Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Feb 1.
Published in final edited form as: Curr Opin Otolaryngol Head Neck Surg. 2022 Feb 1;30(1):33–39. doi: 10.1097/MOO.0000000000000776

Emerging Concepts in Sinonasal Tumor Research

Diana C Lopez 1,2, Andrew E Wadley 1,3, Nyall R London Jr 1,4,5,*
PMCID: PMC9512446  NIHMSID: NIHMS1837145  PMID: 34958321

Abstract

Purpose of review:

Sinonasal malignancies are rare and understudied, often diagnosed at late stages, and may behave aggressively. This review explores investigative diagnostic, therapeutic, and scientific advances specific to sinonasal undifferentiated carcinoma (SNUC), intestinal-type adenocarcinoma (ITAC), and olfactory neuroblastoma (ONB).

Recent findings:

A number of studies have recently contributed more robust knowledge of the genetic and molecular landscapes of SNUC, ITAC, and ONB. These analyses have identified SMARCB1 and IDH2 mutations in SNUC, potentially allowing for the tumor’s subdivision. Recent studies have also defined a role for induction chemotherapy in SNUC. Somatic mutations for ITAC have been identified and may be potentially targetable with FDA approved therapies. Studies defining the tumor microenvironment for ITAC and ONB have introduced the possibility of immune checkpoint inhibition for these tumor types.

Summary:

Studies reviewed here detail promising results of the most current and novel characterization of SNUC, ITAC, and ONB genetic and molecular landscapes, which have informed ongoing therapeutic discovery. With continued multi-institutional efforts, the field of sinonasal tumor research will achieve higher disease control and improved treatment outcomes for patients afflicted with these rare cancers.

Keywords: Sinonasal malignancies, Sinonasal Undifferentiated Carcinoma, Intestinal-Type Adenocarcinoma, Olfactory Neuroblastoma

Introduction

Sinonasal malignancies comprise less than five percent of all head and neck tumors with a cumulative incidence of 0.5–1.0 per 100,000 individuals per year1,2. Although survival outcomes vary with tumor type, sinonasal malignancies may behave aggressively and are often diagnosed at late stages3. Treatment consists of endoscopic or open surgery, induction and/or definitive chemotherapy, and radiotherapy, with therapeutic decision-making hinging on tumor pathology, cancer stage, and implications for potential damage to anatomically adjacent neurovascular structures. Sinonasal squamous cell carcinoma (SNSCC) and adenocarcinoma, including intestinal-type adenocarcinoma (ITAC), represent the most common of the sinonasal tumor types4,5. Additional malignancies within the sinonasal tract include olfactory neuroblastoma (ONB), sinonasal undifferentiated carcinoma (SNUC), adenoid cystic carcinoma, and others2,68. Sinonasal tumors are rare and understudied3, but recent advancements have heralded improved genetic and molecular classification and the elucidation of potential targeted treatment strategies. The goal of this article is to explore these advances with a focus on SNUC, ITAC, and ONB.

Sinonasal Undifferentiated Carcinoma

Sinonasal undifferentiated carcinoma is an uncommon and aggressive cancer defined by the World Health Organization as an undifferentiated tumor lacking glandular and squamous features, and to date has been effectively a diagnosis of exclusion9,10. SNUC comprises merely 3–5% of all sinonasal carcinomas9. Its incidence is low at 0.02 per 100,000 individuals11. SNUC has a male predominance of ∼2–3:1 and typically presents in the fifth decade of life11,12. Although most cases originate within the ethmoid and maxillary sinuses13, patients often present with advanced, invasive disease that pervades dura in 50–74% of cases, and the orbit in 30–63% of cases14,15. The tumor has high metastatic potential with unfavorable outcomes9,13,15,16. Median survival is a bleak 22 months as reported by Surveillance, Epidemiology, End Results Program (SEER) database analysis of 318 cases11. While SNUC is known to arise from nasal and paranasal sinus epithelium, its immunohistologic profile remains largely uncharacterized17,18. Nevertheless, with the elucidation of genetic mutations such as SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily b, member 1 (SMARCB1) and isocitrate dehydrogenase 2 (IDH2), this tumor’s diagnosis is becoming more granular19,20.

Genetic and molecular characterization of SNUC identified potential therapeutic targets

Development of targeted therapy will come only with an improved understanding of the genetics and biologic behavior of this heterogenous carcinoma9; therefore, the establishment of the first human-derived SNUC cell lines by Takahashi et al. in 2012 proved a pivotal advance21. This group demonstrated that these SNUC cell lines express pan-keratin, cytokeratin 8, cytokeratin 19, and epithelial markers of E-cadherin and B-catenin, but lack mesenchymal markers including N-cadherin, vimentin, and alpha-SMA21. Twelve translocations spanning diverse chromosomes were also reported, further contributing to SNUC characterization21. Using the novel cell line, Takahashi et al. discovered the ERBB2 gene to be highly amplified, and thus, human epidermal growth factor receptor 2 to be overexpressed and phosphorylated within SNUC22. Subsequent HER2 signaling pathway inhibition with FDA-approved dual small molecule EGFR/HER2 inhibitor, lapatinib23, stifled cell growth, suggesting HER2 as a potentially worthwhile molecular target22.

