Open Access

Impact of Amplified Oncogenes on Salivary Gland Carcinomas

VASILEIOS PAPANIKOLAOU 1
ARISTEIDIS CHRYSOVERGIS 1
ASIMAKIS D. ASIMAKOPOULOS 2
DESPOINA SPYROPOULOU 3
VASILEIOS RAGOS 4
GEORGE PAPANASTASIOU 4
SOTIRIOS PAPOULIAKOS 5
SOFIANIKI MASTRONIKOLI 6
DIMITRIOS ROUKAS 7
EVANGELOS TSIAMBAS 8 13
PAVLOS PANTOS 1
DIMITRIOS PESCHOS 9
NICHOLAS MASTRONIKOLIS 10
LOUKAS MANAIOS 11
PANAGIOTIS FOTIADES 12
ANTONIS VYLLIOTIS 13
  &  
EFTHYMIOS KYRODIMOS 1

1First Department of Otolaryngology, Hippocration Hospital, University of Athens, Athens, Greece

2Department of Otorhinolaryngology, Centre Hospital, Sion, Switzerland

3Department of Radiation Oncology, Medical School, University of Patras, Patras, Greece

4Department of Maxillofacial, Medical School, University of Ioannina, Ioannina, Greece

5Department of Otolaryngology, Gennimatas GNA Hospital, Athens, Greece

6Brighton and Sussex Medical School, Brighton, U.K.

7Department of Psychiatry, 417 Veterans Army Hospital (NIMTS), Athens, Greece

8Department of Cytology, 417 Veterans Army Hospital (NIMTS), Athens, Greece

13Diagnostic and Research Lab BIOCLAB, Athens, Greece

9Department of Physiology, Medical School, University of Ioannina, Ioannina, Greece

10Department of Otolaryngology, Medical School, University of Patras, Patras, Greece

11Department of Surgery, BIOCLINIC, Thessaloniki, Greece

12Department of Surgery, 424 Army Hospital, Thessaloniki, Greece

Cancer Diagnosis & Prognosis Sep-Oct; 3(5): 528-532 DOI: 10.21873/cdp.10250
Received 03 June 2023 | Revised 10 December 2024 | Accepted 19 July 2023
Corresponding author
Evangelos Tsiambas, MD, MSc, Ph.D., Cytologist, 17 Patriarchou Grigoriou E΄ Street, Ag. Paraskevi 15341, Athens, Greece. E-mail: tsiambasecyto@yahoo.gr
pdf image icon

Abstract

In normal epithelia, proto-oncogenes regulate critical intra- or intercellular functions, including cell growth and proliferation, apoptosis, and signaling transduction from the cell periphery (extracellular space) to the nucleus mediated by different pathways. Oncogenes are the mutated or amplified forms of the corresponding proto-oncogenes that are crucially involved in cell neoplastic and malignant transformation during carcinogenesis. Salivary gland carcinomas (SGCs) demonstrate a variety of histogenetic types. They are characterized by a broad spectrum of chromosomal and gene alterations. In particular, amplifications in specific genes [human epidermal growth factor receptor 2 (HER2), human epidermal growth factor receptor 4 (HER4), epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), Mouse double minute 2 homolog (MDM2), androgen receptor (AR), programmed death (ligand 1 (PD-L1), neurogenic differentiation factor 2 (NEUROD2), phosphatidylinositol 3,4,5-trisphosphate-dependent RAC exchanger 1 protein (PREX1), cyclin-dependent kinase4/6 (CDK4/6), proline-rich acidic protein 1 (PRAP1), kell antigen system (KEL), glutamate receptor subunit epsilon 2 (GRIN2D), Ewing sarcoma RNA-binding protein 1 (EWSR1), MYC proto-oncogene (MYC)] combined or not with chromosomal numerical imbalances (aneuploidy/ polysomy/monosomy) form different genetic signatures affecting the response to monoclonal antibody-based, oncologicaly targeted regimens. Different SGC histotypes demonstrate specific combinations of mutated/amplified genes that modify their clinicohistological features. In the current molecular review, we present the most important amplified oncogenes and their impact on the biological behavior of SGCS.
Keywords: Salivary, gland, oncogene, amplification, carcinoma, review

