Ji, X, Li, Y, Estilo, C, Yegnanarayana, R, Singh, B, et al.. Comparison of oral microbiota in tumor and non-tumor tissues of patients with oral squamous cell carcinoma. BMC Microbiol 2012;12:144. https://doi.org/10.1186/1471-2180-12-144.Search in Google Scholar PubMed PubMed Central

236. Yost, S, Stashenko, P, Choi, Y, Kukuruzinska, M, Genco, CA, Salama, A, et al.. Increased virulence of the oral microbiome in oral squamous cell carcinoma revealed by metatranscriptome analyses. Int J Oral Sci 2018;10:32. https://doi.org/10.1038/s41368-018-0037-7.Search in Google Scholar PubMed PubMed Central

237. Yang, CY, Yeh, YM, Yu, HY, Chin, CY, Hsu, CW, Liu, H, et al.. Oral microbiota community dynamics associated with oral squamous cell carcinoma staging. Front Microbiol 2018;9:862. https://doi.org/10.3389/fmicb.2018.00862.Search in Google Scholar PubMed PubMed Central

238. Chang, C, Wang, H, Liu, J, Pan, C, Zhang, D, Li, X, et al.. Porphyromonas gingivalis infection promoted the proliferation of oral squamous cell carcinoma cells through the miR-21/PDCD4/AP-1 negative signaling pathway. ACS Infect Dis 2019;5:1336–47. https://doi.org/10.1021/acsinfecdis.9b00032.Search in Google Scholar PubMed

239. Wang, Y, Liu, S, Li, B, Jiang, Y, Zhou, X, Chen, J, et al.. Staphylococcus aureus induces COX-2-dependent proliferation and malignant transformation in oral keratinocytes. J Oral Microbiol 2019;11:1643205. https://doi.org/10.1080/20002297.2019.1643205.Search in Google Scholar PubMed PubMed Central

240. Zhang, S, Li, C, Liu, J, Geng, F, Shi, X, Li, Q, et al.. Fusobacterium nucleatum promotes epithelial-mesenchymal transiton through regulation of the lncRNA MIR4435-2HG/miR-296-5p/Akt2/SNAI1 signaling pathway. FEBS J 2020;287:4032–47. https://doi.org/10.1111/febs.15233.Search in Google Scholar PubMed PubMed Central

241. Alemar, B, Izetti, P, Gregorio, C, Macedo, GS, Castro, MA, Osvaldt, AB, et al.. miRNA-21 and miRNA-34a are potential minimally invasive biomarkers for the diagnosis of pancreatic ductal adenocarcinoma. Pancreas 2016;45:84–92. https://doi.org/10.1097/mpa.0000000000000383.Search in Google Scholar

242. Torres, PJ, Fletcher, EM, Gibbons, SM, Bouvet, M, Doran, KS, Kelley, ST. Characterization of the salivary microbiome in patients with pancreatic cancer. PeerJ 2015;3:e1373. https://doi.org/10.7717/peerj.1373.Search in Google Scholar PubMed PubMed Central

243. Hoshino, I. The usefulness of microRNA in urine and saliva as a biomarker of gastroenterological cancer. Int J Clin Oncol 2021;26:1431–40. https://doi.org/10.1007/s10147-021-01911-1.Search in Google Scholar PubMed

244. Huang, K, Gao, X, Wu, L, Yan, B, Wang, Z, Zhang, X, et al.. Salivary microbiota for gastric cancer prediction: an exploratory study. Front Cell Infect Microbiol 2021;11:640309. https://doi.org/10.3389/fcimb.2021.640309.Search in Google Scholar PubMed PubMed Central

245. Agha-Hosseini, F, Mirzaii-Dizgah, I, Rahimi, A. Correlation of serum and salivary CA15-3 levels in patients with breast cancer. Med Oral Patol Oral Cir Bucal 2009;14:e521–4. https://doi.org/10.4317/medoral.14.e521.Search in Google Scholar PubMed

246. Fuzery, AK, Levin, J, Chan, MM, Chan, DW. Translation of proteomic biomarkers into FDA approved cancer diagnostics: issues and challenges. Clin Proteonomics 2013;10:13. https://doi.org/10.1186/1559-0275-10-13.Search in Google Scholar PubMed PubMed Central

247. Da, CG, Shepherd, FA, Tsao, MS. EGFR mutations and lung cancer. Annu Rev Pathol 2011;6:49–69. https://doi.org/10.1146/annurev-pathol-011110-130206.Search in Google Scholar PubMed

248. Liao, BC, Lin, CC, Yang, JC. Second and third-generation epidermal growth factor receptor tyrosine kinase inhibitors in advanced nonsmall cell lung cancer. Curr Opin Oncol 2015;27:94–101. https://doi.org/10.1097/cco.0000000000000164.Search in Google Scholar PubMed

249. Sun, Y, Liu, S, Qiao, Z, Shang, Z, Xia, Z, Niu, X, et al.. Systematic comparison of exosomal proteomes from human saliva and serum for the detection of lung cancer. Anal Chim Acta 2017;982:84–95. https://doi.org/10.1016/j.aca.2017.06.005.Search in Google Scholar PubMed

