Next Article in Journal
The Effectiveness and Safety of Pharmaceutical-Grade Cannabidiol in the Treatment of Mastocytosis-Associated Pain: A Pilot Study
Previous Article in Journal
Weighted Gene Co-Expression Network Analysis (WGCNA) Discovered Novel Long Non-Coding RNAs for Polycystic Ovary Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Molecular Targets in Salivary Gland Cancers: A Comprehensive Genomic Analysis of 118 Mucoepidermoid Carcinoma Tumors

by
Maroun Bou Zerdan
1,
Prashanth Ashok Kumar
2,
Daniel Zaccarini
3,
Jeffrey Ross
4,5,
Richard Huang
3 and
Abirami Sivapiragasam
2,*
1
Department of Internal Medicine, SUNY Upstate Medical University, Syracuse, NY 13210, USA
2
Division of Hematology/Oncology, Department of Medicine, SUNY Upstate Medical University, Syracuse, NY 13210, USA
3
Department of Pathology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
4
Foundation Medicine, Inc., Morrisville, NC 27560, USA
5
Departments of Pathology and Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(2), 519; https://doi.org/10.3390/biomedicines11020519
Submission received: 26 December 2022 / Revised: 24 January 2023 / Accepted: 6 February 2023 / Published: 10 February 2023
(This article belongs to the Section Cancer Biology and Oncology)

Abstract

:
Introduction: Salivary gland carcinomas (SGC) are histologically diverse cancers and next-generation sequencing (NGS) to identify key molecular targets is an important aspect in the management of advanced cases. Methods: DNA was extracted from paraffin embedded tissues of advanced SGC and comprehensive genomic profiling (CGP) was carried out to evaluate for base substitutions, short insertions, deletions, copy number changes, gene fusions and rearrangements. Tumor mutation burden (TMB) was calculated on approximately 1.25 Mb. Some 324 genes in the FoundationOne CDX panel were analyzed. Results: Mucoepidermoid carcinoma (MECa) mutations were assessed. CDKN2A and CDKN2B GA were common in mucoepidermoid carcinoma (MECa) (52.5 and 30.5%). PIK3CA was also common in MECa (16.9%). ERBB2 amplification/short variants (amp/SV) were found in MECa (5.9/0%). HRAS GA was common in MECa (14.4%) as well. Other targets, including BAP1, PTEN, and KRAS, were noted but had a low incidence. In terms of immunotherapy (IO)-predictive markers, TMB > 10 was more common in MECa (16.9%). PDL1 high was also seen in MECa (4.20%). Conclusion: SGC are rare tumors with no FDA-approved treatment options. This large dataset reveals many opportunities for IO and targeted therapy contributing to the continuously increased precision in the selection of treatment for these patients.

1. Introduction

Malignant salivary gland carcinomas (SGC) are an uncommon group of head and neck cancers [1,2,3]. One of the most frequent types of SGC is mucoepidermoid carcinoma (MECa). Due to the rarity of the disease, data are often derived from case reports and retrospective series, rather than prospectively performed clinical trials. Thus, it has been challenging to define the role of chemotherapy or immunotherapy in management of metastatic or recurrent SGC.
Many possible biological targets have been identified. These include but are not limited to c-Kit [4], EGFR [5], HER2 [6,7], androgen, estrogen and progesterone receptor protein expression by immunohistochemistry [8], and PIK3CA [9] and BRAF mutations [7].
Although targeted therapies with kinase inhibitors [10,11,12] and monoclonal antibodies [13,14] have generally had low response rates, these therapies were given to unselected patients rather than matched to individuals whose tumors harbored equivalent aberrations [10,11,12,13,14]. On the contrary, when patients were appropriately treated for the presence of PIK3CA or ERBB2/HER2 alterations, significant responses have been described with alpelisib, trastuzumab and lapatinib [15], or mTOR inhibitors [16].
Since we are moving away from a one-size-fits-all approach to a tailored approach to SGC, further molecular understanding of salivary gland tumors is necessary. Here, we report the most frequent aberrations in two subtypes of SGC.

2. Methods Institutional Review Board (IRB)

Approval for this study was obtained from the Western IRB (Protocol No. 619478). A retrospective database search of a CLIA-certified and CAP-accredited reference molecular laboratory was performed for 1757 of SGC tissue samples. Clinicopathological data confirming that all cases were clinically advanced and metastatic SGC, including patient age and gender, routine histology and immunohistochemical staining results and confirmation of the diagnosis, were extracted from medical records and pathology reports. One (hematoxylin and eosin) H&E slide, corresponding to the tissue that was submitted for genomic profiling, was available for each case to review morphological features.

2.1. Cohort

Comprehensive genomic profiling of all 1757 SGC tissue samples was performed using a commercially available FDA-approved hybridization-captured, adaptor ligation–based libraries using DNA and RNA extracted from formalin-fixed paraffin-embedded tumor in a CLIA- and CAP-certified laboratory (FoundationOneCDx, Foundation Medicine, Inc., Cambridge, MA, USA). On central pathology review, all samples forwarded for DNA extraction contained a minimum of 20% tumor cells. The samples were assayed using adaptor-ligation and hybrid capture next-generation sequencing for all coding exons from up to 324 cancer related genes, plus select introns from up to 31 genes frequently rearranged in cancer. Patient samples were sequenced and evaluated for genomic alterations including base substitutions, insertions, deletions, copy number alterations (amplifications and homozygous deletions), and for select gene fusions/rearrangements, as previously described [17]. The bioinformatics processes used in this study included Bayesian algorithms to detect base substitutions, local assembly algorithms to detect short insertions and deletions, a comparison with process-matched normal control samples to detect gene copy number alterations and an analysis of chimeric read pairs to identify gene fusions, as previously described [17]. The tumor mutational burden was determined on 0.83–1.14 Mb of sequenced DNA using a mutation burden estimation algorithm that, based on the genomic alterations detected, extrapolates to the exome or the genome as a whole, as previously described [18]. In this study, low tumor mutational burden scores were defined as mean TMB, median TMB, percentage of cases with TMB ≥ 10 mutations/Mb, and percentage of cases with TMB ≥ 20 mutations/Mb. Assessment of microsatellite instability was performed from DNA sequencing across 114 loci, as previously described [19]. Each microsatellite locus had repeat length of 7–39 bp. The next-generation sequencing- based “microsatellite instability score” was translated into categorical microsatellite instability high, microsatellite instability intermediate, or microsatellite stable by unsupervised clustering of specimens, for which microsatellite instability status was previously assessed via gold standard methods. Only cases related to muco-epidermoid carcinoma will be discussed in this excerpt.

2.2. Laboratory Methods

PD-L1 expression was determined on subsets of the tumors using the DAKO 22C3 assay, with low positive tumor cell scoring defined as 1–49% staining and high positive tumor cell scoring defined as ≥50% staining.

2.3. Statistical Analysis

Fisher’s exact test was used in the statistical comparisons of the MECa with the other previously published groups of salivary gland carcinomas. Statistical significance was defined as p < 0.05.

3. Results

Mucoepidermoid Carcinoma

Out of a total of 1757 cases, there were a total of 118 MECa tumors having 4.88 genomic alterations (GA)/tumor (Table 1). The median age of patients was 64, with a range of 16 to 89 years. Multiple mutations were found to be present in genes related to cell cycle regulation, chromosome, and chromatin remodeling along with different pathway-related genes. Concerning GA related to cell cycle regulation, 40.70% (n = 48) TP53 GA, 52.50% (n = 62) CDKN2A GA, 30.50% (n = 36) CDKN2B GA, 3.40% (n = 4) were found in each of CCND1 and RB1 GA, while chromosome remodeling related genets such as TERT and ARID1A were found in 15% (n = 18) and 2.50% (n = 3), respectively. BRCA1, BRCA2, ATM, PALB2, and BAP1 were the DNA damage response GA found in our tumor samples. Only 0.80% (n = 1) tumor was found to have PALB2, while 18.60% (n = 22) were found to have BAP1. BRCA1, BRCA2, ATM were found in 1.70% (n = 2), 5.90% (n = 7), 4.20% (n = 5), respectively. Nine tumors, 7.60%, were found to have PTEN, and 20 tumors, 16.90%, were found to have PIK3CA mutations. NF1 and TSC2 were found to be present in 5 and 1 tumors, respectively. ERBB2 and EGFR were found in 5.9% (n = 7) and 0.8% (n = 1), respectively compared to FGFR1 and KIT, which were found in 5.1% (n = 6) and 0.80% (n = 1), respectively. NOTCH1 and NOTCH2 were found in 4.20% (n = 5) and 7.60% (n = 9), respectively. MTAP and MYC were, respectively found in 16 tumors, 13.70%, and 2 tumors, 1.70%. GA related to RAS-RAF pathway were as follows: 3 tumors had KRAS G12C as a GA, while 17 had HRAS GA and 2 had BRAF GA.
A total of 100 cases of MECa tumors had their immuno-oncology biomarkers investigated. None had a reported MSI high frequency. Similarly, CD274 (PD-L1) Amp was not detected in any tumors as well. The median TMB observed was 2.6; TMB > 10% was 16.90% and TMB > 20% was 12.70%. PD-L1 low expression was seen in 26 cases vs. high expression, which was 4.20%. MDM2 Amp and STK11 inactivating GA were each observed in 3.4%, n = 3 tumors.