Gross chromosomal instability and numerical or structural gene modifications represent the main mechanisms of genetic alterations that are involved in epithelial neoplastic and progressively malignant transformation (1,2). Gene mutations, rearrangements and translocations are described as structural changes, whereas gene amplification or gain is the result of an excessive production of gene copies leading to protein overexpression (3,4). Gene amplification and mutations affect a broad spectrum of (proto-) oncogenes and suppressor genes respectively. Concerning the molecular mechanisms that lead to these alterations, viral-dependent carcinogenesis is an excellent model for understanding them (5,6). Proto-oncogenes promote a variety of intra- or intercellular functions in normal epithelia (7). Cell growth, proliferation and apoptotic death that secure tissue homeostasis are regulated by them (8). Additionally, signaling transduction from the cell periphery (extracellular space) to the nucleus is controlled by proto-oncogenes that are present in specific molecular pathways (9,10). Oncogenes represent the mutated or amplified forms of the corresponding proto-oncogenes during epithelial malignant transformation, as occurs in breast adenocarcinoma (11,12).

Salivary gland carcinomas (SGCs) are malignancies that belong to the head and neck carcinoma super family of tumors. Mucoepidermoid (MEC), adenoid cystic (ADCC) and salivary duct (SDC) carcinomas are the prominent histotypes, according to the updated World Health Organization histogenetic categorization (13). MEC, ADCC and SDC are characterized by differences regarding their grade of differentiation, recurrence rates, and metastatic potential (14-16). Interestingly, polymorphous adenocarcinoma, sebaceous adenocarcinoma, acinic-cell carcinoma, basal cell adenocarcinoma, secretory carcinoma, mucinous adenocarcinoma clear-cell carcinoma, cystadenocarcinoma, sebaceous lymphadenocarcinoma, and cystadenocarcinoma represent infrequent SGC histotypes (17,18). Moreover, carcinosarcoma, epithelial–myoepithelial carcinoma, secretory carcinoma, squamous cell carcinoma, anaplastic small-cell carcinoma, undifferentiated carcinomas, and intraductal carcinoma are ‘exotic’ types, very rare but, in the majority of cases, aggressive forms of SGC (19). In the current review, we focused on the oncogenes that have been found to be amplified in SGCs, forming specific genetic signatures in them.

Oncogenes Amplified in SGC Histotypes

SGC histotypes harbor a variety of mutated, fused/ translocated and amplified oncogenes (20,21). This genetic landscape mainly includes amplification of human epidermal growth factor receptor 2 (HER2), human epidermal growth factor receptor 4 (HER4), epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), Mouse double minute 2 homolog (MDM2), androgen receptor (AR), programmed death ligand 1 (PD-L1), neurogenic differentiation factor 2 (NEUROD2), phosphatidylinositol 3,4,5-trisphosphate-dependent RAC exchanger 1 protein (PREX1), cyclin-dependent kinase4/6 (CDK4/6), proline-rich acidic protein 1 (PRAP1), kell antigen system (KEL), glutamate receptor subunit epsilon 2 (GRIN2D), Ewing sarcoma RNA-binding protein 1 (EWSR1), MYC proto-oncogene (MYC) genes at different frequencies (Table I).

