Next Article in Journal
Oxidative DNA Damage and Arterial Hypertension in Light of Current ESC Guidelines
Previous Article in Journal
AAindexNC: Estimating the Physicochemical Properties of Non-Canonical Amino Acids, Including Those Derived from the PDB and PDBeChem Databank
Previous Article in Special Issue
New Insights into the Fanconi Anemia Pathogenesis: A Crosstalk Between Inflammation and Oxidative Stress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Inflammation on the Etiopathogenesis of Benign Salivary Gland Tumors: A Scoping Review

by
Konrad Szydłowski
1,2,*,
Michał Puchalski
3,
Stanisław Ołdziej
3,
Agnieszka Kasprzyk-Tryk
1,2,
Andrzej Skorek
1,2 and
Dmitry Tretiakow
1,2
1
Department of Otolaryngology, Academy of Applied Medical and Social Sciences, 82-300 Elblag, Poland
2
The Nicolaus Copernicus Hospital in Gdansk, Copernicus Healthcare Entity, Powstańców Warszawskich 1/2, 80-152 Gdansk, Poland
3
Intercollegiate Faculty of Biotechnology UG&MUG, University of Gdańsk, Abrahama 58, 80-307 Gdańsk, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(23), 12558; https://doi.org/10.3390/ijms252312558
Submission received: 30 October 2024 / Revised: 20 November 2024 / Accepted: 20 November 2024 / Published: 22 November 2024

Abstract

:
Salivary gland tumors are rare head and neck tumors constituting up to 6% of all head and neck neoplasms; despite being mostly benign, these tumors present in diverse histological subtypes, making them challenging to diagnose and treat. Our research aims to investigate the link between inflammation and tumorigenesis within the salivary glands based on the literature regarding the etiopathogenesis of benign salivary gland tumors. This scoping review was conducted following the PRISMA extension for scoping reviews and reporting guidelines. The search was conducted using the Pubmed and Embase databases. Articles published between 2004 and May 2024 were included in the review. A total of 1097 papers were collected and identified. After 271 duplicates were removed, 826 titles and abstracts were independently reviewed by two researchers. Based on the title and abstract, 735 citations were excluded, and 91 articles were assessed for eligibility. Data were extracted from 46 articles that met the inclusion criteria. The review highlights the significance of inflammation-related factors and its relations with benign salivary gland tumors (SGTs). Knowledge of the etiopathogenesis of these tumors remains insufficient, and their rich immunological background poses challenges in diagnosis. The findings also point to directions for further clinical research, which will be necessary to implement these molecules in clinical practice.

1. Introduction

Salivary gland tumors (SGTs) are diverse tumors that can be benign or malignant and mainly originate in the parotid salivary glands. SGTs are rare head and neck tumors constituting up to 6% of all head and neck neoplasms. Overall, almost 80% of tumors originate in parotid glands, of which 75% are benign and 25% are malignant [1]. According to the 2022 WHO classification, there are fifteen benign and twenty-one malignant epithelial neoplasms, one benign mesenchymal tumor, and two non-neoplastic epithelial lesions [2]. Due to their wide variety and complex histological composition, these tumors present in diverse histopathological subtypes, making them challenging to diagnose and treat [2]. The two most common parotid gland tumors are pleomorphic adenoma (PA), also known as a mixed tumor, and Warthin’s tumor (WT), also known as papillary cystadenoma lymphomatosum. These tumors account for almost 93% of all benign neoplasms originating in parotid glands [3,4]. Although benign, both of these tumors have malignancy potential, significantly higher in WT (up to 15%) than in PA (1%) [1,4]. The etiopathogenesis of benign parotid gland tumors is not clearly understood. Still, there are several risk factors, including chronic alcohol intake, age, and tobacco smoking [5,6]. Diagnosis using fine-needle aspiration biopsy has an average sensitivity of about 60 to 90% for lesions in the salivary glands, including differentiating between benign and malignant tumors [7,8,9,10]. Our research aims to investigate the link between inflammation and tumorigenesis within the salivary glands based on the literature regarding the etiopathogenesis of benign salivary gland tumors. The implementation of new biomarkers for the diagnosis of these tumors could increase their sensitivity and also allow for further development toward targeted biological treatment as an alternative to surgical intervention, which potentially carries the risk of damaging the facial nerve.

Chronic Inflammation and Tumorigenesis

Chronic inflammation has been linked to neoplasm formation since the 19th century, firstly by the German pathologist Rudolf Virchow, who described inflammatory factors inside the tumor tissue [11]. Recently, many studies have shown a link between chronic inflammation and various stages of tumorigenesis [12,13,14,15]. Chronic inflammation may not only be a predisposing factor of tumorigenesis due to a pre-existing inflammatory state but is also responsible for the development and growth within the tumor itself as a tumor-induced inflammation. Inflammation, whether parallel to chronic inflammatory disease or in connection with tumor-induced inflammation, significantly affects the structure of the tumor microenvironment (TME) and the adaptability of both the tumor and surrounding cells. Growing tumors create a continuous cycle of inflammation-induced signaling, facilitating the recruitment of inflammatory molecules. This contrasts with the regular inflammatory pathway, which resolves after limiting the initiating factor. One of the most critical steps in tumorigenesis is tumor initiation. The inflammatory response has the potential to accumulate gene mutations and epigenetic modifications. These changes can deactivate the immune system to suppress tumors and activate pathways that promote cancer. Inflammation unequivocally increases mutagenesis and predisposes normal tissue to accumulate mutations. Another essential part of the development of neoplasms is tumor promotion. Again, the inflammatory process plays a significant role, as research shows that inhibiting inflammation can alter tumor growth. As in tumor initiation, inflammatory cells and their signaling molecules can act as growth factors for growing tumors. Moreover, inflammatory factors play a crucial role in shaping cell plasticity within the tumor microenvironment, ultimately influencing the growth of tumors [13]. The tumor microenvironment is the specific cellular environment in which a tumor exists, consisting of different immune factors, signaling molecules, and extracellular matrix components. It is believed it plays a crucial role in the pathogenesis of tumor growth and its ability to develop. Numerous pathways and molecules, including pro-inflammatory cytokines and chemokines, can trigger and sustain tumorigenesis, creating an environment that helps neoplastic cells grow and thrive [16,17,18].
Additionally, chronic inflammation can have a significant impact on the development of cancer, affecting everything from tumor initiation and promotion to progression and metastasis [12].

2. Methods

This scoping review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) extension for scoping reviews and reporting guidelines [19].

2.1. Search Strategy

The search was conducted using Pubmed and Embase databases. Articles published between 2004 and May 2024 were included in the review. The search terms used were ”salivary gland neoplasms” and “salivary gland tumors”, combined with terms including “benign”, “inflammation”, and “pathogenesis”.

2.2. Inclusion and Exclusion Criteria

Studies investigating salivary gland tumors involving benign tumors were included. The search was restricted to articles written in English. Only human studies were included in the review. Articles analyzing chromosomal translocations, genetic disturbances, and infectious diseases as etiology of benign salivary gland tumors were excluded.

2.3. Data Extraction

After duplicates were removed using Mendeley’s reference management software 2.110.2, two authors (K.S. and D.T.) independently reviewed articles. The titles and abstracts of selected articles were screened, and those articles underwent evaluation for eligibility.

3. Results

As shown in Figure 1, 1097 papers were collected and identified. After 271 duplicates were removed, 826 titles and abstracts were independently reviewed by two researchers. Based on the title and abstract, 735 citations were excluded, and 91 articles were assessed for eligibility. Data were extracted from 46 articles (Table 1) that met the inclusion criteria.
The following main research directions were observed: angiogenesis factors, IgG4, pro-inflammatory molecules, oxidative stress, cell surface, and adhesion molecules, tumor stem cells, cytokines and lymphocytes, SOX-10, proteomic analysis, and others (Figure 2). Most of the analyzed studies were retrospective, randomized, observational studies using healthy tissues or tissues obtained from patients with malignant tumors as control groups. The review also included several case series studies. Of the forty-six studies included in the literature review, eight investigated substance concentrations in plasma, two involved both plasma and tumor tissues, and the remaining studies focused on the expression of molecules in removed tumor tissues or the impact of these molecules on tumor development. The main method for determining markers among selected articles was immunohistochemistry. In some of the studies, molecular research also included the assessment of mRNA expression.