4. Discussion

Mucoepidermoid carcinoma (MECa), the most common malignant salivary gland neoplasms across all ages, are histologically composed of squamoid, mucin-producing and intermediate cell types, which may form various patterns, including cystic and/or solid areas [20,21]. Some of the variants which have been identified include oncocytic, clear cell, sebaceous, and sclerosing variants [21]. According to the World Health Organization (WHO), MECa are divided into three groups: low, intermediate, and high histologic grade, with different clinical courses [21]. Several systems suggest stratifying MECa according to prognostic significance. Some of these include the Brandwein system, the Armed Forces Institute of Pathology scoring system, the Katabi stratification method, and the modified Hailey system [22,23].
In a study performed by Kato et al., 117 patients with salivary gland tumors were assessed, only five of which had MECa tumors [24]. Out of these patients, GA in each of TP53, PI3K, BAP1, and PTEN were found in two patients. GA in PIK3CA, HRAS, NF1 and the cyclin-dependent pathway, which included the following aberrations: CCNDI, CDK4/6 or CDKN2A/B, were found in one patient. Compared to this, our study had a higher percentage of the aforementioned mutations.

4.1. Genetic Alterations

TP53 is a marker of aggressive behavior and poor overall response in all malignancies in general [25]. TP53 alterations were historically reported in 25–33% of MEC, although the sample size was limited in these studies [26]. Compared to this, our study reported TP53 alteration in 40.7% of the patients. TP53 alterations are preferentially found in high grade tumors, indicating that they may represent a switch for transformation from low to high grade [27].
Some reports have indicated that EGFR overexpression may be found in 73% of high grade MEC and connotes a poor prognosis. However, EGFR alterations are generally rare in salivary gland tumors overall and were barely noted in our dataset [27]. Drugs such as Sotorosib have promising anti-cancer activity in KRAS G12C mutated advanced solid cancers [28]. KRAS G12C represents around 14.5% of all KRAS alterations in all tumors [29]. This was found in 2.5% of our cohort of MEC. PIK3CA with agents such as Alpelisib [R6] was found in 16.8% in our cohort. AR alteration, which is frequent in other salivary gland tumors such as salivary ductal carcinoma, was not found in MEC in our analysis [30].
The most significant genetic alteration in MECa is characterized by t(11;19)(q21;p13) translocation which results in the fusion of CREB regulated transcription coactivator 1(CRTC1)- mammalian mastermind-like gene (MAML2) [31]. This translocation, which acts as a potential main driver for mutations, can be detected in 55–88% of MECa [32,33,34]. CAMP response element-binding protein (CREB) is encoded by the CRTC1 gene which is located on chromosome 19. CRTC1, which is also known as MECT1, TORC1, and WAMTP1, modulates cellular pathways mediated by activating protein 1 (AP-1) [32,34,35,36]. CRTC1’s binding to CREB enhances its transcription, thus allowing it to carry out its role in gene cellular proliferation and differentiation. Here, it is worth mentioning that when a fusion occurs in exon 1 of the CRTC1 gene and exon 2 to 5 of the MAML2 gene, the CREB-binding domain of CRTC1 replaces the notch-binding domain of MAML2, thus activating the notch pathway [33,34,35,36]. This newly formed fusion protein activates transcription of cAMP target genes, including PEPCK1, AREG, MMP10, IL6, NR4A2, and NR4A3. AREG detection using immunohistochemistry may help identify fusion-positive MECa [37]. MECa tumorigenesis is thought to arise from the interaction of the fusion gene with MYC and AP-1 [38,39]. Another translocation in rare types of MECa occurs between the EWSR1 and POU5F1 genes [40].
CRTC3-MAML2 gene fusion is less commonly detected in a small subset (around 5%) of MECa [39]. Studies have shown that CRTC1-MAML2 and CRTC3-MAML2 mutations are mutually exclusive [36]. An unfavorable prognosis has been reported in CRTC1-MAML2 fusion-positive MECa with CDKN2A deletions [41].

4.2. Diagnostic Markers

MECa diagnosis can be carried out solely using histology, especially when the tumor shows the typical admixture of squamoid, mucinous, and intermediate cells. When histology is not sufficient for diagnosis, genetic or molecular tests can be utilized. Genetic tests for rearrangements can be helpful to distinguish a Warthin tumor from the Warthin-like variant of MECa and other clear cell maligancies of the salivary gland [42]. Molecular tests, such as a reverse transcription polymerase chain reaction or fluorescent in situ hybridization (FISH) aid in diagnosing variants of MECa, such as oncocytic or clear cell types [43,44,45]. A translocation can be detected by these techniques, especially when FISH and break-apart probes are designed to detect genes. Furthermore, using these techniques, the MAML2 rearrangement can be detected. The CRTC1/3-MAML2 fusions are the only accepted diagnostic markers for MECa. This trans-location may also be detected in cutaneous hidroadenomas that have histologic similarities to MECa [46].

4.3. Prognostic Markers

Earlier, several studies have revealed that a favorable prognosis is associated with CRTC1/3-MAML2 gene rearrangements in younger people, and with a tumor of low to intermediate grade [38,47,48,49,50]. Recently, studies have shown that these mutations are not independent prognostic markers and are not related to prognosis or tumor grade. This can be explained by several reasons. First, some tumors are not diagnosed as MECa due to strict diagnostic criteria. Second, the prevalence of the translocation in MECa varies based on stringency of histologic diagnosis. Last, previous studies likely did not account for the confounding effect of grade on survival [51,52,53,54].
Purkinje cell protein (PCP) 4/peptide (PEP) 19 is a calmodulin-binding antiapoptotic peptide. Yoshimura et al. found that PCP4/PEP19 expression was related to a better prognosis, while HER2 expression was associated with a worse prognosis [55].

4.4. Treatment

Treatment of advanced salivary gland tumors remains limited to systemic chemotherapy, especially in the absence of molecular targets. Given the low response and survival rates with conventional systems, it becomes even more important to identify potential targets. Although our study does not provide any information on treatment outcomes, identifying and describing potential GAs can be of help in the future to identify potential targeted agents [56].
The mainstay of treatment of MECa, similar to any other salivary gland tumor, is surgical resection with disease-free margins [57,58,59]. Better regional control was achieved with combined chemoradiotherapy, but no survival difference was found compared to patients receiving radiotherapy alone [60]. As a matter of fact, systemic therapies should be used as palliative treatment for cancer-related symptom relief, or in rapid progression of the disease [61,62,63].

