Next Article in Journal
Ozonized Hydrogels vs. 1% Chlorhexidine Gel for the Clinical and Domiciliary Management of Peri-Implant Mucositis: A Randomized Clinical Trial
Next Article in Special Issue
Patient-Reported Outcome Measures of Psychosocial Quality of Life in Oropharyngeal Cancer Patients: A Scoping Review
Previous Article in Journal
First Nation-Wide Study of the Incidence and Characteristics of Retinal Detachment in Poland during 2013–2019
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances, Systemic Therapy, and Molecular Targets in Adenoid Cystic Carcinoma of the Head and Neck

Department of Oral and Maxillofacial Surgery and Facial Plastic Surgery, Ludwig Maximilians University, D-80337 Munich, Germany
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(4), 1463; https://doi.org/10.3390/jcm12041463
Submission received: 8 January 2023 / Revised: 3 February 2023 / Accepted: 8 February 2023 / Published: 12 February 2023
(This article belongs to the Special Issue Head and Neck Cancer: Recent Advances and Future Directions)

Abstract

:
With an incidence of 3–4.5 cases per million, adenoid cystic carcinoma (ACC) of the head and neck is one of the most common tumors of the parotid and sublingual salivary glands. In the clinical course, ACC is shown to have an aggressive long-term behavior, which leads to the fact that radical surgical resection of the tumor with tumor-free margins remains the “gold standard” in treating ACC. Particle radiation therapy and systemic molecular biological approaches offer new treatment options. However, risk factors for the formation and prognosis of ACC have not yet been clearly identified. The aim of the present review was to investigate long-term experience of diagnosis and treatment as well as risk and prognostic factors for occurrence and outcome of ACC.

1. Introduction

Adenoid cystic carcinoma (ACC) of the head and neck is a rare neoplasm frequently arising in salivary glands and related tissue [1], accounting for 1% of all head and neck malignancies [2]. An incidence of 3–4.5 cases per million makes ACC one of the most common malignant tumors of the parotid and sublingual salivary glands [3] as well as the most commonly reported tumor of the minor salivary glands [4]. ACC has also been observed in other regions of the head and neck including the trachea, larynx [5], nasal and paranasal sinus, and lacrimal glands [5,6,7]. Slow local growth and perineural invasion, sporadic regional metastasis, and progressive distant metastasis are classical characteristics of ACC [8]. Correlations between clinical outcome and histological grade have been described [9], but risk factors for the formation and prognosis of ACC have still not yet been clearly identified. As ACC is frequently described as “wolf in sheep’s clothing” due to its slow but uncompromisingly growth and dissemination [10], the identification of molecular biological and clinical-pathological predictors of recurrence and survival are needed for a maximization of treatment outcome.

2. Epidemiology

The age at which ACC of the head and neck is diagnosed ranges from 11 to 99 years, with the mean age at diagnosis between 50 and 60 years [11,12,13]. A marginally higher prevalence among women has been observed [14], with a male/female ratio of 0.85:1 [15]. In large population-based studies, ACC accounts for 28% of the total incidence of malignant salivary gland tumors [4], with ACC found in one out of every eight malignant parotid gland tumors [16] and accounting for 40% of submandibular gland carcinomas [17].

3. Clinical Appearance

ACC of the head and neck exhibits an aggressive long-term behavior. In early stages, ACC is associated with slow growth and an indolent course. Pain occurs in later onsets due to perineural invasion [18]. Depending on the localization of the tumor, symptoms may vary from facial nerve paralysis (parotid gland) to tumor masses showing ulceration or oroantral fistulas (palate) and nasal obstruction (nasal and paranasal sinus) [19,20]. Staging at the time of diagnosis varies. Around one-third of patients are diagnosed with T4 tumor size initially. In addition, 15 percent of patients show lymph nodal invasion or distant metastases at the time of diagnosis. This results in more than 40 percent of patients with a TNM stage 4 tumor at the time of primary diagnosis. Locoregional and distant recurrence is reported in 30 percent of patients. The latest clinical studies reported a mean time period of 3.4 years between the initial diagnosis of ACC and recurrence. In cases of death due to ACC, the mean time between diagnosis and the registration of death was 5.6 years with a range of 0.4–14.1 years [15]. In summary, ACC is clinically characterized by aggressive local growth, high recurrence rates, and decreased overall survival. Age, BMI, and N stage are described as the three main clinical prognostic factors identified determining event free survival in a prospective multicenter study of 470 patients with ACC [14].

4. Diagnostic Imaging

Standard preoperative diagnostic imaging includes magnetic resonance imaging (MRI) or computed tomography (CT). Bone invasion imaging is regularly done by CT diagnostics, whereas soft tissue extension and perineural invasion are assessed in MRI [21,22]. The latest studies have shown that conventional MRI of the head and neck should be complemented by whole-body PET/CT to detect local tumor recurrence, lymph node metastases, or distant metastases [23]. Even more PET/CT imaging using 68Ga-labelled FAP inhibitors (68Ga-FAPI-PET/CT) detected additional metastases, thereby increasing the accuracy of staging and radiotherapy planning volumes relative to conventional CT/MRI [24]. In addition, 99mTc-MIBI SPECT/CT and FDG PET/CT showed promising results in the detection of distant metastases in ACC of the head and neck [25].

5. Histopathology

Three histological patterns (tubular, cibriform, solid) have been described in ACC: The tubular pattern is characterized by simple tubules composed of inner ductal and outer myoepithelial cells. The cibriform pattern predominantly consists of myoepithelial cells with myxoid or hyalinized globules along with isolated solitary ductal elements. Solid nests composed of sheets of basaloid cells are typical for the solid pattern [26]. A combination of the cribriform and tubular pattern is often observed in classical ACC. Solid patterns have been associated with a more advanced tumor stage and worse prognosis [27]. Two histological grading systems have been proposed, reflecting the clinical outcome due to the percentage of the solid component in ACC. Perzin [27] and Szanto [28] defined a staging system including three different stages: low grade (stage 1) without a solid component, intermediate grade (stage 2) with a 1–29% solid component, and high grade (stage 3) with a more than 30% solid component. A more recent staging system proposed by Van Weert [29] and Zhang [30] differentiated only two stages: grade one with no solid component and grade two with a solid component. Cervical nodal metastases as well as distant metastases in ACC patients can be histopathologically detected [31].
Another histological property associated with patient outcomes in ACC is perineural invasion (PNI). PNI is significantly correlated with both distant metastasis and unfavorable disease outcomes [32]. However, due to its microscopic appearance, 3D imaging often fails to detect PNI in patients with ACC [33].

