Next Article in Journal
Restriction of Manganese Intake Prevents the Onset of Brain Manganese Overload in Zip14−/− Mice
Next Article in Special Issue
Differential Expression of Peroxisomal Proteins in Distinct Types of Parotid Gland Tumors
Previous Article in Journal
A Transcription Factor Regulates Gene Expression in Chloroplasts
Previous Article in Special Issue
Aging-Related Metabolic Dysfunction in the Salivary Gland: A Review of the Literature
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

2021 Update on Diagnostic Markers and Translocation in Salivary Gland Tumors

1
Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, Ebene -1, 35392 Giessen, Germany
2
Institute for Pathology, Justus Liebig University, Langhansstrasse 10, 35392 Gießen, Germany
3
Institute for Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Koellikerstrasse 6, 97070 Würzburg, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(13), 6771; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22136771
Submission received: 31 May 2021 / Revised: 20 June 2021 / Accepted: 21 June 2021 / Published: 24 June 2021
(This article belongs to the Special Issue Recent Advances in Salivary Gland and Their Function)

Abstract

:
Salivary gland tumors are a rare tumor entity within malignant tumors of all tissues. The most common are malignant mucoepidermoid carcinoma, adenoid cystic carcinoma, and acinic cell carcinoma. Pleomorphic adenoma is the most recurrent form of benign salivary gland tumor. Due to their low incidence rates and complex histological patterns, they are difficult to diagnose accurately. Malignant tumors of the salivary glands are challenging in terms of differentiation because of their variability in histochemistry and translocations. Therefore, the primary goal of the study was to review the current literature to identify the recent developments in histochemical diagnostics and translocations for differentiating salivary gland tumors.

1. Introduction

Salivary gland tumors are rare neoplasias of the head and neck that have an annual incidence of 7.03 to 8.58/100,000 [1]. The prevalence of salivary gland cancer is even lower, with 16/1,000,000 [2]. There are 33 existing tumor entities of epithelial origin, and 2–5% of the tumors are non-epithelial. Based on the known entities, it can be highlighted that epithelial salivary gland tumors consist of 10 benign and 23 malignant subtypes, which are the most commonly prevailing in daily clinical life [3]. The primary problem lies in diagnosing a salivary gland tumor, as it is a relatively uncommon form of lesion with wide variations in histological and biological aspects [4]. These tumors have a significant morphological diversity with multiple overlapping features and low incidence rates [5]. The research also elaborates the present situation of the scientific research in immunohistochemistry with paraffin-embedded slides and translocation with fluorescence in situ hybridization (FISH).

1.1. Histology of the Healthy Salivary Gland

The salivary glands can be divided into two groups: the minor and major salivary glands—the latter consisting of the parotid, the submandibular, and the sublingual glands. However, the origins of the glands vary. The parotid gland is derived from ectoderm in the sixth and the seventh week along with the submandibular gland. In addition, the submandibular and sublingual glands originate from endoderm. The sublingual gland develops two weeks later than the other two in the ninth week. Furthermore, the parotid gland is considered to be a pure serous gland, while the submandibular gland produces mixed, predominantly serous fluid. On the other hand, the sublingual gland is a mixed, predominantly mucous gland, and the minor salivary glands produce seromucous fluid [6].
The histology of the salivary glands can be categorized into two compartments: luminal (acinar and ductal cells) and abluminal cells (myoepithelial and basal cells). Ideally, routine diagnosis of salivary gland tumors is carried out by hematoxylin–eosin (HE) staining, and immunohistochemistry is preferred to enhance the accuracy of the analysis. A normal salivary gland reacts positively to CEA, EMA, gross cystic disease fluid protein-15, Pan-cytokeratin (CK) (AE1/AE3), mitochondrial markers, alpha-amylase, CK14, p63, calponin, MSA, SMA, and vimentin. However, the reaction with Ki-67 (MIB-1), S-100, and HER2 varies, and androgen receptor (AR), EBER in situ hybridization, lymphoid cell markers, Melan A, p53, and the renal cell carcinoma marker CD10 react negatively [7]. Table 1 presents various studies comprising various cell types, the markers used, and the reaction of these markers.

1.2. Current Status of Salivary Gland Tumors and Markers

Salivary gland cancer is categorized as head and neck cancer and constitutes most head and neck cancer diagnoses. Salivary gland tumors differ in glandular cell type, including morphological diversity. Thus, these tumors arise from either the major or several minor salivary glands. Parotid gland tumors are rare and are characterized by heterogeneous entities. The morphological diversity of such tumors and their rarity make them challenging to diagnose. Another factor that adds to the complexity of diagnosis is their subtypes. Diagnosis based on hematoxylin and eosin staining procedures remains the gold standard of salivary gland pathology. Immunohistochemistry combined with hematoxylin and eosin staining can be more useful in specific applications. Ki-67 has the prognostic ability and the potential to differentiate between benign basal cell adenomas and malignant basal cell adenocarcinomas. Similarly, CD43 and Cyclin-A are beneficial for diagnosing adenoid cystic carcinomas and carcinoma ex pleomorphic adenomas. In particular, beta-catenin has two significant functionalities—namely cell–cell adhesion and transcription activation—for various genes that are used for controlling cell proliferation. Platelet-derived growth factor (PDGF) comprises a group of polypeptides responsible for cell proliferation and chemotaxis. Additionally, cyclooxygenase (Cox) enzymes are responsible for catalyzing the synthesis of prostanoids in arachidonic acid metabolism. A study revealed that CEA, Cox-1, Cox-2, PGDF-beta, and WISP-1 and β-catenin are potential diagnostic markers for differentiating benign and malignant parotid gland lesions [8]. This emphasizes that diagnostic markers vary in terms of the type of salivary gland tumor and are used in different forms for accurate diagnosis.

