Abstract
Salivary duct carcinoma (SDC) is an aggressive subtype of primary salivary gland carcinoma, often with an advanced stage at presentation and high rates of metastasis and recurrence. It most commonly arises in the parotid gland of older men and microscopically resembles high-grade breast ductal carcinoma. While 50 years have lapsed since the first report of this entity, recent intensive studies have shed light on its biologic, genetic, and clinical characteristics. The diagnosis of SDC is aided by the immunohistochemical expression of androgen receptor (AR) coupled with its characteristic histomorphology. Fine needle aspiration typically reveals cytologic features of high-grade carcinoma, and ancillary studies using cell block material can facilitate the specific diagnosis of SDC. In surgical specimens, certain histologic features are important prognostic factors, including nuclear pleomorphism, mitotic counts, vascular invasion, and the morphology at the invasion front. Several clinical studies have shown promising results using targeted therapy for AR and human epidermal growth factor receptor 2 (HER2), and the latest version of the National Comprehensive Cancer Network guidelines recommends the evaluation of AR and HER2 status before treatment. Recent molecular analyses have revealed multiple heterogeneous alterations in well-known oncogenes and tumor suppressor genes, including TP53, HRAS, PIK3CA, PTEN, and BRAF. Clinical trials of drugs targeting these genes may broaden the treatment options for SDC in the near future.
Keywords: androgen receptor, fine needle aspiration, human epidermal growth factor receptor 2, salivary duct carcinoma, salivary gland tumor, targeted therapy
INTRODUCTION
Salivary duct carcinoma (SDC) is the most aggressive tumor of the 21 subtypes of primary salivary gland carcinoma in the latest World Health Organization classification (Table 1),1 showing high rates of local recurrence and distant metastases.2,3 SDC mainly affects men over 50 years of age and accounts for approximately 5% to 10% of all salivary gland malignancies.1,2 Most SDCs arise from the major salivary glands, especially the parotid gland, although tumors in the minor salivary glands, sinonasal tract, and lacrimal gland have also been reported.2,4,5
TABLE 1.
Subtypes of Primary Salivary Gland Carcinoma
| Mucoepidermoid carcinoma |
| Adenoid cystic carcinoma |
| Acinic cell carcinoma |
| Polymorphous adenocarcinoma |
| Clear cell carcinoma |
| Basal cell adenocarcinoma |
| Intraductal carcinoma |
| Adenocarcinoma, not otherwise specified |
| Salivary duct carcinoma |
| Myoepithelial carcinoma |
| Epithelial-myoepithelial carcinoma |
| Carcinoma ex pleomorphic adenoma |
| Secretory carcinoma |
| Sebaceous adenocarcinoma |
| Carcinosarcoma |
| Poorly differentiated carcinoma |
| Undifferentiated carcinoma |
| Large cell neuroendocrine carcinoma |
| Small cell neuroendocrine carcinoma |
| Lymphoepithelial carcinoma |
| Squamous cell carcinoma |
| Oncocytic carcinoma |
Adapted from El-Naggar et al.1
Radiologically, most SDCs show features of high-grade malignant tumors. By contrast-enhanced computed tomography, an ill-defined border, invasion into adjacent tissue, and necrosis are more frequently observed in SDC than in other salivary gland tumor subtypes (Fig. 1).6,7 Calcification is also sometimes detected, but this may suggest the presence of a preexisting pleomorphic adenoma rather than SDC itself.6 Magnetic resonance imaging is another useful imaging modality with a high sensitivity and specificity.8,9
FIGURE 1.

Contrast-enhanced computed tomography images of salivary duct carcinoma (SDC) in (A) the parotid gland and (B) the submandibular gland. SDCs (arrowheads) exhibit a heterogeneously enhanced mass with an ill-defined border. Occasional calcification and necrosis are noted.
Studies of SDC were previously limited to small cohorts or mixed populations with other subtypes of salivary gland carcinomas exhibiting different biologic behaviors. However, recent multi-institutional studies and the advent of new immunohistochemical and molecular techniques have better defined the biology of SDC and will likely affect future diagnostic and therapeutic options.
This review provides a comprehensive overview of these updates to raise awareness of SDC when making a cytological diagnosis.
HISTOLOGY
SDC is defined as an aggressive epithelial malignancy resembling high-grade mammary ductal carcinoma characterized by tumor cell proliferation with comedonecrosis and a cribriform growth pattern (Fig. 2A).2 The tumor cells have large pleomorphic nuclei with conspicuous nucleoli and abundant eosinophilic cytoplasm (Fig. 2B). Although SDC histologically resembles breast carcinoma, it generally shows more marked cellular atypia and more mitoses than the latter.10 Several histologic variants of SDC have been reported, including mucin-rich, sarcomatoid, invasive micropapillary, oncocytic, and rhabdoid, but these histomorphological features essentially appear in combination with more typical histologic features of SDC (Fig. 2C).11–15 Sarcomatoid, invasive micropapillary, and rhabdoid SDC are suggested to show a poorer prognosis than conventional SDC.12,13,15 Approximately 20%−59% of SDC cases arise from preexisting pleomorphic adenoma (ex PA), and as such, SDC is the most common subtype of carcinoma ex PA (Fig. 2D). Contrary to a common misunderstanding, previous data have shown that there is no marked difference in the prognosis between SDC ex PA and de novo SDC.3,16–18 Encapsulated and minimally invasive SDC ex PA were shown to exhibit a better prognosis than widely invasive SDC ex PA.19,20 There is no consensus concerning the cutoff value for minimal invasion, and reported values have ranged from 1.5 to 6 mm beyond the fibrous capsule of PA.19,20
FIGURE 2.

Histology and immunohistochemistry of salivary duct carcinoma (SDC). (A) Low-power view of an SDC tumor shows tumor cell proliferation with comedo-type central necrosis resembling breast ductal carcinoma. (B) Tumor cells of SDC have large nuclei with conspicuous nucleoli and abundant eosinophilic cytoplasm. (C) Sarcomatoid SDC. Abrupt transition from conventional SDC (left portion) to fascicular proliferation of highly atypical spindle cells is observed. (D) SDC ex pleomorphic adenoma. Ducts composed of atypical SDC cells (left portion) abut myxochondroid stroma of preexisting pleomorphic adenoma (right portion). (E) Invasive morphology of the tumor front. Many tumor buds (nests comprising <4 cells, indicated with arrows) and poorly differentiated clusters (nests comprising >5 cells, indicated with arrowheads) are observed. (F) Diffuse and strong immunohistochemical staining for androgen receptor (left panel) and human epidermal growth factor receptor 2 (right panel).
