Skip to main content
Laryngoscope Investigative Otolaryngology logoLink to Laryngoscope Investigative Otolaryngology
. 2021 Jun 7;6(4):721–728. doi: 10.1002/lio2.597

The genomics and epigenetics of olfactory neuroblastoma: A systematic review

Raman Preet Kaur 1, Evgeny Izumchenko 2, Dukagjin M Blakaj 3, Nikol Mladkova 3, Matt Lechner 4, Thomas L Beaumont 5, Charalampos S Floudas 6, Gary L Gallia 1,7, Nyall R London Jr 1,7,8,
PMCID: PMC8356883  PMID: 34401496

Abstract

Background

Olfactory neuroblastoma (ONB) or esthesioneuroblastoma (ENB) is a rare malignancy of the nasal cavity believed to arise from the olfactory epithelium. The goal of this study was to systematically review the genomics, epigenetics, and cytogenetics of ONB and to understand the potential clinical implications of these studies.

Methods

A systematic literature review was performed for articles published before May 2020 using Cochrane, Embase, Pubmed, and Scopus databases. Inclusion criteria included genomics, cytogenetics, and epigenetics studies on ONB. Exclusion criteria included studies not in English or systematic reviews. Articles and abstracts were reviewed by two independent reviewers to reduce bias during article selection and synthesis of results. Of the 36 studies included in this review, 24 were research articles and 12 were abstracts.

Results

Although recurrent mutations among ONB tumors are uncommon, alterations in TP53, DMD, PIK3CA, NF1, CDKN2A, CDKN2C, CTNNB1, EGFR, APC, cKIT, cMET, PDGFRA, CDH1, FH, SMAD4, FGFR3 and IDH2 genes have been reported in several recent studies. In addition, cytogenetic studies revealed that the landscape of chromosomal aberrations varies widely amongst ONB tumors.

Conclusions

The rare character of ONB has limited the sample size available for cytogenetic, genomic, and epigenetic studies and contributes to the limitations of this systematic review. Comprehensive genomic and epigenomic studies with larger cohorts are warranted to validate the initial reports summarized in this review and to identify potential therapeutic targets for ONB.

Keywords: cytogenetics, epigenetics, genomics, olfactory neuroblastoma, tumorigenesis

1. INTRODUCTION

Olfactory neuroblastoma (ONB) or esthesioneuroblastoma (ENB) is a rare malignancy of the nasal cavity that likely arises from the olfactory neuro‐epithelium.1 An olfactory epithelial origin is suggested by the tumor anatomical location, morphological features, and characteristic gene expression patterns.2 ONB was first described in 1924 and named “L'esthesioneuroepitheliome olfactif.”3 Due to lack of information on its histological origin, additional names have since been used including olfactory neurocytoma and others.4, 5 The incidence of ONB is 0.4/million/year, accounting for approximately 2%‐3% of nasal cavity tumors.6, 7 To date, no specific risk factors have been identified for ONB.8 Hyams histologic grading and Kadish, modified Kadish, and Dulguerov staging are traditionally used to clinically characterize the tumor and aid in treatment planning.

The rare character of ONB has limited the sample size available for cytogenetic, genomic, and epigenetic studies. Although multiple manuscripts have identified complex genetic and cytogenetic patterns in ONB, only a subset of reported genomic aberrations were consistent across different studies. While these inconsistencies limit the generalizable conclusions regarding the mutational landscape of this disease, it may indicate that ONB is defined by a set of heterogeneous alterations. Previous cytogenetic studies have reported that deletion on chromosome 11 and gain on chromosome 1p were associated with poor prognosis in ONB.9 In addition to cytogenetic analysis, various sequencing approaches such as pyrosequencing, and more recently next generation sequencing (NGS), have also been used to explore genomic alterations in patients with ONB. Although the catalogue of somatic mutations in cancer (COSMIC) database as well as other publications in the field of ONB report alterations in TP53, EGFR, CTNBB1, KIT, RET, APC, FGFR2, KDR, PDGFRA, SMAD4, MET, CDH1, SUZ12, PPP6C, IKZF1, TGFBR2, CARD11 and CDKN2C genes,10, 11, 12 the fraction of ONB with mutations in these genes that present opportunities for targeted therapy is estimated to be up to 50%.10

The goal of this systematic review is to summarize the available ONB cytogenetics, genomics, and epigenetics literature and to suggest potential targetable pathways for precision therapeutics.

