eISSN: 1897-4309
ISSN: 1428-2526
Contemporary Oncology/Współczesna Onkologia
Current issue Archive Manuscripts accepted About the journal Supplements Addendum Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Publication charge Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
3/2024
vol. 28
 
Share:
Share:
Review paper

Molecular landscape of salivary gland malignancies. What is already known?

Julia Pikul
1
,
Anna Rzepakowska
1

  1. Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Warsaw, Warsaw, Poland
Contemp Oncol (Pozn) 2024; 28 (3): 201–216
Online publish date: 2024/10/15
Article file
- Molecular landscape.pdf  [0.15 MB]
Get citation
 
PlumX metrics:
 

Introduction

Malignancies of the salivary glands are rare and account for approximately 5–8.5% of all head and neck cancers (HNC) [13]. Their occurrence is rare, with an annual incidence of 0.69 cases per 100,000 [4, 5]; however, the mortality rate is 40% [1]. Moreover, an increase of approximately 50% in both morbidity and mortality is predicted in the near future [6]. Salivary gland cancers (SGCs) are characterised by miscellaneous disease courses and clinical behaviours that contribute to unfavourable patient outcomes [2]. Among SGCs, more than 20 histopathological varieties have been classified by the World Health Organisation. Mucoepidermoid carcinoma (MEC) is the most common type of cancer, followed by acinic cell carcinoma (AcCC), adenoid cystic carcinoma (AdCC), carcinoma ex-pleomorphic adenoma (Ca ex PA), and adenocarcinoma (AC) [2, 7, 8]. The number of histopathological features interfering with benign lesions might also contribute to misdiagnosis and inappropriate management [911]. The incidence of these tumours is greater in males, and the risk of development increases with age. Former exposure to radiotherapy is also a well-known risk factor [3, 1215]. A history of other cancers, including HNC, and occupational hazards are also associated with SGC incidence [3, 13]. In contrast to HNC risk factors, neither alcohol consumption nor tobacco use increases the risk of salivary gland malignancies [12, 13]. Numerous other causative factors have been proposed; how-ever, studies are limited, and the results are inconclusive. Suspicious lesions, especially those with rapid growth, associated painful swelling, facial nerve palsy, or ulceration, indicate malignancy and should be investigated by imaging methods, preferably multiparametric magnetic resonance imaging.

Preoperative fine-needle aspiration enables the differentiation between benign and malignant tumours as well [2, 3, 16]. Radical surgical excision is the standard management option. Owing to tumour advancement and histopathological features, patients must receive further adjuvant radiotherapy or chemoradiotherapy [5, 16]. Park et al. reported disease recurrence in more than 50% of SGCs, despite radical primary treatment [17]. Distant metastases (DMs) occur in 10–40% of cases, frequently in the lungs (more than 50%), bones (40%), and liver (20%). Metastasis development is related not only to tumour type and stage but also to genetic alterations in tumour cells. These factors are therefore responsible for poor patient outcomes despite radical treatment [1820].

Currently, the value of genetic analysis with next-generation sequencing (NGS) is particularly highlighted in SGCs. This will not only improve the knowledge about the molecular background of the pathologies but also enable the introduction of targeted therapies, especially for recurrent diseases, advanced stages, and drug-resistant cases [16, 2124]. Additionally, it might be a pivotal tool in differential diagnosis, especially in ambiguous cases [25]. A summary of the clinical characteristics of SGCs with respect to incidence, histological subtype, predominant location, and survival is presented in Table 1. The most common genetic rearrangements in SGCs are listed in Table 2. The purpose of this paper was to review genetic variations, including novel findings, in the most known histopathological types of SGCs.

Table 1

Clinicopathological features of salivary gland malignancies

ParametersMucoepi-dermoid carcinomaAdenoid cystic carcinomaAcinic cell carcinomaSalivary duct carcinomaMyoepi-thelial carcinomaEpithelial-myoepithelial carcinomaSecretory carcinomaCarcinoma ex-pleomorphic adenomaClear cell carcinomaIntraductal carcinomaAdeno-carcinomaPoly-morphous adeno-carcinomaMicro-secretory adeno-carcinoma
Histopathological variant/growth patternOncocytic Clear-cell Sclerosing Low-grade Intermediate-grade High-gradeCribriform Tubular SolidSolid papillary-cystic Follicular Microcystic Low-grade Intermediate-grade High-gradeCribriform Solid Cystic PapillarySolid Trabecular ReticularSebaceous Oncocytic Double-clearMicrocystic Tubular SolidMyoepithelial carcinoma Salivary duct carcinoma Adenoid Epithelial-myoepithelial carcinomaSingle cells Nested Solid Sheet-like Cords TrabeculaeIntercalated duct type Apocrine Hybrid Oncocytic Low-grade Intermediate-grade High-gradeVariety of growth patternsLobular Trabecular Microcystic Cribriform PapillaryVariety of growth patterns
Incidence per 1,000,000 (% of all SGC)0.62–1.80 (9–30)0.41–1.72 (6–25)0.41–1.73 (6–17)0.27–0.69 (4–10)0.14–0.83 (2–12)0.34 (< 5)0.14–0.27 (2–4)0.20–1.10 (3–16)< 300 cases were described< 200 cases were described0.14–1.24 (2–18)~1%A few cases
Predominant location Other locationMajor salivary glands (90% in the parotid glands)Submandibular glands or minor salivary glands Lung, breastMajor salivary glands (87% in the parotid glands)Major salivary glandsMajor salivary glandsMajor salivary glandsMajor salivary glandsMajor salivary glandsIntraoral minor salivary glandsMajor salivary glandsMajor salivary glandsMinor salivary glandsMinor salivary glands
5-year survival (%)37.5–10060–9033–9620–5050–6480–96~9525–96No dataNo data43–8175–100No data
References[2, 2628][2, 51, 185, 186][2, 73, 185, 186][2, 185][88, 185, 187][143, 185]  [152, 154, 183, 185][2, 69, 185, 186][168][176, 185][69, 185][2, 105, 185, 188][189]

[i] SGCs – salivary gland carcinomas

Table 2

The most frequent genetic alterations in salivary gland carcinomas

Histopathological typeFusionsOther genetic changesReferences
Mucoepidermoid carcinoma (MEC)CRTC1-MAML2, 56–88%TP53, 21–42% CDKN2A, 42–56% CDKN2B, 31% BAP1, < 21% PIK3CA, 17–21% HRAS, < 14%Saade et al. [31] Kang et al. [34] Seethala et al. [35] Zerdan et al. [47] Wang et al. [48] Morita et al. [49]
Acinic cell carcinoma (AcCC)SCPP gene cluster – NR4A3, > 80%CDKN2A/B high percentage in high-grade tumours and metastases cases ATM, 7–14% PTEN, 10–12%Haller et al. [75] Dogan et al.[78] Ross et al. [69]
Adenoid cystic carcinoma (AdCC)MYB-NFIB, 60–80% MYBL1-NFIB, MYBL1-YTHDF3NOTCH signalling pathway, ~ 40% (NOTCH1, 26%) R/M primary tumours, ~ 13 (NOTCH1, 8.5) KDM6A, ~ 15 BCOR, 13–17 ARID1A, 7–14Wagner et al. [61] Ho et al. [59] Lee et al. [66] Ross et al. [69] Wang et al. [68]
Adenocarcinoma (AC)
   Polymorphous adenocarcinoma (PAC)PRKD1 hotspot mutation, 50–73%Andreasen et al. [108] Weinreb et al. [107]
   Cribriform adenocarcinoma (CA)PRKD1-3 fusions, > 80%Weinreb et al. [115]
   Microsecretory adenocarcinoma (MiAC)MEF2C-SS18, ~ 90%Skálová et al. [39]
   Basal cell adenocarcinoma (BCAC)CYLD mutation, 29%Rito et al. [190]
   Mucinous adenocarcinoma (MAC)AKT1 E17K mutation, 100% TP53 mutation, 88%Rito et al. [190] Rooper et al. [191]
Salivary duct carcinoma (SDC)TP53, 39–60% HRAS, 11–49% ERBB2, 10–100% NF1, 7–20% PIK3CA, 19–47% PTEN, 6–13.5% AR overexpressionDalin et al. [126] Ku et al. [140] Kohsaka et al. [136] Dogan et al. [127] Mueller et al. [123]
Myoepithelial carcinoma (MECA) de novo MECA ex PATGFBR3-PLAG1, 25% FGFR1-PLAG1, 29%Various copy number alternationsDalin et al. [88]
Epithelial-myoepithelial carcinoma (EMC)HRAS, 27–87% PIK3CA, 22–40% AKT1, 6.5–20%Urano et al. [146] Grünewald et al. [148] Chiosea et al. [149] Nakaguro et al. [150]
Secretory carcinoma (SC)ETV6-NTRK3, > 95%Baněčková et al. [192]
Carcinoma ex-pleomorphic adenoma (CA ex PA)PLAG1/HMGA2 rearrangementsTP53, 55–100% ERBB2, 39–57% PIK3CA, 8–42% HRAS, 4–23%Stenman et al. [72] Dalin et al. [88] Chiosea et al. [128] Grünewald et al. [141] Dogan et al. [127] Kohsaka et al. [136]
Clear cell carcinoma (CCC)EWSR1-ATF1, > 90%Antonescu et al. [170]
Intraductal carcinoma (IC)RET rearrangements, ~ 45% NCOA4-RET (mainly in intercalated subtype) MYO18A-ALKHRAS PIK3CA High percentage (only in apocrine subtype)Skálová et al. [179] Weinreb et al. [180] Hsieh et al. [182] Majewska et al. [183]

A comprehensive literature search was performed in the PubMed database. We analysed the full texts of the articles published in English in the period 1984–2024. The exclusion criteria were as follows: languages other than English, only abstracts available, papers concerning HNC holistically without specific analysis of SGCs, and analysis of malignant transformation of benign lesions, e.g. pleomorphic adenoma.

The search was performed with the following keywords: “salivary gland carcinoma”, “genetic alterations’’, “molecular abnormalities”, “NGS”, “targeted therapy”, “precision therapy”, “mucoepidermoid carcinoma”, “acinic cell carcinoma”, “adenoid cystic carcinoma”, “carcinoma ex-pleomorphic adenoma”, “Ca ex PA”, “adenocarcinoma’’, “salivary duct carcinoma”, “myoepithelial carcinoma”, “epithelial-myoepithelial carcinoma”, “secretory carcinoma”, “polymorphous adenocarcinoma”, “cribriform adenocarcinoma”, “microsecretory adenocarcinoma”, “basal cell adenocarcinoma’’, “mucinous adenocarcinoma”, “clear cell carcinoma”, and “intraductal carcinoma”.

The results of the search are presented in relation to the histopathological types of SGCs.

Mucoepidermoid carcinoma

Mucoepidermoid carcinoma is the predominant salivary gland neoplasm and is detected in more than 30% of all salivary malignancies [26]. Generally, it is characte-rised by gradual growth, rare recurrence, and favourable patient outcomes. However, this type of cancer can be highly heterogeneous and can present as low-, intermediate-, or high-grade cancer, with the latter being associated with poor outcomes. Additionally, the mean age at diagnosis is lower than that of other subtypes and ranges from 45 to 49 years [2, 2629].