To differentiate SNUC histopathology from that of similar malignancies, including sinonasal neuroendocrine carcinoma (SNEC) and ONB, a 2018 study employed genome wide copy number profiling of tumor tissue24. SNUC tumors were defined by a cytokeratin positive (CK +) and neuroendocrine negative (Ne −) marker expression pattern and compared to samples previously diagnosed as SNUC by pathology alone. Of all tumors studied, 17 of 54 were reclassified, highlighting the diagnostic challenges in the correct diagnosis of this tumor24. Following this investigation, a 2019 study established seven genes that fully distinguished SNUC from SNSCC. Expression of the human chloride channel accessory-2 (CLCA2) gene, which encodes a calcium-activated chloride channel regulator protein with implications in p53 tumor suppression, was most markedly different between the two malignancies25. Gene ontologies corresponding to DNA repair and cell division were also upregulated in SNUC25. Furthermore, a 2019 MD Anderson study analyzed 13 treatment-naïve SNUC samples to ascertain prognostic markers for treatment response to induction chemotherapy (IC). Twenty-four genes distinguished responders from non-responders, particularly IL20 and FGF20. Sixteen gene pairs were associated with IC positive response, encouraging future treatment decisions that cater towards differential characteristics of diverse SNUC tumors26.

Targeted sequencing in SNUC has allowed for the identification of mutations in IDH2, most of which occur within the protein’s R140 and R172 arginine residues27,28. In 2017, IDH2 R172 mutations were reported within 55–80% of SNUC tumors27,28. This enzyme catalyzes α-ketoglutarate formation, but when mutated, increases (R)-2-hydroxyglutarate (2-HG), an oncometabolite that disturbs cell differentiation via histone and DNA hypermethylation29,30. A 2019 study that examined the monoclonal antibody 11C8B1 to IDH2 R172S in SNUC concluded the antibody may be useful as a supplemental immunohistologic diagnostic marker31. A more recent 2020 evaluation of IDH2 mutation’s specificity to SNUC reported that 11 of 36 (31%) SNUC samples contained IDH2 mutations, although IDH2 mutations also occurred in a minority of neuroendocrine carcinoma, high-grade non-intestinal-type adenocarcinoma, and poorly differentiated squamous cell carcinoma tumors32. The group noted that IDH2-mutant cases had a higher disease specific survival32, a finding corroborated by a 2021 study that highlights the influence IDH2 mutation status has on sinonasal tumor biology19. Similarly, Chitguppi et al. recently proposed a novel SNUC classification based on the expression of SMARCB1, a known tumor suppressor gene and core subunit of the SWI/SNF chromatin remodeling complex20. They noted that SMARCB1 deficiency portended a worse prognosis, with a 50% vs 0% 1-year mortality rate in the SMARCB1-deficient vs -retained group, respectively20. Thus, current research suggests that SNUC may in fact be comprised of multiple subtypes with unique behavioral patterns defined by the presence of genetic mutations, such as IDH2 and SMARCB1. Lastly, a 2020 study sought to characterize SNUC’s immune-oncology gene expression. With next generation sequencing, PRAME (preferentially expressed antigen in melanoma), a testis-selective cancer antigen involved in rendering cancer cell qualities of stemness, invasion, and metastasis, was identified as the most upregulated gene in SNUC and may serve as an important immunotherapeutic target6. Execution of the first whole genome or whole exome sequencing studies in SNUC will allow for continued granular subdivision of SNUC tumors and further precise therapeutic targets based on genomics.

Recent advances in clinical treatment strategies

Multiple studies have demonstrated the value of multimodal therapy for advanced sinonasal tumors, which may consist of radiotherapy, chemotherapy, and/or surgery, via an open craniofacial, endonasal, or combined approach9,12,17,26, in establishing higher disease control9,13,33. Given the aggressive nature of some sinonasal malignancies, induction chemotherapy (IC) is more commonly being incorporated prior to definitive therapy12,17,3436. Proposed benefits of IC may include possible protection against distant metastasis and potential orbital preservation, although such notions continue to be investigated, most notably via a phase II IC, (NCT00707473), trial that is currently underway17,34,3638. Response status to IC may also serve as a guide for likely efficacy of ensuing definitive chemoradiation12,15.

A landmark MD Anderson study by Amit et al. assessed the role of IC in guiding definitive therapy for SNUC in 201917. Ninety-five treatment-naive SNUC patients were treated with platinum-based IC regimen of cisplatin and etoposide17. In patients with a favorable IC response, improved survival was achieved with definitive chemotherapy and radiation (CRT), whereas in those without an IC response, surgery proved the preferred method for disease control17. 5-year disease specific survival in individuals with partial or complete IC response was 81% after treatment with CRT, whereas in individuals without IC response was 0% following CRT and 39% following surgery with chemoradiation17. This study’s sample size contains the greatest number of untreated SNUC tumors to date, a significant feat considering the rarity of SNUC. It also represents a milestone in establishing therapeutic sequence guidelines for a thus far elusive cancer. Similarly, at The Ohio State University, a 2020 retrospective study of 21 SNUC cases by London et al. highlighted an institutional trend towards use of TPF (docetaxel, cisplatin, fluorouracil) IC in the treatment algorithm for treatment-naïve SNUC37.

Progress in understanding SNUC genetic and molecular composition, pathways relevant to carcinogenesis, and favorable treatment sequences is ongoing and flourishing. The continued development of potential genetic, molecular, and immunotherapeutic targets, as well as multi-institutional establishment of optimal treatment regimens is crucial to achieving sound control over this aggressive cancer, and thus, securing improved patient treatment outcomes.