1.2.1. Acinar and Adenoid Cell Carcinomas

An adenoid cystic carcinoma (ADCC) is a malignant form of biphasic epithelial tumor comprising myoepithelial and ductal cells with a high recurrence rate. It is most often encountered at 50 to 70 years of age in the central and minor salivary glands [9]. ADCC constitutes 4.4% of all salivary gland tumors and 11.8% of salivary gland malignancies [9]. These tumors have different patterns such as cribriform, tubular, and solid patterns and mixed forms of growth patterns within the same lesion. Adenoid cystic carcinomas also express ductal and myoepithelial/basal cell markers such as CK7, CAM 5.2, calponin, p63, SOX10, S100, and SMA. The acinar or ductal epithelial cells are generally positive for keratins (CK7 and CAM 5.2) and epithelial membrane antigen (EMA). Usually, they tend to be focally positive or negative for high-molecular-weight keratins (HMWK, CK5/6, and 34BE12). Additionally, they are negative for p63, myoid markers (smooth muscle myosin heavy chain (SMMHC), smooth muscle actin (SMA), calponin), and CK20 (weak focal expression can be observed in rare salivary gland carcinomas). However, myoepithelial cells were found to be positive for p63, myoid markers (SMMHC, SMA, calponin), vimentin, S100, and HMWKs (CK5/6, 34BE12), while they show a weak expression for CK7 and CAM 5.2 and no expression for EMA [10]. ADCCs react positively with calponin, CD43, CK7, CK8, CK14, CK17, CK19, C-Kit, DOG-1, KIT, Maspin, Mcl-1, MCM2, Mit, MYB, NM23, p63, p73, S-100, SMA, SMM, SOX4, and SOX10. They sometimes react positively to caldesmon and GFAP. Apocrine, carbohydrate Ag19-9, carbonic anhydrase VI, CEA, CD9, CK20, HMGA-2, LPLUNC1, PLAG1, SPLUNC1, and SPLUNC2 usually react negatively.
Table 2 and Table 3 present a summary of the literature in terms of adenoid cell carcinomas (ACCs) and acinic cell carcinomas and the corresponding reactions of the patients’ probes to the markers.
Acinic cell carcinomas (ACCs) demonstrate serous acinar differentiation alongside intercalated ductal epithelial differentiation. These tumors have various growth patterns: solid/lobular, microcystic, papillary cystic, and follicular. It is more frequently diagnosed in women (58.8%) than in men (41.2%) [29]. Acinic cell carcinomas usually express CK7 and CAM 5.2. In addition, normal acinar cells express amylase, and most acinic cell carcinomas are found to be negative for this marker. Most reactions of ACC with alpha-1-antitrypsin, carbonic anhydrase VI, chymotrypsin, CK8, CK19, DOG-1, KIT, MCM2, p53, p73, SOX10, synaptophysin, and vimentin are positive. Apocrine, maspin, Mit, and S-100 sometimes react negatively. For amylase, caldesmon, calponin, chromogranin, CK14, CK17, CK20, GFAP, HMGA-2, LPLUNC1, MYB, p63, PLAG1, SMA, SMM, SPLUNC1, and SPLUNC2, adverse effects were observed.
Table 3. Analysis of results for acinic cell carcinoma patient probes.
Table 3. Analysis of results for acinic cell carcinoma patient probes.
StudySample Size ACCMarkerReactionin %
[30]SixAlpha-1-Antitrypsin3 of 650%
[31]Three of parotid originAmylase0 of 30%
[11]Four of parotid originApocrine1 of 425%
[11]Four of parotid originCaldesmon0 of 40%
[11,12]Eight of salivary gland and head and neck origin; four of parotid originCalponin0 of 8; 0 of 40%
[14]28 of salivary gland originCarbonic anhydrase VI (CA6)28 of 28100%
[30]SixChromogranin0 of 60%
[30]SixChymotrypsin4 of 667%
[11]Four of parotid gland originCK74 of 4100%
[11]Four of parotid originCK 84 of 4100%
[11]Four of parotid originCK140 of 40%//+
[11]Four of parotid originCK 170 of 40%//+
[11]Four of parotid originCK194 of 4100%
[11]Four of parotid originCK200 of 40%
[14,18]28 of salivary gland origin; 28 of salivary gland originDOG-128 of 28; 28 of 28100%; 100%
[12]Eight of salivary gland and head and neck originGFAP0 of 80%
[19]One of salivary gland origin or potential mimickersHMGA-20 of 10%
[21]Seven of upper aerodigestive tract originKIT (CD117)6 of 786%
[22]Nine of salivary gland originLPLUNC10 of 90%
[23,24]Five of salivary gland origin; eleven of salivary gland originMaspin5 of 5; 0 of 11100%; 0%
[23]Five of salivary gland originMCM25 of 5100%
[11]Four of parotid originMit1 of 425%
[21]Seven of upper aerodigestive tract originMYB0 of 70%
[13]Fivep532 of 540%
[12,27]Eight of salivary gland and head and neck origin; eightp630 of 8; 0 of 80%; 0%
[27]Eightp735 of 863%
[19]One of salivary gland origin or potential mimickersPLAG10 of 10%
[12,14]Eight of salivary gland and head and neck origin; 28 of salivary gland originS-1001 of 8; 2 of 2813%; 7.1%
[11,12]Eight of salivary gland and head and neck origin; four of parotid originSMA0 of 8; 0 of 40%
[11]Four of parotid originSMM0 of 40%
[22]Nine of salivary gland originSPLUNC10 of 90%
[22]Nine of salivary gland originSPLUNC20 of 90%
[12]Eight of salivary gland and head and neck originSOX108 of 8100%
[30]SixSynaptophysin4 of 667%
[14]28 of salivary gland originVimentin23 of 2882%

1.2.2. Mucoepidermoid Carcinomas

Mucoepidermoid carcinomas (MECs) are malignant epithelial neoplasms composed of mucous, epidermoid, intermediate columnar, clear, and oncolytic cells. Approximately 26% of malignant salivary gland tumors are mucoepidermoid carcinoma [1]. It is usually discovered in the fifth decade of life with a slightly elevated prevalence in the female population [32]. Recently, it was discovered that early-stage MECs may grow predominantly intracystically and are thus easier to resect. This assumption emphasizes the importance of an early diagnosis for a good outcome [33]. MECs appear positive for CK5, CK6, CK7, CK8, CK14, CK18, CK19, EMA, CEA, and p63. Similarly, they are negative for CK20, SMA, MSA, and S100. However, p63 is an efficient marker used to differentiate acinic cell carcinomas from mucoepidermoid carcinomas. On the other hand, myoepithelial carcinomas are malignant tumors formed in salivary glands with high myoepithelial differentiation. These tumor cells are diverse, and include spindled, stellate, epithelioid and clear cells with some resemblance to sarcoma, melanoma or other tumors (Figure 1). Immunoreactivity for both types of keratins and a minimum of one myoepithelial marker reaction is required to diagnose such tumors. Myoepithelial carcinomas mainly express vimentin (100%), calponin (75–100%), S100 (82–100%), CAM 5.2 (89%), EMA (27%), and SMA (35–50%), which shows that some markers are more frequently expressed than others, while some are occasionally expressed. Calponin is sensitive to myoepithelial carcinomas and is considered to be the most specific marker for these carcinomas. However, a combination of markers, such as CK AE/13, CAM5.2, CK5/6, SMA, S100, and calponin, can be effective for accurate diagnosis [8]. Table 4 provides insights on works in the literature that have used a wide range of markers and shows the reaction of the patients to those markers.

1.2.3. Polymorphous Adenocarcinomas

Polymorphous adenocarcinomas are low-grade malignant epithelial carcinomas. The formation of these tumors takes place mainly in the minor salivary glands with mild cytologic features. These tumors consist of diverse growth patterns in different areas such as lobular, papillary, cribriform, ductal, and tubular. Such tumors express CK AE1/3, CAM 5.2, EMA, 34BE12, p53, p63, vimentin, and S100 with an infrequent level of SMA [8]. It was observed that 96% of canalicular adenomas show weak and strong cytoplasmic staining for GFAP in the ductal and myoepithelial cells. GFAP is considered to be more helpful in distinguishing pleomorphic low-grade adenocarcinomas from pleomorphic adenomas [38].

