Next Article in Journal
STAT3-Mediated Promoter-Enhancer Interaction Up-Regulates Inhibitor of DNA Binding 1 (ID1) to Promote Colon Cancer Progression
Previous Article in Journal
Drug Target Identification and Drug Repurposing in Psoriasis through Systems Biology Approach, DNN-Based DTI Model and Genome-Wide Microarray Data
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Histone Deacetylases in the Pathogenesis of Salivary Gland Tumors and Therapeutic Targeting Options

by
Maria Manou
1,†,
Dimitrios S. Kanakoglou
1,†,
Theodoros Loupis
2,
Dimitrios M. Vrachnos
2,
Stamatios Theocharis
3,
Athanasios G. Papavassiliou
1,* and
Christina Piperi
1,*
1
Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Haematology Research Laboratory, Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
3
First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(12), 10038; https://doi.org/10.3390/ijms241210038
Submission received: 12 May 2023 / Revised: 9 June 2023 / Accepted: 10 June 2023 / Published: 12 June 2023
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
Salivary gland tumors (SGTs) comprise a rare and heterogenous category of benign/malignant neoplasms with progressively increasing knowledge of the molecular mechanisms underpinning their pathogenesis, poor prognosis, and therapeutic treatment efficacy. Emerging data are pointing toward an interplay of genetic and epigenetic factors contributing to their heterogeneity and diverse clinical phenotypes. Post-translational histone modifications such as histone acetylation/deacetylation have been shown to actively participate in the pathobiology of SGTs, further suggesting that histone deacetylating factors (HDACs), selective or pan-HDAC inhibitors (HDACis), might present effective treatment options for these neoplasms. Herein, we describe the molecular and epigenetic mechanisms underlying the pathology of the different types of SGTs, focusing on histone acetylation/deacetylation effects on gene expression as well as the progress of HDACis in SGT therapy and the current status of relevant clinical trials.

1. Introduction

Salivary gland tumors (SGTs) are rare neoplasms, representing 3–6% of head and neck (HN) tumors and constituting a wide range of subtypes (Table 1) [1,2]. Overall, 80% of SGTs fall into the category of benign tumors [1,2]. Pleomorphic adenoma (PA) is a frequent benign SGT regardless of the site of origin with incidence of 60–80%, which is followed by Warthin’s tumor (WT) (papillary cystadenolymphoma), basal cell adenoma (BCA), and myoepithelioma (MYE) [2,3,4]. The benign/malignant SGT ratio is 3:1 counting 55,000 cases per year [5]. Malignant cases usually arise in parotid. However, most sublingual gland tumors, and almost 50% of SGTs of the minor glands are also malignant [1,2,3,4,5,6,7]. Around 10% of SGTs arise in the submandibular gland, and 50% of them are diagnosed as PAs [1,2,3,4,5,6]. Mucoepidermoid carcinoma (MEC) refers to the most frequent malignant SGT with a reported incidence of 30% of all malignant SGTs. However, a small number of studies report that adenoid cystic carcinoma (AdCC) is more common [3,4]. These tumors are followed by acinic cell carcinoma (AciCC) and polymorphous adenocarcinoma (PAC) along with adenocarcinomas not-otherwise specified (AC-NOS) as the most frequent malignant SGTs.
Benign SGTs show predilection for the 4th–6th decade of life, whereas most malignant SGTs occur in the 6th–7th decade. Females are more prone to PAs than males, whilst the opposite occurs for MEC. Opposed to adults, acinic cell carcinoma remains the second most frequent aggressive SGT after MEC in children [2,3,4].
The symptoms of malignant tumors include painless swelling or facial nerve paralysis [5,6], erythema and dysesthesia [8], or fluctuant masses accompanied by pain, numbness, and ulcerations of the swelling [9]. Pre-treatment magnetic resonance imaging (MRI) is the first choice toward identifying SGTs and their features (localization/anatomical spread) [10], which is followed by computed tomography and ultrasonography. Recent MRI instrumentation has been helpful in differentiating benign from malignant tumors [11].
The 5th WHO classification encompasses “mucinous adenocarcinoma”, which is characterized by AKT1 (E17K) mutations and histological heterogeneity indicating intraductal papillary mucinous neoplasms of salivary glands. Newly added malignancies enfold “sclerosing microcystic adenocarcinoma” as well as “microsecretory adenocarcinoma” [7].
Most malignant SGTs, except for salivary duct carcinomas (SDC), are considered low-grade malignancies. High-grade transformation is characterized by architectural and cytologic changes, most prominently necrosis, loss of classic architectural patterns such as duct formation, increased mitotic rate and anaplastic cells pleomorphic nuclei with enlarged nucleoli [5,12].
Recurrence is not uncommon in SGTs, with metastatic events being usually grade-related and occurring in almost half of the cases (25–50%) [13]. High-grade neoplasms are linked to high morbidity due to distant metastases via hematogenous spread. Tumors such as AdCC and SDC display distant metastases in lung, liver, or bone [14] with the 5-year survival ratio of relapsed SGTs being just about 20% [13].
Relapse might be preventable, but appropriate therapy requires risk determination, which is conditional to factors including clinical manifestation, imaging, histologic type-characteristic and molecular biomarkers. Primary radiotherapy is employed for inoperable tumors, and adjuvant radiotherapy is selected in extensive nodal and/or locoregional involvement, when there is the presence of residual tumor foci, relapsing disease, and in high-grade tumors that infiltrate vascular channels or neural parenchyma [15,16]. Intensity modulated RT (IMRT) is currently chosen for malignant SGTs’ treatment, offering protection to nerves and relative maintenance of salivary gland function [17].
The use of chemotherapeutic agents is limited and is employed as a palliative mode of treatment or in cases of advanced and/or symptomatic metastatic disease with platinum-based agents being the main option [13,15]. Other treatment options include androgen deprivation therapy (ADT) for androgen receptor positive malignant SGTs, Herceptin plus Docetaxel and ado-Trastuzumab for HER2 positive tumors as well as Entrectinib and Crizotinib in SDCs with ETV-NTRK fusion, and Cabozantinib in intraductal carcinoma with NCOA4-RET fusion [13,15].
Despite their heterogeneity, SGTs are often jointly included in clinical studies. Upon a lack of systematic SGT treatments, repetitive and metastatic tumors remain cureless on most occasions. Owing to their rareness, the genetic and epigenetic characteristics of SGTs are just emerging, and their hallmarks are not well elucidated, harnessing their therapeutic targeting. Thus, further insights on vulnerabilities and molecular landscape are necessary for these life-threatening and hard to treat tumors.

2. Molecular Aspects of SGT Pathology

2.1. Genetic Changes in SGTs

SGTs compose a diverse mosaic of various neoplasms. Their genetic profiling is currently underway, and the main genetic changes characterizing these tumors are described below. However, the elucidation of their molecular landscape remains a challenge (Table 2).
Benign BCA encompasses mutations on the CTNNB1 gene (40%) coding for β-catenin, and part of BCAs manifest a nuclear expression of β-catenin [18]. Approximately 10% of WTs carry genetic variations, with the most common being deletions on chromosome 8, which are accompanied by deletions on 9p [19].
PA and carcinoma ex-pleomorphic adenoma (CAexPA) are characterized by translocations associated to the high-mobility group AT-hook 2 (HMGA2), gene which are frequent (~96%) on PAs and less common in PA and CAexPA (~30%) [20]. Moreover, HMGA2 and pleomorphic adenoma gene 1 (PLAG1) fusions are exclusively occurring in PA and CAexPA and have not been detected in other SGT subtypes [21].
MECs exhibit a slow growing pattern [22], and the majority are marked by repetitive translocations, which are specific for MECs [22,23]. The CREB Regulated Transcription Coactivator 1/Mastermind Like Transcriptional Coactivator 2 (CRTC1/MAML2) fusion has a better prognostic potential [23] than CRTC3/MAML2 fusion that occurs at younger ages (~35 years) [24].
AciCC exhibits activation of the EGFR/Akt/mTOR pathway [25], and a small percentage (<5%) carry rearrangements on the Myb/SANT DNA Binding Domain Containing 3 (MSANTD3) gene [26].
In AdCC, the fusion of the MYB/NFIB gene has been detected [27] followed by 1p36 deletion, which adds to dismal prognosis [28]. AdCC point mutations are related to BRAF, KRAS, SPEN, FGFR2 and NOTCH signaling. Mutations of p53 occur later in the progression of high-grade AdCCs [29].
The mammary analogue secretory carcinomas (MASCs) of SGTs, likewise to breast-type, are characterized by a recurrent translocation which results in ETV6/NTRK3 gene fusion [30] that is not observed in other SGTs.
The clear cell carcinoma (CCC) is exhibiting the EWSR1/ATF1 gene fusion [31], which is also harbored in clear cell types of myoepithelial carcinoma, sarcoma, and odontogenic carcinoma; thus, it is possibly associated with clear cell malignancies [27].
To date, the precise molecular mechanisms underlying these malignancies remain limited. Distinct types of malignancies might express alterative genetic and epigenetic defects. The regulation of gene expression is complicated, and there are diverse mechanisms of epigenetic modifications that might interact, such as histone modifications, DNA methylation and miRNAs. A notable difference of epigenetic patterns is the fact that in contrast to genetic alterations, these changes are potentially reversible and may be restored in vivo as well as in vitro [32,33]. Therefore, the screening of distinct SGTs is essential for their diagnosis and targeted management.
Table 2. Characteristic/recurrent genetic aberrations on SGTs and their clinical course.
Table 2. Characteristic/recurrent genetic aberrations on SGTs and their clinical course.
Tumor BehaviorTumor TypeGenetic AberrationPrevalenceClinical Course
MalignantMECsMECT1/MAML2 fusions40–80%Favorable prognosis [25]
CRTC3/MAML2 fusions6%/younger agesFavorable prognosis [24]
AdCCsMYB/NFIB gene fusions50%Unfavorable prognosis [25]
AciCCsHTN3/MSANTD3 fusions<5%Indolent course [26]
MSANTD3 aberrations4–15%
MASCsETV6/NTRK3 fusionsMajorityFavorable prognosis [34]
(Hyalinizing) CCCsEWSR1/ATF1 fusions93%Indolent course [35]
EWSR1 rearrangement82%
Intraductal carcinomaNCOA4-RET35%Mean OS: 5 years [36]
TRIM27-RET~10%Aggressive/apocrine variant [36]
Microsecretory adenocarcinomaMEF2C-SS18 fusionMajorityMalignant nature [37]
PAC and CASGPRKD1/2/3 translocations or mutationsPRKD1/2/3 fusions: 13% of PACs;
PRKD1 mutations: 56–73% PACs
Indolent course [38]
PRKD1/2/3 fusions: 43–80% CASGs;
PRKD1 mutations: 20% CASGs
BCACPIK3CA mutations~10–30%Unfavorable prognosis [39,40]
CAexPAPLAG1/HMGA2 fusions36%Indolent course [41]
BenignPAsPLAG1 translocations50%Indolent course [42]
HMGA2 translocations34–40%
BCACTNNB1 mutations60%Indolent course [43]