Intestinal-Type Sinonasal Adenocarcinoma

Intestinal-type sinonasal adenocarcinoma (ITAC) is a rare form of cancer arising in the epithelium of the paranasal sinuses and the nasal cavities. This cancer forms most often in the ethmoid sinuses (85%) and comprises 8–25% of all malignant sinonasal tumors39. Distant or lymph node metastasis is relatively uncommon while local recurrence and intracranial invasion account for the primary causes of mortality among these patients4,39,40. ITAC prevalence represents an important occupational health problem as the tumor’s etiology is strongly related to wood dust exposure, making it a disease more commonly seen in carpenters and furniture makers. Individuals in professions with prolonged exposure to wood dust have up to 500–900 times greater risk of developing ITAC compared to the wider community4. Contrarily, tobacco smoke exposure does not seem to play a major role in ITAC development4.

Five distinct histopathologic subtypes of ITAC have been described: colonic (40%), solid (20%), papillary (18%), and mucinous and mixed type (22% combined)39. Although it is known that papillary and colonic subtypes are associated with better clinical outcomes39 while solid and mucinous subtypes possess more aggressive characteristics41, the events leading up to subtype differentiation remain a mystery.

Potential mechanisms of tumorigenesis in ITAC

Currently there is no established pathogenic mechanism or precursor lesion to explain the development of ITAC42. Wood dust, though not a direct mutagen, is thought be an irritant to the nasal mucosa that precipitates prolonged inflammation to noxious stimuli, and thus, cell proliferation with long-term exposure39,42. An upregulation of inflammatory cytokine release of transcription factors, such as tumor necrosis factor and nuclear factor κB, has been demonstrated in sinonasal cancer studies4,43,44. Other markers, such as increased COX2 expression, and TP53 G>A missense mutations possibly linked with the presence of reactive oxygen species, offer further evidence for prolonged inflammation as a potential player in ITAC tumorigenesis39,44,45. Another hypothesized mechanism to explain the origin of this sinonasal cancer is that of cancer stem cells (CSC) capable of differentiating into various cell types4. This model has also been suggested for other head and neck tumors and could provide a more intuitive explanation for ITAC’s ability to form mixed histological types and recur as distinct tumor types post-resection1,4. However, to-date little research has investigated this possibility.

Protein expression and genetic profiling of ITAC

ITACs are named for their histomorphologic resemblance to adenocarcinoma of the intestinal tract, colorectal carcinoma (CRC)45. Indeed, one characteristic trait shared by all ITAC subtypes is that of intra- or extracellular mucin production39. However, recent studies have shown that although these tumors appear similar, ITAC and CRC likely arise from separate pathways. For instance, microsatellite instability (MSI) is an important process in the pathogenesis of CRC but unlikely to play an important role in that of sinonasal ITAC1,7. Additionally, while expression of anaplastic lymphoma kinase (ALK) protein was suspected in ITAC, as seen in lung adenocarcinoma, experiments using break-apart fluorescent in-situ hybridization and immunohistochemistry in 96 sinonasal adenocarcinoma samples offered no evidence of ALK protein expression46.

Other efforts have focused on linking detectable genomic markers and protein expression patterns with histologic or clinical outcomes41,45,4749. Studies using direct sequencing, fluorescence in situ hybridization (FISH), multiplex ligation-dependent probe amplification (MLPA), and microarray comparative genomic hybridization (CGH) have indicated that ITACs possess ample genetic aberrations throughout the genome. However, the papillary subtype seems to consistently possess relatively few copy number alterations as compared to other subtypes47,50, and notably, is often associated with more favorable clinical outcomes39. Other genetic profiling investigations showed worse overall survival for tumors carrying CGH losses at 4q32-ter and gains at 1q22, 6p22, and 3q29, or carrying MLPA losses of TIMP247,48. With methylation-specific MLPA, Costales et al. demonstrated that ITAC carries a greater number of gene methylations than SNSCC, and, more specifically, that papillary and colonic ITAC subtypes possess more gene methylations than solid and mucinous ITAC subtypes, with a mean of 1.26 gene methylations per tumor versus 0.63, respectively5. This finding may suggest that gene methylation plays a larger, or at least distinct, role in the differential development of the less aggressive papillary and colonic tumors5. Using immunohistochemistry, positive p16 protein expression was found to be associated with shorter overall survival41, and with the same technique, an absence of annexin A2 expression was correlated with the aggressive mucinous subtype, and therefore, with decreased survival51. Direct sequencing of proto-oncogenes such as EGFR, KRAS, and BRAF have not proven as informative since few to no ITACs studied carried prognostic mutations in these genes49,52. Overall, such findings are helpful in characterizing ITAC genetic subgroups and have implications in future therapeutic decision making and personalized therapy development.

Therapeutic advancements

Standard treatment is surgical resection with post-operative radiotherapy4,40. However, resection with wide tumor-free margins may be challenging due to the close proximity to critical neurovascular structures4. The first immortal ITAC tumor cell line derived was established by Pérez-Escuredo et al. in 201040. Multiple genetic alterations characteristic of ITAC were described within this cell line, validating its similarity to primary tumor samples40. An in vitro model such as this, which preserves the biological properties of the original cancer, has the potential to be a powerful tool for both functional studies and for the evaluation of novel therapeutic agents. In a study using next generation sequencing, Sánchez-Fernández et al. found one or more potentially actionable somatic mutations in 20 of 27 ITAC cases. Eight of these represent biomarkers for existing FDA-approved targeted therapies53. Other research looking at tumor infiltrating lymphocytes and PD-L1 expression optimistically describe that a subset of ITAC, especially papillary and colonic subtypes, may be candidates for immune checkpoint inhibition8,54.

