Next Article in Journal
ONT-Based Alternative Assemblies Impact on the Annotations of Unique versus Repetitive Features in the Genome of a Romanian Strain of Drosophila melanogaster
Next Article in Special Issue
DNA Damage Response Mechanisms in Head and Neck Cancer: Significant Implications for Therapy and Survival
Previous Article in Journal
Evaluation of HIF-1 Involvement in the BDNF and ProBDNF Signaling Pathways among Obstructive Sleep Apnea Patients
Previous Article in Special Issue
The Hypothalamic–Pituitary–Thyroid Axis Equivalent in Normal and Cancerous Oral Tissues: A Scoping Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Therapeutic Landscape of Salivary Gland Malignancies—Where Are We Now?

by
Robbert Cleymaet
1,†,
Tijl Vermassen
2,3,4,*,†,
Renaat Coopman
1,4,
Hubert Vermeersch
1,
Stijn De Keukeleire
5 and
Sylvie Rottey
2,3,4,6
1
Department of Oromaxillofacial and Plastic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
2
Department Medical Oncology, University Hospital Ghent, 9000 Ghent, Belgium
3
Department Basic and Applied Medical Sciences, Ghent University, 9000 Ghent, Belgium
4
Cancer Research Institute Ghent, 9000 Ghent, Belgium
5
Department Internal Medicine, University Hospital Brussels, 1090 Brussels, Belgium
6
Drug Research Unit Ghent, University Hospital Ghent, 9000 Ghent, Belgium
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(23), 14891; https://doi.org/10.3390/ijms232314891
Submission received: 14 November 2022 / Revised: 23 November 2022 / Accepted: 25 November 2022 / Published: 28 November 2022
(This article belongs to the Special Issue Advances in Molecular Mechanism of Head and Neck Cancer)

Abstract

:
Salivary gland malignancies (SGMs) account for less than 5% of new diagnoses in head and neck tumors. If feasible, surgery is the preferred treatment modality. Nevertheless, some malignancies have a tendency of recurrence, with possible distant metastasis. Alternative treatment strategies, such as primary radiation or chemotherapeutics, often present low response rates. As a result, there is an unmet need for novel therapeutic approaches. Nowadays, target-based therapies (e.g., small inhibitors and immunotherapy) are used by the medical oncologist for possible treatment of advanced SGMs. Based on recent published trials, some novel treatments may provide additional disease control for some patients. However, sample sizes are small, the general findings are unsatisfactory, and a lot of uncertainties remain to be elucidated. Nevertheless, research shows that patients do not benefit from blind administration of systemic treatments and therefore a more personalized approach is highly needed. The aim of this review paper is to summarize the most recent advances in the biological understanding and molecular pathways of salivary gland cancers, the association of these pathways with the current treatments used and their implications for more personalized targeted-based therapies.

1. Introduction

Salivary gland malignancies (SGMs) are rare malignancies in the head and neck region representing fewer than 5% of the newly diagnosed head and neck neoplasms [1,2]. The WHO describes almost 40 different tumors in the salivary gland region of which the most frequent malignant SGMs are muco-epidermoid carcinoma (MEC) [3,4,5,6,7,8], adenoid cystic carcinoma (AdCC) [5,9,10,11,12], salivary duct adenocarcinoma (SDC) [5,13,14,15] and acinic cell carcinoma (AcCC) [5,16,17,18,19] (Table 1).
Swelling at the salivary gland’s location is often the first clinical symptom indicative of a tumoral process, as well as peripheral facial nerve loss, ingrowth and fixation at the skin or the presence of lymph nodes in the neck region. Preliminary diagnosis is routinely done via ultra-sound guided fine needle aspiration cytology (FNAC). However, its use is limited when it comes to identifying the subtype and grade of the malignant tumor [20].
Table 1. Overview of the most common SGMs.
Table 1. Overview of the most common SGMs.
SubtypePrevalenceTreatmentPrognosis
MEC29–35% [3,4,6]- Surgery in case of resectable tumor
- Adjuvant radiotherapy in case of perineural invasion, lymph node involvement, advanced high-grade tumors, positive margins after resection and extra-glandular extension [8]
Depends on grading
5-year OS = 79.3%
5-year DFS = 76.5% [7]
AdCC20–22% [9,10,11]- Complete surgical resection
- Postoperative radiotherapy is almost always used for AdCC [12]
5-year OS = 55–98%
15-year OS = 23–40% [11]
SDC5–10% [13,14]- Surgery and radiation therapy [13]5-year OS = 30–48% [15]
AcCC10–25% [16,17,18,19]- Complete surgical removal via total or subtotal parotidectomy
- Postoperative radiotherapy in all cases except T1N0 or T2N0 [16]
5-year OS = 91%
10-year OS = 88% [16]
AcCC, acinic cell carcinoma AdCC, adenoid cystic carcinoma; DFS, disease-free survival; MEC, mucoepidermoid carcinoma; OS, overall survival; SDC, salivary duct carcinoma; SGMs, salivary gland malignancies.
Ultrasound-guided core needle biopsy provides a more precise preoperative diagnosis, mostly because the technique obtains larger tissue samples, preserves the histologic architecture, and can thereby provide a more accurate insight in tumor typing, grading and immunohistochemistry [21]. Initial diagnosis is often supplemented with a computed tomography (CT) scan or magnetic resonance imaging (MRI). Importantly, these imaging techniques can be leveraged to evaluate local extent and perineural extension, both of which are critical for a correct TNM staging [22]. Novel MRI techniques, including dynamic contrast-enhanced MRI and diffusion-weighted MRI, are being investigated to differentiate between benign and malignant processes [23]. Finally, to detect tumor extension, local recurrence, nodal involvement, and distant metastasis, positron emission tomography CT (PET-CT) is more accurate than classical CT [24]. However, PET-CT is not useful in classifying SGMs as benign or malignant [25]. Although great improvements in imaging have been reported in recent years, it remains difficult to fully rely on imaging for the final SGM diagnosis, thus leaving biopsy as the gold standard.
Upon conclusive diagnosis, a broad local excision with negative margins is the first-line curative therapy for SGMs in the absence of distant metastases. This can be combined with concurrent/adjuvant radiotherapy and/or chemotherapy, depending on lymph node involvement, recurrence, or metastasis of the SGM. The treatment strategies for localized SGMs have extensively been described in other reviews and will not be covered in the current review [26,27,28,29].
Once the patient is diagnosed with locally advanced, inoperable recurrent, or metastatic disease, systemic treatment is mostly the therapy of choice. Classic chemotherapeutic agents have proven only limited durable response. Thus, the necessity has arisen of implementing targeted molecular agents into the armamentarium. However, clinical trials introducing novel treatment strategies in SGMs are scanty [30].
In the current review paper, we will provide an extensive overview on the available systemic treatment regimens in patients with advanced salivary gland carcinoma. In addition, we elaborate on the molecular pathways involved in the carcinogenesis of SGMs and the consequent importance of more personalized and targeted-based therapies. The latter will be illustrated by introducing a case-report from our university hospital.

2. Systemic Therapies

2.1. Chemotherapy

Prior to the advent of targeted therapies, only chemotherapy was available for as palliative treatment in inoperable and advanced/metastatic malignancies of the salivary gland [31]. Various chemotherapy regimens, both in monotherapy or as combination, have been tested in salivary gland carcinoma with objective response rates (ORR) ranging between 0% and 70% [32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54]. An overview of the chemotherapeutic options tested in SGMs is given in Table 2. However, there are presently no conventional chemotherapeutic options recommended in the (neo-)adjuvant nor metastatic setting.

2.2. Target-Based Therapy

Nowadays, researchers are attempting to fully elucidate the molecular etiology of SGMs. Angiogenesis, proliferation, and survival are three hallmarks of carcinogenesis. To enable the tumor to grow and survive, multiple receptors and pathways, such as PI3K, PLC, JAK, and RAS pathways; are activated or overexpressed. This is also the case for SGMs (Figure 1).
Targeting specific receptors or intracellular pathways may therefore be the way to go for SGMs, due to the numerous subtype-specific genomic alterations and upregulations. Nevertheless, molecular variation among different types of SGMs also complicates the selection of the appropriate drug according to a review paper from Sahara et al. [55]. Below, we summarize which receptors, intracellular molecules, and epigenetic factors play an important role in SGMs, and which targeted therapies are currently available to inhibit the aberrant expression/alteration in SGMs.

2.2.1. Receptors

Several overexpressing receptors, e.g., receptor tyrosine kinases, have been indicated for SGMs, especially AdCC. Overexpression of c-KIT has been identified as a hallmark of AdCC histology as 80–90% of cases of AdCC overexpress c-KIT [56]. VEGF, and consequently the VEGFR, is highly expressed in AdCC. This increased expression correlates with stage, tumor size, vascular invasion, recurrence, and metastasis thus considering VEGFR to be the crucial receptor involved in the initiation of the formation and maintenance of tumor vasculature in AdCC [57,58,59,60]. Fibroblast growth factor (FGF) signaling is associated with the progression of AdCC. Fibroblast growth factor (FGF) 1, FGF2, and FGF receptor 1 (FGFR1) have been shown to be overexpressed in AdCC and implicated in the carcinogenesis of AdCC upregulation of extracellular signal-regulated kinase and cyclin D1 and P21 signaling pathway [61,62]. EGFR overexpression rate is seen in about 70% of salivary gland carcinoma, with a maximum of 85% in AcCC, making it an attractive therapeutic target [63,64,65]. Additionally, signaling via insulin-like growth factors (IGF)-1R has been demonstrated to play a key role in malignant transformation, anti-apoptosis, and metastatic behavior in a variety of cancers [66,67,68]. In non-small cell lung cancer, IGF-1R is often overexpressed and can mediate the cancer proliferation [69]. In addition, acquired resistance to EGFR tyrosine kinase inhibitors (TKIs) is associated with enhanced dependency on IGF-1R signaling [70,71,72,73]. Moreover, evidence is accumulating that EGFR/IGF-1R heterodimerization as well as crosstalk between the IGF-1R and EGFR pathways can induce resistance to EGFR inhibition mediated by IGF-1R and vice versa [70].
Both the androgen receptor (AR) and human epidermal growth factor receptor 2 (HER2 or ErBB2) play a role in SGMs. More particularly in SDC, as up to 90% of patients are positive for the AR, and up to 40% are positive for HER2 [13,14,15,74,75,76,77,78,79], with the rate of co-expressing AR and HER2 was found to vary between 35% and 58% [79,80]. Based on the morphologic resemblance and molecular profiling (AR expression; TP53 mutations, HRAS mutations, PIK3CA mutations, and HER2 amplification), SDC is remarkable similar to high-grade breast ductal carcinoma [75]. Next to SDC, other SGMs found to express AR include adenocarcinoma, carcinoma ex pleomorphic adenoma, MEC, and basal cell adenocarcinoma [81,82]. The biologic significance of concomitant HER2 and AR expression remains to be elucidated, although these patients seem to have a worse outcome [79,80].
Oncogenic activation of the Notch pathway, either through mutation, gene rearrangement or over-expression, is implicated in solid tumours [83,84]. Previous studies demonstrate poor prognostic outcomes in AdCC patients with NOTCH mutations [85,86].
Immune-related receptors, such as the programmed death (PD)-1/PD-Ligand (L)1 axis, have shown to play an important role in cancer homeostasis as upregulation of the PD-1 pathway leads to suppression of immune response in many tumors [87]. An analysis of 217 surgically resected SGM specimens indicated high PD-L1 expression in high-grade SGM subtypes associated with aggressive behavior (e.g., SDC). An association between PD-L1 positivity and inferior disease-free survival was also observed [88]. In addition, PD-L2 expression has been found in several tumor types, including AdCC [89,90].