2. MATERIALS AND METHODS

2.1. Search of publications

A systematic review was conducted following the guidelines of the preferred reporting items for systematic reviews and meta‐analysis (PRISMA) statement. We conducted a systematic literature search using Cochrane, Embase, Pubmed, and Scopus databases. Articles published before May 2020 were included in the review. The key words used for the search included Esthesioneuroblastoma, Olfactory Neuroblastoma, nasal glial heterotopia, nasal glioma, neuroblastoma, nose glioma, olfactory esthesioneuroblastoma, olfactory neuroblastomas, paranasal sinus‐nasal cavity, nasal glial heterotopia, olfactory, genomics, genetics, epigenomic, pharmacogenomics, phylogenomic, epigenetic, epigenetics, miRNA, histone, methylation, acetylation, and epigenesis.

2.2. Selection criteria

PubMed, Cochrane, Embase, and Scopus search results were selected as they provided the most comprehensive literature search for the topic. Duplicate articles were removed using Covidence. The title and abstract of all potentially relevant studies were screened for their contents ensuring adherence to the following inclusion and exclusion criteria for the systematic review.

Inclusion criteria:

  1. Cytogenetic studies on clinical samples for ONB patients

  2. Genomics studies on clinical samples of ONB patients

  3. Epigenetics studies on clinical samples of ONB patients

Exclusion criteria:

  1. Studies without full text

  2. Studies not published in English

  3. Review articles

2.3. Data extraction

Data was extracted from all the eligible studies for systematic review. Experimental data involving study design, sample type, and experimental technique was collected from the research articles exploring genetic make‐up of patients using various genomics and cytogenetic techniques. Cytogenetic and genomic alterations as well as epigenetic data were extracted from all previous studies.

2.4. Quality assessment

Two independent reviewers critically assessed the content of all the studies included in this systematic review. A total of 115 abstracts were categorized into three groups: “yes,” “no,” and “maybe”; where “yes” indicated abstracts deciphering genetic and cytogenetic changes, “no” stood for abstracts depicting that study was a review article or was not published in English, whereas “maybe” defined studies focused on head and neck cancers or other cancers of the sinonasal cavity. Among the 115 screened abstracts, 79 were found to be unrelated and were excluded. The full texts of the remaining 36 studies were screened and marked as “yes” and “no.” This time, “yes” indicated studies that explored genetics and cytogenetics of ONB tumors, and “no” was assigned to the studies focusing on head and neck as well as sinonasal cancers that did not include ONB samples. Of the 36 studies, 24 research articles and 12 abstracts were included in this review. These 24 studies were directly related to ONB. The 12 abstracts included in our overview were published in conferences and meetings and explored the genetic and cytogenetic makeup of ONB patients.

3. RESULTS

3.1. Cytogenetic landscape of ONB patients

Cytogenetic changes in ONB patients were studied by using different techniques but were limited in number. Initial research hypothesized that a (11;22) (q24;q12) translocation suggested that ONB was related to Ewing Sarcoma.13, 14 However, a later study analyzing 5 ONB tumors could not identify the EWS gene rearrangement by FISH or the Ewing sarcoma‐associated MIC2 antigen by immunohistochemistry.15 Furthermore, an additional study that performed RT‐PCR on two ONB specimens did not identify EWS‐FLI1 or EWS‐ERG translocations,16 suggesting that ONB is not a member of the primitive peripheral neuroectodermal tumor‐Ewing's group.2, 14, 15, 16

In a later study of 22 patients, comparative genomic hybridization identified deletion of chromosome 3p and high copy number amplifications on 17q in 90%‐100% of tumors analyzed.9 However, despite the high frequency of these chromosomal changes, cytogenetic studies have ultimately revealed that ONB tumors are cytogenetically complex and the landscape of chromosomal aberrations varies widely. The inconclusive reports may stem from the very small number of cases analyzed, with many of the cytogenetic studies having been carried out on only a single patient. The list of detected chromosomal aberrations is summarized in Table 1. While these cytogenetic alterations can potentially be used as predictive and prognostic biomarkers, their biological relevance remains to be validated.

TABLE 1.

Summary of reported cytogenetic alterations

Author Type of alteration # of patients Reported alterations
Bockmühl et al, 20049 Loss/gain—frequency of alteration > 50% of cases or pronounced deletion/gain defined by ratio profile that exceeded the threshold of 0.5 12

Loss: 1p21‐31, 1q24‐q32, 2q22‐q32, 3p/q, 3p12‐p14, 4p/q, 4p13‐p15, 5p14, 5q, 6q14‐q23, 9p, 9q22‐q33, 10p/q, 10q26, 12p11.2‐p12, 12q21, 13q, 13q21‐q23, 18q, 21q21

Gain: 1p34, 1q12, 1q23‐q31, 7p21, 7q11.2, 7q31, 9p23‐p24, 11q13, 14q, 14q32.2, 16p11.2, 16q, 16p13.3, 17p13, 17q11‐q22, 17q12, 17q21‐q24, 17q24‐q25, 17q25, 19, 19p/q, 20p, 20q, 20q13, 22q11.2, 22q13