Olfactory Neuroblastoma

Olfactory neuroblastoma, or esthesioneuroblastoma, is a rare nasal cavity and anterior skull base malignancy first described in 19245557. ONB is thought to originate from olfactory neuro-epithelium in the upper nasal cavity at the region of the cribriform plate58,59. ONB incidence is low at 0.4 per million per year58. ONB progresses insidiously and has a 5-year survival rate of 80%60, although late local-regional recurrence is well-documented, for which long-term patient follow up is advisable58,61. Difficulty in identifying targeted therapies in ONB stems in part from a lack of precise oncogenic driver elucidation62.

Investigation of the ONB genetic and molecular landscape

A 2019 study by Classe et al. evaluated Ki67 proliferation index and tumor infiltrating lymphocytes (TILs) as prognostic alternates to the current Hyams grading system63,64. They demonstrated an association of both high Ki67 proliferation index and elevated intratumoral TILs with high grade ONB. Furthermore, a Ki67 PI greater than 25%, and a CD4/CD8 ratio of greater than two corresponded with poor survival, confirming Ki67 PI and TILs as advantageous prognostic markers63. Although a broad variety of ONB cytogenetic and genomic alterations are reported in the literature, common findings include positive association of chromosome 11 deletion and chromosome 1p gain with poor ONB survival, and TP53 gene alterations that account for the tumor’s most frequent mutations 55,6567. An important 2018 study further molecularly classified ONB into two etiologic groups: neural-like and basal-like62. Although, a third of this study’s basal-like tumors were comprised of an IDH2 R172 mutant-enriched subgroup characterized by pervasive DNA hypermethylation,62 prior studies have detected a low incidence of IDH2 mutation in ONB, ranging between 0–4%27,28,32,65. Finally, Cracolici et al. recently reported on uniform somatostatin receptor 2 (SSTR2) expression in ONB68. Following a finding of significantly greater SSTR2 staining in ONB than in histologically related neoplasms, they suggested SSTR2 likely represents a diagnostically important ONB marker68. Moreover, imaging and therapy based in a somatostatin analog have been applied to extensive or metastatic ONB6972.

Treatment targets in ONB

Therapeutics directed towards the immune checkpoint pathway could be applicable for ONB. Programmed cell death ligand 1 (PD-L1), a transmembrane glycoprotein that interrelates with PD-1, contributes to tumor immune system evasion. A recent 2020 study showed PD-L1 expression in 40% of primary ONB and in 75% of cervical metastases derived from a primary PD-L1 negative ONB56. Greater PD-1+ and CD8+ lymphocyte presence was also identified within tumor and stroma of PD-L1+ tumors as compared to PD-L1- tumors; overall suggesting that PD-L1/PD-1 inhibition may improve ONB management56. ONB research has underscored the olfactory cancer’s genetic and cytogenetic heterogeneity. Considering the rarity of ONB, a multi-institutional investigational approach to further genetic, epigenetic, and molecular stratification, and similarly, to therapeutic discovery, will facilitate a more comprehensive understanding of this malignancy for patients’ benefit. In fact, a phase II immunotherapy trial specifically for recurrent or metastatic ONB, Bintrafusp Alfa in Recurrent/Metastatic Olfactory Neuroblastoma (BARON), (NCT05012098), was recently approved at the National Institutes of Health73.

Conclusion

Studies reviewed here detail promising results of the most current and novel characterization of SNUC, ITAC, and ONB genetic and molecular landscapes, which have informed ongoing therapeutic discovery. With continued multi-institutional efforts, the field of sinonasal tumor research will achieve higher disease control and improved treatment outcomes for patients afflicted with these rare cancers.

Key Bullet Points.

  • Sinonasal tumors are rare and understudied, but recent advancements have enabled improved genetic and molecular classification with progress in targeted treatment development.

  • The improved genetic characterization of SNUC tumors by the presence of SMARCB1 or IDH2 mutations, along with efforts to establish robust clinical therapeutic sequence guidelines, have contributed to a better understanding of the disease and its management.

  • Work aimed at more granular characterization of ITAC genetic subgroups and discovery of somatic mutations targetable by FDA approved therapies is advancing the field of ITAC tumor research.

  • Immune checkpoint inhibition may unveil an avenue of promising treatment options for sinonasal malignancies.

Acknowledgements

The authors thank Dr. Carter Van Waes and Dr. Clint Allen for critical review of the manuscript.

Financial Support and Sponsorship

This research was supported (in part) by the Intramural Research Program of the NIH, NIDCD. This research was made possible through the NIH Medical Research Scholars Program, a public-private partnership supported jointly by the NIH and contributions to the NIH from the Doris Duke Charitable Foundation (DDCF Grant #2014194), the American Association for Dental Research, the Colgate-Palmolive Company, Genentech, Elsevier, and other private donors.

Conflicts of Interest

N. London receives research funding from Merck, holds stock in Navigen Pharmaceuticals, and was a consultant for Cooltech Inc., none of which are relevant to the present manuscript. All other authors declare no competing interests.

Competing interests:

N. London receives research funding from Merck, holds stock in Navigen Pharmaceuticals, and was a consultant for Cooltech Inc., none of which are relevant to the present manuscript. All other authors declare no competing interests.