Chromosomal translocation t(11;19)(q14-21; p12-13) is unique for MEC and results in CREB regulator transcriptional coactivator (CRTC1) (also known as MECT1)-mastermind-like transcriptional coactivator 2 (MAML2) oncogene fusion. It has been detected in more than 80% of patients with this cancer subtype. This alteration leads to cell proliferation and survival through autocrine amphiregulin (AREG)/epidermal growth factor receptor (EGFR) signalling [3035]. Chen et al. revealed that aberrantly activated AREG-EGFR signalling in CRTC1-MAML2-positive MEC cells made them highly sensitive to EGFR inhibition, suggesting benefit from EGFR-targeted therapies, e.g. cetuximab [36]. However, the results of further studies were unsatisfactory, and Ni et al. proposed simultaneous therapy with erlotinib-EGFR inhibitors and Notch inhibitors as more effective [32]. Since MAML2 is involved in NOTCH signalling pathway activation [33, 37, 38], this drug combination becomes more target specific. The other translocation, t(11;19)(q21;q26), results in a CRTC3-MAML2 fusion product that is detected in 6% of cases [30, 39, 40]. Another rare change is the translocation t(6;22)(p21;q12), which promotes ESWR1‒POU5F1 fusion [40]. Previously, the CRTC1-MAML2 fusion product was considered a positive prognostic factor [4143]. However, further research did not reveal significant differences in survival between patients with and without the translocation [31, 44, 45]. In contrast, Anzick et al. revealed that adverse outcomes in patients with translocations might be related to other genetic alterations, such as CDKN2A deletion [46]. How-ever, copy number variations (CNVs) and somatic mutations associated with this alteration have not been frequently analysed in MEC. Zerdan et al. performed NGS analysis of 118 MEC tumours and reported CDKN2A abnormalities in 53% of the cohort. Other frequent changes included those in TP53 (41%), CDKN2B (31%), BAP1 (19%), PIK3CA (17%), TERT (15%), and HRAS (14,5%) [47]. Similar observations regarding the most common variations were reported by Wang et al. [48]. In contrast, the analysis of comparable sample sizes by Morita et al. revealed that HRAS mutations are rarely detected [49]. On the other hand, Kang et al. reported whole-exome sequencing results for 18 MEC tumours, and the second most frequent variation after TP53 was the POU6F2 gene (17%) [34]. In addition, alterations in BRCA2 and ERBB2 are quite common in MEC (17% and 13%, respectively) [30]. Although NF1 alterations are not frequently detected, Kato et al. reported NF1 and TP53 commutation [47, 50]. However, the significance of these findings remains unclear. Further studies are needed to obtain a more in-depth molecular inquiry into MEC molecular pathogenesis, especially in cases with poor outcomes.

Adenoid cystic carcinoma

Adenoid cystic carcinoma frequently arises in the submandibular or minor salivary glands. Its occurrence in the parotid gland is rare. Although AdCC is known as a histopathological type with indolent growth, it tends to recur, with perineural invasion and DM, especially to the lungs [5154]. Cases of relapse and metastasis (R/M) are frequently incurable because of a lack of effective systemic therapies, despite ongoing clinical trials. Therefore, there is an urgent need to verify the possibility of using targeted treatment.

The activating neurogenic locus notch homologue protein 1 (NOTCH1) mutation and v-myb avian myeloblastosis viral oncogene homologue (MYB) overexpression are related to AdCC development, progression, perineural invasion, and even chemoresistance, which predisposes patients to unfavourable outcomes [30, 5558]. In contrast, Ho et al. did not find a correlation between mutational MYBs and either R/M or survival [59]. In approximately 80% of cases, MYB alternations present as the t(6;9)(q22-23;p23-24) translocation, which involves the MYB proto-oncogene and the nuclear factor 1B gene (NFIB) transcription factor, leading to overexpression of the fusion product and worsening the prognosis [30, 60, 61]. MYB NFIB translocation is associated with high MYB expression. This translocation disrupts the MYB 3΄ UTR, a microRNA regulatory site responsible for downregulating MYB. The existence of additional mechanisms for MYB overexpression in AdCC was investigated, revealing alternate rearrangements that translocate super-enhancers in the NFIB and TGFBR3 loci to the MYB locus. The MYB protein binds these super- enhancers, which in turn physically interact with the MYB promoter, drive its overexpression, and establish a positive feedback loop [62].

To emphasise the importance of MYB gene activity, it coordinates the upregulation of pivotal targetable genes involved in several functions related to carcinogenesis, such as apoptosis (API5, BCL2, BIRC3, HSPA8, and SET), cell cycle control (CCNB1, CDC2, and MAD1L1), cell growth and angiogenesis (MYC, KIT, VEGFA, FGF2, and CD53), and cell adhesion (CD34) [63, 64]. Notably, in 35% of MYB-NFIB fusion-negative tumours, MYBL1 alterations were identified [65]. Interestingly, MYB/MYBL1 rearrangements were not very common in R/M AdCCs (22%). In contrast, NOTCH signalling pathway alterations were noted in approximately 40% of R/M cases (with NOTCH1 mutations observed in 26% of these), while only 13% of primary tumours demonstrate increased signalling in the pathway (NOTCH1 mutations in 8.5%) [59, 66].

Notably, Ho et al. also reported frequent alterations in R/M AdCC among genes involved in chromatin remodelling: KDM6A, KMT2C/MLL3, ARID1A, ARID1B, BCOR, MLL2/KMT2D, and CREBBP, with increased frequency compared with primary tumours. TERT promoter mutations were found in > 10% of the R/M patients. Interestingly, NOTCH1 and MYB/MYBL1 fusions are practically undetectable in these lesions [59]. In parallel, Stephens et al., in addition to significant MYB activation, reported SPEN gene alterations (negative NOTCH signalling regulators) in more than 20% of the study cohort [67]. Similar findings regarding NOTCH1, KDM6A, ARID1A, BCOR, CREEB, and TERT have been previously reported. Less frequently detected alterations were in MLL2, RUNX1, PTEN, BAP1, PIK3CA, CDKN2A, ACTB, MGA, CTNNB1, FOXD1, IGFR1, MUC5B, OBSCN, PIK3R1, SPHKAP, TTN FGFR2, and BRAF [68, 69]. In contrast, TP53 mutations are rarely found in AdCCs, including R/M cases. Compared with tumours with favourable outcomes, recurrent and metastatic tumours harbour notably greater loads of mutations. Thus, the options of targeted therapies are quite extensive for verifying their efficiency in advanced stages [56, 70, 71].

Acinic cell carcinoma

The characteristics of AcCC are generally similar to those of MEC. However, some cases of aggressive metastatic AcCC have been reported recently [7274]. Current knowledge regarding the molecular alterations in AcCC has not yet been properly established.

Haller et al. detected rearrangement t(4;9)(q13;q31), which results in secretory Ca-binding phosphoprotein (SCPP) gene cluster (STATH, HTN1, HTN3, ODAM, FDCSP, and MUC7) and nuclear receptor subfamily 4 group A member 3 (NR4A3) fusion in most tumours of the ana-lysed cohort (more than 80%). The former translocation is unique to AcCC and allows for differentiation of AcCC from mammary analogue secretory carcinoma (MASC), parti-cularly in cases with high-grade transformation. Moreover, the resulting fusion gene acts as an oncogenic driver, with the NR4A3 transcription factor being upregulated due to the translocation of active enhancers from the SCPP gene cluster (which is highly expressed in salivary glands) to the region upstream of NR4A3 [75, 76]. The second most common fusion involves the histatin 3 and Myb/SANT-like DNA-binding domain containing 3 genes (HTN3-MSANTD3) (t(4;9)(q13.3;q31.1)), which have been described in a few cases (4–8%) [7577]. According to the authors, the former translocation is exceptional for AcCC and provides an effective differential diagnosis of MASC, especially in cases with high-grade transformation. Moreover, NR4A3 might be considered an oncogenic driver through enhancer hijacking, whereby NR4A3 is upregulated [75, 77]. In a recent study, Ross et al. reported CDKN2A and CDKN2B alterations in 76% and 45% of patients with relapses or metastases, respectively [69]. Simultaneously, Dogan et al. performed a genetic analysis and reported that the CDKN2A/B gene changed solely in high-grade tumours (58% of this group), whereas in the disease course with distant metastasis, these rearrangements were found in nearly 90% of the patients [78], confirming them as a negative prognostic factor. Notably, for tumours with identified negative markers, there are targetable treatment options based on CDK4/6 inhibitors, immunotherapy, or DNA methyltransferase inhibitors [79, 80]. Moreover, in advanced AcCC, other genetic changes have also been observed [78]. The most common rearrangements were related to ATM (7–14%), PTEN (10–12%), FBXW7, and TP53 rearrangements, whereas alterations in BRAF, NF1, HRAS, NOTCH1, TERT, ARID2, BIRC3, MTAP, and FAT1 were less common [69, 78]. Importantly, some of these alterations may provide opportunities for utilising precision therapy.

Carcinoma ex-pleomorphic adenoma

Carcinoma ex PA is a rare primary SGC arising from a preexisting PA. It is estimated that 5–15% of benign pleomorphic adenomas undergo malignant transformation to carcinoma (Ca ex PA) [81, 82]. Thus, the detection of the benign part of the tumour might lead to a final misdiagnosis, but rapid growth and other symptoms should indicate suspicion of malignancy [83]. Although salivary duct carcinoma, myoepithelial carcinoma (MECA), and adenocarcinoma not otherwise specified (NOS) are considered the most commonly detected malignant components of Ca ex PA, other types of SGC histopathology have also been described [8489]. The pleomorphic adenoma gene 1 (PLAG1) and the high-mobility group AT-hook 2 (HMGA2) genes are most frequently altered in both PAs and Ca ex PAs [90], but not typical for primary salivary duct carcinoma (SDC), MECA, or AC. Katabi et al. presumed that rearrangements in these genes were specific to both PA or Ca ex PA and could distinguish Ca ex PA from its de novo counterparts [91]. Nonetheless, further investigations have shown their occurrence in de novo lesions [88]. Carcinoma ex PA tumours have abundant copy number alterations (CNAs) that are suspected to be involved in the malignant transformation from benign lesions. The most common loss of heterozygosity is the amplification of 12q genes (HMGA2, MDM2), deletions of 5q, gains of 8q12.1 (PLAG1) and 8q22.1-q24.1 (MYC), and amplification of 17 chromosomes (ERBB2) [88, 9294]. Table 3 lists the most commonly detected genetic alterations, including fusions and histopathological subtypes of Ca ex PA, reported in the literature.