6. Therapy and Prognosis

Radical surgical resection of the tumor with tumor-free margins remains the “gold standard” in treating ACC of the head and neck. The possibility of achieving tumor-free margins is limited by factors such as tumor localization (proximity to skull base), histopathologic pattern, and previous treatment. The role of surgical margins was examined by Amit et al. who analyzed 507 cases of head and neck ACC in an international multi-center study. Positive margins were associated with the worst outcome, whereas negative and close margins were associated with improved outcome, regardless of the distance from the tumor. Except the oral cavity, the presence of close margin status resulted in similar outcomes as negative margins for ACC of all sites examined. Therefore, negative margins should be achieved whenever possible [34]. Due to the abovementioned limitations, a combination of surgery with radiation therapy has led to superior results in many studies [35], with a disease-specific survival of 92 percent after five years, and 88 percent after ten years. Disease-free survival (DFS) amounted to 67 percent after 5 years and 61 percent after 10 years. Patients with unresectable ACC or positive surgical margins showed a worse disease-specific survival of 78 percent after five years and 32 percent after ten years, as well as worse disease-free survival (35 percent after 5 years, 8 percent after 10 years) [15]. Chemotherapy as a treatment procedure in ACC has also been reported. Common monotherapy strategies have been implemented with 5-FU [36], cisplatin [37], gemcitabine [38], mitoxantrone [39], epirubicin [40], vinorelbine [41], and paclitaxel [42]. In these studies, objective response ranged from 0 to 70% with a median of 16% [43]. Combination chemotherapy such as the combination of cisplatin, doxorubicin, and cyclophosphamide (CAP) has also been examined in several studies [44,45]. The combination of CAP with 5-fluoruracil showed the longest duration of response in patients [46], whereas the combination of cyclophosphamide, vincristine, and 5-FU [47] showed a rather long duration of response. Still, combined chemotherapy led to a relatively poor outcome compared to the treatment side effects (nausea, vomiting, myelosuppression, and neuropathy). An optimum treatment for ACC has not yet been established, probably because of the neurotropic and infiltrative characteristics of the tumor. Disease treatment should be adapted to the initial diagnosis, taking tumor localization, pathological staging, and grading into account [48].

7. Particle Radiation Therapy

Heavy-particle radiotherapy using protons, neutrons, or carbon ions has recently emerged as a promising treatment option. Proton treatment has been performed in several studies, using 60 to 76.4 Gray equivalent (GyE) [49,50], giving even patients with unresectable ACC of the head and neck a promising treatment perspective [51]. Case studies observing survival and outcome of inoperable ACC of the head and neck using proton radiation therapy report overall survival (OS) rates of 80% and a three-year progression-free survival of 63% [52]. Furthermore, patients receiving postoperative intensity-modulated proton therapy to a median of 60 GyE in combination with platinum-based chemotherapy showed no evidence of disease at a median of 24.9 months [50]. Recent studies concluded that there is no significant survival advantage in operated patients relative to patients with unresectable ACC when treated with pencil beam scanning proton therapy (PBS PT) [53].
Effects of carbon ion radiotherapy for head-and-neck cancer, especially ACC, have been evaluated in phase II clinical trials, proving the therapeutic effectiveness without severe morbidity of the normal tissues [54,55,56]. More phase II clinical trials are in progress [57]. Results of a combination treatment with intensity-modulated radiotherapy and active raster-scanning carbon ion boost for adenoid cystic carcinoma of the minor salivary glands of the nasopharynx resulted in adequate local control and OS rates with moderate toxicity [58]. Carbon ion radiotherapy with simultaneous integrated boost for head and neck ACC improved the tumor dose conformation while reducing the unintended dose to the low-risk volume and related probability of complications [59]. Even though fast neutron therapy (FNT) seemed to be a promising treatment procedure, it received less consideration in clinical trials due to limited availability, high costs, and challenges with healthy tissue toxicity in treatment cohorts. Side effects such as posttreatment trismus, acute mucositis, and acute xerostomia have been reported [60,61]. Still, the latest case studies including neutron radiation treatment in ACC report a 10-year OS of 62% and osteoradionecrosis rates comparable to that of photon radiation treatment (2–7%) [62]. Recently, early outcomes for a hybrid neutron–proton approach were found to be promising, describing an OS of 93.1% and a PFS of 79.3%. Still, further clinical trials with longer follow-ups and larger patient cohorts are needed for validation [63]. Table 1 shows clinical trials using particle radiation therapy.

8. Molecular Biomarkers as a Treatment Target

Molecular markers as a prognostic and therapeutical approach have been of great interest in the last years. Prognostic biomolecular factors can help to identify patients that would benefit from more intense treatment and point out new therapeutic targets. In the following, different gene expression patterns are discussed.

8.1. Oncogenes

SOX4, C-KIT, VEGF, BDNF, MYB, Wnt/b-Catenin

Cell cycle and apoptosis regulators together with other oncogenes are discussed as having an impact in the pathogenesis of ACC. It was observed that the Sry-related high mobility group (HMG) box 4 (SOX4) is one of the most highly overexpressed genes in ACC [67]. SOX4 is an essential developmental transcription factor that regulates stemness, differentiation, progenitor development, and multiple developmental pathways [68,69]. Another protein involved in malignant transformation in ACC is the transmembrane tyrosine kinase receptor c-kit. The activation of c-kit promotes cell growth and differentiation [70], and its expression has been shown to correlate with ACC tumor grade [71]. However, clinical trials investigating the effects of c-kit inhibitors in patients have not yet been promising [72].
Furthermore the association of VEGF expression with advanced stage and worse disease-specific survival has been reported [73], along with enhanced microvessel density in ACC, which makes VEGF an important prognosticator of survival and outcome [74,75]. Younes et al. have shown that concomitant inhibition of epidermal growth factor and vascular endothelial growth factor receptor tyrosine kinases reduces the growth and metastasis of human salivary ACC in a mouse model [76], highlighting VEGF and EGF as important therapeutic targets.
As perineural invasion is a frequent complication in ACC, brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) have been suspected to facilitate perineural invasion. BDNF has been reported to be uniformly expressed by ACC [77]. Elevated expression of BDNF was found in salivary ACC specimens, which was significantly correlated with the invasion and metastasis [78]. Overexpression of NGF and TrkA in human salivary ACC tissues has also been reported and may constitute a reason for perineural invasion [79]. Furthermore, overexpression of NGF in combination with solid subtype, advanced stage, perineural invasion, recurrence, and extended resection alone have been reported to have worse survival rates showing the relevance of NGF as a prognostic marker [80].
The proto-oncogene MYB is associated with apoptosis, cell cycle control, cell adhesion, growth, and differentiation [81]. MYB gene fusion or MYB protein overexpression occurs in the majority of ACCs, suggesting that MYB might be a potential therapeutic target, but early results have not been promising [82]. Moreover, no significant prognostic differences have been observed between MYB-positive and MYB-negative ACCs, which indicates that MYB is not a good prognostic marker for ACC [83].
Signaling pathways are another target for therapeutic approaches in ACC. Mutations in components of the canonical Wnt/β-catenin signaling pathway have been found in various neoplasms [84,85]. Mutations in components of the Wnt signaling pathway in ACC of the head and neck have already been described [86,87]. In addition reduced membranous expression of β-catenin can be associated with ACC metastasis [88]. The aberrant Wnt/β-catenin signaling pathway enables cancer stem cell renewal, cell proliferation, and differentiation, thus enforcing tumorigenesis and therefore officiating as a therapeutical target in ACC and other head and neck tumors.

8.2. Tumorsuppressorgenes

p53, p16

Cell cycle regulatory proteins as p53 and p16 and their genes are among the most commonly mutated genes identified in human neoplasms [89]. The p53 protein plays a central role in the expression of genes involved in the regulation of apoptosis and DNA repair. In case damaged DNA is detected in the cell, p53 is phosphorylated, which prevents it from being broken down. The accumulation of p53 induces cell cycle interruption and, if damaged DNA is not repaired properly, apoptosis [90]. Hence, mutations in p53 that limit the protein function ultimately cause replication of damaged DNA, which may result in neoplasms. Studies have shown that p53 mutations in ACC correlate with metastasis and recurrence [91].
The protein p16CDNK2a (p16) is encoded by the gene CDKN2A [92,93] and is produced in response to cellular stress signals preventing entry into the S-phase by inhibiting cyclin-dependent kinases (Cdks), which are necessary for the phosphorylation of the retinoblastoma protein (Rb) [94]. Immunohistochemical studies have shown that p16 expression was reduced in ACC cases of higher histological grade of malignancy [95] due to hypermethylation and homozygous deletion as the main inactivation mechanisms of p16 gene [96]. Therefore, p16 could be used as a prognostic marker for worse survival in ACC of the head and neck and should thus be considered in individual therapy strategies.