References

  • 1.Hermsen MA, Riobello C, García-Marín R, et al. Translational genomics of sinonasal cancers. Semin Cancer Biol 2020;61:101–9. [DOI] [PubMed] [Google Scholar]
  • 2.Turner JH, Reh DD. Incidence and survival in patients with sinonasal cancer: a historical analysis of population-based data. Head Neck 2012;34(6):877–85. [DOI] [PubMed] [Google Scholar]
  • 3.Bossi P, Hermsen M, Lechner M, Franchi A. Precision medicine in rare tumors and the need for multicenter trials and international collaboratives: Sinonasal cancer as paradigm. Oral Oncol 2020;109:104737. [DOI] [PubMed] [Google Scholar]
  • 4.Llorente JL, López F, Suárez C, Hermsen MA. Sinonasal carcinoma: clinical, pathological, genetic and therapeutic advances. Nat Rev Clin Oncol 2014;11(8):460–72. [DOI] [PubMed] [Google Scholar]
  • 5.Costales M, López-Hernández A, García-Inclán C, et al. Gene Methylation Profiling in Sinonasal Adenocarcinoma and Squamous Cell Carcinoma. Otolaryngol--Head Neck Surg Off J Am Acad Otolaryngol-Head Neck Surg 2016;155(5):808–15. [DOI] [PubMed] [Google Scholar]
  • 6.Bell D, Bell A, Ferrarotto R, et al. High-grade sinonasal carcinomas and surveillance of differential expression in immune related transcriptome. Ann Diagn Pathol 2020;49:151622. [DOI] [PubMed] [Google Scholar]
  • 7.Martínez JG, Pérez-Escuredo J, López F, et al. Microsatellite instability analysis of sinonasal carcinomas. Otolaryngol--Head Neck Surg Off J Am Acad Otolaryngol-Head Neck Surg 2009;140(1):55–60. [DOI] [PubMed] [Google Scholar]
  • 8.Riobello C, Vivanco B, Reda S, et al. Programmed death ligand-1 expression as immunotherapeutic target in sinonasal cancer. Head Neck 2018;40(4):818–27. [DOI] [PubMed] [Google Scholar]
  • 9.Tyler MA, Holmes B, Patel ZM. Oncologic management of sinonasal undifferentiated carcinoma. Curr Opin Otolaryngol Head Neck Surg 2019;27(1):59–66. [DOI] [PubMed] [Google Scholar]
  • 10.Lewis JS, Bishop JA, Gillison M, Westra WH, Yarbrough WG. Sinonasal undifferentiated carcinoma. WHO Classif Head Neck Tumours 2017;18–20. [Google Scholar]
  • 11.Chambers KJ, Lehmann AE, Remenschneider A, et al. Incidence and survival patterns of sinonasal undifferentiated carcinoma in the United States. J Neurol Surg Part B Skull Base 2015;76(2):94–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Abdelmeguid AS, Bell D, Hanna EY. Sinonasal Undifferentiated Carcinoma. Curr Oncol Rep 2019;21(3):26. [DOI] [PubMed] [Google Scholar]
  • 13.Al-Mamgani A, van Rooij P, Mehilal R, Tans L, Levendag PC. Combined-modality treatment improved outcome in sinonasal undifferentiated carcinoma: single-institutional experience of 21 patients and review of the literature. Eur Arch Oto-Rhino-Laryngol Off J Eur Fed Oto-Rhino-Laryngol Soc EUFOS Affil Ger Soc Oto-Rhino-Laryngol - Head Neck Surg 2013;270(1):293–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gray ST, Herr MW, Sethi RKV, et al. Treatment outcomes and prognostic factors, including human papillomavirus, for sinonasal undifferentiated carcinoma: a retrospective review. Head Neck 2015;37(3):366–74. [DOI] [PubMed] [Google Scholar]
  • 15.Musy PY, Reibel JF, Levine PA. Sinonasal undifferentiated carcinoma: the search for a better outcome. The Laryngoscope 2002;112(8 Pt 1):1450–5. [DOI] [PubMed] [Google Scholar]
  • 16.Reiersen DA, Pahilan ME, Devaiah AK. Meta-analysis of Treatment Outcomes for Sinonasal Undifferentiated Carcinoma. Otolaryngol Neck Surg 2012;147(1):7–14. [DOI] [PubMed] [Google Scholar]
  • 17. Amit M, Abdelmeguid AS, Watcherporn T, et al. Induction Chemotherapy Response as a Guide for Treatment Optimization in Sinonasal Undifferentiated Carcinoma. J Clin Oncol 2019;37(6):504–12. This landmark study contains the largest number of treatment-naïve SNUC patients to date and defined the role for induction chemotherapy in the treatment of SNUC. In patients with a favorable IC response, improved survival was achieved with definitive chemotherapy and radiation, whereas in those without an IC response, surgery proved the preferred method for disease control.
  • 18.Wadsworth B, Bumpous JM, Martin AW, Nowacki MR, Jenson AB, Farghaly H. Expression of p16 in Sinonasal Undifferentiated Carcinoma (SNUC) Without Associated Human Papillomavirus (HPV). Head Neck Pathol 2011;5(4):349–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Glöss S, Jurmeister P, Thieme A, et al. IDH2 R172 Mutations Across Poorly Differentiated Sinonasal Tract Malignancies: Forty Molecularly Homogenous and Histologically Variable Cases With Favorable Outcome. Am J Surg Pathol 2021;45(9):1190–204. This study compared IDH2-mutated sinonasal tumors to IDH2 wild-type sinonasal tumors using light microscopy, immunohistochemistry, and genome-wide DNA methylation. The outcomes highlight the distinct influence IDH2 mutation status has on sinonasal tumor biology.