2.2.2. Intracellular and Epigenetic Alterations

Intracellular alterations in pathways and/or protein expression has been shown to be an important player in the carcinogenesis of SGMs.
The myeloblastosis (MYB) transcription factor regulates multiple transcriptional pathways including cellular differentiation and proliferation. Alterations and overexpression of the myeloblastosis (MYB) signaling pathway (65%) is a hallmark of AdCC [85,91]. The functional significance of this MYB overexpression is an upregulation of several growth and angiogenic factors, including VEGFA, FGF2, and c-KIT. Of note, the upregulation of VEGFA, FGF2, and c-KIT is not solely MYB-induced, suggesting dependent and independent mechanisms of VEGFR/c-KIT/PDGFR activation [92,93,94,95].
Limited data is available on the phosphatidylinositol 3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) pathway. Younes et al. [96], described that AdCC cell lines exhibited increased phosphorylated Akt activity when stimulated with EGF, which could be countered by treatment with an EGFR/VEGFR inhibitor. Interestingly, only the phosphorylated form of Akt decreased whereas the total level of Akt remained unchanged, indicating the importance of the phosphorylation step by PI3K in the carcinogenesis of SGMs.
NF-κB represents a family of inducible transcription factors which is involved in most gene activations. It was found that NF-κB is highly expressed in AdCC and relates to angiogenesis and poor patient outcome [60,97]. Lastly, DNA methylation and histone hypoacetylation lead to silencing of key genes, including those involved in immune recognition. Preclinical observations of various proposed mechanisms relating to altered gene expression in malignant cells, lymphocytes and cells within the tumor microenvironment support the synergistic activity of epigenetic modification and immune check-point inhibition [98].
Seen the involvement of the above-mentioned targets in SGMs, there is strong rationale for the development and testing of specific inhibitors as anti-cancer therapy in SGMs. Multiple clinical studies, of which the majority phase II studies (listed in Table 3), have been conducted in order to determine the effectiveness of these inhibitors in heterogeneous populations of SGMs [81,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145].
It is however clear that, despite various specific inhibitors at our disposal, the response to therapy in advanced and metastatic SGMs remains poor.
This is especially true for the use of multiple TKIs, with most studies only indicating limit to no responses with occasional ORR reaching up to 46%. The low response to multiple TKIs can be explained by the fact that various receptors and pathways, as indicated above, can induce cancer proliferation and survival (Figure 1). As such, this multitude in pathways provide the tumor with an excellent mechanism of evading the anti-tumoral effect, induced by the TKIs. This might also explain why important differences are observed between studies using the same targeted agent. In this matter, Feeney et al. [111] reported no response to lenvatinib used in patients with AdCC whereas Locati et al. [132] and Tchekmedyian et al. [141] reported an ORR of 12% and 16%; respectively.
Use of the anti-HER2 monoclonal antibody trastuzumab on the other hand, in combination with pertuzumab (anti-HER2) or docetaxel chemotherapy showed a nice effectiveness in patients with HER2+ SGMs, displaying an ORR of 56% and 70%; respectively [128,140]. This reached even efficacy of 90% for use of the HER2-targeted antibody-drug conjugate ado-trastuzumab emtansine in HER2+ patients [129]. This illustrates the high importance for assessing the tumor pathological and molecular profile, as HER2 overexpression and alterations are typically seen in SDC. Nevertheless, this will not always be a measurement for success as the phase II study for trastuzumab monotherapy in HER2+ SGMs only reached an ORR of 8% [116]. It is therefore clear that SGMs attempt to induce drug resistance by activating different receptors and subsequent pathways thus rendering the anti-HER2-therapy useless, as was indicated for multiple TKIs.
The same holds true for therapies directed at the AR. Despite the strong rationale, ORR for enzalutamide (non-steroidal AR antagonist), abiraterone acetate (androgen biosynthesis inhibitors) and leuprorelin acetate + bicalutamide (gonadotropin-releasing hormone receptor agonist + non-steroidal AR antagonist) was only 15%, 21% and 42%, respectively [113,119,131]. The combination of leuprorelin acetate + bicalutamide did although result in a clinical benefit of 75% and a statistically significant increase in three-year disease-free survival (48% versus 28%).
The introduction of immune check-point inhibition (ICI) has shown modest response in treatment-resistant salivary gland cancers. One possible target is PD-L1, as its expression is associated with malignant behavior in SGMs. In a phase Ib trial, the fully humanized monoclonal anti-PD-1 therapeutic antibody pembrolizumab has been investigated during 24 months in 26 PD-L1+ (combined positive score ≥ 1) patients with advanced salivary gland cancers. Twelve percent of this cohort achieved a partial response while an additional 46% of patients had a stable disease course. This means that a total of 58% were classified as having disease control over 24 months [106], indicating the potential of immune checkpoint inhibitors in this patient population.
Due to the rather limited efficacy of blocking receptors involved in SGMs, it is therefore reasonable that targeting intracellular molecules, more downstream in the pathway, might harbor better effects. Although the literature on these intracellular drugs is sparse, also these drugs do not present with the highly needed efficacy for treatment of metastatic SGMs. Inhibition of mTOR, Akt, and NF-κB namely [101,121,126] showed no response in SGMs. Next, cancer stem cell-targeted systemic treatments are also now being extensively assessed. Inhibition of NOTCH signaling has generated a response rate of 0% to 17% in phase I studies [102,110,112]. Focusing more on the epigenetic alterations, by inhibiting DNA-methylation through PRMT5 inhibitors or by inhibiting histone hypoacetylation via histone deacetylase inhibitors, showed a respective ORR 21% and 9% [115,139,146]. Clinical trials utilizing Myb-targeted inhibitors are ongoing.
About outcome in the metastatic setting, if reported and defined, median PFS ranged between 2.3 months and 19.7 months, whereas median OS was between 7.0 months and 39.7 months. The substantial difference between lowest and highest reported outcomes most likely the result of which patients were included into the trials, as in most studies, number of prior systemic lines was no exclusion criteria. In addition, it is important to emphasize that the outcome is also skewed due to the heterogenous populations included into these clinical trials. In this matter, patients with metastatic AdCC or low grade adenocarcinoma have a much slower disease progression in comparison to patients with more aggressive SGMs, such as SDC and mucinous adenocarcinoma. This is clearly demonstrated by the survival data of the clinical trials with lapatinib, sorafenib, gefitinib, cabozantinib and eribulin mesylate. In these trials, the median OS for patients with AdCC is not reached, 26.4 months, 25.9 months, 27.5 months and 20.0 months, whereas OS equals 13.8 months, 12.3 months, 16.0 months, 14.2 months and 15.5 months for patients with non-AdCC; respectively. In these studies, the non-AdCC population consisted mostly from a mix of MEC, SDC, AcCC and adenocarcinoma [100,123,133,137,143].
Next, it is however also clear that highest outcomes were achieved for latest-generation TKIs (e.g., apatinib, cabozantinib, and lenvatinib) [132,141,143,145], or for combination therapies such as imatinib + cisplatin, leuprorelin acetate + bicalutamide or trastuzumab + docetaxel [113,114,140]. The latter would suggest a rational combination of targeted inhibitors of these pathways might improve patient outcome. Of course, treating physicians should be aware of the enhanced safety profile when combining these drugs to increase the success rate in salivary gland carcinoma.
So, although promising therapies have/are being investigated, which can provide the oncologist with new treatment avenues, not all aforementioned therapies are readily used as standard-of-care. For now, the unmet need for therapeutic strategies to treat advanced malignant SGMs still remains. Several clinical studies (monotherapeutic and combination therapies) are currently ongoing (Table 4). However, as we stipulated that several pathways can be used in salivary gland carcinoma to secure tumor proliferation, neo-angiogenesis and survival, the questions remain whether these trials will be successful.
It is therefore imperative that future studies not only validate the above-mentioned therapeutic efficacy findings, but also focus on the tumor biology in order to discover novel targets with higher diagnostic and/or therapeutic potential [55].
Table 3. Overview of targeted therapies tested in SGMs.
Table 3. Overview of targeted therapies tested in SGMs.
PhaseSettingAgentTargetPts, nSubtype *ORR, n (%) *Median PFS (Months)Median OS (Months)
A. Tyrosine kinase inhibitors
II [145]R/M
Any line
ApatinibVEGFR, RET, c-KIT65AdCC30 (46)19.7Not reached
II [118]R/M
Any line
AxitinibVEGFR, PDGFR, c-KIT33AdCC3 (9)5.7
II [130]R/M
First- or second-line
AxitinibVEGFR, PDGFR, c-KIT26AdCC: 6
Non-AdCC: 20
(1 SDC, 3 AcCC, 6 adeno, 2 clear cell, 1 ex pleomorphic adenoma, 1 myoepithelial, 1 epithelial-myoepithelial, 5 undifferentiated)
ITT: 2 (8)
AdCC: 1 (17)
Non-AdCC: 1 (5)
(1 undifferentiated)
5.526.2
II [124]R/M
Any line
AxitinibVEGFR, PDGFR, c-KIT27AdCC0 (0)10.8NR
II [124]R/M
Any line
Axitinib
(after corss-over)
VEGFR, PDGFR, c-KIT26AdCC3 (12)14.527.2
II [143]R/M
Any line
CabozantinibMET, RET, AXL, VEGFR2, FLT3, c-KIT.21AdCC: 15
Non-AdCC: 6
(1 MEC, 4 SDC, 1 AcCC, 1 ex pleomorphic adenoma)
ITT: 2 (10)
AdCC: 1 (7)
Non-AdCC: 1 (17)
(1 SDC)
AdCC: 9.4
Non-AdCC: 7.2
AdCC: 27.5
Non-AdCC: 14.2
II [81]R/M
First-, second- or third-line
CetuximabEGFR30AdCC: 23
Non-AdCC: 7
(2 MEC, 1 AcCC, 1 adeno, 3 myoepithelial)
ITT: 0 (0)
AdCC: 0 (0)
Non-AdCC: 0 (0)
ITT: 6.0
AdCC: 6.0
Non-AdCC: 2.0
I/II [99]Non-R/M
No prior line
Cetuximab
(+ IMRT)
EGFR23AdCC8 (35)NR (DFS)54.0
II [117]Locally advancedCetuximab + cisplatinCetuximab: EGFR
Cisplatin: cytostatic drug
9EGFR+ AdCC4 (44)64.0Not reached
II [117]R/MCetuximab + cisplatin + 5-fluorouracilCetuximab: EGFR
Cisplatin: cytostatic drug
5-fluorouracil: cytostatic drug
12EGFR+ AdCC5 (42)13.024.0
II [144]R/M
Any line
Dasatinibc-kit, BCR-ABL, SRC family, PDGFβ, EPHA254c-kit+
AdCC: 40
Non-AdCC: 14
ITT: 1 (2)
AdCC: 1 (3)
Non-AdCC: 0 (0)
AdCC: 4.8AdCC: 14.5
II [125]R/M
Any line
DovotinibVEGFR, c-Kit, PDGFR, CSF-1R, RET, TrkA, FLT332AdCC1 (3)6.0Not reached
II [107]R/M
Any line
DovotinibVEGFR, c-Kit, PDGFR, CSF-1R, RET, TrkA, FLT334AdCC2 (6)8.220.6
I [104]R/M
Any line
Figitumumab + dacomitinibFigitumumab: IGF1R
Dacomitinib: HER1/EGFR, HER2, HER4
5AdCC: 3
Non-AdCC: 2
1 (20)
II [123]R/M
Any line
GefitinibEGFR36AdCC: 19
Non-AdCC: 17
(2 MEC, 3 SDC, 2 AcCC, 9 adeno, 1 myoepithelial)
ITT: 0 (0)
AdCC: 0 (0)
Non-AdCC: 0 (0)
AdCC: 4.3
Non-AdCC: 2.1
AdCC: 25.9
Non-AdCC: 16.0
II [122]R/M
Any line
Imatinibc-kit, BCR-ABL, PDGFR16c-kit+ AdCC0 (0)2.37.0
II [136]R/M
Any line
Imatinibc-kit, BCR-ABL, PDGFR10AdCC0 (0)6.0
Retro [103]R/M
First-line
Imatinibc-kit, BCR-ABL, PDGFR8c-kit+ AdCC0 (0)3.0
II [114]R/M
Any line
Imatinib + cisplatinImatinib: c-kit, BCR-ABL, PDGFR
Cisplatin: cytostatic drug
28c-kit+ AdCC3 (11)15.035.0
II [100]R/M
Any line
LapatinibHER2, EGFR40EGFR+ or HER2+
AdCC: 20
Non-AdCC: 20
(2 MEC, 4 SDC, 1 AcCC, 7 adeno, 3 SCC, 3 undifferentiated)
ITT: 0 (0)
AdCC: 0 (0)
Non-AdCC: 0 (0)
ITT: 3.4
AdCC: 3.5
Non-AdCC: 2.1
ITT: Not reached
AdCC: Not reached
Non-AdCC: 13.8
II [141]R/M
Any line
LenvatinibVEGFR, FGFR, PDGFR, RET, KIT32AdCC5 (16)17.5
II [132]R/M
Any line
LenvatinibVEGFR, FGFR, PDGFR, RET, KIT26AdCC3 (12)9.127.0
Retro [111]R/M
Any line
LenvatinibVEGFR, FGFR, PDGFR, RET, KIT21AdCC0 (0)4.512.0
II [127]R/M
Any line
NintedanibVEGFR, FGFR, PDGFR20AdCC: 13
Non-AdCC: 7
(2 MEC, 1 SDC, 1 AcCC, 3 adeno)
ITT: 0 (0)
AdCC: 0 (0)
Non-AdCC: 0 (0)
ITT: 7.9
AdCC: 7.9
Non-AdCC: 6.3
Not reached
II [120]R/M
Any line
RegorafenibVEGFR, FGFR, PDGFR38AdCC0 (0)
II [142]R/M
Any line
Sorafenibb-Raf, c-Raf, c- KIT, VEGFR, PDGFR, FLT323AdCC2 (11)11.319.6
II [133]R/M
Any line
Sorafenibb-Raf, c-Raf, c- KIT, VEGFR, PDGFR, FLT337AdCC: 19
Non-AdCC: 18
(5 MEC, 2 SDC, 7 adeno, 3 myoepithelial, 1 undifferentiated)
ITT: 6 (16)
AdCC: 2 (11)
Non-AdCC: 4 (22)
(1 MEC, 1 SDC, 1 adeno, 1 undifferentiated)
ITT: 5.9
AdCC: 8.9
Non-AdCC: 4.2
ITT: 23.4
AdCC: 26.4
Non-AdCC: 12.3
I [134]R/M
Any line
Sorafenib + R1507Sorafenib: b-Raf, c-Raf, c- KIT, VEGFR, PDGFR, FLT3
R1507: IGF1R
?AdCC1
II [105]R/M
Any line
SunitinibVEGFR, c-KIT, PDGFR, RET, FLT313AdCC0 (0)7.218.7
B. Immune checkpoint inhibition
Ib [106]R/M
Any line
PembrolizumabPD-126PD-L1+ (CPS ≥ 1)
AdCC: 2
Non-AdCC: 24
(3 MEC, 1 SDC, 1 AcCC, 11 adeno, 1 myoepithelial, 1 carcinoid, 2 SCC, 1 serous, 3 undifferentiated)
ITT: 3 (12)
AdCC: 0 (0)
Non-AdCC: 3 (13)
(2 adeno, 1 serous)
4.013.0
II [135]R/M
Any line
PembrolizumabPD-110AdCC0 (0)6.627.2
II [135]R/M
Any line
Pembrolizumab
(+ IMRT)
PD-110AdCC0 (0)4.5Not reached
II [109]R/M
Any line
PembrolizumabPD-1109AdCC: 59
Non-AdCC: 50
(4 MEC, 2 SDC, 1 AcCC, 25 adeno, 5 mucinous, 2 SCC, 3 cystadeno, 2 ex pleomorphic adenoma, 1 cystic, 2 myoepithelial, 1 epithelial-myoepithelial, 2 undifferentiated)
5 (5)
PD-L1 (CPS 0): 2/77 (3)
PD-L1+ (CPS ≥ 1): 3/28 (11)
4.021.1
II [138]R/M
Any line
Pembrolizumab + VorinostatPembrolizumab: PD-1
Vorinostat: histone deacetylase
25AdCC: 12
Non-AdCC: 13
(3 MEC, 1 SDC, 3 AcCC, 1 adeno, 1 myoepithelial, 2 ex pleomorphic adenoma, 1 lymphoepithelioma, 1 clear cell)
ITT: 4 (16)
AdCC: 1 (8)
Non-AdCC: 3 (23)
(2 AcCC, 1 lymphoepithelioma)
6.914.0
C. HER2-targeted therapy
II [116]R/M
First-, second- or third-line
TrastazumabHER214HER2+
AdCC: 2
Non-AdCC: 12
(3 MEC, 7 adeno, 2 SCC)
AdCC: 0 (0)
Non-AdCC: 1 (8)
(1 MEC)
4.2
II [128]R/M
Any line
Trastuzumab + PertuzumabTrastuzumab: HER2
Pertuzumab: HER2
16HER2+ non-AdCC
(1 MEC, 3 SDC, 8 adeno, 4 unspecified)
9 (56)9.120.4
II [140]R/M
Any line
Trastuzumab + DocetaxelTrastuzumab: HER2
Docetaxel: cytostatic drug
57HER2+ SDC40 (70)8.939.7
II [129]R/M
Any line
Ado-trastuzumab emtansineHER2-targeted ADC10HER2+9 (90)Not reachedNot reached
D. AR-targeted therapy
II [119]R/M
Any line
EnzalutamideAR46AR+
AdCC:1
Non-AdCC: 45
(41 SDC, 1 adeno, 3 ex pleomorphic adenoma)
7 (15)5.617.0
II [131]R/M
Any line
Abiraterone acetateCYP17A124AR+ non-AdCC
(19 SDC, 5 adeno)
5 (21)ITT: 3.7
SDC: 4.0
Adeno: 2.5
ITT: 22.5
SDC: Not reached
Adeno: 8.8
II [113]R/M
Any line
Leuprorelin acetate + BicalutamideLeuprorelin acetate: GnRH receptor agonist
Bicalutamide: AR
36AR+ non-AdCC
(34 SDC, 2 adeno)
15 (42)8.830.5
E. Notch
I [112]R/M
Any line
BrontictuzumabNOTCH112AdCC2 (17)
I [102]R/M
Any line
BMS-986115pan-NOTCH5AdCC0 (0)
I [110]R/M
Any line
Crenigacestatpan-NOTCH22AdCC1 (5)ITT: 5.3
1st-line: Not reached
2nd-line: 7.7
3rd+-line: 2.4
F. Other therpies
II [121]R/M
Any line
NelfinavirAkt pathway15AdCC0 (0)5.5
II [126]R/M
Any line
EverolimusmTOR34AdCC0 (0)11.223.7
II [101]R/M
Any line
BortezomibNF-κB21AdCC0 (0)6.421.0
II [101]R/M
Any line
Bortezomib + doxorubcinBortezomib: NF-κB
Doxorubcin: cytostatic drug
10AdCC1 (10)6.421.0
II [137]R/M
Any line
Eribulin mesylateMicrotubule inhibitor29AdCC: 11
Non-AdCC: 18
(2 MEC, 1 SDC, 2 AcCC, 4 adeno, 3 myoepithelial, 2 ex pleomorphic adenoma, 1 mammary analogue, 1 clear cell, 2 undifferentiated)
ITT: 3 (10)
AdCC: 2 (18)
Non-AdCC: 1 (6)
(1 ex pleomorphic adenoma)
ITT: 3.5
AdCC: 3.5
Non-AdCC: 3.3
ITT: 16.0
AdCC: 20.0
Non-AdCC: 15.5
I [139]R/M
Any line
GSK3326595PRMT514AdCC3 (21)
I [146]R/M
Any line
ChidamideHistone deacetylase3Undefined1 (33)
II [115]R/M
Any line
VorinostatHistone deacetylase30AdCC2 (7)10.011.5
* If specified in the original study, the subtyping of the non-AdCC tumors is given. AcCC, acinic cell carcinoma; ADC, antibody-drug conjugate; AdCC, adenoid cystic carcinoma; AR, androgen receptor; BCR-ABL, breakpoint cluster region-Abelson; CPS, combined positive score; CSF-1R, colony stimulating factor-1 receptor; CYP17A1, 17α-hydroxylase; DFS, disease-free survival; EGFR, epidermal growth factor receptor; EPHA2, Ephrin A2; FGFR, fibroblast growth factor receptor; FLT3, fms-like tyrosine kinase 3; GnRH, gonadotropin hormone-releasing hormone; HER, human epidermal growth factor receptor; IGF1R, insulin-like growth factor 1 receptor; ITT, intention-to-treat; MEC, mucoepidermoid carcinoma; MET, hepatocyte growth factor receptor; mTOR, mammalian target of rapamycin; n, number; NF-κB, nuclear factor-κB; NOTCH, Neurogenic locus notch homolog protein; ORR, objective response rate; OS, overall survival; PD-1, programmed cell death-1; PDGFR, platelet-derived growth factor receptor; PDGF, platelet-derived growth factor; PD-L1, programmed cell death-ligand 1; PFS, progression-free survival; PRMT5, protein arginine methyltransferase 5; Pts, patients; R/M, recurrent or metastatic; RET, rearranged during transfection; Retro, retrospective; SCC, squamous cell carcinoma; SDC, salivary duct carcinoma; SGMs, salivary gland malignancies; SRC, steroid receptor coactivator; TrkA, tropomyosin receptor kinase A; VEGFR, vascular endothelial growth factor receptor.
Table 4. Ongoing clinical trials in SGMs.
Table 4. Ongoing clinical trials in SGMs.
PhaseTrial IdentificationSettingAgentTargetPts, nSubtype
INCT03886831R/M-Any linePRT543PRMT5227AdCC (+other indications)
INCT03291002R/M-Any lineCV8102 + anti-PD-1 therapyVMD-928: TLR7/8 agonist
anti-PD-1 therapy: PD-1
98AdCC (+other indications)
INCT03556228R/M-Any lineVMD-928VMD-928: TrkA74AdCC (+other indications)
INCT03287427R/M-Any lineTetMYB vaccine + tislelizumabTetMYB vaccine: cancer therapy vaccine
Tislelizumab: PD-1
32AdCC (+CRC)
IINCT03999684R/M-Any lineTretinoinretinoic acid receptor27AdCC
IINCT03691207R/M-Any lineAL101pan-NOTCH87AdCC
IINCT03422679LA or R/M-Any lineCB-103pan-NOTCH200AdCC (+other indications)
I/IINCT03781986R/M-Any lineAPG-115 ± carboplatinAPG-115: MDM2
Carboplatin: cytostatic drug
32AdCC
Non-AdCC
IINCT03639168R/M-Any linechidamide + cisplatinchidamide: histone deacetylase
cisplatin: cytostatic drug
22AdCC
IINCT04209660R/M-Any lineLenvatinib + pembrolizumabLenvatinib: VEGFR, FGFR, PDGFR, RET, KIT
Pembrolizumab: PD-1
64AdCC
Non-AdCC
IINCT04119453R/M-Any lineApatinibVEGFR, RET, c-KIT80AdCC
This list is not comprehensive. AdCC, adenoid cystic carcinoma; FGFR, fibroblast growth factor receptor; LA, locally advanced; NOTCH, Neurogenic locus notch homolog protein; PD-1, programmed cell death-1; PDGFR, platelet-derived growth factor receptor; PRMT5, protein arginine methyltransferase 5; Pts, patients; R/M, recurrent or metastatic; RET, rearranged during transfection; TrkA, tropomyosin receptor kinase A; SGMs, salivary gland malignancies; VEGFR, vascular endothelial growth factor receptor.