8.3. Mitochondrial Alterations

The occurrence of mitochondrial alterations and oxidative stress is an important hallmark of tumorigenesis and the development of cancers, including head and neck carcinoma. Mithani et al. showed that 17 out of 22 ACCs carried mitochondrial mutations, most of them occurring in the NADH complex [97]. Along with this finding, it was observed that superoxide dismutase 2 (SOD2) was deregulated in patients with salivary ACC. Up-regulation of SOD2 was associated with distant metastasis and reduced OS and disease-free survival [98]. Reactive oxygen species in high concentrations can be responsible for cell damage, mutations, and tumors [99,100]. For example, hydroxyl anions damage sections of DNA by reacting with purine and pyrimidine bases and with the deoxyribose framework [101], or they are initiators of lipid peroxidation, as a result of which membrane fluidity is reduced, membrane permeability is increased, membrane proteins are damaged, and receptors or ion channels are inactivated or dysregulated [102]. Even more ROS have a significant impact on physiological cell processes such as cellular signal transduction, transcription, and induction of apoptosis [103] and thus affect tumor genesis. Still, further research is needed to determine the effects of the altered antioxidant defense system (as SOD) in ACC.

8.4. Immune Microenvironment and Evasion Mechanisms

Expression analysis of immune checkpoints (PD-L1, PD-L2, PD-1, and CTLA-4), immune inhibitory molecule HLA-G, and markers of tumor-infiltrating lymphocytes (TIL) and dendritic cells (DC) in ACC showed low CD8+, GrB+ TIL, CD1a, and CD83 populations, as well as scarce positivity for CTLA-4 and PD-1. In contrast, PD-L2 and HLA-G expression was increased, which suggests that the ACC microenvironment exhibits low immunogenicity, represented by low TIL and DC density. Moreover, an activation of the immune inhibitory proteins/PD-L2 and HLA-G may favor tumor escape from the immune system and partially explain the poor prognosis of ACC [10].
Tumor markers and prognostic markers are expressed differently in each individual, showing that each patient should be considered and treated individually according to personalized medicine.

9. Gene Panel Examination and Further Potential Therapeutic Targets

Potential targets identified in adenoid cystic carcinoma point out new directions for further research: the Examinations of the Gene Expression Omnibus (GEO) database was used to explore abnormal coexpression of genes in ACC compared with their expression in normal tissue. The analysis showed that ITGA9 (integrin alpha9) and LAMB1 (laminin subunit beta 1) are important factors regulating the PI3K-Akt pathway. In addition, BAMBI (BMP and Activin membrane-bound inhibitor), a suppressor of TGFβ, serves as an important factor that is involved in the TGF-β signaling pathway and thus acts as a potential target for molecular therapeutic approaches. Further targets described for future studies are SLC22A3 (solute carrier family 22 member 3), FOXP2 (Forkhead box P2), Cdc42EP3 (CDC42 effector protein 3), COL27A1 (collagen type XXVII alpha 1 chain), DUSP1 (dual specificity phosphatase 1), HSPB8 (heat shock protein family B (Small) member 8), ST3Gal4 (ST3 beta-galactoside alpha-2,3-sialyltransferase 4), SPARC (secreted protein acidic and cysteine rich), COL4A2 (collagen type IV alpha 2 chain), PRELP (proline- and arginine-rich end leucine-rich repeat protein), hsa-miR-29-3p, hsa-miR-132-3p, and hsa-miR-708-5p due to regulation of tumorigenesis in ACC [104].
Even more DNA-based next-generation sequencing identified Notch-activating mutations as a promising target in ACC [105]. Notch is involved in both pro- and anti-tumoral effects in the different populations composing the tumor and takes an important role in regulating the crosstalk between the different compartments of the tumor microenvironment [106]. A precise understanding of the contribution of Notch signaling in the different compartments of the TME in ACC is needed in order to design future therapeutic approaches targeting Notch signaling.

10. Systemic Molecular Biological Approaches

Within the last 10 years, outcomes of systemic molecular therapy in advanced ACC have been reported in less than 300 patients. Due to the relatively low prevalence of ACC in the head and neck region, most of the clinical trials are small and are only conducted by a single institution. Initial studies using multi-kinase inhibitors such as sunitinib showed no significant observed responses, but the majority of patients showed stable disease with moderate toxic effects [107]. Targeting mTOR resulted in no complete or partial response with a median progression-free survival (PFS) of 11.2 months, with observed tumor shrinkage described in 44% of treated patients [108]. The use of sorafenib (400 mg) showed modest activity in ACC with a 12-month PFS of 46.2%. Sorafenib was associated with significant toxicity, and considering the limited effectiveness, could not be recommended for further evaluation [109]. Therapy using the Akt signaling inhibitor nelfinavir as monotherapy also did not result in a meaningful improvement in clinical outcome [110]. Treating recurrent/metastatic ACC with regorafenib, a tyrosine kinase inhibitor, resulted in stable disease of 6 months in 17 out of 38 patients [111]. In contrast, therapy with dovitinib (another tyrosine kinase inhibitor) showed promising results: tumor shrinkage was observed in 22 out of 32 metastatic or unresectable ACC patients, with one patient with confirmed partial response. Moreover, metabolic activity of the tumor was reduced in 13 patients after dovitinib treatment [112]. In addition, treatment with the multi-kinase inhibitor lenvatinib resulted in partial response in 5 out of 33 enrolled patients and 24 patients (75%) with stable disease, showing promising results while using molecular target therapy [113]. The results are shown in Table 2.
Especially in the latest trials, stable disease was a common result. Still, not all observed patients showed progressive disease initially when included in the study, leading to difficulties in assessment if periods of stable disease are attributable to drug activity or to a non-active period of the disease itself. In all listed studies, median survival was low, which is related to the initial staging (unresectable, distant metastases). Moreover, all listed trials were addressed to advanced and metastatic ACC (none of the included studies addressed to resectable ACC). Therefore, it is not possible to evaluate whether therapeutic response is detectable in earlier stages of ACC. The results are consistent with earlier analyses, which up until now it remains an open question if ACC benefits from treatment with systemic therapy as the clinical outcome by best supportive care (radical surgery with postoperative radiotherapy) may be the same as that by any intervention targeting molecular mechanisms [115]. Still, in patients with unresectable ACC or distant metastases, a participation in clinical trials might be a suitable option that needs to be taken into account individually. As gene panel examination and DNA-based next-generation sequencing offer opportunities to identify future targets, further prospective clinical trials with larger patient cohorts are needed to evaluate more therapeutic potentials.