Although SDC is basically a highly malignant tumor, some histologic features are prognosticators independent of TNM stage.3,10,21–24 Because no screening system for its early detection is available, most cases of SDC present at an advanced stage, and reported cases of noninvasive in situ SDC are limited.25,26 The histomorphology of the invasive front is an important feature predicting its malignant potential; cases with a high number of tumor buds (tumor cell clusters composed of up to 4 cells) and poorly differentiated clusters (tumor cell clusters composed of more than 5 cells without gland formation) show a poorer prognosis than others (Fig. 2E).10 In a recent study, Nakaguro et al10 proposed a risk stratification model based on 4 histological features: prominent nuclear pleomorphism, mitoses, vascular invasion, and poorly differentiated clusters (Table 2).
TABLE 2.
Histologic Risk Stratification Model for Salivary Duct Carcinoma
| Adverse prognostic factors |
| Prominent nuclear pleomorphism |
| ≥30 Mitoses/10 high-power fields |
| Vascular invasion (H&E stain) |
| ≥5 Poorly differentiated clusters |
| Total number of positive factors |
| 0, 1: Low risk |
| 2, 3: Intermediate risk |
| 4: High risk |
Adapted from Nakaguro et al.10
CYTOLOGY
Aspirates of SDC show overt nuclear features of high-grade carcinoma, making them readily identifiable by fine needle aspiration (FNA) as “malignant” in the Milan System for Reporting Salivary Gland Cytopathology (MSRSGC).27,28 FNA specimens of SDC are composed of large tumor cells with abundant eosinophilic cytoplasm and large, round to oval atypical nuclei with prominent nucleoli. Tumor cells are either isolated or occur in cohesive 3-dimensional clusters that sometimes exhibit a papillary cytoarchitecture (Fig. 3A–D).29,30 The background of the FNA may contain necrotic cellular debris reflecting the surgical pathology correlate of comedonecrosis (Fig. 3C). The cytological diagnosis of malignancy is straightforward when the sample is adequate; however, a specific diagnosis of SDC based solely on cytomorphologic features is not always possible. The differential diagnosis of SDC in FNA samples includes high-grade mucoepidermoid carcinoma; high-grade transformation of various primary salivary gland carcinomas; adenocarcinoma, not otherwise specified; and metastatic cancers from other anatomic sites.30 The features of the histological variants of SDC can be observed in FNA (Fig. 3E).
FIGURE 3.

Fine needle aspiration of salivary duct carcinoma (SDC). (A) A low-power view shows 3-dimensional pseudopapillary clusters of tumor cells. (B) Tightly cohesive cluster with nuclear crowding. Tumor cells show nuclear pleomorphism suggestive of high-grade malignant tumor. (C) Loose cell clusters are observed in a necrotic background. Tumor cells have large pleomorphic nuclei with coarse chromatin and conspicuous nucleoli and abundant eosinophilic cytoplasm. (D) Isolated tumor cells in the aspirates have round to oval nuclei and abundant cytoplasm. Markedly enlarged nucleus, mutinucleation, and cannibalism are noted. (E) Aspirate from a sarcomatoid SDC tumor shows isolated atypical spindle cells on a hemorrhagic background. (F) Immunohistochemical staining of a formalin-fixed cell block specimen. Tumor cells express androgen receptor (left panel) and human epidermal growth factor receptor 2 (right panel).
One pitfall in the FNA diagnosis of SDC is SDC ex PA. As described in the Histology section, SDC is among the most common types of carcinoma ex PA. A cytologic diagnosis of malignancy is sometimes difficult in cases of carcinoma ex PA and the sensitivity is variable depending upon sampling of the malignant component (29%−80%).31–33 In many cases of SDC ex PA, the SDC component will predominate to the near exclusion of any residual classical PA component, thus limiting the ability to recognize the aspirate as SDC ex PA versus a de novo SDC. However, this distinction is usually of less clinical importance, except perhaps for noninvasive examples. It is sufficient to recognize the aspirate as a high-grade carcinoma to guide the proper clinical management.
Most FNA cases of SDC should be readily diagnosed as high-grade carcinoma and classified appropriately using the MSRSGC; however, ancillary immunohistochemical studies on formalin-fixed paraffin-embedded cell block material can be used to more specifically classify the FNA as SDC. Characteristic immunohistochemical findings in the context of the corresponding high-grade cytomorphologic features are sufficient to render a diagnosis of SDC (Fig. 3F).34–36
IMMUNOHISTOCHEMISTRY
Immunohistochemically, a strong and diffuse AR expression is a characteristic feature and a practical tool in the diagnosis of SDC (Fig. 2F), especially when paired with positive staining for GATA3 or GCDFP-15.16,22,37–39 Recent cohorts with a large number of patients showed AR expression in over 90% of cases, and the diagnosis of “AR-negative SDC” requires the careful exclusion of other entities, such as high-grade mucoepidermoid carcinoma and high-grade transformation of other subtypes of salivary carcinomas.18,22,37,40–47 The p63 expression is helpful for differentiating mucoepidermoid carcinoma from SDC, which is usually negative for p63.48,49
Conversely, AR expression has been reported in approximately 10% of cases of salivary gland tumors other than SDC, including pleomorphic adenoma, adenoid cystic carcinoma, and acinic cell carcinoma; however, even if AR expression is present, it is weak to intermediate and focal. The reported sensitivities and specificities of AR expression for diagnosing SDC were 73% to 100% and 76% to 100%, respectively.37,40,48 The diagnosis of SDC should be based primarily on the morphologic features with the support of AR immunohistochemistry.40
By definition, SDC morphologically resembles invasive ductal carcinoma of the breast, and AR is also expressed in 85% to 100% of cases of invasive ductal breast carcinoma.46,50 The differential diagnosis between SDC and metastatic carcinoma of breast origin can thus be challenging from histopathological aspects alone. The lack of estrogen receptor or progesterone receptor expression in SDC may be helpful in such cases.46
In SDC, 16% to 83% of cases show human epidermal growth factor receptor 2 (HER2) expression by immunohistochemistry or HER2 amplification using FISH or next-generation sequencing (Fig. 2F).3,18,23,39–44,46,47,51–55 Although most benign salivary gland tumors are negative for HER2, some malignant tumors—including mucoepidermoid carcinoma and adenoid cystic carcinoma, acinic cell carcinoma, and squamous cell carcinoma—may show expression or amplification of HER2.40,51 AR and HER2 expression is not specific for SDC; however, evaluation of AR or HER2 status is recommended to help guide therapeutic strategies.56 The biomarker classification of SDC based on the AR and HER2 status and Ki-67 labeling index is reminiscent of that of breast carcinoma.41,42 The system suggested by Takase et al42 is useful for predicting the patient survival or selecting appropriate therapy (Table 3). Other immunohistochemical markers of SDCs are listed in Table 4.16,23,38,39,41,42,46–48,52,57
TABLE 3.