3. Novel Targets and Biomarkers

Overall, promising research is being undertaken to further unravel the etiology of SGMs by focusing o.a. on specific protein targets and molecular pathways.
Firstly, prostate-specific membrane antigen (PSMA), a type II transmembrane glycoprotein, is of high interest in SGMs. Three studies explored PSMA-targeted PET/CT performance in SGMs, majority being AdCC [147,148,149]. The only prospective study showed an added diagnostic value for PSMA radioligand PET/CT in 4 out of the 15 AdCC patients; for identification of bone metastases (2), an AdCC local recurrence in the Bartholin gland (1) and additional lymph node metastases (1). Further investigations are ongoing.
Secondly, molecular profiling of the tumor is becoming of increasing interest for all solid malignancy. Recently, decreased expression of c-kit, an epithelial to mesenchymal transition-related protein has been found in MEC. These proteins promote invasion and metastasis. Conversely, c-kit expression was associated with perineural infiltration and tumor grade in AdCC [56,150]. Pan-cancer analysis has shown an activating IGF-1R hotspot non-frameshift insertions, which were significantly enriched in AdCC. This may represent a novel subtype of AdCC which may be more susceptible to existing IGF1R inhibitors [151]. Next generation sequencing targeting NOTCH1/2/3 genes and/or NOTCH1 intra-cellular domain (NICD1) has indicated that patients with NOTCH pathway activated AdCC have worse clinical outcomes [152,153]. Novel treatment options, combined with Notch inhibitors, are therefore needed in this clinically poor group. In addition, multiple research groups are focusing on molecular abnormalities (DNA as well as RNA) which may be of use for SGMs [154,155,156,157,158,159,160]. Selecting patients based on the tumor molecular profile can be considered a major advantage and should be recommended as this can guide the development of novel therapeutic strategies according to disease biology and further personalize patient treatment. On top of this, several fusion genes are known for their involvement in salivary gland carcinoma. It is known that MYB/MYBL1-NFIB fusions are being detected in less than 60% of all AdCC [94], remaining 40% of AdCC without clear oncogenic driver mutations. Recently, Shibata et al. [161] identified six new gene fusions. Of these, NFIB-EPB41L2 and NFIB-MCMDC2 are considered to activate MYB and MYBL1 expression, thus involved in the carcinogenesis of AdCC. These targets may prove useful as potential future target for treatment of this malignancy. Comparably for MEC, it has been shown that the CRTC1/MAML2 fusion is present in up to 56% of cases [162]. Furthermore, earlier research has demonstrated that the CRTC1/MAML2 fusion is necessary for MEC cell proliferation and survival through stimulation of the EGFR signaling pathway [163,164]. In addition, Ni et al. [165] identified the critical role of Notch signaling in maintaining MEC stem-like cells and tumor growth, suggesting that co-targeting of Notch and EGFR might be a potential effective anti-MEC treatment.
Thirdly, we have already indicated that several pathways can be used by the tumor to maintain the homeostasis. Recent genetic analysis has shown that upregulation of hypoxia-inducible factor 1α (HIF-1α) upregulation enhances transcriptional activity and expression of nidogen 1, which subsequently promotes AdCC metastasis via PI3K/Akt pathway activation [166]. Comparably, Sphk1 is overexpressed in AdCC and promotes salivary tumorigenesis by activating the PI3K/Akt pathway [167]. On the other hand, the type III TGF-β receptor is significantly decreased in AdCC, allowing for NF-κB to induced cell proliferation and migration [168]. Therefore, Nidogen 1, Sphk1 and the type III TGF-β receptor could therefore serve as a novel theragnostic in AdCC, potentially in combination with an Akt- or NF-κB-inhibitor; respectively.
Fourthly, only a small proportion of patients seem to profit from ICI with the know anti PD-(L)1 inhibitors. Recently, it has been shown that activation of adenosine-signaling can result in a tolerogenic protumoral microenvironment. [169,170,171]. CD39 and CD73, two adenosine-signaling pathway markers are shown to be present in up to 48.2% and 42.9%, respectively [172]. This adenosine-signaling signature can therefore play a decisive role in tumor immunity and presents itself as a promising therapeutic option for patients with an activated adenosine-signaling pathway. Next, in their review, Witte et al. [160] have provided a nice summary of the current knowledge on the TME in SGMs (according to entity). This can further guide the development of novel (immune-) targeted therapeutic strategies per patient group. This has also been highlighted by Alame et al. [173] who stipulate that ICI or even macrophage- or NK cell-directed therapy can be highly beneficial in immune-infiltrated SDC whereas the focus should lie on molecular targets in immune-poor SDC.
Fifthly, also proteomics can provide us with some interesting targets in case of SGMs. Several proteins have been revealed to potentially aid in the diagnosis and prognosis of SGMs in a systematic review of mass spectrometry-based proteomics [174]. Annexins (ANXA) are a family of calcium-dependent, phospholipid-binding proteins that play a role in cell growth, inflammation, and signaling. Annexins A1 and A4 have been linked to benign tumors, whilst ANXA5 has been linked to malignant salivary tumors. Furthermore, ANXA5 is related with progression, invasion, and metastasis in malignant salivary gland cancers. CRYAB are small heat shock proteins that prevent denatured proteins from aggregating and sustain apoptosis and inflammation in diseases. Salivary gland cancers have been found to exhibit positive regulation of these proteins. In addition, this protein has also been found in other cancers such as breast cancer, lung cancer, prostate cancer, and ovarian cancer. It serves as an anti-apoptotic protein that downregulates the proapoptotic signaling molecules Bcl-2, Bax and caspase 3 [175]. Fibrinogen beta chain (FGB), a component of fibrinogen, is a biomarker for hepatic metastatic colorectal cancer. This protein has found to be overexpressed in malignant salivary tumors and is linked to tumor growth, invasion, and metastasis, whereas reduced expression is more prevalent in benign tumors. GNB2L1 is an intracellular scaffold protein that assists in cell growth, migration, and differentiation. Pleomorphic adenoma as well as malignant SGMs appear to have an overexpression of GNB2L1, and tis protein might therefore be used as a diagnostic marker. The glycoprotein IGHG1, which is produced by B lymphocytes, was shown to be more abundant in benign Warthin tumors and downregulated in malignant SGMs.
Despite these encouraging results, there is a lack of uniformity of proteomics data as well as the inability to determine the number of proteins in each lesion. Thus, further in-depth evaluation is highly recommended to use these proteins in a diagnostic fashion or as potential target in novel systemic therapies.
Based on this overview, various research groups are looking for ways to enhance the efficacy of the therapeutic options administered in salivary gland carcinoma. Although the full etiology remains to be elaborated, it is becoming more obvious that a deeper characterization of each separate case is needed to maximize and personalize patient care.

4. Case Description

To illustrate the high need for a more personalized approach, by use of molecular diagnostics and novel treatment targets, we provide the following case from our university center.
In November 2017, a 57-year-old patient presented with sudden peripheral paralysis of the left facial nerve of the left side on the emergency department. The patient recalled an infection with cutaneous Herpes Zoster some weeks before. An MRI of the brain observed hypercaptation of the left facial nerve in the tympanic and mastoid segment. Altogether, results indicated diagnosis of Bell’s palsy. The patient was sequentially treated with corticosteroids, electrostimulation, mime therapy and massage-therapy but no significant clinical improvement was noticed.
In August 2018, the patients’ general practitioner noticed a painless swelling and retraction of the left retroauricular region on which a CT scan was done in our institution. A necrotic, tumor mass originating from the deep lobe of the left parotic gland infiltrating the skin and facial nerve up to the stylomastoid foramen was discovered. Bilateral cervical lymph node involvement was observed. A superficial and deep biopsy revealed presence of an invasive high-grade salivary duct adenocarcinoma originating from the parotic gland with lymphovascular and perineural invasion. The immunohistochemical profile showed Cytokeratin 7 expression and diffuse androgen receptor (AR) expression, of which additional progesterone and estrogen receptors were found both negative. Her2Neu immune expression was 2+ but without amplification covered by FISH (ratio HER2/CEP17 1.42) on 20 invasive cell cores. NTRK-gene fusion was absent.
Multidisciplinary oncology consultation concluded a stage cT4a cN3b cM0. As surgical treatment was considered unfeasible, the patient was alternatively treated with primary radiotherapy combined with maximal androgene blockade of bicalutamide (Casodex®) 50 mg PO QD + leuproreline injection (Depo-Eligard®) 7.5 mg QM). A total dose of 69.12 Gy on the primary tumor and 56 Gy on the elective neck in 32 fractions was administered with curative intent. PET-CT scan viewed a regression in volume and metabolic activity of primary tumor and lymph nodes, concluding a partial remission. As tolerability to androgen blockers was unfavorable, bicalutamide (Casodex®) and leuproreline injection (Depo-Eligard®) were temporarily ceased and the patient was subsequently treated with adjuvant chemotherapy, Paclitaxel (Taxol®) 80 mg/m2 IV Q1W combined with trastuzumab (Herceptine®) 600 mg IV Q3W. Again, partial remission was attained after four cycles with an acceptable tolerance profile. Current treatment regimen was continued for another four cycles. During treatment, the patient was diagnosed with a treatment-related venous thromboembolic event with lung embolism infarction (for which low-molecular-weight heparins were initiated) but also experienced a significant recuperation of the facial nerve function. After eight cycles of therapy, stable disease was reached, and the patient was treated with trastuzumab as maintenance therapy. The patient remained in remission from July 2019 until May 2020, as progressive disease of the primary tumor was again observed. An in-house NGS sequencing panel of 69 genes was performed, revealing TP53 and PTEN mutations, and CDK4 amplification. The latter was considered an interesting target-based therapeutic approach and after several round-table discussions with clinicians and patient we decided to commence treatment with CDK4/6 inhibitor, palbociclib (Ibrands®) 125 mg PO QD combined with a reintroduction of bicalutamide (Casodex®) 50 mg PO QD. Patient shows excellent tolerability to the combination therapy. After over 80 weeks of therapy, patient presented with intermittent paresthesia in the left retroauricular region, probably due to a discrete growth of the salivary duct adenocarcinoma along the facial nerve. There was no evidence of distant metastases. Patient received methylprednisolone 64 mg QD PO for the intermittent paresthesia and patient was rechallenged, due to the successfulness in the past, to Paclitaxel (Taxol®) 80 mg/m2 IV Q1W combined with trastuzumab (Herceptine®) 600 mg IV Q3W in April 2022. However, imaging after 3 cycles of therapy indicated progressive disease with a mass increase in the left parotid lodge, further perineural invasion into the facial and auriculotemporal nerve and local lymph node involvement. Next, patient was referred for treatment with 177Lutetium-PSMA, although therapy was not administered due to an absent PSMA expression on the SDC tissue samples. Subsequently, patient recently started with Trastuzumab-Emtansine (Kadcyla®) 3.6 mg/kg IV Q3W, an antibody drug conjugate which shows excellent response in patients with HER2+ SDC, especially when treated previously with trastuzumab (Herceptine®). At current time, patient has received 2 cycles and shows only limited toxicity to the therapy (grade I AST, ALT and LDH increase).