11. Conclusions

There are recommended therapy regimens for ACC of the head and neck, but as shown in the above data, those recommendations show limited efficacy. The activation of immune inhibitory proteins such as PD-L2 and HLA-G may favor tumor escape from the immune system and partially explain the poor prognosis of ACC. Various markers such as SOX4, VEGF, BDNF, and targets of the Wnt/β-catenin signaling pathway and the Notch pathway are expressed in ACC, but not equally in all tumor patients. For this reason, expression profiles of patients should be examined individually, and target therapies should be considered according to a personalized medicine approach. The preferred treatment strategy for the majority of patients with ACC of the head and neck remains as radical surgery with postoperative radiotherapy. Particle radiation therapy, especially proton and carbon ion radiation, were found to improve OS and PFS significantly, thus being a promising future treatment procedure. Systemic molecular biological therapeutic approaches as well as the identification of important prognosis factors offer new treatment options, still requiring further clinical observations.

Author Contributions

Conceptualization, I.D. and K.T.O.; methodology, S.O. and I.D., formal analysis, K.T.O. and T.K.K.; investigation, I.D. and K.T.O.; writing—original draft preparation, I.D.; writing—review and editing, K.T.O., W.S., S.O.; supervision, K.T.O. and S.O.; project administration, K.T.O., W.S. and I.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Moskaluk, C.A. Adenoid cystic carcinoma: Clinical and molecular features. Head Neck Pathol. 2013, 7, 17–22. [Google Scholar] [CrossRef]
  2. Dodd, R.; Slevin, N. Salivary gland adenoid cystic carcinoma: A review of chemotherapy and molecular therapies. Oral Oncol. 2006, 42, 759–769. [Google Scholar] [CrossRef] [PubMed]
  3. Spiro, R.H. Salivary neoplasms: Overview of a 35-year experience with 2807 patients. Head Neck Surg. 1986, 8, 177–184. [Google Scholar] [CrossRef]
  4. Bjorndal, K.; Krogdahl, A.; Therkildsen, M.H.; Overgaard, J.; Johansen, J.; Kristensen, C.A.; Homoe, P.; Sorensen, C.H.; Andersen, E.; Bundgaard, T.; et al. Salivary gland carcinoma in Denmark 1990–2005: A national study of incidence, site and histology. Results of the Danish Head and Neck Cancer Group (DAHANCA). Oral Oncol. 2011, 47, 677–682. [Google Scholar] [CrossRef]
  5. Eschwege, F.; Cachin, Y.; Micheau, C. Treatment of adenocarcinomas of the larynx. Can. J. Otolaryngol. 1975, 4, 290–292. [Google Scholar]
  6. Azar, T.; Abdul-Karim, F.W.; Tucker, H.M. Adenoid cystic carcinoma of the trachea. Laryngoscope 1998, 108, 1297–1300. [Google Scholar] [CrossRef] [PubMed]
  7. Mine, T.; Yamada, R. A case of adenoid cystic carcinoma of the lacrimal gland. Nihon Ganka Kiyo 1967, 18, 499–503. [Google Scholar] [PubMed]
  8. Hamper, K.; Lazar, F.; Dietel, M.; Caselitz, J.; Berger, J.; Arps, H.; Falkmer, U.; Auer, G.; Seifert, G. Prognostic factors for adenoid cystic carcinoma of the head and neck: A retrospective evaluation of 96 cases. J. Oral Pathol. Med. 1990, 19, 101–107. [Google Scholar] [CrossRef]
  9. Nochomovitz, L.E.; Kahn, L.B. Adenoid cystic carcinoma of the salivary gland and its histologic variants: A clinicopathologic study of thirty cases. Oral Surg. Oral Med. Oral Pathol. 1977, 44, 394–404. [Google Scholar] [CrossRef] [PubMed]
  10. Mosconi, C.; de Arruda, J.A.A.; de Farias, A.C.R.; Oliveira, G.A.Q.; de Paula, H.M.; Fonseca, F.P.; Mesquita, R.A.; Silva, T.A.; Mendonca, E.F.; Batista, A.C. Immune microenvironment and evasion mechanisms in adenoid cystic carcinomas of salivary glands. Oral Oncol. 2018, 88, 95–101. [Google Scholar] [CrossRef]
  11. Ellington, C.L.; Goodman, M.; Kono, S.A.; Grist, W.; Wadsworth, T.; Chen, A.Y.; Owonikoko, T.; Ramalingam, S.; Shin, D.M.; Khuri, F.R.; et al. Adenoid cystic carcinoma of the head and neck: Incidence and survival trends based on 1973–2007 Surveillance, Epidemiology, and End Results data. Cancer 2012, 118, 4444–4451. [Google Scholar] [CrossRef]
  12. Takebayashi, S.; Shinohara, S.; Tamaki, H.; Tateya, I.; Kitamura, M.; Mizuta, M.; Tanaka, S.; Kojima, T.; Asato, R.; Maetani, T.; et al. Adenoid cystic carcinoma of the head and neck: A retrospective multicenter study. Acta Otolaryngol. 2018, 138, 73–79. [Google Scholar] [CrossRef]
  13. Gamboa-Hoil, S.I.; Silva-Godinez, J.C.; Abrego-Vasquez, A. Adenoid cystic carcinoma of head and neck. A 5-year retrospective study: Experience in a single third-level reference center. Cir. Cir. 2020, 88, 34–40. [Google Scholar] [CrossRef] [PubMed]
  14. Atallah, S.; Casiraghi, O.; Fakhry, N.; Wassef, M.; Uro-Coste, E.; Espitalier, F.; Sudaka, A.; Kaminsky, M.C.; Dakpe, S.; Digue, L.; et al. A prospective multicentre REFCOR study of 470 cases of head and neck Adenoid cystic carcinoma: Epidemiology and prognostic factors. Eur. J. Cancer 2020, 130, 241–249. [Google Scholar] [CrossRef] [PubMed]
  15. 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]
  16. Poorten, V.V.; Hart, A.; Vauterin, T.; Jeunen, G.; Schoenaers, J.; Hamoir, M.; Balm, A.; Stennert, E.; Guntinas-Lichius, O.; Delaere, P. Prognostic index for patients with parotid carcinoma: International external validation in a Belgian-German database. Cancer 2009, 115, 540–550. [Google Scholar] [CrossRef]
  17. Batsakis, J.G. Carcinomas of the submandibular and sublingual glands. Ann. Otol. Rhinol. Laryngol. 1986, 95 Pt 1, 211–212. [Google Scholar] [CrossRef] [PubMed]
  18. Vander Poorten, V.L.; Balm, A.J.; Hilgers, F.J.; Tan, I.B.; Loftus-Coll, B.M.; Keus, R.B.; van Leeuwen, F.E.; Hart, A.A. The development of a prognostic score for patients with parotid carcinoma. Cancer 1999, 85, 2057–2067. [Google Scholar] [CrossRef]
  19. Biswas, K.D.; Saha, J.; Sen, I.; Biswas, G.; Sinha, R.; Saha, D.; Dutta, S. Unusual presentations of adenoid cystic carcinoma in extra-salivary gland subsites in head and neck region: A case series. Indian J. Otolaryngol. Head Neck Surg. 2014, 66 (Suppl. 1), 286–290. [Google Scholar] [CrossRef] [PubMed]