4. Discussion

4.1. Angiogenesis Factors

Among the selected group of studies, the most commonly investigated factor was VEGF or its receptors. These molecules play a crucial role in stimulating the process of angiogenesis, which is key to tumor development and also plays an important role in inflammation [66,67]. Most studies analyzing the role of VEGF reported its overexpression in tumor tissues and elevated levels in the plasma of patients with salivary gland tumors [23,28,36,63]. In the study by Jabbar et al., VEGF overexpression was detected in 29 out of 30 benign salivary gland tumors [63]. Similarly, Faur et al. found an increased VEGF expression and mean microvascular density (MVD) in benign tumors compared to the control group, although these values were generally lower than in malignant tumors [36]. However, Błochowiak et al. found no differences in VEGF expression or its receptors between benign tumors and healthy tissue surrounding the tumor (control group) [49]. These results may be due to the study’s focus on specific isoforms, such as VEGF165 and VEGF165b, which were analyzed. Tampouris et al. demonstrated that pleomorphic adenomas had the strongest predilection for the VEGF C/D–VEGFR3 (flt-4) axis compared to other benign and malignant salivary gland tumors [23]. Tadbir et al. analyzed VEGF levels in the plasma of patients with salivary gland tumors [28]. The average plasma VEGF concentration was the highest in the malignant tumor group (727.3 ± 441.8 pg/mL), slightly lower in the benign tumor group (442.2 ± 343.3 pg/mL), and the lowest in the control group (263.9 ± 310.0 pg/mL) [28]. Gaonkar et al. investigated the expression of endoglin as an angiogenesis marker, showing its increased expression in both benign and malignant tumors compared to the control group [60]. The authors suggest that endoglin concentration in tissue may correlate with the degree of tumor malignancy [60]. The role of endoglin in inflammation, including its involvement in leukocyte adhesion and the formation of new blood vessels, has also been described [68,69]. Other factors involved in regulating tumor angiogenesis include semaphorins (acting as VEGF antagonists) and neuropilins, which function as VEGF coreceptors [42]. However, in a study of 248 benign and malignant tumors, no statistical differences were found in the expression of these molecules, limiting their use in diagnostics [42].

4.2. IgG4

Several studies focused on the presence of IgG4 in plasma or detecting these globulins in Warthin’s tumors and lymphadenomas [29,35,44]. Aga et al. described a case of a patient with Warthin’s tumor and IgG4-related autoimmune pancreatitis [29]. In addition to elevated IgG4 levels in the blood, immunoglobulins were also present in the tumor stroma [29]. In another study, Aga et al. evaluated blood plasma and tissue samples from patients with benign salivary gland tumors for the presence of IgG4, comparing their levels between patients with Warthin’s tumor and pleomorphic adenomas [35]. Elevated IgG4 levels were found in the plasma of five out of eighteen patients with Warthin’s tumor, and, in four of these, IgG4 was also present in tumor tissue, while no IgG4 was detected in the plasma or tumor tissue of patients with pleomorphic adenoma [35]. The study also analyzed the mRNA expression of these immunoglobulins in salivary gland tumor tissues and in a patient with IgG4-related disease (IgG4RD). IgG4 mRNA overexpression was detected in two of three analyzed Warthin’s tumors, similar to IgG4RD, but was not found in pleomorphic adenomas [35]. Kim J et al. analyzed IgG4 levels in tissues from fifteen lymphoepithelial tumors, including twelve benign tumors, eight of which were lymphadenomas and four were lymphoepithelial cysts [44]. Elevated IgG4 levels were statistically significant in two out of eight lymphadenomas, while no correlation was found in other types of lesions [44].

4.3. Pro-Inflammatory Molecules

In a study by Loy et al., an increased expression of COX-2 was observed in all 21 analyzed patients with Warthin’s tumor (WT) [20]. The enzyme was present in the epithelial component of the tumor, excretory ducts, and healthy salivary tissue adjacent to the tumor, while no increased expression was found in the remaining healthy salivary gland parenchyma [20]. Tenorio et al. compared 62 benign and malignant salivary gland tumors for COX-2 expression in their tissues [50]. A strong COX-2 overexpression was observed in all mucoepidermoid carcinoma tissues, while, in benign tumors, a weak expression was observed in 39.5% of pleomorphic adenomas (PAs), and in 60.5%, it was not detected at all [50]. The difference in COX-2 expression between these studies may stem from the different types of benign tumors analyzed (WT vs. PA), suggesting a predilection towards inflammatory pathway via COX-2 in Warthin’s tumor development.
In three studies, researchers collected blood samples from patients undergoing surgery for salivary gland tumors and analyzed leukocyte, platelet, neutrophil, and monocyte counts to calculate indexes representing the intensity of inflammation [57,61,64]. Abbate et al. analyzed inflammatory biomarkers (NLR, PLR, SII) in 191 patients with benign salivary gland tumors compared to 90 control patients [61]. The NLR and SII indexes were significantly higher in patients with salivary gland tumors compared to the control group. In contrast, the PLR index was elevated only in patients with pleomorphic adenomas. Sahin et al. evaluated whether the SII index correlated with the malignancy of salivary gland tumors [57]. Comparing 185 patients with benign tumors to 52 with malignant tumors, a statistically significant difference in SII levels was found, with higher values in malignant tumors. However, no cutoff value was established for using this biomarker in clinical practice [57]. In another study, Abbate et al. examined the use of inflammatory biomarkers (SII, SIRI, PLR, and NLR) in diagnosing salivary gland tumors alongside cytological results from fine-needle aspiration biopsies [64]. SIRI proved to be the most diagnostically useful biomarker, with the combination of SIRI and FNAC results increasing diagnostic sensitivity to 82.8% compared to 59.6% with FNAC alone and 66.7% with SIRI alone [64]. These findings suggest a strong association between inflammation and salivary gland tumors, including benign tumors, and highlight the usefulness of inflammatory biomarkers in diagnosing these tumors.

4.4. Oxidative Stress

Two of the studies analyzed plasma concentrations of substances that serve as markers of oxidative stress [48,62]. In the study by Sowa et al., the results indicate that patients with salivary gland tumors, including benign ones, exhibit a low level of whole-body oxidative stress [48]. This is evidenced by decreased levels of Total Antioxidant Capacity of Blood Serum (FRAP) and thiol groups and increased levels of Advanced Oxidation Protein Products (AOPP). In another study by Sowa et al., which involved 61 patients with benign and malignant salivary gland tumors (52 of whom had benign tumors), elevated blood lipofuscin levels were observed in all patients with salivary gland tumors compared to the control group, regardless of tumor type [62]. Additionally, the study showed a decrease in the antioxidant enzyme Cu-Zn SOD, but only in patients with Warthin’s tumors, suggesting the involvement of this antioxidant enzyme in the pathogenesis of this type of tumor [62]. The findings indicate that oxidative stress, commonly linked to tumor development and chronic inflammation in the literature, is present in patients with salivary gland tumors, including benign ones [70,71,72].

4.5. Cell Surface and Adhesion Molecules/Tumor Stem Cells

Many of the studies focused on detecting adhesion molecules and various receptors and ligands, including CD group receptors and cadherins [21,25,32,33,38,54,58,59]. These molecules, in addition to helping identify cell types in immunophenotyping studies, play significant roles in signaling pathways, including tumorigenesis and inflammation development [73,74]. Tumor stem cells, identified using biomarkers, were also evaluated [32,54,58,59]. These cells can initiate tumorigenesis and may be responsible for treatment failures and tumor recurrence [75].
Certain molecules may act as markers of malignant transformation risk, such as CD166. Tadbir et al. showed increased expression of CD166 in both benign and malignant tumors, with higher levels observed in malignant tumors [38]. In another study, the expression of CD44 was analyzed in benign and malignant salivary gland tumors [59]. CD44 is widely studied in the context of various types of cancer due to its role in cell adhesion and migration and its use as a tumor stem cell marker [76,77,78]. In a study of 38 patients, including 13 with benign tumors, CD44 and CD44v6 isoforms were found to be upregulated in benign tumors compared to the control group, though their levels were slightly lower than in malignant tumors [59]. Ianez et al. compared the expression of CD24 and CD44 between 101 surgically removed pleomorphic adenomas and 20 fetal salivary gland tissues [32]. The study assessed whether these molecules could be markers for salivary gland cancer stem cells. CD24 and CD44 were detected in all pleomorphic adenomas and fetal salivary glands using immunohistochemistry, but no characteristic expression pattern was found within the pleomorphic adenomas. The expression pattern for both markers was consistent between fetal salivary glands and normal human salivary glands used as a control group. Molecular analysis using RT-PCR revealed increased CD44 expression only in pleomorphic adenomas, with no difference in CD24 expression compared to the control group. The authors concluded that CD24 and CD44 could not be used as specific cancer stem cell markers for prognostic or diagnostic purposes [32]. Haghshenas et al. performed a proteomic analysis of five benign and five malignant salivary gland tumors to investigate proteins associated with mesenchymal stem cells [58]. They found overexpression of several markers and receptors, including CD44, CD73, CD90, CD105, and CD166, present in both benign and malignant tumors. Notably, an increased expression of heat shock protein 70 (Hsp70), keratin, and type II cytoskeletal 7 (CK-7) was observed in benign compared to malignant tumors [58]. These findings confirm the distinct architecture and etiopathogenesis of benign and malignant tumors. Two studies focused on cadherin expression, molecules involved in cell adhesion whose role in tumorigenesis has been investigated for many benign and malignant tumors [21,25,73]. In a study of 110 benign and malignant salivary gland tumors, a strong E-cadherin expression was observed in all 54 benign tumors, while malignant tumors showed a loss of expression as their cytological malignancy increased [21]. A study evaluating N-cadherin expression in 18 benign and 31 malignant salivary gland tumors found no expression in the control group tissues, pleomorphic adenomas, or myoepitheliomas [25]. However, N-cadherin was present in the epithelium of one Warthin’s tumor and the germinal centers of lymphoid tissue in four Warthin’s tumors [25]. Additionally, the study demonstrated a correlation between increased N-cadherin expression and histological malignancy and neuroinvasion in malignant tumors [25], suggesting its potential as a diagnostic biomarker. Karbanova et al. found an increased expression of Prominin (CD133) in both benign and malignant salivary gland tumors [33]. This overexpression was also present in the peritumoral tissues and inflamed salivary glands, suggesting a possible link between chronic irritation and tumor development. The authors hypothesize that tumor growth leads to cell compression, disrupting blood flow and causing hypoxia, which results in inflammation [33]. ALDH-1, an enzyme used as a marker for tumor stem cells, was analyzed in 154 salivary gland tumors by Da Silva et al. [54]. They found increased expression of ALDH-1 in all 51 benign tumors and 85.6% of the 103 malignant tumors [54]. The results confirm the presence of tumor stem cells in salivary gland cancers, which may impact disease progression and prognosis. In some malignant tumors, ALDH-1 expression in the stroma correlated with shorter overall survival, suggesting a prognostic role for tumor stem cells in malignant tumors, though further research is needed to understand their role in benign tumors [54].