2.2. Epigenetic Changes in SGTs

Epigenetics refers to inherited alterations in gene expression that can take place independently of the primary DNA sequence [44]. They are mediated by key mechanisms including DNA methylation, post-translational alterations of histones, such as acetylation/deacetylation, methylation, ubiquitination or sumoylation and the binding of microRNAs [45]. Epigenetic alterations are involved in many physiological processes such as cell differentiation and embryogenesis, but they are also implicated in altered cell states and pathological processes such as cancer [45].
Over the years and through the evolving use of different research approaches, the role of epigenetic modifications in SGTs has emerged, clarifying the landscape upon their etiopathogenetic and biologic characteristics, being associated with tumor initiation, growth, and metastatic potential (Table 3).
With respect to DNA methylation, although physiological salivary specimens lack a methylation index, benign tumors (PAs and WTs) exhibit methylation on RASSF1, MGMT and DAPK genes [46].
Transition to carcinoma or higher grade is an event whose molecular trigger is uncertain. The hypermethylation of p16(INK4) and p14(ARF) occurs in CAexPA and PA, whereas the hypermethylation of RASSF1 was less prominent in PA but more evident at CAexPA [47]. Compared to non-cancerous cases, the p16 (CDKN2a) gene shows higher methylation rates in CAexPA [48]. CDH1 is frequently silenced in CaexPA by promoter methylation (57%), indicating that its methylation is essential for the trigger and progression of CaexPA [49].
DNA methylation has also been detected in MEC and AdCC [50]. Specifically, p14 hypermethylation is a hallmark of MEC progress followed by p16, TP53 and hTERT [50]. The normal tissue lacks modifications of p16(INK4a), whereas its hypermethylation was greater in higher-grade compared to lower-grade MECs [51]. RUNX3 silencing through hypermethylation is prominent on MEC (87%), AdCC (75%) and PA (25%) compared to non-malignant specimens, adding to SGTs’ progression and poor prognosis [52]. In AdCC, the methylation of CK genes has been frequently detected [53,54], and a high methylation rate on p21, p19, p18 and p15 (69–92%) was reported [55]. In metastatic and high-grade AdCC, the hypermethylation of RASSF1 has been detected [46].
Kanazawa et al. demonstrated that galanin receptors’ hypermethylation correlated with decreasing OS in SDCs; thus, GALR-1/-2 gene silencing through methylation dictates a crucial epigenetic event with potentially important therapeutic and prognostic significance [56].
Wang et al. found that in CRTC1/MAML2 fusion positive MECS, only CLIC3 demonstrated semantic hypomethylation as well as protein overexpression; therefore, CLIC3 was suggested as an auspicious epigenetic biomarker for MECs [57]. Epigenetic E-cadherin (CDH1) silencing through hypermethylation is a significant pathway in MECs pathogenesis [58] as well as in AdCC [59].
Bell and colleagues found that EN1 shows constant overexpression in AdCCs [60], whereas it is positivity correlated with lymph node metastasis and poor prognosis [61], indicating the diagnostic utility of EN1 on distinct SGTs (AdCCs, PAC) [62]. Additionally, the hypermethylation of RASSF1A is associated with initiation and unfavorable prognosis in AdCCs [63], whereas a higher index of H3K9me3 is prognostic of brisk growth and outlying dissemination in AdCCs [64]. Through epigenetic approaches, Shao and colleagues distinguished AQP1 as an intrant oncogene concerning AdCCs; thus, its overexpression and hypomethylation suggest a signature role in AdCCs [65]. Additionally, Stratifin (14-3-3 s) is recurrently downregulated in AdCC due to hypermethylation, constituting an important mark for the evolvement of AdCC and the radiosensitization of SGTs as well [66]. Lee et al. reported that the RARb2 and RASSF1A genes show regular methylation rates in distinct entities such as AciCC and AdCC [67].
In benign tumors (such as PA), histone methyltransferases (HMTs) SET08, SETDB1, and Eu are elevated compared to physiological tissue [68], further indicating that HMTs can be associated to PA differentiation. Therefore, epigenetic events affecting histones modifications are essential regulators of related genes that participate in tumor transformation [68].
Contrary to benign SGTs, malignant SGTs show low acetylation activity and therefore display a tighter chromatin format. Additionally, an inversely proportional relationship has been detected between H3 (lys9) acetylation and SGTs’ cell proliferation [69]. The methylation and hyperacetylation of H3 is linked to the aggressive phenotype of AdCC [64,70,71], and H3K9 trimethylation co-exists with high grade and invasive features of MECs. In both MECs and AdCCs, higher rates of histone H3 modifications are linked to higher cancer proliferation [71]. Particularly, the upregulation of H3K9Ac, H3K18Ac, and H3K9me3 in AdCC and upregulation of H3K9me3 in MECs were linked with increased Ki-67 expression and malignant pathological features, including aggressiveness [71]. This event is attributed to H3K9me3 and H3K9Ac due to their interaction with genes that are involved in cell proliferation (p21Cip1/Notch1/ERK) [72,73,74]. Additionally, a study demonstrated that 50% of AdCC harbor modifications in genes associated with histone alterations and chromatin remodeling (CHD2, BRD2, ARID5B, KDM5A) [75].
In the context of SGT pathogenesis and their association to epigenetic features, it has been detected that over 3600 mRNA, 280 circRNAs and 3090 lncRNAs are modified [76]. As mentioned above, CRTC1/MAML2 fusion is a well-characterized tumorigenic starting event in the pathogenesis of MECs. Recently, it was reported that lncRNA/LINC00473 is a significant moderator of the CRTC1/MAML2 oncogene [77]. Additionally, miR-302 is upregulated in MEC, and this event enhanced cancer cell migration in vitro [78]. To analyze the expression of apoptotic miRNAs in SGT, a study reported that miR-34a and miR-21 are overexpressed in MEC (74% and 91%, respectively). In the same study, various apoptosis-associated microRNAs (miR-5p, miR-15a, miR-16, miR-17, miR-21, miR-29, miR-34a) were upregulated in PA cases. The analysis revealed that the apoptotic modulator miR-20a was downregulated in MEC (57%) and PA (75%) [79]. These data showed that there is a modified expression of relative genes (BAD, APAF1, BAX, BCL2, DIABLO, CASP8, TP53) in benign PAs and in malignant MECs (TP53, BAX, APAF1, DIABLO, BID, BCL2, CASP-2/3/6/8) [79]. Therefore, in SGTs, modifications in apoptotic miRNAs might be tightly associated with oncogenesis. Likewise, mRNAs and lncRNAs are differentially co-expressed while interacting in PAs [80]. In particular, mRNAs are associated with events of PAs growth, and lncRNAs may affect PAs’ tumorigenesis through the modification of gene expression (IGFBP5, IGF2R and IGF2 mRNAs). Notably, this outcome shows the involvement of different epigenetic modifications on PA, taking part either individually or in combination within SGTs [80].
Moreover, in PLAG1 transgenic mice, the abnormal expression of lncRNAs and mRNAs was indicative for PAs pathogenesis through interfering with a plethora of genes, suggesting a new biomarker in PAs management [80]. The miR-17-92 cluster (miR-17/-20a) was found to be particularly upregulated and related to the aggressive biology of AdCCs, indicating a potential therapeutic target on this entity [81]. Similarly, related to MECs pathogenesis, an aberrant expression of distinct ncRNAs (ON-HSAT154433.1 upregulation/hsa_circ_00123 downregulation) could indicate prospective biomarkers and therapeutical factors on MECs [76,82].
Taken together, epigenetic alterations play a significant role in SGTs’ pathogenesis with a prognostic and diagnostic potential, enabling target therapies.
In this review, we are interested in the role of histone modifications in SGTs and particularly on histone deacetylation mechanisms that might be effective on neoplasms with relevant epigenetic signatures.
Table 3. Epigenetic modifications of SGTs with therapeutic/prognostic marker potential.
Table 3. Epigenetic modifications of SGTs with therapeutic/prognostic marker potential.
SG EntityAberration FormRelated MoleculeReferences
AdCCs methylationEN1, RASSF1A, RECK, Stratifin (14-3-3 s), RUNX3[63,66,83,84,85]
methylationSBSN, AQP1[65]
Promoter methylationE-cadherin, RASSF1A, RARb2[54,59,67,86]
Histone trimethylationH3K9me3[64,87]
acetylationH3K9Ac, H3K18Ac[71,88]
miR-17-92 regulationmiR-17/-20a[81]
MECsAberrant expression of ncRNAsNONHSAT154433.1, has-circ-0012342[76,82]
methylationRUNX3, H3K9Me3[52,85,88]
acetylationH3K9Ac, H3K18Ac[71,88]
SDCs methylationGALR-1/-2[56]
Promoter methylationRASSF1A, RARb2,[67]
PAsDysregulated expression (PLAG1 gene related)9110 lncRNAs/7750 mRNAs [80]
CAexPAPromoter methylationCDH1 (E-cadherin)[49,89]
ACCsPromoter methylationRASSF1A, RARb2[67]
↑ refers to increased, ↓ refers to decreased.

3. Role of Histone Deacetylation in SGTs

3.1. Biochemical and Functional Aspects of Histone Acetylation/Deacetylation

The abundance of lysine residues in histones creates an ideal environment for modifications, especially acetylation and its opposite reaction, deacetylation (Figure 1). The former occurs when an acetyl group from an acyl-CoA donor is placed in the ε-amino group of the lysine side chain via the action of specialized enzyme called histone acetyltransferases (HATs) or recently renamed as lysine acetyltransferases (KATs) since acetylation has been also discovered in non-histone proteins (6). By adding acetyl residues, they may neutralize the positive charge of lysine residues eventually “loosening” the bond of histones with DNA, allowing recruitment and binding of transcriptional factors. There are more than 20 types of HATs/KATs that are freely distributed either in the nucleus or the cytoplasm, acting as transcriptional co-activators [90].
Both acetylation and deacetylation are highly reversible processes, thus for effective gene regulation, there is necessity for balance between the addition and the removal of acetyl groups [90,91]. Removal of acetyl groups (deacetylation) is achieved by histone deacetylases (HDACs) or lysine deacetylases (KDACs) [90]. HDACs function in “re-packing” tightly the DNA with histones via positively charging the lysine residues inducing a close chromatin complex and blocking RNA polymerases from initiating DNA transcription [92].
To this day, 18 HDACs have been identified. The interaction with several co-factors and the existence of a deacetylase domain are the two main characteristics taken into account for the subcategorization of HDACs in two large families: the deacetylase family which comprises of zinc-dependent enzymes and is furtherly subclassified in type-I (HDAC1, -2, -3, -8), type-II (HDAC4, -5, -6, -7, -9, -10) and type-IV (HDAC11), with type-II consisting of two subfamilies of type-IIa and type-IIb due to differences in their domain composition [92].
Each class is further divided into subtypes, and each HDAC subtype has distinct cellular locations and substrate specificities. These enzymes are involved in various cellular processes, such as gene expression regulation, cell cycle progression, apoptosis, and cellular differentiation. The following table summarizes HDAC classes, subtypes, cellular locations, substrate specificities, and main functions (Table 4).
Studies investigating HDACs expression and activity in carcinogenesis are progressively increasing [91,95]. More specifically, Type I HDACs are often dysregulated in cancer regulating cell proliferation and apoptosis [96]. Regarding class-IIa HDACs, HDAC4 may exhibit either a tumor suppressive or oncogenic role [97] whereas HDAC5 and HDAC9 play a significant role in medulloblastomas [98]. In lung cancer, HDAC6 has been linked to epithelial-mesenchymal transition (EMT) via the SMAD3 pathway [99] and HDAC10 was shown to induce proliferation via the AKT signaling pathway [100].
SIRT1 has been upregulated in prostate cancer and leukemia [101,102] while SIRT2 was shown to possess tumor suppressive properties [103]. In urothelial cancer, increased SIRT3 levels were linked to cancer cell cycle arrest. Increased SIRT3 and SIRT6 levels have been detected in breast cancer [103,104] and reduced in gliomas as well as in ovarian cancer [105].

3.2. Role of HDACs in Different SGT Types

HDAC family members have been detected in various types of malignancies such as oral cavity cancer, but the SGTs remain an entity which has not been thoroughly investigated [106].
Based on the Human Protein Atlas (HPA), HDACs have moderate to increased protein-expression levels in breast, cervical, colon, endometrial, liver, lung, ovarian, pancreatic, prostate, renal, glioma, lymphoma, and HN cancer [107]. In HN malignancies, HDAC class-I and -II expressions vary (Table 5). The majority of HDACs members are expressed to almost 100% (HDAC2, -9, -10, SIRT3, -5, -6, -7), ~75% (HDAC1, SIRT1) and ~25% (HDAC3, -6). HDAC5 is not included in the above due to a lack of expression [107]. Almost 39% of NUT midline carcinomas (NMCs) are located in HN [108]. In the case of NMC present in the parotid gland, immunohistochemical analysis showed strong to moderate immuno-expression of HDAC2, -4 and -6, and phospho-HDAC4, -5, -7, in the neoplastic cells [109].
Moreover, an immunohistochemical study from our group evaluated the expression of HDACs (-1, -2, -4 and -6) in SGTs [110]. HDAC subtypes were abundantly expressed in both benign and malignant samples. In malignant tumors, HDAC1, 2, 4 and HDAC6 expression was observed in 14%, 82%, 36% and 18%, respectively. High HDAC expression was present in 25% to 89% of HDAC-positive SGTs and was not associated with patients’ age and gender. HDAC2 expression was suggested as a positive prognostic factor and was associated to extended overall survival (OS) on high-grade malignant SGTs. HDAC6 overexpression, on the other hand, might have a negative impact on the OS of high-grade SGT cases given that HDAC6 knockdown was linked to extended OS [110].
Wagner et al. studied the immunohistochemical expression of acetylated histone H3 and Ki67 in 84 cases of SGTs with tissue microarrays (TMAs). The analysis showed that aggressive SGTs were under-acetylated, in contrast to benign SGTs, leading to chromatin condensation. Moreover, H3 was oppositely associated with Ki67 expression on SGTs, indicating that HDACs increase cell proliferation [69].
Chidamide (HBI-8000/Tucidinostat), which induces H3 and H4 acetylation, has shown a selective inhibitory effect to HDACs (class-I and -II) [111]; another study reported on the efficacious stability/durability and response of a patient with advanced AdCC during a phase-I trial [112]. This was the case for the preclinical administration of Chidamide in AdCC that exhibited significant inhibition and satisfactory results, suggesting its potential effectiveness for AdCC patients [113].
Preclinically, HDAC6i/KA2507 showed antitumor efficacy in 93 human cancer cell lines and in in vivo cancer models, while its antitumor efficiency was increased upon the co-administration of a checkpoint inhibitor and subsequently allocated to the first clinical study where SGTs (AdCC, SDCs) were also included [114]. Based on prior NGS (next-generation sequencing) analysis in AdCC [75,115] and the supporting radiological/clinical findings of two salivary AdCC patients in a clinical study of vorinostat evaluation in advanced malignancies [116], vorinostat was further evaluated in a phase-II trial of AdCCs with promising results [117].
Studies using in vitro and in vivo models demonstrate the synergistic effect of different mechanisms such are checkpoint and HDAC inhibitors [118]. Several trial data are also encouraging upon solid and blood malignancies [119]. The Keynote-028 study depicted 3/26 SGT respondents with PDL-1 expression (>1%) [120]. A phase-II/vorinostat study in 30 relapsed/metastatic AdCCs reported 97% clinical advantage and 2 impartial responses [121]. In this frame, Rodrigues et al., performed a phase-II study that investigated the combinatory effect of pan-HDACi/vorinostat together with the anti-PD1i/Pembrolizumab in different SGTs, reporting important clinical outcomes in different types (lymphoepithelioma-like/AciCC/AdCC carcinomas) [122]. Τhe underlying mechanisms and types of inhibition in the various subtypes of SGTs are discussed in detail in the following section.
The integration of complementary bioinformatics approaches is crucial for the comprehensive identification and analysis of HDAC expression in a variety of SGT types, providing insights into their functional roles, molecular mechanisms, and potential as therapeutic targets. Leveraging computational methods and tools, such as artificial intelligence (AI) and machine learning (ML), researchers can obtain crucial insights into the roles of HDACs in SGT biology and explore their potential as therapeutic targets. Additionally, in silico analyses of publicly available datasets, such as The Cancer Genome Atlas (TCGA) [123] or the Gene Expression Omnibus (GEO) [124], can provide a wealth of information on HDAC expression patterns as well as their association with other proteins and molecular pathways.
Gene expression profiling techniques, such as microarrays, RNA-Seq, and single-cell RNA-sequencing (scRNA-Seq), enable the assessment of global gene expression levels, including HDACs, across various tumor types [124]. ScRNA-Seq, in particular, provides a higher resolution of the cellular heterogeneity within SGTs, allowing for the identification of HDAC expression patterns in specific cell populations [125]. By comparing the expression profiles between tumor and normal tissues or among distinct tumor types, scientists can identify differentially expressed HDACs that may contribute to tumor development or progression [126]. Furthermore, co-expression analysis can help identify genes that are co-expressed with HDACs in SGTs, potentially uncovering functionally related or co-regulated genes and providing insights into the molecular mechanisms of HDACs in these tumors [127]. Further downstream, gene set enrichment analysis (GSEA, also called functional enrichment analysis) can be employed to delineate the biological processes and molecular pathways associated with differentially expressed HDACs, offering a deeper understanding of their roles in SGTs [128]. Protein–protein interaction network (PPIN) analysis can explore the molecular interactions between HDACs and other proteins in SGTs, potentially revealing novel therapeutic targets or emphasizing the role of HDACs in the context of other relevant proteins [129].
Lastly, AI and ML techniques can be applied to classify SGT types based on HDAC expression patterns or predict patient outcomes [130]. By incorporating multi-omics data, such as genomics, transcriptomics, and proteomics, ML algorithms such as support vector machines (SVMs), random forests (RF), or deep learning (DL) models can offer a more comprehensive understanding of the molecular landscape of SGTs, facilitating the identification of potential biomarkers for diagnostic or prognostic purposes and aiding in the development of personalized therapeutic strategies [131].