1.2.4. Salivary Duct Carcinomas

Salivary duct carcinomas are aggressive and malignant epithelial tumors that form from intralobular and interlobular excretory ducts. These tumors express AR, GCDFP-15, CK AE1/3, CK7, 34BE12, CEA, and EMA. However, they can occasionally be positive for ER, PR, and S-100. Ki-67 shows an increased expression of more than 25%. Androgen receptors occur more frequently in men compared to women in salivary duct carcinomas. A rate of 80% overexpression of HER2/neu and p53 for salivary duct carcinomas is connected to a weaker level of prognosis [8].

1.2.5. Pleomorphic Adenomas

Pleomorphic adenomas (PMAs) are the most common benign salivary gland tumors with an occurrence rate of 86% [39]. They were first described in 1859 by Billroth [6]. Histologically, PMAs consist of myoepithelial and epithelial cells in different morphological patterns (Figure 1) [40]. A PMA can develop into a benign but metastasizing subtype, the metastasizing pleomorphic adenoma (MPA), or a malignant mixed tumor called carcinoma ex-pleomorphic adenoma and carcinosarcoma. The probability of malignant transformation rises with longer existence of the tumor and increased patient age [41,42]. PMAs react mostly positively to calponin, CD9, GFAP, Mcl-2, NM23, p63, S-100, SMA, and Sox10. PLAG1 is specific for PMAs and is therefore widely used as diagnostic marker. Amylase, DOG1, HMGA-2, KIT, and MYB are sometimes positive. The usually negatively reacting markers are carbonic anhydrase VI, LPLUNC1, SPLUNC1, and SPLUNC2. Table 5 lists the relevant studies including the sample sizes of PMAs, the marker used, and the reactions.

2. State-of-the-Art Methods in Pre-Operative Diagnosis of Salivary Gland Tumors

Salivary gland tumors are rare and account for about 2% to 6.5% of all head and neck neoplasms. The rarity and morphological variations make them difficult to diagnose. Therefore, state-of-the-art diagnostic imaging methods have been adopted over the years for better differential diagnosis. Imaging provides crucial information for accurate localization of salivary gland tumors (e.g., localization in superficial and deep lobes) and differentiation between benignancy and malignancy and plays an important role in the staging procedure of salivary gland cancer. In addition, imaging also allows for differentiating recurrent malignant tumors from post-treatment changes and monitoring patient health after therapy. Magnetic resonance (MR) imaging is the most preferred modality for the evaluation of salivary gland tumors. Routine pre-contrast MR imaging plays an important role in accurate localization and assessment of locoregional extension of salivary gland tumors. Contrast-based MR imaging assesses the perineural spread of salivary cancer malignancy, which commonly occurs in patients with adenoid cystic carcinomas. Advanced MR imaging procedures such as diffusion-weighted MR imaging and dynamic contrast-enhanced (DCE) imaging effectively characterize particular salivary gland tumors. MR imaging is also practical for differentiating recurrence from post-treatment changes. Similarly, routine post-contrast computed tomography (CT) scans help to evaluate skull base cortical invasions, whereas PET-CT effectively detects distant metastases of salivary gland cancer [44].
It is essential to identify whether mass stems from the deep or superficial lobe of the parotid gland, as the surgical approaches for these tumors vary. Therefore, tumor localization is of utmost importance for accurate diagnosis. Another crucial aspect of diagnosis is identifying the intraparotidal pathway of the facial nerve, which is performed using anatomical landmarks. The three-dimensional cross-sectional imaging technique proposed by Atkinson highlights how the proposed approach can help demonstrate the facial nerve. A T2-weighted MR sequence can distinguish between ducts and nerves of low signal intensity. Generally, pleomorphic adenomas have a progressive radiocontrast agent enhancement, whereas Warthin tumors appear to be washed out. These tumors require significant enhancement, which is achievable using DCE and ADC MR imaging techniques. Similarly, T1- and T2-weighted imaging and contrast-enhanced imaging are used to identify the signal intensities and densities based on MR and CT imaging. Myoepitheliomas appear to be small and round tumors with smooth contours [44].

3. Fine Needle Aspiration of Salivary Gland Tumors

Fine needle aspiration (FNA) of salivary gland tumors is an important diagnostic tool for preoperative risk stratification. It helps to distinguish benign from malignant tumors and is, therefore, indispensable for therapeutic decisions [45]. FNA also helps to limit the need for surgical intervention and can thereby reduce treatment costs [46]. However, FNA has a lower sensitivity with high heterogeneity; therefore, it is advised to use as an additional tool for the detection of malignancies [45,47]. For example, in PMA, at least 25% of the aspirate smear consists of fibrillary stroma with intermixed myoepithelial cells surrounding gland ductal structures [48]. As MECs are very heterogeneous, diagnosis by FNA is difficult. Depending on the aspirated portion, false diagnoses such as abscesses or PMA may occur. To confirm a low-grade MEC by FNA, cells such as intermediate cells, mucin-secreting cells, and squamous cells must be present [49]. ACC often presents as normal acinar cells in FNA and is, thus, underdiagnosed. However, if the tumor is of predominant papillary architecture type, it resembles adenocarcinoma. Typically, acinar-cell-like tumor cells are seen in the FNA of ACCs, taking over their pattern. In some cases, one also sees capillary plexus or even papillary formations around a fibrovascular core [50]. In summary, FNA is suitable to diagnose homogeneous tumors with a high probability. For heterogeneous tumors such as salivary gland tumors, FNA is not suitable as the sole diagnostic tool, but can be used as an additional one.

4. Translocations

Translocations are considered to account for about 20% of all forms of cancers. In recent years, translocations and their resultant fusion oncogenes were found to be rare for epithelial tumors. These are genetic aberrations that are mainly found in hematolymphoid and soft-tissue neoplasms and are rare in other tumors. Difficulty arises in finding epithelial tumors with translocations because of growing carcinoma or acquiring the karyotypes of these tumors. It has recently been found that multiple salivary cancers comprise translocations, namely MEC, ACC, and other forms of carcinomas such as MASC and HCCC, among several more. Translocation emergence detection is more feasible with next-generation molecular markers, gene fusions, and whole gene sequencing. Salivary glands are appropriate for finding translocations as they show low-grade malignancy with relative homogeneity. Translocations become essential as every tumor with a specific subtype looks histologically similar. At the same time, they also appear different from typical salivary gland elements. Certain tumors with a complex nature of genomic abnormalities occur in the background of preneoplastic dysplasias. Considering these factors, it can be highlighted that tumors such as epithelial-myoepithelial carcinomas and basal cell adenocarcinomas may have translocations [51].