Revised Salivary Duct Carcinoma Classification Based on Biomarker Immunoprofiling
| HER2 | ||
|---|---|---|
| (+) | (−) | |
| AR | ||
| (+) | Apocrine HER2 | Apocrine A (Ki-67-low) |
| Apocrine B (Ki-67-high) | ||
| (−) | HER2-enriched | Double negative |
Abbreviations: AR, androgen receptor; HER2, human epidermal growth factor receptor 2.
A case was considered positive for AR when ≥20% of the tumor cell nuclei showed strong staining. A Ki-67 labeling index <40% was considered Ki-67–low, while ≥40% was Ki-67–high.
Adapted from Takase et al.42
TABLE 4.
Immunohistochemical Findings in Salivary Duct Carcinoma
| Markers | Positive Cases |
|---|---|
| AR | 73%–100% |
| HER2 (IHC or FISH) | 16%–83% |
| GATA3 | 73%–100% |
| GCDFP-15 | 76%–100% |
| CEA | 71%–72% |
| CK5/6 | 14%–76% |
| EGFR | 12%–33% |
| p53 (extreme negative or positive) | 42% |
| p63 | Usually negative |
| Average Ki-67 labeling index | 35%–46% |
Abbreviations: AR, androgen receptor; CEA, carcinoembryonic antigen; EGFR, epidermal growth factor receptor; FISH, fluorescence in situ hybridization; HER2, human epidermal growth factor receptor 2; IHC, immunohistochemistry.
Regarding the relationship between immunohistochemical markers and prognosis, an improved prognosis in patients with AR and its coregulatory protein forkhead box protein A1 (FOXA1) expression has been reported,42,58 but this finding was not observed in another large cohort.18 Although HER2 status does not affect patient survival, p53 extreme negative/positive staining (null or diffuse strong expression), which suggests the presence of a TP53 point mutation, and CK5/6 staining are significantly associated with a worse prognosis.18,42 “Apocrine A” type SDC (AR+, HER2-, Ki-67 low) on biomarker classification (Table 3) showed a better prognosis than other SDC tumors.42
The expression of PD-L1 in SDC has been evaluated in some studies; however, the clones of PD-L1 antibodies and the evaluation criteria varied from study to study, and the rate of positive cases ranged from 6% to 50%.59–64 The association between PD-L1 expression and prognosis were inconsistent between studies,60,64 and further investigations are therefore warranted.
MOLECULAR BIOLOGY
An increasing number of studies have reported the molecular characteristics of SDC. In addition to HER2 amplification, various hotspot mutations and fusion genes have been detected (Table 5).43,59,65–73 In comparison with other subtypes of salivary gland carcinomas, the results are heterogeneous, and most of the recurrent mutations are well-known oncogenes or tumor suppressor genes whose mutations are also observed in malignancies of other organs. Regarding the results of next-generation sequencing, because the panels, threshold, and analysis and validation methods have differed among studies, the assessment and comparison of the true mutation frequency is difficult, but TP53, PIK3CA, HRAS, and PTEN mutations are commonly and described frequently (Table 5). TP53 mutations, including missense and truncating mutations, and PI3K/AKT pathway activation evaluated by the p-Akt expression are associated with poor prognosis.67,74
TABLE 5.
Genetic Alterations Reported in Salivary Duct Carcinoma
| Pathway | Genes |
|---|---|
| Gene mutation | |
| DNA damage | TP53 (38%–68%), ATM |
| MAPK | HRAS (13%–27%), BRAF (3%–13%), KRAS, NRAS |
| RTK | HER2 (7%–20%), EGFR, HER4, RET, PDGFRA, KDR, FGFR1, FLT3 |
| PI3K/AKT | PIK3CA (18%–53%), PTEN (10%–53%), AKT1 (1.5%–19%) |
| Cell cycle | CDKN5A, FBXW7, RB1 |
| Wnt | APC, CTNNB1 |
| Other | ALK (2.7%–7%), KMT2C, ABL1, CDH1, SMO, VHL, STK11, PTPN11, SMAD4, NOTCH1, FOXA1 |
| Amplification | HER2 (30%−40%), NOTCH1, AKT1, SMARCA4 |
| Fusion gene | ETV6-NTRK, BCL6-TRADD, ABL1-PPP2R2C, CTNNB1-PLAG1, LIFR-PLAG1, NCOA4-RET, HNRNPH3-ALK |
Abbreviations: MAPK, mitogen-activated protein kinases; PI3K, phosphoinositide 3-kinase; RTK, receptor tyrosine kinases.
Values within parentheses indicate the incidence of genetic alterations.
Interestingly, the genetic changes observed specifically in certain subtypes of salivary gland carcinoma, such as the HRAS mutations in epithelial-myoepithelial carcinoma,75 CTNNB1 mutation in basal cell adenoma/adenocarcinoma,76–78 BRAF mutation in sialadenoma papilliferum,79 ETV6-NTRK3 fusion gene in secretory carcinoma,80 and NCOA4-RET in intraductal carcinoma,81 have also been detected in SDC without any histologic evidence of these tumor subtypes.65,70 The presence of subtype-specific genetic changes in SDC may suggest the origin of SDC, but evidence supporting this hypothesis remains sparse.