4. Targeting Options of HDACis in SGTs

4.1. HDAC Inhibition

HDACis are natural or semi-factitial derivatives of natural products [10]. Based on their structure, they are classified into small-chain fatty acids such as valproic acid (VPA) and butyrate acid, cyclic peptides/depsipeptide-FK228 (Romidepsin), hydroxamic acids (SAHA), LBH589 (Panobinostat), TSA (Trichostatin A), AR-42 (a hydroxamate-phenylbutyrate derivative), cyclic tetra-peptide (Trapoxin A) and benzamides (Entinostat/MS-275 and MGCD0103) [11]. Lately, the role of synthetic alkyl 4HR as a new HDACi has emerged [132,133]. Additionally, tumor angiogenesis is inhibited by a variety of actions that the hydroxamic derivative LBH589 has on endothelial cells [134].
HDACis might promote oncogenic mechanisms through various gene signaling pathways. SAHA acetylates tumor suppressors such p53 and miR-15/16, and it favors the alteration of mutant into wild-type p53 cancer cells [135]. MPT0G009 administration enhances TRAIL expression [136], and valproic acid prompts the activation of p21 and PARP [137]. Likewise, HDACis lead to histone refracturing on the p21 domain, modulate the cell cycle genes (Cdk-1, Cdk-4, Cdk-6) expression, and regulate serine/threonine kinases [136,137].
HDACis might also affect several biological processes in cancer cells such as apoptotic activity by balancing gene expression via histone methylation or/and acetylation as well as by acetylation of diverse non-histone proteins such as STAT-3, p53 and tubulin. Additionally, HDACis can regulate apoptosis by inducing ubiquitin (UBB) gene expression or Mcl-1 and Bcl-2 [138], modulate caspase proteins by inducing TRAIL expression, downregulate c-FLIP factors and regulate cell-cycle arrest at active or pathologically proliferative malignant cells [139,140,141].
Over the years, SGTs’ treatment relies on combination therapy with platinum, single-agent, HER-2, EGFR, c-kit, tyrosine kinase and microtubular inhibitors, fusion transcripts targets, hormonal or combination therapy with biologics and immune checkpoint inhibitors [142]. Immunotherapy in combination with other agents for SGTs such as HDAC inhibitors is a field that is now beginning to be explored (Table 6).

4.1.1. HDACis in Mucoepidermoid Carcinoma (MEC)

Unlike combination therapies, multi-target agents are in favor of more predictable pharmacokinetics and may reduce therapeutical cross-reactions [150]. CUDC-101 is a molecule composed of combined EGFR and HDAC inhibitors that demonstrates efficacy in comparison to combined therapies including EGFRi (Erlotinib) and HDACis (SAHA). This agent is adequate toward almost 40 various cell lines among 16 types of cancers (in vitro/in vivo) [151,152] and shows efficacious tolerance on clinical studies [153]. CUDC-101 might blunt MEC cancer stem-cell tumorigenicity, shows irrevocable and robust cytotoxicity on MEC cells and affects apoptosis through caspase 3/7 activation [147]. As proposed by Sharma and colleagues, together, HDACi and EGFRi suggest a promising agent against MEC tumors. Additionally, CUDC-101 is effective in simultaneously downregulating p-EGFR and upregulating histone acetylation (H3K9ac and H4K12ac) compared to EGFR and SAHA. Therefore, coupling EGFRi and HDACi might bring up to fivefold cell cytotoxicity compared to EGFR or HDAC single treatment [147].
HDAC7 modulates cell proliferation, differentiation, apoptosis, and migration in physiological and pathological conditions [154]. The aberrant expression of HDAC7 has been observed in gastric cancer [155], breast cancer [156], leukemia and lymphoma [157], glioma cancer [158], lung cancer [159], ovarian cancer [160] and nasopharyngeal carcinoma [161]. The broad expression of HDAC7 is related to poor prognosis and metastasis [155,159]. In addition, various HDACis such as Trichostatin A [162], valproic acid [163] and vorinostat [164] target HDAC7. The role and the underlying mechanism of HDAC7 in MEC is yet to be clarified. Ahn and colleagues studied the association between HDAC7 and autophagy in cancer cells [144]. YD-15 and Mc3 cells were treated with distinct HDAC7 siRNAs. HDAC silencing induced autophagy in MEC cells, by regulating LC3-II and p62 expression, possibly through a pro-survival mechanism which resembles the apicidin-induced autophagy. Moreover, HDAC7 suppression affected apoptosis through activating caspases [144]. Additionally, MAPK (mitogen-activated protein kinase) activity is associated with a reduction in ERK phosphorylation, hypothesizing that HDAC7 silencing allies with ERK inactivation in MEC. Interestingly, HDAC7 might play a robust oncogenic role in MEC cells by inhibiting cell proliferation (G2/M arrest), downregulating c-Myc and upregulating p27 accumulation [144].
Apicidin might inhibit the proliferative potential of MEC cells [143]. Apicidin-prompted apoptosis and autophagy might act in a competitive way; therefore, autophagy may act protectively on trigger apicidin-prompted apoptosis in MEC. Additionally, apicidin induces an apoptotic effect by inactivating ERK and AKT/mTOR signaling and activating JNK (c-Jun NH2-terminal kinase). The autophagic response of apicidin is also mediated by inactivating AKT/mTOR pathways. Apicidin cytotoxicity is limited by JNK inhibition and enhanced by AKT and ERK inhibition. These events are probably caused by the downregulation of IGF1R, which upregulates the AKT/mTOR and MAPK signaling pathways [143].
Sodium butyrate or NaBu refers to an apoptotic HDACi [165]. Τhe route which NaBu uses in order to implement its apoptotic activity in MEC cells is unclear. Survivin (BIRC5) is an adaptor protein that affects mitosis and apoptotic inhibition [166]. Survivin’s function depends on its cellular location [167,168]. In SGTs, when expressed in cytoplasm, it is associated with poor prognosis [165]. In the nucleus of malignant cells, survivin binds with STAT3, thus inhibiting the transcriptional potential of downstream factors of the STAT3 pathway [148,169,170]. In oral YD15 and MC3-MEC cell lines, NaBu might decrease cell viability, inhibit the transcriptional potency of the Bcl-XL/STAT3 molecule, and enhance the cleavage of caspase 9 and caspase 3. In addition, NaBu acetylates survivin, causing its nuclear translocation, which in turn represses STAT3 oncogenic activity and prompts caspase-reliant apoptosis in MEC [148]. In vivo, NaBu might attenuate tumor proliferation without hepatic or renal toxicity [148].

4.1.2. HDACis in Adenoid Cystic Carcinoma (AdCC)

AdCC is an infrequent tumor often occurring in salivary glands. Almost half (35–50%) of AdCC patients bring gene mutations associated with the remodeling of chromatin and histone alterations [75,113,115]. Hitherto, there is no targeted agent approved by the FDA for its treatment [117]. In in vivo as well as in vitro trials, vorinostat usage minimized the CSCs rate [146]. Despite the total depletion in tumor cell viability, the prolonged administration of vorinostat led to CSCs proliferation in AdCC primary cells. Nevertheless, synergistically, cisplatin along with vorinostat decreased the viability of HACC2A as well as HACC6 tumor cells by 73.5% and 77.5%, respectively. Vorinostat stimulated cancer cells’ sensitization in cisplatin through a mechanism that couples CSCs depletion and triggering cell senescence [146].
Chidamide is a new HDACi that falls into the benzamide family. Specifically, it increases the acetylation potential in the H3 histone, is more stable than SAHA and eclectically intercepts the effect of HDAC1, -2, -3 and -10 [171]. To date, it has been only authorized by China’s FDA as a choice over relapsed or refractory PTCL management [172]. Chidamide implements its antitumor effect through various mechanisms. Additionally, it might reverse the EMT and treatment resistance of neoplastic cells [171], induce apoptosis and tumor cell growth arrest [173] and amplify apoptotic activity as well as platinum-based DNA-damage response in non-small cell lung cancer (NSCLC) [174]. In phase-I and -II studies, chidamide has shown favorable pharmacokinetics and pharmaco-dynamics, notable tolerability, and antitumor efficiency [112,175].
In preclinical study, chidamide and cisplatin (CDDP) restrained the proliferation of AdCC xenografts [113]. In AdCC-2 and AdCC-3 cells, the cleavage of caspase 3 was not observed after chidamide administration, whilst the chidamide-induced apoptotic path is still unclear [113]. Chidamide may also inhibit cell development and proliferation through a dosage and time-dependent mode and affect the G2/M phase of cells. Moreover, it can decrease p-AKT expression and upregulate Ac-H3 protein expression in AdCC. Thus, chidamide inhibits cell growth by increasing H3 acetylation and intervening on AKT phosphorylation and G2/M cell-cycle arrest on AdCC cells [113].

4.1.3. HDACis in Salivary Gland Ductal Adenocarcinoma (SGDA)

The p53 family member p63 maintains two characteristic isoforms, which are crucial for proliferating and differentiating epithelial cells. TAp63 disposes a pro-apoptotic effect, whereas DNp63 is oncogenic [176]. p63 is upregulated in various malignancies such as SGTs [177]. Diagnostically, DNp63 (p63/p40) applies for histologically differentiating SGTs [178]. So far, the roles and mechanisms of p63 on SGTs are not well known. Nakano and colleagues transfected A253 (SGDA) cells with siRNA-p63 or injected with an HDACi (Trichostatin A and Quisinostat) [149] (Figure 2). As shown, the knockdown of p63 with siRNA-p63 may induce tight junction (TJ) proteins, differentiation markers and aberrant cell metabolism in human SGDA A253 cells. Both p63 inhibition and HDACis hinder the proliferation of A253 cells. In addition, HDACis may inhibit p63 protein expression, enhance TJ proteins expression and prevent cell migration in SGDA cells, suggesting their potential therapeutic utility [149].

4.2. Phase-I Clinical Studies of HDACis in SGTs

4.2.1. Vorinostat in a Phase I Study

Vorinostat inhibits class-I as well as class-II HDACs and holds FDA approval for administration at refractory CTCL [179]. In phase-I trials, vorinostat was administered on patients with hepatic malfunction and solid malignancies [116]. Before entering the trial, all AdCC patients had disease advancement in the former treatment. A salivary AdCC patients’ response to treatment led to the enrollment of a further four patients with AdCC. Additionally, one patient with AdCC had a partial response, and four reached disease steadiness in a period of 4, 12, 12 and 13 months [116].

4.2.2. KA2507 Selective HDAC6 Inhibitor

Ubiquitously expressed HDAC6 is located on the cytoplasm [180] and affects tumor cell modifications by inducing anchorage-independent proliferation in transmuted cells [181]. Via post-translational modifications of its substrates and cytoplasmic-interacting molecules (such as α-tubulin, cortactin and HSP90), HDAC6 affects a variety of cellular processes such as intracellular signaling, DNA-damage response, gene transcription, and cell movement. Disruption of the physiological expression and function of HDAC6 affects tumorigenic modifications, cancer cell growth, mitosis, invasion, and migration [182]. So far, there is no FDA approval for HDAC6 inhibitors except for the pan-HDACis such as LBH589, PXD101, Romidepsin and SAHA [183,184].
KA2507 is hydroxamic acid family inhibitor of nano-molecular weight that eclectically targets HDAC6. From 2017 to 2019, in a phase-I trial, KA2507 was administrated in 20 patients with solid tumors including heavily pretreated, treatment-refractory patients [114]. Patients with salivary AdCC (15%) and SDC (5%) were included. Administrating KA2507 had positive tolerance and showed no evidence of mechanism-based toxicity (myelosuppression, cardiac toxicity). Stable disease was observed in a patient with AdCC.
In vitro, KA2507 lacks inhibition potency on proliferative malignant cells (human/mouse). On the other hand, KA2507 demonstrated efficacy in in vivo preclinical cancer types [114]. The selective inhibition of HDAC6, barren of off-target class-I HDAC effect, correlated with increased α-tubulin acetylation and the downregulation of histone H3 acetylation. Moreover, in mouse models, KA2507 can modulate antitumor response through the alteration of several immune biomarkers. It was shown to reduce STAT3 activation, decrease the expression of PD-L1, increase type-I MHC proteins and show efficacy when co-administered with a checkpoint inhibitor [114].

4.3. Phase-II Trials of HDACis in SGTs

4.3.1. SAHA Treatment in AdCC

AdCC is an uncommon type of malignant SGT, encompassing 1% of HN cancers and 5–10% of the malignant SGTs [185]. Surgical intervention is the preferred way of management toward localized AdCC arising from major and minor salivary glands with postoperative radiotherapy to optimize local disease control. AdCC has a primary slow-moving profile but increased migration likelihood. In metastatic stages, AdCC shows low responsiveness to available therapies [186].
In a multi-institutional clinical trial (phase-II) of SAHA administration, 30 patients with progressed or metastatic AdCC were included [117], exhibiting in most cases (20) decreased tumor size. In addition, patients exhibited a median stable disease (8 months), progression-free survival (10 months) and overall survival (11.5 months). Partial response was noted in two cases, whilst three patients withdrew due to toxicity. Toxicities (grade-III) included lymphopenia (23%), hypertension (10%), fatigue (10%), oral achiness, cephalalgy and thromboembolic incidents. Moreover, two AdCC patients who underwent vorinostat therapy manifested early (after 2–3 cycles) clinical improvement including pain and tiredness reduction, weight advantage, radiological improvement, reduced liver lesion and normalization of liver enzymes. The late responses were attributed to vorinostat immunostimulation, which possibly requires prolonged time to manifest when compared to other therapeutic agents [117].

4.3.2. Combination of Pembrolizumab and Vorinostat in SGTs

Despite the encouraging response in HER2-positive SDC treatment [187], most SGT trials are rare, have a number of limitations and have low success rates. In a phase-II clinical trial (2015–2017), vorinostat and Pembrolizumab were administrated in recurring/metastatic SCCHN (squamous cell carcinomas of head and neck of HN/25 patients) together with SGTs (25 patients) [122]. Stable disease was met in 14 patients (56%), while responses were prolonged and observed in SGTs (16%) including a lymphoepithelioma-like parotid carcinoma, an AdCC and two acinic cell carcinoma patients. Toxicities were broader than Pembrolizumab alone, and adverse events were observed in 13 SGT patients (52%). Moreover, vorinostat-related dose reductions included nine patients (36%). Vorinostat-related toxicity included fatigue and weight loss (1), malaise (1), severe fatigue (2) and increased serum creatinine (5). The response was promising, although the dual treatment can exhibit lower responses in SGTs than HN [122]. A possible interpretation of the lower response rate in SGTs is their reduced rates of PD-L1 expression, reduced tumor mutational profile and non-association to previous tobacco usage, alcohol, or viral exposure.

5. Conclusions—Outlook

Malignant cells exploit epigenetic modifications in order to regulate tumorigenic proliferation, migration, and drug effectiveness [33,36]. Accordingly, curative agents tackling epigenetic mechanisms may be novel frontiers in cancer therapy. HDACis induce chromatin acetylation, but the molecular pathways that determine HDACis efficiency as sensitizing agents remain elusive. Alone or in synergy with known therapies, HDACis may inhibit resistance to antitumor treatment.
In vitro, EGFRi and HDACi (CUDC-101) display synergistic effects on MEC cells. Additionally, the inhibition of AKT and ERK amplifies apicidin-related cytotoxicity in MEC cells. Vorinostat and cisplatin synergistically reduce the viability of ACC cells. Vorinostat boosts cancer cells’ sensitization to cisplatin, depletes CSCs, and induces the activation of cellular senescence. Chidamide in synergy with cisplatin can hinder the growth of AdCC cells. HDACis, such as Quisinostat and TSA, may enhance TJ protein expression, downregulate p63 expression, and prevent cell migration in SGDA cells. Moreover, in an in vivo preclinical model, KA2507’s effectiveness was boosted by the co-administration of an immune checkpoint inhibitor. Clinically, HDAC6-selective inhibitor KA2507 is well tolerated with no significant toxicities. Therefore, KA2507 optimal use may be preferable in a combinatorial immunotherapy approach.
In a phase-I trial, vorinostat treatment of AdCC cells showed partial response and disease stabilization. In a phase-II study with vorinostat, most patients showed disease stability and decreased tumor size. HDAC7 inhibition triggers the apoptotic cell death and autophagy in MEC cells. In addition, NaBu can cause the nuclear translocation of survivin, inhibit growth, and induce apoptosis in MEC cells. HDAC2 is related to extended OS on high-grade SGT tumors. Conversely, HDAC6 upregulation is negatively correlated with the OS of high-grade malignant SGT patients, suggesting that HDAC6 inhibition holds promise in SGT treatment. Therefore, HDACis may be potent agents that exhibit antitumor activities.
Conclusively, SGT trials are currently infrequent and may have a number of limitations as well as low success rates. Nevertheless, the continuous investigation of molecular targets holds the key to successful cancer treatments, and a detailed profiling of HDAC modifications along with their inhibition mechanisms might prove to be a promising approach toward a more effective SGT treatment.