Loss associated with worse prognosis or metastasis: 4p/q, 5p/q, 6q, 7q31‐q32, 9p, 11p/q, 11p12‐p14, 15q21

Gain associated with worse prognosis or metastasis: 1p32‐34, 1q12, 2p22‐p24, 8q, 20q

Castañeda et al, 199140 Ploidy 1 Near pseudotetraploid, chromosome 5 present in multiples of eight
Guled et al, 200824 Loss/gain—frequency of alteration > 20% of cases 13

Loss: 2q31.1, 2q33.3, 2q37.1, 4p13, 5q31.2, 6p12.3, 6p21.33, 6p22.1, 6q16.3, 6q21, 6q22.1, 15q11.2‐q24.1, 15q13.1, 18q12.2‐q12.3, 19q12, 19q13.11, 19q13.32, 19q13.43, 22q11.23, 22q12.1, 22q11.1‐q11.21, Xp/q

Gain: 1p36.31, 1p35.3, 4p12‐p15.31, 4p16.2‐p16.3, 4q12, 4q21.22‐q22.1, 4q27‐q35.2, 5q34, 5q35.1‐q35.3, 6p12.3, 7q11.23, 7q21.11, 9p13.3, 10p12.31, 12q23.1, 12q24.31, 13q, 13q14.2‐14.3, 13q31.1, 13q34, 15q13.3, 16q12.1, 20p/q, 20p13.3‐p12.2, 20q11.21‐q11.23, 20q13.32‐q13.33, 21q, 22q12.1, Xp/q

Holland et al, 200741 Loss/gain—G‐banding 1

Loss: 1p12‐p21, 1p22‐p32, 1p31‐p33, 2q31‐q33, 2q37, 3p11‐p13, 3p12‐p14, 3p25, 3q25, 3q26, 6p21, 6q12‐q14, 6q22‐q24, 10q26, 11q23, 15q26, 20q11.1‐q12, 21q22, 22q13

Gain: 1q25‐q32, 1q25‐q41, 16p13.3, 16q13‐q22, 17p12, 17p13, 17q25

Holland et al, 200741 Loss/gain—SNP array karyotyping 1

Loss: 2q37.3, 3p21.3, 3q27.2, 4p12, 4q31.3, 7q36.1, 8q24.3, 10p26.11, 11p11.2, 12q24.31, 14q32.33, 16p11.2, 21q22.11

Gain: 2q14.3, 3p21.3, 6q25.3, 6q27, 7q11.21, 7q11.23, 7q36.1, 7q36.3, 8p11.21, 9p13.3, 10q11.23, 11p14.1, 11q15.3, 11q23.3, 11q24.3, 13q12.11, 13.q33.3, 13q34, 14q32.31, 15q12, 15q13.1, 16p11.2, 16p13.11, 17q12, 17q21.31, 17q25.3, 19q13.42, 20q13.31, 21q22.3, 22q13.1, 22q13.31, 22q13.33

Jin et al, 199542 Ploidy—G‐banding of short‐term culture 1 Hyperdiploid karyotype with unbalanced structural rearrangements
Kristensen et al, 199143 Ploidy 1 polyploidy, aneuploidy and marker chromosomes
Lopez‐Hernandez et al, 201844 Loss/gain 11

Loss: 1p, 2p25.1‐14.3, 3, 4, 5q, 13q14.3‐q21.1, 16p12.3, 22q12.2‐12.3

Gain: 1q, 3q, 4p14‐13, 4q13‐31, 6p, 7p, 7q11.23‐33, 9p24, 11p14.3‐p12, 11q13.4‐q14.2, 12p, 12q, 14q, 16q23.3‐24.3, 17q22‐25, 18q, 20

Mezzelani et al, 199915 Gain 5 Gain: (1/5 patients) 8p/q
Riazimand et al, 200245 Loss/gain 3

Loss: (3/3 patients): 4q, (2/3 patients): 13q, (1/3 patients): 6p

Gain: (3/3 patients): 8q24.1, 15q25, 19p/q, 22q, (2/3 patients): 1p32, 9q34.1, (1/3 patients): 10q24.3

Sorensen et al, 199613 Translocation 6 + 2 cell lines t(11;22) (q24;q12)
Szymas et al, 199746 Loss/gain 1

Loss: 5q, 16p/q, 18p/q, 17p, 19p/q, Xp/q

Gain: 1q, 4p/q, 8p/q, 11p/q, 14p/q, 17q

Valli et al, 201527 Loss/gain—frequency of alteration ≥ 3/11 samples 10 (11 samples)