Table 3

Malignant component of carcinoma ex pleomorphic adenoma as reported in respective studies

GeneIdentified malignant component in Ca ex PAReferences
Genes fusions
CTNNB1-PLAG1MECA, SDC ~ 30%Asahina et al. [193], Skálová et al. [194], Dalin et al. [126]
FBXO32-PLAG1NDBubola et al. [195]
FGFR1-PLAG1MECA, SDC, NDDalin et al. [88], Chiosea et al. [128], Skálová et al. [194], Bubola et al. [195]
LIFR-PLAG1MECA, SDCSkálová et al. [194], Dalin et al. [126]
MEG3-PLAG1NDBubola et al. [195]
ND4-PLAG1MECADalin et al. [88]
PLAG1-NFIBNDBubola et al. [195]
TGFBR3-PLAG1MECADalin et al. [88], Rupp et al. [196]
HMGA2-CNOT2NDBubola et al. [195]
HMGA2-NFIBNDBubola et al. [195]
HMGA2 fusionsMECADalin et al. [88]
Oher PLAG1 fusionsMECADalin et al. [88]
HMGA2-WIF1ND, Adenoid cystic carcinoma with sarcomatoid transformation, MECAPersson et al. [92] Katabi et al. [197]
ETV6-RETSCSmith et al. [198]
ZCCHC7-NTRK2ND (recurrence and metastatic case)Pircher et al. [199]
Somatic gene mutations
TP53SDC, MECAChiosea et al. [128], Grünewald et al. [141], Dogan et al. [127], Rupp et al. [196], Dalin et al. [126], Kohsaka et al. [136], Mueller et al. [123]
PIK3CASDC, MECA, EMCChiosea et al. [128], Dogan et al. [127], Dalin et al. [88], Hallani et al. [144], Dalin et al. [126], Kohsaka et al. [136], Mueller et al. [123]
HRASSDC, MECA, EMCChiosea et al. [128], Dogan et al. [127], Dalin et al. [88], Hallani et al. [144], Dalin et al. [126]
ERBB2SDC (gain/amp)Chiosea et al. [128], Dogan et al. [127], Dalin et al. [126], Kohsaka et al. [136], Mueller et al. [123]
AKT1SDCDalin et al. [126]
ALKSDCMueller et al. [123]
APCSDCDogan et al. [127], Mueller et al. [123]
ARSDCDogan et al. [127]
ARID1ASDCKohsaka et al. [136]
ASXL1SDCDogan et al. [127]
ATMSDC, MECAChiosea et al. [128], Dalin et al. [88], Mueller et al. [123]
ATRMECADalin et al. [88]
AURKASDCDogan et al. [127]
BAP1SDCDogan et al. [127]
BRAFSDCChiosea et al. [128], Kohsaka et al. [136]
BRCA1MECADalin et al [88]
BRCA2SDCDogan et al. [127], Kohsaka et al. [136]
BTKSDCDogan et al. [127]
CCNE1SDCDogan et al. [127], Mueller et al. [123]
CCND3SDCMueller et al. [123]
CDH1SDCDogan et al. [127]
CDK4SDCGrünewald et al. [141], Mueller et al. [123]
CDK6SDCMueller et al. [123]
CDK12SDCDogan et al. [127]
CDKN1BSDCDogan et al. [127]
CDKN2ASDCChiosea et al. [128], Mueller et al. [123]
CHEK2SDCMueller et al. [123]
CREBBPMECA, SDCDalin et al. [88], Mueller et al. [123]
CTCFSDCDogan et al. [127]
DNMT1, DNMT3A, NMT3BSDCDogan et al. [127]
DOCK7SDCDalin et al. [126]
EGFRSDCDogan et al. [127]
EP300SDCMueller et al. [123]
ERBB3SDCDogan et al. [127]
EWSR1MECA (clear cell)Skálová et al. [194]
FANCA, FANCCSDCDogan et al. [127]
FASNSDCDalin et al. [126]
FAT1SDC, MECADogan et al. [127], Dalin et al. [88]
FAT4MECADalin et al [88]
FBXW7SDCDogan et al. [127], Mueller et al. [123]
FGFR1MECA, SDCDalin et al. [88], Dalin et al. [126], Mueller et al. [123]
FGFR2MECADalin et al. [88]
FGFR3SDCChiosea et al. [128]
FGFR4SDCMueller et al. [123]
FHSDCDogan et al. [127]
FLCNSDCDogan et al. [127]
FOXA1SDCDalin et al. [126], Kohsaka et al. [136]
GATA2SDCDogan et al. [127]
HMGA2NDPersson et al. [92]
HNF1ASDCDogan et al. [127]
JUNSDCDogan et al. [127]
KDRSDCDalin et al. [126]
KITSDCMueller et al. [123]
KMT2ASDCDogan et al. [127], Kohsaka et al. [136]
KMT2BSDCDalin et al. [126]
KMT2CSDCDogan et al. [127], Dalin et al. [126], Kohsaka et al. [136]
KMT2DSDCKohsaka et al. [136]
LIFRMECADalin et al. [88]
MAP2K2SDCKohsaka et al. [136]
MAP3K1SDCDogan et al. [127]
MDM2ND, SDCPersson et al. [92], Mueller et al. [123]
METMECADalin et al. [88]
MLH3SDCDalin et al. [126]
MML2MECADalin et al. [88]
MN1MECADalin et al. [88]
MSH5SDCDalin et al. [126]
MTORSDCDalin et al. [126]
MYCSDCDogan et al. [127]
NCOA1, NCOA2MECADalin et al. [88]
NCOR1SDCDogan et al. [127], Dalin et al. [126]
NF1SDCDogan et al. [127], Dalin et al. [126], Kohsaka et al. [136], Mueller et al. [123]
NOTCH1MECA, SDCDalin et al. [88], Mueller et al. [123]
NOTCH2-3SDCMueller et al. [123]
NSD1SDCDalin et al. [126]
PIK3R1SDCDogan et al. [127]
PTENSDCChiosea et al. [128], Dogan et al. [127], Kohsaka et al. [136]
PTPN11SDCDogan et al. [127]
PTPRSSDCDogan et al. [127]
RAD51CSDCDogan et al. [127]
RETSDCDalin et al. [126]
RICTORSDCMueller et al. [123]
ROS1SDCMueller et al. [123]
RTEL1SDCDogan et al. [127]
SF3B1SDCDalin et al. [126]
SMAD4SDCDalin et al. [126]
SMARCA4MECA, SDCDalin et al. [88], Dalin et al. [126]
TSC2SDCMueller et al. [123]
ZFHX3SDCKohsaka et al. [136]

[i] EMC – epithelial-myoepithelial carcinoma, MECA – mucoepidermoid carcinoma, ND – no data available, SDC – salivary duct carcinoma

Myoepithelial carcinoma

The incidence of MECA is estimated to be very low, at 2% among all SGCs. Nonetheless, because of the difficulty of proper diagnosis, the actual number of cases is predicted to be greater [10, 95]. The tumour might occur as a de novo lesion or arise from the malignant transformation of a PA or myoepithelioma [96]. These data suggest that MECA ex PAs are more frequently detected than de novo lesions [88, 97]. However, the conclusion regarding which component is characterised by more aggressive behaviour or poorer patient outcomes remains debatable [95, 97100]. In most cases, this subtype of cancer is associated with adverse patient results, including early local and DM [10, 88, 95]. Myoepithelial carcinoma is one of the most commonly confirmed components of Ca ex PAs [89, 101].

Salivary gland MECA rarely occurs; therefore, few genetic studies of this type are available. Dalin et al. analysed 40 tumours with divisions on either the MECA de novo or the MECA ex PA, as well as cases with and without recurrence. In MECA ex PA, more genetic alterations, including fusions, somatic mutations, and CNVs, were found. According to the authors, CNVs are responsible for the malignant transformation of the PA into the MECA ex PA and are also associated with a worse prognosis. FGFR1-PLAG1 fusion was the most commonly (18%) identified in the MECA ex PA, followed by TGFBR3-PLAG1 but with no evidence of their prognostic value. Furthermore, EWSR1-ATF1 was described only in the MECA de novo, with or without recurrence [88]. In contrast to the research conducted by Skálová et al., EWSR1 rearrangements were found frequently in the clear cell component of MECA both in de novo cases and those arising from the PA, but the fusion partner genes were not identified [102]. In the aforementioned study, PIK3CA was present only in patients without relapse, whereas FGFR2 mutations were found in patients with recurrence [88]. The findings are summarised in Table 4. FGFR2 mutations were also described in 2 patients after radical PA excision, in which the MECA rapidly developed. In both PAs and MECAs (without the PA component), FGFR2 point mutations were confirmed, which might be indicative of an aggressive disease course [103]. Recently, Gandhi et al. reported a novel CTCF-NCOA2 fusion in a single MECA patient [104]. Furthermore, Cormier et al. presented a novel TERT promoter mutation in metastatic MECA ex PA (the tumour was previously misdiagnosed as PA) [9].

Table 4

Genetic rearrangements in the mucoepidermoid carcinoma de novo and the mucoepidermoid carcinoma ex pleomorphic adenoma presented in the study by Dalin et al. in relation to oncological outcomes

MECA de novoMECA ex PA
No recurrenceRecurrenceNo recurrenceRecurrence
TGFBR3-PLAG1HMGA2 fusionsTGFBR3-PLAG1FGFR1-PLAG1
Other PLAG1 fusionsEWSR1-ATF1FGFR1-PLAG1Other PLAG1 fusions
EWSR1-ATF1FGFR1Other PLAG1 fusionsHMGA2 fusions
MSN-ALKFGFR2HMGA2 fusionsFGFR2
PIK3CASMARCA4HRASMAML2
MAML2PCM1PIK3CANOTCH1
NOTCH1TRIP11FGFR1ATM
ATMLIFRATR
KMT2CMETBRCA1
SETD2MAML2MN1
ATRCOL2A1
CREBBPFAT1
NCOA1FAT4
NCOA2

[i] MECA – mucoepidermoid carcinoma, PA – pleomorphic adenoma

Adenocarcinoma

Polymorphous adenocarcinoma

Polymorphous adenocarcinoma (PAC) is a rare, slow- growing malignant tumour. It mainly arises from the minor salivary glands (second most common histopathological type), particularly those localised on the hard palate. There is a higher prevalence in women than in men, and patient outcomes are defined as one of the most favourable outcomes among SGCs [105, 106].

Weinreb et al. revealed a PRKD1 p.E710 hotspot mutation in nearly 73% of tumours, and these observations were not identified in other SGCs. Thus, this alteration is unique to PAC and may be useful for differentiating it from its mimics [107, 108]. Notably, in cribriform adenocarcinoma (CA), PRKD1-3 fusions are the most common. CA is classified as an aggressive variant of PAC with a high predisposition to metastasis [109112]. Among the fusion partners ARID1A, ATL2, DDX3X, PPP2R2A, PRKAR2A, SNX9, and STRN3 (cases with high-grade transformation) should be mentioned [113116]. However, the type of genomic alteration is not specific for any AC subtype, and occasionally, either PRKD1-3 fusions or PRKD1 rearrangements are found in PAC and CA, respectively [109]. Therefore, differentiation between these 2 variants with various behaviours might be challenging.

Adenocarcinoma not otherwise specified

Tumours with a histopathological diagnosis of adenocarcinoma NOS constitute a heterogeneous group that has not yet been well characterised. For example, NTRK2-ZCCHC7 and SS18-ZBTB7A fusions have been described [116, 117]. In R/M cases, TP53 (55%), PIK3CA, HRAS, CDKN2A, ERBB2, PTEN, NF1, and ARID1A alterations were observed with considerable frequency [69].

On the basis of genetic pattern analysis, microsecretory adenocarcinoma has been distinguished from NOS. Microsecretory adenocarcinoma harbours MEF2C-SS18 fusion in approximately 90% of cases [39, 118].

The most common alterations in basal cell adenocarcinoma and mucinous adenocarcinoma are shown in Table 1.