250. Jiang, X, Chen, X, Chen, Z, Yu, J, Lou, H, Wu, J. High-throughput salivary metabolite profiling on an ultralow noise tip-enhanced laser desorption ionization mass spectrometry platform for noninvasive diagnosis of early lung cancer. J Proteome Res 2021;20:4346–56. https://doi.org/10.1021/acs.jproteome.1c00310.Search in Google Scholar PubMed

251. Miller, CS, Foley, JR, Floriano, PN, Christodoulides, N, Ebersole, JL, Campbell, CL, et al.. Utility of salivary biomarkers for demonstrating acute myocardial infarction. J Dent Res 2014;93(7 Suppl):72S–79S. https://doi.org/10.1177/0022034514537522.Search in Google Scholar PubMed PubMed Central

252. Floriano, PN, Christodoulides, N, Miller, CS, Ebersole, JL, Spertus, J, Rose, BG, et al.. Use of saliva-based nano-biochip tests for acute myocardial infarction at the point of care: a feasibility study. Clin Chem 2009;55:1530–8. https://doi.org/10.1373/clinchem.2008.117713.Search in Google Scholar PubMed PubMed Central

253. Foley, JR, Sneed, JD, Steinhubl, SR, Kolasa, JR, Ebersole, JL, Lin, Y, et al.. Salivary biomarkers associated with myocardial necrosis: results from an alcohol septal ablation model. Oral Surg Oral Med Oral Pathol Oral Radiol 2012;114:616–23. https://doi.org/10.1016/j.oooo.2012.05.024.Search in Google Scholar PubMed PubMed Central

254. Rao, PV, Reddy, AP, Lu, X, Dasari, S, Krishnaprasad, A, Biggs, E, et al.. Proteomic identification of salivary biomarkers of type-2 diabetes. J Proteome Res 2009;8:239–45. https://doi.org/10.1021/pr8003776.Search in Google Scholar PubMed

255. Gonzalez-Sanchez, M, Bartolome, F, Antequera, D, Puertas-Martin, V, Gonzalez, P, Gomez-Grande, A, et al.. Decreased salivary lactoferrin levels are specific to Alzheimer’s disease. EBioMedicine 2020;57:102834. https://doi.org/10.1016/j.ebiom.2020.102834.Search in Google Scholar PubMed PubMed Central

256. Mota, F, Nascimento, KS, Oliveira, MV, Osterne, V, Clemente, J, Correia-Neto, C, et al.. Potential protein markers in children with Autistic spectrum disorder (ASD) revealed by salivary proteomics. Int J Biol Macromol 2022;199:243–51. https://doi.org/10.1016/j.ijbiomac.2022.01.011.Search in Google Scholar PubMed

257. Dantzer, R, Kalin, N. Salivary biomarkers of stress: cortisol and alpha-amylase. Psychoneuroendocrinology 2009;34:1.10.1016/j.psyneuen.2008.11.002Search in Google Scholar PubMed

258. Genuino, AJ, Chaikledkaew, U, The, DO, Reungwetwattana, T, Thakkinstian, A. Adjuvant trastuzumab regimen for HER2-positive early-stage breast cancer: a systematic review and meta-analysis. Expet Rev Clin Pharmacol 2019;12:815–24. https://doi.org/10.1080/17512433.2019.1637252.Search in Google Scholar PubMed PubMed Central

259. Lee, A, Ghaname, CB, Braun, TM, Sugai, JV, Teles, RP, Loesche, WJ, et al.. Bacterial and salivary biomarkers predict the gingival inflammatory profile. J Periodontol 2012;83:79–89. https://doi.org/10.1902/jop.2011.110060.Search in Google Scholar PubMed

260. Sexton, WM, Lin, Y, Kryscio, RJ, Dawson, DR, Ebersole, JL, Miller, CS. Salivary biomarkers of periodontal disease in response to treatment. J Clin Periodontol 2011;38:434–41. https://doi.org/10.1111/j.1600-051x.2011.01706.x.Search in Google Scholar PubMed PubMed Central

261. Butler-Laporte, G, Lawandi, A, Schiller, I, Yao, M, Dendukuri, N, McDonald, EG, et al.. Comparison of saliva and nasopharyngeal swab nucleic acid amplification testing for detection of SARS-CoV-2: a systematic review and meta-analysis. JAMA Intern Med 2021;181:353–60. https://doi.org/10.1001/jamainternmed.2020.8876.Search in Google Scholar PubMed PubMed Central

262. Huang, X, Tang, G, Ismail, N, Wang, X. Developing RT-LAMP assays for rapid diagnosis of SARS-CoV-2 in saliva. EBioMedicine 2022;75:103736. https://doi.org/10.1016/j.ebiom.2021.103736.Search in Google Scholar PubMed PubMed Central

263. Wood, BR, Kochan, K, Bedolla, DE, Salazar-Quiroz, N, Grimley, SL, Perez-Guaita, D, et al.. Infrared based saliva screening test for COVID-19. Angew Chem Int Ed Engl 2021;60:17102–7. https://doi.org/10.1002/anie.202104453.Search in Google Scholar PubMed PubMed Central

Received: 2022-08-11
Accepted: 2022-09-29
Published Online: 2022-10-27
Published in Print: 2023-01-27

© 2022 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 2.5.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2022-0793/html
Scroll to top button