Loss: Yp11.31, Yp11.2, Yq11.21‐q11.23

Gain: 2p/q, 5p/q, 6p/q, 7p/q, 11p/q, 13p/q, 14p/q, 15p/q, 16p/q, 17p/q, 18p/q, 19p/q, 20 p/q, 22p/q, Xp/q, Yp/q

VanDevanter et al, 199147 Gain—short term culture 1 Gain: 8p/q
Lazo de la Vega et al, 201721 Loss/gain 18

Loss: 1p/q, 3p/q, 8p/q, 12p/q

Gain: 5p/q, 7p/q, 11p/q, 20p/q

Weiss et al, 201248 Loss/gain/amplification 1

Loss: 5q15, 6 p25.1, 7p15.3, 7p21.3, 11q24.2, 19p12, 21q.1

Gain/amplification: 5p15.33, 7p13, 8p, 8q24.3, 9q22.31, 9q34.3, 16q22.1, 16q24.3

Whang‐Peng et al, 198749 Translocation, loss/gain 1 cell line

t(2;14)(p25;p12), t(8;17)(q12;p12), t(11;22)(q24;q12)

Loss: 18p/q

Gain: 12p/q, del(8)(q12)

3.2. Genetic landscape of ONB patients

Compared to cytogenetic approaches, exploring the genome by high throughput NGS technologies provides a more detailed view of the tumor genome at the nucleotide level. As ONB is rare, there are no large genomic studies to date, with the largest genomic study conducted in a cohort of 41 ONB specimens.10 Among the 41 tumors, 28 harbored genomic alterations. The study reported TP53 gene as the most commonly altered along with genomic alterations in PIK3CA, NF1, CDK2NA and CDKN2C genes. Genetic changes in TP53 were detected in the DNA binding domain of this tumor suppressor gene, and 17% of patients carried splice site, missense (P278S, R248W, G245C and P278R) and truncation (P190del) alterations. It was also observed that 7% of tumors in this study had alterations in PIK3CA (missense mutations at positions E545Q and E542K) and truncation alterations in NF1, CDKN2A, and CDKN2C genes.10 Another case report has also documented the presence of a TP53 missense mutation and CDKN2C loss‐of‐function alteration in a patient with ONB.17 Notably, the gain of function alteration detected in PIK3CA gene were located in the accessory domain, which has been suggested to be associated with substrate presentation according to the Single Nucleotide Polymorphism Database (dbSNP).

Gallia et al found deletions involving the dystrophin (DMD) locus in 12 of 14 (86%) of ONB tumors analyzed in their study.18 Alterations in DMD gene were detected in both exonic region as well as 5′UTR. DMD has been identified as a tumor suppressor in other tumor types.19 Interestingly, one of the two tumors that did not display alterations in the Xp21.1 locus, showed a somatic homozygous deletion in LAMA2, bringing the number of ONBs with deletions of genes involved in the development of muscular dystrophies to 13 of 14 (93%).18

Classe et al screened DNA extracted from 14 ONB samples and matched germline DNA from blood or adjacent normal tissue by whole exome sequencing and found that 21.4% of patients carried single nucleotide alterations in the TP53 gene, with 7.14% of these mutations reported to be stop‐gain, resulting in a premature transcription termination.12

Topcagic et al, have retrospectively analyzed 23 ONB patient samples by using targeted DNA sequencing, in situ hybridization and gene fusion assays.20 DNA sequencing of 15 patient samples was carried out; that included a panel of 46 genes in 10 cases, and an extended panel of 592 genes by NGS in 5 cases. In these 15 patients, the study detected alterations in TP53 (H214Y, c673‐1G>T and T155_V157del), CTNNB1 (S33_H36del), EGFR (Q276R and T572R), APC (A1474T), cKIT (G565V), cMET (L1321I), PDGFRA (V546L), CDH1 (D756Y), FH (K477dup) and SMAD4 (N468fs) genes.20 Among the 15 patients analyzed in this study, 20% exhibited missense mutation, single nucleotide variation or deletion in DNA binding domain of the TP53 gene.20

Lazo de la Vega et al, studied 20 ONB samples using targeted multiplexed PCR based NGS. While no recurrent alterations were detected in their study, copy number alterations affecting chromosomes 5, 7, 11 and 20 were identified in a subset of patients.21 In two tumors, co‐amplification of CCND1 at 11q13 and FGFR3 at 4p16 was observed, resulting in potential driver oncogenic event. CCND1 is a core cell cycle regulator that is frequently overexpressed in many solid tumors including head and neck cancers. Missense mutation V272M in DNA binding domain of TP53 gene was detected in one patient. PTEN gene exhibited nonsense mutation Y16X in one patient, resulting in a truncated PTEN protein which is unable to inhibit the PIK3/mTOR pathway.21 Although there was little overlap of genetic alterations identified by different studies, most of the genomic studies have reported the TP53 gene as frequently mutated in ONB patients, suggesting that a complex signaling network regulated by p53 can be a potential therapeutic target (Table 2).