4.6. Cytokines and Lymphocytes

Numerous studies have analyzed cytokine concentrations and expression in both tumor tissues and plasma from patients with benign salivary gland tumors [24,26,40,43,45,46,47,55,56].
One of the chemokines studied was CCL28, which is involved in lymphocyte recruitment and plays a vital role in the immune response, particularly in the oral cavity. Liu G et al. analyzed 72 benign salivary gland tumors, including pleomorphic adenomas and adenolymphomas, for the presence of CCL28 in the tissues. They found decreased expression of CCL28 in tumors compared to healthy surrounding tissues, suggesting that reduced recruitment of anti-tumor factors could be a mechanism in the tumorigenesis of these neoplasms [24]. Biological drugs, specifically monoclonal antibodies against various pro-inflammatory factors, are a groundbreaking class of medications used in treating many inflammatory and autoimmune diseases [79,80]. Carlesimo M et al. presented cases of two patients with psoriasis treated with Adalimumab (an anti-TNF alpha antibody) who developed benign salivary gland tumors during the treatment [26]. One patient developed bilateral Warthin’s tumors, while the other was diagnosed with a pleomorphic adenoma of the parotid gland. Although the limited number of cases does not allow for a definitive cause-and-effect relationship, this raises the possibility of a link between TNF alpha activity and tumorigenesis in benign salivary gland tumors. The distribution of T regulatory cells (Tregs), IL-17-producing lymphocytes, and CTLA4+ lymphocytes was analyzed in the plasma of patients with nineteen benign and eight malignant salivary gland tumors and compared to a control group [40]. An increased proportion of Tregs and CTLA4+CD4+ lymphocytes was found in both benign and malignant tumor patients compared to the control group, with the ratio being significantly higher in the malignant tumor group (7.74 ± 1.1 for Tregs and 8.18 ± 1.77 for CTLA4+CD4+ cells in malignant tumors, vs. 4.38 ± 0.56 for Tregs and 3.83 ± 0.56 for CTLA4+CD4+ cells in benign tumors, and 2.34 ± 0.28 for Tregs and 2.22 ± 0.25 for CTLA4+CD4+ cells in controls). Meanwhile, the percentage of Th17 cells and the Th17/Treg ratio was lower in both benign and malignant tumor groups compared to controls (Th17 cells: 0.84 ± 0.14 in malignant tumors, 2.09 ± 0.31 in benign tumors, and 2.31 ± 0.23 in controls; Th17/Treg ratio: 0.12 ± 0.03 in malignant tumors, 0.48 ± 0.09 in benign tumors, and 1.26 ± 0.23 in controls). These differences in lymphocyte subpopulations may suggest their role in the pathogenesis of tumors, with benign tumors appearing as an intermediate stage between malignant tumors and normal salivary gland tissues [40].
In another study, Haghshenas et al. analyzed the concentrations of lymphocyte subtypes (Th1, Th2, Tc1, and Tc2) in the peripheral blood of patients with benign and malignant salivary gland tumors [45]. No difference was observed in the Th1 and Th2 lymphocyte percentages between the benign tumor group and controls. However, the Th1/Th2 ratio in the benign tumor group (8.19 ± 1.56) showed a statistically significant decline compared to controls (11.13 ± 1.60). Similarly, no differences were found in the Tc1 and Tc2 percentages, though the Tc1/Tc2 ratio was lower in the benign tumor group (13.79 ± 4.16) compared to controls (16.40 ± 2.70), although this difference was not statistically significant (p = 0.12). Khademi B et al. [43] analyzed cytokine levels, specifically IFN-gamma (a Th1 marker) and IL-4 (a Th2 marker), in 55 benign tumors, 14 malignant tumors, and a control group. No significant differences were found in these cytokines between groups, suggesting no role in the etiology of salivary gland tumors [43]. However, Zare R et al. found a slightly higher expression of IL-33 in benign pleomorphic adenomas compared to healthy salivary tissue, though the expression was lower than in malignant tumors [46]. These results indicate that IL-33 may have potential as a biomarker for salivary gland tumor diagnostics.
Adipocytokines are a group of bioactive proteins involved in metabolic processes [81]. Sowa et al. analyzed circulating adipocytokines in the plasma of salivary gland tumor patients [47]. They found increased levels of adiponectin and visfatin in all 51 patients with benign tumors, regardless of gender. Additionally, higher leptin levels were observed in men with benign salivary gland tumors but not in women, with no difference in IL-6 levels between male and female patients compared to the control group [47].
Kobayashi et al. evaluated the potential role of helper T cells and follicular helper T cells in tissues from 64 patients with Warthin’s tumors, dividing the tumors into solid (25 tumors) and cystic (39 tumors) subtypes based on imaging studies [56]. Cystic tumors showed atrophy of germinal centers compared to solid tumors, resulting in a decrease in follicular helper T cells in the germinal centers. Cystic tumors also demonstrated an increase in T-bet-positive lymphocytes in the epithelium compared to solid tumors. Furthermore, Th2-induced responses were suppressed in cystic tumors, with a dominance of Th1 responses, leading to chronic inflammation and more significant tumor tissue destruction compared to solid tumors [56]. These differences in lymphocyte concentrations and ratios suggest that each subtype presents a unique microenvironment and architecture, which may reflect slightly different pathogenesis and behavior [56].
Mochizuki et al. compared the expression of CXCL12, CXCL10, and CCL18 between 20 Warthin’s tumors and 20 pleomorphic adenomas using immunohistochemical analysis and RT-PCR [55]. All Warthin’s tumors were immunopositive for CXCL12 and CXCL10, with most also positive for CCL18, while nearly all pleomorphic adenomas showed negative results [55]. The findings suggest that these chemokines are linked to the lymphatic stroma of Warthin’s tumors, playing a role in their pathogenesis and highlighting their histological distinction from pleomorphic adenomas.

4.7. SOX-10

Ohtomo et al. analyzed a total of 90 benign and malignant salivary gland tumors for the expression of SOX-10 protein, a transcription factor involved in the differentiation of various cell types, which is also present in the healthy tissue of major salivary glands [30,82]. Among the 14 benign tumors analyzed significant overexpression of SOX-10 protein was found only in pleomorphic adenomas, with no overexpression observed in Warthin’s tumors or oncocytomas [30]. The results confirm these tumor types’ distinct pathogenesis and nature and highlight SOX-10 as a valuable diagnostic marker. In the study by Baneckova et al. SOX-10 and S1009 protein expression was evaluated in 89 benign tumors, including 74 oncocytomas, 15 pleomorphic adenomas (PA), and myoepitheliomas (ME) with oncocytic metaplasia. SOX-10 and S1009 expression was detected in all pleomorphic adenomas and myoepitheliomas, and in 11 of the 74 oncocytomas [53]. Further histopathological analysis based on these findings suggests that tumors with positive expression of these proteins, initially diagnosed as oncocytoma, were oncocytic variants of PA and ME, which may help differentiate these tumors and shed new light on their pathogenesis [53].

4.8. Proteomic Analysis

Donadio et al. performed proteomic analysis on fine needle biopsy material from 36 benign parotid gland tumors, including 22 pleomorphic adenomas and 14 Warthin’s tumors [31]. A total of 26 different proteins were detected. An analysis of the identified proteins using dedicated computational software revealed two distinct protein networks for pleomorphic adenomas and Warthin’s tumors [31]. Among the proteins with increased expression in pleomorphic adenoma tissues were ANXA1, ANXA4, ApoE, and CRYAB, which are directly linked to the process of tumorigenesis [31,83,84]. On the other hand, the protein network detected in Warthin’s tumors (e.g., IGHG1, IGKC, IGHA1, and S100A9) was associated with immunological and inflammatory diseases [85]. These detected differences in tumorigenesis pathways between these tumors and identifying specific proteins offer hope for their use as biomarkers and could lead to improved preoperative diagnostics.