SDCs are reported to be microsatellite-stable.59,63 The overall tumor mutational burden (TMB) is 1.7 to 2.5 mutations/Mb, and a high TMB has been reported in only a small number of cases.59,70,82 The overall TMB value is higher than that in adenoid cystic carcinoma but close to that of breast, pancreas, prostate, and kidney cancers.70,73,82
TREATMENT
General Aspects
The rarity of SDC has made it impossible to conduct large definitive randomized trials to establish standard approaches to treatment. The current treatment options and trends for patients with SDC have been reviewed by various researchers.83–87 Because there is a considerable variety in the treatment of SDC between institutions, we herein mainly discuss the contents of the recent updates to the National Comprehensive Cancer Network (NCCN) guidelines.56 The major therapeutic approach for SDC is adequate and appropriate surgical resection. Elective neck dissection is recommended for patients with neck metastases and T3–4 patients without neck metastasis. Postoperative radiation therapy is an effective and appropriate therapeutic option according to the NCCN guidelines, regardless of the T stage and margin status.56,88–90 For recurrent unresectable and/or metastatic SDC, there are no randomized trials that have established the most effective cytotoxic chemotherapy, but smaller trials have shown. overall response rate of taxane (paclitaxel or docetaxel) plus platinum (carboplatin) chemotherapy ranging from 39% to 50% for SDC.91,92 Other chemotherapeutic agents have also been studied, including taxanes, platinum, cyclophosphamide, doxorubicin, gemcitabine, vinorelbine and eribulin; however, these study cohorts included other carcinoma subtypes, so the results cannot be simply applied to SDC.93–98
AR- and HER2-Targeted Therapy
Based on biomarker profile of SDC, treatments targeting AR and HER2 have been introduced. Small cohort studies have provided promising results for targeted therapy in SDC, including recent data from a larger cohorts comprising more than 35 patients with histological reviews from Japan and the Netherlands.99–102 These data have confirmed the efficacy of targeted therapy, and the latest NCCN guidelines include AR therapy and anti-HER2 therapy as therapeutic options for AR-positive and HER2-overexpressing tumors, respectively.56 The NCCN guidelines further recommend the evaluation of the AR and HER2 status for SDC and adenocarcinoma, not otherwise specified.56
Drugs used for androgen deprivation therapy include AR blockers (eg, bicalutamide) and luteinizing hormone-releasing hormone analogues (eg, leuprolide). The response rate for advanced or recurrent patients was shown to be 17% to 65%, and median overall survival was 17 to 44 months.99,101,103,104 AR-V7, an AR therapy–resistant splicing variant of AR in prostate cancer, is also expressed in 15% to 50% of SDC patients, but the relationship between the expression of AR-V7 and AR therapy resistance has not been proven in SDC.59,70
Protocols of HER2-targeted therapy for HER2-overexpressing SDC tumors are based on those for breast cancer treatment. Combination therapy of an anti-HER2 antibody (trastuzumab, ado-trastuzumab emtansine, or pertuzumab) with conventional taxane-based chemotherapy increased the response rates and time to disease progression. The response rates ranged from 67% to 100%, and the median progression-free survival was 3 to 18 months.54,55,100,105–108
Immune Checkpoint Inhibitor Therapy and Other New Treatment Options
In SDC, the predictive biomarkers for immune checkpoint inhibitors indicate a small potential for achieving therapeutic benefit.59 Most SDC cases are microsatellite-stable and have a low TMB.59,63,70,73,82 There are no consistent data on PD-L1 expression.59–64 A study of immune checkpoint inhibitor (pembrolizumab) treatment for advanced salivary gland carcinoma reported a 12% response rate, and another study of pembrolizumab included 1 SDC case in the cohort.109,110 As mentioned above, recent molecular analyses have revealed additional “druggable” genetic alterations, including PIK3CA, HRAS, BRAF, PTEN, NTRK, and RET. Basket trials targeting these molecules have shown therapeutic activity in some salivary gland tumors.65,111–114 It may therefore be worthwhile for recurrent and/or metastatic SDC patients to undergo molecular testing to identify pertinent targeted therapies and consider their suitability for enrollment in basket trials, such as the NCI-MATCH Trial.115
CONCLUSIONS
The cytologic and histopathologic diagnosis of primary salivary gland tumors is often a challenging issue for general pathologists because of their histomorphologic diversity, the plethora of histologic subtypes, and their low incidence (<1% of malignancies from all body sites).116 Recent research on SDC has improved our understanding of its biology and clinicopathologic characteristics, which has led to more accurate diagnosis of SDC and has resulted in new treatment strategies, although some are not yet globally available. Considering the aggressiveness of SDC, future basic and clinical studies will be useful for the development of more effective therapeutic strategies.
FUNDING SUPPORT
No specific funding was disclosed.
Footnotes
CONFLICT OF INTEREST DISCLOSURES
Lori J. Wirth is a consulting or advisory board member for Bayer, Blueprint, Cue Biopharma, Exelexis, Genetech, Lilly, Loxo Oncology, Merck, and Rakuten Medical. The other authors made no disclosures.
REFERENCES
- 1.El-Naggar AK, Chan JKC, Grandis JR, Takata T, Slootweg PJ, eds. WHO Classification of Head and Neck Tumours. International Agency for Research on Cancer; 2017. [Google Scholar]
- 2.Nagao T, Licitra L, Loening T, Vielh P, Williams MD. Salivary duct carcinoma In: El-Naggar AK, Chan JKC, Grandis JR, Takata T, Slootweg PJ, eds. WHO Classification of Head and Neck Tumours. International Agency for Research on Cancer; 2017;173–174. [Google Scholar]