The HER2/neu proto-oncogene is located on the long arm of chromosome 17 (gene locus: 17q21) and encodes for a transmembranous glycoprotein that acts as a tyrosine kinase receptor. It is a member of the epidermal growth factor receptor super-family that includes EGFR (HER1), HER2, HER3 and HER4 genes (22). Signalling transduction is mediated by intracellular proteins following HER2 activation. ΗER2 protein overexpression is frequently detected in a variety of solid malignancies, including of the breast, lung, stomach and colon (23,24). Concerning HNCs, especially SGCs, there is a variety of monoclonal antibodies, such as alpelisib, trastuzumab and lapatinib, that target HER2 protein on cancerous cell membranes (25). Furthermore, HER2/neu overactivation seems to be correlated to an aggressive SGC phenotype, especially MEC (26,27). A study group explored the role of the two main molecular techniques that are applied for detecting HER2 gene amplification, namely fluorescence in situ hybridization, and next-generation sequencing (28). They reported a fine agreement comparing these two methods for the assessment of gene copy numbers in protein overexpressing cases. Interestingly, although amplification is the main mechanism of HER2 oncogenic activation, this gene is also mutated in a subset of SGCs. Another molecular analysis identified synchronous HER2 and tumor protein 53 (TP53) mutations in a series of oncocytic SGCs, which represent a distinct histogenetic variant (29). Based on the previously referred molecular data regarding the HER2 amplification /mutation status in SGCs, anti-HER2 targeted agents, including afatinib and trastuzumab, were used to inhibit the receptor, thereby preventing excessive signaling transduction (30,31). Interestingly, a novel monoclonal antibody–drug conjugate, fam-trastuzumab deruxtecan, seems to be a very promising agent because it provides simultaneous inhibition of HER2 and topoisomerase I in selected breast adenocarcinoma and SGC cases. In one of them, a HER2-positive metastatic parotid gland carcinoma, the malignancy demonstrated no recurrence as a result of a complete, well-tolerated response (32). Interestingly, increased HER2 copy numbers combined with PD-L1 overexpression have been detected in some SDC cases and correlated with a better prognosis and response rate to immunotherapeutic targeted anti-PD-L1 strategies (33).

Besides HER2 amplification, extensive gene-scanning analyses, mainly based on whole-genome sequencing platforms, have revealed focal amplifications on 8q21-q23 gene loci (MYC/MYB genes) in the majority of SGC subtypes, including MECs and ADCCs (34). Additionally, ALK amplification was detected in a molecular study with combined fluorescent in situ hybridization and next-generation sequencing analyses. This is an alternative to the ALK rearrangement mechanism found in a very limited subgroup of patients with SDC (35). Interestingly, in subsets of clear-cell myoepithelial carcinomas, the EWSR1 gene has been found to be amplified, although rearrangement is the prominent mechanism of its alteration in SGCs (36). Concerning another gene, AR, there is limited, but strong evidence that its protein overexpression in rare cases of SDCs is a result of its gene amplification (37). Finally, actinin 4 (ACTN4) gene has been found to be overexpressed due to amplification in solid malignancies of different histotypes including SGCs (38). In contrast to HER2 and EWSR1 genes, MDM2 and CDK4 oncogenes, which share the chromosomal region 12q13-15, demonstrate a low amplification rate in SDCs. A molecular study showed that although rare, the combination of MDM and CDK4 amplification is correlated to TP53 mutations and high-mobility group protein 2 (HMG2) rearrangement/amplification in subgroups of ex-pleomorphic adenomas (39). Similarly, another study group analyzed a series of SDCs by implementing a next-generation sequencing assay (40). They reported only one MDM2-amplified case compared to HER2-amplified ones. The majority of the previous referred studies concluded that HER2 but not MDM2 amplification is correlated to an aggressive phenotype and poor survival rates in subgroups of patients. For this reason, they suggest that targeting the HER2-mediated pathway by monoclonal antibodies is an optimal oncological approach that could provide benefits for these patients.

In conclusion, amplification affects specific genes (HER2/HER4/EGFR, ALK, MDM2, AR, PD-L1, NEUROD2, PREX, CDK4/6, PRAP1, KEL, GRIN2D, EWSR1, MYC) in SGCs, combined or not with numerical imbalances (aneuploidy/polysomy/monosomy) in the corresponding chromosomes. Amplification of these oncogenes is associated with an excessive expression of their products (oncoproteins). As with mutations in specific genes, these amplified genes lead to different genetic signatures and modify the rate of response to targeted, monoclonal antibody-based therapeutic strategies in the corresponding patients improving their survival potential.