Standard Therapy

i.
Surgery
The most critical factor in determining the prolonged outcome and disease-specific survival is locoregional disease control [64]. To enhance locoregional control, postoperative radiotherapy is recommended, and it is typically reserved for cancers with high-risk characteristics. High-risk characteristics are defined by close or positive surgical margins, nodal metastases, extracapsular spread (ECS), perineural invasion, lymph vascular invasion, advanced tumor (T) stage, and high-grade histopathology [65,66]. In cases of deep lobe and recurrent cancers, radiation therapy may also be utilized. Doses greater than 60 Gy are necessary to achieve local tumor control. Higher doses, along with chemotherapy or alone, may be used to treat medical or technically unresectable tumors [67].
To our knowledge, there have only been two retrospective studies solely observing MECa, one describing the epidemiology and the other dealing with prognostic factors [68,69]. Most salivary gland tumor studies have been on a small series of patients with different salivary gland tumors treated uniformly with single agent chemotherapy or a combination of agents.
ii.
Chemotherapy
Airoldi et al. evaluated cisplatin plus vinorelbine (VNB) in 16 patients with recurrent metastatic salivary gland tumors; only one had MECa, vs. VNB alone in 20 patients. The combination of cisplatin plus VNB was found to be superior to VNB, but a prognosis related to the sole MECa tumor could not be singled out [70]. Gilbert et al. conducted a phase II evaluation of single-agent paclitaxel 200 mg/m2 every 3 weeks, which resulted in eight partial responses among 31 patients with mucoepidermoid or adenocarcinoma histologic subtypes, giving a response rate of 26% with a 95% confidence interval of 10% to 41% [71]. The last chemotherapeutic regimen which was also found to be useful consisted of docetaxel. Raguse et al. determined the role of docetaxel in four patients with high-grade mucoepidermoid cancer of the major salivary glands. After six cycles, complete remission was noticed in two patients, and partial remission was seen in the other two patients [72].
iii.
Monoclonal antibody
Many studies suggest amplification/overexpression of HER2/neu in mucoepidermoid carcinomas [73,74,75,76]. HER2 positivity found by IHC has been reported in 5.5% of MECs [77]. In our study, HER2 amplification was found in 5/9% of patients. Haddad et al. assessed the use of Herceptin (trastuzumab) on 14 patients with salivary gland tumors (SGC), three of which had MECa tumors with overexpressed HER2/neu in their salivary gland tumors. A partial response was seen only in one out of three patients, and lasted more than 2 years [14].
A case report by Gazola et al. revealed how a patient who was refractory to trastuzumab, ado-trastuzumab emtansine and tucatinib actually achieved excellent clinical benefit from Fam-trastuzumab deruxtecan [78]. Tsurutani et al. assessed the safety and tolerability of trastuzumab deruxtecan in pretreated, Her2-expressing non-breast/non-gastric or Her2-mutatnt solid tumors. Out of 60 patients, 8 patients had salivary gland tumors. Unfortunately, the results were not broken down into distinct tumor types. However, it was reported that tumor types including HER2-expressing or HER2-amplified salivary gland cancer, biliary tract cancer, and endometrial cancer, and HER2-mutant nonamplified breast cancer had a median progression-free survival of 11.0 months (95% CI, 2.8-Not evaluated) [79]. Currently, there are 8 clinical trials assessing trastuzumab deruxtecan or trastuzumab emtansine in salivary gland tumors.
The National Cancer Institute—Molecular Analysis for Therapy Choice (NCI-MATCH) assessed the efficacy of ado-trastuzumab emtansine (T-DM1) in HER2-amplified histologies in MECa. A partial response was observed in a patient with MECa [80]. Similarly, a case report of an elderly male with HER-2 Neu-overexpressing metastatic MECa demonstrated a prompt and sustained disease response to targeted therapies directed against HER-2 Neu, with a long survival interrupted by hepatoxicity to TDM-1 treatment [81].
iv.
Targeted therapies
1.
Sorafenib
It is known that MECa has high angiogenic activity with increased expression of vascular endothelial growth factor (VEGF) and angiopietin-2 (ANG2) [82,83,84]. In a phase II trial with recurrent and malignant salivary gland tumors, Locati et al. reported that sorafenib demonstrated a rapid decrease in disease progression in two patients with MECa [85].
2.
Nintedanib
In a single-arm, phase II trial carried out by Kim et al., 20 patients with SGT were enrolled, of which two patients (10%) had MECa tumors [86]. Results were not segregated by histological subtype, but 75% of patients had stable disease.
3.
Lapatinib
According to Lujan et al., the EGFR pathway is activated in high-grade MECs with aggressive behavior [87]. However, our report revealed very small numbers. Inhibiting both EGFR and receptor ERBB2 receptors, lapatinib was found to have cytostatic effect. In a phase II study performed by Agulnik et al., no objective response was noticed in the two MECa patients; however, stable disease (>6 months) was observed in 36% of all the eligible patients, n = 62 [12].
4.
Vorinostat
Pouloudi et al. were the first to assess immunohistochemically the expression of Hdi in SGC [88]. Wagner et al. have recently assessed acetyl-histone H3 expression (AH3) and Ki67 index in tissue microarrays (TMAs) of 84 cases of SGC [89]. Ahn et al. assessed the inhibition of histone deacetylase (HD)-7 expression in apicidin-treated MEC cell lines, providing evidence that HD-7 downregulation inhibits cell proliferation and induces autophagy in MEC cells [90]. Wagner et al. also investigated the benefits of Hdi and NFκB combined inhibition in MECa cell lines with the administration of Vorinostat and Emetine, respectively [89]. Vorinostat failed in significantly reducing the total number of tumor cells. Alone, Vorinostat efficiently disrupted the population of cancer stem cells. However, when combined with Emetine, Vorinostat provided an effective regimen for managing MECa [89].
5.
ANA-12
ANA-12 is a tyrosine receptor kinase B (TrkB) inhibitor. The brain-derived neutropenic factor (BDNF) is a growth factor that binds to TrkB and activates downstream pathways such asPI3K/Akt, which has a crucial role in tumorigenesis [91]. Perineural invasion in high-grade MECa, another negative prognostic marker, is associated with expression of BDNF and TrkB [92,93]. In vitro studies have shown that inhibiting TrkB decreases invasion and delays migration of MECa cells. Combining ANA-12 with cisplatin, however, caused the recovery and re-accumulation of cancer stem cells [92,93].
v.
Immunotherapy
TMB is a measure of neoantigen burden and predicts responsiveness to immune checkpoint therapy [94]. Similarly, PD-L1 expression estimated by immunohistochemistry assays can also predict immunotherapy responsiveness [95]. The multi-cohort phase 2 KEYNOTE-158 study established the role of pembrolizumab in patients with TMB > 10, resulting in the FDA issuing a blanket approval in this setting, regardless of the tumor type [96]. Upon analysis of a cohort of 109 advanced salivary gland carcinomas from the KEYNOTE-158 trial, pembrolizumab had an objective response rate of 4.6% in the overall population and 10.7% in the PD-L1 positive (CPS ≥ 1) cohort. Overall survival was 21 months and progression-free survival was 4 months [97]. KEYNOTE-028 was a phase 1 trial of 26 advanced salivary gland tumors with PD-L1 ≥ 1%. The objective response rate was 12%, and median duration of response was 4 months [98]. While trials specifically studying the role of checkpoint inhibitors in MECa are lacking [64], individual case reports have demonstrated durable clinical responses with checkpoint therapy [99]. Our report highlights that in some patients with advanced or refractory MECa having TMB > 10 or PD-L1 positivity, immune checkpoint therapy may be a viable option.
vi.
Novel studies
CRTC1-MAML2 positive cells were sensitive to epidermal growth factor receptor (EGFR) tyrosine kinase inhibition pre-clinically, suggesting a potential role for such drugs [100]. ERBB2 amplifications, though infrequent, may be amenable to Her-2 targeted therapy [101].
Last, Wein et al. reported that the combination of the notch inhibitor, dnMAML1 and the γ-secretase inhibitor, and the EGFR inhibitor Erlotinib enhanced MECa suppression as compared to individual treatment [102].

5. Synopsis

Treatment requires a multidisciplinary approach, and surgery to negative margins is a mainstay, supplemented by radiation for better locoregional control and chemotherapy, usually in the palliative recurrent or metastatic setting. Currently, surgical management and adjuvant radiotherapy are the best options for achieving disease control because conventional chemotherapies are ineffective against this disease due to resistance [103,104,105,106].
The high recurrence rate seen in most studies suggests that initial surgery should be radical with sufficient normal tissue margins [107]. Preservation of the facial nerve should be suggested only when the parotid tumor is small and is situated far away from the nerve. Otherwise, in cases of extensive tumor invasion, total parotidectomy with partial resection of the facial nerve is preferred [107].
Lately, there has been a huge emphasis on targeted therapy. Ross et al. conducted a comprehensive analysis of genomic profiles of metastatic and relapsed salivary gland carcinomas [108]. The MECa in the current study featured GA that were most similar to ductal adenocarcinomas and adenocarcinomas not otherwise specified including similar frequencies of alterations in ERBB2, PIK3CA, BRAF and TP53 (all differences not significant) [108].
In the literature, some cases of response to immune checkpoint inhibitors exist. Despite having no PD-L1 expression, two cases of metastatic high-grade MECa with prolonged response to immune checkpoint inhibitor pembrolizumab were reported by Pharaon et al. [99]. This shows the necessity of conducing further genomic profiling to unravel the connection between immunotherapy and the tumor microenvironment.