4.1. Translocations in Pleomorphic Adenomas (PAs)

Pleomorphic adenomas (PAs) comprise a myxoid matrix, chondroid, and other elements related to myoepithelial cells. These cells are often in the forms of epithelioid, spindled, plasmacytoid, and clear cells. Pleomorphic adenomas are known to conceal translocations that involve PLAG1 or HMGA2 in most cases. Such tumors have high morbidity rates and are subject to frequent recurrence if not resected promptly with wide safety margins. These tumors are an aggressive form of malignant salivary gland tumors with a significantly increased mortality rate. Abnormalities in PA occur in the 8q12 region, while some have balanced reciprocal translocations, including PLAG1, and some show wide variations in abnormalities with various fusions. The most commonly known fusion partner is CTNNB1, which is the gene coding for beta-catenin. PLAG1 upregulations are usually detected using immunohistochemistry but are not confined to those cases. Another fusion partner is the LIFR, which has similar upregulations as CTNNB1. Another minor known partner is SII that has been cloned. The ring chromosomes in specific PAs have been identified as fused with FGFR1 to PLAG1, which is one of the causes of similar-appearing tumors. PAs with numerous aberrations and the ones without any identifiable aberrations usually have a similar appearance. This suggests that FISH procedures for such gene types have limited diagnostic utility without prognostic significance [51].

4.2. Translocations in Mucoepidermoid Carcinomas (MECs)

MECs are some of the most common salivary gland carcinomas. They can be found in major or minor salivary glands. These tumors are also likely to occur in other locations such as the tracheobronchial tree, cervix, skin, or the upper respiratory tract. MECs have large ducts and comprise three cell types: mucinous cells, epidermoid cells, and intermediate cells. Typically, the occurrence of MECs should alert the clinicians to perform additional diagnostic tests. These tumors can mimic the mucinous components or have cystic variations. There were no prior diagnostic or prognostic markers until CRTC1 and MAML2 fusion came into use. This fusion is often used for prognosis, and about 55% of cases tended to have a positive outcome. Fusion-positive cases are more likely to be observed in younger patients with decreased recurrence rates. MAMLM2 FISH has been used for higher-grade translocation-positive MECs for prognostic purposes in some studies [51]. Another study has pointed out that 46% of high-grade MECs are found to be translocation-positive with minimal anaplasia and were high grade due to additional factors. On the contrary, high-grade translocation-negative MECs appear to have significant anaplasias with frequent mucinous differentiation. The MAML2 FISH fusion provides better outcomes, but the results are less convincing [52].

4.3. Translocations in Adenoid Cystic Carcinomas (ACCs)

ACCs are the most common form of salivary gland carcinoma after MECs. They are of intercalated duct origin, which signifies that there is a participation of both ductal and myoepithelial cells. ACCs comprise a combination of tubular and cribriform elements with bilayers and visible myoepithelial layers on the outer region. Recent studies include t (6;9) (q22–23; p23–24) in various karyotypes of ACCs. Cloning of the fusion of MYB oncogene and NFIB transcription factor gene has been tested for salivary glands, head and neck, and breast sites. However, MYB activation is required for fusion-negative cases. MYB overexpression shown with RT-PCR appeared more in fusion-positive cases and was confirmed by immunohistochemistry for the MYB protein. MYB protein expression tended to be negative or focal in non-ACC salivary cases. It has been noted that MYB expression in specific ACC cases has been strong but not very sensitive overall.
On the other hand, MYB has been found to play a crucial role in ACCs irrespective of the type of fusion. Additionally, MYB RNA has appeared to be fusion-positive for ACCs in all anatomic sites. Nevertheless, it is unclear whether translocation in ACC can be a useful diagnostic marker [51]. Table 6 offers an overview of translocation types and prevalence assessed in various studies.

5. Analysis of Markers

In recent years, many new immunohistochemical and molecular markers have been proposed for diagnosis, some of them being successful in distinguishing between the many different entities. In these papers, following markers proved to be helpful: carbonic anhydrase VI is only positive in ACC, PLAG1 in PMA, S-100 and SOX10 in PMA and ADCC, and LPLUNC and SPLUNC2 in MEC. Figure 2, Figure 3, Figure 4 and Figure 5 depict the possible markers that have made it easy to differentiate between tumors.

6. Future Directives

Salivary gland tumors are complex due to the variations in their morphological characteristics. In order to achieve accurate detection and dose delivery, there is a need for the combination of emerging biological markers and next-generation diagnostic technologies. Furthermore, several types of research have highlighted the importance of saliva becoming a significant source of insights on possible diagnosis and prognosis.
In the newest edition of the WHO Classification of Head and Neck Tumors from 2017, the increasing importance of translocation diagnostics is underlined. Regarding the selected tumors in this paper, eight molecular alterations are mentionable: PMA has a PLAG1 fusion in over 50% of cases and an HMGA2 fusion in approximately 15%. MECs possess a CRTC1-MAML2 alteration with a probability of 40–80% and a CRTC3-MAML2 alteration with around 5% probability. A CDKN2A deletion happens in 35% of MECs. MYB alterations are frequent in 80% of ADCC cases, followed by MYBL1 in 10% and NOTCH1 in 5–10% [41].
At present, there are about 100 types of salivary biomarkers that currently exist, ranging from non-organic compound biomarkers (sodium, calcium); protein biomarkers (p53, alpha-amylase); and DNA-, RNA-, and microRNA-related biomarkers (p53 gene codon 63, miR-125a) to metabolomics biomarkers (valine and lactic acid) and other miscellaneous biomarkers. Salivary-based biomarkers have gained importance for assessing the risk of malignant diseases. DNA- or RNA-based methods help to identify the tumors without traditional approaches. PCR identification techniques ensure accurate measurement, and new techniques such as saliva-based identification and salivary epithelial cells are being developed to improve accurate diagnosis. Salivary epithelial cells are known to secrete proteins in the bloodstream. Hence, they are considered a potential marker for the target site. Emerging salivary gland markers include molecular and protein-based markers that can provide critical information on the disease status. The increasing number of cases of oral cancer in recent years has led to research in salivary biomarkers for oral cancers such as salivary gland carcinomas [53].

7. Conclusions

Molecular analysis is an emerging approach of sequencing that helps clinicians to characterize tumors effectively. Evidence of translocation by molecular markers is not essential for diagnosis. However, the number of molecular alterations can refine the diagnosis. Use of the present molecular analysis and translocation needs to become more established in daily clinical life. On the other hand, technological advancements have introduced powerful diagnostic imaging techniques that can identify and aid in further prognosis of the disease and classification according to the malignancies. Thus, in time, several novel molecular alterations are expected be discovered, which might potentially increase the importance of markers.