5. Conclusions

Treatment options for advanced SGMs are currently extensively being investigated, and pathological and therapeutic progress is being made. If surgery is considered unfeasible, radiotherapy and chemotherapy are currently the standard of care. These treatments, however, are not effective in case of recurrent or metastatic disease, and the medical oncologist is left with limited treatment options. Based on published trials, drugs based on the expression of specific molecular markers may give extra disease control. Nevertheless, data from large clinical studies are lacking, therefore leaving the question open which is the most appropriate treatment option in SGMs. By means of our case report, we have clearly showed that research into novel genetic alternations as well as combination therapies in these cancers is highly recommended achieving the best possible disease control. Selecting patients based on their molecular profile should be endorsed to guide patient stratification as well as the development of novel therapeutic strategies. This especially as we pointed out that various pathways can be used by the tumor to create resistance to the specific therapies administered. This developed drug-resistance implies that timely examinations, f.i. through liquid biopsies, are a necessity in SGMs to improve patient follow-up. This will allow the medical oncologist to switch therapy based on the patient’s molecular profile, even before progression has been detected.

Author Contributions

Conceptualization, R.C. (Robbert Cleymaet), T.V., S.D.K. and S.R.; Data curation, R.C. (Robbert Cleymaet), T.V. and S.D.K.; Writing—original draft preparation, R.C. (Robbert Cleymaet) and T.V.; Writing—review and editing, R.C. (Robbert Cleymaet), T.V., R.C. (Renaat Coopman), H.V., S.D.K. and S.R.; Visualization, T.V.; Supervision, R.C. (Renaat Coopman), H.V. and S.R.; Project administration, R.C. (Robbert Cleymaet) and T.V. 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

Written informed consent has been obtained from the patient to publish this paper.

Data Availability Statement

Not applicable.