Salivary duct carcinoma

Salivary duct carcinoma is one of the most aggressive SGCs, with either early relapse or frequent DM. It is also associated with significant mortality. Predilection in elderly males with a smoking history is usually combined with advanced-stage presentation and parotid gland localisation [119123]. The estimated morbidity is 5.5–12% [124, 125]. Moreover, SDCs ex PAs have also been detected [122, 126128]. Table 2 provides genetic information for this subtype.

In addition to the microscopic structure resembling high-grade ductal carcinoma of the breast, SDC is also characterised by the overexpression of human epidermal growth factor receptor 2 (HER2). Instead of oestrogen and progesterone receptor positivity, androgen receptor (AR) expression is detected in 75–98% of cases [122, 126, 129, 130]. Notably, AR is seldom detectable in other SGCs [131]. However, studies are inconclusive regarding the prognostic value of the AR [129, 131, 132]. Nevertheless, Kawakita et al. showed in a retrospective study that the utilisation of HER2-targeted therapy and androgen deprivation therapy significantly improved patients results compared with conventional therapy management [133]. The anti-HER2 therapies that induce improvement in clinical responses in SDC patients use trastuzumab in combination with chemotherapy (i.e. taxanes, capecitabine, carboplatin, eribulin) or with another anti-HER2 targeted agent (i.e. pertuzumab). Further expectations and therapeutic advances are related to novel anti-HER2 drugs such as antibody-drug conjugates (i.e. trastuzumab emtansine, trastuzumab deruxtecan) introduced in this setting [134].

In recent years, genetic knowledge about SDC has increased profoundly, but it still has not been comprehensively investigated. The tumour mutation burden is extremely high in most SDC cases, in contrast to other SGCs. Vos et al. evaluated therapy with nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4) in patients with metastatic SGC. Although the efficacy was limited in AdCC, with infrequent responses, they found it promising for non-AdCC SGCs, particularly salivary duct carcinomas [135]. Genetic fusions are not recurrent events in this subtype, whereas somatic mutations as well as CNVs are considerably more common [123, 126, 136]. Moreover, most of them provide opportunities for the utilisation of targeted treatment for this unpredictable cancer [30, 127, 137139]. TP53, HRAS, PIK3CA, and ERBB2 (HER) rearrangements are the most common, and some of them are related to poor outcomes [123, 126128, 136, 140, 141]. Interestingly, although HRAS mutations constitute the majority of de novo lesions, they are rare in SDC ex PAs [123, 126, 127, 136]. Data regarding the molecular landscape of SDCs are presented in Table 5.

Table 5

The genetic pathways most commonly affected in salivary duct carcinoma

PathwayGenesReferences
DNA damageTP53 (39–60%), ATM, BRCA2, CHEK2, MDM2, MDM4, MLH3, MLH5[123, 126, 127, 136, 140, 141]
MAPKHRAS (11–49%), NF1 (7–20%), BRAF, KRAS, NRAS[123, 125, 126, 127, 128, 136, 137, 140]
RTKERBB2 (10–100%), ALK, EGFR, ERBB3-4, FGFR1-2, FGFR4, FLT3, JAK2, KDR, KIT, MET, NTRK2, PDGFRA, RET[123, 126, 127, 136, 137, 140]
PI3K/AKT/mTORPIK3CA (19–47%), PTEN (6–13.5%), AKT1-3, PIK3R1, RICTOR, RPTOR, TSC2[123, 125, 126, 127, 128, 136, 137, 140]
Androgen signallingAR, FASN, FOXA1[126, 136]
Histone modificationKDM6A, KMT2A, KMT2C, KMT2D, KMT2E, NSD1[126, 127, 136, 140]
Cell cycleCDK4, CDK6, CDK12, CDKN1A, CDKN1B, CDKN2A, CCNE1, CCND1-3, RB1[123, 126, 127, 136, 140, 141]
NOTCHCREBBP, EP300, FBXW7, NOTCH1-3[123, 140]
SWI/SNF complexARID1A, SMARCA4, SMARCB1[123, 126, 127, 136]
WNT-β-cateninAPC, CDH1, CTNNB1, FAT1[123, 126, 140]
OtherABL1, AURKA, BCOR, CCND1, CCNE1, FLCN, GNAS, HMGA2, IDH1-2, IGFR1, IKBKE, KLF5, AMP, MAP2K1, MAP2K4, MITF, MPL, MYC, PRDM1, SMAD4, SMO, STK11, TNIK, VHL, ZFHX3[123, 126, 127, 136, 140, 141]
FusionsETV6-NTRK3, ABL1-PPP2R2C, BCL6-TRADD, HNRNPH3-ALK, EML4-ALK, RAPGEF6-ACSL6[126, 127, 195]

Epithelial-myoepithelial carcinoma

Epithelial-myoepithelial carcinoma (EMC) is rarely detected, and it was first reported by Donath et al. in 1972. Previously, it appeared under other terminology of adenomyoepithelioma or clear cell adenoma. The tumour consists of a dual cell population that forms a double layer: inner ductal cells and outer myoepithelial cells [142144]. Notably, various histological subtypes of EMCs exist, including sebaceous, oncocytic, and double-clear subtypes. Thus, the differential diagnosis could pose difficulties [145, 146]. Morbidity predominates in females more than males. Most commonly, the parotid gland is affected, and the tumour is characterised by a high overall survival rate. Although DM rarely occur, relapses are common [143, 147].

HRAS (27–87%) was described as the most frequently mutated gene in EMC [146, 148150]. In the studies conducted by Urano et al. and Nakaguro et al., these findings were not detected in EMCs ex PAs [146, 150]. In parallel, Hallani et al. did not prove HRAS alterations for de novo EMC [144]. PIK3CA and AKT1 have been reported quite commonly in EMC (22–40% and 6.5–20%, respectively) [146, 148]. CTNNB1, FBXW7, and TP53 rearrangements and SMARCB1 deletions have been reported in single cases (the last 3 in high-grade tumours) [144, 148]. Mäkelä et al. described rare metastatic EMC in a 36-year-old woman, where in addition to HRAS mutation, ARID1B, ATR, CDK12, ERBB4, MAPK1, NANOG, NOTCH2, PIK3R1, and RPTOR alterations were detected [151].

Secretory carcinoma

Secretory carcinoma (SC) (previously known as mammary analogue secretory carcinoma) is a novel salivary gland tumour that was described by Skálová et al. in 2010 [152]. Most of these tumours were previously classified as AcCC [153]. The age at diagnosis is relatively low (mean 45 years), including paediatric patients. There is a greater predilection in men, and the disease course is indolent, with favourable patient outcomes [154, 155].

Secretory carcinoma has a significant histological and molecular resemblance to breast secretory carcinoma. It is characterised by harbouring the same translocation t(12;15)(p13;q25), resulting in the ETV6-NTRK3 fusion gene encoding a chimeric oncoprotein-tyrosine kinase (unlike AcCC) [152, 155, 156]. Other ETV6 fusion partners have also been discovered, including ETV6-MAML3 [157], ETV6-MET [158], and ETV6-RET [157, 159]. Notably, some of these genes remain unknown (ETV6-X) [160]. Recently, other novel fusions, such as VIM-RET [161], CTNNA1-ALK [162], and dual fusion, ETV6-RET and EGFR-SEPT14, were identified in an 18-year-old male [159]. ETV6-NTRK3 and MYB- SMR3B fusions were found in recurrent high-grade submandibular tumours [161]. Only a few studies have analysed genetic rearrangements other than fusions. Na et al. identified pathogenic PRSS1 mutations, mainly in patients with an aggressive disease course and recurrence, whereas other findings were classified as likely pathogenic or of uncertain significance [163]. In contrast, Skálová et al. analysed 3 tumours with high-grade transformation and did not detect the most commonly occurring genetic alterations associated with poor outcomes (TP53, CTNNB1, EGFR, CCND1) [164].

Testing for ETV6-NTRK3 gene rearrangements is critical for SC patients care since entrectinib, an inhibitor of tropomyosin receptor kinase (TRKs), has been reported to be effective and safe in treating solid tumours with NTRK fusion genes. In an integrated analysis of phase 1–2 trials (STARTRK-1, STARTRK-2, and ALKA-372-001) of solid tumours with the NTRK fusion gene, the response rate to the TRK inhibitor entrectinib was 57%, and the median progression-free survival was 11.2 months [165]. Another TRK inhibitor, larotrectinib, is also effective in the treatment of solid tumours with the NTRK fusion gene [166]. Other potential therapies for SC patients with identified oncogenic RET fusions, namely ETV6-RET , are selpercatinib and pralsetinib selective RET inhibitors, currently under preclinical and clinical testing [167].

Clear cell carcinoma

Clear cell carcinoma (CCC) (previously known as hyali-nising clear cell carcinoma) is an indolent low-grade tumour that typically arises from the intraoral minor salivary glands. There is a higher prevalence in females, whereas relapses and metastases are rare [168].

Considering the occurrence of clear cells in other SGCs, differential diagnosis may be a challenge [169]. Antonescu et al. first described genetic rearrangement in the CCC-EWSR1-ATF1 fusion t(12;22)(q13;q12). It occurs in more than 90% of cases, and, being unique for CCC, it is therefore a helpful differentiation tool [170]. EWSR1-CREB1, EWSR1-CREM, and SMARCA2-CREM fusions have been reported in single cases thus far [171173].

Intraductal carcinoma

Intraductal carcinoma (IC) is a rare salivary gland tumour that affects mainly the parotid gland, with features similar to mammary atypical ductal hyperplasia or ductal carcinoma in situ of the breast [174, 175]. Recent studies have classified 4 distinctive subtypes: intercalated duct type, apocrine, hybrid, and oncocytic [176].

RET rearrangements, including recurrent NCOA4-RET (intercalated, oncocytic, seldom hybrid), TRIM27-RET (hybrid, apocrine), and TRIM33-RET (oncocytic) rearrangements, have been detected [177179]. In contrast, RET gene altera-tions have not yet been confirmed in the apocrine subtype [180].

The relationship between IC and SDC remains controversial, even though they are considered diverse entities. Intraductal carcinoma, especially invasive apocrine IC, is a precursor for more aggressive cancers, such as SDC [174, 176, 180]. Nevertheless, this issue requires further investigation. Molecular evidence of resemblance to SDC revealed a high occurrence of HRAS and PIK3CA hotspot mutations in apocrine IC [174, 180182]. Additionally, ATM, SPEN, and TP53 mutations and either DFFA-ARID1A or KIF13B-EPB41L4B fusions were found in this subtype [174,180]. In parallel, BRAF V600E mutations in the oncocytic subtype and novel fusions of TUT1-ETV5 and KIAA1217-RET in intercalated duct variants and hybrid intercalated duct tumours with invasive growth have also been identified [178, 179].

Furthermore, Majewska et al. reported an MYO18A-ALK fusion in intercalated duct-type IC in elderly patients after radical excision and no disease relapse during follow-up [183].

Recently, Watanabe et al. presented a case of a 59-year-old male with high-grade intercalated-type IC and DM. Despite radical excision and postoperative radiotherapy, the patient developed multiple DM. Genetic analysis revealed a CTNNA1-ALK fusion and TP53 mutation. Despite further ALK-TKI therapy, the patient’s condition declined, and NGS analysis of the blood samples revealed a novel PIK3CA mutation (ALK fusion was not detected). The importance of this shift remains uncertain. Nevertheless, treatment failure might be related to novel alterations and the predominance of other abnormalities in recurrent tumour tissue [184].

