Next Article in Journal
Photodynamic Synergistic Effect of Pheophorbide a and Doxorubicin in Combined Treatment against Tumoral Cells
Next Article in Special Issue
Androgen Receptor Signaling in Bladder Cancer
Previous Article in Journal
Epidermal Growth Factor Pathway Signaling in Drosophila Embryogenesis: Tools for Understanding Cancer
Previous Article in Special Issue
A Tale of Two Signals: AR and WNT in Development and Tumorigenesis of Prostate and Mammary Gland
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Androgen Receptor Signaling in Salivary Gland Cancer

1
Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
2
Department of Pediatrics, Institution for Clinical Sciences, University of Gothenburg, Gothenburg SE-416 86, Sweden
3
Head and Neck Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
4
Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
*
Authors to whom correspondence should be addressed.
Cancers 2017, 9(2), 17; https://doi.org/10.3390/cancers9020017
Submission received: 22 December 2016 / Revised: 28 January 2017 / Accepted: 3 February 2017 / Published: 8 February 2017
(This article belongs to the Special Issue AR Signaling in Human Malignancies: Prostate Cancer and Beyond)

Abstract

:
Salivary gland cancers comprise a small subset of human malignancies, and are classified into multiple subtypes that exhibit diverse histology, molecular biology and clinical presentation. Local disease is potentially curable with surgery, which may be combined with adjuvant radiotherapy. However, metastatic or unresectable tumors rarely respond to chemotherapy and carry a poorer prognosis. Recent molecular studies have shown evidence of androgen receptor signaling in several types of salivary gland cancer, mainly salivary duct carcinoma. Successful treatment with anti-androgen therapy in other androgen receptor-positive malignancies such as prostate and breast cancer has inspired researchers to investigate this treatment in salivary gland cancer as well. In this review, we describe the prevalence, biology, and therapeutic implications of androgen receptor signaling in salivary gland cancer.

1. Introduction

Salivary gland cancers (SGCs) are a group of uncommon, heterogeneous tumors that account for 0.3% of all malignancies and 6% of head and neck cancers in the United States [1]. The majority of SGCs are found in the parotid gland (59%–81% of cases), but they also arise in the submandibular gland (6%–21%), or in minor salivary glands (7%–22%) that populate the upper aerodigestive tract [2,3,4]. The World Health Organization classifies 24 subtypes of SGC, which show significant variation in histological and clinical features [1]. SGC is generally treated with surgery and, in selected cases, adjuvant radiotherapy (RT) [5]. Systemic therapy has not been adequately tested in many SGC subtypes, and cytotoxic chemotherapy has shown a limited effect in SGCs in general. As a consequence, the prognosis of recurrent or metastatic SGC can be poor [2,6,7]. However, recent studies have investigated the molecular landscape of several types of SGCs, such as adenoid cystic carcinoma (ACC), mucoepidermoid carcinoma (MEC), polymorphous low grade adenocarcinoma (PLGA), secretory carcinoma and salivary duct carcinoma (SDC), and uncovered molecular targets of interest in selected patients [8,9,10,11,12,13].
The androgen receptor (AR) is a nuclear steroid hormone receptor that is physiologically expressed at low levels in many human tissues [14]. Its main ligands are testosterone and 5α-dihydrotestosterone (DHT). AR regulates the transcription of multiple effector genes through direct DNA binding or interaction with other transcription factors, leading to increased cell growth, differentiation, and survival [15]. Overactive AR signaling is an important oncogenic driver in several tumor types, including prostate cancer and a subset of breast cancers [16,17]. Androgen-deprivation therapy (ADT) has been used in patients with prostate cancer since the 1940s [18], and has more recently gained interest in a growing number of malignancies [17,19,20,21]. ADT may be achieved by direct inhibition of AR (known as anti-androgen therapy), or by downregulating the gonadotropin-releasing hormone (GnRH) receptor signaling output, which leads to reduced serum testosterone levels (known as chemical castration). These two methods are often combined to achieve what has been termed maximum or complete androgen blockade [22].