Acknowledgments

We like to acknowledge the patient for allowing to use his personal case to be illustrated in this review article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. McHugh, J.B.; Visscher, D.W.; Barnes, E.L. Update on selected salivary gland neoplasms. Arch. Pathol. Lab. Med. 2009, 133, 1763–1774. [Google Scholar] [CrossRef]
  2. Horn-Ross, P.L.; Ljung, B.M.; Morrow, M. Environmental factors and the risk of salivary gland cancer. Epidemiology 1997, 8, 414–419. [Google Scholar] [CrossRef] [PubMed]
  3. Mesolella, M.; Iengo, M.; Testa, D.; Di Lullo, A.M.; Salzano, G.; Salzano, F.A. Mucoepidermoid carcinoma of the base of tongue. Acta Otorhinolaryngol. Ital. 2015, 35, 58–61. [Google Scholar]
  4. Peraza, A.; Gomez, R.; Beltran, J.; Amarista, F.J. Mucoepidermoid carcinoma. An update and review of the literature. J. Stomatol. Oral Maxillofac. Surg. 2020, 121, 713–720. [Google Scholar] [CrossRef]
  5. Spiro, R.H. Salivary neoplasms: Overview of a 35-year experience with 2,807 patients. Head Neck Surg. 1986, 8, 177–184. [Google Scholar] [CrossRef] [PubMed]
  6. Spiro, R.H.; Huvos, A.G.; Berk, R.; Strong, E.W. Mucoepidermoid carcinoma of salivary gland origin: A clinicopathologic study of 367 cases. Am. J. Surg. 1978, 136, 461–468. [Google Scholar] [CrossRef] [PubMed]
  7. McHugh, C.H.; Roberts, D.B.; El-Naggar, A.K.; Hanna, E.Y.; Garden, A.S.; Kies, M.S.; Weber, R.S.; Kupferman, M.E. Prognostic factors in mucoepidermoid carcinoma of the salivary glands. Cancer 2012, 118, 3928–3936. [Google Scholar] [CrossRef] [PubMed]
  8. Sama, S.; Komiya, T.; Guddati, A.K. Advances in the Treatment of Mucoepidermoid Carcinoma. World J. Oncol. 2022, 13, 1–7. [Google Scholar] [CrossRef] [PubMed]
  9. De Morais, E.F.; da Silva, L.P.; Moreira, D.G.L.; Mafra, R.P.; Rolim, L.S.A.; de Moura Santos, E.; de Souza, L.B.; de Almeida Freitas, R. Prognostic Factors and Survival in Adenoid Cystic Carcinoma of the Head and Neck: A Retrospective Clinical and Histopathological Analysis of Patients Seen at a Cancer Center. Head Neck Pathol. 2021, 15, 416–424. [Google Scholar] [CrossRef] [PubMed]
  10. Srivastava, A.C.; Barpande, S.R.; Bhavthankar, J.D.; Mandale, M.S. Adenoid cystic carcinoma of palate: Report of a solid variant. J. Oral Maxillofac. Pathol. JOMFP 2018, 22 (Suppl. 1), S65–S68. [Google Scholar] [CrossRef]
  11. Xu, B.; Drill, E.; Ho, A.; Ho, A.; Dunn, L.; Prieto-Granada, C.N.; Chan, T.; Ganly, I.; Ghossein, R.; Katabi, N. Predictors of Outcome in Adenoid Cystic Carcinoma of Salivary Glands: A Clinicopathologic Study with Correlation Between MYB Fusion and Protein Expression. Am. J. Surg. Pathol. 2017, 41, 1422–1432. [Google Scholar] [CrossRef]
  12. Cantu, G. Adenoid cystic carcinoma. An indolent but aggressive tumour. Part B: Treatment and prognosis. Acta Otorhinolaryngol. Ital. 2021, 41, 296–307. [Google Scholar] [CrossRef] [PubMed]
  13. Gilbert, M.R.; Sharma, A.; Schmitt, N.C.; Johnson, J.T.; Ferris, R.L.; Duvvuri, U.; Kim, S. A 20-Year Review of 75 Cases of Salivary Duct Carcinoma. JAMA Otolaryngol. Head Neck Surg. 2016, 142, 489–495. [Google Scholar] [CrossRef] [PubMed]
  14. Nakaguro, M.; Tada, Y.; Faquin, W.C.; Sadow, P.M.; Wirth, L.J.; Nagao, T. Salivary duct carcinoma: Updates in histology, cytology, molecular biology, and treatment. Cancer Cytopathol. 2020, 128, 693–703. [Google Scholar] [CrossRef] [PubMed]
  15. D’Heygere, E.; Meulemans, J.; Vander Poorten, V. Salivary duct carcinoma. Curr. Opin. Otolaryngol. Head Neck Surg. 2018, 26, 142–151. [Google Scholar] [CrossRef]
  16. Al-Zaher, N.; Obeid, A.; Al-Salam, S.; Al-Kayyali, B.S. Acinic cell carcinoma of the salivary glands: A literature review. Hematol./Oncol. Stem Cell Ther. 2009, 2, 259–264. [Google Scholar] [CrossRef] [Green Version]
  17. Ammad Ud Din, M.; Shaikh, H. Adenoid Cystic Cancer; StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
  18. Li, N.; Xu, L.; Zhao, H.; El-Naggar, A.K.; Sturgis, E.M. A comparison of the demographics, clinical features, and survival of patients with adenoid cystic carcinoma of major and minor salivary glands versus less common sites within the Surveillance, Epidemiology, and End Results registry. Cancer 2012, 118, 3945–3953. [Google Scholar] [CrossRef]
  19. Pang, Y.; Sun, L.; Liu, H.; Ma, J. Differential diagnosis and treatment of salivary secretory carcinoma and acinic cell carcinoma. Oral Oncol. 2021, 119, 105370. [Google Scholar] [CrossRef] [PubMed]
  20. Colella, G.; Cannavale, R.; Flamminio, F.; Foschini, M.P. Fine-needle aspiration cytology of salivary gland lesions: A systematic review. J. Oral Maxillofac. Surg. 2010, 68, 2146–2153. [Google Scholar] [CrossRef]
  21. Eom, H.J.; Lee, J.H.; Ko, M.S.; Choi, Y.J.; Yoon, R.G.; Cho, K.J.; Nam, S.Y.; Baek, J.H. Comparison of fine-needle aspiration and core needle biopsy under ultrasonographic guidance for detecting malignancy and for the tissue-specific diagnosis of salivary gland tumors. Am. J. Neuroradiol. 2015, 36, 1188–1193. [Google Scholar] [CrossRef] [Green Version]
  22. Lee, Y.Y.; Wong, K.T.; King, A.D.; Ahuja, A.T. Imaging of salivary gland tumours. Eur. J. Radiol. 2008, 66, 419–436. [Google Scholar] [CrossRef]
  23. Eida, S.; Sumi, M.; Nakamura, T. Multiparametric magnetic resonance imaging for the differentiation between benign and malignant salivary gland tumors. J. Magn. Reson. Imaging 2010, 31, 673–679. [Google Scholar] [CrossRef] [PubMed]
  24. Roh, J.L.; Ryu, C.H.; Choi, S.H.; Kim, J.S.; Lee, J.H.; Cho, K.J.; Nam, S.Y.; Kim, S.Y. Clinical utility of 18F-FDG PET for patients with salivary gland malignancies. J. Nucl. Med. 2007, 48, 240–246. [Google Scholar] [PubMed]
  25. Keyes, J.W., Jr.; Harkness, B.A.; Greven, K.M.; Williams, D.W., III; Watson, N.E., Jr.; McGuirt, W.F. Salivary gland tumors: Pretherapy evaluation with PET. Radiology 1994, 192, 99–102. [Google Scholar] [CrossRef] [PubMed]
  26. Geiger, J.L.; Ismaila, N.; Beadle, B.; Caudell, J.J.; Chau, N.; Deschler, D.; Glastonbury, C.; Kaufman, M.; Lamarre, E.; Lau, H.Y.; et al. Management of Salivary Gland Malignancy: ASCO Guideline. J. Clin. Oncol. 2021, 39, 1909–1941. [Google Scholar] [CrossRef]
  27. Lombardi, D.; McGurk, M.; Vander Poorten, V.; Guzzo, M.; Accorona, R.; Rampinelli, V.; Nicolai, P. Surgical treatment of salivary malignant tumors. Oral Oncol. 2017, 65, 102–113. [Google Scholar] [CrossRef]
  28. Mannelli, G.; Comini, L.V.; Sacchetto, A.; Santoro, R.; Spinelli, G.; Bonomo, P.; Desideri, I.; Bossi, P.; Orlandi, E.; Alderotti, G.; et al. Estimating survival after salvage surgery for recurrent salivary gland cancers: Systematic review. Head Neck 2022, 44, 1961–1975. [Google Scholar] [CrossRef] [PubMed]
  29. Yu, G.; Peng, X. Conservative and functional surgery in the treatment of salivary gland tumours. Int. J. Oral Sci. 2019, 11, 22. [Google Scholar] [CrossRef] [Green Version]
  30. Lewis, A.G.; Tong, T.; Maghami, E. Diagnosis and Management of Malignant Salivary Gland Tumors of the Parotid Gland. Otolaryngol. Clin. North Am. 2016, 49, 343–380. [Google Scholar] [CrossRef]
  31. Voulgarelis, M.; Ziakas, P.D.; Papageorgiou, A.; Baimpa, E.; Tzioufas, A.G.; Moutsopoulos, H.M. Prognosis and outcome of non-Hodgkin lymphoma in primary Sjogren syndrome. Medicine 2012, 91, 1–9. [Google Scholar] [CrossRef] [PubMed]
  32. Airoldi, M.; Fornari, G.; Pedani, F.; Marchionatti, S.; Gabriele, P.; Succo, G.; Bumma, C. Paclitaxel and carboplatin for recurrent salivary gland malignancies. Anticancer Res. 2000, 20, 3781–3783. [Google Scholar] [PubMed]
  33. Airoldi, M.; Garzaro, M.; Pedani, F.; Ostellino, O.; Succo, G.; Riva, G.; Sensini, M.; Naqe, N.; Bellini, E.; Raimondo, L.; et al. Cisplatin+Vinorelbine Treatment of Recurrent or Metastatic Salivary Gland Malignancies (RMSGM): A Final Report on 60 Cases. Am. J. Clin. Oncol. 2017, 40, 86–90. [Google Scholar] [CrossRef]
  34. Airoldi, M.; Pedani, F.; Succo, G.; Gabriele, A.M.; Ragona, R.; Marchionatti, S.; Bumma, C. Phase II randomized trial comparing vinorelbine versus vinorelbine plus cisplatin in patients with recurrent salivary gland malignancies. Cancer 2001, 91, 541–547. [Google Scholar] [CrossRef] [PubMed]
  35. Belani, C.P.; Eisenberger, M.A.; Gray, W.C. Preliminary experience with chemotherapy in advanced salivary gland neoplasms. Med. Pediatr. Oncol. 1988, 16, 197–202. [Google Scholar] [CrossRef] [PubMed]
  36. Creagan, E.T.; Woods, J.E.; Rubin, J.; Schaid, D.J. Cisplatin-based chemotherapy for neoplasms arising from salivary glands and contiguous structures in the head and neck. Cancer 1988, 62, 2313–2319. [Google Scholar] [CrossRef] [PubMed]
  37. De Haan, L.D.; De Mulder, P.H.; Vermorken, J.B.; Schornagel, J.H.; Vermey, A.; Verweij, J. Cisplatin-based chemotherapy in advanced adenoid cystic carcinoma of the head and neck. Head Neck 1992, 14, 273–277. [Google Scholar] [CrossRef]
  38. Dimery, I.W.; Legha, S.S.; Shirinian, M.; Hong, W.K. Fluorouracil, doxorubicin, cyclophosphamide, and cisplatin combination chemotherapy in advanced or recurrent salivary gland carcinoma. J. Clin. Oncol. 1990, 8, 1056–1062. [Google Scholar] [CrossRef] [PubMed]
  39. Dreyfuss, A.I.; Clark, J.R.; Fallon, B.G.; Posner, M.R.; Norris, C.M., Jr.; Miller, D. Cyclophosphamide, doxorubicin, and cisplatin combination chemotherapy for advanced carcinomas of salivary gland origin. Cancer 1987, 60, 2869–2872. [Google Scholar] [CrossRef] [PubMed]
  40. Gilbert, J.; Li, Y.; Pinto, H.A.; Jennings, T.; Kies, M.S.; Silverman, P.; Forastiere, A.A. Phase II trial of taxol in salivary gland malignancies (E1394): A trial of the Eastern Cooperative Oncology Group. Head Neck 2006, 28, 197–204. [Google Scholar] [CrossRef]
  41. Hill, M.E.; Constenla, D.O.; A’Hern, R.P.; Henk, J.M.; Rhys-Evans, P.; Breach, N.; Archer, D.; Gore, M.E. Cisplatin and 5-fluorouracil for symptom control in advanced salivary adenoid cystic carcinoma. Oral Oncol. 1997, 33, 275–278. [Google Scholar] [CrossRef]
  42. Hong, M.H.; Kim, C.G.; Koh, Y.W.; Choi, E.C.; Kim, J.; Yoon, S.O.; Kim, H.R.; Cho, B.C. Efficacy and safety of vinorelbine plus cisplatin chemotherapy for patients with recurrent and/or metastatic salivary gland cancer of the head and neck. Head Neck 2018, 40, 55–62. [Google Scholar] [CrossRef]
  43. Kim, H.R.; Lee, S.J.; Park, S.; Jung, H.A.; Lee, S.H.; Jeong, H.S.; Chung, M.K.; Ahn, M.J. A Single-Arm, Prospective, Phase II Study of Cisplatin Plus Weekly Docetaxel as First-Line Therapy in Patients with Metastatic or Recurrent Salivary Gland Cancer. Cancer Res. Treat. 2022, 54, 719–727. [Google Scholar] [CrossRef] [PubMed]
  44. Laurie, S.A.; Siu, L.L.; Winquist, E.; Maksymiuk, A.; Harnett, E.L.; Walsh, W.; Tu, D.; Parulekar, W.R. A phase 2 study of platinum and gemcitabine in patients with advanced salivary gland cancer: A trial of the NCIC Clinical Trials Group. Cancer 2010, 116, 362–368. [Google Scholar] [CrossRef]
  45. Licitra, L.; Cavina, R.; Grandi, C.; Palma, S.D.; Guzzo, M.; Demicheli, R.; Molinari, R. Cisplatin, doxorubicin and cyclophosphamide in advanced salivary gland carcinoma. Ann. Oncol. 1996, 7, 640–642. [Google Scholar] [CrossRef]
  46. Licitra, L.; Marchini, S.; Spinazze, S.; Rossi, A.; Rocca, A.; Grandi, C.; Molinari, R. Cisplatin in advanced salivary gland carcinoma. A phase II study of 25 patients. Cancer 1991, 68, 1874–1877. [Google Scholar] [CrossRef]
  47. Mattox, D.E.; Von Hoff, D.D.; Balcerzak, S.P. Southwest Oncology Group study of mitoxantrone for treatment of patients with advanced adenoid cystic carcinoma of the head and neck. Investig. New Drugs 1990, 8, 105–107. [Google Scholar] [CrossRef] [PubMed]
  48. Nakano, K.; Sato, Y.; Sasaki, T.; Shimbashi, W.; Fukushima, H.; Yonekawa, H.; Mitani, H.; Kawabata, K.; Takahashi, S. Combination chemotherapy of carboplatin and paclitaxel for advanced/metastatic salivary gland carcinoma patients: Differences in responses by different pathological diagnoses. Acta Oto-Laryngol. 2016, 136, 948–951. [Google Scholar] [CrossRef] [PubMed]
  49. Schramm, V.L., Jr.; Srodes, C.; Myers, E.N. Cisplatin therapy for adenoid cystic carcinoma. Arch. Otolaryngol. 1981, 107, 739–741. [Google Scholar] [CrossRef]
  50. Tannock, I.F.; Sutherland, D.J. Chemotherapy for adenocystic carcinoma. Cancer 1980, 46, 452–454. [Google Scholar] [CrossRef] [PubMed]
  51. Triozzi, P.L.; Brantley, A.; Fisher, S.; Cole, T.B.; Crocker, I.; Huang, A.T. 5-Fluorouracil, cyclophosphamide, and vincristine for adenoid cystic carcinoma of the head and neck. Cancer 1987, 59, 887–890. [Google Scholar] [CrossRef] [PubMed]
  52. Van Herpen, C.M.; Locati, L.D.; Buter, J.; Thomas, J.; Bogaerts, J.; Lacombe, D.; de Mulder, P.; Awada, A.; Licitra, L.; Bernier, J.; et al. Phase II study on gemcitabine in recurrent and/or metastatic adenoid cystic carcinoma of the head and neck (EORTC 24982). Eur. J. Cancer 2008, 44, 2542–2545. [Google Scholar] [CrossRef] [PubMed]
  53. Vermorken, J.B.; Verweij, J.; de Mulder, P.H.; Cognetti, F.; Clavel, M.; Rodenhuis, S.; Kirkpatrick, A.; Snow, G.B. Epirubicin in patients with advanced or recurrent adenoid cystic carcinoma of the head and neck: A phase II study of the EORTC Head and Neck Cancer Cooperative Group. Ann. Oncol. 1993, 4, 785–788. [Google Scholar] [CrossRef]
  54. Verweij, J.; de Mulder, P.H.; de Graeff, A.; Vermorken, J.B.; Wildiers, J.; Kerger, J.; Schornagel, J.; Cognetti, F.; Kirkpatrick, A.; Sahmoud, T.; et al. Phase II study on mitoxantrone in adenoid cystic carcinomas of the head and neck. Ann. Oncol. 1996, 7, 867–869. [Google Scholar] [CrossRef] [PubMed]
  55. Sahara, S.; Herzog, A.E.; Nör, J.E. Systemic therapies for salivary gland adenoid cystic carcinoma. Am. J. Cancer Res. 2021, 11, 4092–4110. [Google Scholar] [PubMed]
  56. Holst, V.A.; Marshall, C.E.; Moskaluk, C.A.; Frierson, H.F., Jr. KIT protein expression and analysis of c-kit gene mutation in adenoid cystic carcinoma. Mod. Pathol. 1999, 12, 956–960. [Google Scholar]
  57. Kondo, S.; Mukudai, Y.; Soga, D.; Nishida, T.; Takigawa, M.; Shirota, T. Differential expression of vascular endothelial growth factor in high- and low-metastasis cell lines of salivary gland adenoid cystic carcinoma. Anticancer Res. 2014, 34, 671–677. [Google Scholar] [PubMed]
  58. Park, S.; Nam, S.J.; Keam, B.; Kim, T.M.; Jeon, Y.K.; Lee, S.H.; Hah, J.H.; Kwon, T.K.; Kim, D.W.; Sung, M.W.; et al. VEGF and Ki-67 Overexpression in Predicting Poor Overall Survival in Adenoid Cystic Carcinoma. Cancer Res. Treat. 2016, 48, 518–526. [Google Scholar] [CrossRef] [Green Version]