Conflict of Interest

The Authors have no conflicts of interest to declare.

Authors’ Contributions

ET, AC, and VP: Design of the study and article writing; DS, DP, VR, NM and EK: academic advisors; PP, AA, SP, DR, GP and SM: collection and management of references and published data. All Authors read and approved the final article.

References

1 Stratton MR Campbell PJ & Futreal PA The cancer genome. Nature. 458(7239) 719 - 724 2009. DOI: 10.1038/nature07943
2 Albertson DG Collins C McCormick F & Gray JW Chromosome aberrations in solid tumors. Nat Genet. 34(4) 369 - 376 2003. DOI: 10.1038/ng1215
3 Hanahan D & Weinberg R Hallmarks of cancer: The next generation. Cell. 144(5) 646 - 674 2011. DOI: 10.1016/j.cell.2011.02.013
4 Arora S Molecular genetics of head and neck cancer (Review). Mol Med Rep. 19 - 22 2012. DOI: 10.3892/mmr.2012.889
5 Hanahan D Hallmarks of cancer: New dimensions. Cancer Discov. 12(1) 31 - 46 2022. DOI: 10.1158/2159-8290.CD-21-1059
6 Mersi EA Fietelson MA & Munger K Human viral oncogenesis: A cancer hallmarks analysis. Cell Host Microbe. 15(3) 266 - 282 2014. DOI: 10.1016/j.chrom.2014.02.011
7 Cline MJ Keynote address: The role of proto-oncogenes in human cancer: Implications for diagnosis and treatment. Int J Radiat Oncol Biol Phys. 13(9) 1297 - 1301 1987. DOI: 10.1016/0360-3016(87)90219-7
8 Anderson MW Reynolds SH You M & Maronpot RM Role of proto-oncogene activation in carcinogenesis. Environ Health Perspect. 98 13 - 24 1992.
9 Brown G Oncogenes, proto-oncogenes, and lineage restriction of cancer stem cells. Int J Mol Sci. 22(18) 9667 2021. DOI: 10.3390/ijms22189667
10 Blanpain C Tracing the cellular origin of cancer. Nat Cell Biol. 15(2) 126 - 134 2013.
11 Kontomanolis EN Koutras A Syllaios A Schizas D Mastoraki A Garmpis N Diakosavvas M Angelou K Tsatsaris G Pagkalos A Ntounis T & Fasoulakis Z Role of oncogenes and tumor-suppressor genes in carcinogenesis: A review. Anticancer Res. 40(11) 6009 - 6015 2020. DOI: 10.21873/anticanres.14622
12 Shimazaki A Kubo M Kurata K Takao Y Hayashi S Harada Y Kawaji H Kaneshiro K Yamada M Kai M & Nakamura M CCND1 copy number variation in circulating tumor DNA from luminal B breast cancer patients. Anticancer Res. 42(8) 4071 - 4077 2022.
13 Seethala RR & Stenman G Update from the 4th edition of the World Health Organization classification of head and neck tumours: Tumors of the salivary gland. Head Neck Pathol. 11(1) 55 - 67 2017. DOI: 10.1007/s12105-017-0795-0
14 Skálová A Hyrcza MD & Leivo I Update from the 5th edition of the World Health Organization classification of head and neck tumors: Salivary Glands. Head Neck Pathol. 16(1) 40 - 53 2022. DOI: 10.1007/s12105-022-01420-1
15 Nishida H Kusaba T Kawamura K Oyama Y & Daa T Histopathological aspects of the prognostic factors for salivary gland cancers. Cancers (Basel). 15(4) 1236 2023. DOI: 10.3390/cancers15041236
16 Young A & Okuyemi OT Malignant salivary gland tumors. Treasure Island, FL, USA, StatPearls Publishing. DOI: 10.7759/cureus.35722