Author Contributions

Conceptualization, S.K.; methodology, M.T.M., T.D. and S.K.; validation, M.T.M., C.W., S.K.; formal analysis, M.T.M., and C.W.; investigation, M.T.M., T.D., S.K.; resources, S.K., and S.E.; data curation, M.T.M.; writing—original draft preparation, M.T.M. and S.K.; writing—review and editing, M.T.M., C.W. and S.K.; visualization M.T.M.; and S.K., supervision, S.K.; project administration, S.K.; funding acquisition, S.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bradley, P.J.; McGurk, M. Incidence of Salivary Gland Neoplasms in a Defined UK Population. Br. J. Oral Maxillofac. Surg. 2013, 51, 399–403. [Google Scholar] [CrossRef]
  2. Del Signore, A.G.; Megwalu, U.C. The Rising Incidence of Major Salivary Gland Cancer in the United States. Ear Nose Throat J. 2017, 96, E13–E16. [Google Scholar] [CrossRef]
  3. Rousseau, A.; Badoual, C. Head and Neck: Salivary Gland Tumors: An Overview. Atlas Genet. Cytogenet. Oncol. Haematol. 2011, 15. [Google Scholar] [CrossRef]
  4. Cunha, J.L.S.; Coimbra, A.C.P.; Silva, J.V.R.; Nascimento, I.S.; Andrade, M.E.; Oliveira, C.R.; Almeida, O.; Soares, C.; Sousa, S.F.; Albuquerque-Júnior, R.L. Epidemiologic Analysis of Salivary Gland Tumors over a 10-Years Period Diagnosed in a Northeast Brazilian Population. Med. Oral Patol. Oral Cirugia Bucal 2020, 25, e516–e522. [Google Scholar] [CrossRef]
  5. Speight, P.M.; Barrett, A.W. Salivary Gland Tumours: Diagnostic Challenges and an Update on the Latest WHO Classification. Diagn. Histopathol. 2020, 26, 147–158. [Google Scholar] [CrossRef]
  6. Hellquist, H.; Skalova, A. Histopathology of the Salivary Glands; Springer: Berlin/Heidelberg, Germany, 2014; ISBN 978-3-540-46912-4. [Google Scholar]
  7. Nagao, T.; Sato, E.; Inoue, R.; Oshiro, H.; Takahashi, R.H.; Nagai, T.; Yoshida, M.; Suzuki, F.; Obikane, H.; Yamashina, M.; et al. Immunohistochemical Analysis of Salivary Gland Tumors: Application for Surgical Pathology Practice. ACTA Histochem. Cytochem. 2012, 45, 269–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Curran, A.E.; Allen, C.M.; Beck, F.M.; Damm, D.D.; Murrah, V.A. Distinctive Pattern of Glial Fibrillary Acidic Protein Immunoreactivity Useful in Distinguishing Fragmented Pleomorphic Adenoma, Canalicular Adenoma and Polymorphous Low Grade Adenocarcinoma of Minor Salivary Glands. Head Neck Pathol. 2007, 1, 27–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Eveson, J.W.; Cawson, R.A. Salivary gland tumours. A review of 2410 cases with particular reference to histological types, site, age and sex distribution. J. Pathol. 1985, 146, 51–58. [Google Scholar] [CrossRef] [PubMed]
  10. Schneider, S.; Kloimstein, P.; Pammer, J.; Brannath, W.; Grasl, M.C.; Erovic, B.M. New Diagnostic Markers in Salivary Gland Tumors. Eur. Arch. Otorhinolaryngol. 2014, 271, 1999–2007. [Google Scholar] [CrossRef]
  11. Foschini, M.P.; Marucci, G.; Eusebi, V. Low-grade mucoepidermoid carcinoma of salivary glands: Characteristic immunohistochemical profile and evidence of striated duct differentiation. Virchows Arch. 2002, 440, 536–542. [Google Scholar] [CrossRef]
  12. 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] [PubMed] [Green Version]
  13. Tsubochi, H.; Suzuki, T.; Suzuki, S.; Ohashi, Y.; Ishibashi, S.; Moriya, T.; Fujimura, S.; Sasano, H. Immunohistochemical study of basaloid squamous cell carcinoma, adenoid cystic and mucoepidermoid carcinoma in the upper aerodigestive tract. Anticancer Res. 2000, 20, 1205–1211. [Google Scholar]
  14. Hsieh, M.S.; Jeng, Y.M.; Jhuang, Y.L.; Chou, Y.H.; Lin, C.Y. Carbonic anhydrase VI: A novel marker for salivary serous acinar differentiation and its application to discriminate acinic cell carcinoma from mammary analogue secretory carcinoma of the salivary gland. Histopathology 2016, 68, 641–647. [Google Scholar] [CrossRef]
  15. Sakamoto, K.; Nakamura, Y.; Nakashima, T. Immunohistochemical distribution of CD9 in parotid gland tumors. Auris Nasus Larynx 2004, 31, 49–55. [Google Scholar] [CrossRef]
  16. Woo, V.L.; Bhuiya, T.; Kelsch, R. Assessment of CD43 expression in adenoid cystic carcinomas, polymorphous low-grade adenocarcinomas, and monomorphic adenomas. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2006, 102, 495–500. [Google Scholar] [CrossRef] [PubMed]
  17. Edwards, P.C.; Bhuiya, T.; Kelsch, R.D. C-kit expression in the salivary gland neoplasms adenoid cystic carcinoma, polymorphous low-grade adenocarcinoma, and monomorphic adenoma. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2003, 95, 586–593. [Google Scholar] [CrossRef] [Green Version]
  18. Chenevert, J.; Duvvuri, U.; Chiosea, S.; Dacic, S.; Cieply, K.; Kim, J.; Shiwarsk, D.; Seethala, R.R. DOG1: A novel marker of salivary acinar and intercalated duct differentiation. Mod. Pathol. 2012, 25, 919–929. [Google Scholar] [CrossRef]
  19. Foo, W.C.; Jo, V.Y.; Krane, J.F. Usefulness of translocation-associated immunohistochemical stains in the fine-needle aspiration diagnosis of salivary gland neoplasms. Cancer Cytopathol. 2016, 124, 397–405. [Google Scholar] [CrossRef]
  20. Mino, M.; Pilch, B.Z.; Faquin, W.C. Expression of KIT (CD117) in neoplasms of the head and neck: An ancillary marker for adenoid cystic carcinoma. Mod. Pathol. 2003, 16, 1224–1231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. West, R.B.; Kong, C.; Clarke, N.; Gilks, T.; Lipsick, J.; Cao, H.; Kwok, S.; Montgomery, K.D.; Varma, S.; Le, Q.T. MYB expression and translocation in adenoid cystic carcinomas and other salivary gland tumors with clinicopathologic correlation. Am. J. Surg. Pathol. 2011, 35, 92–99. [Google Scholar] [CrossRef] [Green Version]
  22. Vargas, P.A.; Speight, P.M.; Bingle, C.D.; Barrett, A.W.; Bingle, L. Expression of PLUNC family members in benign and malignant salivary gland tumours. Oral Dis. 2008, 14, 613–619. [Google Scholar] [CrossRef] [Green Version]