  59. Zhang, J.; Peng, B. In vitro angiogenesis and expression of nuclear factor kappaB and VEGF in high and low metastasis cell lines of salivary gland Adenoid Cystic Carcinoma. BMC Cancer 2007, 7, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Zhang, J.; Peng, B.; Chen, X. Expressions of nuclear factor kappaB, inducible nitric oxide synthase, and vascular endothelial growth factor in adenoid cystic carcinoma of salivary glands: Correlations with the angiogenesis and clinical outcome. Clin. Cancer Res. 2005, 11, 7334–7343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Myoken, Y.; Myoken, Y.; Okamoto, T.; Sato, J.D.; Kan, M.; McKeehan, W.L.; Nakahara, M.; Takada, K. Immunohistochemical study of overexpression of fibroblast growth factor-1 (FGF-1), FGF-2, and FGF receptor-1 in human malignant salivary gland tumours. J. Pathol. 1996, 178, 429–436. [Google Scholar] [CrossRef]
  62. Vekony, H.; Ylstra, B.; Wilting, S.M.; Meijer, G.A.; van de Wiel, M.A.; Leemans, C.R.; van der Waal, I.; Bloemena, E. DNA copy number gains at loci of growth factors and their receptors in salivary gland adenoid cystic carcinoma. Clin. Cancer Res. 2007, 13, 3133–3139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Gibbons, M.D.; Manne, U.; Carroll, W.R.; Peters, G.E.; Weiss, H.L.; Grizzle, W.E. Molecular differences in mucoepidermoid carcinoma and adenoid cystic carcinoma of the major salivary glands. Laryngoscope 2001, 111, 1373–1378. [Google Scholar] [CrossRef] [PubMed]
  64. Vered, M.; Braunstein, E.; Buchner, A. Immunohistochemical study of epidermal growth factor receptor in adenoid cystic carcinoma of salivary gland origin. Head Neck 2002, 24, 632–636. [Google Scholar] [CrossRef] [PubMed]
  65. Yamada, K.; Iwai, K.; Okada, Y.; Mori, M. Immunohistochemical expression of epidermal growth factor receptor in salivary gland tumours. Virchows. Arch. A 1989, 415, 523–531. [Google Scholar] [CrossRef]
  66. Baserga, R. The insulin-like growth factor-I receptor as a target for cancer therapy. Expert Opin. Ther. Targets 2005, 9, 753–768. [Google Scholar] [CrossRef]
  67. Sachdev, D.; Yee, D. Disrupting insulin-like growth factor signaling as a potential cancer therapy. Mol. Cancer Ther. 2007, 6, 1–12. [Google Scholar] [CrossRef] [Green Version]
  68. Tao, Y.; Pinzi, V.; Bourhis, J.; Deutsch, E. Mechanisms of disease: Signaling of the insulin-like growth factor 1 receptor pathway—Therapeutic perspectives in cancer. Nat. Clin. Pract. Oncol. 2007, 4, 591–602. [Google Scholar] [CrossRef]
  69. Kato, T.; Hayama, S.; Yamabuki, T.; Ishikawa, N.; Miyamoto, M.; Ito, T.; Tsuchiya, E.; Kondo, S.; Nakamura, Y.; Daigo, Y. Increased expression of insulin-like growth factor-II messenger RNA–binding protein 1 is associated with tumor progression in patients with lung cancer. Clin. Cancer Res. 2007, 13, 434–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Jones, H.E.; Gee, J.M.; Hutcheson, I.R.; Knowlden, J.M.; Barrow, D.; Nicholson, R.I. Growth factor receptor interplay and resistance in cancer. Endocr. Relat. Cancer 2006, 13 (Suppl. 1), S45–S51. [Google Scholar] [CrossRef]
  71. Suda, K.; Mizuuchi, H.; Sato, K.; Takemoto, T.; Iwasaki, T.; Mitsudomi, T. The insulin-like growth factor 1 receptor causes acquired resistance to erlotinib in lung cancer cells with the wild-type epidermal growth factor receptor. Int. J. Cancer 2014, 135, 1002–1006. [Google Scholar] [CrossRef]
  72. Morgillo, F.; Woo, J.K.; Kim, E.S.; Hong, W.K.; Lee, H.Y. Heterodimerization of insulin-like growth factor receptor/epidermal growth factor receptor and induction of survivin expression counteract the antitumor action of erlotinib. Cancer Res. 2006, 66, 10100–10111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Yang, L.; Li, J.; Ran, L.; Pan, F.; Zhao, X.; Ding, Z.; Chen, Y.; Peng, Q.; Liang, H. Phosphorylated insulin-like growth factor 1 receptor is implicated in resistance to the cytostatic effect of gefitinib in colorectal cancer cells. J. Gastrointest. Surg. 2011, 15, 942–957. [Google Scholar] [CrossRef] [PubMed]
  74. Boon, E.; Bel, M.; van Boxtel, W.; van der Graaf, W.T.A.; van Es, R.J.J.; Eerenstein, S.E.J.; Baatenburg de Jong, R.J.; van den Brekel, M.W.M.; van der Velden, L.A.; Witjes, M.J.H.; et al. A clinicopathological study and prognostic factor analysis of 177 salivary duct carcinoma patients from The Netherlands. Int. J. Cancer 2018, 143, 758–766. [Google Scholar] [CrossRef] [PubMed]
  75. Dalin, M.G.; Desrichard, A.; Katabi, N.; Makarov, V.; Walsh, L.A.; Lee, K.W.; Wang, Q.; Armenia, J.; West, L.; Dogan, S.; et al. Comprehensive Molecular Characterization of Salivary Duct Carcinoma Reveals Actionable Targets and Similarity to Apocrine Breast Cancer. Clin. Cancer Res. 2016, 22, 4623–4633. [Google Scholar] [CrossRef] [Green Version]
  76. Masubuchi, T.; Tada, Y.; Maruya, S.; Osamura, Y.; Kamata, S.E.; Miura, K.; Fushimi, C.; Takahashi, H.; Kawakita, D.; Kishimoto, S.; et al. Clinicopathological significance of androgen receptor, HER2, Ki-67 and EGFR expressions in salivary duct carcinoma. Int. J. Clin. Oncol. 2015, 20, 35–44. [Google Scholar] [CrossRef] [PubMed]
  77. Ross, J.S.; Gay, L.M.; Wang, K.; Vergilio, J.A.; Suh, J.; Ramkissoon, S.; Somerset, H.; Johnson, J.M.; Russell, J.; Ali, S.; et al. Comprehensive genomic profiles of metastatic and relapsed salivary gland carcinomas are associated with tumor type and reveal new routes to targeted therapies. Ann. Oncol. 2017, 28, 2539–2546. [Google Scholar] [CrossRef] [PubMed]
  78. Shimura, T.; Tada, Y.; Hirai, H.; Kawakita, D.; Kano, S.; Tsukahara, K.; Shimizu, A.; Takase, S.; Imanishi, Y.; Ozawa, H.; et al. Prognostic and histogenetic roles of gene alteration and the expression of key potentially actionable targets in salivary duct carcinomas. Oncotarget 2018, 9, 1852–1867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Takase, S.; Kano, S.; Tada, Y.; Kawakita, D.; Shimura, T.; Hirai, H.; Tsukahara, K.; Shimizu, A.; Imanishi, Y.; Ozawa, H.; et al. Biomarker immunoprofile in salivary duct carcinomas: Clinicopathological and prognostic implications with evaluation of the revised classification. Oncotarget 2017, 8, 59023–59035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Santana, T.; Pavel, A.; Martinek, P.; Steiner, P.; Grossmann, P.; Baneckova, M.; Skalova, A. Biomarker immunoprofile and molecular characteristics in salivary duct carcinoma: Clinicopathological and prognostic implications. Hum. Pathol. 2019, 93, 37–47. [Google Scholar] [CrossRef]
  81. Locati, L.D.; Perrone, F.; Losa, M.; Mela, M.; Casieri, P.; Orsenigo, M.; Cortelazzi, B.; Negri, T.; Tamborini, E.; Quattrone, P.; et al. Treatment relevant target immunophenotyping of 139 salivary gland carcinomas (SGCs). Oral Oncol. 2009, 45, 986–990. [Google Scholar] [CrossRef] [PubMed]
  82. Nasser, S.M.; Faquin, W.C.; Dayal, Y. Expression of androgen, estrogen, and progesterone receptors in salivary gland tumors: Frequent expression of androgen receptor in a subset of malignant salivary gland tumors. Am. J. Clin. Pathol. 2003, 119, 801–806. [Google Scholar] [CrossRef] [PubMed]
  83. Koch, U.; Radtke, F. Notch signaling in solid tumors. Curr. Top. Dev. Biol. 2010, 92, 411–455. [Google Scholar] [PubMed]
  84. Miele, L.; Golde, T.; Osborne, B. Notch signaling in cancer. Curr. Mol. Med. 2006, 6, 905–918. [Google Scholar] [CrossRef] [PubMed]
  85. Ho, A.S.; Ochoa, A.; Jayakumaran, G.; Zehir, A.; Valero Mayor, C.; Tepe, J.; Makarov, V.; Dalin, M.G.; He, J.; Bailey, M.; et al. Genetic hallmarks of recurrent/metastatic adenoid cystic carcinoma. J. Clin. Investig. 2019, 129, 4276–4289. [Google Scholar] [CrossRef]
  86. Sajed, D.P.; Faquin, W.C.; Carey, C.; Severson, E.A.; Afrogheh, A.; Johnson, C.; Blacklow, S.C.; Chau, N.G.; Lin, D.T.; Krane, J.F.; et al. Diffuse Staining for Activated NOTCH1 Correlates with NOTCH1 Mutation Status and Is Associated with Worse Outcome in Adenoid Cystic Carcinoma. Am. J. Surg. Pathol. 2017, 41, 1473–1482. [Google Scholar] [CrossRef] [PubMed]
  87. Iwai, Y.; Ishida, M.; Tanaka, Y.; Okazaki, T.; Honjo, T.; Minato, N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl. Acad. Sci. USA 2002, 99, 12293–12297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Mukaigawa, T.; Hayashi, R.; Hashimoto, K.; Ugumori, T.; Hato, N.; Fujii, S. Programmed death ligand-1 expression is associated with poor disease free survival in salivary gland carcinomas. J. Surg. Oncol. 2016, 114, 36–43. [Google Scholar] [CrossRef] [PubMed]
  89. Yearley, J.H.; Gibson, C.; Yu, N.; Moon, C.; Murphy, E.; Juco, J.; Lunceford, J.; Cheng, J.; Chow, L.Q.M.; Seiwert, T.Y.; et al. PD-L2 Expression in Human Tumors: Relevance to Anti-PD-1 Therapy in Cancer. Clin. Cancer Res. 2017, 23, 3158–3167. [Google Scholar] [CrossRef] [Green Version]
  90. Sridharan, V.; Gjini, E.; Liao, X.; Chau, N.G.; Haddad, R.I.; Severgnini, M.; Hammerman, P.; El-Naggar, A.; Freeman, G.J.; Hodi, F.S.; et al. Immune Profiling of Adenoid Cystic Carcinoma: PD-L2 Expression and Associations with Tumor-Infiltrating Lymphocytes. Cancer Immunol. Res. 2016, 4, 679–687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. 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] [PubMed]
  92. Drier, Y.; Cotton, M.J.; Williamson, K.E.; Gillespie, S.M.; Ryan, R.J.; Kluk, M.J.; Carey, C.D.; Rodig, S.J.; Sholl, L.M.; Afrogheh, A.H.; et al. An oncogenic MYB feedback loop drives alternate cell fates in adenoid cystic carcinoma. Nat. Genet. 2016, 48, 265–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Mitani, Y.; Li, J.; Rao, P.H.; Zhao, Y.J.; Bell, D.; Lippman, S.M.; Weber, R.S.; Caulin, C.; El-Naggar, A.K. Comprehensive analysis of the MYB-NFIB gene fusion in salivary adenoid cystic carcinoma: Incidence, variability, and clinicopathologic significance. Clin. Cancer Res. 2010, 16, 4722–4731. [Google Scholar] [CrossRef]
  94. Persson, M.; Andren, Y.; Mark, J.; Horlings, H.M.; Persson, F.; Stenman, G. Recurrent fusion of MYB and NFIB transcription factor genes in carcinomas of the breast and head and neck. Proc. Natl. Acad. Sci. USA 2009, 106, 18740–18744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Xu, L.H.; Zhao, F.; Yang, W.W.; Chen, C.W.; Du, Z.H.; Fu, M.; Ge, X.Y.; Li, S.L. MYB promotes the growth and metastasis of salivary adenoid cystic carcinoma. Int. J. Oncol. 2019, 54, 1579–1590. [Google Scholar] [CrossRef] [Green Version]
  96. Younes, M.N.; Park, Y.W.; Yazici, Y.D.; Gu, M.; Santillan, A.A.; Nong, X.; Kim, S.; Jasser, S.A.; El-Naggar, A.K.; Myers, J.N. Concomitant inhibition of epidermal growth factor and vascular endothelial growth factor receptor tyrosine kinases reduces growth and metastasis of human salivary adenoid cystic carcinoma in an orthotopic nude mouse model. Mol. Cancer Ther. 2006, 5, 2696–2705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Zhang, J.; Peng, B. NF-kappaB promotes iNOS and VEGF expression in salivary gland adenoid cystic carcinoma cells and enhances endothelial cell motility in vitro. Cell Prolif. 2009, 42, 150–161. [Google Scholar] [CrossRef] [PubMed]
  98. Magner, W.J.; Kazim, A.L.; Stewart, C.; Romano, M.A.; Catalano, G.; Grande, C.; Keiser, N.; Santaniello, F.; Tomasi, T.B. Activation of MHC class I, II, and CD40 gene expression by histone deacetylase inhibitors. J. Immunol. 2000, 165, 7017–7024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Adeberg, S.; Akbaba, S.; Lang, K.; Held, T.; Verma, V.; Nikoghosyan, A.; Bernhardt, D.; Münter, M.; Freier, K.; Plinkert, P.; et al. The Phase 1/2 ACCEPT Trial: Concurrent Cetuximab and Intensity Modulated Radiation Therapy with Carbon Ion Boost for Adenoid Cystic Carcinoma of the Head and Neck. Int. J. Radiat. Oncol. Biol. Phys. 2020, 106, 167–173. [Google Scholar] [CrossRef] [Green Version]
  100. Agulnik, M.; Cohen, E.W.; Cohen, R.B.; Chen, E.X.; Vokes, E.E.; Hotte, S.J.; Winquist, E.; Laurie, S.; Hayes, D.N.; Dancey, J.E.; et al. Phase II study of lapatinib in recurrent or metastatic epidermal growth factor receptor and/or erbB2 expressing adenoid cystic carcinoma and non–adenoid cystic carcinoma malignant tumors of the salivary glands. J. Clin. Oncol. 2007, 25, 3978–3984. [Google Scholar] [CrossRef]
  101. Argiris, A.; Ghebremichael, M.; Burtness, B.; Axelrod, R.S.; Deconti, R.C.; Forastiere, A.A. A phase 2 trial of bortezomib followed by the addition of doxorubicin at progression in patients with recurrent or metastatic adenoid cystic carcinoma of the head and neck: A trial of the Eastern Cooperative Oncology Group (E1303). Cancer 2011, 117, 3374–3382. [Google Scholar] [CrossRef] [Green Version]
  102. Aung, K.L.; El-Khoueiry, A.B.; Gelmon, K.; Tran, B.; Bajaj, G.; He, B.; Chen, T.; Zhu, L.; Poojary, S.; Basak, S.; et al. A multi-arm phase I dose escalating study of an oral NOTCH inhibitor BMS-986115 in patients with advanced solid tumours. Investig. New Drugs 2018, 36, 1026–1036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Bahl, A.; Panda, N.K.; Elangovan, A.; Bakshi, J.; Verma, R.; Mohindra, S.; Gupta, R.; Oinam, A.S.; Kaur, S.; Vashishta, R.K.; et al. Evaluation of Multimodality Management of Adenoid Cystic Carcinoma of the Head and Neck. Indian J. Otolaryngol. Head Neck Surg. 2019, 71 (Suppl. 1), 628–632. [Google Scholar] [CrossRef]
  104. Calvo, E.; Soria, J.C.; Ma, W.W.; Wang, T.; Bahleda, R.; Tolcher, A.W.; Gernhardt, D.; O’Connell, J.; Millham, R.; Giri, N.; et al. A Phase I Clinical Trial and Independent Patient-Derived Xenograft Study of Combined Targeted Treatment with Dacomitinib and Figitumumab in Advanced Solid Tumors. Clin. Cancer Res. 2017, 23, 1177–1185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Chau, N.G.; Hotte, S.J.; Chen, E.X.; Chin, S.F.; Turner, S.; Wang, L.; Siu, L.L. A phase II study of sunitinib in recurrent and/or metastatic adenoid cystic carcinoma (ACC) of the salivary glands: Current progress and challenges in evaluating molecularly targeted agents in ACC. Ann. Oncol. 2012, 23, 1562–1570. [Google Scholar] [CrossRef] [PubMed]
  106. 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] [PubMed]
  107. Dillon, P.M.; Petroni, G.R.; Horton, B.J.; Moskaluk, C.A.; Fracasso, P.M.; Douvas, M.G.; Varhegyi, N.; Zaja-Milatovic, S.; Thomas, C.Y. A Phase II Study of Dovitinib in Patients with Recurrent or Metastatic Adenoid Cystic Carcinoma. Clin. Cancer Res. 2017, 23, 4138–4145. [Google Scholar] [CrossRef] [Green Version]
  108. Dogan, S.; Ng, C.K.Y.; Xu, B.; Kumar, R.; Wang, L.; Edelweiss, M.; Scott, S.N.; Zehir, A.; Drilon, A.; Morris, L.G.T.; et al. The repertoire of genetic alterations in salivary duct carcinoma including a novel HNRNPH3-ALK rearrangement. Hum. Pathol. 2019, 88, 66–77. [Google Scholar] [CrossRef]
  109. Even, C.; Delord, J.P.; Price, K.A.; Nakagawa, K.; Oh, D.Y.; Burge, M.; Chung, H.C.; Doi, T.; Fakih, M.; Takahashi, S.; et al. Evaluation of pembrolizumab monotherapy in patients with previously treated advanced salivary gland carcinoma in the phase 2 KEYNOTE-158 study. Eur. J. Cancer 2022, 171, 259–268. [Google Scholar] [CrossRef]