17 Chen Y Li G Jiang W Nie RC Deng H Chen Y Li H & Chen Y Prognostic risk factor of major salivary gland carcinomas and survival prediction model based on random survival forests. Cancer Med. 12(9) 10899 - 10907 2023. DOI: 10.1002/cam4.5801
18 Hamamoto Y Harada H Suzuki M Fujii T & Nakatsuka SI Salivary duct carcinoma of the parotid gland originating from an epithelial-myoepithelial carcinoma: Report of a rare case. Head Neck Pathol. 14(1) 283 - 289 2020. DOI: 10.1007/s12105-019-01034-0
19 Andersson MK & Stenman G The landscape of gene fusions and somatic mutations in salivary gland neoplasms – Implications for diagnosis and therapy. Oral Oncol. 57 63 - 69 2016. DOI: 10.1016/j.oraloncology.2016.04.002
20 Persson F Andren Y Winnes M Wedell B Nordkvist A & Gudnadottir G High-resolution genomic profiling of adenomas and carcinomas of the salivary glands reveals amplification, rearrangement, and fusion of HMGA2. Genes Chromosomes Cancer. 48(1) 69 - 82 2009. DOI: 10.1002/gcc.20619
21 Harari PM Epidermal growth factor receptor inhibition strategies in oncology. Endocr Relat Cancer. 11 689 - 708 2004.
22 Valabrega G Montemurro F & Aglietta M Trastuzumab: mechanism of action, resistance and future perspectives in HER2-overexpressing breast cancer. Ann Oncol. 18(6) 977 - 984 2007. DOI: 10.1093/annonc/mdl475
23 Takenaka M Hanagiri T & Shinohara S The prognostic significance of HER2 overexpression in non-small cell lung cancer. Anticancer Res. 31 4631 - 4636 2011.
24 Falchook GS Lippman SM Bastida CC & Kurzrock R Human epidermal receptor 2-amplified salivary duct carcinoma: regression with dual human epidermal receptor 2 inhibition and anti-vascular endothelial growth factor combination treatment. Head Neck. 36(3) E25 - E27 2014. DOI: 10.1002/hed.23429
25 Yoshimura T Higashi S Yamada S Noguchi H Nomoto M Suzuki H Ishida T Takayama H Hirano Y & Yamashita M PCP4/PEP19 and HER2 are novel prognostic markers in mucoepidermoid carcinoma of the salivary gland. Cancers. 14 54 - 62 2021. DOI: 10.3390/cancers14010054
26 Bou Zerdan M Kumar PA Zaccarini D Ross J Huang R & Sivapiragasam A Molecular targets in salivary gland cancers: A comprehensive genomic analysis of 118 mucoepidermoid carcinoma tumors. Biomedicines. 11(2) 519 2023. DOI: 10.3390/biomedicines11020519
27 Ferguson DC Momeni Boroujeni A Zheng T Mohanty AS Ho AL Arcila ME Ross DS & Dogan S ERBB2 amplification status in 67 salivary duct carcinomas assessed by immunohistochemistry, fluorescence in situ hybridization, and targeted exome sequencing. Mod Pathol. 35(7) 895 - 902 2022. DOI: 10.1038/s41379-021-00999-0
28 Zaccarini DJ Sivapiragasam A Sokol E Huang RS Pavlick DC Janovitz T Nasr MR & Ross JS Comprehensive molecular profiling of oncocytic salivary gland malignancies. Appl Immunohistochem Mol Morphol. 30(9) 609 - 613 2022. DOI: 10.1097/PAI.0000000000001048
29 Salawu A Hansen AR Spreafico A Al-Ezzi E Webster S Bedard PL Doi J Wang L Siu LL & Abdul Razak AR A Phase 2 Trial of afatinib in patients with solid tumors that harbor genomic aberrations in the HER family: The MOBILITY3 basket study. Target Oncol. 17(3) 271 - 281 2022. DOI: 10.1007/s11523-022-00884-z