  20. Husain, Q.; Kanumuri, V.V.; Svider, P.F.; Radvansky, B.M.; Boghani, Z.; Liu, J.K.; Eloy, J.A. Sinonasal adenoid cystic carcinoma: Systematic review of survival and treatment strategies. Otolaryngol. Head Neck Surg. 2013, 148, 29–39. [Google Scholar] [CrossRef] [PubMed]
  21. Matsuzaki, H.; Yanagi, Y.; Hara, M.; Katase, N.; Asaumi, J.; Hisatomi, M.; Unetsubo, T.; Konouchi, H.; Takenobu, T.; Nagatsuka, H. Minor salivary gland tumors in the oral cavity: Diagnostic value of dynamic contrast-enhanced MRI. Eur. J. Radiol. 2012, 81, 2684–2691. [Google Scholar] [CrossRef] [Green Version]
  22. Hanna, E.; Vural, E.; Prokopakis, E.; Carrau, R.; Snyderman, C.; Weissman, J. The sensitivity and specificity of high-resolution imaging in evaluating perineural spread of adenoid cystic carcinoma to the skull base. Arch. Otolaryngol. Head Neck Surg. 2007, 133, 541–545. [Google Scholar] [CrossRef] [PubMed]
  23. Ruhlmann, V.; Poeppel, T.D.; Veit, J.; Nagarajah, J.; Umutlu, L.; Hoffmann, T.K.; Bockisch, A.; Herrmann, K.; Sauerwein, W. Diagnostic accuracy of (18)F-FDG PET/CT and MR imaging in patients with adenoid cystic carcinoma. BMC Cancer 2017, 17, 887. [Google Scholar] [CrossRef]
  24. Rohrich, M.; Syed, M.; Liew, D.P.; Giesel, F.L.; Liermann, J.; Choyke, P.L.; Wefers, A.K.; Ritz, T.; Szymbara, M.; Schillings, L.; et al. (68)Ga-FAPI-PET/CT improves diagnostic staging and radiotherapy planning of adenoid cystic carcinomas—Imaging analysis and histological validation. Radiother Oncol. 2021, 160, 192–201. [Google Scholar] [CrossRef] [PubMed]
  25. Zhao, Q.; Dong, A.; Ye, H.; Zuo, C. 99mTc-MIBI SPECT/CT and FDG PET/CT in Isolated Bilateral Renal Metastases From Adenoid Cystic Carcinoma of the Maxilla. Clin. Nucl. Med. 2022, 47, e205–e207. [Google Scholar] [CrossRef] [PubMed]
  26. Matsuba, H.M.; Simpson, J.R.; Mauney, M.; Thawley, S.E. Adenoid cystic salivary gland carcinoma: A clinicopathologic correlation. Head Neck Surg. 1986, 8, 200–204. [Google Scholar] [CrossRef] [PubMed]
  27. Perzin, K.H.; Gullane, P.; Clairmont, A.C. Adenoid cystic carcinomas arising in salivary glands: A correlation of histologic features and clinical course. Cancer 1978, 42, 265–282. [Google Scholar] [CrossRef]
  28. Szanto, P.A.; Luna, M.A.; Tortoledo, M.E.; White, R.A. Histologic grading of adenoid cystic carcinoma of the salivary glands. Cancer 1984, 54, 1062–1069. [Google Scholar] [CrossRef] [PubMed]
  29. Van Weert, S.; Reinhard, R.; Bloemena, E.; Buter, J.; Witte, B.I.; Vergeer, M.R.; Leemans, C.R. Differences in patterns of survival in metastatic adenoid cystic carcinoma of the head and neck. Head Neck 2017, 39, 456–463. [Google Scholar] [CrossRef] [PubMed]
  30. Zhang, C.Y.; Xia, R.H.; Han, J.; Wang, B.S.; Tian, W.D.; Zhong, L.P.; Tian, Z.; Wang, L.Z.; Hu, Y.H.; Li, J. Adenoid cystic carcinoma of the head and neck: Clin.icopathologic analysis of 218 cases in a Chinese population. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2013, 115, 368–375. [Google Scholar] [CrossRef]
  31. Coca-Pelaz, A.; Rodrigo, J.P.; Bradley, P.J.; Vander Poorten, V.; Triantafyllou, A.; Hunt, J.L.; Strojan, P.; Rinaldo, A.; Haigentz, M., Jr.; Takes, R.P.; et al. Adenoid cystic carcinoma of the head and neck—An update. Oral Oncol. 2015, 51, 652–661. [Google Scholar] [CrossRef]
  32. Stelow, E.B.; Bishop, J.A. Update from the 4th Edition of the World Health Organization Classification of Head and Neck Tumours: Tumors of the Nasal Cavity, Paranasal Sinuses and Skull Base. Head Neck Pathol. 2017, 11, 3–15. [Google Scholar] [CrossRef] [PubMed]
  33. Liu, X.; Yang, X.; Zhan, C.; Zhang, Y.; Hou, J.; Yin, X. Perineural Invasion in Adenoid Cystic Carcinoma of the Salivary Glands: Where We Are and Where We Need to Go. Front. Oncol. 2020, 10, 1493. [Google Scholar] [CrossRef]
  34. Amit, M.; Na’ara, S.; Trejo-Leider, L.; Ramer, N.; Burstein, D.; Yue, M.; Miles, B.; Yang, X.; Lei, D.; Bjoerndal, K.; et al. Defining the surgical margins of adenoid cystic carcinoma and their impact on outcome: An international collaborative study. Head Neck 2017, 39, 1008–1014. [Google Scholar] [CrossRef]
  35. Mendenhall, W.M.; Morris, C.G.; Amdur, R.J.; Werning, J.W.; Hinerman, R.W.; Villaret, D.B. Radiotherapy alone or combined with surgery for adenoid cystic carcinoma of the head and neck. Head Neck 2004, 26, 154–162. [Google Scholar] [CrossRef]
  36. Tannock, I.F.; Sutherland, D.J. Chemotherapy for adenocystic carcinoma. Cancer 1980, 46, 452–454. [Google Scholar] [CrossRef] [PubMed]
  37. Schramm, V.L., Jr.; Srodes, C.; Myers, E.N. Cisplatin therapy for adenoid cystic carcinoma. Arch. Otolaryngol. 1981, 107, 739–741. [Google Scholar] [CrossRef] [PubMed]
  38. 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]
  39. 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. EORTC Head and Neck Cancer Cooperative Group. Ann. Oncol. 1996, 7, 867–869. [Google Scholar] [CrossRef] [PubMed]
  40. 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]
  41. 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]
  42. 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]
  43. Sahara, S.; Herzog, A.E.; Nor, J.E. Systemic therapies for salivary gland adenoid cystic carcinoma. Am. J. Cancer Res. 2021, 11, 4092–4110. [Google Scholar] [PubMed]
  44. Licitra, L.; Cavina, R.; Grandi, C.; Palma, S.D.; Guzzo, M.; Demicheli, R.; Molinari, R. Cisplatin, doxorubicin and cyclophosphamide in advanced salivary gland carcinoma. A phase II trial of 22 patients. Ann. Oncol. 1996, 7, 640–642. [Google Scholar] [CrossRef] [PubMed]
  45. 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]
  46. 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]
  47. 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]
  48. Van Weert, S.; Bloemena, E.; van der Waal, I.; de Bree, R.; Rietveld, D.H.; Kuik, J.D.; Leemans, C.R. Adenoid cystic carcinoma of the head and neck: A single-center analysis of 105 consecutive cases over a 30-year period. Oral Oncol. 2013, 49, 824–829. [Google Scholar] [CrossRef]