  • 20. Chitguppi C, Rabinowitz MR, Johnson J, et al. Loss of SMARCB1 Expression Confers Poor Prognosis to Sinonasal Undifferentiated Carcinoma. J Neurol Surg Part B Skull Base 2020;81(6):610–9. This study showed that SMARCB1, a known tumor suppressor gene and core subunit of the SWI/SNF chromatin remodeling complex, deficiency portends a worse prognosis in SNUC. Its findings suggest a novel SNUC sub-classification based on the expression of SMARCB1.
  • 21.Takahashi Y, Kupferman ME, Bell D, et al. Establishment and characterization of novel cell lines from sinonasal undifferentiated carcinoma. Clin Cancer Res Off J Am Assoc Cancer Res 2012;18(22):6178–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Takahashi Y, Lee J, Pickering C, et al. Human epidermal growth factor receptor 2/neu as a novel therapeutic target in sinonasal undifferentiated carcinoma. Head Neck 2016;38 Suppl 1:E1926–1934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ryan Q, Ibrahim A, Cohen MH, et al. FDA drug approval summary: lapatinib in combination with capecitabine for previously treated metastatic breast cancer that overexpresses HER-2. The Oncologist 2008;13(10):1114–9. [DOI] [PubMed] [Google Scholar]
  • 24.López-Hernández A, Vivanco B, Franchi A, et al. Genetic profiling of poorly differentiated sinonasal tumours. Sci Rep 2018;8(1):3998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Takahashi Y, Gleber-Netto FO, Bell D, et al. Identification of novel diagnostic markers for sinonasal undifferentiated carcinoma. Head Neck 2019;41(8):2688–95. [DOI] [PubMed] [Google Scholar]
  • 26.Takahashi Y, Gleber-Netto FO, Bell D, et al. Identification of markers predictive for response to induction chemotherapy in patients with sinonasal undifferentiated carcinoma. Oral Oncol 2019;97:56–61. [DOI] [PubMed] [Google Scholar]
  • 27.Jo VY, Chau NG, Hornick JL, Krane JF, Sholl LM. Recurrent IDH2 R172X mutations in sinonasal undifferentiated carcinoma. Mod Pathol Off J U S Can Acad Pathol Inc 2017;30(5):650–9. [DOI] [PubMed] [Google Scholar]
  • 28.Dogan S, Chute DJ, Xu B, et al. Frequent IDH2 R172 mutations in undifferentiated and poorly-differentiated sinonasal carcinomas. J Pathol 2017;242(4):400–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Stein EM, DiNardo CD, Fathi AT, et al. Molecular remission and response patterns in patients with mutant-IDH2 acute myeloid leukemia treated with enasidenib. Blood 2019;133(7):676–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lu C, Ward PS, Kapoor GS, et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 2012;483(7390):474–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Dogan S, Frosina D, Fayad M, et al. The role of a monoclonal antibody 11C8B1 as a diagnostic marker of IDH2 -mutated sinonasal undifferentiated carcinoma. Mod Pathol 2019;32(2):205–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Riobello C, López-Hernández A, Cabal VN, et al. IDH2 Mutation Analysis in Undifferentiated and Poorly Differentiated Sinonasal Carcinomas for Diagnosis and Clinical Management. Am J Surg Pathol 2020;44(3):396–405. This study evaluated IDH2 mutations’ specificity to poorly differentiated sinonasal carcinomas and reported that 31% of SNUC samples contained IDH2 mutations, with IDH2-mutant cases possessing a higher disease specific survival. These results further affirmed the notion that IDH2-mutated sinonasal tumors may represent a unique subtype with distinct behavioral patterns.
  • 33.Bonnecaze G de, Verillaud B, Chaltiel L, et al. Clinical characteristics and prognostic factors of sinonasal undifferentiated carcinoma: a multicenter study. Int Forum Allergy Rhinol 2018;8(9):1065–72. [DOI] [PubMed] [Google Scholar]
  • 34.Khoury T, Jang D, Carrau R, Ready N, Barak I, Hachem RA. Role of induction chemotherapy in sinonasal malignancies: a systematic review. Int Forum Allergy Rhinol 2019;9(2):212–9. [DOI] [PubMed] [Google Scholar]
  • 35.Watanabe S, Honma Y, Murakami N, et al. Induction chemotherapy with docetaxel, cisplatin and fluorouracil followed by concurrent chemoradiotherapy for unresectable sinonasal undifferentiated carcinoma: Two cases of report. World J Clin Cases 2019;7(6):765–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gamez ME, Lal D, Halyard MY, et al. Outcomes and patterns of failure for sinonasal undifferentiated carcinoma (SNUC): The Mayo Clinic Experience. Head Neck 2017;39(9):1819–24. [DOI] [PubMed] [Google Scholar]
  • 37.London NR, Mohyeldin A, Daoud G, et al. Sinonasal undifferentiated carcinoma: Institutional trend toward induction chemotherapy followed by definitive chemoradiation. Head Neck 2020;42(11):3197–205. [DOI] [PubMed] [Google Scholar]