6. Conclusions

As rare and highly malignant tumors, SGC have a few proven target therapeutics. As mentioned above, several predictive molecular markers are being unraveled. Under the current treatment guidelines, most unresectable tumors become resistant to treatment within a short period of time. Our data suggest that MECa is a heterogeneous disease at the genomic level. Closer examination of the genomic profile also suggests that many of the MECa samples possess at least one target gene with direct or related targeted inhibitors currently available. Thus, it is important to translate knowledge obtained from genomic analysis of SGC samples into clinical cancer sequencing, combined with precision oncology clinical trials.

Author Contributions

J.R., D.Z. and A.S. contributed to conception and design of the study. R.H. and J.R. organized the database. R.H. performed the statistical analysis. M.B.Z. wrote the first draft of the manuscript. D.Z. and P.A.K. wrote sections of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The original contributions presented in the study are included in the article, further inquiries can be directed to the corresponding author/s.

Conflicts of Interest

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

Abbreviations

ANG-2Angiopietin-2
ABLAbelson
BDNFbrain-derived neutropenic factor
CREBCAMP Response Element-Binding Protein
CRTC1CREB Regulated Transcription Coactivator 1
EGFRepidermal growth factor receptor
erbB2receptor tyrosine-protein kinase
HNSCCsquamous cell carcinoma of the head and neck
FGFRfibroblast growth factor receptor
FISHFluorescent in situ hybridization
MECaMucoepidermoid carcinoma
NCI-MATCHNational Cancer Institute—Molecular Analysis for Therapy Choice
PDGFRplatelet-derived growth factor receptor
T-DM1ado-trastuzumab emtansine
TrkBtyrosine receptor kinase B
VEGFVascular endothelial growth factor
WHOWorld Health Organization