TABLE 2.

Summary of genomic alterations

Author Type of alteration # of patients Genes
Cha et al, 201617 Mutations 1 TP53 (missense), CDKN2C (loss of function)
Classe et al, 201812 Insertion/deletion 27 CDKN2A, CTR9
Stop/gain RB1, TP53
Non‐synonymous SNV ARID1A, BRCA2, BRIP1, CHD7, DNMT3A, DNMT3B, EXO1, FZD9, GLI2, GLI3, HERC2, IDH2, KDM2B, KIT, KMT2A, KMT2C, KMT2D, LEF1, MSH3, NAA10, NUMA1, RBBP4, SMARCA4, SMARCC1, SMC2, TOPBP1, TP53, XRCC1, YWHAE
Gallia et al, 201818 Deletions 14 Dystrophin, LAMA2
Gay et al, 201710 Short variants, truncations, and rearrangements 41 ARID1A, ARID2, ATM, AXL‐ARHGEF fusion, BCOR, CDKN1B, CDKN2C, CTCF, CTNNB1, DAXX, IDH2, KDM5C, LRP1B deletion, NF1, NRAS, PBRM1, PIK3CA, PIK3R2, PTCH1, PTEN, SMARCA4, TET2, TNFAIP3, TP53, TSC1,
Focal copy number alterations

Amplification: BCL2L1, BCL2L2, CDK6, HGF, FGF14, GNAS, IRS2, KIT, MDM4, MYC, PIK3C2B, RICTOR

Loss: ARID2, CDKN2A, CDKN2B, CDKN2C, FAF1, MLH1, NF1, PTPRD, TP53, RB1,

Non focal amplifications ARFRP1, AURKA, FGF10, FGFR4, FLT4, GATA6, LYN, PDGFRB, RICTOR, SRC, TOP1, ZNF217,
Kim et al, 201750 Recurrent and/or pathogenic somatic mutations 6 ANKHD1, ARHGEF9, ATM, CNOT10, CSDM1, DCTN1, ITSN1, LMTK2, MACF1 (recurrently mutated), MAP2K1, MMRN2, MYOCD, RCE1, RORB, SDCCAG8, ZNF471
Inter‐chromosomal in‐frame fusion gene DHRS2‐GSTM2 (recurrent in two samples)
Topcagic et al, 201820 Mutations 15 APC, c‐KIT, CTNNB1, FH, SMAD4, TP53,
Variants of unknown significance CDH1, cMet, EGFR, PDGFRA,
Lazo de la Vega et al, 201721 Mutations 18 CTNNB1, KMT2A, KMT2C, PTEN, TP53
Copy number alterations

Loss: CDKN2A, CDKN2B

Gain: FGFR3, CCND1

Frameshift insertion DCC, RNF213
Wang et al, 201651 Missense mutations 1 EGFR, FGFR2, KDR, RET
Weiss et al, 201248 Insertion/deletions /gain/loss 1

Deletions: ARID4B, CCDC120, CYP4A22

Insertions: KCNA10, OBSCN

Gene mutations KDR, MAP4K2, MYC, NLRC4, SIN3B, TAOK2, TP53,

Several studies have also detected IDH2 gene alterations in ONB samples sequencing. However, the true frequency of IDH2 aberrations remains controversial. For example, Dogan et al, screened 9 ONB samples using targeted exome sequencing and none of these ONB samples showed alterations in IDH2.22, 23 In contrast, Classe et al, have detected R172 IDH2 mutation in 16.7% of ONB specimens,12 which were further validated by pyrosequencing. Gay et al, screened 41 patients and reported two missense mutations, R172S and R172T, in different patients each carrying one alteration.10 Various studies have reported short variants, truncations, rearrangements, focal copy number alterations and non‐focal number alterations in a variety of other genes, but at much lower frequency (Table 2).

3.3. Epigenetic landscape of ONB

Currently, epigenetic data on ONB are very limited and focus predominantly on DNA methylation. Capper et al, carried out a microarray‐based methylation profiling of 66 ONB samples and distinguished four unique subgroups within the histopathological diagnosis of ONB.11 A total of 64% of ONB tumors were used to define the “core ONB” genome‐wide methylation profile, while a total of 7 samples formed a second group with global hypermethylation features that were also associated with R172 IDH2 hotspot mutations in all cases. The third group consisted of only 4 tumors with a high level of overall methylation but absence CpG island methylation and lacking IDH1 or IDH2 mutations. The group was heterogeneous and did not fit any of the above inclusion criteria.