Author Contributions

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

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AciCC: acinic cell carcinoma; AC-NOS: adenocarcinomas non-otherwise specified; AdCC: adenoid cystic carcinoma; ADT: androgen deprivation therapy; AI: artificial intelligence; BCA: basal cell adenoma; BCAC: basal cell adenocarcinoma; CAexPA: carcinoma ex-pleomorphic adenoma; CASG: cribriform adenocarcinoma of salivary gland; CCC: clear cell carcinoma; CDDP: Cisplatin; CTCL: cutaneous T-cell lymphoma; DL: deep learning; DHS: deoxyhypusine synthase; EMT: epithelial-mesenchymal transition; GEO: gene expression omnibus; GSEA: gene set enrichment analysis; HAT: histone acetyltransferase; HDAC: histone deacetylase; HDACi: histone deacetylase inhibitor; HMT: histone methyltransferase; HN: head and neck tumor; HPA: human protein atlas; IMRT: intensity modulated RT; JNK: c-Jun NH2-terminal kinase; KAT: lysine acetyltransferase; KDAC: lysine deacetylase; MAPK: mitogen-activated protein kinase; MASC: mammary analogue secretory carcinoma; MEC: mucoepidermoid carcinoma; ML: machine learning; MRI: magnetic resonance imaging; MYE: myoepithelioma; NAD: nicotinamide adenine dinucleotide; NGS: next-generation sequencing; NMC: NUT midline carcinoma; NSCLC: non-small cell lung cancer; NTD: N-terminus domain; OS: overall survival; PA: pleomorphic adenoma; PAC: polymorphous adenocarcinoma; PPIN: protein–protein interaction network; PTCL: peripheral T-cell lymphoma; RF: random forests; SCCHN: squamous cell carcinomas of head and neck; SAHA: vorinostat; scRNA-Seq: single-cell RNA sequencing; SDC: salivary duct carcinoma; SGDA: salivary gland ductal adenocarcinoma; SGT: salivary gland tumor; SVMs: support vector machines; TCGA: The Cancer Genome Atlas; TJ: tight junction; TMA: tissue microarray; TSA: trichostatin A; UBB: ubiquitin gene; VPA: valproic acid; WT: Warthin’s.