References

  1. Barnes, L.; Eveson, J.W.; Sidransky, D.; Reichart, P. Pathology and Genetics of Head and Neck Tumours; IARC: Lyon, France, 2005; Volume 9. [Google Scholar]
  2. Wahlberg, P.; Anderson, H.; Biörklund, A.; Möller, T.; Perfekt, R. Carcinoma of the parotid and submandibular glands—A study of survival in 2465 patients. Oral Oncol. 2002, 38, 706–713. [Google Scholar] [CrossRef] [PubMed]
  3. Guzzo, M.; Locati, L.D.; Prott, F.J.; Gatta, G.; McGurk, M.; Licitra, L. Major and minor salivary gland tumors. Crit. Rev. Oncol. Hematol. 2010, 74, 134–148. [Google Scholar] [CrossRef] [PubMed]
  4. Holst, V.A.; Marshall, C.E.; Moskaluk, C.A.; Frierson, H., Jr. KIT protein expression and analysis of c-kit gene mutation in adenoid cystic carcinoma. Mod. Pathol. Off. J. U.S. Can. Acad. Pathol. Inc. 1999, 12, 956–960. [Google Scholar]
  5. Vered, M.; Braunstein, E.; Buchner, A. Immunohistochemical study of epidermal growth factor receptor in adenoid cystic carcinoma of salivary gland origin. Head Neck J. Sci. Spec. Head Neck 2002, 24, 632–636. [Google Scholar] [CrossRef]
  6. Glisson, B.; Colevas, A.D.; Haddad, R.; Krane, J.; El-Naggar, A.; Kies, M.; Costello, R.; Summey, C.; Arquette, M.; Langer, C. HER2 expression in salivary gland carcinomas: Dependence on histological subtype. Clin. Cancer Res. 2004, 10, 944–946. [Google Scholar] [CrossRef]
  7. Jaehne, M.; Roeser, K.; Jaekel, T.; Schepers, J.D.; Albert, N.; Löning, T. Clinical and immunohistologic typing of salivary duct carcinoma: A report of 50 cases. Cancer 2005, 103, 2526–2533. [Google Scholar] [CrossRef]
  8. Nasser, S.M.; Faquin, W.C.; Dayal, Y. Expression of androgen, estrogen, and progesterone receptors in salivary gland tumors: Frequent expression of androgen receptor in a subset of malignant salivary gland tumors. Am. J. Clin. Pathol. 2003, 119, 801–806. [Google Scholar] [CrossRef]
  9. Nardi, V.; Sadow, P.M.; Juric, D.; Zhao, D.; Cosper, A.K.; Bergethon, K.; Scialabba, V.L.; Batten, J.M.; Borger, D.R.; Iafrate, A.J. Detection of Novel Actionable Genetic Changes in Salivary Duct Carcinoma Helps Direct Patient TreatmentMolecular Characterization of Salivary Duct Carcinoma. Clin. Cancer Res. 2013, 19, 480–490. [Google Scholar] [CrossRef]
  10. Pfeffer, M.R.; Talmi, Y.; Catane, R.; Symon, Z.; Yosepovitch, A.; Levitt, M. A phase II study of Imatinib for advanced adenoid cystic carcinoma of head and neck salivary glands. Oral Oncol. 2007, 43, 33–36. [Google Scholar] [CrossRef]
  11. Jakob, J.A.; Kies, M.S.; Glisson, B.S.; Kupferman, M.E.; Liu, D.D.; Lee, J.J.; El-Naggar, A.K.; Gonzalez–Angulo, A.M.; Blumenschein, G.R., Jr. Phase II study of gefitinib in patients with advanced salivary gland cancers. Head Neck 2015, 37, 644–649. [Google Scholar] [CrossRef]
  12. Agulnik, M.; Cohen, E.W.; Cohen, R.B.; Chen, E.X.; Vokes, E.E.; Hotte, S.J.; Winquist, E.; Laurie, S.; Hayes, D.N.; Dancey, J.E. Phase II study of lapatinib in recurrent or metastatic epidermal growth factor receptor and/or erbB2 expressing adenoid cystic carcinoma and non-adenoid cystic carcinoma malignant tumors of the salivary glands. J. Clin. Oncol. 2007, 25, 3978–3984. [Google Scholar] [CrossRef]
  13. Locati, L.; Bossi, P.; Perrone, F.; Potepan, P.; Crippa, F.; Mariani, L.; Casieri, P.; Orsenigo, M.; Losa, M.; Bergamini, C. Cetuximab in recurrent and/or metastatic salivary gland carcinomas: A phase II study. Oral Oncol. 2009, 45, 574–578. [Google Scholar] [CrossRef]
  14. Haddad, R.; Colevas, A.D.; Krane, J.F.; Cooper, D.; Glisson, B.; Amrein, P.C.; Weeks, L.; Costello, R.; Posner, M. Herceptin in patients with advanced or metastatic salivary gland carcinomas. A phase II study. Oral Oncol. 2003, 39, 724–727. [Google Scholar] [CrossRef] [PubMed]
  15. Falchook, G.S.; Lippman, S.M.; Bastida, C.C.; Kurzrock, R. Human epidermal receptor 2–amplified salivary duct carcinoma: Regression with dual human epidermal receptor 2 inhibition and anti–vascular endothelial growth factor combination treatment. Head Neck 2014, 36, E25–E27. [Google Scholar] [CrossRef]
  16. Piha-Paul, S.A.; Cohen, P.R.; Kurzrock, R. Salivary duct carcinoma: Targeting the phosphatidylinositol 3-kinase pathway by blocking mammalian target of rapamycin with temsirolimus. J. Clin. Oncol. 2011, 29, e727–e730. [Google Scholar] [CrossRef] [PubMed]
  17. Frampton, G.M.; Fichtenholtz, A.; Otto, G.A.; Wang, K.; Downing, S.R.; He, J.; Schnall-Levin, M.; White, J.; Sanford, E.M.; An, P. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat. Biotechnol. 2013, 31, 1023–1031. [Google Scholar] [CrossRef] [PubMed]
  18. Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34. [Google Scholar] [CrossRef]
  19. Trabucco, S.E.; Gowen, K.; Maund, S.L.; Sanford, E.; Fabrizio, D.A.; Hall, M.J.; Yakirevich, E.; Gregg, J.P.; Stephens, P.J.; Frampton, G.M. A novel next-generation sequencing approach to detecting microsatellite instability and pan-tumor characterization of 1000 microsatellite instability–high cases in 67,000 patient samples. J. Mol. Diagn. 2019, 21, 1053–1066. [Google Scholar] [CrossRef]
  20. Toper, M.H.; Sarioglu, S. Molecular pathology of salivary gland neoplasms: Diagnostic, prognostic, and predictive perspective. Adv. Anat. Pathol. 2021, 28, 81–93. [Google Scholar] [CrossRef]
  21. El-Naggar, A.K.; Chan, J.K.; Grandis, J.R. WHO Classification of Head and Neck Tumours. 2017. Available online: https://publications.iarc.fr/Book-And-Report-Series/Who-Classification-Of-Tumours/WHO-Classification-Of-Head-And-Neck-Tumours-2017 (accessed on 25 December 2022).
  22. Shafique, K.; Zhang, P.J.; Montone, K.T.; Song, S.; Livolsi, V.A.; Baloch, Z. Pathologic grading of mucoepidermoid carcinomas of the salivary gland and its effect on clinicopathologic follow-up: An institutional experience. Hum. Pathol. 2020, 98, 89–97. [Google Scholar] [CrossRef]
  23. 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]
  24. Kato, S.; Elkin, S.K.; Schwaederle, M.; Tomson, B.N.; Helsten, T.; Carter, J.L.; Kurzrock, R. Genomic landscape of salivary gland tumors. Oncotarget 2015, 6, 25631. [Google Scholar] [CrossRef] [PubMed]
  25. Olivier, T. mutations in human cancers: Origins, consequences, and clinical use. Cold Spring Harb. Perspect. Biol 2010, 2, a001008. [Google Scholar] [CrossRef] [PubMed]
  26. Kang, H.; Tan, M.; Bishop, J.A.; Jones, S.; Sausen, M.; Ha, P.K.; Agrawal, N. Whole-Exome Sequencing of Salivary Gland Mucoepidermoid CarcinomaWhole-Exome Sequencing of Salivary Mucoepidermoid Carcinoma. Clin. Cancer Res. 2017, 23, 283–288. [Google Scholar] [CrossRef]
  27. Yan, K.; Yesensky, J.; Hasina, R.; Agrawal, N. Genomics of mucoepidermoid and adenoid cystic carcinomas. Laryngoscope Investig. Otolaryngol. 2018, 3, 56–61. [Google Scholar] [CrossRef]
  28. Hong, D.S.; Fakih, M.G.; Strickler, J.H.; Desai, J.; Durm, G.A.; Shapiro, G.I.; Falchook, G.S.; Price, T.J.; Sacher, A.; Denlinger, C.S. KRASG12C inhibition with sotorasib in advanced solid tumors. N. Engl. J. Med. 2020, 383, 1207–1217. [Google Scholar] [CrossRef]
  29. Loong, H.H.-F.; Du, N.; Cheng, C.; Lin, H.; Guo, J.; Lin, G.; Li, M.; Jiang, T.; Shi, Z.; Cui, Y. KRAS G12C mutations in Asia: A landscape analysis of 11,951 Chinese tumor samples. Transl. Lung Cancer Res. 2020, 9, 1759. [Google Scholar] [CrossRef] [PubMed]
  30. Sheth, H.; Kumar, P.; Shreenivas, A.; Sambath, J.; Pragya, R.; Madre, C.; Athikari, N.; Khandare, H.; Peshattiwar, V.; Datar, R. Excellent response with alpelisib and bicalutamide for advanced salivary duct carcinoma with PIK3CA mutation and high androgen receptor expression—A case report. JCO Precis. Oncol. 2021, 5. [Google Scholar] [CrossRef]