The unique methylome profile of ONB was further confirmed in a study by Dogan et al, in a series of sinonasal tumors that included ONB, using a DNA methylation microarray. While only 4 ONB samples were included, they formed a distinct cluster based on a semi‐supervised hierarchical clustering analysis.23

4. DISCUSSION

The current review systematically summarizes the genetic, cytogenetic, and epigenetic alterations that have been previously reported in ONB. Although heterogeneous chromosomal instability in ONB were predicted by various studies, more cytogenetic alterations were observed in high grade tumors in comparison to low grade malignancies,24 which is not surprising and has been previously observed in other cancers, such as cutaneous squamous cell carcinoma.25 Loss of 3p was found to be the most common chromosomal alteration among patients with ONB and this alteration has been reported to be associated with resistance to chemotherapy or radiotherapy in other settings.9, 26 Additional studies have predicted metastasis and poor prognosis in patients with deletion in chromosome 11 and gain on chromosome 1p.9, 24 While frequent cytogenetic alterations were detected in all chromosomes besides 8 and 20, Valli et al, reported that neither Kadish stage nor Hymans grade was significantly associated with any of the cytogenetic alterations detected in their study.27 One cytogenetic study that analyzed primary and recurrent malignancies, found that there were decreased trisomies and increased number of partial gains in relapsed tumor in comparison to the primary disease.27 Patients exhibiting aneuploidic alterations have been treated with energy and proteotoxic stress‐inducing compounds such as AICAR (5‐aminoimidazole‐4‐carboxamide riboside), 17‐AAG (17‐allylamino‐17‐demethoxy‐geldanamycin) and chloroquinone, however, the applicability of these treatment modalities to ONB requires further investigation.28 More importantly, platinum‐based agents were reported to be efficient against cancer cells with heterogeneous chromosomal instability,29 suggesting that platinum‐based chemotherapy may be an effective approach in treating this disease among the patients with chromosomal instability. In fact, cisplatin has shown efficacy as induction chemotherapy for ONB.30

Although comprehensive molecular profiling for patients with advanced and metastatic disease can be useful in designing defined therapeutic strategies, sequencing of ONB has been limited by low sample sizes due to rarity of the disease. The COSMIC database and published studies have reported alterations in a large number genes, including TP53, EGFR, CTNNB1, KIT, RET, APC, FGFR2, KDR, PDGFRA, SMAD4, MET, PIK3CA, NF1, CDKN2A, CDH1, SUZ12, PP6C, IKZF1, TGFBR2, CARD11, CDKN2C, SND1, cMET and CREB3L1, with subset of these genes being either known tumor drivers or tumor suppressors. For example, alterations in cKIT and PGDFRA have been implicated in the pathogenesis of gastrointestinal tumors. Tyrosine kinase inhibitors such as Imatinib, have been investigated in patients with gastrointestinal stromal tumors harboring the gain‐of‐function mutations in aforementioned genes,31 and may be considered for treating ONB patients given the high frequency of cases harboring such alterations. Gay et al have reported mutations in genes associated with mTOR, CDK and growth factor signaling pathways.10 Inhibitors of these pathways have been used in various cancers including breast, head and neck, lymphoma, and lung cancer.32, 33

The findings of the current review and data extracted from COSMIC database suggest that TP53 is the most frequently mutated gene in ONB.10, 12, 17, 20, 21, 34 Known alterations in TP53 include stop/gain, short variants, truncations, rearrangements and SNPs in the DNA binding domain. These alterations are commonly associated with loss of protein function and have been reported to be involved in driving tumorigenesis of various tumor types, including breast, lung, colorectal and other solid malignancies. In general, ~50% of human cancers carry a loss of function mutation resulting in abolished or reduced p53 protein function.35 TP53 gene mutations are associated with multiple oncogenic processes, such as loss of cell cycle arrest, apoptosis, inhibition of angiogenesis and metastasis, DNA repair, as well as inhibition of mTOR signaling axis. The enhanced number of alterations in TP53 among patients with ONB can pave the path for designing new therapies focused on restoring or enhancing the p53 tumor suppression activities. Various molecules including nutlins, MI series, RO5693, PRIMA‐1, and RITA have been developed for treatment of cancers with TP53 mutations. Nutlins function by targeting negative regulators of p53 protein, MI series function by reactivation of p53 by p21 mediated cell cycle arrest, whereas PRIMA‐1 binds to p53 by Michael addition after getting converted into methylene quinuclidinone (MQ), and its binding at cysteine residues induces apoptosis in tumor cells. Another molecule, COTI‐1, induces DNA damage signaling. Many of these molecules are in the preclinical phase, except for two compounds APR‐246 and COTI‐2 that have progressed to clinical trials.36 None of these approaches have been tested in ONB patients. Of note, tumors harboring TP53 alterations may respond better to WEE kinase inhibitors that function by acting against G2‐M checkpoint regulators of the cell cycle, WEE1 and CHK1.37