2. AR Expression in SGC

Nuclear AR expression based on immunohistochemistry (IHC) is the most widely used marker of active AR signaling, and correlates with the response to ADT in prostate cancer [23]. The prevalence of AR expression varies substantially between different subtypes of SGC (see Table 1 for a summary of published IHC data). AR overexpression is most frequently associated with salivary duct carcinomas (SDC), the majority of which are positive for AR. Several studies have shown AR immunoreactivity in 64%–77% of cases [8,24,25,26,27,28,29,30], whereas a recent large report detected AR expression in as many as 98% of SDCs [31]. In that study, several tumors initially diagnosed as AR-negative SDCs were reclassified as other tumor entities after a second evaluation by salivary pathologists. Also, for tumors with conventional SDC morphology and a negative first AR IHC, the staining was repeated and showed AR expression the second time in several cases. This may suggest that the prevalence of AR-positive SDC was previously underestimated due to technical issues or diagnostic difficulties.
Our group recently identified AR positivity by IHC in 75% of SDCs, and RNA sequencing confirmed extremely low but detectable levels of AR mRNA in AR IHC–negative cases, all of which had typical SDC morphology at the time of pathologic re-evaluation [8]. Interestingly, three of four AR IHC-negative cases showed AR signaling activity at levels equivalent to AR IHC-positive cases, as measured by expression of AR-regulated genes. Both AR-negative and AR-positive SDCs showed global gene expression patterns highly similar to AR-positive (also termed molecular apocrine) breast cancers. This raises the possibility that some SDCs with low levels of AR may have acquired alternative mechanisms to activate AR signaling pathways. Furthermore, the remarkable biological similarity between the two cancer types may suggest that treatment results in patients with molecular apocrine breast cancer could be of interest for the design of clinical trials in SDC.
The prognostic relevance of AR expression in SDC is difficult to assess, due to the rarity of the disease and the low number of AR-negative cases. Some investigators have identified a trend suggestive of better disease-free survival in AR-positive compared to AR-negative SDC patients [26,29], but this association has not been identified by other groups [8,24,25]. Similarly, one study detected a higher prevalence of AR expression in men than in women with SDC [30], a finding that has not been replicated in other reports [8,26].
In other subtypes of SGC, nuclear AR expression is found at lower rates. Adenocarcinoma, not otherwise specified (AC NOS) and acinic cell carcinoma (AcCC) are AR-positive in 26% and 15% of the cases, respectively [28,32,33,34,35]. On the other hand, only a small subset of MEC and ACC have detectable expression of AR [27,28,32,33,34,36,37], and some of these cases show weak AR expression (5%–15% stained nuclei) which may not be relevant for the biology of the tumors [32]. Among the rare types of SGC, AR expression has been reported in PLGA and basal cell adenocarcinoma (BCAC) [28,32], whereas all published cases of myoepithelial carcinoma (MECA) have been AR-negative [28,33]. Five cases of AR-positive epithelial-myoepithelial carcinoma (EMC) were reported and suggested to represent a specific variant of the disease, denoted apocrine EMC [38]. However, one study of six unselected EMCs did not detect AR [28], and the prevalence of AR expression in EMC is unknown. Given the challenging nature of salivary gland pathology, it is possible that some of these AR-positive entities in fact represent SDC.
A subset of SGCs result from the malignant transformation of a pre-existing pleomorphic adenoma (PA). PA is the most prevalent salivary gland tumor, and is typically benign and non-metastatic. Around 6% of PAs develop into different types of carcinoma, denoted carcinoma ex-PA [39]. Whereas PAs are AR-positive in 30% of the cases, 90% of carcinoma ex-PAs express AR. This difference may suggest that AR expression is a risk factor for the malignant transformation of PAs. Alternatively, overexpression of AR may act as an oncogenic event in some carcinomas ex-PA [40].

3. Expression of AR Splice Variants

The full-length AR (AR-FL) gene consists of eight exons, of which exons 4–8 encode the ligand-binding domain. Expression of alternative AR isoforms lacking the ligand-binding domain (which normally serves as a binding site for anti-androgens, such as enzalutamide) is associated with ADT resistance in prostate cancer [46,47,48,49,50]. AR-V7, a constitutively active AR splice variant that includes only exons 1–3 and a cryptic exon 3, is detected in 37%–50% of SDCs (Figure 1) [8,26]. On average, AR-V7 is expressed at around 5% of AR-FL RNA levels [8], which is similar to the AR-V7/AR-FL ratio seen in prostate cancer [16]. Another AR isoform, AR-V3, including only exons 1, 2 and a cryptic exon 2, is also found in SDC but at lower rates and only in male patients [26]. AR-45, which lacks the majority of exon 1, including the N-terminal domain that mediates ligand-independent transactivation of AR [51], is detected in a minority of SDCs [26]. However, the association between the alternative AR isoforms and response to ADT in SDC, and the prevalence of AR-V7, AR-V3, and AR-45 in other types of SGC, remains unknown.

4. Genetic Alterations Affecting AR Signaling

An extra copy of chromosome X, which includes the AR gene, is found in almost 40% of SDCs. This may contribute to overexpression of AR, although some of the tumors with an extra chromosome X are negative for AR in IHC [26]. Unlike in prostate cancer, focal amplification or protein-altering somatic mutations of AR have not been found in SDC or ACC [8,9,26].
Forkhead box protein A1 (FOXA1) is a transcription factor that mediates the transcription of AR target genes by facilitating the AR/chromatin interaction [52]. FOXA1 mutations may potentially be associated with ADT resistance in prostate cancer, although this is being actively investigated [53]. In a recent exome sequencing study reported by our group, we identified alteration (either somatic mutations in the DNA-binding domain or high-level amplification) of FOXA1 in four of 12 AR-positive SDCs. Conversely, no FOXA1 alterations were found in four AR-negative SDCs [8].
Fatty acid synthase (FASN) is an enzyme that controls fatty acid synthesis and has been shown to promote the growth of prostate cancer as a result of AR signaling. Experimental studies suggest that FASN overexpression can mediate resistance to ADT in prostate cancer, although no clinical data are yet available [54]. In our exome study of SDC, alterations (missense mutations, a frameshift insertion, and high-level amplification) of FASN were found in four of 12 AR-positive but not in AR-negative tumors [8].
In ACC, which rarely expresses AR, no significant genetic alterations affecting AR signaling have been detected [9]. In other subtypes of SGC, the prevalence of AR-related genetic alterations is unknown.