  49. Linton, O.R.; Moore, M.G.; Brigance, J.S.; Summerlin, D.J.; McDonald, M.W. Proton therapy for head and neck adenoid cystic carcinoma: Initial clinical outcomes. Head Neck 2015, 37, 117–124. [Google Scholar] [CrossRef]
  50. Holliday, E.; Bhattasali, O.; Kies, M.S.; Hanna, E.; Garden, A.S.; Rosenthal, D.I.; Morrison, W.H.; Gunn, G.B.; Phan, J.; Zhu, X.R.; et al. Postoperative Intensity-Modulated Proton Therapy for Head and Neck Adenoid Cystic Carcinoma. Int. J. Part Ther. 2016, 2, 533–543. [Google Scholar] [CrossRef]
  51. Bhattasali, O.; Holliday, E.; Kies, M.S.; Hanna, E.Y.; Garden, A.S.; Rosenthal, D.I.; Morrison, W.H.; Gunn, G.B.; Fuller, C.D.; Zhu, X.R.; et al. Definitive proton radiation therapy and concurrent cisplatin for unresectable head and neck adenoid cystic carcinoma: A series of 9 cases and a critical review of the literature. Head Neck 2016, 38 (Suppl. 1), E1472–E1480. [Google Scholar] [CrossRef] [PubMed]
  52. Morimoto, K.; Demizu, Y.; Hashimoto, N.; Mima, M.; Terashima, K.; Fujii, O.; Otsuki, N.; Murakami, M.; Fuwa, N.; Nibu, K. Particle radiotherapy using protons or carbon ions for unresectable locally advanced head and neck cancers with skull base invasion. Jpn J. Clin. Oncol. 2014, 44, 428–434. [Google Scholar] [CrossRef]
  53. Pelak, M.J.; Walser, M.; Bachtiary, B.; Hrbacek, J.; Lomax, A.J.; Kliebsch, U.L.; Beer, J.; Pica, A.; Malyapa, R.; Weber, D.C. Clinical outcomes of head and neck adenoid cystic carcinoma patients treated with pencil beam-scanning proton therapy. Oral Oncol. 2020, 107, 104752. [Google Scholar] [CrossRef]
  54. Pommier, P.; Liebsch, N.J.; Deschler, D.G.; Lin, D.T.; McIntyre, J.F.; Barker, F.G., 2nd; Adams, J.A.; Lopes, V.V.; Varvares, M.; Loeffler, J.S.; et al. Proton beam radiation therapy for skull base adenoid cystic carcinoma. Arch. Otolaryngol. Head Neck Surg. 2006, 132, 1242–1249. [Google Scholar] [CrossRef]
  55. Mizoe, J.E.; Hasegawa, A.; Jingu, K.; Takagi, R.; Bessyo, H.; Morikawa, T.; Tonoki, M.; Tsuji, H.; Kamada, T.; Tsujii, H.; et al. Results of carbon ion radiotherapy for head and neck cancer. Radiother Oncol. 2012, 103, 32–37. [Google Scholar] [CrossRef] [PubMed]
  56. Koto, M.; Hasegawa, A.; Takagi, R.; Ikawa, H.; Naganawa, K.; Mizoe, J.E.; Jingu, K.; Tsujii, H.; Tsuji, H.; Kamada, T.; et al. Evaluation of the safety and efficacy of carbon ion radiotherapy for locally advanced adenoid cystic carcinoma of the tongue base. Head Neck 2016, 38 (Suppl. 1), E2122–E2126. [Google Scholar] [CrossRef]
  57. Lang, K.; Adeberg, S.; Harrabi, S.; Held, T.; Kieser, M.; Debus, J.; Herfarth, K. Adenoid cystic Carcinoma and Carbon ion Only irradiation (ACCO): Study protocol for a prospective, open, randomized, two-armed, phase II study. BMC Cancer 2021, 21, 812. [Google Scholar] [CrossRef]
  58. Akbaba, S.; Ahmed, D.; Lang, K.; Held, T.; Mattke, M.; Hoerner-Rieber, J.; Herfarth, K.; Rieken, S.; Plinkert, P.; Debus, J.; et al. Results of a combination treatment with intensity modulated radiotherapy and active raster-scanning carbon ion boost for adenoid cystic carcinoma of the minor salivary glands of the nasopharynx. Oral Oncol. 2019, 91, 39–46. [Google Scholar] [CrossRef]
  59. Mastella, E.; Molinelli, S.; Magro, G.; Russo, S.; Bonora, M.; Ronchi, S.; Ingargiola, R.; Jensen, A.D.; Ciocca, M.; Vischioni, B.; et al. In Silico Feasibility Study of Carbon Ion Radiotherapy With Simultaneous Integrated Boost for Head and Neck Adenoid Cystic Carcinoma. Front. Oncol. 2021, 11, 772580. [Google Scholar] [CrossRef]
  60. Davis, C.; Sikes, J.; Namaranian, P.; Laramore, G.; Dillon, J.K. Neutron Beam Radiation Therapy: An Overview of Treatment and Oral Complications When Treating Salivary Gland Malignancies. J. Oral Maxillofac. Surg. 2016, 74, 830–835. [Google Scholar]
  61. Ebner, D.K.; Malouff, T.D.; Frank, S.J.; Koto, M. The Role of Particle Therapy in Adenoid Cystic Carcinoma and Mucosal Melanoma of the Head and Neck. Int. J. Part Ther. 2021, 8, 273–284. [Google Scholar] [CrossRef]
  62. Timoshchuk, M.A.; Dekker, P.; Hippe, D.S.; Parvathaneni, U.; Liao, J.J.; Laramore, G.E.; Dillon, J.K. The efficacy of neutron radiation therapy in treating salivary gland malignancies. Oral Oncol. 2019, 88, 51–57. [Google Scholar] [CrossRef]
  63. Aljabab, S.; Lui, A.; Wong, T.; Liao, J.; Laramore, G.; Parvathaneni, U. A Combined Neutron and Proton Regimen for Advanced Salivary Tumors: Early Clinical Experience. Cureus 2021, 13, e14844. [Google Scholar] [CrossRef]
  64. Takagi, M.; Demizu, Y.; Hashimoto, N.; Mima, M.; Terashima, K.; Fujii, O.; Jin, D.; Niwa, Y.; Morimoto, K.; Akagi, T.; et al. Treatment outcomes of particle radiotherapy using protons or carbon ions as a single-modality therapy for adenoid cystic carcinoma of the head and neck. Radiother. Oncol. 2014, 113, 364–370. [Google Scholar] [CrossRef] [PubMed]
  65. Sulaiman, N.S.; Demizu, Y.; Koto, M.; Saitoh, J.I.; Suefuji, H.; Tsuji, H.; Ohno, T.; Shioyama, Y.; Okimoto, T.; Daimon, T.; et al. Multicenter Study of Carbon-Ion Radiation Therapy for Adenoid Cystic Carcinoma of the Head and Neck: Subanalysis of the Japan Carbon-Ion Radiation Oncology Study Group (J-CROS) Study (1402 HN). Int. J. Radiat. Oncol. Biol. Phys. 2018, 100, 639–646. [Google Scholar] [CrossRef] [PubMed]
  66. Douglas, J.G.; Laramore, G.E.; Austin-Seymour, M.; Koh, W.; Stelzer, K.; Griffin, T.W. Treatment of locally advanced adenoid cystic carcinoma of the head and neck with neutron radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 2000, 46, 551–557. [Google Scholar] [CrossRef]
  67. Frierson, H.F., Jr.; El-Naggar, A.K.; Welsh, J.B.; Sapinoso, L.M.; Su, A.I.; Cheng, J.; Saku, T.; Moskaluk, C.A.; Hampton, G.M. Large scale molecular analysis identifies genes with altered expression in salivary adenoid cystic carcinoma. Am. J. Pathol. 2002, 161, 1315–1323. [Google Scholar] [CrossRef]