- 3.Gilbert MR, Sharma A, Schmitt NC, et al. A 20-year review of 75 cases of salivary duct carcinoma. JAMA Otolaryngol Head Neck Surg. 2016;142:489–495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Müller S, Mantsopoulos K, Iro H, Agaimy A. Salivary duct carcinoma of the sinonasal cavity: a case report and review of the literature. Head Neck. 2016;38:E2464–E2466. [DOI] [PubMed] [Google Scholar]
- 5.Rahimi S, Lambiase A, Brennan PA, Abdolrahimzadeh S. An androgen receptor-positive carcinoma of the lacrimal drainage system resembling salivary duct carcinoma: case report and review of the literature. Appl Immunohistochem Mol Morphol. 2016;24:e69–e71. [DOI] [PubMed] [Google Scholar]
- 6.Weon YC, Park SW, Kim HJ, et al. Salivary duct carcinomas: clinical and CT and MR imaging features in 20 patients. Neuroradiology. 2012;54:631–640. [DOI] [PubMed] [Google Scholar]
- 7.Wu WL, Wang CL, Li D, Luo J, Ye JY, Xu SS. Multiphase contrast-enhanced computed tomography imaging features of salivary duct carcinoma: differentiation from other salivary gland malignancies. Oral Surg Oral Med Oral Pathol Oral Radiol. 2019;128:543–551. [DOI] [PubMed] [Google Scholar]
- 8.Lee YY, Wong KT, King AD, Ahuja AT. Imaging of salivary gland tumours. Eur J Radiol. 2008;66:419–436. [DOI] [PubMed] [Google Scholar]
- 9.Liu Y, Li J, Tan YR, Xiong P, Zhong LP. Accuracy of diagnosis of salivary gland tumors with the use of ultrasonography, computed tomography, and magnetic resonance imaging: a meta-analysis. Oral Surg Oral Med Oral Pathol Oral Radiol. 2015;119:238–245.e232. [DOI] [PubMed] [Google Scholar]
- 10.Nakaguro M, Sato Y, Tada Y, et al. Prognostic implication of histopathologic indicators in salivary duct carcinoma: proposal of a novel histologic risk stratification model. Am J Surg Pathol. 2020;44:526–535. [DOI] [PubMed] [Google Scholar]
- 11.Simpson RHW, Prasad AR, Lewis JE, Skálová A, David L. Mucin-rich variant of salivary duct carcinoma: a clinicopathologic and immunohistochemical study of four cases. Am J Surg Pathol. 2003;27: 1070–1079. [DOI] [PubMed] [Google Scholar]
- 12.Nagao T, Gaffey TA, Serizawa H, et al. Sarcomatoid variant of salivary duct carcinoma: clinicopathologic and immunohistochemical study of eight cases with review of the literature. Am J Clin Pathol. 2004;122:222–231. [DOI] [PubMed] [Google Scholar]
- 13.Nagao T, Gaffey TA, Visscher DW, et al. Invasive micropapillary salivary duct carcinoma: a distinct histologic variant with biologic significance. Am J Surg Pathol. 2004;28:319–326. [DOI] [PubMed] [Google Scholar]
- 14.Sekhri R, Ortanca I, Boals C, Agarwal S. Salivary duct carcinoma: a case report of oncocytic variant with possible treatment implications and review of literature. Pathol Res Pract. 2019;215:152549. [DOI] [PubMed] [Google Scholar]
- 15.Kusafuka K, Kawasaki T, Maeda M, et al. Salivary duct carcinoma with rhabdoid features: a salivary counterpart of pleomorphic lobular carcinoma of the breast. Histopathology. 2017;70:164–173. [DOI] [PubMed] [Google Scholar]
- 16.Lewis JE, McKinney BC, Weiland LH, Ferreiro JA, Olsen KD. Salivary duct carcinoma. Clinicopathologic and immunohistochemical review of 26 cases. Cancer. 1996;77:223–230. [DOI] [PubMed] [Google Scholar]
- 17.Lim CM, Hobson C, Kim S, Johnson JT. Clinical outcome of patients with carcinoma ex pleomorphic adenoma of the parotid gland: a comparative study from a single tertiary center. Head Neck. 2015;37:543–547. [DOI] [PubMed] [Google Scholar]
- 18.Boon E, Bel M, van Boxtel W, et al. A clinicopathological study and prognostic factor analysis of 177 salivary duct carcinoma patients from The Netherlands. Int J Cancer. 2018;143:758–766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Rito M, Fonseca I. Carcinoma ex-pleomorphic adenoma of the salivary glands has a high risk of progression when the tumor invades more than 2.5 mm beyond the capsule of the residual pleomorphic adenoma. Virchows Arch. 2016;468:297–303. [DOI] [PubMed] [Google Scholar]
- 20.Griffith CC, Thompson LDR, Assaad A, et al. Salivary duct carcinoma and the concept of early carcinoma ex pleomorphic adenoma. Histopathology. 2014;65:854–860. [DOI] [PubMed] [Google Scholar]
- 21.Otsuka K, Imanishi Y, Tada Y, et al. Clinical outcomes and prognostic factors for salivary duct carcinoma: a multi-institutional analysis of 141 patients. Ann Surg Oncol. 2016;23:2038–2045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Williams L, Thompson LDR, Seethala RR, et al. Salivary duct carcinoma: the predominance of apocrine morphology, prevalence of histologic variants, and androgen receptor expression. Am J Surg Pathol. 2015;39:705–713. [DOI] [PubMed] [Google Scholar]
- 23.Jaehne M, Roeser K, Jaekel T, Schepers JD, Albert N, Löning T. Clinical and immunohistologic typing of salivary duct carcinoma: a report of 50 cases. Cancer. 2005;103:2526–2533. [DOI] [PubMed] [Google Scholar]
- 24.Han MW, Roh J-L, Choi S-H, et al. Prognostic factors and outcome analysis of salivary duct carcinoma. Auris Nasus Larynx. 2015;42:472–477. [DOI] [PubMed] [Google Scholar]
- 25.Simpson RHW, Desai S, Di Palma S. Salivary duct carcinoma in situ of the parotid gland. Histopathology. 2008;53:416–425. [DOI] [PubMed] [Google Scholar]
- 26.Simpson RHW. Salivary duct carcinoma: new developments—morphological variants including pure in situ high grade lesions; proposed molecular classification. Head Neck Pathol. 2013;7(suppl 1):S48–S58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Faquin WC, Rossi ED, Baloch Z, et al. The Milan System for Reporting Salivary Gland Cytopathology. Springer; 2018. [Google Scholar]
- 28.Rossi ED, Faquin WC. The Milan System for Reporting Salivary Gland Cytopathology (MSRSGC): an international effort toward improved patient care—when the roots might be inspired by Leonardo da Vinci. Cancer Cytopathol. 2018;126:756–766. [DOI] [PubMed] [Google Scholar]
- 29.Klijanienko J, Vielh P. Cytologic characteristics and histomorphologic correlations of 21 salivary duct carcinomas. Diagn Cytopathol. 1998;19:333–337. [DOI] [PubMed] [Google Scholar]
- 30.Gilcrease MZ, Guzman-Paz M, Froberg K, Pambuccian S. Salivary duct carcinoma. Is a specific diagnosis possible by fine needle aspiration cytology? Acta Cytol. 1998;42:1389–1396. [DOI] [PubMed] [Google Scholar]