5. Anti-Androgen Therapy in Patients with SGC

Several ADT drugs have been developed and tested clinically, mainly in patients with prostate cancer. Abiraterone is a CYP17A1 inhibitor which reduces circulating levels of androgen by ultimately blocking the conversion of pregnenolone to DHT. Bicalutamide and flutamide are competitive inhibitors of the AR ligand-binding domain, as is enzalutamide, which was developed more recently and has greater AR affinity compared to the earlier anti-androgens, and may inhibit AR activity via a variety of different mechanisms [55]. Triptorelin and goserelin are GnRH agonists which eventually cause downregulation of luteinizing hormone (LH) and thereby reduced serum testosterone levels [22].
Inspired by results from other cancers [17,56] and functional studies showing AR-dependency in cultured SGC cells [26,57], a number of patients with AR-positive SGC have been treated with different ADT regimens (see Table 2 for a summary of reported cases). In a retrospective analysis of 17 patients with recurrent or metastatic AR-positive SGC, of which the majority had SDC or AC NOS, the overall response rate was 65%. Treatment was generally well tolerated in these patients, both men and women. However, relapse was commonly seen, leading to a three-year progression-free survival (PFS) of 12%, and a five-year overall survival of 19% [58]. Smaller studies of AR-positive SDC patients have reported somewhat less favorable outcomes, with an overall ADT response rate of 25%–50% [8,59]. Several case reports have shown a good effect of ADT alone in patients with AR-positive SDC or AC NOS, including stable disease for several months as well as cases of complete remission [43,60,61]. A few patients with SDC or AC NOS, who initially responded to a combination of bicalutamide and triptorelin but had a relapse, then showed a response to subsequent abiraterone, suggesting resistance mediated by the reactivation of AR signaling during ADT treatment [62,63]. ADT has also been combined with either definitive RT or palliative chemotherapy with robust responses in several single case reports of SGC [64,65].
Patients with AC NOS have been found to respond well to ADT, with partial or complete response in 10 of 11 reported cases, and a median PFS of 20 months. SDC patients appear to have a lower response rate, with partial or complete response in 11 of 26 (42%) reported cases and a median PFS of eight months (Table 2).
Of note, several dramatic responses to ADT in SGC patients were published only as case reports of extraordinary responders. A recent preliminary study including all SDC patients treated with ADT in the Netherlands showed somewhat more modest results, with partial response in four (13%) cases, stable disease in 10 (32%) cases, and progressive disease in 17 (55%) cases, and a median PFS of 3.8 months [45]. On the other hand, since the majority of SGCs are chemotherapy-resistant, the treatment options for patients with generalized disease are limited and AR is the most promising target for these patients with otherwise incurable disease. Several clinical trials are currently ongoing, investigating the efficacy of ADT in patients with recurrent/metastatic AR-positive SGC, using abiraterone, bicalutamide or enzalutamide in male and female patients (NCT02749903, NCT01969578, NCT02867852). In addition to providing valuable clinical response information, these trials will also collect tumor tissue for correlative research, facilitating further understanding of molecular determinants of response to ADT in AR-positive SGC.

6. Conclusions

AR is expressed in a majority of SDCs and in a minority of other SGCs such as AC NOS, and ADT has emerged as a promising therapy in patients with AR-positive SGC. Several potential mechanisms of resistance to ADT have been described, including the expression of AR splice variants and mutations in FOXA1 and FASN. Ongoing and future clinical trials will likely shed light on the clinical benefit and limitations of ADT in AR-positive SGC.

Acknowledgments

Martin G. Dalin was supported by Sahlgrenska University Hospital, The Swedish Medical Society, and Svensson’s Fund for Medical Research. Luc G. T. Morris was supported by NIH K08 DE024774, the Society of MSKCC, the Damon Runyon Cancer Research Foundation, and the Jayme and Peter Flowers Fund.