  68. Sinner, D.; Kordich, J.J.; Spence, J.R.; Opoka, R.; Rankin, S.; Lin, S.C.; Jonatan, D.; Zorn, A.M.; Wells, J.M. Sox17 and Sox4 differentially regulate beta-catenin/T-cell factor activity and proliferation of colon carcinoma cells. Mol. Cell Biol. 2007, 27, 7802–7815. [Google Scholar] [CrossRef]
  69. Cheung, M.; Abu-Elmagd, M.; Clevers, H.; Scotting, P.J. Roles of Sox4 in central nervous system development. Brain Res. Mol. Brain Res. 2000, 79, 180–191. [Google Scholar] [CrossRef]
  70. Funasaka, Y.; Boulton, T.; Cobb, M.; Yarden, Y.; Fan, B.; Lyman, S.D.; Williams, D.E.; Anderson, D.M.; Zakut, R.; Mishima, Y.; et al. c-Kit-kinase induces a cascade of protein tyrosine phosphorylation in normal human melanocytes in response to mast cell growth factor and stimulates mitogen-activated protein kinase but is down-regulated in melanomas. Mol. Biol. Cell 1992, 3, 197–209. [Google Scholar] [CrossRef]
  71. 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] [PubMed]
  72. 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]
  73. Lim, J.J.; Kang, S.; Lee, M.R.; Pai, H.K.; Yoon, H.J.; Lee, J.I.; Hong, S.P.; Lim, C.Y. Expression of vascular endothelial growth factor in salivary gland carcinomas and its relation to p53, Ki-67 and prognosis. J. Oral Pathol. Med. 2003, 32, 552–561. [Google Scholar] [CrossRef]
  74. Yu, F.; Jiang, X.Z.; Chen, W.T.; Zhao, Y.F.; Zhou, X.J. Microvessel density and expression of vascular endothelial growth factor in adenoid cystic carcinoma of salivary gland. Shanghai Kou Qiang Yi Xue 2003, 12, 443–446. [Google Scholar]
  75. Li, Z.; Tang, P.; Xu, Z. Clinico-pathological significance of microvessel density and vascular endothelial growth factor expression in adenoid cystic carcinoma of salivary glands. Zhonghua Kou Qiang Yi Xue Za Zhi 2001, 36, 212–214. [Google Scholar]
  76. 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]
  77. Kowalski, P.J.; Paulino, A.F. Perineural invasion in adenoid cystic carcinoma: Its causation/promotion by brain-derived neurotrophic factor. Hum. Pathol. 2002, 33, 933–936. [Google Scholar] [CrossRef]
  78. Jia, S.; Wang, W.; Hu, Z.; Shan, C.; Wang, L.; Wu, B.; Yang, Z.; Yang, X.; Lei, D. BDNF mediated TrkB activation contributes to the EMT progression and the poor prognosis in human salivary adenoid cystic carcinoma. Oral Oncol. 2015, 51, 64–70. [Google Scholar] [CrossRef]
  79. Wang, L.; Sun, M.; Jiang, Y.; Yang, L.; Lei, D.; Lu, C.; Zhao, Y.; Zhang, P.; Yang, Y.; Li, J. Nerve growth factor and tyrosine kinase A in human salivary adenoid cystic carcinoma: Expression patterns and effects on in vitro invasive behavior. J. Oral Maxillofac. Surg. 2006, 64, 636–641. [Google Scholar] [CrossRef]
  80. Hao, L.; Xiao-lin, N.; Qi, C.; Yi-ping, Y.; Jia-quan, L.; Yan-ning, L. Nerve growth factor and vascular endothelial growth factor: Retrospective analysis of 63 patients with salivary adenoid cystic carcinoma. Int. J. Oral Sci. 2010, 2, 35–44. [Google Scholar] [PubMed]
  81. Shen-Ong, G.L. The myb oncogene. Biochim. Biophys. Acta 1990, 1032, 39–52. [Google Scholar] [CrossRef]
  82. Stenman, G.; Persson, F.; Andersson, M.K. Diagnostic and therapeutic implications of new molecular biomarkers in salivary gland cancers. Oral Oncol. 2014, 50, 683–690. [Google Scholar] [CrossRef]
  83. Liu, X.; Chen, D.; Lao, X.; Liang, Y. The value of MYB as a prognostic marker for adenoid cystic carcinoma: Meta-analysis. Head Neck 2019, 41, 1517–1524. [Google Scholar] [CrossRef] [PubMed]
  84. Bienz, M.; Clevers, H. Linking colorectal cancer to Wnt signaling. Cell 2000, 103, 311–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Lustig, B.; Behrens, J. The Wnt signaling pathway and its role in tumor development. J. Cancer Res. Clin. Oncol. 2003, 129, 199–221. [Google Scholar] [CrossRef] [PubMed]
  86. Daa, T.; Kashima, K.; Kaku, N.; Suzuki, M.; Yokoyama, S. Mutations in components of the Wnt signaling pathway in adenoid cystic carcinoma. Mod. Pathol. 2004, 17, 1475–1482. [Google Scholar] [CrossRef]
  87. Daa, T.; Kaku, N.; Kashima, K.; Nakayama, I.; Yokoyama, S. Expression of beta-catenin, E-cadherin and cyclin D1 in adenoid cystic carcinoma of the salivary gland. J. Exp. Clin. Cancer Res. 2005, 24, 83–87. [Google Scholar] [PubMed]
  88. Zhou, C.X.; Gao, Y. Aberrant expression of beta-catenin, Pin1 and cylin D1 in salivary adenoid cystic carcinoma: Relation to tumor proliferation and metastasis. Oncol. Rep. 2006, 16, 505–511. [Google Scholar]
  89. El-Deiry, W.S.; Harper, J.W.; O’Connor, P.M.; Velculescu, V.E.; Canman, C.E.; Jackman, J.; Pietenpol, J.A.; Burrell, M.; Hill, D.E.; Wang, Y.; et al. WAF1/CIP1 is induced in p53-mediated G1 arrest and apoptosis. Cancer Res. 1994, 54, 1169–1174. [Google Scholar]
  90. Haupt, S.; Berger, M.; Goldberg, Z.; Haupt, Y. Apoptosis—The p53 network. J. Cell Sci. 2003, 116 Pt 20, 4077–4085. [Google Scholar] [CrossRef]
  91. Kiyoshima, T.; Shima, K.; Kobayashi, I.; Matsuo, K.; Okamura, K.; Komatsu, S.; Rasul, A.M.; Sakai, H. Expression of p53 tumor suppressor gene in adenoid cystic and mucoepidermoid carcinomas of the salivary glands. Oral Oncol. 2001, 37, 315–322. [Google Scholar] [CrossRef]
  92. Bressac-de Paillerets, B. Discovery of a new gene frequently inactivated in human cancers: Protein p16, a cell cycle regulator. Bull. Cancer 1994, 81, 853–856. [Google Scholar] [PubMed]
  93. Foulkes, W.D.; Flanders, T.Y.; Pollock, P.M.; Hayward, N.K. The CDKN2A (p16) gene and human cancer. Mol. Med. 1997, 3, 5–20. [Google Scholar] [CrossRef] [PubMed]
  94. Sherr, C.J. Cancer cell cycles. Science 1996, 274, 1672–1677. [Google Scholar] [CrossRef]
  95. Maruya, S.; Kurotaki, H.; Shimoyama, N.; Kaimori, M.; Shinkawa, H.; Yagihashi, S. Expression of p16 protein and hypermethylation status of its promoter gene in adenoid cystic carcinoma of the head and neck. ORL J. Otorhinolaryngol. Relat. Spec. 2003, 65, 26–32. [Google Scholar] [CrossRef] [PubMed]