  23. Ghazy, S.E.; Helmy, I.M.; Baghdadi, H.M. Maspin and MCM2 Immunoprofiling in Salivary Gland Carcinomas. Diagn. Pathol. 2011, 6, 1–8. [Google Scholar] [CrossRef] [Green Version]
  24. Schwarz, S.; Ettl, T.; Kleinsasser, N.; Hartmann, A.; Reichert, T.-E.; Driemel, O. Loss of Maspin Expression Is a Negative Prognostic Factor in Common Salivary Gland Tumors. Oral Oncol. 2008, 44, 563–570. [Google Scholar] [CrossRef] [PubMed]
  25. Heiduschka, G.; Erovic, B.M.; Pammer, J.; Kotowski, U.; Kaider, A.; Ch. Grasl, M.; Thurnher, D. Mcl-1 expression is up-regulated in malignancies of the parotid gland. Dis. Markers 2011, 30, 229–233. [Google Scholar] [CrossRef]
  26. do Nascimento, K.C.; de Faria, P.R.; Dib, L.L.; Ferreira de Aguiar, M.C.; Cardoso, S.V.; Chen, J.; Loyola, A.M. Immunohistochemical Localization of the NM23 Protein in Salivary Gland Neoplasms with Distinct Biological Behavior. Virchows Arch. 2006, 449, 660–666. [Google Scholar] [CrossRef]
  27. Seethala, R.R.; LiVolsi, V.A.; Zhang, P.J.; Pasha, T.L.; Baloch, Z.W. Comparison of P63 and P73 Expression in Benign and Malignant Salivary Gland Lesions. Head Neck 2005, 27, 696–702. [Google Scholar] [CrossRef]
  28. Pramoonjago, P.; Baras, A.S.; Moskaluk, C.A. Knockdown of Sox4 Expression by RNAi Induces Apoptosis in ACC3 Cells. Oncogene 2006, 25, 5626–5639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Stewart, A.K.; Bland, K.I.; McGinnis, L.S.; Morrow, M.; Eyre, H.J. Clinical highlights from the National Cancer Data Base, 2000. CA: A Cancer J. Clin. 2000, 50, 171–183. [Google Scholar] [CrossRef] [PubMed]
  30. Skacel, M.; Ormsby, A.H.; Petras, R.E.; McMahon, J.T.; Henricks, W.H. Immunohistochemistry in the Differential Diagnosis of Acinar and Endocrine Pancreatic Neoplasms. Appl. Immunohistochem. Mol. Morphol. 2000, 8, 203–209. [Google Scholar] [CrossRef]
  31. Morely, D.J.; Hodes, M.E. Amylase Expression in Human Parotid Neoplasms: Evidence by In Situ Hybridization for Lack of Transcription of the Amylase Gene. J. Histochem. Cytochem. 1988, 36, 487–491. [Google Scholar] [CrossRef]
  32. Kokemueller, H.; Brueggemann, N.; Swennen, G.; Eckardt, A. Mucoepidermoid carcinoma of the salivary glands--clinical review of 42 cases. Oral Oncol. 2005, 41, 3–10. [Google Scholar] [CrossRef]
  33. Capodiferro, S.; Ingravallo, G.; Limongelli, L.; Mastropasqua, M.G.; Tempesta, A.; Favia, G.; Maiorano, E. Intra-Cystic (In Situ) Mucoepidermoid Carcinoma: A Clinico-Pathological Study of 14 Cases. J. Clin. Med. 2020, 9, 1157. [Google Scholar] [CrossRef] [Green Version]
  34. Butler, R.T.; Spector, M.E.; Thomas, D.; McDaniel, A.S.; McHugh, J.B. An immunohistochemical panel for reliable differentiation of salivary duct carcinoma and mucoepidermoid carcinoma. Head Neck Pathol. 2014, 8, 133–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Pires, F.R.; De Almeida, O.P.; De Araujo, V.C.; Kowalski, L.P. Prognostic factors in head and neck mucoepidermoid carcinoma. Arch. Otolaryngol. Head Neck Surg. 2004, 130, 174–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Zyada, M.M.; Grawish, M.E.; Elsabaa, H.M. Predictive value of cyclooxygenase 2 and Bcl-2 for cervical lymph node metastasis in mucoepidermoid carcinoma. Ann. Diagn. Pathol. 2009, 13, 313–321. [Google Scholar] [CrossRef] [PubMed]
  37. Alos, L.; Lujan, B.; Castillo, M.; Nadal, A.; Carreras, M.; Caballero, M.; de Bolos, C.; Cardesa, A. Expression of Membrane-Bound Mucins (MUC1 and MUC4) and Secreted Mucins (MUC2, MUC5AC, MUC5B, MUC6 and MUC7) in Mucoepidermoid Carcinomas of Salivary Glands. Am. J. Surg. Pathol. 2005, 29, 806–813. [Google Scholar] [CrossRef]
  38. Zhu, S.; Schuerch, C.; Hunt, J. Review and Updates of Immunohistochemistry in Selected Salivary Gland and Head and Neck Tumors. Arch. Pathol. Lab. Med. 2015, 139, 55–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Subhashraj, K. Salivary Gland Tumors: A Single Institution Experience in India. Br. J. Oral Maxillofac. Surg. 2008, 46, 635–638. [Google Scholar] [CrossRef]
  40. Seifert, G.; Langrock, I.; Donath, K. A pathological classification of pleomorphic adenoma of the salivary glands (author’s transl). Hno 1976, 24, 415–426. [Google Scholar]
  41. Seethala, R.R.; Stenman, G. Update from the 4th Edition of the World Health Organization Classification of Head and Neck Tumours: Tumors of the Salivary Gland. Head Neck Pathol. 2017, 11, 55–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Bradley, P.J. Metastasizing pleomorphic salivary adenoma’ should now be considered a low-grade malignancy with a lethal potential. Curr. Opin. Otolaryngol. Head Neck Surg. 2005, 13, 123–126. [Google Scholar] [CrossRef]
  43. Matsuyama, A.; Hisaoka, M.; Nagao, Y.; Hashimoto, H. Aberrant PLAG1 expression in pleomorphic adenomas of the salivary gland: A molecular genetic and immunohistochemical study. Virchows Arch. 2011, 458, 583–592. [Google Scholar] [CrossRef]
  44. Abdel Razek, A.A.K.; Mukherji, S.K. State-of-the-Art Imaging of Salivary Gland Tumors. Neuroimaging Clin. N. Am. 2018, 28, 303–317. [Google Scholar] [CrossRef] [PubMed]
  45. Schmidt, R.L.; Hall, B.J.; Wilson, A.R.; Layfield, L.J. A systematic review and meta-analysis of the diagnostic accuracy of fine-needle aspiration cytology for parotid gland lesions. Am. J. Clin. Pathol. 2011, 136, 45–59. [Google Scholar] [CrossRef] [Green Version]
  46. Layfield, L.J.; Gopez, E.; Hirschowitz, S. Cost efficiency analysis for fine-needle aspiration in the workup of parotid and submandibular gland nodules. Diagn. Cytopathol. 2006, 34, 734–738. [Google Scholar] [CrossRef] [PubMed]
  47. Schmidt, R.L.; Hunt, J.P.; Hall, B.J.; Wilson, A.R.; Layfield, L.J. A systematic review and meta-analysis of the diagnostic accuracy of frozen section for parotid gland lesions. Am. J. Clin. Pathol. 2011, 136, 729–738. [Google Scholar] [CrossRef] [PubMed]