Author Contributions

Manuscript concept: Martin G. Dalin, Luc G. T. Morris. Literature review: Martin G. Dalin, Philip A. Watson, Alan L. Ho, Luc G. T. Morris. Analysis of data: Martin G. Dalin, Luc G. T. Morris. Interpretation of data: Martin G. Dalin, Philip A. Watson, Alan L. Ho, Luc G. T. Morris. Graphic design: Martin G. Dalin. Preparation of manuscript: Martin G. Dalin, Philip A. Watson, Alan L. Ho, Luc G. T. Morris.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Eveson, J.W.; Auclair, P.; Gnepp, D.R.; El-Naggar, A.K. Tumors of the salivary glands. In Pathology and Genetics of Head and Neck Tumours: International Agency for Research on Cancer; World Health Organization: Geneva, Switzerland, 2005; pp. 210–281. [Google Scholar]
  2. Terhaard, C.H.; Lubsen, H.; Van der Tweel, I.; Hilgers, F.J.; Eijkenboom, W.M.; Marres, H.A.; Tjho-Heslinga, R.E.; de Jong, J.M.; Roodenburg, J.L. Salivary gland carcinoma: Independent prognostic factors for locoregional control, distant metastases, and overall survival: Results of the dutch head and neck oncology cooperative group. Head Neck 2004, 26, 681–693. [Google Scholar] [CrossRef] [PubMed]
  3. Renehan, A.; Gleave, E.N.; Hancock, B.D.; Smith, P.; McGurk, M. Long-term follow-up of over 1000 patients with salivary gland tumours treated in a single centre. Br. J. Surg. 1996, 83, 1750–1754. [Google Scholar] [CrossRef] [PubMed]
  4. Fitzpatrick, P.J.; Theriault, C. Malignant salivary gland tumors. Int. J. Radiat. Oncol. Biol. Phys. 1986, 12, 1743–1747. [Google Scholar] [CrossRef]
  5. Panwar, A.; Kozel, J.A.; Lydiatt, W.M. Cancers of major salivary glands. Surg. Oncol. Clin. N. Am. 2015, 24, 615–633. [Google Scholar] [CrossRef] [PubMed]
  6. Laurie, S.A.; Licitra, L. Systemic therapy in the palliative management of advanced salivary gland cancers. J. Clin. Oncol. 2006, 24, 2673–2678. [Google Scholar] [CrossRef] [PubMed]
  7. Licitra, L.; Grandi, C.; Prott, F.J.; Schornagel, J.H.; Bruzzi, P.; Molinari, R. Major and minor salivary glands tumours. Crit. Rev. Oncol. Hematol. 2003, 45, 215–225. [Google Scholar] [CrossRef]
  8. Dalin, M.G.; Desrichard, A.; Katabi, N.; Makarov, V.; Walsh, L.A.; Lee, K.W.; Wang, Q.; Armenia, J.; West, L.; Dogan, S.; et al. Comprehensive molecular characterization of salivary duct carcinoma reveals actionable targets and similarity to apocrine breast cancer. Clin. Cancer Res. 2016, 22, 4623–4633. [Google Scholar] [CrossRef] [PubMed]
  9. Ho, A.S.; Kannan, K.; Roy, D.M.; Morris, L.G.; Ganly, I.; Katabi, N.; Ramaswami, D.; Walsh, L.A.; Eng, S.; Huse, J.T.; et al. The mutational landscape of adenoid cystic carcinoma. Nat. Genet. 2013, 45, 791–798. [Google Scholar] [CrossRef] [PubMed]
  10. Ku, B.M.; Jung, H.A.; Sun, J.M.; Ko, Y.H.; Jeong, H.S.; Son, Y.I.; Baek, C.H.; Park, K.; Ahn, M.J. High-throughput profiling identifies clinically actionable mutations in salivary duct carcinoma. J. Trans. Med. 2014. [Google Scholar] [CrossRef] [PubMed]
  11. Weinreb, I.; Piscuoglio, S.; Martelotto, L.G.; Waggott, D.; Ng, C.K.; Perez-Ordonez, B.; Harding, N.J.; Alfaro, J.; Chu, K.C.; Viale, A.; et al. Hotspot activating PRKD1 somatic mutations in polymorphous low-grade adenocarcinomas of the salivary glands. Nat. Genet. 2014, 46, 1166–1169. [Google Scholar] [CrossRef] [PubMed]
  12. Kang, H.; Tan, M.; Bishop, J.A.; Jones, S.; Sausen, M.; Ha, P.K.; Agrawal, N. Whole-exome sequencing of salivary gland mucoepidermoid carcinoma. Clin. Cancer Res. 2017, 23, 283–288. [Google Scholar] [CrossRef] [PubMed]
  13. Skalova, A.; Vanecek, T.; Simpson, R.H.; Laco, J.; Majewska, H.; Baneckova, M.; Steiner, P.; Michal, M. Mammary analogue secretory carcinoma of salivary glands: Molecular analysis of 25 ETV6 gene rearranged tumors with lack of detection of classical ETV6-NTRK3 fusion transcript by standard RT-PCR: Report of 4 cases harboring ETV6-X gene fusion. Am. J. Surg. Pathol. 2016, 40, 3–13. [Google Scholar] [CrossRef] [PubMed]
  14. Ruizeveld de Winter, J.A.; Trapman, J.; Vermey, M.; Mulder, E.; Zegers, N.D.; van der Kwast, T.H. Androgen receptor expression in human tissues: An immunohistochemical study. J. Histochem. Cytochem. 1991, 39, 927–936. [Google Scholar] [CrossRef] [PubMed]