30 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. 33(6) 2587 - 2591 2013. DOI: 10.6004/jnccn.2021.7089
31 Chatzopoulos K Collins AR Sotiriou S Keeney MG Visscher DW Rivera M Schembri-Wismayer DJ Lewis JE Greipp PT Sukov WR Chintakuntlawar AV Price KA & Garcia JJ Increased ERBB2 gene copy numbers reveal a subset of salivary duct carcinomas with high densities of tumor infiltrating lymphocytes and PD-L1 expression. Head Neck Pathol. 14(4) 951 - 965 2020. DOI: 10.1007/s12105-020-01163-x
32 Karpinets TV Mitani Y Liu B Zhang J Pytynia KB Sellen LD Karagiannis DT Ferrarotto R Futreal AP & El-Naggar AK Whole-genome sequencing of common salivary gland carcinomas: Subtype-restricted and shared genetic alterations. Clin Cancer Res. 27(14) 3960 - 3969 2021. DOI: 10.1158/1078-0432.CCR-20-4071
33 Majewska H Gorczyński A Czapiewski P Menon R Mueller J Lakis S Heuckmann JM Laco J Gupta R Andreasen S Stodulski D Iliszko M Dziadziuszko R Jassem J Heukamp LC & Biernat W ALK alterations in salivary gland carcinomas. Virchows Arch. 478(5) 933 - 941 2021. DOI: 10.1007/s00428-020-02971-w
34 Skálová A Agaimy A Vanecek T Baněčková M Laco J Ptáková N Šteiner P Majewska H Biernat W Corcione L Eis V Koshyk O Vondrák J Michal M & Leivo I Molecular profiling of clear cell myoepithelial carcinoma of salivary glands with ewsr1 rearrangement identifies frequent PLAG1 gene fusions but no EWSR1 fusion transcripts. Am J Surg Pathol. 45(1) 1 - 13 2021. DOI: 10.1097/PAS.0000000000001591
35 You Y Wang P Wan X Xu L Gong Y & Zhang W Androgen deprivation therapy for patients with androgen-receptor-positive metastatic salivary duct carcinoma: A case report and review of the literature. Onco Targets Ther. 14 3481 - 3486 2021. DOI: 10.2147/OTT.S304900
36 Honda K Development of biomarkers to predict recurrence by determining the metastatic ability of cancer cells. J Nippon Med Sch. 89(1) 24 - 32 2022. DOI: 10.1272/jnms.JNMS.2022_89-118
37 Bou Zerdan M Kumar PA Zaccarini D Ross J Huang R & Sivapiragasam A Molecular targets in salivary gland cancers: A comprehensive genomic analysis of 118 mucoepidermoid carcinoma tumors. Biomedicines. 11(2) 519 2023. DOI: 10.3390/biomedicines11020519
38 Crhysovergis A Papanikolaou V Spyropoulou D Roukas D Asimakopoulos AA Papanastasiou G Mastronikoli S Falidas E Papouliakos S Tsiambas E Pantos P Ragos V Peschos D Mastronikolis N & Kyrodimos E Mutational signatures in salivary gland carcinomas. Cancer Diagn Progn. 3(4) 411 - 415 2023. DOI: 10.21873/cdp.10233
39 Grünewald I Trautmann M Busch A Bauer L Huss S Schweinshaupt P Vollbrecht C Odenthal M Quaas A Büttner R Meyer MF Beutner D Hüttenbrink KB Wardelmann E Stenner M & Hartmann W MDM2 and CDK4 amplifications are rare events in salivary duct carcinomas. Oncotarget. 7(46) 75261 - 75272 2016. DOI: 10.18632/oncotarget.12127
40 Santana T Pavel A Martinek P Steiner P Grossmann P Baněčková M & Skálová A Biomarker immunoprofile and molecular characteristics in salivary duct carcinoma: clinicopathological and prognostic implications. Hum Pathol. 93 37 - 47 2019. DOI: 10.1016/j.humpath.2019.08.009