While epigenetics research in ONB is recently emerging, it has already demonstrated a promising practical implementation in clinical practice, as ONB harbors unique genome‐wide methylation profiles in comparison with other sinonasal malignancies.11 A pattern of global hypermethylation was described in a subgroup of ONB tumors that harbor R172 IDH2 hotspot mutations, corroborating the complex interplay between genomic and epigenomic regulation. It has been previously described that IDH mutations lead to neomorphic enzymatic activity, and acute myeloid leukemia patients that harbor IDH mutations also demonstrate a hypermethylation phenotype.38 This represents an attractive area for pharmaceutical targeting, as in vitro data using IDH2 R140Q expression in TF‐1 cells that reflects global hypermethylation of clinical acute myeloid leukemia, was reversed by an IDH2 small molecule inhibitor, AGI‐6780.39 It is likely that additional epigenetic regulatory mechanisms such as miRNAs and long non‐coding RNAs play a role in the onset, progression, and ultimately therapeutic response of ONB, but those remain yet to be determined.

The rare character of ONB has limited the sample size available for cytogenetic, genomic, and epigenetic studies and contributes to the limitations of this systematic review. Furthermore, the exclusion criteria included studies not in English or without full text which may have limited the scope of articles reviewed. Given its rarity, studies characterizing the landscape of genomic alterations in ONB are still limited, and targeted therapies are lacking for this disease. Thus, additional studies with larger sample sizes are warranted to further advance our understanding of the molecular processes underlying the progression of ONB. An increase in available molecular data will assist in detecting potential correlations between changes in the ONB specific genetic landscape and a clinical course of the disease. Furthermore, alterations specific to high grade and low grade ONB may be used for designing novel therapeutic approaches aiming to improve the overall survival of patients with this rare but aggressive disease. As the number of fully sequenced ONB cases continues to grow, comprehensive, well‐powered molecular analyses will allow international collaborative networks to correlate the genomic and epigenomic profiles with clinical outcomes and create a map of novel targets for therapeutic exploitation. Not only will this significantly improve our understanding of ONB pathogenesis, but also allow us to guide targeted therapies and potentially adopt these techniques in the routine diagnostic work‐up of our patients.

CONFLICT OF INTEREST

N. London holds stock in Navigen Pharmaceuticals, was a consultant for Cooltech Inc., and receives research funding from Merck, Inc., none of which are relevant to this study. All other authors declare no conflicts of interest.

ACKNOWLEDGMENTS

This research was supported (in part) by the Intramural Research Program of the NIH, NIDCD.

Kaur RP, Izumchenko E, Blakaj DM, et al. The genomics and epigenetics of olfactory neuroblastoma: A systematic review. Laryngoscope Investigative Otolaryngology. 2021;6(4):721–728. 10.1002/lio2.597

Funding information NIH Clinical Center; NIDCD; Intramural Research Program of the NIH