  15. Lu, N.Z.; Wardell, S.E.; Burnstein, K.L.; Defranco, D.; Fuller, P.J.; Giguere, V.; Hochberg, R.B.; McKay, L.; Renoir, J.M.; Weigel, N.L.; et al. International union of pharmacology. Lxv. The pharmacology and classification of the nuclear receptor superfamily: Glucocorticoid, mineralocorticoid, progesterone, and androgen receptors. Pharmacol. Rev. 2006, 58, 782–797. [Google Scholar] [CrossRef] [PubMed]
  16. Robinson, D.; Van Allen, E.M.; Wu, Y.M.; Schultz, N.; Lonigro, R.J.; Mosquera, J.M.; Montgomery, B.; Taplin, M.E.; Pritchard, C.C.; Attard, G.; et al. Integrative clinical genomics of advanced prostate cancer. Cell 2015, 161, 1215–1228. [Google Scholar] [CrossRef] [PubMed]
  17. Narayanan, R.; Dalton, J.T. Androgen receptor: A complex therapeutic target for breast cancer. Cancers 2016. [Google Scholar] [CrossRef] [PubMed]
  18. Perlmutter, M.A.; Lepor, H. Androgen deprivation therapy in the treatment of advanced prostate cancer. Rev. Urol. 2007, 9, S3–S8. [Google Scholar] [PubMed]
  19. Godoy, G.; Gakis, G.; Smith, C.L.; Fahmy, O. Effects of androgen and estrogen receptor signaling pathways on bladder cancer initiation and progression. Bladder Cancer 2016, 2, 127–137. [Google Scholar] [CrossRef] [PubMed]
  20. Di Lauro, L.; Barba, M.; Pizzuti, L.; Vici, P.; Sergi, D.; Di Benedetto, A.; Mottolese, M.; Speirs, V.; Santini, D.; De Maria, R.; et al. Androgen receptor and antiandrogen therapy in male breast cancer. Cancer lett. 2015, 368, 20–25. [Google Scholar] [CrossRef] [PubMed]
  21. Kanda, T.; Jiang, X.; Yokosuka, O. Androgen receptor signaling in hepatocellular carcinoma and pancreatic cancers. World J. Gastroenterol. 2014, 20, 9229–9236. [Google Scholar] [PubMed]
  22. Gomella, L.G. Effective testosterone suppression for prostate cancer: Is there a best castration therapy? Rev. Urol. 2009, 11, 52–60. [Google Scholar] [PubMed]
  23. Grivas, P.D.; Robins, D.M.; Hussain, M. Predicting response to hormonal therapy and survival in men with hormone sensitive metastatic prostate cancer. Crit. Rev. Oncol. Hematol. 2013, 85, 82–93. [Google Scholar] [CrossRef] [PubMed]
  24. Luk, P.P.; Weston, J.D.; Yu, B.; Selinger, C.I.; Ekmejian, R.; Eviston, T.J.; Lum, T.; Gao, K.; Boyer, M.; O’Toole, S.A.; et al. Salivary duct carcinoma: Clinicopathologic features, morphologic spectrum, and somatic mutations. Head Neck 2016, 38, E1838–E1847. [Google Scholar] [CrossRef] [PubMed]
  25. Huang, X.; Hao, J.; Chen, S.; Deng, R. Salivary duct carcinoma: A clinopathological report of 11 cases. Oncol. Lett. 2015, 10, 337–341. [Google Scholar] [CrossRef] [PubMed]
  26. Mitani, Y.; Rao, P.H.; Maity, S.N.; Lee, Y.C.; Ferrarotto, R.; Post, J.C.; Licitra, L.; Lippman, S.M.; Kies, M.S.; Weber, R.S.; et al. Alterations associated with androgen receptor gene activation in salivary duct carcinoma of both sexes: Potential therapeutic ramifications. Clin. Cancer Res. 2014, 20, 6570–6581. [Google Scholar] [CrossRef] [PubMed]
  27. Butler, R.T.; Spector, M.E.; Thomas, D.; McDaniel, A.S.; McHugh, J.B. An immunohistochemical panel for reliable differentiation of salivary duct carcinoma and mucoepidermoid carcinoma. Head Neck Pathol. 2014, 8, 133–140. [Google Scholar] [CrossRef] [PubMed]
  28. Cros, J.; Sbidian, E.; Hans, S.; Roussel, H.; Scotte, F.; Tartour, E.; Brasnu, D.; Laurent-Puig, P.; Bruneval, P.; Blons, H.; et al. Expression and mutational status of treatment-relevant targets and key oncogenes in 123 malignant salivary gland tumours. Ann. Oncol. 2013, 24, 2624–2629. [Google Scholar] [CrossRef] [PubMed]
  29. Masubuchi, T.; Tada, Y.; Maruya, S.; Osamura, Y.; Kamata, S.E.; Miura, K.; Fushimi, C.; Takahashi, H.; Kawakita, D.; Kishimoto, S.; et al. Clinicopathological significance of androgen receptor, HER2, KI-67 and EGFR expressions in salivary duct carcinoma. Int. J. Clin. Oncol. 2015, 20, 35–44. [Google Scholar] [CrossRef] [PubMed]
  30. Williams, M.D.; Roberts, D.; Blumenschein, G.R., Jr.; Temam, S.; Kies, M.S.; Rosenthal, D.I.; Weber, R.S.; El-Naggar, A.K. Differential expression of hormonal and growth factor receptors in salivary duct carcinomas: Biologic significance and potential role in therapeutic stratification of patients. Am. J. Surg. Pathol. 2007, 31, 1645–1652. [Google Scholar] [CrossRef] [PubMed]
  31. Williams, L.; Thompson, L.D.; Seethala, R.R.; Weinreb, I.; Assaad, A.M.; Tuluc, M.; Ud Din, N.; Purgina, B.; Lai, C.; Griffith, C.C.; 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. [Google Scholar] [CrossRef] [PubMed]