  31. Kaye, F.J. Mutation-associated fusion cancer genes in solid tumors. Mol. Cancer Ther. 2009, 8, 1399–1408. [Google Scholar] [CrossRef]
  32. Stenman, G. Fusion oncogenes and tumor type specificity—Insights from salivary gland tumors. In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2005; Volume 15, pp. 224–235. [Google Scholar]
  33. O’Neill, I.D. t (11; 19) translocation and CRTC1-MAML2 fusion oncogene in mucoepidermoid carcinoma. Oral Oncol. 2009, 45, 2–9. [Google Scholar] [CrossRef]
  34. Conkright, M.D.; Canettieri, G.; Screaton, R.; Guzman, E.; Miraglia, L.; Hogenesch, J.B.; Montminy, M. TORCs: Transducers of regulated CREB activity. Mol. Cell 2003, 12, 413–423. [Google Scholar] [CrossRef]
  35. Wu, L.; Liu, J.; Gao, P.; Nakamura, M.; Cao, Y.; Shen, H.; Griffin, J.D. Transforming activity of MECT1-MAML2 fusion oncoprotein is mediated by constitutive CREB activation. EMBO J. 2005, 24, 2391–2402. [Google Scholar] [CrossRef]
  36. Tonon, G.; Modi, S.; Wu, L.; Kubo, A.; Coxon, A.B.; Komiya, T.; O’Neil, K.; Stover, K.; El-Naggar, A.; Griffin, J.D. 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. [Google Scholar] [CrossRef]
  37. Shinomiya, H.; Ito, Y.; Kubo, M.; Yonezawa, K.; Otsuki, N.; Iwae, S.; Inagaki, H.; Nibu, K.-I. Expression of amphiregulin in mucoepidermoid carcinoma of the major salivary glands: A molecular and clinicopathological study. Hum. Pathol. 2016, 57, 37–44. [Google Scholar] [CrossRef]
  38. Okumura, Y.; Miyabe, S.; Nakayama, T.; Fujiyoshi, Y.; Hattori, H.; Shimozato, K.; Inagaki, H. Impact of CRTC1/3–MAML2 fusions on histological classification and prognosis of mucoepidermoid carcinoma. Histopathology 2011, 59, 90–97. [Google Scholar] [CrossRef] [PubMed]
  39. Nakayama, T.; Miyabe, S.; Okabe, M.; Sakuma, H.; Ijichi, K.; Hasegawa, Y.; Nagatsuka, H.; Shimozato, K.; Inagaki, H. Clinicopathological significance of the CRTC3–MAML2 fusion transcript in mucoepidermoid carcinoma. Mod. Pathol. 2009, 22, 1575–1581. [Google Scholar] [CrossRef] [PubMed]
  40. Möller, E.; Stenman, G.; Mandahl, N.; Hamberg, H.; Mölne, L.; Van Den Oord, J.; Brosjö, O.; Mertens, F.; Panagopoulos, I. POU5F1, encoding a key regulator of stem cell pluripotency, is fused to EWSR1 in hidradenoma of the skin and mucoepidermoid carcinoma of the salivary glands. J. Pathol. A J. Pathol. Soc. Great Br. Irel. 2008, 215, 78–86. [Google Scholar] [CrossRef] [PubMed]
  41. Anzick, S.L.; Chen, W.D.; Park, Y.; Meltzer, P.; Bell, D.; El-Naggar, A.K.; Kaye, F.J. Unfavorable prognosis of CRTC1-MAML2 positive mucoepidermoid tumors with CDKN2A deletions. Genes Chromosomes Cancer 2010, 49, 59–69. [Google Scholar] [CrossRef]
  42. Jee, K.J.; Persson, M.; Heikinheimo, K.; Passador-Santos, F.; Aro, K.; Knuutila, S.; Odell, E.W.; Mäkitie, A.; Sundelin, K.; Stenman, G. Genomic profiles and CRTC1–MAML2 fusion distinguish different subtypes of mucoepidermoid carcinoma. Mod. Pathol. 2013, 26, 213–222. [Google Scholar] [CrossRef]
  43. García, J.J.; Hunt, J.L.; Weinreb, I.; McHugh, J.B.; Barnes, E.L.; Cieply, K.; Dacic, S.; Seethala, R.R. Fluorescence in situ hybridization for detection of MAML2 rearrangements in oncocytic mucoepidermoid carcinomas: Utility as a diagnostic test. Hum. Pathol. 2011, 42, 2001–2009. [Google Scholar] [CrossRef]
  44. Bishop, J.A.; Cowan, M.L.; Shum, C.H.; Westra, W.H. MAML2 rearrangements in variant forms of mucoepidermoid carcinoma: Ancillary diagnostic testing for the ciliated and warthin-like variants. Am. J. Surg. Pathol. 2018, 42, 130. [Google Scholar] [CrossRef]
  45. Tajima, S.; Namiki, I.; Koda, K. A clear cell variant of mucoepidermoid carcinoma harboring CRTC1-MAML2 fusion gene found in buccal mucosa: Report of a case showing a large clear cell component and lacking typical epidermoid cells and intermediate cells. Med. Mol. Morphol. 2017, 50, 117–121. [Google Scholar] [CrossRef]
  46. Kuma, Y.; Yamada, Y.; Yamamoto, H.; Kohashi, K.; Ito, T.; Furue, M.; Oda, Y. A novel fusion gene CRTC3-MAML2 in hidradenoma: Histopathological significance. Hum. Pathol. 2017, 70, 55–61. [Google Scholar] [CrossRef]
  47. Behboudi, A.; Enlund, F.; Winnes, M.; Andrén, Y.; Nordkvist, A.; Leivo, I.; Flaberg, E.; Szekely, L.; Mäkitie, A.; Grenman, R. Molecular classification of mucoepidermoid carcinomas—Prognostic significance of the MECT1–MAML2 fusion oncogene. Genes Chromosomes Cancer 2006, 45, 470–481. [Google Scholar] [CrossRef] [PubMed]
  48. Okabe, M.; Miyabe, S.; Nagatsuka, H.; Terada, A.; Hanai, N.; Yokoi, M.; Shimozato, K.; Eimoto, T.; Nakamura, S.; Nagai, N. MECT1-MAML2 fusion transcript defines a favorable subset of mucoepidermoid carcinoma. Clin. Cancer Res. 2006, 12, 3902–3907. [Google Scholar] [CrossRef] [PubMed]
  49. Luk, P.P.; Wykes, J.; Selinger, C.I.; Ekmejian, R.; Tay, J.; Gao, K.; Eviston, T.J.; Lum, T.; O’Toole, S.A.; Clark, J.R. Diagnostic and prognostic utility of Mastermind-like 2 (MAML2) gene rearrangement detection by fluorescent in situ hybridization (FISH) in mucoepidermoid carcinoma of the salivary glands. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2016, 121, 530–541. [Google Scholar] [CrossRef]
  50. Tirado, Y.; Williams, M.D.; Hanna, E.Y.; Kaye, F.J.; Batsakis, J.G.; El-Naggar, A.K. CRTC1/MAML2 fusion transcript in high grade mucoepidermoid carcinomas of salivary and thyroid glands and Warthin’s tumors: Implications for histogenesis and biologic behavior. Genes Chromosomes Cancer 2007, 46, 708–715. [Google Scholar] [CrossRef]
  51. Birkeland, A.C.; Foltin, S.K.; Michmerhuizen, N.L.; Hoesli, R.C.; Rosko, A.J.; Byrd, S.; Yanik, M.; Nor, J.E.; Bradford, C.R.; Prince, M.E. Correlation of Crtc1/3-Maml2 fusion status, grade and survival in mucoepidermoid carcinoma. Oral Oncol. 2017, 68, 5–8. [Google Scholar] [CrossRef]
  52. Saade, R.E.; 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. [Google Scholar] [CrossRef] [PubMed]
  53. Chiosea, S.I.; Dacic, S.; Nikiforova, M.N.; Seethala, R.R. Prospective testing of mucoepidermoid carcinoma for the MAML2 translocation: Clinical implications. Laryngoscope 2012, 122, 1690–1694. [Google Scholar] [CrossRef]
  54. Cipriani, N.A.; Lusardi, J.J.; McElherne, J.; Pearson, A.T.; Olivas, A.D.; Fitzpatrick, C.; Lingen, M.W.; Blair, E.A. Mucoepidermoid carcinoma: A comparison of histologic grading systems and relationship to MAML2 rearrangement and prognosis. Am. J. Surg. Pathol. 2019, 43, 885. [Google Scholar] [CrossRef]
  55. Yoshimura, T.; Higashi, S.; Yamada, S.; Noguchi, H.; Nomoto, M.; Suzuki, H.; Ishida, T.; Takayama, H.; Hirano, Y.; Yamashita, M. PCP4/PEP19 and HER2 Are Novel Prognostic Markers in Mucoepidermoid Carcinoma of the Salivary Gland. Cancers 2021, 14, 54. [Google Scholar] [CrossRef] [PubMed]
  56. Di Villeneuve, L.; Souza, I.L.; Tolentino, F.D.S.; Ferrarotto, R.; Schvartsman, G. Salivary gland carcinoma: Novel targets to overcome treatment resistance in advanced disease. Front. Oncol. 2020, 10, 580141. [Google Scholar] [CrossRef]
  57. Yih, W.-Y.; Kratochvil, F.J.; Stewart, J.C. Intraoral minor salivary gland neoplasms: Review of 213 cases. J. Oral Maxillofac. Surg. 2005, 63, 805–810. [Google Scholar] [CrossRef] [PubMed]
  58. Mendenhall, W.M.; Morris, C.G.; Amdur, R.J.; Werning, J.W.; Villaret, D.B. Radiotherapy alone or combined with surgery for salivary gland carcinoma. Cancer 2005, 103, 2544–2550. [Google Scholar] [CrossRef] [PubMed]
  59. Roh, J.L.; Choi, S.H.; Lee, S.W.; Cho, K.J.; Nam, S.Y.; Kim, S.Y. Carcinomas arising in the submandibular gland: High propensity for systemic failure. J. Surg. Oncol. 2008, 97, 533–537. [Google Scholar] [CrossRef]