Conclusions

Salivary gland carcinomas are rare entities with unpredictable disease courses. The diversity of both the histological architecture and molecular alterations is distinct among individual subtypes, which leads to diagnostic difficulties. Moreover, because of the rare incidence of SGCs, multicentre clinical trials are urgently needed to provide targeted therapeutic options. Currently, the value of gene-tic analysis has been highlighted, particularly in terms of the possibilities of precision therapies and in light of the insufficient effectiveness of standard treatment options. Knowledge of the molecular landscape of SGC, especially related to outcome predictors, will provide novel and precise methods for diagnosis and therapy in the future.

Disclosures

Institutional review board statement

Not applicable.

Assistance with the article

None.

Financial support and sponsorship

None.

Conflicts of interest

None.

References

1 

Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021; 71: 209-249.

2 

Young A, Okuyemi OT. Malignant Salivary Gland Tumors. StatPearls. StatPearls Publishing Copyright © 2022, StatPearls Publishing LLC.: Treasure Island (FL), 2022.

3 

Gatta G, Guzzo M, Locati LD, McGurk M, Prott FJ. Major and minor salivary gland tumours. Crit Rev Oncol Hematol 2020; 152: 102959.

4 

Global Cancer Observatory. International Agency for Research on Cancer. Cancer Today.

5 

Van Herpen C, Poorten VV, Skalova A, et al. Salivary gland cancer: ESMO-European Reference Network on Rare Adult Solid Cancers (EURACAN) Clinical Practice Guideline for diagnosis, treatment and follow-up. ESMO Open 2022; 7: 100602.

6 

Global Cancer Observatory. International Agency for Research on Cancer. Cancer Tomorrow. Available from: https://gco.iarc.fr/tomorrow/en.

7 

Son E, Panwar A, Mosher CH, Lydiatt D. Cancers of the major sali-vary gland. J Oncol Pract 2018; 14: 99-108.

8 

Speight PM, Barrett AW. Salivary gland tumours: diagnostic challenges and an update on the latest WHO classification. Diagn Histopathol 2020; 26: 147-158.

9 

Cormier C, Agarwal S. Myoepithelial carcinoma ex-pleomorphic adenoma: a rare pathology misdiagnosed as pleomorphic ade-noma; with a novel TERT promoter mutation and high PD-L1 expression. Head Neck Pathol 2022; 16: 322-330.

10 

Xu B, Mneimneh W, Torrence DE, et al. Misinterpreted myoepithelial carcinoma of salivary gland: a challenging and potentially significant pitfall. Am J Surg Pathol 2019; 43: 601-609.

11 

Hernandez-Prera JC, Skálová A, Franchi A, et al. Pleomorphic ade-noma: the great mimicker of malignancy. Histopathology 2021; 79: 279-290.

12 

Spitz MR, Tilley BC, Batsakis JG, Gibeau JM, Newell GR. Risk factors for major salivary gland carcinoma. A case-comparison study. Cancer 1984; 54: 1854-1859.

13 

Radoï L, Barul C, Menvielle G, et al. Risk factors for salivary gland cancers in France: Results from a case-control study, the ICARE study. Oral Oncol 2018; 80: 56-63.

14 

Spitz MR, Fueger JJ, Goepfert H, Newell GR. Salivary gland cancer. A case-control investigation of risk factors. Arch Otolaryngol Head Neck Surg 1990; 116: 1163-1166.

15 

Salivary Gland Cancer: Risk Factors. CancerNet Editorial Board 05/2020.

16 

Geiger JL, Ismaila N, Beadle B, et al. Management of salivary gland malignancy: ASCO Guideline. J Clin Oncol 2021; 39: 1909-1941.

17 

Park GC, Roh JL, Cho KJ, et al. Incidence and risk factors of late recurrence in patients with salivary gland cancer. Clin Otolaryngol 2017; 42: 416-424.

18 

Glazer TA, Shuman AG. Distant metastases and palliative care. Adv Otorhinolaryngol 2016; 78: 182-188.

19 

Nam SJ, Roh JL, Cho KJ, Choi SH, Nam SY, Kim SY. Risk factors and survival associated with distant metastasis in patients with carcinoma of the salivary gland. Ann Surg Oncol 2016; 23: 4376-4383.

20 

Mimica X, McGill M, Hay A, et al. Distant metastasis of salivary gland cancer: Incidence, management, and outcomes. Cancer 2020; 126: 2153-2162.

21 

Morganti S, Tarantino P, Ferraro E, D’Amico P, Duso BA, Curigliano G. Next Generation Sequencing (NGS): A Revolutionary Technology in Pharmacogenomics and Personalized Medicine in Cancer. Adv Exp Med Biol 2019; 1168: 9-30.

22 

Mosele F, Remon J, Mateo et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2020; 31: 1491-1505.

23 

Hussen BM, Abdullah ST, Salihi A, et al. The emerging roles of NGS in clinical oncology and personalized medicine. Pathol Res Pract 2022; 230: 153760.

24 

Kamps R, Brandão RD, Bosch BJ, et al. Next-generation sequencing in oncology: genetic diagnosis, risk prediction and cancer classification. Int J Mol Sci 2017; 18.

25 

Skálová A, Stenman G, Simpson RHW, et al. The role of molecular testing in the differential diagnosis of salivary gland carcinomas. Am J Surg Pathol 2018; 42: e11-e27.

26 

Sama S, Komiya T, Guddati AK. Advances in the treatment of mucoepidermoid carcinoma. World J Oncol 2022; 13: 1-7.

27 

Peraza A, Gómez R, Beltran J, Amarista FJ. Mucoepidermoid carcinoma. An update and review of the literature. J Stomatol Oral Maxillofac Surg 2020; 121: 713-720.

28 

Coca-Pelaz A, Rodrigo JP, Triantafyllou A, et al. Salivary mucoepidermoid carcinoma revisited. Eur Arch Otorhinolaryngol 2015; 272: 799-819.

29 

Spiro RH. Salivary neoplasms: overview of a 35-year experience with 2,807 patients. Head Neck Surg 1986; 8: 177-184.

30 

Vathiotis IA, Johnson JM, Argiris A. New systemic therapies in salivary gland cancer. In: Vermorken JB, Budach V, Leemans CR, Machiels J-P, Nicolai P, O’Sullivan B (eds). Critical issues in head and neck oncology. Springer International Publishing, Cham 2023, 327-345.

31 

Saade RE, Bell D, Garcia J, Roberts D, Weber R. Role of CRTC1/MAML2 translocation in the prognosis and clinical outcomes of mucoepidermoid carcinoma. JAMA Otolaryngol Head Neck Surg 2016; 142: 234-240.

32 

Ni W, Chen Z, Zhou X, et al. Targeting Notch and EGFR signaling in human mucoepidermoid carcinoma. Signal Transduct Target Ther 2021; 6: 27.

33 

Coxon A, Rozenblum E, Park YS, et al. Mect1-Maml2 fusion oncogene linked to the aberrant activation of cyclic AMP/CREB regulated genes. Cancer Res 2005; 65: 7137-7144.

34 

Kang H, Tan M, Bishop JA, et al. Whole-exome sequencing of sali-vary gland mucoepidermoid carcinoma. Clin Cancer Res 2017; 23: 283-288.

35 

Seethala RR, Dacic S, Cieply K, Kelly LM, Nikiforova MN. A reappraisal of the MECT1/MAML2 translocation in salivary mucoepidermoid carcinomas. Am J Surg Pathol 2010; 34: 1106-1121.

36 

Chen Z, Chen J, Gu Y, et al. Aberrantly activated AREG-EGFR signaling is required for the growth and survival of CRTC1-MAML2 fusion-positive mucoepidermoid carcinoma cells. Oncogene 2014; 33: 3869-3877.

37 

Tonon G, Modi S, Wu L, et al. t(11;19)(q21;p13) translocation in mucoepidermoid carcinoma creates a novel fusion product that disrupts a Notch signaling pathway. Nat Genet 2003; 33: 208-213.

38 

O’Neill ID. t(11;19) translocation and CRTC1-MAML2 fusion oncogene in mucoepidermoid carcinoma. Oral Oncol 2009; 45: 2-9.

39 

Skálová A, Hyrcza MD, 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.

40 

Kaur K, Mehta S, Vanik S, et al. The evolving role of molecular pathology in the diagnosis of salivary gland tumours with potential pitfalls. Eur Arch Otorhinolaryngol 2022; 279: 3769-3783.

41 

Okabe M, Miyabe S, Nagatsuka H, et al. MECT1-MAML2 fusion transcript defines a favorable subset of mucoepidermoid carcinoma. Clin Cancer Res 2006; 12: 3902-3907.

42 

Miyabe S, Okabe M, Nagatsuka H, et al. Prognostic significance of p27Kip1, Ki-67, and CRTC1 MAML2 fusion transcript in mucoepidermoid carcinoma: a molecular and clinicopathologic study of 101 cases. J Oral Maxillofac Surg 2009; 67: 1432-1441.

43 

Behboudi A, Enlund F, Winnes M, et al. Molecular classification of mucoepidermoid carcinomas prognostic significance of the MECT1-MAML2 fusion oncogene. Genes Chromosomes Cancer 2006; 45: 470-481.

44 

Birkeland AC, Foltin SK, Michmerhuizen NL, et al. Correlation of Crtc1/3-Maml2 fusion status, grade and survival in mucoepidermoid carcinoma. Oral Oncol 2017; 68: 5-8.

45 

van Weert S, Lissenberg-Witte BI, Bloemena E, Leemans CR. Mucoepidermoid carcinoma of the head and neck: CRTC1/3 MAML 2 translocation and its prognosticators. Eur Arch Otorhinolaryngol 2022; 279: 2573-2581.

46 

Anzick SL, Chen WD, Park Y, et al. Unfavorable prognosis of CRTC1-MAML2 positive mucoepidermoid tumors with CDKN2A deletions. Genes Chromosomes Cancer 2010; 49: 59-69.

47 

Bou Zerdan M, Kumar PA, Zaccarini D, Ross J, Huang R, Sivapiragasam A. Molecular targets in salivary gland cancers: a comprehensive genomic analysis of 118 mucoepidermoid carcinoma tumors. Biomedicines 2023; 11.

48 

Wang K, McDermott JD, Schrock AB, et al. Comprehensive genomic profiling of salivary mucoepidermoid carcinomas reveals frequent BAP1, PIK3CA, and other actionable genomic alterations. Ann Oncol 2017; 28: 748-753.

49 

Morita M, Murase T, Okumura Y, et al. Clinicopathological signi-ficance of EGFR pathway gene mutations and CRTC1/3-MAML2 fusions in salivary gland mucoepidermoid carcinoma. Histopathology 2020; 76: 1013-1022.

50 

Kato S, Elkin SK, Schwaederle M, et al. Genomic landscape of sali-vary gland tumors. Oncotarget 2015; 6: 25631-25645.

51 

Cantù G. Adenoid cystic carcinoma. An indolent but aggressive tumour. Part A: from aetiopathogenesis to diagnosis. Acta Otorhinolaryngol Ital 2021; 41: 206-214.