- 31.Klijanienko J, El-Naggar AK, Vielh P. Fine-needle sampling findings in 26 carcinoma ex pleomorphic adenomas: diagnostic pitfalls and clinical considerations. Diagn Cytopathol. 1999;21:163–166. [DOI] [PubMed] [Google Scholar]
- 32.Okano K, Ishida M, Sandoh K, Fujisawa T, Iwai H, Tsuta K. Cytological features of carcinoma ex pleomorphic adenoma of the salivary glands: a diagnostic challenge. Diagn Cytopathol. 2020;48: 149–153. [DOI] [PubMed] [Google Scholar]
- 33.Nouraei SAR, Hope KL, Kelly CG, McLean NR, Soames JV. Carcinoma ex benign pleomorphic adenoma of the parotid gland. Plast Reconstr Surg. 2005;116:1206–1213. [DOI] [PubMed] [Google Scholar]
- 34.Moriki T, Ueta S, Takahashi T, Mitani M, Ichien M. Salivary duct carcinoma: cytologic characteristics and application of androgen receptor immunostaining for diagnosis. Cancer. 2001;93:344–350. [DOI] [PubMed] [Google Scholar]
- 35.Jo VY, Krane JF. Ancillary testing in salivary gland cytology: A practical guide. Cancer Cytopathol. 2018;126(suppl 8):627–642. [DOI] [PubMed] [Google Scholar]
- 36.Kawahara A, Harada H, Akiba J, Kage M. Salivary duct carcinoma cytologically diagnosed distinctly from salivary gland carcinomas with squamous differentiation. Diagn Cytopathol. 2008;36:485–493. [DOI] [PubMed] [Google Scholar]
- 37.Xu B, Dogan S, Haroon Al Rasheed MR, Ghossein R, Katabi N. Androgen receptor immunohistochemistry in salivary duct carcinoma: a retrospective study of 188 cases focusing on tumoral heterogeneity and temporal concordance. Hum Pathol. 2019;93:30–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Schwartz LE, Begum S, Westra WH, Bishop JA. GATA3 immunohistochemical expression in salivary gland neoplasms. Head Neck Pathol. 2013;7:311–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kapadia SB, Barnes L. Expression of androgen receptor, gross cystic disease fluid protein, and CD44 in salivary duct carcinoma. Mod Pathol. 1998;11:1033–1038. [PubMed] [Google Scholar]
- 40.Can NT, Lingen MW, Mashek H, et al. Expression of hormone receptors and HER-2 in benign and malignant salivary gland tumors. Head Neck Pathol. 2018;12:95–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Di Palma S, Simpson RH, Marchio C, et al. Salivary duct carcinomas can be classified into luminal androgen receptor-positive, HER2 and basal-like phenotypes. Histopathology. 2012;61:629–643. [DOI] [PubMed] [Google Scholar]
- 42.Takase S, Kano S, Tada Y, et al. Biomarker immunoprofile in salivary duct carcinomas: clinicopathological and prognostic implications with evaluation of the revised classification. Oncotarget. 2017;8:59023–59035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Luk PP, Weston JD, Yu B, et al. Salivary duct carcinoma: clinicopathologic features, morphologic spectrum, and somatic mutations. Head Neck. 2016;38(suppl 1):E1838–E1847. [DOI] [PubMed] [Google Scholar]
- 44.Villepelet A, Lefèvre M, Verillaud B, et al. Salivary duct carcinoma: prospective multicenter study of 61 cases of the Réseau d’Expertise Français des Cancers ORL Rares. Head Neck. 2019;41:584–591. [DOI] [PubMed] [Google Scholar]
- 45.Fan CY, Wang J, Barnes EL. Expression of androgen receptor and prostatic specific markers in salivary duct carcinoma: an immunohistochemical analysis of 13 cases and review of the literature. Am J Surg Pathol. 2000;24:579–586. [DOI] [PubMed] [Google Scholar]
- 46.Jalaly JB, Sanati S, Chernock RD, Dibe DG, El-Mofty SK. Salivary duct carcinoma and invasive ductal carcinoma of the breast: a comparative immunohistochemical study. Head Neck Pathol. 2018;12: 488–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Santana T, Pavel A, Martinek P, et al. Biomarker immunoprofile and molecular characteristics in salivary duct carcinoma: clinicopathological and prognostic implications. Hum Pathol. 2019;93:37–47. [DOI] [PubMed] [Google Scholar]
- 48.Butler RT, Spector ME, Thomas D, McDaniel AS, McHugh JB. An immunohistochemical panel for reliable differentiation of salivary duct carcinoma and mucoepidermoid carcinoma. Head Neck Pathol. 2014;8:133–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Thompson L, Bishop J. Head and Neck Pathology. 3rd ed Elsevier; 2018. [Google Scholar]
- 50.Elebro K, Bendahl PO, Jernström H, Borgquist S. Androgen receptor expression and breast cancer mortality in a population-based prospective cohort. Breast Cancer Res Treat. 2017;165:645–657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Glisson B, Colevas AD, Haddad R, et al. HER2 expression in salivary gland carcinomas: dependence on histological subtype. Clin Cancer Res. 2004;10:944–946. [DOI] [PubMed] [Google Scholar]
- 52.Wick MR, Ockner DM, Mills SE, Ritter JH, Swanson PE. Homologous carcinomas of the breasts, skin, and salivary glands. A histologic and immunohistochemical comparison of ductal mammary carcinoma, ductal sweat gland carcinoma, and salivary duct carcinoma. Am J Clin Pathol. 1998;109:75–84. [DOI] [PubMed] [Google Scholar]
- 53.Saintigny P, Mitani Y, Pytynia KB, et al. Frequent PTEN loss and differential HER2/PI3K signaling pathway alterations in salivary duct carcinoma: implications for targeted therapy. Cancer. 2018;124:3693–3705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hainsworth JD, Meric-Bernstam F, Swanton C, et al. Targeted therapy for advanced solid tumors on the basis of molecular profiles: results from MyPathway, an open-label, phase IIa multiple basket study. J Clin Oncol. 2018;36:536–542. [DOI] [PubMed] [Google Scholar]
- 55.Li BT SR, Offin M, Buonocore D, Myers ML, Venkatesh A, Razavi P. Ado-trastuzumab emtansine in patients with HER2 amplified salivary gland cancers (SGCs): results from a phase II basket trial [abstract]. J Clin Oncol. 2019;37(suppl):6001. [Google Scholar]
- 56.National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology, head and neck cancers. Accessed February 14, 2020 https://www.nccn.org/professionals/physician_gls/pdf/head-and-neck.pdf. [DOI] [PubMed]
- 57.Griffith CC, Seethala RR, Luvison A, Miller M, Chiosea SI. PIK3CA mutations and PTEN loss in salivary duct carcinomas. Am J Surg Pathol. 2013;37:1201–1207. [DOI] [PubMed] [Google Scholar]