  60. de Souza, L.B.; de Oliveira, L.C.; Nonaka, C.F.W.; Lopes, M.L.D.d.S.; Pinto, L.P.; Queiroz, L.M.G. Immunoexpression of GLUT-1 and angiogenic index in pleomorphic adenomas, adenoid cystic carcinomas, and mucoepidermoid carcinomas of the salivary glands. Eur. Arch. Otorhinolaryngol. 2017, 274, 2549–2556. [Google Scholar] [CrossRef]
  61. Lagha, A.; Chraiet, N.; Ayadi, M.; Krimi, S.; Allani, B.; Rifi, H.; Raies, H.; Mezlini, A. RETRACTED: Systemic therapy in the management of metastatic or advanced salivary gland cancers. Oral Oncol. 2012, 48, 948–957. [Google Scholar] [CrossRef]
  62. Limaye, S.A.; Posner, M.R.; Krane, J.F.; Fonfria, M.; Lorch, J.H.; Dillon, D.A.; Shreenivas, A.V.; Tishler, R.B.; Haddad, R.I. Trastuzumab for the treatment of salivary duct carcinoma. Oncologist 2013, 18, 294–300. [Google Scholar] [CrossRef]
  63. Lewis, A.G.; Tong, T.; Maghami, E. Diagnosis and management of malignant salivary gland tumors of the parotid gland. Otolaryngol. Clin. North Am. 2016, 49, 343–380. [Google Scholar] [CrossRef]
  64. Sama, S.; Komiya, T.; Guddati, A.K. Advances in the Treatment of Mucoepidermoid Carcinoma. World J. Oncol. 2022, 13, 1–7. [Google Scholar] [CrossRef]
  65. Young, A.; Okuyemi, O.T. Malignant Salivary Gland Tumors. In StatPearls; StatPearls Publishing: Tampa, FL, USA, 2021. [Google Scholar]
  66. Ries, F.; Klastersky, J. Nephrotoxicity induced by cancer chemotherapy with special emphasis on cisplatin toxicity. Am. J. Kidney Dis. 1986, 8, 368–379. [Google Scholar] [CrossRef] [PubMed]
  67. Pederson, A.W.; Salama, J.K.; Haraf, D.J.; Witt, M.E.; Stenson, K.M.; Portugal, L.; Seiwert, T.; Villaflor, V.M.; Cohen, E.E.; Vokes, E.E. Adjuvant chemoradiotherapy for locoregionally advanced and high-risk salivary gland malignancies. Head Neck Oncol. 2011, 3, 31. [Google Scholar] [CrossRef] [PubMed]
  68. Guevara-Canales, J.O.; Morales-Vadillo, R.; Guzmán-Arias, G.; Cava-Vergiú, C.E.; Guerra-Miller, H.; Montes-Gil, J.E. Mucoepidermoid carcinoma of the salivary glands. A retrospective study of 51 cases and review of the literatura. Acta Odontológica Latinoam. 2016, 29, 230–238. [Google Scholar]
  69. Terauchi, M.; Michi, Y.; Hirai, H.; Sugiyama, K.; Wada, A.; Harada, H.; Yoda, T. Prognostic factors in mucoepidermoid carcinoma of the minor salivary glands: A single-center retrospective study. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2021, 131, 209–216. [Google Scholar] [CrossRef]
  70. Airoldi, M.; Pedani, F.; Succo, G.; Gabriele, A.M.; Ragona, R.; Marchionatti, S.; Bumma, C. Phase II randomized trial comparing vinorelbine versus vinorelbine plus cisplatin in patients with recurrent salivary gland malignancies. Cancer 2001, 91, 541–547. [Google Scholar] [CrossRef]
  71. Gilbert, J.; Li, Y.; Pinto, H.A.; Jennings, T.; Kies, M.S.; Silverman, P.; Forastiere, A.A. Phase II trial of taxol in salivary gland malignancies (E1394): A trial of the Eastern Cooperative Oncology Group. Head Neck J. Sci. Spec. Head Neck 2006, 28, 197–204. [Google Scholar] [CrossRef]
  72. Raguse, J.-D.; Gath, H.J.; Bier, J.; Riess, H.; Oettle, H. Docetaxel (Taxotere) in recurrent high grade mucoepidermoid carcinoma of the major salivary glands. Oral Oncol. Extra 2004, 40, 5–7. [Google Scholar] [CrossRef]
  73. Nakano, T.; Yamamoto, H.; Hashimoto, K.; Yasumatsu, R.; Nakashima, T.; Oda, Y.; Komune, S. HER2 and EGFR Gene Copy Number Alterations Are Predominant in High-Grade Salivary Mucoepidermoid Carcinoma. Otolaryngol. Head Neck Surg. 2013, 149, P174. [Google Scholar] [CrossRef]
  74. Nguyen, L.H.; Black, M.J.; Hier, M.; Chauvin, P.; Rochon, L. HER2/neu and Ki-67 as prognostic indicators in mucoepidermoid carcinoma of salivary glands. J. Otolaryngol. 2003, 32. [Google Scholar] [CrossRef]
  75. Alotaibi, A.M.; Alqarni, M.A.; Alnobi, A.; Tarakji, B. Human epidermal growth factor receptor 2 (HER2/neu) in salivary gland carcinomas: A review of literature. J. Clin. Diagn. Res. 2015, 9, ZE04. [Google Scholar] [CrossRef] [PubMed]
  76. Kurzrock, R.; Bowles, D.; Kang, H.; Meric-Bernstam, F.; Hainsworth, J.; Spigel, D.; Bose, R.; Burris, H.; Sweeney, C.; Beattie, M. Targeted therapy for advanced salivary gland carcinoma based on molecular profiling: Results from MyPathway, a phase IIa multiple basket study. Ann. Oncol. 2020, 31, 412–421. [Google Scholar] [CrossRef] [PubMed]
  77. Egebjerg, K.; Harwood, C.D.; Woller, N.C.; Kristensen, C.A.; Mau-Sørensen, M. Her2 positivity in histological subtypes of salivary gland carcinoma: A systematic review and meta-analysis. Front. Oncol. 2021, 11, 2154. [Google Scholar] [CrossRef] [PubMed]
  78. Gazola, A.A.; Wong, D.; Chin, R.; Nikitas, J.; Kim, C.J.; Hart, S.D.; St John, M.; Chai-Ho, W. Excellent Response to Fam-Trastuzumab Deruxtecan for Human Epidermal Growth Factor Receptor 2–Positive Salivary Duct Carcinoma With CNS Metastasis: A Case Report. JCO Precis. Oncol. 2022, 6, e2200399. [Google Scholar] [CrossRef]
  79. Tsurutani, J.; Iwata, H.; Krop, I.; Jänne, P.A.; Takahashi, S.; Park, H.; Redfern, C.; Tamura, K.; Wise-Draper, T.M.; Saito, K. Targeting HER2 with Trastuzumab Deruxtecan: A Dose-Expansion, Phase I Study in Multiple Advanced Solid TumorsT-DXd in Advanced Solid Tumors. Cancer Discov. 2020, 10, 688–701. [Google Scholar] [CrossRef]
  80. Jhaveri, K.; Wang, X.; Makker, V.; Luoh, S.-W.; Mitchell, E.; Zwiebel, J.; Sharon, E.; Gray, R.; Li, S.; McShane, L. Ado-trastuzumab emtansine (T-DM1) in patients with HER2-amplified tumors excluding breast and gastric/gastroesophageal junction (GEJ) adenocarcinomas: Results from the NCI-MATCH trial (EAY131) subprotocol Q. Ann. Oncol. 2019, 30, 1821–1830. [Google Scholar] [CrossRef]
  81. Mohammed, T.; Mangeshkar, S.; Desai, A.; Hegde, U. HER-2 Neu gene: A valuable therapeutic target in metastatic mucoepidermoid carcinoma. J. Oncol. Pharm. Pract. 2021, 27, 1806–1809. [Google Scholar] [CrossRef]
  82. Cardoso, S.V.; Souza, K.C.N.; Faria, P.R.; Eisenberg, A.L.A.; Dias, F.L.; Loyola, A.M. Assessment of angiogenesis by CD105 antigen in epithelial salivary gland neoplasms with diverse metastatic behavior. BMC Cancer 2009, 9, 391. [Google Scholar] [CrossRef]
  83. Gleber-Netto, F.O.; Florêncio, T.N.G.; de Sousa, S.F.; Abreu, M.H.N.G.; Mendonça, E.F.; Aguiar, M.C.F. Angiogenesis and lymphangiogenesis in mucoepidermoid carcinoma of minor salivary glands. J. Oral Pathol. Med. 2012, 41, 603–609. [Google Scholar] [CrossRef]
  84. Demasi, A.P.; Silva, C.A.; Silva, A.D.; Furuse, C.; Soares, A.B.; Altemani, A.; Napimoga, M.H.; Araújo, V.C. Expression of the vascular endothelial growth factor and angiopoietins in mucoepidermoid carcinoma of salivary gland. Head Neck Pathol. 2012, 6, 10–15. [Google Scholar] [CrossRef]
  85. Locati, L.D.; Bossi, P.; Civelli, E.M.; Perrone, F.; Bergamini, C.; Cortelazzi, B.; Quattrone, P.; Imbimbo, M.; Mirabile, A.; Granata, R. Sorafenib in recurrent and/or metastatic salivary gland carcinomas (RMSGCs): An investigator-initiated phase II trial (NCT01703455). Am. Soc. Clin. Oncol. 2013, 31, 6020. [Google Scholar] [CrossRef]
  86. Kim, Y.; Lee, S.J.; Lee, J.Y.; Lee, S.H.; Sun, J.M.; Park, K.; An, H.J.; Cho, J.Y.; Kang, E.J.; Lee, H.Y. Clinical trial of nintedanib in patients with recurrent or metastatic salivary gland cancer of the head and neck: A multicenter phase 2 study (Korean Cancer Study Group HN14-01). Cancer 2017, 123, 1958–1964. [Google Scholar] [CrossRef] [PubMed]