  96. Guo, X.L.; Sun, S.Z.; Wei, F.C. Mechanisms of p16 gene inactivation salivary adenoid cystic carcinoma. Hua Xi Kou Qiang Yi Xue Za Zhi 2005, 23, 418–420. [Google Scholar]
  97. Mithani, S.K.; Shao, C.; Tan, M.; Smith, I.M.; Califano, J.A.; El-Naggar, A.K.; Ha, P.K. Mitochondrial mutations in adenoid cystic carcinoma of the salivary glands. PLoS ONE 2009, 4, e8493. [Google Scholar] [CrossRef] [Green Version]
  98. Chang, B.; Yang, H.; Jiao, Y.; Wang, K.; Liu, Z.; Wu, P.; Li, S.; Wang, A. SOD2 deregulation enhances migration, invasion and has poor prognosis in salivary adenoid cystic carcinoma. Sci. Rep. 2016, 6, 25918. [Google Scholar] [CrossRef] [PubMed]
  99. Harman, D. Aging: A theory based on free radical and radiation chemistry. J. Gerontol. 1956, 11, 298–300. [Google Scholar] [CrossRef]
  100. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef]
  101. Halliwell, B. Oxygen and nitrogen are pro-carcinogens. Damage to DNA by reactive oxygen, chlorine and nitrogen species: Measurement, mechanism and the effects of nutrition. Mutat. Res. 1999, 443, 37–52. [Google Scholar] [CrossRef]
  102. Halliwell, B.; Gutteridge, J.M. Gutteridge, Lipid peroxidation, oxygen radicals, cell damage, and antioxidant therapy. Lancet 1984, 1, 1396–1397. [Google Scholar] [CrossRef]
  103. Trachootham, D.; Lu, W.; Ogasawara, M.A.; Nilsa, R.D.; Huang, P. Redox regulation of cell survival. Antioxid. Redox Signal. 2008, 10, 1343–1374. [Google Scholar] [CrossRef] [PubMed]
  104. Liu, Z.; Gao, J.; Yang, Y.; Zhao, H.; Ma, C.; Yu, T. Potential targets identified in adenoid cystic carcinoma point out new directions for further research. Am. J. Transl. Res. 2021, 13, 1085–1108. [Google Scholar]
  105. Rack, S.; Feeney, L.; Hapuarachi, B.; Adderley, H.; Woodhouse, L.; Betts, G.; Burghel, G.J.; Harrington, K.J.; Metcalf, R. Evaluation of the Clinical Utility of Genomic Profiling to Inform Selection of Clinical Trial Therapy in Salivary Gland Cancer. Cancers 2022, 14, 1133. [Google Scholar] [CrossRef]
  106. Meurette, O.; Mehlen, P. Notch Signaling in the Tumor Microenvironment. Cancer Cell 2018, 34, 536–548. [Google Scholar] [CrossRef] [PubMed]
  107. 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]
  108. 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] [Green Version]
  109. 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]
  110. 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]
  111. Ho, A.L.; Sherman, E.J.; Baxi, S.S.; Haque, S.; Ni, A.; Antonescu, C.R.; Katabi, N.; Morris, L.G.; Chan, T.A.-t.; Pfister, D.G. Phase II study of regorafenib in progressive, recurrent/metastatic adenoid cystic carcinoma. J. Clin. Oncol. 2016, 34 (Suppl. 15), 6096. [Google Scholar] [CrossRef]
  112. 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]
  113. Tchekmedyian, V.; Sherman, E.; Dunn, L.; Tran, C.; Baxi, S.; Katabi, N.; Antonescu, C.; Ostrovnaya, I.; Haque, S.; Pfister, D.; 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]
  114. 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]
  115. Laurie, S.A.; Ho, A.L.; Fury, M.G.; Sherman, E.; Pfister, D.G. Systemic therapy in the management of metastatic or locally recurrent adenoid cystic carcinoma of the salivary glands: A systematic review. Lancet Oncol. 2011, 12, 815–824. [Google Scholar] [CrossRef] [PubMed]
Table 1. Particle radiation therapy. # means number.
Table 1. Particle radiation therapy. # means number.
TherapyAuthors# of Patients with ACCLocal
Control
OSDisease-Free SurvivalProgression-Free Survival
ProtonsLinton et al. [49]262 y LC = 86% (recurrent disease)
95% (primary disease)
57% (recurrent disease), 93% (primary disease)--
Protons + photonsPommier et al. [54]235 y LC = 93%77%56%-
Protons vs. CIRTTakagi et al. [64]3475%63%-39%
CIRTMizoe [55]695 y LC = 73%68%--
CIRTKoto [56]18 5 y LC = 92% 72%44%-
CIRTAkbaba [58]592 y LC = 83%87%--
CIRTMastella [59]10----
CIRTSulaiman [65]289 2 y = 88% 94%-68%
NeutronsDouglas [66]1515 y LC = 57%72%--
Table 2. Clinical phase II trials investigating systemic molecular biological therapeutic approaches between 2012 and 2022. # means number.
Table 2. Clinical phase II trials investigating systemic molecular biological therapeutic approaches between 2012 and 2022. # means number.
Phase II TrialTargetAuthorsYear# of Patients with ACCObjective ResponseProgression-Free SurvivalMedian SurvivalStudy DesignNOS Score
Sunitinib 37.5 mgMultikinase inhibitorChau et al. [107]20121407.2 months18.7 monthsClinical trial4
Everolismus mTORKim et al. [108]201434011.2 months-Clinical trial4
Sorafenib 400 mg Multikinase inhibitorThomson et al. [109]201523011.3 months19.6 monthsClinical trial5
Nelfinavir 1250 mgInhibitor of Akt signallingHoover et al. [110]20151505.5 months-Clinical trial3.5
Dovitinib 500 mg FGRF, VEGFRKeam et al. [112]20153216.0 months-Clinical trial4
Regorafenib 120 mg FGRF, VEGFRHo et al. [111]2017380--Clinical trial3
Lenvatinib FGRF, VEGFRTchekmedyian [113]201932517.5 months-Clinical trial4
LenvatinibMultikinase inhibitorLocati et al. [114]20202839.1 months27 monthsClinical trial6
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dewenter, I.; Otto, S.; Kakoschke, T.K.; Smolka, W.; Obermeier, K.T. Recent Advances, Systemic Therapy, and Molecular Targets in Adenoid Cystic Carcinoma of the Head and Neck. J. Clin. Med. 2023, 12, 1463. https://doi.org/10.3390/jcm12041463

AMA Style

Dewenter I, Otto S, Kakoschke TK, Smolka W, Obermeier KT. Recent Advances, Systemic Therapy, and Molecular Targets in Adenoid Cystic Carcinoma of the Head and Neck. Journal of Clinical Medicine. 2023; 12(4):1463. https://doi.org/10.3390/jcm12041463

Chicago/Turabian Style

Dewenter, Ina, Sven Otto, Tamara Katharina Kakoschke, Wenko Smolka, and Katharina Theresa Obermeier. 2023. "Recent Advances, Systemic Therapy, and Molecular Targets in Adenoid Cystic Carcinoma of the Head and Neck" Journal of Clinical Medicine 12, no. 4: 1463. https://doi.org/10.3390/jcm12041463

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