References

  1. Cunha, J.L.S.; Hernandez-Guerrero, J.C.; de Almeida, O.P.; Soares, C.D.; Mosqueda-Taylor, A. Salivary Gland Tumors: A Retrospective Study of 164 Cases from a Single Private Practice Service in Mexico and Literature Review. Head Neck Pathol. 2021, 15, 523–531. [Google Scholar] [CrossRef] [PubMed]
  2. Gao, M.; Hao, Y.; Huang, M.X.; Ma, D.Q.; Chen, Y.; Luo, H.Y.; Gao, Y.; Cao, Z.Q.; Peng, X.; Yu, G.Y. Salivary gland tumours in a northern Chinese population: A 50-year retrospective study of 7190 cases. Int. J. Oral Maxillofac. Surg. 2017, 46, 343–349. [Google Scholar] [CrossRef] [PubMed]
  3. Sentani, K.; Ogawa, I.; Ozasa, K.; Sadakane, A.; Utada, M.; Tsuya, T.; Kajihara, H.; Yonehara, S.; Takeshima, Y.; Yasui, W. Characteristics of 5015 Salivary Gland Neoplasms Registered in the Hiroshima Tumor Tissue Registry over a Period of 39 Years. J. Clin. Med. 2019, 8, 566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Lawal, A.O.; Adisa, A.O.; Kolude, B.; Adeyemi, B.F.; Olajide, M.A. A review of 413 salivary gland tumours in the head and neck region. J. Clin. Exp. Dent. 2013, 5, e218–e222. [Google Scholar] [CrossRef] [Green Version]
  5. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. 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. [Google Scholar] [CrossRef]
  6. Rooper, L.M. Challenges in Minor Salivary Gland Biopsies: A Practical Approach to Problematic Histologic Patterns. Head Neck Pathol. 2019, 13, 476–484. [Google Scholar] [CrossRef]
  7. Skalova, A.; Hyrcza, M.D.; Leivo, I. Update from the 5th Edition of the World Health Organization Classification of Head and Neck Tumors: Salivary Glands. Head Neck Pathol. 2022, 16, 40–53. [Google Scholar] [CrossRef]
  8. da Silva, L.P.; Serpa, M.S.; Viveiros, S.K.; Sena, D.A.C.; de Carvalho Pinho, R.F.; de Abreu Guimaraes, L.D.; de Sousa Andrade, E.S.; Dias Pereira, J.R.; Silveira, M.; Sobral, A.P.V.; et al. Salivary gland tumors in a Brazilian population: A 20-year retrospective and multicentric study of 2292 cases. J. Craniomaxillofac. Surg. 2018, 46, 2227–2233. [Google Scholar] [CrossRef]
  9. Mariz, B.; do Socorro Queiroz Feio, P.; Roza, A.; de Andrade, B.A.B.; Agostini, M.; Romanach, M.J.; Fonseca, F.P.; Pontes, H.A.R.; Ribeiro, A.C.P.; Brandao, T.B.; et al. Clinical predictors of malignancy in palatal salivary gland tumors. Oral Dis. 2019, 25, 1919–1924. [Google Scholar] [CrossRef]
  10. Vielh, P.; Klijanienko, J. Salivary Gland Tumours; S. Karger AG: Basel, Switzerland, 2000; ISBN 978-3-318-00544-8. [Google Scholar] [CrossRef]
  11. Habermann, C.R.; Gossrau, P.; Graessner, J.; Arndt, C.; Cramer, M.C.; Reitmeier, F.; Jaehne, M.; Adam, G. Diffusion-weighted echo-planar MRI: A valuable tool for differentiating primary parotid gland tumors? Rofo 2005, 177, 940–945. [Google Scholar] [CrossRef]
  12. Skalova, A.; Leivo, I.; Hellquist, H.; Agaimy, A.; Simpson, R.H.W.; Stenman, G.; Vander Poorten, V.; Bishop, J.A.; Franchi, A.; Hernandez-Prera, J.C.; et al. High-grade Transformation/Dedifferentiation in Salivary Gland Carcinomas: Occurrence Across Subtypes and Clinical Significance. Adv. Anat. Pathol. 2021, 28, 107–118. [Google Scholar] [CrossRef]
  13. Alfieri, S.; Granata, R.; Bergamini, C.; Resteghini, C.; Bossi, P.; Licitra, L.F.; Locati, L.D. Systemic therapy in metastatic salivary gland carcinomas: A pathology-driven paradigm? Oral Oncol. 2017, 66, 58–63. [Google Scholar] [CrossRef]
  14. Hellquist, H.; Skalova, A.; Barnes, L.; Cardesa, A.; Thompson, L.D.; Triantafyllou, A.; Williams, M.D.; Devaney, K.O.; Gnepp, D.R.; Bishop, J.A.; et al. Cervical Lymph Node Metastasis in High-Grade Transformation of Head and Neck Adenoid Cystic Carcinoma: A Collective International Review. Adv. Ther. 2016, 33, 357–368. [Google Scholar] [CrossRef] [Green Version]
  15. Gatta, G.; Guzzo, M.; Locati, L.D.; McGurk, M.; Prott, F.J. Major and minor salivary gland tumours. Crit. Rev. Oncol. Hematol. 2020, 152, 102959. [Google Scholar] [CrossRef]
  16. Herman, M.P.; Werning, J.W.; Morris, C.G.; Kirwan, J.M.; Amdur, R.J.; Mendenhall, W.M. Elective neck management for high-grade salivary gland carcinoma. Am. J. Otolaryngol. 2013, 34, 205–208. [Google Scholar] [CrossRef] [PubMed]
  17. Saarilahti, K.; Kouri, M.; Collan, J.; Hamalainen, T.; Atula, T.; Joensuu, H.; Tenhunen, M. Intensity modulated radiotherapy for head and neck cancer: Evidence for preserved salivary gland function. Radiother. Oncol. 2005, 74, 251–258. [Google Scholar] [CrossRef] [PubMed]
  18. Sato, M.; Yamamoto, H.; Hatanaka, Y.; Nishijima, T.; Jiromaru, R.; Yasumatsu, R.; Taguchi, K.; Masuda, M.; Nakagawa, T.; Oda, Y. Wnt/beta-catenin signal alteration and its diagnostic utility in basal cell adenoma and histologically similar tumors of the salivary gland. Pathol. Res. Pract. 2018, 214, 586–592. [Google Scholar] [CrossRef] [PubMed]
  19. Wemmert, S.; Willnecker, V.; Sauter, B.; Schuh, S.; Brunner, C.; Bohle, R.M.; Urbschat, S.; Schick, B. Genomic alterations in Warthin tumors of the parotid gland. Oncol. Rep. 2014, 31, 1899–1904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Mito, J.K.; Jo, V.Y.; Chiosea, S.I.; Dal Cin, P.; Krane, J.F. HMGA2 is a specific immunohistochemical marker for pleomorphic adenoma and carcinoma ex-pleomorphic adenoma. Histopathology 2017, 71, 511–521. [Google Scholar] [CrossRef]
  21. Stenman, G. Fusion oncogenes in salivary gland tumors: Molecular and clinical consequences. Head Neck Pathol. 2013, 7 (Suppl. 1), S12–S19. [Google Scholar] [CrossRef]
  22. Skalova, A.; Vanecek, T.; Simpson, R.H.; Vazmitsel, M.A.; Majewska, H.; Mukensnabl, P.; Hauer, L.; Andrle, P.; Hosticka, L.; Grossmann, P.; et al. CRTC1-MAML2 and CRTC3-MAML2 fusions were not detected in metaplastic Warthin tumor and metaplastic pleomorphic adenoma of salivary glands. Am. J. Surg. Pathol. 2013, 37, 1743–1750. [Google Scholar] [CrossRef] [PubMed]
  23. Seethala, R.R.; Dacic, S.; Cieply, K.; Kelly, L.M.; Nikiforova, M.N. A reappraisal of the MECT1/MAML2 translocation in salivary mucoepidermoid carcinomas. Am. J. Surg. Pathol. 2010, 34, 1106–1121. [Google Scholar] [CrossRef] [PubMed]
  24. 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] [Green Version]
  25. Yin, L.X.; Ha, P.K. Genetic alterations in salivary gland cancers. Cancer 2016, 122, 1822–1831. [Google Scholar] [CrossRef] [Green Version]
  26. Andreasen, S.; Varma, S.; Barasch, N.; Thompson, L.D.R.; Miettinen, M.; Rooper, L.; Stelow, E.B.; Agander, T.K.; Seethala, R.R.; Chiosea, S.I.; 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. [Google Scholar] [CrossRef] [PubMed]
  27. Andersson, M.K.; Stenman, G. The landscape of gene fusions and somatic mutations in salivary gland neoplasms—Implications for diagnosis and therapy. Oral Oncol. 2016, 57, 63–69. [Google Scholar] [CrossRef]
  28. Steiner, P.; Andreasen, S.; Grossmann, P.; Hauer, L.; Vanecek, T.; Miesbauerova, M.; Santana, T.; Kiss, K.; Slouka, D.; Skalova, A. Prognostic significance of 1p36 locus deletion in adenoid cystic carcinoma of the salivary glands. Virchows Arch. 2018, 473, 471–480. [Google Scholar] [CrossRef]
  29. Papadaki, H.; Finkelstein, S.D.; Kounelis, S.; Bakker, A.; Swalsky, P.A.; Kapadia, S.B. The role of p53 mutation and protein expression in primary and recurrent adenoid cystic carcinoma. Hum. Pathol. 1996, 27, 567–572. [Google Scholar] [CrossRef]
  30. Skalova, A.; Vanecek, T.; Sima, R.; Laco, J.; Weinreb, I.; Perez-Ordonez, B.; Starek, I.; Geierova, M.; Simpson, R.H.; Passador-Santos, F.; 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. [Google Scholar] [CrossRef]
  31. Chapman, E.; Skalova, A.; Ptakova, N.; Martinek, P.; Goytain, A.; Tucker, T.; Xiong, W.; Leader, M.; Kudlow, B.A.; Haimes, J.D.; 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. Pathol. 2018, 42, 1182–1189. [Google Scholar] [CrossRef]
  32. Garcia-Manero, G.; Issa, J.P. Histone deacetylase inhibitors: A review of their clinical status as antineoplastic agents. Cancer Investig. 2005, 23, 635–642. [Google Scholar] [CrossRef] [PubMed]
  33. Toyota, M.; Issa, J.P. Epigenetic changes in solid and hematopoietic tumors. Semin. Oncol. 2005, 32, 521–530. [Google Scholar] [CrossRef] [PubMed]
  34. Khalele, B.A. Systematic review of mammary analog secretory carcinoma of salivary glands at 7 years after description. Head Neck 2017, 39, 1243–1248. [Google Scholar] [CrossRef]
  35. Antonescu, C.R.; Katabi, N.; Zhang, L.; Sung, Y.S.; Seethala, R.R.; Jordan, R.C.; Perez-Ordoñez, B.; Have, C.; Asa, S.L.; Leong, I.T. EWSR1-ATF1 fusion is a novel and consistent finding in hyalinizing clear-cell carcinoma of salivary gland. Genes Chromosomes Cancer 2011, 50, 559–570. [Google Scholar] [CrossRef]
  36. Skalova, A.; Vanecek, T.; Uro-Coste, E.; Bishop, J.A.; Weinreb, I.; Thompson, L.D.R.; de Sanctis, S.; Schiavo-Lena, M.; Laco, J.; Badoual, C.; 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. [Google Scholar] [CrossRef]
  37. Bishop, J.A.; Weinreb, I.; Swanson, D.; Westra, W.H.; Qureshi, H.S.; Sciubba, J.; MacMillan, C.; Rooper, L.M.; Dickson, B.C. Microsecretory Adenocarcinoma: A Novel Salivary Gland Tumor Characterized by a Recurrent MEF2C-SS18 Fusion. Am. J. Surg. Pathol. 2019, 43, 1023–1032. [Google Scholar] [CrossRef]
  38. de Jager, V.D.; de Visscher, S.A.; Schuuring, E.; Doff, J.J.; van Kempen, L.C. A novel PPP2R2A:: PRKD1 fusion in a cribriform adenocarcinoma of salivary gland. Genes Chromosomes Cancer 2023, 62, 297–300. [Google Scholar] [CrossRef]
  39. Jo, V.Y.; Sholl, L.M.; Krane, J.F. Distinctive Patterns of CTNNB1 (beta-Catenin) Alterations in Salivary Gland Basal Cell Adenoma and Basal Cell Adenocarcinoma. Am. J. Surg. Pathol. 2016, 40, 1143–1150. [Google Scholar] [CrossRef]
  40. Wilson, T.C.; Ma, D.; Tilak, A.; Tesdahl, B.; Robinson, R.A. Next-generation sequencing in salivary gland basal cell adenocarcinoma and basal cell adenoma. Head Neck Pathol. 2016, 10, 494–500. [Google Scholar]
  41. de Brito, B.S.; Giovanelli, N.; Egal, E.S.; Sanchez-Romero, C.; Nascimento, J.S.; Martins, A.S.; Tincani, A.J.; Del Negro, A.; Gondak, R.O.; Almeida, O.P.; et al. Loss of expression of Plag1 in malignant transformation from pleomorphic adenoma to carcinoma ex pleomorphic adenoma. Hum. Pathol. 2016, 57, 152–159. [Google Scholar] [CrossRef] [PubMed]
  42. Owosho, A.A.; Adesina, O.M.; Odujoko, O.; Akinyemi, H.; Komolafe, A.; Tadros, S.; Bauer, R.; Summersgill, K.F. HMGA2 Immunoexpression is frequent in salivary gland pleomorphic adenoma: Immunohistochemical and molecular analyses of PLAG1 and HMGA2 in 25 cases. Int. J. Clin. Exp. Pathol. 2022, 15, 63–71. [Google Scholar]
  43. Lee, Y.-H.; Huang, W.-C.; Hsieh, M.-S. CTNNB1 mutations in basal cell adenoma of the salivary gland. J. Formos. Med. Assoc. 2018, 117, 894–901. [Google Scholar] [PubMed]
  44. Waddington, C.H. The epigenotype. 1942. Int. J. Epidemiol. 2012, 41, 10–13. [Google Scholar] [CrossRef] [Green Version]
  45. Sharma, S.; Kelly, T.K.; Jones, P.A. Epigenetics in cancer. Carcinogenesis 2010, 31, 27–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Williams, M.D.; Chakravarti, N.; Kies, M.S.; Maruya, S.; Myers, J.N.; Haviland, J.C.; Weber, R.S.; Lotan, R.; El-Naggar, A.K. Implications of methylation patterns of cancer genes in salivary gland tumors. Clin. Cancer Res. 2006, 12, 7353–7358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Schache, A.G.; Hall, G.; Woolgar, J.A.; Nikolaidis, G.; Triantafyllou, A.; Lowe, D.; Risk, J.M.; Shaw, R.J.; Liloglou, T. Quantitative promoter methylation differentiates carcinoma ex pleomorphic adenoma from pleomorphic salivary adenoma. Br. J. Cancer 2010, 103, 1846–1851. [Google Scholar] [CrossRef]
  48. Hu, Y.H.; Zhang, C.Y.; Tian, Z.; Wang, L.Z.; Li, J. Aberrant protein expression and promoter methylation of p16 gene are correlated with malignant transformation of salivary pleomorphic adenoma. Arch. Pathol. Lab. Med. 2011, 135, 882–889. [Google Scholar] [CrossRef]
  49. Xia, L.; Hu, Y.; Gu, T.; Wang, L.; Tian, Z. Promoter hypermethylation may contribute to E-cadherin repression in the human salivary carcinoma ex pleomorphic adenoma. Int. J. Oncol. 2018, 52, 496–504. [Google Scholar] [CrossRef] [Green Version]
  50. Nikolic, N.; Carkic, J.; Ilic Dimitrijevic, I.; Eljabo, N.; Radunovic, M.; Anicic, B.; Tanic, N.; Falk, M.; Milasin, J. P14 methylation: An epigenetic signature of salivary gland mucoepidermoid carcinoma in the Serbian population. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2018, 125, 52–58. [Google Scholar] [CrossRef] [Green Version]
  51. Guo, X.L.; Sun, S.Z.; Wang, W.X.; Wei, F.C.; Yu, H.B.; Ma, B.L. Alterations of p16INK4a tumour suppressor gene in mucoepidermoid carcinoma of the salivary glands. Int. J. Oral Maxillofac. Surg. 2007, 36, 350–353. [Google Scholar] [CrossRef]
  52. Sasahira, T.; Kurihara, M.; Yamamoto, K.; Bhawal, U.K.; Kirita, T.; Kuniyasu, H. Downregulation of runt-related transcription factor 3 associated with poor prognosis of adenoid cystic and mucoepidermoid carcinomas of the salivary gland. Cancer Sci. 2011, 102, 492–497. [Google Scholar] [CrossRef] [PubMed]
  53. Maruya, S.; Kurotaki, H.; Shimoyama, N.; Kaimori, M.; Shinkawa, H.; Yagihashi, S. Expression of p16 protein and hypermethylation status of its promoter gene in adenoid cystic carcinoma of the head and neck. ORL J. Otorhinolaryngol. Relat. Spec. 2003, 65, 26–32. [Google Scholar] [CrossRef]
  54. Li, J.; El-Naggar, A.; Mao, L. Promoter methylation of p16INK4a, RASSF1A, and DAPK is frequent in salivary adenoid cystic carcinoma. Cancer 2005, 104, 771–776. [Google Scholar] [CrossRef] [PubMed]
  55. Daa, T.; Kashima, K.; Kondo, Y.; Yada, N.; Suzuki, M.; Yokoyama, S. Aberrant methylation in promoter regions of cyclin-dependent kinase inhibitor genes in adenoid cystic carcinoma of the salivary gland. APMIS 2008, 116, 21–26. [Google Scholar] [CrossRef] [PubMed]
  56. Kanazawa, T.; Misawa, K.; Fukushima, H.; Misawa, Y.; Sato, Y.; Maruta, M.; Imayoshi, S.; Kusaka, G.; Kawabata, K.; Mineta, H. Epigenetic inactivation of galanin receptors in salivary duct carcinoma of the parotid gland: Potential utility as biomarkers for prognosis. Oncol. Lett. 2018, 15, 9043–9050. [Google Scholar] [CrossRef] [Green Version]