  48. Griffith, C.C.; Pai, R.K.; Schneider, F.; Duvvuri, U.; Ferris, R.L.; Johnson, J.T.; Seethala, R.R. Salivary gland tumor fine-needle aspiration cytology: A proposal for a risk stratification classification. Am. J. Clin. Pathol. 2015, 143, 839–853. [Google Scholar] [CrossRef] [Green Version]
  49. Vasudevan, G.; Bishnu, A.; Singh, B.M.K.; Singh, V.K. Mucoepidermoid Carcinoma of Salivary Gland: Limitations and Pitfalls on FNA. J. Clin. Diagn. Res. 2017, 11, ER04–ER06. [Google Scholar] [CrossRef]
  50. Bansal, A.; Kaur, M.; Dalal, V.; Siraj, F. Papillary Pattern in Acinic Cell Carcinoma of Parotid Gland: A Potential Diagnostic Pitfall on FNAC. J. Cytol. 2018, 35, 57–59. [Google Scholar] [CrossRef]
  51. Weinreb, I. Translocation-Associated Salivary Gland Tumors: A Review and Update. Adv. Anat. Pathol. 2013, 20, 367–377. [Google Scholar] [CrossRef]
  52. Seethala, R.R.; Dacic, S.; Cieply, K.; Kelly, L.M.; Nikiforova, M.N. A Reappraisal of the MECT1/MAML2 Translocation in Salivary Mucoepidermoid Carcinomas. Am. J. Surg. Pathol. 2010, 34, 1106–1121. [Google Scholar] [CrossRef] [PubMed]
  53. Krishnamurthy, S. Salivary Gland Disorders: A Comprehensive Review. World J. Stomatol. 2015, 4, 56–71. [Google Scholar] [CrossRef]
Figure 1. Comparison of the structural differences between control, pleomorphic adenoma (PMA), mucoepidermoid carcinoma (MEC), and acinar cell carcinoma (ACC) parotid gland tissue stained with HE staining. Control parotid tissue (A) is compared with PMA, which is composed of different cells of epithelial and mesenchymal lineage differentiations (B). ACC tumor cells resemble acinar cells in structure and pattern (C). MEC consists mostly of squamous epithelium and mucus-forming epithelium (D). Bars represent (AD) 100 μm.
Figure 1. Comparison of the structural differences between control, pleomorphic adenoma (PMA), mucoepidermoid carcinoma (MEC), and acinar cell carcinoma (ACC) parotid gland tissue stained with HE staining. Control parotid tissue (A) is compared with PMA, which is composed of different cells of epithelial and mesenchymal lineage differentiations (B). ACC tumor cells resemble acinar cells in structure and pattern (C). MEC consists mostly of squamous epithelium and mucus-forming epithelium (D). Bars represent (AD) 100 μm.
Ijms 22 06771 g001
Figure 2. PMA marker reaction.
Figure 2. PMA marker reaction.
Ijms 22 06771 g002
Figure 3. MEC marker reaction.
Figure 3. MEC marker reaction.
Ijms 22 06771 g003
Figure 4. ADCC marker reaction.
Figure 4. ADCC marker reaction.
Ijms 22 06771 g004
Figure 5. ACC marker reaction.
Figure 5. ACC marker reaction.
Ijms 22 06771 g005
Table 1. Normal salivary gland.
Table 1. Normal salivary gland.
Normal Salivary GlandMarkerReaction
GeneralAndrogen receptorNegative
EBER in situ hybridizationNegative
Ki-67 (MIB-1)Few cells positive
GeneralMelan ANegative
P53Negative
GeneralRenal cell carcinoma/CD10Negative
GeneralS-100Variable
GeneralCEAPositive
Acinar cellsCK14Positive
Abluminal cellsP63Positive
Abluminal cellsCK, AE1/AE3Positive
Myoepithelial cellsCalponinPositive
Myoepithelial cellsGFAPPositive (variable)
Myoepithelial cellsMSAPositive
Myoepithelial cellsSMAPositive
Myoepithelial cellsVimentinPositive
Luminal cellsEMAPositive
Gross cystic disease fluid proteinPositive
CK, AE1/AE3Positive
HER2Negative to
weakly positive
Striated duct cellsMitochondrialPositive
Alpha-amylasePositive
Table 2. Adenoid cell carcinomas.
Table 2. Adenoid cell carcinomas.
StudySample Size ADCCMarkerReactionin %
[11]Three of upper aerodigestive tract originApocrine0 of 30%
[11]Three of upper aerodigestive tract originCaldesmon1 of 333%
[11,12]23 of salivary gland and head and neck origin; three of upper aerodigestive tract originCalponin18 of 23; 3 of 378%; 100%
[13]FiveCarbohydrate Ag 19-90 of 50%
[14]Six of salivary gland originCarbonic anhydrase VI (CA6)0 of 60%
[13]FiveCEA0 of 50%
[15]Three of parotid gland originCD90 of 30%
[16]Four of parotid originCD434 of 4100%
[13]FiveCEA0 of 50%
[11]Three of upper aerodigestive tract originCK73 of 3100%
[11]Three of upper aerodigestive tract originCK83 of 3100%
[11]Three of upper aerodigestive tract originCK143 of 3100%
[11]Three of upper aerodigestive tract originCK173 of 3100%
[11]Three of upper aerodigestive tract originCK193 of 3100%
[11]Three of upper aerodigestive tract originCK200 of 30%
[17]15 of salivary gland originC-Kit15 of 15100%
[18]24 of salivary gland originDOG-117 of 2470%
[12]23 of salivary gland and head and neck originGFAP5 of 2321%
[19]Nine of salivary gland origin or potential mimickersHMGA-20 of 90%
[19,20,21]11 of salivary gland origin or potential mimickers; 66 of the head and neck area and breast; 37 of salivary gland originKIT4 of 11; 62 of 66 (H300 (54 of 66)/A4502 (58 of 66)); 37 of 3736%;
94%;
100%
[22]13 of salivary gland originLPLUNC10 of 130%
[23,24]14 of salivary gland origin; 25 of salivary gland originMaspin12 of 14; 19 of 2586%; 76%
[25]13 of parotid originMcl-112 of 1392%
[23]14 of salivary gland originMCM212 of 1486%
[11]Three of upper aerodigestive tract originMit2 of 366%
[19,21]Nine; 37 of salivary gland originMYB4 of 11; 24 of 3736%; 65%
[26]Six of upper aerodigestive tract and lung origin, 14 of salivary gland origin, one of maxilla originNM23 68,70%
[12,27]23 of salivary gland and head and neck origin; 16 of salivary glandp6321 of 23; 16 of 1691%; 100%
[27]16 of salivary glandp7316 of 16100%
[19]Eleven of salivary gland origin or potential mimickersPLAG10 of 110%
[12]23 of salivary gland and head and neck originS-10021 of 2391%
[11,12]23 of salivary gland and head and neck origin; three of upper aerodigestive tract originSMA21 of 23;
3 of 3
91%;
100%
[11]Three of upper aerodigestive tract originSMM2 of 366%
[22]13 of salivary gland originSPLUNC10 of 130%
[22]13 of salivary gland originSPLUNC20 of 130%
[28]28SOX428 of 28100%
[12]23 of salivary gland and head and neck originSOX1022 of 2396%
Table 4. Results of patient probes with mucoepidermoid carcinomas to various markers.