  • 38.National Cancer Institute. Phase II trial of induction therapy with docetaxel, cisplatin and fluorouracil in previously untreated patients with locally advanced squamous cell car cinoma and/or poorly differentiated carcinoma of the nasal cavity and/or paranasal sinuses. NCT00707473. [Internet]. [cited 2021 Aug 18];Available from: https://clinicaltrials.gov/ct2/show/NCT00707473
  • 39.Llorente JL, Pérez-Escuredo J, Alvarez-Marcos C, Suárez C, Hermsen M. Genetic and clinical aspects of wood dust related intestinal-type sinonasal adenocarcinoma: a review. Eur Arch Oto-Rhino-Laryngol Off J Eur Fed Oto-Rhino-Laryngol Soc EUFOS Affil Ger Soc Oto-Rhino-Laryngol - Head Neck Surg 2009;266(1):1–7. [DOI] [PubMed] [Google Scholar]
  • 40.Pérez-Escuredo J, García Martínez J, García-Inclán C, et al. Establishment and genetic characterization of an immortal tumor cell line derived from intestinal-type sinonasal adenocarcinoma. Cell Oncol Dordr 2011;34(1):23–31. [DOI] [PubMed] [Google Scholar]
  • 41.Vivanco Allende B, Perez-Escuredo J, Fuentes Martínez N, Fresno Forcelledo MF, Llorente Pendás JL, Hermsen M. Intestinal-type sinonasal adenocarcinomas. Immunohistochemical profile of 66 cases. Acta Otorrinolaringol Esp 2013;64(2):115–23. [DOI] [PubMed] [Google Scholar]
  • 42.Vivanco B, Llorente JL, Perez-Escuredo J, Alvarez Marcos C, Fresno MF, Hermsen MA. Benign lesions in mucosa adjacent to intestinal-type sinonasal adenocarcinoma. Pathol Res Int 2011;2011:230147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ben-Neriah Y, Karin M. Inflammation meets cancer, with NF-κB as the matchmaker. Nat Immunol 2011;12(8):715–23. [DOI] [PubMed] [Google Scholar]
  • 44.Holmila R, Cyr D, Luce D, et al. COX-2 and p53 in human sinonasal cancer: COX-2 expression is associated with adenocarcinoma histology and wood-dust exposure. Int J Cancer J Int Cancer 2008;122(9):2154–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Pérez-Escuredo J, Martínez JG, Vivanco B, et al. Wood dust-related mutational profile of TP53 in intestinal-type sinonasal adenocarcinoma. Hum Pathol 2012;43(11):1894–901. [DOI] [PubMed] [Google Scholar]
  • 46.Pacheco E, Llorente JL, López-Hernández A, et al. Absence of chromosomal translocations and protein expression of ALK in sinonasal adenocarcinomas. Acta Otorrinolaringol Esp 2017;68(1):9–14. [DOI] [PubMed] [Google Scholar]
  • 47.López-Hernández A, Pérez-Escuredo J, Vivanco B, et al. Genomic profiling of intestinal-type sinonasal adenocarcinoma reveals subgroups of patients with distinct clinical outcomes. Head Neck 2018;40(2):259–73. [DOI] [PubMed] [Google Scholar]
  • 48.Perez-Escuredo J, Lopez-Hernandez A, Costales M, et al. Recurrent DNA copy number alterations in intestinal-type sinonasal adenocarcinoma. Rhinology 2016;54(3):278–86. [DOI] [PubMed] [Google Scholar]
  • 49.García-Inclán C, López F, Pérez-Escuredo J, et al. EGFR status and KRAS/BRAF mutations in intestinal-type sinonasal adenocarcinomas. Cell Oncol Dordr 2012;35(6):443–50. [DOI] [PubMed] [Google Scholar]
  • 50.Hermsen MA, Llorente JL, Pérez-Escuredo J, et al. Genome-wide analysis of genetic changes in intestinal-type sinonasal adenocarcinoma. Head Neck 2009;31(3):290–7. [DOI] [PubMed] [Google Scholar]
  • 51.Rodrigo JP, García-Pedrero JM, Llorente JL, et al. Down-regulation of annexin A1 and A2 protein expression in intestinal-type sinonasal adenocarcinomas. Hum Pathol 2011;42(1):88–94. [DOI] [PubMed] [Google Scholar]
  • 52.López F, García Inclán C, Pérez-Escuredo J, et al. KRAS and BRAF mutations in sinonasal cancer. Oral Oncol 2012;48(8):692–7. [DOI] [PubMed] [Google Scholar]
  • 53. Sánchez-Fernández P, Riobello C, Costales M, et al. Next-generation sequencing for identification of actionable gene mutations in intestinal-type sinonasal adenocarcinoma. Sci Rep 2021;11(1):2247. Using next generation sequencing, this study found one or more potentially actionable somatic mutations in 20 of 27 ITAC cases, with eight of these representing biomarkers for existing FDA-approved targeted therapies. This study represents a promising avenue to pursue in the development of personalized therapy for ITAC.
  • 54. García-Marín R, Reda S, Riobello C, et al. CD8+ Tumour-Infiltrating Lymphocytes and Tumour Microenvironment Immune Types as Biomarkers for Immunotherapy in Sinonasal Intestinal-Type Adenocarcinoma. Vaccines 2020;8(2):202. This study evaluated tumor infiltrating lymphocytes and PD-L1 expression in ITAC as biomarkers for immunotherapy. It concluded that subsets of ITAC, especially papillary and colonic subtypes, may represent candidates for immune checkpoint inhibition.