  57. Wang, Z.; Ling, S.; Rettig, E.; Sobel, R.; Tan, M.; Fertig, E.J.; Considine, M.; El-Naggar, A.K.; Brait, M.; Fakhry, C. Epigenetic screening of salivary gland mucoepidermoid carcinoma identifies hypomethylation of CLIC3 as a common alteration. Oral Oncol. 2015, 51, 1120. [Google Scholar] [CrossRef] [Green Version]
  58. Shieh, Y.S.; Shiah, S.G.; Jeng, H.H.; Lee, H.S.; Wu, C.W.; Chang, L.C. DNA methyltransferase 1 expression and promoter methylation of E-cadherin in mucoepidermoid carcinoma. Cancer 2005, 104, 1013–1021. [Google Scholar]
  59. Zhang, C.Y.; Mao, L.; Li, L.; Tian, Z.; Zhou, X.J.; Zhang, Z.Y.; Li, J. Promoter methylation as a common mechanism for inactivating E-cadherin in human salivary gland adenoid cystic carcinoma. Cancer 2007, 110, 87–95. [Google Scholar] [CrossRef]
  60. Bell, D.; Bell, A.H.; Bondaruk, J.; Hanna, E.Y.; Weber, R.S. In-depth characterization of the salivary adenoid cystic carcinoma transcriptome with emphasis on dominant cell type. Cancer 2016, 122, 1513–1522. [Google Scholar] [CrossRef] [Green Version]
  61. Bell, D.; Bell, A.; Roberts, D.; Weber, R.S.; El-Naggar, A.K. Developmental transcription factor EN1--a novel biomarker in human salivary gland adenoid cystic carcinoma. Cancer 2012, 118, 1288–1292. [Google Scholar] [CrossRef] [Green Version]
  62. Baba, S.; Akashi, T.; Kayamori, K.; Ohuchi, T.; Ogawa, I.; Kubota, N.; Nakano, K.; Nagatsuka, H.; Hasegawa, H.; Matsuzaka, K. Homeobox transcription factor engrailed homeobox 1 is a possible diagnostic marker for adenoid cystic carcinoma and polymorphous adenocarcinoma. Pathol. Int. 2021, 71, 113–123. [Google Scholar] [CrossRef]
  63. Zhang, C.-Y.; Zhao, Y.-X.; Xia, R.-H.; Han, J.; Wang, B.-S.; Tian, Z.; Wang, L.-Z.; Hu, Y.-H.; Li, J. RASSF1A promoter hypermethylation is a strong biomarker of poor survival in patients with salivary adenoid cystic carcinoma in a Chinese population. PLoS ONE 2014, 9, e110159. [Google Scholar]
  64. Xia, R.; Zhou, R.; Tian, Z.; Zhang, C.; Wang, L.; Hu, Y.; Han, J.; Li, J. High expression of H3K9me3 is a strong predictor of poor survival in patients with salivary adenoid cystic carcinoma. Arch. Pathol. Lab. Med. 2013, 137, 1761–1769. [Google Scholar] [CrossRef]
  65. Shao, C.; Sun, W.; Tan, M.; Glazer, C.A.; Bhan, S.; Zhong, X.; Fakhry, C.; Sharma, R.; Westra, W.H.; Hoque, M.O. Integrated, Genome-Wide Screening for Hypomethylated Oncogenes in Salivary Gland Adenoid Cystic CarcinomaScreening for Hypomethylated Oncogenes in Salivary Gland ACC. Clin. Cancer Res. 2011, 17, 4320–4330. [Google Scholar] [PubMed] [Green Version]
  66. Uchida, D.; Begum, N.; Almofti, A.; Kawamata, H.; Yoshida, H.; Sato, M. Frequent downregulation of 14-3-3 sigma protein and hypermethylation of 14-3-3 sigma gene in salivary gland adenoid cystic carcinoma. Br. J. Cancer 2004, 91, 1131–1138. [Google Scholar] [CrossRef] [Green Version]
  67. Lee, E.-S.; Issa, J.-P.; Roberts, D.B.; Williams, M.D.; Weber, R.S.; Kies, M.S.; El-Naggar, A.K. Quantitative promoter hypermethylation analysis of cancer-related genes in salivary gland carcinomas: Comparison with methylation-specific PCR technique and clinical significance. Clin. Cancer Res. 2008, 14, 2664–2672. [Google Scholar]
  68. Aniello, F.; Colella, G.; Muscariello, G.; Lanza, A.; Ferrara, D.; Branno, M.; Minucci, S. Expression of four histone lysine-methyltransferases in parotid gland tumors. Anticancer. Res. 2006, 26, 2063–2067. [Google Scholar]
  69. Wagner, V.P.; Martins, M.D.; Guimaraes, D.M.; Vasconcelos, A.C.; Meurer, L.; Vargas, P.A.; Fonseca, F.P.; Squarize, C.H.; Castilho, R.M. Reduced chromatin acetylation of malignant salivary gland tumors correlates with enhanced proliferation. J. Oral Pathol. Med. 2017, 46, 792–797. [Google Scholar] [CrossRef] [PubMed]
  70. Mat Lazim, N.; Yousaf, A.; Abusalah, M.A.H.; Sulong, S.; Mohd Ismail, Z.I.; Mohamud, R.; Abu-Harirah, H.A.; AlRamadneh, T.N.; Hassan, R.; Abdullah, B. The Epigenesis of Salivary Glands Carcinoma: From Field Cancerization to Carcinogenesis. Cancers 2023, 15, 2111. [Google Scholar]
  71. Lam-Ubol, A.; Phattarataratip, E. Distinct histone H3 modification profiles correlate with aggressive characteristics of salivary gland neoplasms. Sci. Rep. 2022, 12, 15063. [Google Scholar] [CrossRef] [PubMed]
  72. Nandakumar, V.; Vaid, M.; Katiyar, S.K. (-)-Epigallocatechin-3-gallate reactivates silenced tumor suppressor genes, Cip1/p21 and p16INK4a, by reducing DNA methylation and increasing histones acetylation in human skin cancer cells. Carcinogenesis 2011, 32, 537–544. [Google Scholar] [CrossRef] [Green Version]
  73. Martin, C.; Chen, S.; Heilos, D.; Sauer, G.; Hunt, J.; Shaw, A.G.; Sims, P.F.; Jackson, D.A.; Lovric, J. Changed genome heterochromatinization upon prolonged activation of the Raf/ERK signaling pathway. PLoS ONE 2010, 5, e13322. [Google Scholar] [CrossRef] [Green Version]
  74. Mysliwiec, M.R.; Carlson, C.D.; Tietjen, J.; Hung, H.; Ansari, A.Z.; Lee, Y. Jarid2 (Jumonji, AT rich interactive domain 2) regulates NOTCH1 expression via histone modification in the developing heart. J. Biol. Chem. 2012, 287, 1235–1241. [Google Scholar] [CrossRef] [Green Version]
  75. Stephens, P.J.; Davies, H.R.; Mitani, Y.; Van Loo, P.; Shlien, A.; Tarpey, P.S.; Papaemmanuil, E.; Cheverton, A.; Bignell, G.R.; Butler, A.P.; et al. Whole exome sequencing of adenoid cystic carcinoma. J. Clin. Investig. 2013, 123, 2965–2968. [Google Scholar] [CrossRef]
  76. Lu, H.; Han, N.; Xu, W.; Zhu, Y.; Liu, L.; Liu, S.; Yang, W. Screening and bioinformatics analysis of mRNA, long non-coding RNA and circular RNA expression profiles in mucoepidermoid carcinoma of salivary gland. Biochem. Biophys. Res. Commun. 2019, 508, 66–71. [Google Scholar] [CrossRef]
  77. Chen, Z.; Lin, S.; Li, J.L.; Ni, W.; Guo, R.; Lu, J.; Kaye, F.J.; Wu, L. CRTC1-MAML2 fusion-induced lncRNA LINC00473 expression maintains the growth and survival of human mucoepidermoid carcinoma cells. Oncogene 2018, 37, 1885–1895. [Google Scholar] [CrossRef]
  78. Binmadi, N.O.; Basile, J.R.; Perez, P.; Gallo, A.; Tandon, M.; Elias, W.; Jang, S.I.; Alevizos, I. miRNA expression profile of mucoepidermoid carcinoma. Oral Dis. 2018, 24, 537–543. [Google Scholar] [CrossRef]
  79. Flores, B.C.; Lourenco, S.V.; Damascena, A.S.; Kowaslki, L.P.; Soares, F.A.; Coutinho-Camillo, C.M. Altered expression of apoptosis-regulating miRNAs in salivary gland tumors suggests their involvement in salivary gland tumorigenesis. Virchows. Arch. 2017, 470, 291–299. [Google Scholar] [CrossRef]
  80. Xu, W.; Liu, L.; Lu, H.; Fu, J.; Zhang, C.; Yang, W.; Shen, S. Dysregulated long non-coding RNAs in pleomorphic adenoma tissues of pleomorphic adenoma gene 1 transgenic mice. Mol. Med. Rep. 2019, 19, 4735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Mitani, Y.; Roberts, D.B.; Fatani, H.; Weber, R.S.; Kies, M.S.; Lippman, S.M.; El-Naggar, A.K. MicroRNA profiling of salivary adenoid cystic carcinoma: Association of miR-17-92 upregulation with poor outcome. PLoS ONE 2013, 8, e66778. [Google Scholar]
  82. Ramírez-Martínez, C.M.; Jacinto-Alemán, L.F.; Cruz-Hervert, L.P.; Portilla-Robertson, J.; Leyva-Huerta, E.R. Bioinformatic Analysis for Mucoepidermoid and Adenoid Cystic Carcinoma of Therapeutic Targets. Vaccines 2022, 10, 1557. [Google Scholar] [CrossRef]
  83. Liu, J.; Shao, C.; Tan, M.L.; Mu, D.; Ferris, R.L.; Ha, P.K. Molecular biology of adenoid cystic carcinoma. Head Neck 2012, 34, 1665–1677. [Google Scholar] [CrossRef]
  84. Zhou, X.; Huang, S.; Zhang, D.; Zhang, S.; Li, W.; Chen, Z.; Wu, H. Effects of 5-aza-2′ deoxycytidine on RECK gene expression and tumor invasion in salivary adenoid cystic carcinoma. Braz. J. Med. Biol. Res. 2015, 48, 254. [Google Scholar] [CrossRef] [Green Version]
  85. Nikolic, N.; Carkic, J.; Jacimovic, J.; Jakovljevic, A.; Anicic, B.; Jezdic, Z.; Milasin, J. Methylation of tumour suppressor genes in benign and malignant salivary gland tumours: A systematic review and meta-analysis. Epigenetics. 2022, 17, 1661–1676. [Google Scholar] [CrossRef]
  86. Camuzi, D.; Simão, T.A.; Dias, F.; Ribeiro Pinto, L.F.; Soares-Lima, S.C. Head and Neck Cancers Are Not Alike When Tarred with the Same Brush: An Epigenetic Perspective from the Cancerization Field to Prognosis. Cancers. 2021, 13, 5630. [Google Scholar] [CrossRef]
  87. Huang, C.G.; Li, M.Z.; Wang, S.H.; Liu, Y.; Zhang, H.L.; Haybaeck, J.; Yang, Z.H. Analysis of Cytological Misdiagnosis and Oversight of Adenoid Cystic Carcinoma of Salivary Gland. Cancer Control. 2023, 30, 10732748221131652. [Google Scholar] [CrossRef] [PubMed]
  88. Nishida, H.; Kusaba, T.; Kawamura, K.; Oyama, Y.; Daa, T. Histopathological Aspects of the Prognostic Factors for Salivary Gland Cancers. Cancers. 2023, 15, 1236. [Google Scholar] [CrossRef]
  89. Kiwerska, K.; Kowal-Wisniewska, E.; Ustaszewski, A.; Bartkowiak, E.; Jarmuz-Szymczak, M.; Wierzbicka, M.; Giefing, M. Global DNA Methylation Profiling Reveals Differentially Methylated CpGs between Salivary Gland Pleomorphic Adenomas with Distinct Clinical Course. Int. J. Mol. Sci. 2022, 23, 5962. [Google Scholar] [CrossRef] [PubMed]
  90. Xia, C.; Tao, Y.; Li, M.; Che, T.; Qu, J. Protein acetylation and deacetylation: An important regulatory modification in gene transcription (Review). Exp. Ther. Med. 2020, 20, 2923–2940. [Google Scholar] [CrossRef] [PubMed]
  91. Meier, K.; Brehm, A. Chromatin regulation: How complex does it get? Epigenetics 2014, 9, 1485–1495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Seto, E.; Yoshida, M. Erasers of histone acetylation: The histone deacetylase enzymes. Cold Spring Harb. Perspect. Biol. 2014, 6, a018713. [Google Scholar] [CrossRef] [Green Version]
  93. Ruijter, A.J.M.d.; Gennip, A.H.v.; Caron, H.N.; Kemp, S.; Kuilenburg, A.B.P.v. Histone deacetylases (HDACs): Characterization of the classical HDAC family. Biochem. J. 2003, 370, 737–749. [Google Scholar] [CrossRef] [Green Version]
  94. Haberland, M.; Montgomery, R.L.; Olson, E.N. The many roles of histone deacetylases in development and physiology: Implications for disease and therapy. Nat. Rev. Genet. 2009, 10, 32–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Bruhn, C.; Bastianello, G.; Foiani, M. Cancer cell histone density links global histone acetylation, mitochondrial proteome and histone acetylase inhibitor sensitivity. Commun Biol. 2022, 5, 882. [Google Scholar] [CrossRef]
  96. Rettig, I.; Koeneke, E.; Trippel, F.; Mueller, W.C.; Burhenne, J.; Kopp-Schneider, A.; Fabian, J.; Schober, A.; Fernekorn, U.; von Deimling, A.; et al. Selective inhibition of HDAC8 decreases neuroblastoma growth in vitro and in vivo and enhances retinoic acid-mediated differentiation. Cell Death Dis. 2015, 6, e1657. [Google Scholar] [CrossRef] [Green Version]
  97. Cuttini, E.; Goi, C.; Pellarin, E.; Vida, R.; Brancolini, C. HDAC4 in cancer: A multitasking platform to drive not only epigenetic modifications. Front. Mol. Biosci. 2023, 10, 24. [Google Scholar] [CrossRef] [PubMed]
  98. Milde, T.; Oehme, I.; Korshunov, A.; Kopp-Schneider, A.; Remke, M.; Northcott, P.; Deubzer, H.E.; Lodrini, M.; Taylor, M.D.; Von Deimling, A. HDAC5 and HDAC9 in Medulloblastoma: Novel Markers for Risk Stratification and Role in Tumor Cell GrowthHDAC5 and HDAC9 in Medulloblastoma. Clin. Cancer Res. 2010, 16, 3240–3252. [Google Scholar] [CrossRef] [Green Version]
  99. Deskin, B.; Lasky, J.; Zhuang, Y.; Shan, B. Requirement of HDAC6 for activation of Notch1 by TGF-beta1. Sci. Rep. 2016, 6, 31086. [Google Scholar] [CrossRef]
  100. Yang, Y.; Huang, Y.; Wang, Z.; Wang, H.T.; Duan, B.; Ye, D.; Wang, C.; Jing, R.; Leng, Y.; Xi, J.; et al. HDAC10 promotes lung cancer proliferation via AKT phosphorylation. Oncotarget 2016, 7, 59388–59401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Huffman, D.M.; Grizzle, W.E.; Bamman, M.M.; Kim, J.S.; Eltoum, I.A.; Elgavish, A.; Nagy, T.R. SIRT1 is significantly elevated in mouse and human prostate cancer. Cancer Res. 2007, 67, 6612–6618. [Google Scholar] [CrossRef] [Green Version]
  102. Bradbury, C.A.; Khanim, F.L.; Hayden, R.; Bunce, C.M.; White, D.A.; Drayson, M.T.; Craddock, C.; Turner, B.M. Histone deacetylases in acute myeloid leukaemia show a distinctive pattern of expression that changes selectively in response to deacetylase inhibitors. Leukemia 2005, 19, 1751–1759. [Google Scholar] [CrossRef] [Green Version]
  103. Singh, A.K.; Bishayee, A.; Pandey, A.K. Targeting Histone Deacetylases with Natural and Synthetic Agents: An Emerging Anticancer Strategy. Nutrients 2018, 10, 731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Ashraf, N.; Zino, S.; Macintyre, A.; Kingsmore, D.; Payne, A.P.; George, W.D.; Shiels, P.G. Altered sirtuin expression is associated with node-positive breast cancer. Br. J. Cancer 2006, 95, 1056–1061. [Google Scholar] [CrossRef] [Green Version]
  105. de Ceu Teixeira, M.; Sanchez-Lopez, E.; Espina, M.; Garcia, M.L.; Durazzo, A.; Lucarini, M.; Novellino, E.; Souto, S.B.; Santini, A.; Souto, E.B. Sirtuins and SIRT6 in Carcinogenesis and in Diet. Int. J. Mol. Sci. 2019, 20, 4945. [Google Scholar] [CrossRef] [Green Version]
  106. Chang, H.H.; Chiang, C.P.; Hung, H.C.; Lin, C.Y.; Deng, Y.T.; Kuo, M.Y. Histone deacetylase 2 expression predicts poorer prognosis in oral cancer patients. Oral Oncol. 2009, 45, 610–614. [Google Scholar] [CrossRef]
  107. Wawruszak, A.; Kalafut, J.; Okon, E.; Czapinski, J.; Halasa, M.; Przybyszewska, A.; Miziak, P.; Okla, K.; Rivero-Muller, A.; Stepulak, A. Histone Deacetylase Inhibitors and Phenotypical Transformation of Cancer Cells. Cancers 2019, 11, 148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Chau, N.G.; Hurwitz, S.; Mitchell, C.M.; Aserlind, A.; Grunfeld, N.; Kaplan, L.; Hsi, P.; Bauer, D.E.; Lathan, C.S.; Rodriguez-Galindo, C.; et al. Intensive treatment and survival outcomes in NUT midline carcinoma of the head and neck. Cancer 2016, 122, 3632–3640. [Google Scholar] [CrossRef] [PubMed]
  109. Esteves, G.; Ferreira, J.; Afonso, R.; Martins, C.; Zagalo, C.; Felix, A. HDAC Overexpression in a NUT Midline Carcinoma of the Parotid Gland with Exceptional Survival: A Case Report. Head Neck Pathol. 2020, 14, 1117–1122. [Google Scholar] [CrossRef] [PubMed]