4.9. Miscellaneous

Human beta-defensin is a protein described in the literature as involved in tumor pathogenesis, potentially functioning as a tumor suppressor or acting as a proto-oncogene depending on the protein isoform and cancer type [86]. In the analyzed study, the expression of three hBD (1, 2, 3) subtypes was compared between healthy salivary gland tissue, chronically inflamed salivary glands, pleomorphic adenoma tissues, and adjacent adenoma tissues [22]. Pantelis et al. demonstrated statistically reduced expression of hBD1, with no statistical difference for hBD 2/3 in all five analyzed pleomorphic adenomas compared to healthy salivary gland tissue [22]. However, hBD1 and hBD3 expression levels were significantly elevated in inflamed salivary glands and adjacent adenoma tissues compared to healthy salivary tissue. The results suggest an important role for hBD1 as a tumor suppressor in pleomorphic adenoma tumorigenesis.
Cyclin D1 and p16INK4A proteins are involved in cell proliferation via regulation dependent on the Rb protein, and disturbances in this regulation can lead to tumor development. Jour et al. analyzed p16 and cyclin D1 protein expression in 44 salivary gland tumors, including 14 benign tumors. The overexpression of both proteins was observed in nearly all benign and malignant tumors compared to healthy tissue, but no statistically significant differences were found between salivary gland tumor types. The results argue against using these factors as diagnostic markers for distinguishing malignancy in salivary gland tumors [27].
Maspin is a protease inhibitor protein that acts as a tumor suppressor by inhibiting tumorigenesis processes [87]. Its role has been analyzed in many cancers, where its expression has been associated with either better or worse patient prognosis depending on the tumor type [34]. Reshma et al. compared maspin expression between benign and malignant salivary gland tumors, demonstrating the increased expression of this protein in all benign salivary gland tumors, while it was present in only a little over half of malignant tumors [34]. This could indicate a less aggressive nature for some malignant tumors. The results suggest the potential use of maspin as a diagnostic or prognostic marker in salivary gland tumors, given the milder nature of tumors expressing this protein.
Lipids are a significant component of the tumor microenvironment and have a notable impact on the process of carcinogenesis [88]. He Q et al. analyzed the distribution pattern of phosphatidylcholine subtypes within Warthin’s tumors compared to normal salivary gland tissue using imaging mass spectrometry [37]. Five different phosphatidylcholines were detected in all analyzed Warthin’s tumors, with only PC (16:0/16:0) being significantly elevated in the lymphoid stroma region of the Warthin’s tumors, which may indicate a distinct metabolism in this tumor region. The authors suggest a possible connection between this localization of PC (16:0/16:0) and the role of inflammation in the etiopathogenesis of this tumor [37].
PRDM1 is a molecule involved in the modulation of immune cells, including B and T lymphocytes [89]. Wang et al. analyzed PRDM1 protein in 40 Warthin’s tumors (WT), comparing its expression between the epithelial component of the tumor, surrounding healthy tissues, germinal centers of WT, and palatine tonsil tissues with follicular hyperplasia, which served as a reference for the lymphoid stroma of the Warthin’s tumor. They detected an overexpression of PRDM1 protein in all analyzed tumors within the epithelial component and palatine tonsil tissues, but no expression was observed in the lymphoid germinal centers or tissues surrounding the tumor [39]. The results indicate a significant role of the PRDM1 protein in the development of Warthin’s tumors, which may serve as a target for diagnosis and treatment.
Caveolin-1 is a protein that stimulates the process of tumorigenesis but can also play a suppressive role depending on the type of tumor [90,91]. A study by Jaafari-Ashkavandi et al. assessed the expression of Caveolin-1 between benign (15 pleomorphic adenomas) and malignant tumors compared to normal salivary gland tissue. They demonstrated statistically significant overexpression of Caveolin-1 in both benign and malignant tumors compared to the control group, with no differences in expression between benign and malignant tumors. Additionally, for pleomorphic adenomas, they observed an inverse correlation between Caveolin-1 expression and the Ki67 proliferation index. According to the authors, these findings indicate a significant role of Caveolin-1 in the development of salivary gland tumors and its potential use as a diagnostic biomarker [41].
Glypican-3 (GPC-3) is a proteoglycan that regulates cell proliferation, abundantly found in embryonic tissues but sparse in adult tissues [92]. Disruptions in the expression of this molecule may contribute to tumorigenesis [93]. In a study by Tadbir et al., they assessed GPC-3 expression in salivary gland tumors, including 17 benign tumors (pleomorphic adenomas), compared to the normal parenchyma of the surrounding salivary gland [51]. They demonstrated an overexpression of GPC-3 in tissues from salivary gland tumors, including in 11 out of 17 pleomorphic adenomas, compared to healthy tissue; however, this expression was more frequent and more substantial in malignant tumors than in benign ones (64.7% for PA vs. 82.4% for ACC vs. 87.5% for MEC) [51]. The authors suggest using GPC-3 as a target for potentially targeted salivary gland tumor therapy [51].
The RANK receptor and its ligand RANKL are factors involved in bone metabolism [94,95] and have also been widely described in the literature as significant factors in the carcinogenesis of many malignant tumors being detected in the tumor microenvironment [95]. In a study by Aslan et al., they compared the expression of RANK and RANKL between 50 malignant and 38 benign salivary gland tumors [52]. RANK expression was detected in 82% of malignant tumors compared to 34.2% in benign tumors, and in the case of RANKL, 28% vs. 5.3% in favor of malignant tumors. This may indicate the potential of RANK and RANKL as biomarkers that could be used in the differential diagnosis of benign and malignant salivary gland tumors.
Periostin is an extracellular matrix protein that plays structural roles and regulates fibrosis processes [96]. It is also associated with chronic inflammation [97]. Tateda et al. used specific antibodies to study periostin expression in 38 benign parotid gland tumors removed during surgery [65]. An increased expression of this protein was found in 32 out of 38 (84.2%) tissue samples, suggesting its involvement in the pathogenesis of benign salivary gland tumors and its potential use as a diagnostic biomarker [65].

5. Conclusions

In summary, the above literature review highlights the significant role of factors related to inflammation and benign salivary gland tumors. Knowledge of the etiopathogenesis of these tumors remains insufficient, and the rich immunological background of these tumors poses challenges in diagnosis. Despite the benign nature of these tumors, surgical treatment carries the risk of potentially significant complications, such as facial nerve paralysis. Changing the approach to diagnosis and treatment could minimize the risk of misdiagnosis, and new potential targeted drugs could serve as alternatives or support surgical treatment. The findings also point to directions for further clinical research, which will be necessary to implement these molecules in clinical practice.

Author Contributions

Conceptualization, K.S. and M.P.; methodology, K.S., D.T., and M.P., investigation, K.S. and D.T., writing—original draft preparation, K.S., M.P., A.K.-T., D.T., and S.O.; writing—review and editing, K.S., M.P., D.T., A.S., and S.O.; supervision, A.S. and D.T.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted following the Declaration of Helsinki, and the implementation of this study did not require the Institutional Review Board Statement (or Ethics Committee).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

WT—Warthin’s tumor, PA—pleomorphic adenoma, TME—tumor microenviroment, ME—myoepithelioma.