52 

Sung MW, Kim KH, Kim JW, et al. Clinicopathologic predictors and impact of distant metastasis from adenoid cystic carcinoma of the head and neck. Arch Otolaryngol Head Neck Surg 2003; 129: 1193-1197.

53 

Liu X, Yang X, Zhan C, Zhang Y, Hou J, Yin X. Perineural invasion in adenoid cystic carcinoma of the salivary glands: where we are and where we need to go. Front Oncol 2020; 10: 1493.

54 

Cordesmeyer R, Schliephake H, Kauffmann P, et al. Clinical prognostic factors of salivary adenoid cystic carcinoma: A single-center analysis of 61 patients. J Craniomaxillofac Surg 2017; 45: 1784-1787.

55 

Ferrarotto R, Mitani Y, Diao L, et al. Activating NOTCH1 mutations define a distinct subgroup of patients with adenoid cystic carcinoma who have poor prognosis, propensity to bone and liver metastasis, and potential responsiveness to Notch1 inhibitors. J Clin Oncol 2017; 35: 352-360.

56 

Sahara S, Herzog AE, Nör JE. Systemic therapies for salivary gland adenoid cystic carcinoma. Am J Cancer Res 2021; 11: 4092-4110.

57 

Ferrarotto R, Mitani Y, McGrail DJ, et al. Proteogenomic analysis of salivary adenoid cystic carcinomas defines molecular subtypes and identifies therapeutic targets. Clin Cancer Res 2021; 27: 852-864.

58 

Xu LH, Zhao F, Yang WW, et al. MYB promotes the growth and metastasis of salivary adenoid cystic carcinoma. Int J Oncol 2019; 54: 1579-1590.

59 

Ho AS, Ochoa A, Jayakumaran G, et al. Genetic hallmarks of recurrent/metastatic adenoid cystic carcinoma. J Clin Invest 2019; 129: 4276-4289.

60 

Weaver AN, Lakritz S, Mandair D, Ulanja MB, Bowles DW. A mole-cular guide to systemic therapy in salivary gland carcinoma. Head Neck 2023; 45: 1315-1326.

61 

Wagner VP, Bingle CD, Bingle L. MYB-NFIB fusion transcript in adenoid cystic carcinoma: current state of knowledge and future directions. Crit Rev Oncol Hematol 2022; 176: 103745.

62 

Drier Y, Cotton MJ, Williamson KE, et al. An oncogenic MYB feedback loop drives alternate cell fates in adenoid cystic carcinoma. Nature Genetics 2016; 48: 265-272.

63 

Stenman G, Andersson MK, Andrén Y. New tricks from an old oncogene: gene fusion and copy number alterations of MYB in human cancer. Cell Cycle 2010; 9: 2986-2995.

64 

Persson M, Andrén Y, Mark J, Horlings HM, Persson F, Stenman G. Recurrent fusion of MYB and NFIB transcription factor genes in carcinomas of the breast and head and neck. Proc Natl Acad Sci U S A 2009; 106: 18740-18744.

65 

Mitani Y, Liu B, Rao PH, et al. Novel MYBL1 Gene Rearrangements with Recurrent MYBL1-NFIB Fusions in Salivary Adenoid Cystic Carcinomas Lacking t(6;9) Translocations. Clin Cancer Res 2016; 22: 725 733.

66 

Lee RH, Wai KC, Chan JW, Ha PK, Kang H. Approaches to the mana-gement of metastatic adenoid cystic carcinoma. Cancers (Basel) 2022; 14.

67 

Stephens PJ, Davies HR, Mitani Y, et al. Whole exome sequencing of adenoid cystic carcinoma. J Clin Invest 2013; 123: 2965-2968.

68 

Wang S, Yu Y, Fang Y, et al. Whole-exome sequencing reveals genetic underpinnings of salivary adenoid cystic carcinoma in the Chinese population. J Genet Genomics 2020; 47: 397-401.

69 

Ross JS, Gay LM, Wang K, et al. Comprehensive genomic profiles of metastatic and relapsed salivary gland carcinomas are associated with tumor type and reveal new routes to targeted therapies. Ann Oncol 2017; 28: 2539-2546.

70 

Chae YK, Chung SY, Davis AA, et al. Adenoid cystic carcinoma: current therapy and potential therapeutic advances based on genomic profiling. Oncotarget 2015; 6: 37117-37134.

71 

Dillon PM, Chakraborty S, Moskaluk CA, Joshi PJ, Thomas CY. Adenoid cystic carcinoma: a review of recent advances, molecular targets, and clinical trials. Head Neck 2016; 38: 620-627.

72 

Vander Poorten V, Triantafyllou A, Thompson LD, et al. Salivary acinic cell carcinoma: reappraisal and update. Eur Arch Otorhinolaryngol 2016; 273: 3511-3531.

73 

Al-Zaher N, Obeid A, Al-Salam S, Al-Kayyali BS. Acinic cell carcinoma of the salivary glands: a literature review. Hematol Oncol Stem Cell Ther 2009; 2: 259-264.

74 

Dublin JC, Oliver JR, Tam MM, et al. Nodal metastases in pediatric and adult acinic cell carcinoma of the major salivary glands. Otolaryngol Head Neck Surg 2022; 167: 941-951.

75 

Haller F, Bieg M, Will R, et al. Enhancer hijacking activates oncogenic transcription factor NR4A3 in acinic cell carcinomas of the salivary glands. Nat Commun 2019; 10: 368.

76 

Andreasen S, Varma S, Barasch N, et al. The HTN3-MSANTD3 fusion gene defines a subset of acinic cell carcinoma of the salivary gland. Am J Surg Pathol 2019; 43: 489-496.

77 

Haller F, Skálová A, Ihrler S, et al. Nuclear NR4A3 immunostaining is a specific and sensitive novel marker for acinic cell carcinoma of the salivary glands. Am J Surg Pathol 2019; 43: 1264-1272.

78 

Dogan S, Xu B, Rana S, et al. Loss of CDKN2A/B is a molecular marker of high-grade histology and is associated with aggressive behavior in acinic cell carcinoma. Mod Pathol 2023; 36: 100150.

79 

Kreuger IZM, Slieker RC, van Groningen T, van Doorn R. Therapeutic strategies for targeting CDKN2A loss in melanoma. J Invest Dermatol 2023; 143: 18-25.e11.

80 

Zhao R, Choi BY, Lee MH, Bode AM, Dong Z. Implications of genetic and epigenetic alterations of CDKN2A (p16(INK4a)) in cancer. EBioMedicine 2016; 8: 30-39.

81 

Hellquist H, Paiva-Correia A, Vander Poorten V, et al. Analysis of the clinical relevance of histological classification of benign epithelial salivary gland tumours. Adv Ther 2019; 36: 1950-1974.

82 

Valstar MH, de Ridder M, van den Broek EC, et al. Salivary gland pleomorphic adenoma in the Netherlands: A nationwide observational study of primary tumor incidence, malignant transformation, recurrence, and risk factors for recurrence. Oral Oncol 2017; 66: 93-99.

83 

Gupta A, Koochakzadeh S, Neskey DM, Nguyen SA, Lentsch EJ. Carcinoma ex pleomorphic adenoma: A review of incidence, demographics, risk factors, and survival. Am J Otolaryngol 2019; 40: 102279.

84 

Antony J, Gopalan V, Smith RA, Lam AK. Carcinoma ex pleomorphic adenoma: a comprehensive review of clinical, pathological and molecular data. Head Neck Pathol 2012; 6: 1-9.

85 

Park KS, Kim JH, Lee DH, Lee JK, Lim SC. Carcinoma ex pleomorphic adenoma of the parotid gland. Am J Otolaryngol 2022; 43: 103389.

86 

Suzuki M, Matsuzuka T, Saijo S, et al. Carcinoma ex pleomorphic adenoma of the parotid gland: a multi-institutional retrospective analysis in the Northern Japan Head and Neck Cancer So-ciety. Acta Otolaryngol 2016; 136: 1154-1158.

87 

Nandini DB, Singh WT, Aparnadevi P, Ningombam DS. Epithelial-myoepithelial carcinoma ex pleomorphic adenoma of the pa-rotid gland with unique histologic differentiation: a rare case report. J Oral Maxillofac Pathol 2022; 26: S34-s39.

88 

Dalin MG, Katabi N, Persson M, et al. Multi-dimensional genomic analysis of myoepithelial carcinoma identifies prevalent oncogenic gene fusions. Nat Commun 2017; 8: 1197.

89 

Katabi N, Gomez D, Klimstra DS, Carlson DL, Lee N, Ghossein R. Prognostic factors of recurrence in salivary carcinoma ex pleomorphic adenoma, with emphasis on the carcinoma histologic subtype: a clinicopathologic study of 43 cases. Hum Pathol 2010; 41: 927-934.

90 

Stenman G. Fusion oncogenes in salivary gland tumors: molecular and clinical consequences. Head Neck Pathol 2013; 7 Suppl 1: S12-19.

91 

Katabi N, Ghossein R, Ho A, et al. Consistent PLAG1 and HMGA2 abnormalities distinguish carcinoma ex-pleomorphic adenoma from its de novo counterparts. Hum Pathol 2015; 46: 26-33.

92 

Persson F, Andrén Y, Winnes M, et al. High-resolution genomic profiling of adenomas and carcinomas of the salivary glands reveals amplification, rearrangement, and fusion of HMGA2. Genes Chromosomes Cancer 2009; 48: 69-82.

93 

Bell D, J NM, Rao PH, El-Naggar AK. t(3;8) as the sole chromosomal abnormality in a myoepithelial carcinoma ex pleomorphic adenoma: a putative progression event. Head Neck 2013; 35: E181-183.

94 

El-Naggar AK, Callender D, Coombes MM, Hurr K, Luna MA, Batsakis JG. Molecular genetic alterations in carcinoma ex-pleomorphic adenoma: a putative progression model? Genes Chromosomes Cancer 2000; 27: 162-168.

95 

Xu B, Katabi N. Myoepithelial Carcinoma. Surg Pathol Clin 2021; 14: 67-73.

96 

Lavareze L, Scarini JF, de Lima-Souza RA, et al. Clinicopathological and survival profile of patients with salivary gland myoepithelial carcinoma: A systematic review. J Oral Pathol Med 2023; 52: 101-108.

97 

Kong M, Drill EN, Morris L, et al. Prognostic factors in myoepithelial carcinoma of salivary glands: a clinicopathologic study of 48 cases. Am J Surg Pathol 2015; 39: 931-938.

98 

Su YX, Roberts DB, Hanna EY, et al. Risk factors and prognosis for myoepithelial carcinoma of the major salivary glands. Ann Surg Oncol 2015; 22: 3701-3707.

99 

Di Palma S, Guzzo M. Malignant myoepithelioma of salivary glands: clinicopathological features of ten cases. Virchows Arch A Pathol Anat Histopathol 1993; 423: 389-396.

100 

Soluk-Tekkeşin M, Wright JM. The World Health Organization Classification of Odontogenic Lesions: a summary of the changes of the 2017 (4th) edition. Turk Patoloji Derg 2018; 34.

101 

Haltiner CC, Betz S, Smith J, Nelson B, Ambrosio AA. Carcinoma ex-pleomorphic adenoma diagnosis during global health enga-gement operations. Mil Med 2021; 186: 828-832.