  110. 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]
  111. Zhao, S.; Guo, J.; Zhao, Y.; Fei, C.; Zheng, Q.; Li, X.; Chang, C. Chidamide, a novel histone deacetylase inhibitor, inhibits the viability of MDS and AML cells by suppressing JAK2/STAT3 signaling. Am. J. Transl. Res. 2016, 8, 3169–3178. [Google Scholar]
  112. Dong, M.; Ning, Z.Q.; Xing, P.Y.; Xu, J.L.; Cao, H.X.; Dou, G.F.; Meng, Z.Y.; Shi, Y.K.; Lu, X.P.; Feng, F.Y. Phase I study of chidamide (CS055/HBI-8000), a new histone deacetylase inhibitor, in patients with advanced solid tumors and lymphomas. Cancer Chemother. Pharmacol. 2012, 69, 1413–1422. [Google Scholar] [CrossRef]
  113. Yang, S.; Nan, P.; Li, C.; Lin, F.; Li, H.; Wang, T.; Zhou, C.; Zhang, X.; Meng, X.; Qian, H.; et al. Inhibitory effect of chidamide on the growth of human adenoid cystic carcinoma cells. Biomed. Pharmacother. 2018, 99, 608–614. [Google Scholar] [CrossRef]
  114. Tsimberidou, A.M.; Beer, P.A.; Cartwright, C.A.; Haymaker, C.; Vo, H.H.; Kiany, S.; Cecil, A.R.L.; Dow, J.; Haque, K.; Silva, F.A.; et al. Preclinical Development and First-in-Human Study of KA2507, a Selective and Potent Inhibitor of Histone Deacetylase 6, for Patients with Refractory Solid Tumors. Clin. Cancer Res. 2021, 27, 3584–3594. [Google Scholar] [CrossRef]
  115. Ho, A.S.; Kannan, K.; Roy, D.M.; Morris, L.G.; Ganly, I.; Katabi, N.; Ramaswami, D.; Walsh, L.A.; Eng, S.; Huse, J.T.; et al. The mutational landscape of adenoid cystic carcinoma. Nat. Genet. 2013, 45, 791–798. [Google Scholar] [CrossRef]
  116. Ramalingam, S.S.; Kummar, S.; Sarantopoulos, J.; Shibata, S.; LoRusso, P.; Yerk, M.; Holleran, J.; Lin, Y.; Beumer, J.H.; Harvey, R.D.; et al. Phase I study of vorinostat in patients with advanced solid tumors and hepatic dysfunction: A National Cancer Institute Organ Dysfunction Working Group study. J. Clin. Oncol. 2010, 28, 4507–4512. [Google Scholar] [CrossRef] [Green Version]
  117. Goncalves, P.H.; Heilbrun, L.K.; Barrett, M.T.; Kummar, S.; Hansen, A.R.; Siu, L.L.; Piekarz, R.L.; Sukari, A.W.; Chao, J.; Pilat, M.J.; et al. A phase 2 study of vorinostat in locally advanced, recurrent, or metastatic adenoid cystic carcinoma. Oncotarget 2017, 8, 32918–32929. [Google Scholar] [CrossRef] [Green Version]
  118. Woods, D.M.; Sodré, A.L.; Villagra, A.; Sarnaik, A.; Sotomayor, E.M.; Weber, J. HDAC Inhibition Upregulates PD-1 Ligands in Melanoma and Augments Immunotherapy with PD-1 BlockadeHDAC Inhibition Upregulates PD-1 Ligands in Melanoma. Cancer Immunol. Res. 2015, 3, 1375–1385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Hellmann, M.D.; Janne, P.A.; Opyrchal, M.; Hafez, N.; Raez, L.E.; Gabrilovich, D.I.; Wang, F.; Trepel, J.B.; Lee, M.J.; Yuno, A.; et al. Entinostat plus Pembrolizumab in Patients with Metastatic NSCLC Previously Treated with Anti-PD-(L)1 Therapy. Clin. Cancer Res. 2021, 27, 1019–1028. [Google Scholar] [CrossRef] [PubMed]
  120. 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.; et al. Pembrolizumab for the Treatment of Advanced Salivary Gland Carcinoma: Findings of the Phase 1b KEYNOTE-028 Study. Am. J. Clin. Oncol. 2018, 41, 1083–1088. [Google Scholar] [CrossRef]
  121. Goncalves, P.H.; Kummar, S.; Siu, L.L.; Hansen, A.R.; Savvides, P.; Sukari, A.; Chao, J.; Heilbrun, L.K.; Pilat, M.J.; Smith, D.W.; et al. A phase II study of suberoylanilide hydroxamic acid (SAHA) in subjects with locally advanced, recurrent, or metastatic adenoid cystic carcinoma (ACC). J. Clin. Oncol. 2013, 31, 6045. [Google Scholar] [CrossRef]
  122. Rodriguez, C.P.; Wu, Q.V.; Voutsinas, J.; Fromm, J.R.; Jiang, X.; Pillarisetty, V.G.; Lee, S.M.; Santana-Davila, R.; Goulart, B.; Baik, C.S.; et al. A Phase II Trial of Pembrolizumab and Vorinostat in Recurrent Metastatic Head and Neck Squamous Cell Carcinomas and Salivary Gland Cancer. Clin. Cancer Res. 2020, 26, 837–845. [Google Scholar] [CrossRef]
  123. Chang, K.; Creighton, C.J.; Davis, C.; Donehower, L.; Drummond, J.; Wheeler, D.; Ally, A.; Balasundaram, M.; Birol, I.; Butterfield, Y.S.N.; et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 2013, 45, 1113–1120. [Google Scholar] [CrossRef]
  124. Edgar, R.; Domrachev, M.; Lash, A.E. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002, 30, 207–210. [Google Scholar] [CrossRef] [Green Version]
  125. Tang, F.; Barbacioru, C.; Wang, Y.; Nordman, E.; Lee, C.; Xu, N.; Wang, X.; Bodeau, J.; Tuch, B.B.; Siddiqui, A.; et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat. Methods 2009, 6, 377–382. [Google Scholar] [CrossRef]
  126. Wang, Z.; Gerstein, M.; Snyder, M. RNA-Seq: A revolutionary tool for transcriptomics. Nat. Rev. Genet. 2009, 10, 57–63. [Google Scholar] [CrossRef]
  127. Langfelder, P.; Horvath, S. WGCNA: An R package for weighted correlation network analysis. BMC Bioinform. 2008, 9, 559. [Google Scholar] [CrossRef] [Green Version]
  128. Huang, D.W.; Sherman, B.T.; Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009, 4, 44–57. [Google Scholar] [CrossRef]
  129. Szklarczyk, D.; Kirsch, R.; Koutrouli, M.; Nastou, K.; Mehryary, F.; Hachilif, R.; Gable, A.L.; Fang, T.; Doncheva, N.T.; Pyysalo, S.; et al. The STRING database in 2023: Protein-protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic Acids Res. 2023, 51, D638–D646. [Google Scholar] [CrossRef] [PubMed]
  130. Kourou, K.; Exarchos, T.P.; Exarchos, K.P.; Karamouzis, M.V.; Fotiadis, D.I. Machine learning applications in cancer prognosis and prediction. Comput. Struct. Biotechnol. J. 2015, 13, 8–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Hasin, Y.; Seldin, M.; Lusis, A. Multi-omics approaches to disease. Genome Biol. 2017, 18, 83. [Google Scholar] [CrossRef] [PubMed]
  132. Yoon, J.H.; Kim, D.W.; Lee, S.K.; Kim, S.G. Effects of 4-hexylresorcinol administration on the submandibular glands in a growing rat model. Head Face Med. 2022, 18, 16. [Google Scholar] [CrossRef] [PubMed]
  133. Kim, J.-Y.; Kweon, H.-Y.; Kim, D.-W.; Choi, J.-Y.; Kim, S.-G. 4-Hexylresorcinol Inhibits Class I Histone Deacetylases in Human Umbilical Cord Endothelial Cells. Appl. Sci. 2021, 11, 3486. [Google Scholar] [CrossRef]
  134. Qian, D.Z.; Kato, Y.; Shabbeer, S.; Wei, Y.; Verheul, H.M.; Salumbides, B.; Sanni, T.; Atadja, P.; Pili, R. Targeting tumor angiogenesis with histone deacetylase inhibitors: The hydroxamic acid derivative LBH589. Clin. Cancer Res. 2006, 12, 634–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Frohlich, L.F.; Mrakovcic, M.; Smole, C.; Zatloukal, K. Molecular mechanism leading to SAHA-induced autophagy in tumor cells: Evidence for a p53-dependent pathway. Cancer Cell Int. 2016, 16, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Chen, M.C.; Huang, H.H.; Lai, C.Y.; Lin, Y.J.; Liou, J.P.; Lai, M.J.; Li, Y.H.; Teng, C.M.; Yang, C.R. Novel histone deacetylase inhibitor MPT0G009 induces cell apoptosis and synergistic anticancer activity with tumor necrosis factor-related apoptosis-inducing ligand against human hepatocellular carcinoma. Oncotarget 2016, 7, 402–417. [Google Scholar] [CrossRef] [Green Version]
  137. Li, Y.; Liu, T.; Ivan, C.; Huang, J.; Shen, D.Y.; Kavanagh, J.J.; Bast, R.C., Jr.; Fu, S.; Hu, W.; Sood, A.K. Enhanced Cytotoxic Effects of Combined Valproic Acid and the Aurora Kinase Inhibitor VE465 on Gynecologic Cancer Cells. Front. Oncol. 2013, 3, 58. [Google Scholar] [CrossRef] [Green Version]
  138. Wu, P.; Tian, Y.; Chen, G.; Wang, B.; Gui, L.; Xi, L.; Ma, X.; Fang, Y.; Zhu, T.; Wang, D.; et al. Correction: Ubiquitin B: An essential mediator of trichostatin A-induced tumor-selective killing in human cancer cells. Cell Death Differ. 2022, 29, 1299. [Google Scholar] [CrossRef]
  139. Ramaiah, M.J.; Tangutur, A.D.; Manyam, R.R. Epigenetic modulation and understanding of HDAC inhibitors in cancer therapy. Life Sci. 2021, 277, 119504. [Google Scholar] [CrossRef]
  140. Kauh, J.; Fan, S.; Xia, M.; Yue, P.; Yang, L.; Khuri, F.R.; Sun, S.Y. c-FLIP degradation mediates sensitization of pancreatic cancer cells to TRAIL-induced apoptosis by the histone deacetylase inhibitor LBH589. PLoS ONE 2010, 5, e10376. [Google Scholar] [CrossRef] [Green Version]
  141. Liu, J.; Li, G.; Wang, X.; Wang, L.; Zhao, R.; Wang, J.; Kong, Y.; Ding, J.; Li, J.; Zhang, L. Droxinostat, a Histone Deacetylase Inhibitor, Induces Apoptosis in Hepatocellular Carcinoma Cell Lines via Activation of the Mitochondrial Pathway and Downregulation of FLIP. Transl. Oncol. 2016, 9, 70–78. [Google Scholar] [CrossRef] [Green Version]
  142. Chintakuntlawar, A.V.; Okuno, S.H.; Price, K.A. Systemic therapy for recurrent or metastatic salivary gland malignancies. Cancers Head Neck 2016, 1, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Ahn, M.Y.; Ahn, J.W.; Kim, H.S.; Lee, J.; Yoon, J.H. Apicidin inhibits cell growth by downregulating IGF-1R in salivary mucoepidermoid carcinoma cells. Oncol. Rep. 2015, 33, 1899–1907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. 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]
  145. Guimarães, D.M.; Almeida, L.O.; Martins, M.D.; Warner, K.A.; Silva, A.R.; Vargas, P.A.; Nunes, F.D.; Squarize, C.H.; Nör, J.E.; Castilho, R.M. Sensitizing mucoepidermoid carcinomas to chemotherapy by targeted disruption of cancer stem cells. Oncotarget 2016, 7, 42447. [Google Scholar] [CrossRef] [Green Version]
  146. Almeida, L.O.; Guimaraes, D.M.; Martins, M.D.; Martins, M.A.T.; Warner, K.A.; Nor, J.E.; Castilho, R.M.; Squarize, C.H. Unlocking the chromatin of adenoid cystic carcinomas using HDAC inhibitors sensitize cancer stem cells to cisplatin and induces tumor senescence. Stem. Cell Res. 2017, 21, 94–105. [Google Scholar] [CrossRef]
  147. Parag-Sharma, K.; Tasoulas, J.; Musicant, A.M.; do Nascimento-Filho, C.H.V.; Zhu, Z.; Twomey, C.; Liu, P.; Castilho, R.M.; Amelio, A.L. Synergistic efficacy of combined EGFR and HDAC inhibitors overcomes tolerance to EGFR monotherapy in salivary mucoepidermoid carcinoma. Oral Oncol. 2021, 115, 105166. [Google Scholar] [CrossRef] [PubMed]
  148. Jang, B.; Yang, I.H.; Cho, N.P.; Jin, B.; Lee, W.; Jung, Y.C.; Hong, S.D.; Shin, J.A.; Cho, S.D. Down-regulation and nuclear localization of survivin by sodium butyrate induces caspase-dependent apoptosis in human oral mucoepidermoid carcinoma. Oral Oncol. 2019, 88, 160–167. [Google Scholar] [CrossRef] [PubMed]
  149. Nakano, M.; Ohwada, K.; Shindo, Y.; Konno, T.; Kohno, T.; Kikuchi, S.; Tsujiwaki, M.; Ishii, D.; Nishida, S.; Kakuki, T.; et al. Inhibition of HDAC and Signal Transduction Pathways Induces Tight Junctions and Promotes Differentiation in p63-Positive Salivary Duct Adenocarcinoma. Cancers 2022, 14, 2584. [Google Scholar] [CrossRef]
  150. Talevi, A. Multi-target pharmacology: Possibilities and limitations of the “skeleton key approach” from a medicinal chemist perspective. Front. Pharmacol. 2015, 6, 205. [Google Scholar] [CrossRef] [Green Version]
  151. Lai, C.J.; Bao, R.; Tao, X.; Wang, J.; Atoyan, R.; Qu, H.; Wang, D.G.; Yin, L.; Samson, M.; Forrester, J.; et al. CUDC-101, a multitargeted inhibitor of histone deacetylase, epidermal growth factor receptor, and human epidermal growth factor receptor 2, exerts potent anticancer activity. Cancer Res. 2010, 70, 3647–3656. [Google Scholar] [CrossRef] [Green Version]
  152. Wang, J.; Pursell, N.W.; Samson, M.E.; Atoyan, R.; Ma, A.W.; Selmi, A.; Xu, W.; Cai, X.; Voi, M.; Savagner, P.; et al. Potential advantages of CUDC-101, a multitargeted HDAC, EGFR, and HER2 inhibitor, in treating drug resistance and preventing cancer cell migration and invasion. Mol. Cancer Ther. 2013, 12, 925–936. [Google Scholar] [CrossRef] [Green Version]
  153. Galloway, T.J.; Wirth, L.J.; Colevas, A.D.; Gilbert, J.; Bauman, J.E.; Saba, N.F.; Raben, D.; Mehra, R.; Ma, A.W.; Atoyan, R.; et al. A Phase I Study of CUDC-101, a Multitarget Inhibitor of HDACs, EGFR, and HER2, in Combination with Chemoradiation in Patients with Head and Neck Squamous Cell Carcinoma. Clin. Cancer Res. 2015, 21, 1566–1573. [Google Scholar] [CrossRef] [Green Version]
  154. Parra, M. Class IIa HDACs—New insights into their functions in physiology and pathology. FEBS J. 2015, 282, 1736–1744. [Google Scholar] [CrossRef]
  155. Yu, Y.; Cao, F.; Yu, X.; Zhou, P.; Di, Q.; Lei, J.; Tai, Y.; Wu, H.; Li, X.; Wang, X.; et al. The expression of HDAC7 in cancerous gastric tissues is positively associated with distant metastasis and poor patient prognosis. Clin. Transl. Oncol. 2017, 19, 1045–1054. [Google Scholar] [CrossRef] [PubMed]
  156. Wu, M.Y.; Fu, J.; Xiao, X.; Wu, J.; Wu, R.C. MiR-34a regulates therapy resistance by targeting HDAC1 and HDAC7 in breast cancer. Cancer Lett. 2014, 354, 311–319. [Google Scholar] [CrossRef] [PubMed]
  157. Barneda-Zahonero, B.; Collazo, O.; Azagra, A.; Fernandez-Duran, I.; Serra-Musach, J.; Islam, A.B.; Vega-Garcia, N.; Malatesta, R.; Camos, M.; Gomez, A.; et al. The transcriptional repressor HDAC7 promotes apoptosis and c-Myc downregulation in particular types of leukemia and lymphoma. Cell Death Dis. 2015, 6, e1635. [Google Scholar] [CrossRef] [Green Version]
  158. Yu, X.; Wang, M.; Wu, J.; Han, Q.; Zhang, X. Correction to: ZNF326 promotes malignant phenotype of glioma by up-regulating HDAC7 expression and activating Wnt pathway. J. Exp. Clin. Cancer Res. 2020, 39, 14. [Google Scholar] [CrossRef] [Green Version]
  159. Sang, Y.; Sun, L.; Wu, Y.; Yuan, W.; Liu, Y.; Li, S.W. Histone deacetylase 7 inhibits plakoglobin expression to promote lung cancer cell growth and metastasis. Int. J. Oncol. 2019, 54, 1112–1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Witt, A.E.; Lee, C.W.; Lee, T.I.; Azzam, D.J.; Wang, B.; Caslini, C.; Petrocca, F.; Grosso, J.; Jones, M.; Cohick, E.B.; et al. Identification of a cancer stem cell-specific function for the histone deacetylases, HDAC1 and HDAC7, in breast and ovarian cancer. Oncogene 2017, 36, 1707–1720. [Google Scholar] [CrossRef]
  161. Li, Q.G.; Xiao, T.; Zhu, W.; Yu, Z.Z.; Huang, X.P.; Yi, H.; Lu, S.S.; Tang, Y.Y.; Huang, W.; Xiao, Z.Q. HDAC7 promotes the oncogenicity of nasopharyngeal carcinoma cells by miR-4465-EphA2 signaling axis. Cell Death Dis. 2020, 11, 322. [Google Scholar] [CrossRef]