  32. Nasser, S.M.; Faquin, W.C.; Dayal, Y. Expression of androgen, estrogen, and progesterone receptors in salivary gland tumors. Frequent expression of androgen receptor in a subset of malignant salivary gland tumors. Am. J. Clin. Pathol. 2003, 119, 801–806. [Google Scholar] [CrossRef] [PubMed]
  33. Locati, L.D.; Perrone, F.; Losa, M.; Mela, M.; Casieri, P.; Orsenigo, M.; Cortelazzi, B.; Negri, T.; Tamborini, E.; Quattrone, P.; et al. Treatment relevant target immunophenotyping of 139 salivary gland carcinomas (SGCS). Oral Oncol. 2009, 45, 986–990. [Google Scholar] [CrossRef] [PubMed]
  34. Sygut, D.; Bien, S.; Ziolkowska, M.; Sporny, S. Immunohistochemical expression of androgen receptor in salivary gland cancers. Pol. J. Pathol. 2008, 59, 205–210. [Google Scholar] [PubMed]
  35. Thompson, L.D.; Aslam, M.N.; Stall, J.N.; Udager, A.M.; Chiosea, S.; McHugh, J.B. Clinicopathologic and immunophenotypic characterization of 25 cases of acinic cell carcinoma with high-grade transformation. Head Neck Pathol. 2016, 10, 152–160. [Google Scholar] [CrossRef] [PubMed]
  36. Ito, F.A.; Ito, K.; Coletta, R.D.; Vargas, P.A.; Lopes, M.A. Immunohistochemical study of androgen, estrogen and progesterone receptors in salivary gland tumors. Braz. Oral Res. 2009, 23, 393–398. [Google Scholar] [CrossRef] [PubMed]
  37. Ishibashi, K.; Ito, Y.; Fujii, K.; Masaki, A.; Beppu, S.; Kawakita, D.; Ijichi, K.; Shimozato, K.; Inagaki, H. Androgen receptor-positive mucoepidermoid carcinoma: Case report and literature review. Int. J. Surg. Pathol. 2015, 23, 243–247. [Google Scholar] [CrossRef] [PubMed]
  38. Seethala, R.R.; Richmond, J.A.; Hoschar, A.P.; Barnes, E.L. New variants of epithelial-myoepithelial carcinoma: Oncocytic-sebaceous and apocrine. Arch. Pathol. Lab. Med. 2009, 133, 950–959. [Google Scholar] [PubMed]
  39. Di Palma, S. Carcinoma ex pleomorphic adenoma, with particular emphasis on early lesions. Head Neck Pathol. 2013, 7, S68–S76. [Google Scholar] [CrossRef] [PubMed]
  40. Nakajima, Y.; Kishimoto, T.; Nagai, Y.; Yamada, M.; Iida, Y.; Okamoto, Y.; Ishida, Y.; Nakatani, Y.; Ichinose, M. Expressions of androgen receptor and its co-regulators in carcinoma ex pleomorphic adenoma of salivary gland. Pathology 2009, 41, 634–639. [Google Scholar] [CrossRef] [PubMed]
  41. Fan, C.Y.; Melhem, M.F.; Hosal, A.S.; Grandis, J.R.; Barnes, E.L. Expression of androgen receptor, epidermal growth factor receptor, and transforming growth factor alpha in salivary duct carcinoma. Arch. Otolaryngol. Head Neck Surg. 2001, 127, 1075–1079. [Google Scholar] [CrossRef] [PubMed]
  42. Di Palma, S.; Simpson, R.H.; Marchio, C.; Skalova, A.; Ungari, M.; Sandison, A.; Whitaker, S.; Parry, S.; Reis-Filho, J.S. Salivary duct carcinomas can be classified into luminal androgen receptor-positive, HER2 and basal-like phenotypes. Histopathology 2012, 61, 629–643. [Google Scholar] [CrossRef] [PubMed]
  43. Yamamoto, N.; Minami, S.; Fujii, M. Clinicopathologic study of salivary duct carcinoma and the efficacy of androgen deprivation therapy. Am. J. Otolaryngol. 2014, 35, 731–735. [Google Scholar] [CrossRef] [PubMed]
  44. Chiosea, S.I.; Williams, L.; Griffith, C.C.; Thompson, L.D.; Weinreb, I.; Bauman, J.E.; Luvison, A.; Roy, S.; Seethala, R.R.; Nikiforova, M.N. Molecular characterization of apocrine salivary duct carcinoma. Am. J. Surg. Pathol. 2015, 39, 744–752. [Google Scholar] [CrossRef] [PubMed]
  45. Boon, E.; Bel, M.; van der Graaf, W.T.A.; van Es, R.J.J.; Eerenstein, S.; de Jong, R.B.; van den Brekel, M.; van der Velden, L.-A.; Witjes, M.; Hoeben, A.; et al. Salivary duct carcinoma: Clinical outcomes and prognostic factors in 157 patients and results of androgen deprivation therapy in recurrent disease (n = 31)—Study of the dutch head and neck society (DHNS). J. Clin. Oncol. 2016, 34, Suppl. abstract 6016. [Google Scholar]
  46. Antonarakis, E.S.; Lu, C.; Wang, H.; Luber, B.; Nakazawa, M.; Roeser, J.C.; Chen, Y.; Mohammad, T.A.; Chen, Y.; Fedor, H.L.; et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N. Engl. J. Med. 2014, 371, 1028–1038. [Google Scholar] [CrossRef] [PubMed]
  47. Dehm, S.M.; Schmidt, L.J.; Heemers, H.V.; Vessella, R.L.; Tindall, D.J. Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Res. 2008, 68, 5469–5477. [Google Scholar] [CrossRef] [PubMed]
  48. Guo, Z.; Yang, X.; Sun, F.; Jiang, R.; Linn, D.E.; Chen, H.; Chen, H.; Kong, X.; Melamed, J.; Tepper, C.G.; et al. A novel androgen receptor splice variant is up-regulated during prostate cancer progression and promotes androgen depletion-resistant growth. Cancer Res. 2009, 69, 2305–2313. [Google Scholar] [CrossRef] [PubMed]