- 58.Urano M, Hirai H, Tada Y, et al. The high expression of FOXA1 is correlated with a favourable prognosis in salivary duct carcinomas: a study of 142 cases. Histopathology. 2018;73:943–952. [DOI] [PubMed] [Google Scholar]
- 59.Gargano SM, Senarathne W, Feldman R, et al. Novel therapeutic targets in salivary duct carcinoma uncovered by comprehensive molecular profiling. Cancer Med. 2019;8:7322–7329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Vital D, Ikenberg K, Moch H, Rössle M, Huber GF. The expression of PD-L1 in salivary gland carcinomas. Sci Rep. 2019;9:12724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Xu B, Jungbluth AA, Frosina D, et al. The immune microenvironment and expression of PD-L1, PD-1, PRAME and MHC I in salivary duct carcinoma. Histopathology. 2019;75:672–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hamza A, Roberts D, Su S, Weber RS, Bell D, Ferrarotto R. PD-L1 expression by immunohistochemistry in salivary duct carcinoma. Ann Diagn Pathol. 2019;40:49–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Nakano T, Takizawa K, Uezato A, Taguchi K, Toh S, Masuda M. Prognostic value of programed death ligand-1 and ligand-2 co-expression in salivary gland carcinomas. Oral Oncol. 2019;90:30–37. [DOI] [PubMed] [Google Scholar]
- 64.Sato F, Akiba J, Kawahara A, et al. The expression of programed death ligand-1 could be related with unfavorable prognosis in salivary duct carcinoma. J Oral Pathol Med. 2018;47:683–690. [DOI] [PubMed] [Google Scholar]
- 65.Wang K, Russell JS, McDermott JD, et al. Profiling of 149 salivary duct carcinomas, carcinoma ex pleomorphic adenomas, and adenocarcinomas, not otherwise specified reveals actionable genomic alterations. Clin Cancer Res. 2016;22:6061–6068. [DOI] [PubMed] [Google Scholar]
- 66.Nardi V, Sadow PM, Juric D, et al. Detection of novel actionable genetic changes in salivary duct carcinoma helps direct patient treatment. Clin Cancer Res. 2013;19:480–490. [DOI] [PubMed] [Google Scholar]
- 67.Shimura T, Tada Y, Hirai 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ku BM, Jung HA, Sun JM, et al. High-throughput profiling identifies clinically actionable mutations in salivary duct carcinoma. J Transl Med. 2014;12:299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Grunewald I, Vollbrecht C, Meinrath J, et al. Targeted next generation sequencing of parotid gland cancer uncovers genetic heterogeneity. Oncotarget. 2015;6:18224–18237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Dalin MG, Desrichard A, Katabi N, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Khoo TK, Yu B, Smith JA, et al. Somatic mutations in salivary duct carcinoma and potential therapeutic targets. Oncotarget. 2017;8: 75893–75903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Dogan S, Ng CKY, Xu B, et al. The repertoire of genetic alterations in salivary duct carcinoma including a novel HNRNPH3-ALK rearrangement. Hum Pathol. 2019;88:66–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ross JS, Gay LM, Wang K, et al. Comprehensive genomic profiles of metastatic and relapsed salivary gland carcinomas are associated with tumor type and reveal new routes to targeted therapies. Ann Oncol. 2017;28:2539–2546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Grünewald I, Trautmann M, Busch A, et al. MDM2 and CDK4 amplifications are rare events in salivary duct carcinomas. Oncotarget. 2016;7:75261–75272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Urano M, Nakaguro M, Yamamoto Y, et al. Diagnostic significance of HRAS mutations in epithelial-myoepithelial carcinomas exhibiting a broad histopathologic spectrum. Am J Surg Pathol. 2019;43:984–994. [DOI] [PubMed] [Google Scholar]
- 76.Jo VY, Sholl LM, Krane JF. Distinctive patterns of CTNNB1 (β-catenin) alterations in salivary gland basal cell adenoma and basal cell adenocarcinoma. Am J Surg Pathol. 2016;40:1143–1150. [DOI] [PubMed] [Google Scholar]
- 77.Kawahara A, Harada H, Abe H, et al. Nuclear β-catenin expression in basal cell adenomas of salivary gland. J Oral Pathol Med. 2011;40:460–466. [DOI] [PubMed] [Google Scholar]
- 78.Sato M, Yamamoto H, Hatanaka Y, et al. Wnt/β-catenin signal alteration and its diagnostic utility in basal cell adenoma and histologically similar tumors of the salivary gland. Pathol Res Pract. 2018;214:586–592. [DOI] [PubMed] [Google Scholar]
- 79.Nakaguro M, Urano M, Ogawa I, et al. Histopathological evaluation of minor salivary gland papillary-cystic tumours: focus on genetic alterations in sialadenoma papilliferum and intraductal papillary mucinous neoplasm. Histopathology. 2020;76:411–422. [DOI] [PubMed] [Google Scholar]
- 80.Skálová A, Vanecek T, Sima R, et al. Mammary analogue secretory carcinoma of salivary glands, containing the ETV6-NTRK3 fusion gene: a hitherto undescribed salivary gland tumor entity. Am J Surg Pathol. 2010;34:599–608. [DOI] [PubMed] [Google Scholar]
- 81.Skálová A, Vanecek T, Uro-Coste E, et al. Molecular profiling of salivary gland intraductal carcinoma revealed a subset of tumors harboring NCOA4-RET and novel TRIM27-RET fusions: a report of 17 cases. Am J Surg Pathol. 2018;42:1445–1455. [DOI] [PubMed] [Google Scholar]
- 82.Linxweiler M, Kuo F, Katabi N, et al. The immune microenvironment and neoantigen landscape of aggressive salivary gland carcinomas differ by subtype. Clin Cancer Res. Published online February 14, 2020. doi: 10.1158/1078-0432.CCR-19-3758 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.D’Heygere E, Meulemans J, Vander Poorten V. Salivary duct carcinoma. Curr Opin Otolaryngol Head Neck Surg. 2018;26:142–151. [DOI] [PubMed] [Google Scholar]
- 84.Lassche G, van Boxtel W, Ligtenberg MJL, van Engen-van Grunsven ACH, van Herpen CML. Advances and challenges in precision medicine in salivary gland cancer. Cancer Treat Rev. 2019;80:101906. [DOI] [PubMed] [Google Scholar]