  87. Lujan, B.; Hakim, S.; Moyano, S.; Nadal, A.; Caballero, M.; Diaz, A.; Valera, A.; Carrera, M.; Cardesa, A.; Alos, L. Activation of the EGFR/ERK pathway in high-grade mucoepidermoid carcinomas of the salivary glands. Br. J. Cancer 2010, 103, 510–516. [Google Scholar] [CrossRef] [PubMed]
  88. Pouloudi, D.; Manou, M.; Sarantis, P.; Tsoukalas, N.; Tsourouflis, G.; Dana, E.; Karamouzis, M.V.; Klijanienko, J.; Theocharis, S. Clinical Significance of Histone Deacetylase (HDAC)-1,-2,-4 and-6 Expression in Salivary Gland Tumors. Diagnostics 2021, 11, 517. [Google Scholar] [CrossRef]
  89. Wagner, V.P.; Martins, M.D.; Martins, M.A.; Almeida, L.O.; Warner, K.A.; Nör, J.E.; Squarize, C.H.; Castilho, R.M. Targeting histone deacetylase and NFκB signaling as a novel therapy for Mucoepidermoid Carcinomas. Sci. Rep. 2018, 8, 2065. [Google Scholar] [CrossRef]
  90. Ahn, M.Y.; Yoon, J.H. Histone deacetylase 7 silencing induces apoptosis and autophagy in salivary mucoepidermoid carcinoma cells. J. Oral Pathol. Med. 2017, 46, 276–283. [Google Scholar] [CrossRef]
  91. de Moraes, J.K.; Wagner, V.P.; Fonseca, F.P.; Vargas, P.A.; de Farias, C.B.; Roesler, R.; Martins, M.D. Uncovering the role of brain-derived neurotrophic factor/tyrosine kinase receptor B signaling in head and neck malignancies. J. Oral Pathol. Med. 2018, 47, 221–227. [Google Scholar] [CrossRef]
  92. McHugh, C.H.; Roberts, D.B.; El-Naggar, A.K.; Hanna, E.Y.; Garden, A.S.; Kies, M.S.; Weber, R.S.; Kupferman, M.E. Prognostic factors in mucoepidermoid carcinoma of the salivary glands. Cancer 2012, 118, 3928–3936. [Google Scholar] [CrossRef]
  93. Wagner, V.P.; Martins, M.D.; Amoura, E.; Zanella, V.G.; Roesler, R.; de Farias, C.B.; Bingle, C.D.; Vargas, P.A.; Bingle, L. TrkB-targeted therapy for mucoepidermoid carcinoma. Biomedicines 2020, 8, 531. [Google Scholar] [CrossRef]
  94. Klempner, S.J.; Fabrizio, D.; Bane, S.; Reinhart, M.; Peoples, T.; Ali, S.M.; Sokol, E.S.; Frampton, G.; Schrock, A.B.; Anhorn, R. Tumor mutational burden as a predictive biomarker for response to immune checkpoint inhibitors: A review of current evidence. Oncologist 2020, 25, e147–e159. [Google Scholar] [CrossRef]
  95. Doroshow, D.B.; Bhalla, S.; Beasley, M.B.; Sholl, L.M.; Kerr, K.M.; Gnjatic, S.; Wistuba, I.I.; Rimm, D.L.; Tsao, M.S.; Hirsch, F.R. PD-L1 as a biomarker of response to immune-checkpoint inhibitors. Nat. Rev. Clin. Oncol. 2021, 18, 345–362. [Google Scholar] [CrossRef] [PubMed]
  96. Marabelle, A.; Fakih, M.; Lopez, J.; Shah, M.; Shapira-Frommer, R.; Nakagawa, K.; Chung, H.C.; Kindler, H.L.; Lopez-Martin, J.A.; Miller, W.H., Jr. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: Prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020, 21, 1353–1365. [Google Scholar] [CrossRef] [PubMed]
  97. Even, C.; Delord, J.-P.; Price, K.A.; Nakagawa, K.; Oh, D.-Y.; Burge, M.; Chung, H.C.; Doi, T.; Fakih, M.; Takahashi, S. Evaluation of pembrolizumab monotherapy in patients with salivary gland carcinoma in the phase 2 KEYNOTE-158 study. Oral Oncol. 2021, 118, 9. [Google Scholar] [CrossRef]
  98. Cohen, R.B.; Delord, J.-P.; Doi, T.; Piha-Paul, S.A.; Liu, S.V.; Gilbert, J.; Algazi, A.P.; Damian, S.; Hong, R.-L.; Le Tourneau, C. Pembrolizumab for the treatment of advanced salivary gland carcinoma: Findings of the phase 1b KEYNOTE-028 study. Am. J. Clin. Oncol. 2018, 41, 1083. [Google Scholar] [CrossRef]
  99. Pharaon, R.R.; Gernon, T.; Chang, S.; Vora, N.; Villaflor, V.M.; Bell, D.; Afkhami, M.; Amini, A.; Sampath, S.; Kang, R.; et al. Prolonged response to checkpoint inhibitor therapy in two metastatic mucoepidermoid salivary gland carcinoma cases: A research report. Mol. Case Stud. 2022, 8, a006189. [Google Scholar]
  100. Chen, Z.; Chen, J.; Gu, Y.; Hu, C.; Li, J.; Lin, S.; Shen, H.; Cao, C.; Gao, R.; Li, J. 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. [Google Scholar] [CrossRef]
  101. De Block, K.; Vander Poorten, V.; Dormaar, T.; Nuyts, S.; Hauben, E.; Floris, G.; Deroose, C.M.; Schöffski, P.; Clement, P.M. Metastatic HER-2-positive salivary gland carcinoma treated with trastuzumab and a taxane: A series of six patients. Acta Clin. Belg. 2016, 71, 383–388. [Google Scholar] [CrossRef]
  102. Villa, J.C.; Chiu, D.; Brandes, A.H.; Escorcia, F.E.; Villa, C.H.; Maguire, W.F.; Hu, C.-J.; de Stanchina, E.; Simon, M.C.; Sisodia, S.S. Nontranscriptional role of Hif-1α in activation of γ-secretase and notch signaling in breast cancer. Cell Rep. 2014, 8, 1077–1092. [Google Scholar] [CrossRef]
  103. Kaplan, M.J.; Johns, M.E.; Cantrell, R.W. Chemotherapy for salivary gland cancer. Otolaryngol. Head Neck Surg. 1986, 95, 165–170. [Google Scholar] [CrossRef]
  104. Pires, F.R.; De Almeida, O.P.; De Araújo, 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]
  105. Grisanti, S.; Amoroso, V.; Buglione, M.; Rosati, A.; Gatta, R.; Pizzocaro, C.; Ferrari, V.D.; Marini, G. Cetuximab in the treatment of metastatic mucoepidermoid carcinoma of the salivary glands: A case report and review of literature. J. Med. Case Reports 2008, 2, 320. [Google Scholar] [CrossRef] [PubMed]
  106. Guzzo, M.; Andreola, S.; Sirizzotti, G.; Cantu, G. Mucoepidermoid carcinoma of the salivary glands: Clinicopathologic review of 108 patients treated at the National Cancer Institute of Milan. Ann. Surg. Oncol. 2002, 9, 688–695. [Google Scholar] [CrossRef] [PubMed]
  107. Yu, G.; Ma, D.; Sun, K.; Li, T.; Zhang, Y. Myoepithelial carcinoma of the salivary glands: Behavior and management. Chin. Med. J. 2003, 116, 163–165. [Google Scholar] [PubMed]
  108. Ross, J.; Gay, L.; Wang, K.; Vergilio, J.; Suh, J.; Ramkissoon, S.; Somerset, H.; Johnson, J.; Russell, J.; Ali, S. 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. [Google Scholar] [CrossRef] [PubMed]
Table 1. Clinical characteristics and genomic alterations in MECa.
Table 1. Clinical characteristics and genomic alterations in MECa.
Muco-Epidermoid Carcinoma
Number of cases118
Median age (range)64 (16–89+)
Gender (M/F)58%/42%
GA per tumor4.88
Cell Cycle Regulatory GA
TP5340.70%
CDKN2A52.50%
CDKN2B30.50%
CDK41.70%
CCND13.40%
RB13.40%
Chromosomal and Chromatin Related GA
TERT15.00%
ARID1A2.50%
xRAS-RAF Pathway GA
KRAS All5.10%
KRAS G12C2.50%
HRAS14.40%
BRAF1.70%
MTOR Pathway GA
PTEN7.60%
PIK3CA16.90%
NF14.20%
TSC21.00%
DNA Damage Response Associated GA
BRCA11.70%
BRCA25.90%
ATM4.20%
PALB20.80%
BAP118.60%
Receptor Tyrosine Kinase Targetable GA
ERBB2 (amp/SV)5.9%/0%
EGFR (amp/SV)0.8%/0%
FGFR15.10%
FGFR20%
RET0%
ETV6lNTRK3 fusion0%
MET0%
KIT0.80%
Transcription Factor Genomic Alterations
NFIB-MYB Fusion0%
ESR10%
AR0%
MYC1.70%
EWSR0%
Emerging Potentially Genomic Alterations
NOTCH14.20%
NOTCH27.60%
MTAP13.70%
(F1CDx only)(51 cases)
Immuno-Oncology Drug Biomarkers
MSI high frequency0%
(100 cases)
CD274 (PD-L1) amp0%
STK11 inactivating GA3.40%
MDM2 amp3.40%
Median TMB2.6
TMB > 10%16.90%
TMB > 20%12.70%
PD-L1 low expression (≤49%)38.40%
(26 cases)
PD-L1 high expression (>50%)4.20%
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bou Zerdan, M.; Kumar, P.A.; 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, 519. https://doi.org/10.3390/biomedicines11020519

AMA Style

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(2):519. https://doi.org/10.3390/biomedicines11020519

Chicago/Turabian Style

Bou Zerdan, Maroun, Prashanth Ashok Kumar, Daniel Zaccarini, Jeffrey Ross, Richard Huang, and Abirami Sivapiragasam. 2023. "Molecular Targets in Salivary Gland Cancers: A Comprehensive Genomic Analysis of 118 Mucoepidermoid Carcinoma Tumors" Biomedicines 11, no. 2: 519. https://doi.org/10.3390/biomedicines11020519

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