102 

Skálová A, Weinreb I, Hyrcza M, et al. Clear cell myoepithelial carcinoma of salivary glands showing EWSR1 rearrangement: molecular analysis of 94 salivary gland carcinomas with prominent clear cell component. Am J Surg Pathol 2015; 39: 338-348.

103 

Pikul J, Rzepakowska A, Machnicki M, Stokłosa T. FGFR2 point mutation in 2 cases of pleomorphic adenoma progressing to myo-epithelial carcinoma. Contemp Oncol (Pozn) 2023; 27: 211-216.

104 

Gandhi J, Mantilla JG, Ricciotti RW, Chen EY, Liu YJ, Bandhlish A. Myoepithelial carcinoma of theparotid gland with a novel CTCF: NCOA2 fusion. Genes Chromosomes Cancer 2023; 62: 161-166.

105 

Patel TD, Vazquez A, Marchiano E, Park RC, Baredes S, Eloy JA. Polymorphous low-grade adenocarcinoma of the head and neck: A population-based study of 460 cases. Laryngoscope 2015; 125: 1644 1649.

106 

Mimica X, Katabi N, McGill MR, et al. Polymorphous adenocarcinoma of salivary glands. Oral Oncol 2019; 95: 52-58.

107 

Weinreb I, Piscuoglio S, Martelotto LG, et al. Hotspot activating PRKD1 somatic mutations in polymorphous low-grade adenocarcinomas of the salivary glands. Nat Genet 2014; 46: 1166-1169.

108 

Andreasen S, Melchior LC, Kiss K, et al. The PRKD1 E710D hotspot mutation is highly specific in separating polymorphous adeno-carcinoma of the palate from adenoid cystic carcinoma and pleomorphic adenoma on FNA. Cancer Cytopathol 2018; 126: 275-281.

109 

Xu B, Barbieri AL, Bishop JA, et al. Histologic Classification and Molecular Signature of Polymorphous Adenocarcinoma (PAC) and Cribriform Adenocarcinoma of Salivary Gland (CASG): an international interobserver study. Am J Surg Pathol 2020; 44: 545-552.

110 

Skalova A, Sima R, Kaspirkova-Nemcova J, et al. Cribriform ade-nocarcinoma of minor salivary gland origin principally affecting the tongue: characterization of new entity. Am J Surg Pathol 2011; 35: 1168-1176.

111 

Laco J, Kamarádová K, Vítková P, et al. Cribriform adenocarcinoma of minor salivary glands may express galectin-3, cytokera--tin 19, and HBME-1 and contains polymorphisms of RET and H-RAS proto oncogenes. Virchows Arch 2012; 461: 531-540.

112 

Sebastiao APM, Xu B, Lozada JR, et al. Histologic spectrum of polymorphous adenocarcinoma of the salivary gland harbor genetic alterations affecting PRKD genes. Mod Pathol 2020; 33: 65-73.

113 

de Jager VD, de Visscher S, Schuuring E, Doff JJ, van Kempen LC. A novel PPP2R2A::PRKD1 fusion in a cribriform adenocarcinoma of salivary gland. Genes Chromosomes Cancer 2023; 62: 297-300.

114 

Owosho AA, Baker E, Wood CB, Jain R. A novel STRN3::PRKD1 fusion in a cribriform adenocarcinoma of salivary gland with high-grade transformation. Genes Chromosomes Cancer 2023; 62: 624 628.

115 

Weinreb I, Zhang L, Tirunagari LM, et al. Novel PRKD gene rearrangements and variant fusions in cribriform adenocarcinoma of salivary gland origin. Genes Chromosomes Cancer 2014; 53: 845-856.

116 

Freiberger SN, Brada M, Fritz C, et al. SalvGlandDx–a comprehensive salivary gland neoplasm specific next generation sequencing panel to facilitate diagnosis and identify therapeutic targets. Neoplasia 2021; 23: 473-487.

117 

Bishop JA, Weinreb I, Swanson D, et al. Microsecretory adenocarcinoma: a novel salivary gland tumor characterized by a recurrent MEF2C-SS18 fusion. Am J Surg Pathol 2019; 43: 1023-1032.

118 

Bishop JA, Sajed DP, Weinreb I, et al. Microsecretory adenocarcinoma of salivary glands: an expanded series of 24 cases. Head Neck Pathol 2021; 15: 1192-1201.

119 

Schmitt NC, Kang H, Sharma A. Salivary duct carcinoma: an aggressive salivary gland malignancy with opportunities for targeted therapy. Oral Oncol 2017; 74: 40-48.

120 

Johnston ML, Huang SH, Waldron JN, et al. Salivary duct carcinoma: treatment, outcomes, and patterns of failure. Head Neck 2016; 38 Suppl 1: E820-826.

121 

Gilbert MR, Sharma A, Schmitt NC, et al. A 20-year review of 75 cases of salivary duct carcinoma.JAMA Otolaryngol Head Neck Surg 2016; 142: 489-495.

122 

Williams L, Thompson LD, Seethala RR, et al. Salivary duct carcinoma: the predominance of apocrine morphology, prevalence of histologic variants, and androgen receptor expression. Am J Surg Pathol 2015; 39: 705-713.

123 

Mueller SA, Gauthier MA, Blackburn J, et al. Molecular patterns in salivary duct carcinoma identifyprognostic subgroups. Mod Pathol 2020; 33: 1896-1909.

124 

Luukkaa H, Klemi P, Leivo I, et al. Salivary gland cancer in Finland 1991–1996: an evaluation of 237 cases. Acta Otolaryngol 2005; 125: 207-214.

125 

Luk PP, Weston JD, Yu B, et al. Salivary duct carcinoma: clinicopathologic features, morphologic spectrum, and somatic mutations. Head Neck 2016; 38 Suppl 1: E1838-1847.

126 

Dalin MG, Desrichard A, Katabi N, et al. Comprehensive mole-cular characterization of salivary duct carcinoma reveals actionable targets and similarity to apocrine breast cancer. Clin Cancer Res 2016; 22: 4623-4633.

127 

Dogan S, Ng CKY, Xu B, Kumar R, et al. The repertoire of gene-tic alterations in salivary duct carcinoma including a novel HNRNPH3-ALK rearrangement. Hum Pathol 2019; 88: 66-77.

128 

Chiosea SI, Thompson LD, Weinreb I, et al. Subsets of salivary duct carcinoma defined by morphologic evidence of pleomorphic adenoma, PLAG1 or HMGA2 rearrangements, and common genetic alterations. Cancer 2016; 122: 3136-3144.

129 

Boon E, Bel M, van Boxtel W, et al. A clinicopathological study and prognostic factor analysis of 177 salivary duct carcinoma patients from The Netherlands. Int J Cancer 2018; 143: 758-766.

130 

Skálová A, Stárek I, Vanecek T, et al. Expression of HER-2/neu gene and protein in salivary duct carcinomas of parotid gland as revealed by fluorescence in-situ hybridization and immunohistochemistry. Histopathology 2003; 42: 348-356.

131 

Dalin MG, Watson PA, Ho AL, Morris LG. Androgen receptor signaling in salivary gland cancer. Cancers (Basel) 2017; 9.

132 

Williams CYK, Townson AT, Terry N, Schmitt NC, Sharma A. Role of HER2 in prognosis of salivary duct carcinoma: a systematic review and meta-analysis. Laryngoscope 2023; 133: 476-484.

133 

Kawakita D, Nagao T, Takahashi H, et al. Survival benefit of HER2-targeted or androgen deprivation therapy in salivary duct carcinoma. Ther Adv Med Oncol 2022; 14: 17588359221119538.

134 

Filippini DM, Pagani R, Tober N, et al. HER2-targeted therapies for salivary gland cancers. Oral Oncol 2024; 148: 106612.

135 

Vos JL, Burman B, Jain S, et al. Nivolumab plus ipilimumab in advanced salivary gland cancer: a phase 2 trial. Nature Medicine 2023; 29: 3077-3089.

136 

Kohsaka S, Tada Y, Ando M, et al. Identification of novel prognostic and predictive biomarkers in salivary duct carcinoma via comprehensive molecular profiling. NPJ Precis Oncol 2022; 6: 82.

137 

Nardi V, Sadow PM, Juric D, et al. Detection of novel actionable genetic changes in salivary duct carcinoma helps direct patient treatment. Clin Cancer Res 2013; 19: 480-490.

138 

Wang H, Guo M, Wei H, Chen Y. Targeting p53 pathways: mecha-nisms, structures, and advances in therapy. Signal Transduct Target Ther 2023; 8: 92.

139 

Hassin O, Oren M. Drugging p53 in cancer: one protein, many targets. Nat Rev Drug Discov 2023; 22: 127-144.

140 

Ku BM, Jung HA, Sun JM, et al. High-throughput profiling identifies clinically actionable mutations in salivary duct carcinoma. J Transl Med 2014; 12: 299.

141 

Grünewald I, Trautmann M, Busch A, et al. MDM2 and CDK4 amplifications are rare events in salivary duct carcinomas. Oncotarget 2016; 7: 75261-75272.

142 

Politi M, Robiony M, Avellini C, Orsaria M. Epithelial-myoepithelial carcinoma of the parotid gland: Clinicopathological aspect, diagnosis and surgical consideration. Ann Maxillofac Surg 2014; 4: 99-102.

143 

Vázquez A, Patel TD, D’Aguillo CM, et al. Epithelial-myoepithelial carcinoma of the salivary glands:an analysis of 246 cases. Otolaryngol Head Neck Surg 2015; 153: 569-574.

144 

El Hallani S, Udager AM, Bell D, et al. Epithelial-myoepithelial carcinoma: frequent morphologic and molecular evidence of preexisting pleomorphic adenoma, common HRAS mutations in PLAG1-intact and HMGA2-intact Cases, and Occasional TP53, FBXW7, and SMARCB1 alterations in high-grade cases. Am J Surg Pathol 2018; 42: 18-27.

145 

Kusafuka K, Yamashita M, Muramatsu A, Arai K, Suzuki M. Epi-thelial-myoepithelial carcinoma ex pleomorphic adenoma of the parotid gland: report of a rare case with immunohistochemical and genetic analyses. Med Mol Morphol 2021; 54: 173-180.

146 

Urano M, Nakaguro M, Yamamoto Y, et al. Diagnostic significance of HRAS mutations in epithelial myoepithelial carcinomas exhibiting a broad histopathologic spectrum. Am J Surg Pathol 2019; 43: 984-994.

147 

Nakaguro M, Nagao T. Epithelial-myoepithelial carcinoma. Surg Pathol Clin 2021; 14: 97-109.

148 

Grünewald I, Vollbrecht C, Meinrath J, et al. Targeted next gene-ration sequencing of parotid gland cancer uncovers genetic hete-rogeneity. Oncotarget 2015; 6: 18224-18237.

149 

Chiosea SI, Miller M, Seethala RR. HRAS mutations in epithelial-myoepithelial carcinoma. Head Neck Pathol 2014; 8: 146-150.

150 

Nakaguro M, Tanigawa M, Hirai H, et al. The diagnostic utility of RAS Q61R mutation-specific immunohistochemistry in epithelial-myoepithelial carcinoma. Am J Surg Pathol 2021; 45: 885-894.