Table 4. Results of patient probes with mucoepidermoid carcinomas to various markers.
StudySample Size MECMarkerReactionin %
[31]Five of parotid originAmylase0 of 50%
[34]59 of upper aerodigestive tract originAndrogen receptor0 of 590%
[11]10 of upper aerodigestive tract originApocrine0 of 100%
[35]173 of upper aerodigestive tract originBcl-2 63%
[11]10 of upper aerodigestive tract originCaldesmon0 of 100%
[11,12]Six; 10 of salivary gland and head and neck originCalponin0 of 6; 1 of 100%; 10%
[13]SevenCarbohydrate Ag 19-94 of 757.10%
[14]Five of salivary gland originCarbonic anhydrase VI (CA6)0 of 50%
[13,35]173 of upper aerodigestive tract originCEA2 of 728.57%; 68.6%
[35]173 of upper aerodigestive tract originC-erb-2 80%
[15]Six of parotid originCD95 of 683%
[34]59 of upper aerodigestive tract originCK5/660 of 6493.75%
[11]10 of upper aerodigestive tract originCK710 of 10100%
[11]10 of upper aerodigestive tract originCK 810 of 10100%
[11]10 of upper aerodigestive tract originCK1410 of 10100%
[11]10 of upper aerodigestive tract originCK 1710 of 10100%
[11]10 of upper aerodigestive tract originCK1910 of 10100%
[11]10 of upper aerodigestive tract originCK201 of 1010%
[36]11 of upper aerodigestive tract originCox-211 of 11100%
[18]Eight of salivary gland originDOG-13 of 837.50%
[12]Six of salivary gland and head and neck originGFAP2 of 633%
[34]71 of upper aerodigestive tract originHer2/neu26 of 7137%
[19]Three of salivary gland origin or potential mimickersHMGA-20 of 30%
[20,21]Nine of head and neck origin; 23 of upper aerodigestive tract originKIT (CD117)H300 (1 of 9)/A4502 (0 of 9); 10 of 231%; 43%
[22]10 of salivary gland originLPLUNC110 of 10100%
[23,24]15 of salivary gland origin; 15 of salivary gland originMaspin15 of 15;
13 of 15
100%;
86.7%
[25]12 of parotid originMcl-211 of 1292%
[23]15 of salivary gland originMCM215 of 15100%
[11]10 of upper aerodigestive tract originMit10 of 10100%
[37]40 of salivary gland originMUC140 of 40100%
[37]40 of salivary gland originMUC22 of 405%
[37]40 of salivary gland originMUC438 of 4095%
[37]40 of salivary gland originMUC5AC29 of 4072%
[37]40 of salivary gland originMUC5B33 of 4082%
[37]40 of salivary gland originMUC613 of 4032%
[37]40 of salivary gland originMUC72 of 405%
[19,21]23 of the upper aerodigestive tract; three of salivary gland origin or potential mimickersMYB1 of 23;
1 of 3
4%;
33%
[26]Seven of upper aerodigestive tract, nine of salivary gland origin, one of maxilla, and two of mandibula originNM2318 of 1992.80%
[13,35]Seven; seven of upper aerodigestive tract originp534 of 757.1%;
16.4%
[12,27,34]Six of salivary gland and head and neck origin; four; 65 of upper aerodigestive tract originp632 of 6; 4 of 4; 62 of 6533%; 100%; 95.3%
[27]Fourp734 of 4100%
[35]No sample size number availablePCNA 92.9%
[19]Three of salivary gland origin or potential mimickersPLAG10 of 30%
[12]Six of salivary gland and head and neck originS-1001 of 617%
[11,12]10 of upper aerodigestive tract origin; six of salivary gland and head and neck originSMA0 of 10; 0/60%
[11]10 of upper aerodigestive tract originSMM0 of 100%
[22]10 of salivary gland originSPLUNC10 of 100%
[22]10 of salivary gland originSPLUNC210 of 10100%
[12]Six of salivary gland and head and neck originSOX100 of 60%
Table 5. Pleomorphic adenoma result analysis.
Table 5. Pleomorphic adenoma result analysis.
StudySample Size PMAMarkerReaction
[31]10 of parotid originAmylase10%
(1 of 10)
[12]10 (thereof four carcinomas)Calponin70%
(7 of 10)
[19,20,21]30; 16; 16 of salivary gland originKIT10%; 19%; 44%
[14]Five of salivary gland originCarbonic anhydrate VIZero
(0 of 5)
[15]16 of parotid originCD968.75%
(11 of 16)
[18]14 of salivary gland originDOG17%
(1 of 14)
[22]10 of salivary gland originSPLUNC10% (0 of 10)
[22]10
of salivary gland origin
SPLUNC20% (0 of 10)
[22]10
of salivary gland origin
LPLUNC10% (0 of 10)
[12]10 of salivary gland and head and neck origin
(thereof four carcinomas)
GFAP90% (9 of 10)
[26]16NM2375%
[12]10 of salivary gland and head and neck origin
(thereof four carcinomas)
P6380% (8 of 10)
[25]30 of parotid originMcl-273% (22 of 30)
[12]10 of salivary gland and head and neck origin
(thereof four carcinomas)
S100100% (10 of 10)
[12]10 of salivary gland and head and neck origin
(thereof four carcinomas)
SMA70% (7 of 10)
[19,21]29 of salivary gland origin or potential mimickers; 16 of salivary gland originMYB4% (1 of 4); 6% (1 of 16)
[19,43]30 of salivary gland origin or potential mimickers; 45 of salivary gland originPLAG173% (22 of 30); 100% (45 of 45)
Table 6. Overview of translocation types and prevalence.
Table 6. Overview of translocation types and prevalence.
TumorGeneChromosomal RearrangementPrevalence
PMAPLAG1 fusions8q12 translocations>50%
HMGA2 fusions12q13–15 translocations~15%
MECCRTC1-MAML2t (11; 19) (q21; p13)40–80%
CRTC3-MAML2t (11; 15) (q21; q26)~5%
CDKN2A deletion9p21.3~35%
ADCCMYB fusion/activation6q22–23 translocations~80%
MYBL1 fusion/activation8q13 translocations~10%
NOTCH1 mutation-5–10%
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Meyer, M.T.; Watermann, C.; Dreyer, T.; Ergün, S.; Karnati, S. 2021 Update on Diagnostic Markers and Translocation in Salivary Gland Tumors. Int. J. Mol. Sci. 2021, 22, 6771. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22136771

AMA Style

Meyer MT, Watermann C, Dreyer T, Ergün S, Karnati S. 2021 Update on Diagnostic Markers and Translocation in Salivary Gland Tumors. International Journal of Molecular Sciences. 2021; 22(13):6771. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22136771

Chicago/Turabian Style

Meyer, Malin Tordis, Christoph Watermann, Thomas Dreyer, Süleyman Ergün, and Srikanth Karnati. 2021. "2021 Update on Diagnostic Markers and Translocation in Salivary Gland Tumors" International Journal of Molecular Sciences 22, no. 13: 6771. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22136771

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