References

  1. Shah, J.; Bhuvanesh, S.; Patel, S.; Wong, R. Jatin Shah’s Head and Neck Surgery and Oncology; Elsevier: Amsterdam, The Netherlands, 2020; Volume 7, pp. 489–555. [Google Scholar]
  2. Skálová, A.; Hyrcza, M.D.; Leivo, I. Update from the 5th Edition of the World Health Organization Classification of Head and Neck Tumors: Salivary Glands. Head Neck Pathol. 2022, 16, 40–53. [Google Scholar] [CrossRef]
  3. Zhan, K.Y.; Khaja, S.F.; Flack, A.B.; Day, T.A. Benign Parotid Tumors. Otolaryngol. Clin. N. Am. 2016, 49, 327–342. [Google Scholar] [CrossRef] [PubMed]
  4. Bradley, P.J. Frequency and Histopathology by Site, Major Pathologies, Symptoms and Signs of Salivary Gland Neoplasms. Adv. Otorhinolaryngol. 2016, 78, 9–16. [Google Scholar] [CrossRef] [PubMed]
  5. Gao, Y.; Chen, H.; Liu, Y.; Zhang, X.; Qiu, Y.; Huang, D. Modifiable Factors for Benign Salivary Gland Neoplasms: A Mendelian Randomization Study. Oral. Dis. 2024, 30, 2245–2253. [Google Scholar] [CrossRef]
  6. Li, T.I.; Chiang, M.T.; Chiu, K.C.; Lai, C.H.; Liu, S.Y.; Shieh, Y.S. The Association of Betel Quid, Alcohol, and Cigarettes with Salivary Gland Tumor-A Case-Control Study. J. Dent. Sci. 2017, 12, 151–155. [Google Scholar] [CrossRef] [PubMed]
  7. Tretiakow, D.; Skorek, A. Regarding the “Review of Surgical Techniques and Guide for Decision Making in the Treatment of Benign Parotid Tumors. Eur. Arch. Oto-Rhino-Laryngol. 2020, 277, 3537–3538. [Google Scholar] [CrossRef] [PubMed]
  8. Tretiakow, D.; Mikaszewski, B.; Skorek, A. The Role of Fine needle Aspiration Biopsy (FNAB) in the Diagnostic Management of Parotid Gland Masses with Emphasis on Potential Pitfalls. Eur. Arch. Oto-Rhino-Laryngol. 2020, 277, 2939–2940. [Google Scholar] [CrossRef]
  9. Nakaguro, M. Diagnostic Clues and Pitfalls in Salivary Gland Fine-Needle Aspiration Cytology. Semin. Diagn. Pathol. 2024, 41, 207–211. [Google Scholar] [CrossRef] [PubMed]
  10. Reerds, S.T.H.; Van Engen-Van Grunsven, A.C.H.; van den Hoogen, F.J.A.; Takes, R.P.; Marres, H.A.M.; Honings, J. Accuracy of Parotid Gland FNA Cytology and Reliability of the Milan System for Reporting Salivary Gland Cytopathology in Clinical Practice. Cancer Cytopathol. 2021, 129, 719–728. [Google Scholar] [CrossRef]
  11. Balkwill, F.; Mantovani, A. Inflammation and Cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
  12. Hibino, S.; Kawazoe, T.; Kasahara, H.; Itoh, S.; Ishimoto, T.; Sakata-Yanagimoto, M.; Taniguchi, K. Inflammation-Induced Tumorigenesis and Metastasis. Int. J. Mol. Sci. 2021, 22, 5421. [Google Scholar] [CrossRef] [PubMed]
  13. Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef] [PubMed]
  14. Diakos, C.I.; Charles, K.A.; McMillan, D.C.; Clarke, S.J. Cancer-Related Inflammation and Treatment Effectiveness. Lancet Oncol. 2014, 15, e493–e503. [Google Scholar] [CrossRef] [PubMed]
  15. Elinav, E.; Nowarski, R.; Thaiss, C.A.; Hu, B.; Jin, C.; Flavell, R.A. Inflammation-Induced Cancer: Crosstalk between Tumours, Immune Cells and Microorganisms. Nat. Rev. Cancer 2013, 13, 759–771. [Google Scholar] [CrossRef] [PubMed]
  16. Puchalski, M.; Tretiakow, D.; Skorek, A.; Szydłowski, K.; Stodulski, D.; Mikaszewski, B.; Odroniec, A.; Musiał, N.; Thiel, M.; Czaplewska, P.; et al. Comparison of Peptidomes Extracted from Healthy Tissue and Tumor Tissue of the Parotid Glands and Saliva Samples. Int. J. Mol. Sci. 2024, 25, 8799. [Google Scholar] [CrossRef] [PubMed]
  17. Decock, J.; Comito, G.; Zaravinos, A. Editorial: Tumor Microenvironment, Inflammation, and Resistance to Immunotherapies. Front. Oncol. 2023, 13, 1215332. [Google Scholar] [CrossRef]
  18. Niu, T.; Zhou, F. Inflammation and Tumor Microenvironment. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2023, 48, 1899–1913. [Google Scholar] [CrossRef]
  19. Tricco, A.C.; Lillie, E.; Zarin, W.; O’Brien, K.K.; Colquhoun, H.; Levac, D.; Moher, D.; Peters, M.D.J.; Horsley, T.; Weeks, L.; et al. PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. Ann. Intern. Med. 2018, 169, 467–473. [Google Scholar] [CrossRef]
  20. Loy, A.H.C.; Putti, T.C.; Tan, L.K.S. Cyclooxygenase-2 Expression in Warthin’s Tumour. J. Laryngol. Otol. 2005, 119, 515–518. [Google Scholar] [CrossRef]
  21. Andreadis, D.; Epivatianos, A.; Mireas, G.; Nomikos, A.; Poulopoulos, A.; Yiotakis, J.; Barbatis, C. Immunohistochemical Detection of E-Cadherin in Certain Types of Salivary Gland Tumours. J. Laryngol. Otol. 2006, 120, 298–304. [Google Scholar] [CrossRef]
  22. Pantelis, A.; Wenghoefer, M.; Haas, S.; Merkelbach-Bruse, S.; Pantelis, D.; Jepsen, S.; Bootz, F.; Winter, J. Down Regulation and Nuclear Localization of Human β-Defensin-1 in Pleomorphic Adenomas of Salivary Glands. Oral. Oncol. 2009, 45, 526–530. [Google Scholar] [CrossRef] [PubMed]
  23. Tampouris, A.I.; Kandiloros, D.; Giotakis, I.; Gakiopoulou, H.; Lazaris, A.C. The Role of the VEGF-C/-D/Flt-4 Autocrine Loop in the Pathogenesis of Salivary Neoplasms. Pathol. Res. Pract. 2012, 208, 151–156. [Google Scholar] [CrossRef]
  24. Liu, G.X.; Lan, J.; Sun, Y.; Hu, Y.J.; Jiang, G.S. Expression of the Chemokine CCL28 in Pleomorphic Adenoma and Adenolymphoma of the Human Salivary Glands. Exp. Ther. Med. 2012, 4, 65–69. [Google Scholar] [CrossRef] [PubMed]
  25. Kehagias, N.; Epivatianos, A.; Sakas, L.; Andreadis, D.; Markopoulos, A.; Antoniades, K. Expression of N-Cadherin in Salivary Gland Tumors. Med. Princ. Pract. 2012, 22, 59–64. [Google Scholar] [CrossRef] [PubMed]
  26. Carlesimo, M.; Mari, E.; La Pietra, M.; Orsini, D.; Pranteda, G.; Pranteda, G.; Grimaldi, M.; Arcese, A. Occurrence of Salivary Gland Tumours in Two Patients Treated with Biological Agents. Int. J. Immunopathol. Pharmacol. 2012, 25, 297–300. [Google Scholar] [CrossRef] [PubMed]
  27. Jour, G.; West, K.; Ghali, V.; Shank, D.; Ephrem, G.; Wenig, B.M. Differential Expression of P16INK4A and Cyclin D1 in Benign and Malignant Salivary Gland Tumors: A Study of 44 Cases. Head Neck Pathol. 2013, 7, 224–231. [Google Scholar] [CrossRef]
  28. Andisheh Tadbir, A.; Khademi, B.; Malekzadeh, M.; Mardani, M.; Khademi, B. Upregulation of Serum Vascular Endothelial Growth Factor in Patients with Salivary Gland Tumor. Patholog Res. Int. 2013, 2013, 740582. [Google Scholar] [CrossRef]
  29. Aga, M.; Kondo, S.; Yamada, K.; Sawada-Kitamura, S.; Yagi-Nakanishi, S.; Endo, K.; Wakisaka, N.; Murono, S.; Kawano, M.; Yoshizaki, T. Warthin’s Tumor Associated with IgG4-Related Disease. Auris Nasus Larynx 2013, 40, 514–517. [Google Scholar] [CrossRef]
  30. Ohtomo, R.; Mori, T.; Shibata, S.; Tsuta, K.; Maeshima, A.M.; Akazawa, C.; Watabe, Y.; Honda, K.; Yamada, T.; Yoshimoto, S.; et al. SOX10 Is a Novel Marker of Acinus and Intercalated Duct Differentiation in Salivary Gland Tumors: A Clue to the Histogenesis for Tumor Diagnosis. Mod. Pathol. 2013, 26, 1041–1050. [Google Scholar] [CrossRef]
  31. Donadio, E.; Giusti, L.; Seccia, V.; Ciregia, F.; da Valle, Y.; Dallan, I.; Ventroni, T.; Giannaccini, G.; Sellari-Franceschini, S.; Lucacchini, A. New Insight into Benign Tumours of Major Salivary Glands by Proteomic Approach. PLoS ONE 2013, 8, e71874. [Google Scholar] [CrossRef]
  32. Ianez, R.C.F.; Coutinho-Camillo, C.M.; Buim, M.E.; Pinto, C.A.L.; Soares, F.A.; Lourenço, S.V. CD24 and CD44 in Salivary Gland Pleomorphic Adenoma and in Human Salivary Gland Morphogenesis: Differential Markers of Glandular Structure or Stem Cell Indicators? Histopathology 2013, 62, 1075–1082. [Google Scholar] [CrossRef] [PubMed]