  110. Even, C.; Lassen, U.; Merchan, J.; Le Tourneau, C.; Soria, J.C.; Ferte, C.; Ricci, F.; Diener, J.T.; Yuen, E.; Smith, C.; et al. Safety and clinical activity of the Notch inhibitor, crenigacestat (LY3039478), in an open-label phase I trial expansion cohort of advanced or metastatic adenoid cystic carcinoma. Investig. New Drugs 2020, 38, 402–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Feeney, L.; Jain, Y.; Beasley, M.; Donnelly, O.; Kong, A.; Moleron, R.; Nallathambi, C.; Rolles, M.; Sanghera, P.; Tin, A.; et al. Centralised RECIST Assessment and Clinical Outcomes with Lenvatinib Monotherapy in Recurrent and Metastatic Adenoid Cystic Carcinoma. Cancers 2021, 13, 4336. [Google Scholar] [CrossRef]
  112. Ferrarotto, R.; Eckhardt, G.; Patnaik, A.; LoRusso, P.; Faoro, L.; Heymach, J.V.; Kapoun, A.M.; Xu, L.; Munster, P. A phase I dose-escalation and dose-expansion study of brontictuzumab in subjects with selected solid tumors. Ann. Oncol. 2018, 29, 1561–1568. [Google Scholar] [CrossRef]
  113. Fushimi, C.; Tada, Y.; Takahashi, H.; Nagao, T.; Ojiri, H.; Masubuchi, T.; Matsuki, T.; Miura, K.; Kawakita, D.; Hirai, H.; et al. A prospective phase II study of combined androgen blockade in patients with androgen receptor-positive metastatic or locally advanced unresectable salivary gland carcinoma. Ann. Oncol. 2018, 29, 979–984. [Google Scholar] [CrossRef] [PubMed]
  114. Ghosal, N.; Mais, K.; Shenjere, P.; Julyan, P.; Hastings, D.; Ward, T.; Ryder, W.D.; Bruce, I.; Homer, J.; Slevin, N.J. Phase II study of cisplatin and imatinib in advanced salivary adenoid cystic carcinoma. Br. J. Oral Maxillofac. Surg. 2011, 49, 510–515. [Google Scholar] [CrossRef] [PubMed]
  115. 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] [PubMed] [Green Version]
  116. Haddad, R.; Colevas, A.D.; Krane, J.F.; Cooper, D.; Glisson, B.; Amrein, P.C.; Weeks, L.; Costello, R.; Posner, M. Herceptin in patients with advanced or metastatic salivary gland carcinomas. A phase II study. Oral Oncol. 2003, 39, 724–727. [Google Scholar] [CrossRef] [PubMed]
  117. Hitre, E.; Budai, B.; Takácsi-Nagy, Z.; Rubovszky, G.; Tóth, E.; Remenár, É.; Polgár, C.; Láng, I. Cetuximab and platinum-based chemoradio- or chemotherapy of patients with epidermal growth factor receptor expressing adenoid cystic carcinoma: A phase II trial. Br. J. Cancer 2013, 109, 1117–1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Ho, A.L.; Dunn, L.; Sherman, E.J.; Fury, M.G.; Baxi, S.S.; Chandramohan, R.; Dogan, S.; Morris, L.G.; Cullen, G.D.; Haque, S.; et al. A phase II study of axitinib (AG-013736) in patients with incurable adenoid cystic carcinoma. Ann. Oncol. 2016, 27, 1902–1908. [Google Scholar] [CrossRef] [PubMed]
  119. Ho, A.L.; Foster, N.R.; Zoroufy, A.J.; Campbell, J.D.; Worden, F.; Price, K.; Adkins, D.; Bowles, D.W.; Kang, H.; Burtness, B.; et al. Phase II Study of Enzalutamide for Patients with Androgen Receptor–Positive Salivary Gland Cancers (Alliance A091404). J. Clin. Oncol. 2022. online ahead of print. [Google Scholar] [CrossRef]
  120. Ho, A.L.; Sherman, E.J.; Baxi, S.S.; Haque, S.; Ni, A.; Antonescu, C.R.; Katabi, N.; Morris, L.G.; Chan, T.A.; Pfister, D.G. Phase II study of regorafenib in progressive, recurrent/metastatic adenoid cystic carcinoma. J. Clin. Oncol. 2016, 34, 6096. [Google Scholar] [CrossRef]
  121. Hoover, A.C.; Milhem, M.M.; Anderson, C.M.; Sun, W.; Smith, B.J.; Hoffman, H.T.; Buatti, J.M. Efficacy of nelfinavir as monotherapy in refractory adenoid cystic carcinoma: Results of a phase II clinical trial. Head Neck 2015, 37, 722–726. [Google Scholar] [CrossRef] [PubMed]
  122. Hotte, S.J.; Winquist, E.W.; Lamont, E.; MacKenzie, M.; Vokes, E.; Chen, E.X.; Brown, S.; Pond, G.R.; Murgo, A.; Siu, L.L. Imatinib mesylate in patients with adenoid cystic cancers of the salivary glands expressing c-kit: A Princess Margaret Hospital phase II consortium study. J. Clin. Oncol. 2005, 23, 585–590. [Google Scholar] [CrossRef] [PubMed]
  123. Jakob, J.A.; Kies, M.S.; Glisson, B.S.; Kupferman, M.E.; Liu, D.D.; Lee, J.J.; El-Naggar, A.K.; Gonzalez-Angulo, A.M.; Blumenschein, G.R., Jr. Phase II study of gefitinib in patients with advanced salivary gland cancers. Head Neck 2015, 37, 644–649. [Google Scholar] [CrossRef] [PubMed]
  124. Kang, E.J.; Ahn, M.J.; Ock, C.Y.; Lee, K.W.; Kwon, J.H.; Yang, Y.; Choi, Y.H.; Kim, M.K.; Ji, J.H.; Yun, T.; et al. Randomized Phase II Study of Axitinib versus Observation in Patients with Recurred or Metastatic Adenoid Cystic Carcinoma. Clin. Cancer Res. 2021, 27, 5272–5279. [Google Scholar] [CrossRef] [PubMed]
  125. Keam, B.; Kim, S.B.; Shin, S.H.; Cho, B.C.; Lee, K.W.; Kim, M.K.; Yun, H.J.; Lee, S.H.; Yoon, D.H.; Bang, Y.J. Phase 2 study of dovitinib in patients with metastatic or unresectable adenoid cystic carcinoma. Cancer 2015, 121, 2612–2617. [Google Scholar] [CrossRef] [PubMed]
  126. Kim, D.W.; Oh, D.Y.; Shin, S.H.; Kang, J.H.; Cho, B.C.; Chung, J.S.; Kim, H.; Park, K.U.; Kwon, J.H.; Han, J.Y.; et al. A multicenter phase II study of everolimus in patients with progressive unresectable adenoid cystic carcinoma. BMC Cancer 2014, 14, 795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Kim, Y.; Lee, S.J.; Lee, J.Y.; Lee, S.H.; Sun, J.M.; Park, K.; An, H.J.; Cho, J.Y.; Kang, E.J.; Lee, H.Y.; et al. Clinical trial of nintedanib in patients with recurrent or metastatic salivary gland cancer of the head and neck: A multicenter phase 2 study (Korean Cancer Study Group HN14-01). Cancer 2017, 123, 1958–1964. [Google Scholar] [CrossRef] [Green Version]
  128. Kurzrock, R.; Bowles, D.W.; Kang, H.; Meric-Bernstam, F.; Hainsworth, J.; Spigel, D.R.; Bose, R.; Burris, H.; Sweeney, C.J.; Beattie, M.S.; et al. Targeted therapy for advanced salivary gland carcinoma based on molecular profiling: Results from MyPathway, a phase IIa multiple basket study. Ann. Oncol. 2020, 31, 412–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Li, B.T.; Shen, R.; Offin, M.; Buonocore, D.J.; Myers, M.L.; Venkatesh, A.; Razavi, P.; Ginsberg, M.S.; Ulaner, G.A.; Solit, D.B.; et al. Ado-trastuzumab emtansine in patients with HER2 amplified salivary gland cancers (SGCs): Results from a phase II basket trial. J. Clin. Oncol. 2019, 37, 6001. [Google Scholar] [CrossRef]
  130. Locati, L.D.; Cavalieri, S.; Bergamini, C.; Resteghini, C.; Alfieri, S.; Calareso, G.; Bossi, P.; Perrone, F.; Tamborini, E.; Quattrone, P.; et al. Phase II trial with axitinib in recurrent and/or metastatic salivary gland cancers of the upper aerodigestive tract. Head Neck 2019, 41, 3670–3676. [Google Scholar] [CrossRef] [PubMed]
  131. Locati, L.D.; Cavalieri, S.; Bergamini, C.; Resteghini, C.; Colombo, E.; Calareso, G.; Mariani, L.; Quattrone, P.; Alfieri, S.; Bossi, P.; et al. Abiraterone Acetate in Patients with Castration-Resistant, Androgen Receptor-Expressing Salivary Gland Cancer: A Phase II Trial. J. Clin. Oncol. 2021, 39, 4061–4068. [Google Scholar] [CrossRef] [PubMed]
  132. Locati, L.D.; Galbiati, D.; Calareso, G.; Alfieri, S.; Singer, S.; Cavalieri, S.; Bergamini, C.; Bossi, P.; Orlandi, E.; Resteghini, C.; et al. Patients with adenoid cystic carcinomas of the salivary glands treated with lenvatinib: Activity and quality of life. Cancer 2020, 126, 1888–1894. [Google Scholar] [CrossRef]
  133. Locati, L.D.; Perrone, F.; Cortelazzi, B.; Bergamini, C.; Bossi, P.; Civelli, E.; Morosi, C.; Lo Vullo, S.; Imbimbo, M.; Quattrone, P.; et al. A phase II study of sorafenib in recurrent and/or metastatic salivary gland carcinomas: Translational analyses and clinical impact. Eur. J. Cancer 2016, 69, 158–165. [Google Scholar] [CrossRef]
  134. Mahadevan, D.; Sutton, G.R.; Arteta-Bulos, R.; Bowden, C.J.; Miller, P.J.; Swart, R.E.; Walker, M.S.; Haluska, P.; Munster, P.N.; Marshall, J.; et al. Phase 1b study of safety, tolerability and efficacy of R1507, a monoclonal antibody to IGF-1R in combination with multiple standard oncology regimens in patients with advanced solid malignancies. Cancer Chemother. Pharmacol. 2014, 73, 467–473. [Google Scholar] [CrossRef]
  135. Mahmood, U.; Bang, A.; Chen, Y.H.; Mak, R.H.; Lorch, J.H.; Hanna, G.J.; Nishino, M.; Manuszak, C.; Thrash, E.M.; Severgnini, M.; et al. A Randomized Phase 2 Study of Pembrolizumab with or without Radiation in Patients with Recurrent or Metastatic Adenoid Cystic Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2021, 109, 134–144. [Google Scholar] [CrossRef] [PubMed]
  136. Pfeffer, M.R.; Talmi, Y.; Catane, R.; Symon, Z.; Yosepovitch, A.; Levitt, M. A phase II study of Imatinib for advanced adenoid cystic carcinoma of head and neck salivary glands. Oral Oncol. 2007, 43, 33–36. [Google Scholar] [CrossRef]
  137. Rodriguez, C.P.; Martins, R.G.; Baik, C.; Chow, L.Q.; Santana-Davila, R.; Goulart, B.H.; Lee, S.; Eaton, K.D. Phase II trial of eribulin mesylate in recurrent or metastatic salivary gland malignancies. Head Neck 2018, 40, 584–589. [Google Scholar] [CrossRef] [PubMed]
  138. 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] [PubMed]
  139. Siu, L.L.; Rasco, D.W.; Postel Vinay, S.; Martin Romano, P.; Menis, J.; Opdam, F.L.; Heinhuis, K.M.; Egger, J.L.; Gorman, S.A.; Parasrampuria, R.; et al. METEOR-1: A phase I study of GSK3326595, a first-in-class protein arginine methyltransferase 5 (PRMT5) inhibitor, in advanced solid tumours. Ann. Oncol. 2019, 30, v159. [Google Scholar] [CrossRef]
  140. Takahashi, H.; Tada, Y.; Saotome, T.; Akazawa, K.; Ojiri, H.; Fushimi, C.; Masubuchi, T.; Matsuki, T.; Tani, K.; Osamura, R.Y.; et al. Phase II Trial of Trastuzumab and Docetaxel in Patients with Human Epidermal Growth Factor Receptor 2–Positive Salivary Duct Carcinoma. J. Clin. Oncol. 2019, 37, 125–134. [Google Scholar] [CrossRef]
  141. Tchekmedyian, V.; Sherman, E.J.; Dunn, L.; Tran, C.; Baxi, S.; Katabi, N.; Antonescu, C.R.; Ostrovnaya, I.; Haque, S.S.; Pfister, D.G.; et al. Phase II Study of Lenvatinib in Patients with Progressive, Recurrent or Metastatic Adenoid Cystic Carcinoma. J. Clin. Oncol. 2019, 37, 1529–1537. [Google Scholar] [CrossRef] [PubMed]
  142. Thomson, D.J.; Silva, P.; Denton, K.; Bonington, S.; Mak, S.K.; Swindell, R.; Homer, J.; Sykes, A.J.; Lee, L.W.; Yap, B.K.; et al. Phase II trial of sorafenib in advanced salivary adenoid cystic carcinoma of the head and neck. Head Neck 2015, 37, 182–187. [Google Scholar] [CrossRef]
  143. Van Boxtel, W.; Uijen, M.J.M.; Krens, S.D.; Dijkema, T.; Willems, S.M.; Jonker, M.A.; Pegge, S.A.H.; van Engen-van Grunsven, A.C.H.; van Herpen, C.M.L. Excessive toxicity of cabozantinib in a phase II study in patients with recurrent and/or metastatic salivary gland cancer. Eur. J. Cancer 2022, 161, 128–137. [Google Scholar] [CrossRef]
  144. Wong, S.J.; Karrison, T.; Hayes, D.N.; Kies, M.S.; Cullen, K.J.; Tanvetyanon, T.; Argiris, A.; Takebe, N.; Lim, D.; Saba, N.F.; et al. Phase II trial of dasatinib for recurrent or metastatic c-KIT expressing adenoid cystic carcinoma and for nonadenoid cystic malignant salivary tumors. Ann. Oncol. 2016, 27, 318–323. [Google Scholar] [CrossRef] [Green Version]
  145. Zhu, G.; Zhang, L.; Dou, S.; Li, R.; Li, J.; Ye, L.; Jiang, W.; Dong, M.; Ruan, M.; Yang, W.; et al. Apatinib in patients with recurrent or metastatic adenoid cystic carcinoma of the head and neck: A single-arm, phase II prospective study. Ther. Adv. Med. Oncol. 2021, 13, 17588359211013626. [Google Scholar] [CrossRef]
  146. 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]
  147. Klein Nulent, T.J.W.; van Es, R.J.J.; Krijger, G.C.; de Bree, R.; Willems, S.M.; de Keizer, B. Prostate-specific membrane antigen PET imaging and immunohistochemistry in adenoid cystic carcinoma-a preliminary analysis. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 1614–1621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Klein Nulent, T.J.W.; van Es, R.J.J.; Willems, S.M.; Braat, A.; Devriese, L.A.; de Bree, R.; de Keizer, B. First experiences with (177)Lu-PSMA-617 therapy for recurrent or metastatic salivary gland cancer. EJNMMI Res. 2021, 11, 126. [Google Scholar] [CrossRef] [PubMed]
  149. Van Boxtel, W.; Lutje, S.; van Engen-van Grunsven, I.C.H.; Verhaegh, G.W.; Schalken, J.A.; Jonker, M.A.; Nagarajah, J.; Gotthardt, M.; van Herpen, C.M.L. (68)Ga-PSMA-HBED-CC PET/CT imaging for adenoid cystic carcinoma and salivary duct carcinoma: A phase 2 imaging study. Theranostics 2020, 10, 2273–2283. [Google Scholar] [CrossRef] [PubMed]
  150. Kerche, L.E.; de Sousa, E.A.; Squarize, C.H.; Oliveira, K.K.; Marchi, F.A.; Bettim, B.B.; Kowalski, L.P.; Soares, F.A.; Lourenco, S.V.; Coutinho-Camillo, C.M. EMT in salivary gland tumors: The expression of microRNAs miR-155 and miR-200c is associated with clinical-pathological parameters. Mol. Biol. Rep. 2022, 49, 2157–2167. [Google Scholar] [CrossRef]
  151. Margolis, M.; Janovitz, T.; Laird, J.; Mata, D.A.; Montesion, M.; Lee, J.K.; Madison, R.W.; Schrock, A.B.; Tukachinsky, H.; Allen, J.M.; et al. Activating IGF1R hotspot non-frameshift insertions define a novel, potentially targetable molecular subtype of adenoid cystic carcinoma. Mod. Pathol. 2022, 35, 1618–1623. [Google Scholar] [CrossRef]
  152. De Sousa, L.G.; Jovanovic, K.; Ferrarotto, R. Metastatic Adenoid Cystic Carcinoma: Genomic Landscape and Emerging Treatments. Curr. Treat. Options Oncol. 2022, 23, 1135–1150. [Google Scholar] [CrossRef]
  153. Feeney, L.; Hapuarachi, B.; Adderley, H.; Rack, S.; Morgan, D.; Walker, R.; Rauch, R.; Herz, E.; Kaye, J.; Harrington, K.; et al. Clinical disease course and survival outcomes following disease recurrence in adenoid cystic carcinoma with and without NOTCH signaling pathway activation. Oral Oncol. 2022, 133, 106028. [Google Scholar] [CrossRef]
  154. Di Villeneuve, L.; Souza, I.L.; Tolentino, F.D.S.; Ferrarotto, R.; Schvartsman, G. Salivary Gland Carcinoma: Novel Targets to Overcome Treatment Resistance in Advanced Disease. Front. Oncol. 2020, 10, 580141. [Google Scholar] [CrossRef]
  155. Ferrarotto, R.; Mitani, Y.; McGrail, D.J.; Li, K.; Karpinets, T.V.; Bell, D.; Frank, S.J.; Song, X.; Kupferman, M.E.; Liu, B.; et al. Proteogenomic Analysis of Salivary Adenoid Cystic Carcinomas Defines Molecular Subtypes and Identifies Therapeutic Targets. Clin. Cancer Res. 2021, 27, 852–864. [Google Scholar] [CrossRef]
  156. Karpinets, T.V.; Mitani, Y.; Liu, B.; Zhang, J.; Pytynia, K.B.; Sellen, L.D.; Karagiannis, D.T.; Ferrarotto, R.; Futreal, A.P.; El-Naggar, A.K. Whole-Genome Sequencing of Common Salivary Gland Carcinomas: Subtype-Restricted and Shared Genetic Alterations. Clin. Cancer Res. 2021, 27, 3960–3969. [Google Scholar] [CrossRef]