- 85.Schmitt NC, Kang H, Sharma A. Salivary duct carcinoma: an aggressive salivary gland malignancy with opportunities for targeted therapy. Oral Oncol. 2017;74:40–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Alfieri S, Granata R, Bergamini C, et al. Systemic therapy in metastatic salivary gland carcinomas: a pathology-driven paradigm? Oral Oncol. 2017;66:58–63. [DOI] [PubMed] [Google Scholar]
- 87.Schvartsman G, Pinto NA, Bell D, Ferrarotto R. Salivary gland tumors: Molecular characterization and therapeutic advances for metastatic disease. Head Neck. 2019;41:239–247. [DOI] [PubMed] [Google Scholar]
- 88.Lewis AG, Tong T, Maghami E. Diagnosis and management of malignant salivary gland tumors of the parotid gland. Otolaryngol Clin North Am. 2016;49:343–380. [DOI] [PubMed] [Google Scholar]
- 89.Sood S, McGurk M, Vaz F. Management of salivary gland tumours: united kingdom national multidisciplinary guidelines. J Laryngol Otol. 2016;130:S142–S149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Mahmood U, Koshy M, Goloubeva O, Suntharalingam M. Adjuvant radiation therapy for high-grade and/or locally advanced major salivary gland tumors. Arch Otolaryngol Head Neck Surg. 2011;137:1025–1030. [DOI] [PubMed] [Google Scholar]
- 91.Nakano K, Sato Y, Sasaki T, et al. Combination chemotherapy of carboplatin and paclitaxel for advanced/metastatic salivary gland carcinoma patients: differences in responses by different pathological diagnoses. Acta Otolaryngol. 2016;136:948–951. [DOI] [PubMed] [Google Scholar]
- 92.Okada T, Saotome T, Nagao T, et al. Carboplatin and docetaxel in patients with salivary gland carcinoma: a retrospective study. In Vivo. 2019;33:843–853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Licitra L, Cavina R, Grandi C, et al. Cisplatin, doxorubicin and cyclophosphamide in advanced salivary gland carcinoma. A phase II trial of 22 patients. Ann Oncol. 1996;7:640–642. [DOI] [PubMed] [Google Scholar]
- 94.Laurie SA, Siu LL, Winquist E, et al. 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. [DOI] [PubMed] [Google Scholar]
- 95.Airoldi M, Garzaro M, Pedani F, 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. [DOI] [PubMed] [Google Scholar]
- 96.Licitra L, Marchini S, Spinazzè S, et al. Cisplatin in advanced salivary gland carcinoma. A phase II study of 25 patients. Cancer. 1991;68:1874–1877. [DOI] [PubMed] [Google Scholar]
- 97.Gilbert J, Li Y, Pinto HA, et al. Phase II trial of taxol in salivary gland malignancies (E1394): a trial of the Eastern Cooperative Oncology Group. Head Neck. 2006;28:197–204. [DOI] [PubMed] [Google Scholar]
- 98.Debaere D, Vander Poorten V, Nuyts S, et al. Cyclophosphamide, doxorubicin, and cisplatin in advanced salivary gland cancer. B-ENT. 2011;7:1–6. [PubMed] [Google Scholar]
- 99.Fushimi C, Tada Y, Takahashi 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Takahashi H, Tada Y, Saotome T, 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. [DOI] [PubMed] [Google Scholar]
- 101.Boon E, van Boxtel W, Buter J, et al. Androgen deprivation therapy for androgen receptor-positive advanced salivary duct carcinoma: a nationwide case series of 35 patients in The Netherlands. Head Neck. 2018;40:605–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.van Boxtel W, Locati LD, van Engen-van Grunsven ACH, et al. Adjuvant androgen deprivation therapy for poor-risk, androgen receptor-positive salivary duct carcinoma. Eur J Cancer. 2019;110:62–70. [DOI] [PubMed] [Google Scholar]
- 103.Locati LD, Perrone F, Cortelazzi B, et al. Clinical activity of androgen deprivation therapy in patients with metastatic/relapsed androgen receptor-positive salivary gland cancers. Head Neck. 2016;38:724–731. [DOI] [PubMed] [Google Scholar]
- 104.Viscuse PV, Price KA, Garcia JJ, Schembri-Wismayer DJ, Chintakuntlawar AV. First line androgen deprivation therapy vs. chemotherapy for patients with androgen receptor positive recurrent or metastatic salivary gland carcinoma—a retrospective study. Front Oncol. 2019;9:701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Limaye SA, Posner MR, Krane JF, et al. Trastuzumab for the treatment of salivary duct carcinoma. Oncologist. 2013;18:294–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.De Block K, Vander Poorten V, Dormaar T, et al. Metastatic HER-2-positive salivary gland carcinoma treated with trastuzumab and a taxane: a series of six patients. Acta Clin Belg. 2016;71:383–388. [DOI] [PubMed] [Google Scholar]
- 107.Perissinotti AJ, Lee Pierce M, Pace MB, El-Naggar A, Kies MS, Kupferman M. The role of trastuzumab in the management of salivary ductal carcinomas. Anticancer Res. 2013;33:2587–2591. [PubMed] [Google Scholar]
- 108.Jhaveri KL, Wang XV, Makker V, et al. Ado-trastuzumab emtansine (T-DM1) in patients with HER2-amplified tumors excluding breast and gastric/gastroesophageal junction (GEJ) adenocarcinomas: results from the NCI-MATCH trial (EAY131) subprotocol Q. Ann Oncol. 2019;30:1821–1830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Cohen RB, Delord J-P, Doi T, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Rodriguez CP, Wu Q, Voutsinas J, 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. [DOI] [PubMed] [Google Scholar]
- 111.Hyman DM, Puzanov I, Subbiah V, et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N Engl J Med. 2015;373:726–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Klempner SJ, Bordoni R, Gowen K, et al. Identification of BRAF kinase domain duplications across multiple tumor types and response to RAF inhibitor therapy. JAMA Oncol. 2016;2:272–274. [DOI] [PubMed] [Google Scholar]
- 113.Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020;21:271–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Kurzrock R, Bowles DW, Kang H, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.National Cancer Institute. NCI-MATCH trial (molecular analysis for therapy choice). Accessed January 15, 2020 https://www.cancer.gov/about-cancer/treatment/clinical-trials/nci-supported/nci-match. [DOI] [PMC free article] [PubMed]
- 116.Ellis GL, Auclair PL. Armed Forces Institute of Pathology Universities Associated for Research Education in Pathology: Tumors of the Salivary Glands. American Registry of Pathology Press; 1996. [Google Scholar]