  33. Karbanová, J.; Laco, J.; Marzesco, A.M.; Janich, P.; Voborníková, M.; Mokrý, J.; Fargeas, C.A.; Huttner, W.B.; Corbeil, D. Human PROMININ-1 (CD133) Is Detected in Both Neoplastic and Non-Neoplastic Salivary Gland Diseases and Released into Saliva in a Ubiquitinated Form. PLoS ONE 2014, 9, e98927. [Google Scholar] [CrossRef]
  34. Reshma, V.; Rao, K.; Priya, N.; Umadevi, H.; Smitha, T.; Sheethal, H. Expression of Maspin in Benign and Malignant Salivary Gland Tumors: An Immunohistochemical Study. Indian J. Dent. Res. 2014, 25, 346–351. [Google Scholar] [CrossRef] [PubMed]
  35. Aga, M.; Kondo, S.; Yamada, K.; Wakisaka, N.; Yagi-Nakanishi, S.; Tsuji, A.; Endo, K.; Murono, S.; Ito, M.; Muramatsu, M.; et al. Immunoglobulin Class Switching to IgG4 in Warthin Tumor and Analysis of Serum IgG4 Levels and IgG4-Positive Plasma Cells in the Tumor. Hum. Pathol. 2014, 45, 793–801. [Google Scholar] [CrossRef] [PubMed]
  36. Faur, A.C.; Lazar, E.; Cornianu, M. Vascular Endothelial Growth Factor (VEGF) Expression and Microvascular Density in Salivary Gland Tumours. APMIS 2014, 122, 418–426. [Google Scholar] [CrossRef] [PubMed]
  37. He, Q.; Takizawa, Y.; Hayasaka, T.; Masaki, N.; Kusama, Y.; Su, J.; Mineta, H.; Setou, M. Increased Phosphatidylcholine (16:0/16:0) in the Folliculus Lymphaticus of Warthin Tumor. Anal. Bioanal. Chem. 2014, 406, 5815–5825. [Google Scholar] [CrossRef]
  38. Andisheh-Tadbir, A.; Ashraf, M.J.; Khademi, B.; Ahmadi, S. Clinical Implication of CD166 Expression in Salivary Gland Tumor. Tumor Biol. 2015, 36, 2793–2799. [Google Scholar] [CrossRef]
  39. Wang, Y.; Zhou, J.; Zhang, Y.; Wang, L.; Liu, Y.; Fan, L.; Zhu, J.; Xu, X.; Huang, G.; Li, X.; et al. PRDMI Expression on the Epithelial Component but Not on Ectopic Lymphoid Tissues of Warthin Tumour. Oral Dis. 2015, 21, 432–436. [Google Scholar] [CrossRef] [PubMed]
  40. Haghshenas, M.R.; Khademi, B.; Faghih, Z.; Ghaderi, A.; Erfani, N. Immune Regulatory Cells and IL17-Producing Lymphocytes in Patients with Benign and Malignant Salivary Gland Tumors. Immunol. Lett. 2015, 164, 109–116. [Google Scholar] [CrossRef]
  41. Jaafari-Ashkavandi, Z.; Ashraf, M.J.; Nazhvani, A.D.; Azizi, Z. Caveolin-1 Overexpression in Benign and Malignant Salivary Gland Tumors. Tumor Biol. 2016, 37, 1863–1869. [Google Scholar] [CrossRef]
  42. Fonseca, F.P.; Bingle, L.; Santos-Silva, A.R.; Lopes, M.A.; de Almeida, O.P.; de Andrade, B.A.B.; Mariano, F.V.; Kowalski, L.P.; Rangel, A.L.C.A.; Martins, M.D.; et al. Semaphorins and Neuropilins Expression in Salivary Gland Tumors. J. Oral Pathol. Med. 2016, 45, 119–126. [Google Scholar] [CrossRef] [PubMed]
  43. Khademi, B.; Tajvarpour, M.; Mojtahedi, Z.; Haghshenas, M.R.; Erfani, N. T-Helper Type 1 and 2 Cytokine Levels in Patients with Benign and Malignant Salivary Gland Tumors. Iran. J. Immunol. 2016, 13, 9–15. [Google Scholar] [PubMed]
  44. Kim, J.; Song, J.S.; Roh, J.L.; Choi, S.H.; Nam, S.Y.; Kim, S.Y.; Cho, K.J. Increased Immunoglobulin G4-Positive Plasma Cells in Lymphadenoma of the Salivary Gland: An Immunohistochemical Comparison among Lymphoepithelial Lesions. Appl. Immunohistochem. Mol. Morphol. 2018, 26, 420–424. [Google Scholar] [CrossRef] [PubMed]
  45. Haghshenas, M.R.; Khademi, B.; Ashraf, M.J.; Ghaderi, A.; Erfani, N. Helper and Cytotoxic T-Cell Subsets (Th1, Th2, Tc1, and Tc2) in Benign and Malignant Salivary Gland Tumors. Oral. Dis. 2016, 22, 566–572. [Google Scholar] [CrossRef]
  46. Zare, R.; Malekzadeh, M.; Hashemi, M.; Khademi, B.; Andishe-Tadbir, A. Investigation of IL-33 Serum Levels in Patients with Benign and Malignant Salivary Gland Tumors. Cancer Biomark. 2018, 23, 61–65. [Google Scholar] [CrossRef] [PubMed]
  47. Sowa, P.; Misiolek, M.; Orecka, B.; Czecior, E.; Adamczyk-Sowa, M. Serum Levels of Selected Adipocytokines in Benign and Malignant Parotid Gland Tumor Patients. Cytokine 2018, 106, 40–44. [Google Scholar] [CrossRef]
  48. Sowa, P.; Misiolek, M.; Pasinski, B.; Bartosz, G.; Soszynski, M.; Adamczyk-Sowa, M.; Sadowska-Bartosz, I. Oxidative Stress Markers Patients with Parotid Gland Tumors: A Pilot Study. Biomed. Res. Int. 2018, 2018, 4340871. [Google Scholar] [CrossRef]
  49. Błochowiak, K.J.; Sokalski, J.; Bodnar, M.B.; Trzybulska, D.; Marszałek, A.K.; Witmanowski, H. Expression of VEGF165b, VEGFR1, VEGFR2 and CD34 in Benign and Malignant Tumors of Parotid Glands. Adv. Clin. Exp. Med. 2018, 27, 83–90. [Google Scholar] [CrossRef]
  50. Tenório, J.D.R.; da Silva, L.P.; Xavier, M.G.D.A.; Santana, T.; do Nascimento, G.J.F.; Sobral, A.P.V. Differential Expression of Cyclooxygenase-2 and Cyclin D1 in Salivary Gland Tumors. Eur. Arch. Oto-Rhino-Laryngol. 2018, 275, 2341–2347. [Google Scholar] [CrossRef]
  51. Andisheh-Tadbir, A.; Ashraf, M.J.; Gudarzi, A.; Zare, R. Evaluation of Glypican-3 Expression in Benign and Malignant Salivary Gland Tumors. J. Oral Biol. Craniofac Res. 2019, 9, 63–66. [Google Scholar] [CrossRef]
  52. Aslan, F.; Küçük, Ü. RANK and RANKL Expression in Salivary Gland Tumors. Ear Nose Throat J. 2020, 99, 475–480. [Google Scholar] [CrossRef] [PubMed]
  53. Baněčková, M.; Uro-Coste, E.; Ptáková, N.; Šteiner, P.; Stanowska, O.; Benincasa, G.; Colella, G.; Vondrák, J.; Michal, M.; Leivo, I.; et al. What Is Hiding behind S100 Protein and SOX10 Positive Oncocytomas? Oncocytic Pleomorphic Adenoma and Myoepithelioma with Novel Gene Fusions in a Subset of Cases. Hum. Pathol. 2020, 103, 52–62. [Google Scholar] [CrossRef] [PubMed]
  54. da Silva, L.P.; de Sousa Lopes, M.L.D.; Sarmento, A.S.C.; de Albuquerque Borges, M.; de Moura, S.R.S.; Sobral, A.P.V.; de Souza, L.B. Increased Expression of ALDH-1 Is Associated with Clinical Parameters of Salivary Glands Neoplasms. Exp. Mol. Pathol. 2020, 117, 104552. [Google Scholar] [CrossRef] [PubMed]
  55. Mochizuki, K.; Oishi, N.; Kawai, M.; Odate, T.; Tahara, I.; Inoue, T.; Kasai, K.; Kondo, T. Expressions of Cxcl12, Cxcl10 and Ccl18 in Warthin Tumors Characterized Pathologically by Having a Lymphoid Stroma with Germinal Centers. Histol. Histopathol. 2021, 36, 931–938. [Google Scholar] [CrossRef] [PubMed]
  56. Kobayashi, Y.; Kurose, N.; Guo, X.; Shioya, A.; Kitamura, M.; Tsuji, H.; Yamada, S. The Potential Role of Follicular Helper T Cells and Helper T Cells Type 1 in Warthin Tumour. Pathol. Res. Pract. 2021, 220, 153386. [Google Scholar] [CrossRef]
  57. Sahin, A.; Kars, A.; Kilic, K.; Ucar, H.B.; Sakat, M.S. Efficacy of the Systemic Immune Inflammation Index in Malignant and Benign Parotid Neoplasms. Cureus 2022, 14, e31878. [Google Scholar] [CrossRef]
  58. Haghshenas, M.R.; Erfani, N.; Khansalar, S.; Khademi, B.; Ashraf, M.J.; Razmkhah, M.; Ghaderi, A. Proteomics Study of Mesenchymal Stem Cell-Like Cells Obtained from Tumor Microenvironment of Patients with Malignant and Benign Salivary Gland Tumors. Cell J. 2022, 24, 196–203. [Google Scholar] [CrossRef]
  59. Laohavisudhi, F.; Chunchai, T.; Ketchaikosol, N.; Thosaporn, W.; Chattipakorn, N.; Chattipakorn, S.C. Evaluation of CD44s, CD44v6, CXCR2, CXCL1, and IL-1β in Benign and Malignant Tumors of Salivary Glands. Diagnostics 2022, 12, 1275. [Google Scholar] [CrossRef]
  60. Gaonkar, P.P.; Patankar, S.R.; Sridharan, G. Assessment of Angiogenesis Using Endoglin in Salivary Gland Tumors—An Immunohistochemical Study. J. Cancer Res. Ther. 2022, 18, 623–628. [Google Scholar] [CrossRef]