  162. Li, S.; Wang, B.; Xu, Y.; Zhang, J. Autotaxin is induced by TSA through HDAC3 and HDAC7 inhibition and antagonizes the TSA-induced cell apoptosis. Mol. Cancer 2011, 10, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Kwiecinska, P.; Wrobel, A.; Tauboll, E.; Gregoraszczuk, E.L. Valproic acid, but not levetiracetam, selectively decreases HDAC7 and HDAC2 expression in human ovarian cancer cells. Toxicol. Lett. 2014, 224, 225–232. [Google Scholar] [CrossRef] [PubMed]
  164. Dokmanovic, M.; Perez, G.; Xu, W.; Ngo, L.; Clarke, C.; Parmigiani, R.B.; Marks, P.A. Histone deacetylase inhibitors selectively suppress expression of HDAC7. Mol. Cancer Ther. 2007, 6, 2525–2534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Stenner, M.; Weinell, A.; Ponert, T.; Hardt, A.; Hahn, M.; Preuss, S.F.; Guntinas-Lichius, O.; Klussmann, J.P. Cytoplasmic expression of survivin is an independent predictor of poor prognosis in patients with salivary gland cancer. Histopathology 2010, 57, 699–706. [Google Scholar] [CrossRef]
  166. Wheatley, S.P.; Altieri, D.C. Survivin at a glance. J. Cell Sci. 2019, 132, jcs223826. [Google Scholar] [CrossRef] [Green Version]
  167. Stauber, R.H.; Mann, W.; Knauer, S.K. Nuclear and cytoplasmic survivin: Molecular mechanism, prognostic, and therapeutic potential. Cancer Res. 2007, 67, 5999–6002. [Google Scholar] [CrossRef] [Green Version]
  168. Lippert, B.M.; Knauer, S.K.; Fetz, V.; Mann, W.; Stauber, R.H. Dynamic survivin in head and neck cancer: Molecular mechanism and therapeutic potential. Int. J. Cancer 2007, 121, 1169–1174. [Google Scholar] [CrossRef]
  169. Wang, H.; Holloway, M.P.; Ma, L.; Cooper, Z.A.; Riolo, M.; Samkari, A.; Elenitoba-Johnson, K.S.; Chin, Y.E.; Altura, R.A. Acetylation directs survivin nuclear localization to repress STAT3 oncogenic activity. J. Biol. Chem. 2010, 285, 36129–36137. [Google Scholar] [CrossRef] [Green Version]
  170. Sen, N.; Che, X.; Rajamani, J.; Zerboni, L.; Sung, P.; Ptacek, J.; Arvin, A.M. Signal transducer and activator of transcription 3 (STAT3) and survivin induction by varicella-zoster virus promote replication and skin pathogenesis. Proc. Natl. Acad. Sci. USA 2012, 109, 600–605. [Google Scholar] [CrossRef] [Green Version]
  171. Hu, X.; Wang, L.; Lin, L.; Han, X.; Dou, G.; Meng, Z.; Shi, Y. A phase I trial of an oral subtype-selective histone deacetylase inhibitor, chidamide, in combination with paclitaxel and carboplatin in patients with advanced non-small cell lung cancer. Chin. J. Cancer Res. 2016, 28, 444–451. [Google Scholar] [CrossRef] [Green Version]
  172. Shi, Y.; Jia, B.; Xu, W.; Li, W.; Liu, T.; Liu, P.; Zhao, W.; Zhang, H.; Sun, X.; Yang, H.; et al. Chidamide in relapsed or refractory peripheral T cell lymphoma: A multicenter real-world study in China. J. Hematol. Oncol. 2017, 10, 69. [Google Scholar] [CrossRef] [Green Version]
  173. Gong, K.; Xie, J.; Yi, H.; Li, W. CS055 (Chidamide/HBI-8000), a novel histone deacetylase inhibitor, induces G1 arrest, ROS-dependent apoptosis and differentiation in human leukaemia cells. Biochem. J. 2012, 443, 735–746. [Google Scholar] [CrossRef] [Green Version]
  174. Zhou, Y.; Pan, D.S.; Shan, S.; Zhu, J.Z.; Zhang, K.; Yue, X.P.; Nie, L.P.; Wan, J.; Lu, X.P.; Zhang, W.; et al. Non-toxic dose chidamide synergistically enhances platinum-induced DNA damage responses and apoptosis in Non-Small-Cell lung cancer cells. Biomed. Pharmacother. 2014, 68, 483–491. [Google Scholar] [CrossRef] [PubMed]
  175. Shi, Y.; Dong, M.; Hong, X.; Zhang, W.; Feng, J.; Zhu, J.; Yu, L.; Ke, X.; Huang, H.; Shen, Z.; et al. Results from a multicenter, open-label, pivotal phase II study of chidamide in relapsed or refractory peripheral T-cell lymphoma. Ann. Oncol. 2015, 26, 1766–1771. [Google Scholar] [CrossRef] [PubMed]
  176. Koster, M.I.; Kim, S.; Mills, A.A.; DeMayo, F.J.; Roop, D.R. p63 is the molecular switch for initiation of an epithelial stratification program. Genes Dev. 2004, 18, 126–131. [Google Scholar] [CrossRef] [Green Version]
  177. Steurer, S.; Riemann, C.; Buscheck, F.; Luebke, A.M.; Kluth, M.; Hube-Magg, C.; Hinsch, A.; Hoflmayer, D.; Weidemann, S.; Fraune, C.; et al. p63 expression in human tumors and normal tissues: A tissue microarray study on 10,200 tumors. Biomark. Res. 2021, 9, 7. [Google Scholar] [CrossRef]
  178. Sivakumar, N.; Narwal, A.; Pandiar, D.; Devi, A.; Anand, R.; Bansal, D.; Kamboj, M. Diagnostic utility of p63/p40 in the histologic differentiation of salivary gland tumors: A systematic review. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2022, 133, 189–198. [Google Scholar] [CrossRef] [PubMed]
  179. Dokmanovic, M.; Clarke, C.; Marks, P.A. Histone deacetylase inhibitors: Overview and perspectives. Mol. Cancer Res. 2007, 5, 981–989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Boyault, C.; Sadoul, K.; Pabion, M.; Khochbin, S. HDAC6, at the crossroads between cytoskeleton and cell signaling by acetylation and ubiquitination. Oncogene 2007, 26, 5468–5476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Lee, Y.S.; Lim, K.H.; Guo, X.; Kawaguchi, Y.; Gao, Y.; Barrientos, T.; Ordentlich, P.; Wang, X.F.; Counter, C.M.; Yao, T.P. The cytoplasmic deacetylase HDAC6 is required for efficient oncogenic tumorigenesis. Cancer Res. 2008, 68, 7561–7569. [Google Scholar] [CrossRef] [Green Version]
  182. Li, T.; Zhang, C.; Hassan, S.; Liu, X.; Song, F.; Chen, K.; Zhang, W.; Yang, J. Histone deacetylase 6 in cancer. J. Hematol. Oncol. 2018, 11, 111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Schnekenburger, M.; Florean, C.; Dicato, M.; Diederich, M. Epigenetic alterations as a universal feature of cancer hallmarks and a promising target for personalized treatments. Curr. Top. Med. Chem. 2016, 16, 745–776. [Google Scholar] [CrossRef] [PubMed]
  184. Losson, H.; Schnekenburger, M.; Dicato, M.; Diederich, M. HDAC6-an Emerging Target Against Chronic Myeloid Leukemia? Cancers 2020, 12, 318. [Google Scholar] [CrossRef] [Green Version]
  185. Coca-Pelaz, A.; Rodrigo, J.P.; Bradley, P.J.; Vander Poorten, V.; Triantafyllou, A.; Hunt, J.L.; Strojan, P.; Rinaldo, A.; Haigentz, M., Jr.; Takes, R.P.; et al. Adenoid cystic carcinoma of the head and neck--An update. Oral Oncol. 2015, 51, 652–661. [Google Scholar] [CrossRef] [PubMed]
  186. Ellington, C.L.; Goodman, M.; Kono, S.A.; Grist, W.; Wadsworth, T.; Chen, A.Y.; Owonikoko, T.; Ramalingam, S.; Shin, D.M.; Khuri, F.R.; et al. Adenoid cystic carcinoma of the head and neck: Incidence and survival trends based on 1973-2007 Surveillance, Epidemiology, and End Results data. Cancer 2012, 118, 4444–4451. [Google Scholar] [CrossRef] [PubMed]
  187. Uijen, M.J.M.; Lassche, G.; van Engen-van Grunsven, A.C.H.; Driessen, C.M.L.; van Herpen, C.M.L. Case series of docetaxel, trastuzumab, and pertuzumab (DTP) as first line anti-HER2 therapy and ado-trastuzumab emtansine (T-DM1) as second line for recurrent or metastatic HER2-positive salivary duct carcinoma. Oral Oncol. 2022, 125, 105703. [Google Scholar] [CrossRef]
Figure 1. The processes of histone acetylation and deacetylation are fundamental mechanisms of epigenetic regulation, controlling gene expression. Histone acetyltransferases (HATs/KATs) catalyze the addition of acetyl groups to lysine residues of histone proteins, which promotes the relaxation of chromatin structure and enhances accessibility of DNA to transcription factors. Conversely, histone deacetylases (HDACs) remove acetyl groups from histones, leading to chromatin compaction and repression of gene transcription. This figure was created using the tools provided by BioRender.com (accessed on 3 May 2023).
Figure 1. The processes of histone acetylation and deacetylation are fundamental mechanisms of epigenetic regulation, controlling gene expression. Histone acetyltransferases (HATs/KATs) catalyze the addition of acetyl groups to lysine residues of histone proteins, which promotes the relaxation of chromatin structure and enhances accessibility of DNA to transcription factors. Conversely, histone deacetylases (HDACs) remove acetyl groups from histones, leading to chromatin compaction and repression of gene transcription. This figure was created using the tools provided by BioRender.com (accessed on 3 May 2023).
Ijms 24 10038 g001
Figure 2. Effects of HDAC inhibitors TSA and Quisinostat on the cell cycle of salivary gland ductal adenocarcinomas (SGDAs). This figure is based on the work of Nakano et al. [149] and was created using the tools provided by BioRender.com (accessed on 2 May 2023).
Figure 2. Effects of HDAC inhibitors TSA and Quisinostat on the cell cycle of salivary gland ductal adenocarcinomas (SGDAs). This figure is based on the work of Nakano et al. [149] and was created using the tools provided by BioRender.com (accessed on 2 May 2023).
Ijms 24 10038 g002
Table 1. Salivary gland neoplasms according to WHO 2022.
Table 1. Salivary gland neoplasms according to WHO 2022.
Salivary Carcinomas Not Otherwise
Specified (NOS) and Emerging Entities
Malignant Epithelial Tumors
Large-cell neuroendocrine carcinomaAcinic cell carcinoma
Lymphoepithelial carcinomaAdenoid cystic carcinoma
Oncocytic carcinomaBasal cell adenocarcinoma
Small-cell neuroendocrine carcinomaCarcinoma ex pleomorphic adenoma
Squamous cell carcinomaCarcinosarcoma
Undifferentiated carcinomaClear cell carcinoma
Benign Epithelial TumorsEpithelial–myoepithelial carcinoma
Basal cell adenomaIntraductal carcinoma
CystadenomaMicrosecretory adenocarcinoma *
Ductal papillomasMucinous adenocarcinoma *
Intercalated duct adenoma *Mucoepidermoid carcinoma
Keratocystoma *Myoepithelial carcinoma
LymphadenomaPolymorphous adenocarcinoma
MyoepitheliomaSalivary duct carcinoma
OncocytomaSclerosing microcystic adenocarcinoma *
Pleomorphic adenomaSebaceous adenocarcinoma
Sclerosing polycystic adenoma *Secretory carcinoma
Sialadenoma papilliferumUncertain Malignant Potentiality
Striated duct adenoma *Sialoblastoma
Warthin tumor
* New entities included in the newest WHO classification.
Table 4. HDAC classes, subtypes, cellular locations, substrate specificities, and functions.
Table 4. HDAC classes, subtypes, cellular locations, substrate specificities, and functions.
HDAC ClassSubtypesCellular LocationSubstrate SpecificityFunctions and Targets
Class-IHDAC1, HDAC2, HDAC3, HDAC8NucleusAll four core histones (HDAC1,2); H3, H4 (HDAC8)Gene repression; Cell-cycle regulation; Apoptosis; DNA repair [93]
Class-IIaHDAC4, HDAC5, HDAC7, HDAC9Nucleus and CytoplasmAll four core histones (HDAC4,5)Cell differentiation; Apoptosis; Angiogenesis; Response to external signals [94]
Class-IIbHDAC6, HDAC10Cytoplasm (HDAC6); Nucleus (HDAC10)H3K9, H3K56, α-tubulin (HDAC6); Polyamine catabolism (HDAC10)Regulation of microtubule stability; Aggresome formation [94]
Class-IIISIRT1, SIRT2Nucleus and CytoplasmH4K16, H3K9 (SIRT1); H4K16, H3K56 (SIRT2)Aging; Metabolism; DNA repair; Stress resistance [93]
SIRT3, SIRT4, SIRT5MitochondriaH4K16 (SIRT3); Mitochondrial metabolism (SIRT4,5)Energy homeostasis; Oxidative stress response [93]
SIRT6, SIRT7Nucleus (SIRT6); Nucleolus (SIRT7)H3K9, H3K56 (SIRT6); H3K18 (SIRT7)Genome stability; Ribosome biogenesis [93]
Class-IVHDAC11Nucleus and CytoplasmH3K9, H3K14DNA replication regulation; Immune regulation [93]
Table 5. HDAC expression in HN malignancies and SGTs.
Table 5. HDAC expression in HN malignancies and SGTs.
Categorization Study Type/MethodHDAC TypeExpression RateReference
HN malignanciesTissue sections (FFPE)/IHCHDAC2, -9, -10~100%[107]
SIRT3, -5, -6, -7~100%[107]
HDAC1, SIRT1~75%[107]
SGTsTissue sections (FFPE)/IHCHDAC1Benign SGTs: 30%[110]
Malignant SGTs: 14%[110]
Tissue sections (FFPE)/IHCHDAC2Benign SGTs: 86%[110]
Malignant SGTs: 82%[110]
Tissue sections (FFPE)/IHCHDAC4Benign SGTs: 44%[110]
Malignant SGTs: 36%[110]
Tissue sections (FFPE)/IHCHDAC6Benign SGTs: 11%[110]
Malignant SGTs: 18%[110]
Table 6. Research studies evaluating HDACi efficacy in salivary gland malignancies.
Table 6. Research studies evaluating HDACi efficacy in salivary gland malignancies.
Type of Salivary Gland MalignancySubstanceMain OutcomeReference
MEC (cell line)Apicidin (HDACi) proliferative potential[143,144]
MEC (cell line)Vorinostat (HDACi) cancer stem cell population[145]
Vorinostat (HDACi) + cisplatin sensitivity to cisplatin
AdCC (cell line)
Mice transplanted with tumor cells
Vorinostat (HDACi) population of cancer cells and stem cells[146]
Vorinostat (HDACi) + cisplatinFurther cancer stem cell population
AdCC (cell line)Chidamide (HDACi) proliferative potential[113]
Chidamide (HDACi) + cisplatinCell cycle arrest
MEC (cell line)Vorinostat (HDACi) cancer stem cell population[69]
Vorinostat (HDACi) + Εmetin (anti-NF-κB)Achieving combined therapeutic effect
MEC (cell line)CUDC-101
(EGFRi + HDACi)
cytotoxicity
cancer stem cell oncogenicity
[147]
MEC (cell line)
Mice transplanted with tumor cells
Sodium butyrate (HDACi) proliferative potential
no organ toxicities in vivo
[148]
SGDA (cell line)HDACi
(TSA + Quisinostat)
proliferation and migration potential
epithelial barrier function
[149]
↑ refers to increased, ↓ refers to decreased.
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

Manou, M.; Kanakoglou, D.S.; Loupis, T.; Vrachnos, D.M.; Theocharis, S.; Papavassiliou, A.G.; Piperi, C. Role of Histone Deacetylases in the Pathogenesis of Salivary Gland Tumors and Therapeutic Targeting Options. Int. J. Mol. Sci. 2023, 24, 10038. https://doi.org/10.3390/ijms241210038

AMA Style

Manou M, Kanakoglou DS, Loupis T, Vrachnos DM, Theocharis S, Papavassiliou AG, Piperi C. Role of Histone Deacetylases in the Pathogenesis of Salivary Gland Tumors and Therapeutic Targeting Options. International Journal of Molecular Sciences. 2023; 24(12):10038. https://doi.org/10.3390/ijms241210038

Chicago/Turabian Style

Manou, Maria, Dimitrios S. Kanakoglou, Theodoros Loupis, Dimitrios M. Vrachnos, Stamatios Theocharis, Athanasios G. Papavassiliou, and Christina Piperi. 2023. "Role of Histone Deacetylases in the Pathogenesis of Salivary Gland Tumors and Therapeutic Targeting Options" International Journal of Molecular Sciences 24, no. 12: 10038. https://doi.org/10.3390/ijms241210038

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