  • 55.Kaur Raman, Izumchenko E, Blakaj D, et al. The genomics and epigenetics of olfactory neuroblastoma: A systematic review - Kaur - - Laryngoscope Investigative Otolaryngology - Wiley Online Library [Internet]. [cited 2021 Jul 2];Available from: 10.1002/lio2.597 [DOI] [PMC free article] [PubMed]
  • 56. London NR, Rooper LM, Bishop JA, et al. Expression of Programmed Cell Death Ligand 1 and Associated Lymphocyte Infiltration in Olfactory Neuroblastoma. World Neurosurg 2020;135:e187–93. This study showed PD-L1 expression in 40% of primary ONB and in 75% of cervical metastases derived from a primary PD-L1 negative ONB, as well as the presence of increased PD-1+ and CD8+ lymphocytes within tumor and stroma of PD-L1+ tumors. This analysis suggests that PD-L1/PD-1 inhibition may improve ONB management, highlighting the potential for immune checkpoint pathway inhibition in ONB.
  • 57.Berger L, Luc G, Richard D. L’esthesioneuroepitheliome olfatif. Bull Assoc Fr Etud Cancer 1924;13:410–21. [Google Scholar]
  • 58.Su SY, Bell D, Hanna EY. Esthesioneuroblastoma, Neuroendocrine Carcinoma, and Sinonasal Undifferentiated Carcinoma: Differentiation in Diagnosis and Treatment. Int Arch Otorhinolaryngol 2014;18(Suppl 2):S149–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ow TJ, Bell D, Kupferman ME, Demonte F, Hanna EY. Esthesioneuroblastoma. Neurosurg Clin N Am 2013;24(1):51–65. [DOI] [PubMed] [Google Scholar]
  • 60.Song X, Wang J, Wang S, Yan L, Li Y. Prognostic factors and outcomes of multimodality treatment in olfactory neuroblastoma. Oral Oncol 2020;103:104618. [DOI] [PubMed] [Google Scholar]
  • 61.Ow TJ, Hanna EY, Roberts DB, et al. Optimization of long-term outcomes for patients with esthesioneuroblastoma. Head Neck 2014;36(4):524–30. [DOI] [PubMed] [Google Scholar]
  • 62.Classe M, Yao H, Mouawad R, et al. Integrated Multi-omic Analysis of Esthesioneuroblastomas Identifies Two Subgroups Linked to Cell Ontogeny. Cell Rep 2018;25(3):811–821.e5. [DOI] [PubMed] [Google Scholar]
  • 63.Classe M, Burgess A, El Zein S, et al. Evaluating the prognostic potential of the Ki67 proliferation index and tumour-infiltrating lymphocytes in olfactory neuroblastoma. Histopathology 2019;75(6):853–64. [DOI] [PubMed] [Google Scholar]
  • 64.Gallagher KK, Spector ME, Pepper J-P, McKean EL, Marentette LJ, McHugh JB. Esthesioneuroblastoma: updating histologic grading as it relates to prognosis. Ann Otol Rhinol Laryngol 2014;123(5):353–8. [DOI] [PubMed] [Google Scholar]
  • 65.Gay LM, Kim S, Fedorchak K, et al. Comprehensive Genomic Profiling of Esthesioneuroblastoma Reveals Additional Treatment Options. The Oncologist 2017;22(7):834–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Bockmühl U, You X, Pacyna-Gengelbach M, Arps H, Draf W, Petersen I. CGH pattern of esthesioneuroblastoma and their metastases. Brain Pathol Zurich Switz 2004;14(2):158–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Guled M, Myllykangas S, Frierson H, Mills S, Knuutila S, Stelow E. Array comparative genomic hybridization analysis of olfactory neuroblastoma - PubMed. Mod Pathol 2008;21(6):770–8. [DOI] [PubMed] [Google Scholar]
  • 68. Cracolici V, Wang EW, Gardner PA, et al. SSTR2 Expression in Olfactory Neuroblastoma: Clinical and Therapeutic Implications. Head Neck Pathol 2021; This study reported on uniform somatostatin receptor 2 (SSTR2) expression in ONB. A finding of significantly greater SSTR2 staining in ONB than in histologically related neoplasms suggests that SSTR2 represents a likely diagnostically important ONB marker and provides the basis for somatostatin analog therapy, which has been applied to metastatic ONB.
  • 69.Wang L, Tang K, Zhang Q, et al. Somatostatin receptor-based molecular imaging and therapy for neuroendocrine tumors. BioMed Res Int 2013;2013:102819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Savelli G, Bartolomei M, Bignardi M. Somatostatin receptors imaging and therapy in a patient affected by esthesioneuroblastoma with meningeal metastases. A classic example of theranostic approach. J Neurooncol 2016;127(3):617–9. [DOI] [PubMed] [Google Scholar]
  • 71.Makis W, McCann K, McEwan AJB. Esthesioneuroblastoma (olfactory neuroblastoma) treated with 111In-octreotide and 177Lu-DOTATATE PRRT. Clin Nucl Med 2015;40(4):317–21. [DOI] [PubMed] [Google Scholar]
  • 72.Hasan OK, Ravi Kumar AS, Kong G, et al. Efficacy of Peptide Receptor Radionuclide Therapy for Esthesioneuroblastoma. J Nucl Med Off Publ Soc Nucl Med 2020;61(9):1326–30. [DOI] [PubMed] [Google Scholar]
  • 73.National Cancer Institute (NCI). Phase 2 Study of Bintrafusp Alfa in Recurrent/Metastatic Olfactory Neuroblastoma (BARON). [Internet]. clinicaltrials.gov; 2021. [cited 2021 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT05012098

RESOURCES