  61. Abbate, V.; Barone, S.; Orabona, G.D.; Bonavolontà, P.; Galdiero, M.R.; Cristinziano, L.; Modestino, L.; Iaconetta, G.; Califano, L. Role of Inflammation in Benign Salivary Gland Tumor Etiopathogenesis: An Evaluation through the Blood Inflammatory Biomarkers. Eurasian J. Med. Oncol. 2022, 6, 150–155. [Google Scholar] [CrossRef]
  62. Sowa, P.; Kasperczyk, S.; Dadok, A.; Misiołek, M.; Adamczyk-Sowa, M. Low-Intensity Whole-Body Oxidative Stress in Patients Withparotid Gland Tumors. Otolaryngol. Pol. 2022, 77, 19–25. [Google Scholar] [CrossRef] [PubMed]
  63. Jabbar, Z.; Khan, M.M.; Tariq, S.F.; Alam, S.; Arbab, K.N.; Khan, A.A.; Khalid, N.; Abdullah, L.; Jabbar, R. Association of Vascular Endothelial Growth Factor (VEGF) in Salivary Gland Tumors. Pak. J. Med. Health Sci. 2023, 17, 156–159. [Google Scholar] [CrossRef]
  64. Abbate, V.; Barone, S.; Borriello, G.; Troise, S.; Bonavolontà, P.; Pacella, D.; Vaira, L.A.; Turri-Zanoni, M.; Cuéllar, C.N.; Califano, L.; et al. Diagnostic Performance of Inflammatory Biomarkers and Cytological Analysis in Salivary Gland Tumors. Head Neck 2023, 45, 3015–3023. [Google Scholar] [CrossRef] [PubMed]
  65. Tateda, Y.; Suzuki, T.; Sato, T.; Izuhara, K.; Ise, K.; Shimada, H.; Murakami, K.; Murakami, K.; Nakamura, Y.; Ohta, N. Expression of Periostin in Benign Salivary Gland Tumors. Tohoku J. Exp. Med. 2024, 262, 105–113. [Google Scholar] [CrossRef]
  66. Ghalehbandi, S.; Yuzugulen, J.; Pranjol, M.Z.I.; Pourgholami, M.H. The Role of VEGF in Cancer-Induced Angiogenesis and Research Progress of Drugs Targeting VEGF. Eur. J. Pharmacol. 2023, 949, 175586. [Google Scholar] [CrossRef]
  67. Heloterä, H.; Kaarniranta, K. A Linkage between Angiogenesis and Inflammation in Neovascular Age-Related Macular Degeneration. Cells 2022, 11, 3453. [Google Scholar] [CrossRef]
  68. Rossi, E.; Sanz-Rodriguez, F.; Eleno, N.; Düwell, A.; Blanco, F.J.; Langa, C.; Botella, L.M.; Cabañas, C.; Lopez-Novoa, J.M.; Bernabeu, C. Endothelial Endoglin Is Involved in Inflammation: Role in Leukocyte Adhesion and Transmigration. Blood 2013, 121, 403–415. [Google Scholar] [CrossRef]
  69. Meurer, S.K.; Weiskirchen, R. Endoglin: An “Accessory” Receptor Regulating Blood Cell Development and Inflammation. Int. J. Mol. Sci. 2020, 21, 9247. [Google Scholar] [CrossRef]
  70. Jelic, M.D.; Mandic, A.D.; Maricic, S.M.; Srdjenovic, B.U. Oxidative Stress and Its Role in Cancer. J. Cancer Res. Ther. 2021, 17, 22–28. [Google Scholar] [CrossRef]
  71. Caliri, A.W.; Tommasi, S.; Besaratinia, A. Relationships among Smoking, Oxidative Stress, Inflammation, Macromolecular Damage, and Cancer. Mutat. Res. Rev. Mutat. Res. 2021, 787, 108365. [Google Scholar] [CrossRef]
  72. Wigner, P.; Grębowski, R.; Bijak, M.; Saluk-Bijak, J.; Szemraj, J. The Interplay between Oxidative Stress, Inflammation and Angiogenesis in Bladder Cancer Development. Int. J. Mol. Sci. 2021, 22, 4483. [Google Scholar] [CrossRef] [PubMed]
  73. Kaszak, I.; Witkowska-Piłaszewicz, O.; Niewiadomska, Z.; Dworecka-Kaszak, B.; Toka, F.N.; Jurka, P. Role of Cadherins in Cancer-A Review. Int. J. Mol. Sci. 2020, 21, 7624. [Google Scholar] [CrossRef] [PubMed]
  74. Huang, H.W.; Chang, C.C.; Wang, C.S.; Lin, K.H. Association between Inflammation and Function of Cell Adhesion Molecules Influence on Gastrointestinal Cancer Development. Cells 2021, 10, 67. [Google Scholar] [CrossRef] [PubMed]
  75. Walcher, L.; Kistenmacher, A.K.; Suo, H.; Kitte, R.; Dluczek, S.; Strauß, A.; Blaudszun, A.R.; Yevsa, T.; Fricke, S.; Kossatz-Boehlert, U. Cancer Stem Cells-Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Front. Immunol. 2020, 11, 1280. [Google Scholar] [CrossRef] [PubMed]
  76. Wu, S.; Tan, Y.; Li, F.; Han, Y.; Zhang, S.; Lin, X. CD44: A Cancer Stem Cell Marker and Therapeutic Target in Leukemia Treatment. Front. Immunol. 2024, 15, 1354992. [Google Scholar] [CrossRef]
  77. Hou, W.; Kong, L.; Hou, Z.; Ji, H. CD44 Is a Prognostic Biomarker and Correlated with Immune Infiltrates in Gastric Cancer. BMC Med. Genom. 2022, 15, 225. [Google Scholar] [CrossRef]
  78. Mesrati, M.H.; Syafruddin, S.E.; Mohtar, M.A.; Syahir, A. CD44: A Multifunctional Mediator of Cancer Progression. Biomolecules 2021, 11, 1850. [Google Scholar] [CrossRef]
  79. Cushing, K.; Higgins, P.D.R. Management of Crohn Disease: A Review. JAMA 2021, 325, 69–80. [Google Scholar] [CrossRef]
  80. Briani, C.; Visentin, A. Therapeutic Monoclonal Antibody Therapies in Chronic Autoimmune Demyelinating Neuropathies. Neurotherapeutics 2022, 19, 874–884. [Google Scholar] [CrossRef]
  81. Maximus, P.S.; Al Achkar, Z.; Hamid, P.F.; Hasnain, S.S.; Peralta, C.A. Adipocytokines: Are They the Theory of Everything? Cytokine 2020, 133, 155144. [Google Scholar] [CrossRef]
  82. Sy, A.L.; Hoang, M.P. SOX10. J. Clin. Pathol. 2023, 76, 649–653. [Google Scholar] [CrossRef] [PubMed]
  83. Bancaro, N.; Calì, B.; Troiani, M.; Elia, A.R.; Arzola, R.A.; Attanasio, G.; Lai, P.; Crespo, M.; Gurel, B.; Pereira, R.; et al. Apolipoprotein E Induces Pathogenic Senescent-like Myeloid Cells in Prostate Cancer. Cancer Cell 2023, 41, 602–619.e11. [Google Scholar] [CrossRef] [PubMed]
  84. Zhao, R.R.; Mao, X.R.; Wang, X.F.; Zheng, Y.; Wang, Y.P.; Zhou, Y.N. Role of Annexin A Family in Tumorigenesis and Chemoresistance of Gastric Cancer. Neoplasma 2022, 69, 251–263. [Google Scholar] [CrossRef] [PubMed]
  85. Xia, P.; Ji, X.; Yan, L.; Lian, S.; Chen, Z.; Luo, Y. Roles of S100A8, S100A9 and S100A12 in Infection, Inflammation and Immunity. Immunology 2024, 171, 365–376. [Google Scholar] [CrossRef]
  86. Ghosh, S.K.; McCormick, T.S.; Weinberg, A. Human Beta Defensins and Cancer: Contradictions and Common Ground. Front. Oncol. 2019, 9, 314. [Google Scholar] [CrossRef]
  87. Tang, S.; Ling, Z.; Jiang, J.; Gu, X.; Leng, Y.; Wei, C.; Cheng, H.; Li, X. Integrating the Tumor-Suppressive Activity of Maspin with P53 in Retuning the Epithelial Homeostasis: A Working Hypothesis and Applicable Prospects. Front. Oncol. 2022, 12, 1037794. [Google Scholar] [CrossRef]
  88. Corn, K.C.; Windham, M.A.; Rafat, M. Lipids in the Tumor Microenvironment: From Cancer Progression to Treatment. Prog. Lipid Res. 2020, 80, 101055. [Google Scholar] [CrossRef]
  89. Kim, S.J. Immunological Function of Blimp-1 in Dendritic Cells and Relevance to Autoimmune Diseases. Immunol. Res. 2015, 63, 113–120. [Google Scholar] [CrossRef]
  90. Ren, L.; Zhou, P.; Wu, H.; Liang, Y.; Xu, R.; Lu, H.; Chen, Q. Caveolin-1 Is a Prognostic Marker and Suppresses the Proliferation of Breast Cancer. Transl. Cancer Res. 2021, 10, 3797–3810. [Google Scholar] [CrossRef]
  91. Lu, T.; Zhang, Z.; Pan, X.; Zhang, J.; Wang, X.; Wang, M.; Li, H.; Yan, M.; Chen, W. Caveolin-1 Promotes Cancer Progression via Inhibiting Ferroptosis in Head and Neck Squamous Cell Carcinoma. J. Oral. Pathol. Med. 2022, 51, 52–62. [Google Scholar] [CrossRef]
  92. Zheng, X.; Liu, X.; Lei, Y.; Wang, G.; Liu, M. Glypican-3: A Novel and Promising Target for the Treatment of Hepatocellular Carcinoma. Front. Oncol. 2022, 12, 824208. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, D.; Gao, Y.; Zhang, Y.; Wang, L.; Chen, G. Glypican-3 Promotes Cell Proliferation and Tumorigenesis through up-Regulation of β-Catenin Expression in Lung Squamous Cell Carcinoma. Biosci. Rep. 2019, 39, BSR20181147. [Google Scholar] [CrossRef] [PubMed]