151 

Mäkelä R, Arjonen A, Suryo Rahmanto A, et al. Ex vivo assessment of targeted therapies in a rare metastatic epithelial-myoepithelial carcinoma. Neoplasia 2020; 22: 390-398.

152 

Skálová A, Vanecek T, Sima R, et al. Mammary analogue secretory carcinoma of salivary glands, containing the ETV6-NTRK3 fusion gene: a hitherto undescribed salivary gland tumor entity. Am J Surg Pathol 2010; 34: 599-608.

153 

Bishop JA, Yonescu R, Batista D, Eisele DW, Westra WH. Most nonparotid “acinic cell carcinomas” represent mammary analog secretory carcinomas. Am J Surg Pathol 2013; 37: 1053-1057.

154 

Ayre G, Hyrcza M, Wu J, Berthelet E, Skálová A, Thomson T. Secretory carcinoma of the major salivary gland: Provincial population-based analysis of clinical behavior and outcomes. Head Neck 2019; 41: 1227-1236.

155 

Alves LDB, de Melo AC, Farinha TA, et al. A systematic review of secretory carcinoma of the salivarygland: where are we? Oral Surg Oral Med Oral Pathol Oral Radiol 2021; 132: e143-e152.

156 

Tognon C, Knezevich SR, Huntsman D, et al. Expression of the ETV6-NTRK3 gene fusion as aprimary event in human secretory breast carcinoma. Cancer Cell 2002; 2: 367-376.

157 

Guilmette J, Dias-Santagata D, Nosé V, Lennerz JK, Sadow PM. Novel gene fusions in secretory carcinoma of the salivary glands: enlarging the ETV6 family. Hum Pathol 2019; 83: 50-58.

158 

Rooper LM, Karantanos T, Ning Y, Bishop JA, Gordon SW, Kang H. Salivary secretory carcinoma with a novel ETV6-MET fusion: expanding the molecular spectrum of a recently described entity. Am J Surg Pathol 2018; 42: 1121-1126.

159 

Black M, Liu CZ, Onozato M, et al. Concurrent identification of novel EGFR-SEPT14 fusion and ETV6-RET fusion in secretory carcinoma of the salivary gland. Head Neck Pathol 2020; 14: 817-821.

160 

Ito Y, Ishibashi K, Masaki A, et al. Mammary analogue secretory carcinoma of salivary glands: a clinicopathologic and molecular study including 2 cases harboring ETV6-X fusion. Am J Surg Pathol 2015; 39: 602-610.

161 

Skálová A, Banečkova M, Thompson LDR, et al. Expanding the molecular spectrum of secretory carcinoma of salivary glands with a novel VIM-RET fusion. Am J Surg Pathol 2020; 44: 1295-1307.

162 

Sasaki E, Masago K, Fujita S, Suzuki H, Hanai N, Hosoda W. Sali-vary secretory carcinoma harbouring a novel ALK fusion: expanding the molecular characterization of carcinomas beyond the ETV6 gene. Am J Surg Pathol 2020; 44: 962-969.

163 

Na K, Hernandez-Prera JC, Lim JY, Woo HY, Yoon SO. Characteri-zation of novel genetic alterations in salivary gland secretory carcinoma. Mod Pathol 2020; 33: 541-550.

164 

Skálová A, Vanecek T, Majewska H, et al. Mammary analogue secretory carcinoma of salivary glands with high-grade transformation: report of 3 cases with the ETV6-NTRK3 gene fusion and analysis of TP53, β catenin, EGFR, and CCND1 genes. Am J Surg Pathol 2014; 38: 23-33.

165 

Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol 2020; 21: 271-282.

166 

Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 2020; 21: 531-540.

167 

Regua AT, Najjar M, Lo HW. RET signaling pathway and RET inhibitors in human cancer. Front Oncol 2022; 12: 932353.

168 

Desai A, Faquin WC, Iafrate AJ, Rivera MN, Jaquinet A, Troulis MJ. Clear cell carcinoma: a comprehensive literature review of 254 cases. Int J Oral Maxillofac Surg 2022; 51: 705-712.

169 

Weinreb I. Hyalinizing clear cell carcinoma of salivary gland: a review and update. Head Neck Pathol 2013; 7 Suppl 1: S20-29.

170 

Antonescu CR, Katabi N, Zhang L, et al. EWSR1-ATF1 fusion is a novel and consistent finding in hyalinizing clear-cell carcinoma of salivary gland. Genes Chromosomes Cancer 2011; 50: 559-570.

171 

Chapman E, Skalova A, Ptakova N, et al. Molecular profiling of hyalinizing clear cell carcinomas revealed a subset of tumors harboring a novel EWSR1-CREM fusion: report of 3 cases. Am J Surg Patho 2018; 42: 1182-1189.

172 

Lanic MD, Guérin R, Sater V, et al. A novel SMARCA2-CREM fusion expending the molecular spectrum of salivary gland hyalinazing clear cell carcinoma beyond the FET genes. Genes Chromosomes Cancer 2023; 62: 231-236.

173 

Gonzalez MF, Akhtar I. Hyalinising clear cell carcinoma with a novel fusion gene: Insights into the diagnosis. Cytopathology 2023; 34: 611-616.

174 

Bishop JA, Gagan J, Krane JF, Jo VY. Low-grade apocrine intraductal carcinoma: expanding the morphologic and molecular spectrum of an enigmatic salivary gland tumor. Head Neck Pathol 2020; 14: 869

175 

Giovacchini F, Bensi C, Belli S, et al. Low-grade intraductal carcinoma of salivary glands: A systematic review of this rare entity. J Oral Biol Craniofac Res 2019; 9: 96-110.

176 

Thompson LDR, Bishop JA. Salivary gland intraductal carcinoma: how do 183 reported cases fit into a developing classification. Adv Anat Pathol 2023; 30: 112-129.

177 

Skálová A, Vanecek T, Uro-Coste E, et al. Molecular profiling of salivary gland intraductal carcinoma revealed a subset of tumors harboring NCOA4-RET and novel TRIM27-RET fusions: a report of 17 cases. Am J Surg Pathol 2018; 42: 1445-1455.

178 

Bishop JA, Nakaguro M, Whaley RD, et al. Oncocytic intraductal carcinoma of salivary glands: a distinct variant with TRIM33-RET fusions and BRAF V600E mutations. Histopathology 2021; 79: 338-346.

179 

Skálová A, Ptáková N, Santana T, et al. NCOA4-RET and TRIM27-RET are characteristic gene fusions in salivary intraductal carcinoma, including invasive and metastatic tumors: is “intraductal” correct? Am J Surg Pathol 2019; 43: 1303-1313.

180 

Weinreb I, Bishop JA, Chiosea SI, et al. Recurrent RET gene rearrangements in intraductal carcinomas of salivary gland. Am J Surg Pathol 2018; 42: 442-452.

181 

Viswanathan K, Sadow PM, Maleki Z, et al. Cytomorphologic features of intraductal salivary gland carcinoma: A multi-institutional study of 13 FNA cases with histologic, molecular, and clinical correlations. Cancer Cytopathol 2021; 129: 928-946.

182 

Hsieh MS, Lee YH, Jin YT, Kuo YJ. Clinicopathological study of intraductal carcinoma of the salivary gland, with emphasis on the apocrine type. Virchows Arch 2020; 477: 581-592.

183 

Majewska H, Gorczyński A, Czapiewski P, et al. ALK alterations in salivary gland carcinomas. Virchows Arch 2021; 478: 933-941.

184 

Watanabe T, Honma Y, Yonemori K, Sunami K, Yoshimoto S, Mori T. High-grade intraductal carcinoma of the parotid gland harboring CTNNA1::ALK rearrangement: changes in genetic status using genetic testing during treatment with an ALK inhibitor. Head Neck 2024; 46: E26-e31.

185 

WHO Classification of Head and Neck Tumours, vol. 9, 2017.

186 

Żurek M, Rzepakowska A, Jasak K, Niemczyk K. The epidemiology of salivary glands pathologies in adult population over 10 years in poland–cohort study. Int J Env Res Public Health 2022; 19: 179.

187 

Kucharska E, Rzepakowska A, Żurek M, et al. Oncologic outcomes of the most prevalent major salivary gland cancers: retrospective cohort study from single center. Eur Arch Otorhinolaryngol 2024; 281: 4305-4313.

188 

Khosla D, Madan R, Goyal S, Kumar N, Kapoor R. Polymorphous low-grade adenocarcinoma of thesalivary glands–a review. Niger Med J 2021; 62: 49-53.

189 

Bishop JA, Sajed DP. Microsecretory adenocarcinoma of salivary glands. Adv Anatom Pathol 2023; 30: 130-135.

190 

Rito M, Mitani Y, Bell D, et al. Frequent and differential mutations of the CYLD gene in basal cell salivary neoplasms: linkage to tumor development and progression. Mod Pathol 2018; 31: 1064-1072.

191 

Rooper LM, Argyris PP, Thompson LDR, et al. Salivary mucinous adenocarcinoma is a histologically diverse single entity with recurrent AKT1 E17K mutations: clinicopathologic and molecular characterization with proposal for a unified classification. Am J Surg Pathol 2021; 45: 1337-1347.

192 

Baněčková M, Thompson LDR, Hyrcza MD, et al. Salivary gland secretory carcinoma: clinicopathologic and genetic characteristics of 215 cases and proposal for a grading system. Am J Surg Pathol 2023; 47: 661-677.

193 

Asahina M, Saito T, Hayashi T, Fukumura Y, Mitani K, Yao T. Clinicopathological effect of PLAG1 fusion genes in pleomorphic adenoma and carcinoma ex pleomorphic adenoma with special emphasis on histological features. Histopathology 2019; 74: 514-525.

194 

Skálová A, Agaimy A, Vanecek T, et al. Molecular profiling of clear cell myoepithelial carcinoma of salivary glands with ewsr1 rearrangement identifies frequent PLAG1 gene fusions but no EWSR1 fusion transcripts. Am J Surg Pathol 2021; 45: 1-13.

195 

Bubola J, MacMillan CM, Demicco EG, et al. Targeted RNA sequencing in the routine clinical detection of fusion genes in salivary gland tumors. Genes Chromosomes Cancer 2021; 60: 695-708.

196 

Rupp NJ, Höller S, Brada M, et al. Expanding the clinicopathological spectrum of TGFBR3-PLAG1 rearranged salivary gland neoplasms with myoepithelial differentiation including evidence of high-grade transformation. Genes Chromosomes Cancer 2022; 61: 94-104.

197 

Katabi N, Sukhadia P, DiNapoli SE, et al. Expanding the histolo-gical spectrum of salivary gland neoplasms with HMGA2::WIF1 fusion emphasising their malignant potential: a report of eight cases. Histopathology 2024; 84: 387-398.

198 

Smith ME, Surrey LF, Zhang PJ, Weinstein GS, LiVolsi VA. Molecular identification of an ETV6-RET fusion in a secretory carcinoma associated with a pleomorphic adenoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2022; 134: 733-738.

199 

Pircher M, Briner HR, Bonomo M, et al. Mixed response and mechanisms of resistance to larotrectinib in metastatic carcinoma ex pleomorphic adenoma of the parotid harboring an NTRK2 fusion: A case report. Medicine (Baltimore) 2021; 100: e24463.

Copyright: © 2024 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.