  157. Miller, L.E.; Au, V.; Mokhtari, T.E.; Goss, D.; Faden, D.L.; Varvares, M.A. A Contemporary Review of Molecular Therapeutic Targets for Adenoid Cystic Carcinoma. Cancers 2022, 14, 992. [Google Scholar] [CrossRef]
  158. Mueller, S.K.; Haderlein, M.; Lettmaier, S.; Agaimy, A.; Haller, F.; Hecht, M.; Fietkau, R.; Iro, H.; Mantsopoulos, K. Targeted Therapy, Chemotherapy, Immunotherapy and Novel Treatment Options for Different Subtypes of Salivary Gland Cancer. J. Clin. Med. 2022, 11, 720. [Google Scholar] [CrossRef] [PubMed]
  159. Vahabi, M.; Blandino, G.; Di Agostino, S. MicroRNAs in head and neck squamous cell carcinoma: A possible challenge as biomarkers, determinants for the choice of therapy and targets for personalized molecular therapies. Transl. Cancer Res. 2021, 10, 3090–3110. [Google Scholar] [CrossRef] [PubMed]
  160. Witte, H.M.; Gebauer, N.; Steinestel, K. Mutational and immunologic Landscape in malignant Salivary Gland Tumors harbor the potential for novel therapeutic strategies. Crit. Rev. Oncol. Hematol. 2022, 170, 103592. [Google Scholar] [CrossRef] [PubMed]
  161. Shibata, E.; Morita, K.I.; Kayamori, K.; Tange, S.; Shibata, H.; Harazono, Y.; Michi, Y.; Ikeda, T.; Harada, H.; Imoto, I.; et al. Detection of novel fusion genes by next-generation sequencing-based targeted RNA sequencing analysis in adenoid cystic carcinoma of head and neck. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2021, 132, 426–433. [Google Scholar] [CrossRef] [PubMed]
  162. Saade, R.E.; Bell, D.; Garcia, J.; Roberts, D.; Weber, R. Role of CRTC1/MAML2 Translocation in the Prognosis and Clinical Outcomes of Mucoepidermoid Carcinoma. JAMA Otolaryngol.–Head Neck Surg. 2016, 142, 234–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Chen, Z.; Chen, J.; Gu, Y.; Hu, C.; Li, J.L.; Lin, S.; Shen, H.; Cao, C.; Gao, R.; Li, J.; et al. Aberrantly activated AREG–EGFR signaling is required for the growth and survival of CRTC1–MAML2 fusion-positive mucoepidermoid carcinoma cells. Oncogene 2014, 33, 3869–3877. [Google Scholar] [CrossRef]
  164. Chen, Z.; Ni, W.; Li, J.L.; Lin, S.; Zhou, X.; Sun, Y.; Li, J.W.; Leon, M.E.; Hurtado, M.D.; Zolotukhin, S.; et al. The CRTC1-MAML2 fusion is the major oncogenic driver in mucoepidermoid carcinoma. JCI Insight 2021, 6, e139497. [Google Scholar] [CrossRef]
  165. Ni, W.; Chen, Z.; Zhou, X.; Yang, R.; Yu, M.; Lu, J.; Kaye, F.J.; Wu, L. Targeting Notch and EGFR signaling in human mucoepidermoid carcinoma. Signal Transduct. Target. Ther. 2021, 6, 27. [Google Scholar] [CrossRef] [PubMed]
  166. Han, N.; Li, X.; Wang, Y.; Li, H.; Zhang, C.; Zhao, X.; Zhang, Z.; Ruan, M.; Zhang, C. HIF-1alpha induced NID1 expression promotes pulmonary metastases via the PI3K-AKT pathway in salivary gland adenoid cystic carcinoma. Oral Oncol. 2022, 131, 105940. [Google Scholar] [CrossRef] [PubMed]
  167. Hou, C.X.; Wang, L.; Cai, M.; Meng, Y.; Tang, Y.T.; Zhu, Q.H.; Han, W.; Sun, N.N.; Ma, B.; Hu, Y.; et al. Sphk1 promotes salivary adenoid cystic carcinoma progression via PI3K/Akt signaling. Pathol. Res. Pract. 2021, 227, 153620. [Google Scholar] [CrossRef] [PubMed]
  168. Xu, D.; Li, D.; Lu, Z.; Dong, X.; Wang, X. Type III TGF-beta receptor inhibits cell proliferation and migration in salivary glands adenoid cystic carcinoma by suppressing NF-kappaB signaling. Oncol. Rep. 2016, 35, 267–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Allard, B.; Longhi, M.S.; Robson, S.C.; Stagg, J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol. Rev. 2017, 276, 121–144. [Google Scholar] [CrossRef] [Green Version]
  170. Allard, D.; Allard, B.; Stagg, J. On the mechanism of anti-CD39 immune checkpoint therapy. J. Immunother. Cancer 2020, 8, e000186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Elliott, M.R.; Chekeni, F.B.; Trampont, P.C.; Lazarowski, E.R.; Kadl, A.; Walk, S.F.; Park, D.; Woodson, R.I.; Ostankovich, M.; Sharma, P.; et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature 2009, 461, 282–286. [Google Scholar] [CrossRef] [Green Version]
  172. Bauer, A.; Gebauer, N.; Knief, J.; Tharun, L.; Arnold, N.; Riecke, A.; Steinestel, K.; Witte, H.M. The expression of the adenosine pathway markers CD39 and CD73 in salivary gland carcinomas harbors the potential for novel immune checkpoint inhibition. J. Cancer Res. Clin. Oncol. 2022. online ahead of print. [Google Scholar] [CrossRef]
  173. Alame, M.; Cornillot, E.; Cacheux, V.; Tosato, G.; Four, M.; De Oliveira, L.; Gofflot, S.; Delvenne, P.; Turtoi, E.; Cabello-Aguilar, S.; et al. The molecular landscape and microenvironment of salivary duct carcinoma reveal new therapeutic opportunities. Theranostics 2020, 10, 4383–4394. [Google Scholar] [CrossRef] [PubMed]
  174. De Lima-Souza, R.A.; Scarini, J.F.; Lavareze, L.; Emerick, C.; Dos Santos, E.S.; Paes Leme, A.F.; Egal, E.S.A.; Altemani, A.; Mariano, F.V. Protein markers of primary salivary gland tumors: A systematic review of proteomic profiling studies. Arch. Oral Biol. 2022, 136, 105373. [Google Scholar] [CrossRef] [PubMed]
  175. Hu, W.F.; Gong, L.; Cao, Z.; Ma, H.; Ji, W.; Deng, M.; Liu, M.; Hu, X.H.; Chen, P.; Yan, Q.; et al. alphaA- and alphaB-crystallins interact with caspase-3 and Bax to guard mouse lens development. Curr. Mol. Med. 2012, 12, 177–187. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of receptors and molecular pathways involved in SGMs. Illustration of the receptors and intracellular pathways that can initiate angiogenesis, proliferation, and survival in SGMs. Red spheres indicate targeted therapies which have been developed for numerous of these receptors and intracellular molecules: 1: abiraterone, bicalutamide, enzalutamide, leuprorelin; 2: apatinib, axitinib, dasatinib, dovotinib, imatinib, lenvatinib, sorafenib, sunitinib; 3: cetuximab, dacomitinib, gefitinib, lapatinib; 4: dovotinib, nintedanib, lenvatinib, regorafenib; 5: cabozantinib, dovotinib, sorafenib, sunitinib; 6: lapatinib, pertuzumab, trastazumab; 7: figitumumab, R1507; 8: everolimus; 9: bortezomib; 10: AL101, BMS-986115, brontictuzumab, CB-103, crenigacestat; 11: axitinib, dovotinib, imatinib, lenvatinib, nintedanib, regorafenib, sorafenib, sunitinib; 12: apatinib, axitinib, cabozantinib, dovotinib, lenvatinib, nintedanib, regorafenib, sorafenib, sunitinib; 13: apatinib, cabozantinib, dovotinib, lenvatinib, sorafenib, sunitinib; 14: nelfinavir; 15: VMD-928; and 16: APG-115. It is clear that SGMs have a multitude of pathways which can sustain carcinogenesis and therefore provide the carcinoma with an excellent tool for drug resistance to a specific targeted therapy. AR, androgen receptor; EGFR, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; FLT3, fms-like tyrosine kinase 3; HER, human epidermal growth factor receptor; IGF1R, insulin-like growth factor 1 receptor; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor-κB; NOTCH, Neurogenic locus notch homolog protein; PDGFR, platelet-derived growth factor receptor; RET, rearranged during transfection; SGMs, salivary gland malignancies; Trk, tropomyosin receptor kinase; VEGFR, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor.
Figure 1. Overview of receptors and molecular pathways involved in SGMs. Illustration of the receptors and intracellular pathways that can initiate angiogenesis, proliferation, and survival in SGMs. Red spheres indicate targeted therapies which have been developed for numerous of these receptors and intracellular molecules: 1: abiraterone, bicalutamide, enzalutamide, leuprorelin; 2: apatinib, axitinib, dasatinib, dovotinib, imatinib, lenvatinib, sorafenib, sunitinib; 3: cetuximab, dacomitinib, gefitinib, lapatinib; 4: dovotinib, nintedanib, lenvatinib, regorafenib; 5: cabozantinib, dovotinib, sorafenib, sunitinib; 6: lapatinib, pertuzumab, trastazumab; 7: figitumumab, R1507; 8: everolimus; 9: bortezomib; 10: AL101, BMS-986115, brontictuzumab, CB-103, crenigacestat; 11: axitinib, dovotinib, imatinib, lenvatinib, nintedanib, regorafenib, sorafenib, sunitinib; 12: apatinib, axitinib, cabozantinib, dovotinib, lenvatinib, nintedanib, regorafenib, sorafenib, sunitinib; 13: apatinib, cabozantinib, dovotinib, lenvatinib, sorafenib, sunitinib; 14: nelfinavir; 15: VMD-928; and 16: APG-115. It is clear that SGMs have a multitude of pathways which can sustain carcinogenesis and therefore provide the carcinoma with an excellent tool for drug resistance to a specific targeted therapy. AR, androgen receptor; EGFR, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; FLT3, fms-like tyrosine kinase 3; HER, human epidermal growth factor receptor; IGF1R, insulin-like growth factor 1 receptor; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor-κB; NOTCH, Neurogenic locus notch homolog protein; PDGFR, platelet-derived growth factor receptor; RET, rearranged during transfection; SGMs, salivary gland malignancies; Trk, tropomyosin receptor kinase; VEGFR, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor.
Ijms 23 14891 g001
Table 2. Overview of chemotherapies tested in SGMs.
Table 2. Overview of chemotherapies tested in SGMs.
StudySettingAgentTargetPts, nSubtype *ORR, n (%) *Median PFS (Months)Median OS (Months)
A. Monotherapy
Retro [50]R/M
Any line
5-fluorouracilcytostatic drug12AdCC4 (33)ITT: 21.0
II [49]R/M
Any line
Cisplatincytostatic drug10AdCC7 (70)
II [46]R/M
First- or second-line
Cisplatincytostatic drug25AdCC: 13
Non-AdCC: 12
(5 MEC, 5 adeno, 1 AcCC, 1 SCC)
ITT: 4 (16)
AdCC: 2 (15)
Non-AdCC: 2 (17)
(1 MEC, 1 SCC)
7.014.0
II [37]R/M
First- or second-line
Cisplatincytostatic drug10AdCC0 (0)3.021.0
Retro [36]R/M
First-line
Cisplatincytostatic drugs34AdCC: 11
Non-AdCC: 23
(7 MEC, 14 adeno, 2 mixed)
ITT: 13 (38)
AdCC: 2 (18)
Non-AdCC: 11 (48)
(3 MEC, 7 adeno, 1 mixed)
7.015.0
II [53]R/M
First- or second-line
Epirubicincytostatic drug20AdCC2 (10)3.715.6
II [52]R/M
First-line
Gemcitabinecytostatic drug21AdCC0 (0)
Retro [50]R/M
Any line
Methotrexatecytostatic drug7AdCC0 (0)ITT: 21.0
II [54]R/M
First-line
Mitoxantronecytostatic drug32AdCC4 (13)18.0
II [47]R/M
Any line
Mitoxantronecytostatic drug18AdCC1 (5)19.0
II [40]R/M
First-line
Paclitaxelcytostatic drug45AdCC: 14
Non-AdCC: 31
(14 MEC, 17 adeno)
ITT: 8 (18)
AdCC: 0 (0)
Non-AdCC: 8 (26)
(3 MEC, 5 adeno)
4.0ITT: 12.5
II [34]R/M
First- or second line
Vinorelbinecytostatic drug20AdCC: 13
Non-AdCC: 7
(5 adeno, 1 mixed, 1 undifferentiated carcinoma)
ITT: 4 (20)
AdCC: 2 (15)
Non-AdCC: 2 (29)
(2 adeno)
5.08.5
B. Combination therapy
II [32]R/M
Any line
Carboplatin + paclitaxelcytostatic drugs14AdCC: 10
Non-AdCC: 9
(1 MEC, 1 adeno, 2 undifferentiated)
ITT: 2 (14)
AdCC: 2 (20)
Non-AdCC: 0 (0)
6.0–13.512.5
Retro [48]R/M
Any line
Carboplatin + paclitaxelcytostatic drugs38AdCC: 9
Non-AdCC: 29
(1 MEC, 18 SDC, 4 adeno, 1 myoepithelial, 1 epithelial-myoepithelial, 4 ex pleomorphic adenoma)
ITT: 15 (39)
AdCC: 1 (9)
Non-AdCC: 14 (48)
ITT: 6.5
AdCC: 9.7
Non-AdCC: 6.2–6.5
ITT: 26.5
AdCC: 21.9
Non-AdCC: 34.7–44.0
II [41]R/M
Any line
Cisplatin + 5-fluorouracilcytostatic drugs11AdCC0 (0)9.012.0
II [37]R/M
First- or second line
Cisplatin + doxorubicin + bleomycincytostatic drugs9AdCC3 (33)10.012.0
Retro [39]R/M
Any line
Cisplatin + doxorubicin + cyclophosphamidecytostatic drugs13AdCC: 9
Non-AdCC: 4
(4 adeno)
ITT: 6 (46)
AdCC: 3 (33)
Non-AdCC: 3 (75)
(3 adeno)
Retro [35]R/M
Any line
Cisplatin + doxorubicin + cyclophosphamidecytostatic drugs8AdCC: 4
Non-AdCC: 4
(3 MEC, 1 adeno)
ITT: 5 (63)
AdCC: 1 (25)
Non-AdCC: 4 (100)
(3 MEC, 1 adeno)
5.011.0
II [45]R/M
Any line
Cisplatin + doxorubicin + cyclophosphamidecytostatic drugs22AdCC: 12
Non-AdCC: 10
(1 MEC, 2 SDC, 2 adeno, 1 NET, 3 myoepithelioma, 1 undifferentiated)
ITT: 6 (27)
AdCC: 3 (25)
Non-AdCC: 3 (30)
(1 MEC, 1 SDC, 1 NET)
21.0
II [38]R/M
First-line
Cisplatin + doxorubicin + cyclophosphamide + 5-fluorouracilcytostatic drugs16AdCC: 7
Non-AdCC: 9
(1 MEC, 8 adeno)
ITT: 8 (50)
AdCC: 3 (42)
Non-AdCC: 5 (56)
(1 MEC, 4 adeno)
16.8
II [34]R/M
First- or second line
Cisplatin + vinorelbinecytostatic drugs16AdCC: 9
Non-AdCC: 7
(1 MEC, 4 adeno, 2 undifferentiated)
ITT: 7 (44)
AdCC: 4 (44)
Non-AdCC: 3 (43)
(2 adeno, 1 undifferentiated)
7.011.0
II [33]R/M
First-line
Cisplatin + vinorelvinecytostatic drugs42AdCC: 24
Non-AdCC: 18
(2 MEC, 11 adeno,
2 mixed, 3 undifferentiated)
ITT: 13 (31)
AdCC: 7 (17)
Non-AdCC: 7 (39)
(7 adeno)
6.010.0
II [33]R/M
Second-line
Cisplatin + vinorelvinecytostatic drugs18AdCC: 10
Non-AdCC: 8
(9 MEC, 4 adeno, 2 undifferentiated)
ITT: 1 (6)
AdCC: 0 (0)
Non-AdCC: 1 (13)
(1 adeno)
3.54.0
II [42]R/M
Any line
Cisplatin + vinorelvinecytostatic drugs40AdCC: 19
Non-AdCC: 21
(6 MEC, 10 adeno, 2 AcCC, 1 myoepithelial, 1 ex pleomorphic adenoma, 1 undifferentiated)
ITT: 14 (35)
AdCC: 6 (32)
Non-AdCC: 8 (38)
6.316.9
II [51]R/M
First-line
Cyclophosphamide + vincristine + 5-fluorouracilcytostatic drugs8AdCC2 (25)28.062.0
II [43]R/M
First-line
Platin + docetaxelcytostatic drugs41AdCC: 26
Non-AdCC: 15
(1 MEC, 10 SDC, 3 adeno, 1 SCC)
ITT: 19 (46)
AdCC: 6 (23)
Non-AdCC: 13 (87)
(1 MEC, 9 SDC, 2 adeno, 1 SCC
ITT: 9.4
AdCC: 8.9
Non-AdCC: 10.5
ITT: 28.2
AdCC: 27.6
Non-AdCC: 29.3
II [44]R/M
First- or second line
Platin + gemcitabinecytostatic drugs33AdCC: 10
Non-AdCC: 23
(4 MEC, 1 SDC, 5 AcCC, 9 adeno, 4 undifferentiated)
ITT: 8 (27)
AdCC: 2 (20)
Non-AdCC: 6 (26)
(1 MEC, 1 SDC, 1 AcCC, 3 adeno)
13.8
* If specified in the original study, the subtyping of the non-AdCC tumors is given. AcCC, acinic cell carcinoma; AdCC, adenoid cystic carcinoma; ITT, intention-to-treat; MEC, mucoepidermoid carcinoma; n, number; ORR, objective response rate; OS, overall survival; Pts, patients; PFS, progression-free survival; R/M, recurrent or metastatic; retro, retrospective; SCC, squamous cell carcinoma; SDC, salivary duct carcinoma; SGMs, salivary gland malignancies.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cleymaet, R.; Vermassen, T.; Coopman, R.; Vermeersch, H.; De Keukeleire, S.; Rottey, S. The Therapeutic Landscape of Salivary Gland Malignancies—Where Are We Now? Int. J. Mol. Sci. 2022, 23, 14891. https://doi.org/10.3390/ijms232314891

AMA Style

Cleymaet R, Vermassen T, Coopman R, Vermeersch H, De Keukeleire S, Rottey S. The Therapeutic Landscape of Salivary Gland Malignancies—Where Are We Now? International Journal of Molecular Sciences. 2022; 23(23):14891. https://doi.org/10.3390/ijms232314891

Chicago/Turabian Style

Cleymaet, Robbert, Tijl Vermassen, Renaat Coopman, Hubert Vermeersch, Stijn De Keukeleire, and Sylvie Rottey. 2022. "The Therapeutic Landscape of Salivary Gland Malignancies—Where Are We Now?" International Journal of Molecular Sciences 23, no. 23: 14891. https://doi.org/10.3390/ijms232314891

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