  49. Sun, S.; Sprenger, C.C.; Vessella, R.L.; Haugk, K.; Soriano, K.; Mostaghel, E.A.; Page, S.T.; Coleman, I.M.; Nguyen, H.M.; Sun, H.; et al. Castration resistance in human prostate cancer is conferred by a frequently occurring androgen receptor splice variant. J. Clin. Investig. 2010, 120, 2715–2730. [Google Scholar] [CrossRef]
  50. Watson, P.A.; Chen, Y.F.; Balbas, M.D.; Wongvipat, J.; Socci, N.D.; Viale, A.; Kim, K.; Sawyers, C.L. Constitutively active androgen receptor splice variants expressed in castration-resistant prostate cancer require full-length androgen receptor. Proc. Natl. Acad. Sci. USA 2010, 107, 16759–16765. [Google Scholar] [CrossRef] [PubMed]
  51. Metzger, E.; Muller, J.M.; Ferrari, S.; Buettner, R.; Schule, R. A novel inducible transactivation domain in the androgen receptor: Implications for prk in prostate cancer. EMBO J. 2003, 22, 270–280. [Google Scholar] [CrossRef] [PubMed]
  52. Augello, M.A.; Hickey, T.E.; Knudsen, K.E. Foxa1: Master of steroid receptor function in cancer. EMBO J. 2011, 30, 3885–3894. [Google Scholar] [CrossRef] [PubMed]
  53. Robinson, J.L.; Holmes, K.A.; Carroll, J.S. FOXA1 mutations in hormone-dependent cancers. Front. Oncol. 2013. [Google Scholar] [CrossRef] [PubMed]
  54. Wen, S.; Niu, Y.; Lee, S.O.; Yeh, S.; Shang, Z.; Gao, H.; Li, Y.; Chou, F.; Chang, C. Targeting fatty acid synthase with ASC-J9 suppresses proliferation and invasion of prostate cancer cells. Mol. Carcinog. 2016, 55, 2278–2290. [Google Scholar] [CrossRef] [PubMed]
  55. Armstrong, C.M.; Gao, A.C. Drug resistance in castration resistant prostate cancer: Resistance mechanisms and emerging treatment strategies. Am. J. Clin. Exp. Urol. 2015, 3, 64–76. [Google Scholar] [PubMed]
  56. Merseburger, A.S.; Alcaraz, A.; von Klot, C.A. Androgen deprivation therapy as backbone therapy in the management of prostate cancer. OncoTargets Ther. 2016, 9, 7263–7274. [Google Scholar] [CrossRef] [PubMed]
  57. Kamata, Y.U.; Sumida, T.; Murase, R.; Nakano, H.; Yamada, T.; Mori, Y. Blockade of androgen-induced malignant phenotypes by flutamide administration in human salivary duct carcinoma cells. Anticancer Res. 2016, 36, 6071–6075. [Google Scholar] [CrossRef] [PubMed]
  58. Locati, L.D.; Perrone, F.; Cortelazzi, B.; Lo Vullo, S.; Bossi, P.; Dagrada, G.; Quattrone, P.; Bergamini, C.; Potepan, P.; Civelli, E.; 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. [Google Scholar] [CrossRef] [PubMed]
  59. Jaspers, H.C.; Verbist, B.M.; Schoffelen, R.; Mattijssen, V.; Slootweg, P.J.; van der Graaf, W.T.; van Herpen, C.M. Androgen receptor-positive salivary duct carcinoma: A disease entity with promising new treatment options. J. Clin. Oncol. 2011, 29, e473–e476. [Google Scholar] [CrossRef] [PubMed]
  60. Locati, L.D.; Quattrone, P.; Bossi, P.; Marchiano, A.V.; Cantu, G.; Licitra, L. A complete remission with androgen-deprivation therapy in a recurrent androgen receptor-expressing adenocarcinoma of the parotid gland. Ann. Oncol. 2003, 14, 1327–1328. [Google Scholar] [CrossRef] [PubMed]
  61. Van der Hulst, R.W.; van Krieken, J.H.; van der Kwast, T.H.; Gerritsen, J.J.; Baatenburg de Jong, R.J.; Lycklama a Nijeholt, A.A.; Meinders, A.E. Partial remission of parotid gland carcinoma after goserelin. Lancet 1994. [Google Scholar] [CrossRef]
  62. Urban, D.; Rischin, D.; Angel, C.; D’Costa, I.; Solomon, B. Abiraterone in metastatic salivary duct carcinoma. J. Natl. Compr. Cancer Netw. 2015, 13, 288–290. [Google Scholar]
  63. Locati, L.D.; Perrone, F.; Cortelazzi, B.; Imbimbo, M.; Bossi, P.; Potepan, P.; Civelli, E.; Rinaldi, G.; Quattrone, P.; Licitra, L.; et al. Activity of abiraterone in rechallenging two ar-expressing salivary gland adenocarcinomas, resistant to androgen-deprivation therapy. Cancer Biol. Ther. 2014, 15, 678–682. [Google Scholar] [CrossRef] [PubMed]
  64. Soper, M.S.; Iganej, S.; Thompson, L.D. Definitive treatment of androgen receptor-positive salivary duct carcinoma with androgen deprivation therapy and external beam radiotherapy. Head Neck 2014, 36, E4–E7. [Google Scholar] [CrossRef] [PubMed]
  65. Kuroda, H.; Sakurai, T.; Yamada, M.; Uemura, N.; Ono, M.; Abe, T.; Fujii, S.; Maeda, M.; Kohda, K.; Obata, M.; et al. Effective treatment by both anti-androgen therapy and chemotherapy for a patient with advanced salivary duct carcinoma. Jpn. J. Cancer Chemother. 2011, 38, 627–630. [Google Scholar]