  94. Udagawa, N.; Koide, M.; Nakamura, M.; Nakamichi, Y.; Yamashita, T.; Uehara, S.; Kobayashi, Y.; Furuya, Y.; Yasuda, H.; Fukuda, C.; et al. Osteoclast Differentiation by RANKL and OPG Signaling Pathways. J. Bone Miner. Metab. 2021, 39, 19–26. [Google Scholar] [CrossRef]
  95. De Leon-Oliva, D.; Barrena-Blázquez, S.; Jiménez-Álvarez, L.; Fraile-Martinez, O.; García-Montero, C.; López-González, L.; Torres-Carranza, D.; García-Puente, L.M.; Carranza, S.T.; Álvarez-Mon, M.Á.; et al. The RANK-RANKL-OPG System: A Multifaceted Regulator of Homeostasis, Immunity, and Cancer. Medicina 2023, 59, 1752. [Google Scholar] [CrossRef] [PubMed]
  96. Dorafshan, S.; Razmi, M.; Safaei, S.; Gentilin, E.; Madjd, Z.; Ghods, R. Periostin: Biology and Function in Cancer. Cancer Cell Int. 2022, 22, 315. [Google Scholar] [CrossRef]
  97. Sonnenberg-Riethmacher, E.; Miehe, M.; Riethmacher, D. Periostin in Allergy and Inflammation. Front. Immunol. 2021, 12, 722170. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram.
Figure 1. PRISMA flow diagram.
Ijms 25 12558 g001
Figure 2. Main directions of research.
Figure 2. Main directions of research.
Ijms 25 12558 g002
Table 1. Characteristic of included studies.
Table 1. Characteristic of included studies.
StudyNumber of Patients with Benign SGTStudied MoleculeEffect on Benign Tumor Tissues or Serum
Loy et al., 2005 [20]21COX-2Overexpression/
upregulation
Andreadis et al., 2006 [21]54E-cadherinStrong overexpression
Pantelis et al., 2008 [22]5Human beta-defensin 1/2/3Decreased expression of hBD-1,
no significant difference regarding hBD-2 and hBD-3
Tampouris et al., 2011 [23]20VEGF-C/VEGF-D/
VEGFR-3 (flt-4)
All 3 strongly expressed in PA,
VEGF-C/D moderately expressed or not expressed in other benign tumors
flt-4 strongly expressed in all benign tumors
Liu et al., 2012 [24]72Chemokine CCL28Decreased expression
Kehagias et al., 2012 [25]18N-cadherinExpressed in some cases of Warthin’s tumor,
not expressed in other benign samples
Carlesimo et al., 2012 [26]2TNF-alphaNot analyzed in the tissue
Jour et al., 2013 [27]14Cyclin D1 and p16INK4AAlmost all benign tumors expressed p16 and cyclin D1
Andisheh Tadbir et al., 2013 [28]31VEGFHigher concentration in serum
Aga et al., 2013 [29] 1IgG4Serum and tissue levels increased
Ohtomo et al., 2013 [30]14SOX-10Present in all PA, ME, absent in some of WT
Donadio et al., 2013 [31]3626 proteinsCharacteristic separate chains of proteins for PA and WT
Ianez et al., 2013 [32]101CD24/CD44CD24/CD44 positive by immunochemistry,
CD44, also by PCR
Karbanová et al., 2014 [33]10Prominin-1 (CD133)Overexpressed
Reshma et al., 2014 [34]15MaspinOverexpressed
Aga et al., 2014 [35]37IgG4Serum and tissue levels increased in some of WT,
mRNA overexpressed in some of WT, not increased/expressed in PA
Faur et al., 2014 [36]20VEGFModerately positive VEGF expression
He et al., 2014 [37]3PhosphatidylocholineIncreased in WT
Andisheh-Tadbir et al., 2014 [38]15CD166Overexpressed
Wang et al., 2015 [39]40PRDM1Pverexpressed
Haghshenas
et al., 2015 [40]
19IL-17-producing lymphocytes and CTLA4+ lymphocytesIncreased concentration in serum
Jaafari-Ashkavandi
et al., 2015 [41]
15Caveolin-1overexpressed
Fonseca et al., 2015 [42]120Semaphorins and neutrophilinsNo significant difference vs. control group
Khademi et al., 2016 [43]50INF y, IL-4No significant difference vs. control group
Kim et al., 2016 [44]8IgG4Increased concentration in serum
Haghshenas et al., 2016 [45]15Th1, Th2, Tc1, Tc2 lymphocytesNo significant difference vs. control group
Zare et al., 2018 [46]14IL-33Slightly overexpressed
Sowa et al., 2018 [47]51AdipocytokinesSerum levels of adiponectin and visfatin elevated,
leptin elevated in males,
IL-6 no difference
Sowa et al., 2018 [48]26Protein oxidation productsSerum level of Total Antioxidant Capacity of Blood Serum and thiol groups decreased,
Advanced oxidation protein products increased
Błochowiak et al., 2018 [49]45VEGF165b, VEGFR1, VEGFR2 and CD34No significant difference vs. control group
Tenorio et al., 2018 [50]38COX-2, cyclin D1Underexpressed vs. malignant tumors
Andisheh-Tadbir et al., 2019 [51]17Glypican-3 (GPC-3)Overexpressed
Aslan et al., 2020 [52]38RANK, RANKLUnderexpressed vs. malignant tumors
Baneckova et al., 2020 [53]89S1009, SOX-10Expressed in 10% of oncocytic cases of PA/ME,
negative in oncocytoma
Da Silva et al., 2020 [54]51ALDH-1Expressed in parenchyma of all benign tumors
Mochizuki et al., 2021 [55]40CXCL10, CXCL12, CCL18WT mostly positive for expression,
PA mostly negative
Kobayashi et al., 2021 [56]64Th, TfhDifferent for solid-type and cyst-type tumor
Sahin et al., 2022 [57]185Systemic Immune-Inflammation Index (SII)Lower vs. malignant tumors
Haghshenas et al., 2022 [58]5MSC cellsMany cell expressed—CD44, CD73, CD90, CD105, and CD166, heat shock protein 70 (Hsp70), keratin, type II cytoskeletal 7 (CK-7),
Laohavisudhi et al., 2022 [59]13CD44s, CD44v6, CXCR2, CXCL1, and IL-1βCD44s, CD44v6, CXCR2—increased in benign tumors
Gaonkar et al., 2022 [60]15EndoglinHigher expression vs. control group,
lower vs. malignant tumors
Abbate et al., 2022 [61]191NLR, SII, PRLAll markers significantly increased vs. control group
Sowa et al., 2022 [62]52Oxidative stress markersPlasma lipofuscin increased in all benign tumors,
Cu-Zn SOD decreased in WT
Jabbar et al., 2023 [63]30VEGFOverexpression
Abbate et al., 2023 [64]140Inflammatory biomarkers
SII, SIRI, PLR, and NLR
SIRI showed highest accuracy in determining malignancy,
decreased vs. malignant tumors
Tateda et al., 2024 [65]38PeriostinOverexpression in 32 out of 38 benign tumors
Abbreviations: PA—pleomorphic adenoma, WT—Warthin’s tumor, ME—myoepithelioma, VEGF—vascular endothelial growth factor, MSC—mesenchymal stem cells, NLR—neutrophil to lymphocyte ratio, SII—systemic immune-inflammation index, PLR—platelet to lymphocyte ratio, SIRI—systemic inflammation response index.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Szydłowski, K.; Puchalski, M.; Ołdziej, S.; Kasprzyk-Tryk, A.; Skorek, A.; Tretiakow, D. The Impact of Inflammation on the Etiopathogenesis of Benign Salivary Gland Tumors: A Scoping Review. Int. J. Mol. Sci. 2024, 25, 12558. https://doi.org/10.3390/ijms252312558

AMA Style

Szydłowski K, Puchalski M, Ołdziej S, Kasprzyk-Tryk A, Skorek A, Tretiakow D. The Impact of Inflammation on the Etiopathogenesis of Benign Salivary Gland Tumors: A Scoping Review. International Journal of Molecular Sciences. 2024; 25(23):12558. https://doi.org/10.3390/ijms252312558

Chicago/Turabian Style

Szydłowski, Konrad, Michał Puchalski, Stanisław Ołdziej, Agnieszka Kasprzyk-Tryk, Andrzej Skorek, and Dmitry Tretiakow. 2024. "The Impact of Inflammation on the Etiopathogenesis of Benign Salivary Gland Tumors: A Scoping Review" International Journal of Molecular Sciences 25, no. 23: 12558. https://doi.org/10.3390/ijms252312558

APA Style

Szydłowski, K., Puchalski, M., Ołdziej, S., Kasprzyk-Tryk, A., Skorek, A., & Tretiakow, D. (2024). The Impact of Inflammation on the Etiopathogenesis of Benign Salivary Gland Tumors: A Scoping Review. International Journal of Molecular Sciences, 25(23), 12558. https://doi.org/10.3390/ijms252312558

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

Article Metrics

Back to TopTop