Figure 1. Reported prevalence of AR splice variant expression in SDC. References: For AR-FL, [8,24,25,26,27,28,29,30,31,33,34,41,42,43,44,45]; for AR-V7, [8,26]; for AR-V3 and AR-45, [26].
Figure 1. Reported prevalence of AR splice variant expression in SDC. References: For AR-FL, [8,24,25,26,27,28,29,30,31,33,34,41,42,43,44,45]; for AR-V7, [8,26]; for AR-V3 and AR-45, [26].
Cancers 09 00017 g001
Table 1. Prevalence of positive AR immunoreactivity in different types of SGC.
Table 1. Prevalence of positive AR immunoreactivity in different types of SGC.
HistologyAR Positivity 1Reported Range 2References
SDC615/713 (86%)43%–100%[8,24,25,26,27,28,29,30,31,33,34,41,42,43,44,45]
AC NOS11/43 (26%)21%–33%[28,33,34]
AcCC6/40 (15%)0%–31%[28,32,34,35]
MEC7/135 (5%)0%–20%[27,28,32,33,34,36]
ACC7/145 (5%)0%–20%[28,32,33,34,36]
EMC0/6 (0%)N/A[28]
MECA0/7 (0%)N/A[28,33]
BCAC2/2 (100%)N/A[32]
PLGA1/2 (50%)N/A[28]
1 Number of AR-positive cases/total number of cases, in all studies combined; 2 Range of prevalence detected in the different studies. SDC, salivary duct carcinoma; AC NOS, adenocarcinoma not otherwise specified; AcCC, acinic cell carcinoma; MEC, mucoepodermoid carcinoma; ACC, adenoid cystic carcinoma; EMC, epithelial-myoepithelial carcinoma; MECA, myoepithelial carcinoma; BCAC, basal cell adenocarcinoma; PLGA, polymorphous low grade adenocarcinoma.
Table 2. Reported cases of ADT treatment in patients with AR-positive SGC.
Table 2. Reported cases of ADT treatment in patients with AR-positive SGC.
Patient ID 1HistologySexAge 2ADT AgentsResponsePFS (Months)Ref.
1AC NOSm73Bicalutamide + triptorelinCRN.K.[60]
2AC NOSm72Bicalutamide + triptorelinCR2[58]
3AC NOSmN.K.GoserelinPRN.K.[61]
4AC NOSm59Bicalutamide + triptorelinPR12[63]
5AC NOSm44Bicalutamide + triptorelinPR25[63]
6AC NOSm67Bicalutamide + triptorelinPR22[58]
7AC NOSm67Bicalutamide + triptorelinPR22[58]
8AC NOSm46Bicalutamide + triptorelinPR58[58]
9AC NOSm49Bicalutamide + triptorelinPR7[58]
10AC NOSm62Bicalutamide + triptorelinPR9[58]
11AC NOSm69Bicalutamide + triptorelinSD20[58]
12Cyst ACm79Bicalutamide + triptorelinPR14[58]
13Cyst ACf68Triptorelin + cyproteronePD0[58]
14Poor diff.m54Bicalutamide + triptorelinPD0[58]
15SDCf87Bicalutamide + leuprolide 3CR24[64]
16SDCm44Bicalutamide + triptorelinCR39[58]
17SDCm67Bicalutamide + triptorelinCR11[58]
18SDCm66BicalutamidePR14[43]
19SDCm50BicalutamidePR8[59]
20SDCf83BicalutamidePR26[59]
21SDCm45GoserelinPR4[62]
22SDCm45Bicalutamide + goserelinPR10[62]
23SDCm45Abiraterone + goserelinPR10[62]
24SDCm51Bicalutamide + triptorelinPR6[58]
25SDCm67Bicalutamide + triptorelinPR7[58]
26SDCf68Bicalutamide + leuprolideSD17[8]
27SDCm57BicalutamideSD14[59]
28SDCm56Bicalutamide + goserelinSD12[59]
29SDCm67Bicalutamide + goserelinSD8[59]
30SDCm75Bicalutamide + triptorelinSD8[58]
31SDCm54Bicalutamide + triptorelinSD10[58]
32SDCm68Bicalutamide + triptorelinSD23[58]
33SDCf48Bicalutamide + leuprolidePD0[8]
34SDCf69Bicalutamide + leuprolidePD0[8]
35SDCm77Bicalutamide + leuprolidePD0[8]
36SDCm73Bicalutamide + goserelinPD0[59]
37SDCm68Bicalutamide + goserelinPD0[59]
38SDCf64BicalutamidePD0[59]
39SDCm39BicalutamidePD0[59]
40SDCm73BicalutamidePD0[59]
1 Patients are sorted by tumor histology and then best response; 2 At start of ADT; 3 This patient received external beam radiotherapy together with ADT. ADT, androgen deprivation therapy; PFS, progression-free survival; Ref., reference; AC NOS, adenocarcinoma not otherwise specified; Cyst AC, cystadenocarcinoma; Poor diff., poorly differentiated; SDC, salivary duct carcinoma; m, male; f, female; N.K., not known; CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease.

Share and Cite

MDPI and ACS Style

Dalin, M.G.; Watson, P.A.; Ho, A.L.; Morris, L.G.T. Androgen Receptor Signaling in Salivary Gland Cancer. Cancers 2017, 9, 17. https://doi.org/10.3390/cancers9020017

AMA Style

Dalin MG, Watson PA, Ho AL, Morris LGT. Androgen Receptor Signaling in Salivary Gland Cancer. Cancers. 2017; 9(2):17. https://doi.org/10.3390/cancers9020017

Chicago/Turabian Style

Dalin, Martin G., Philip A. Watson, Alan L. Ho, and Luc G. T. Morris. 2017. "Androgen Receptor Signaling in Salivary Gland Cancer" Cancers 9, no. 2: 17. https://doi.org/10.3390/cancers9020017

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