BIBLIOGRAPHY

  • 1.London NR Jr, 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‐e193. [DOI] [PubMed] [Google Scholar]
  • 2.Czapiewski P, Kunc M, Haybaeck J. Genetic and molecular alterations in olfactory neuroblastoma: implications for pathogenesis, prognosis and treatment. Oncotarget. 2016;7(32):52584‐52596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Berger L, Luc G, Richard D. L'esthesioneuroepitheliome olfatif. Bull Assoc Fr Etud Cancer. 1924;13:410‐421. [Google Scholar]
  • 4.King JT. Olfactory neurocytoma ("esthesioneuroepitheliome olfactif") case arising in the nasopharynx. AMA Arch Otolaryngol. 1959;69(6):729. [DOI] [PubMed] [Google Scholar]
  • 5.Sepulveda I, Herrera A, Delgado C, Platin E, Rosel P. Esthesioneuroblastoma: a case report and review of the literature. Int J Odontostomat. 2012;6(3):285‐289. [Google Scholar]
  • 6.Wenig B, Dulguerov P, Kapadia S, Prasad M, Fanburg‐Smith J, Thompson L. Tumours of the Nasal Cavity and Paranasal Sinuses: Neuroectodermal Tumours. IARC Press; 2005. [Google Scholar]
  • 7.Wolfe AR, Blakaj D, London N, et al. Clinical outcomes and multidisciplinary patterns of failure for olfactory neuroblastoma: the Ohio state experience. J Neurol Surg B Skull Base. 2020;81(3):287‐294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Faragalla H, Weinreb I. Olfactory neuroblastoma: a review and update. Adv Anat Pathol. 2009;16(5):322‐331. [DOI] [PubMed] [Google Scholar]
  • 9.Bockmuhl U, You X, Pacyna‐Gengelbach M, Arps H, Draf W, Petersen I. CGH pattern of esthesioneuroblastoma and their metastases. Brain Pathol. 2004;14(2):158‐163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gay LM, Kim S, Fedorchak K, et al. Comprehensive genomic profiling of Esthesioneuroblastoma reveals additional treatment options. Oncologist. 2017;22(7):834‐842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Capper D, Engel NW, Stichel D, et al. DNA methylation‐based reclassification of olfactory neuroblastoma. Acta Neuropathol. 2018;136(2):255‐271. [DOI] [PubMed] [Google Scholar]
  • 12.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. e815. [DOI] [PubMed] [Google Scholar]
  • 13.Sorensen PH, Wu JK, Berean KW, et al. Olfactory neuroblastoma is a peripheral primitive neuroectodermal tumor related to Ewing sarcoma. Proc Natl Acad Sci U S A. 1996;93(3):1038‐1043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cavazzana AO, Navarro S, Noguera R, Reynolds PC, Triche TJ. Olfactory neuroblastoma is not a neuroblastoma but is related to primitive neuroectodermal tumor (PNET). Prog Clin Biol Res. 1988;271:463‐473. [PubMed] [Google Scholar]
  • 15.Mezzelani A, Tornielli S, Minoletti F, Pierotti MA, Sozzi G, Pilotti S. Esthesioneuroblastoma is not a member of the primitive peripheral neuroectodermal tumour‐Ewing's group. Br J Cancer. 1999;81(4):586‐591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Dockhorn‐Dworniczak B, Schafer KL, Blasius S, et al. Assessment of molecular genetic detection of chromosome translocations in the differential diagnosis of pediatric sarcomas. Klin Padiatr. 1997;209(4):156‐164. [DOI] [PubMed] [Google Scholar]
  • 17.Cha S, Lee J, Shin JY, et al. Clinical application of genomic profiling to find druggable targets for adolescent and young adult (AYA) cancer patients with metastasis. BMC Cancer. 2016;16:170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Gallia GL, Zhang M, Ning Y, et al. Genomic analysis identifies frequent deletions of Dystrophin in olfactory neuroblastoma. Nat Commun. 2018;9(1):5410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wang Y, Marino‐Enriquez A, Bennett RR, et al. Dystrophin is a tumor suppressor in human cancers with myogenic programs. Nat Genet. 2014;46(6):601‐606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Topcagic J, Feldman R, Ghazalpour A, Swensen J, Gatalica Z, Vranic S. Comprehensive molecular profiling of advanced/metastatic olfactory neuroblastomas. PLoS One. 2018;13(1):e0191244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lazo de la Vega L, McHugh JB, Cani AK, et al. Comprehensive molecular profiling of olfactory neuroblastoma identifies potentially targetable FGFR3 amplifications. Mol Cancer Res. 2017;15(11):1551‐1557. [DOI] [PubMed] [Google Scholar]
  • 22.Dogan S, Chute DJ, Xu B, et al. Frequent IDH2 R172 mutations in undifferentiated and poorly‐differentiated sinonasal carcinomas. J Pathol. 2017;242(4):400‐408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Dogan S, Vasudevaraja V, Xu B, et al. DNA methylation‐based classification of sinonasal undifferentiated carcinoma. Mod Pathol. 2019;32(10):1447‐1459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Guled M, Myllykangas S, Frierson HF Jr, Mills SE, Knuutila S, Stelow EB. Array comparative genomic hybridization analysis of olfactory neuroblastoma. Mod Pathol. 2008;21(6):770‐778. [DOI] [PubMed] [Google Scholar]
  • 25.Purdie KJ, Harwood CA, Gulati A, et al. Single nucleotide polymorphism array analysis defines a specific genetic fingerprint for well‐differentiated cutaneous SCCs. J Invest Dermatol. 2009;129(6):1562‐1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gollin SM. Cytogenetic alterations and their molecular genetic correlates in head and neck squamous cell carcinoma: a next generation window to the biology of disease. Genes Chromosomes Cancer. 2014;53(12):972‐990. [DOI] [PubMed] [Google Scholar]
  • 27.Valli R, De Bernardi F, Frattini A, et al. Comparative genomic hybridization on microarray (a‐CGH) in olfactory neuroblastoma: analysis of ten cases and review of the literature. Genes Chromosomes Cancer. 2015;54(12):771‐775. [DOI] [PubMed] [Google Scholar]
  • 28.Tang YC, Williams BR, Siegel JJ, Amon A. Identification of aneuploidy‐selective antiproliferation compounds. Cell. 2011;144(4):499‐512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.McClelland SE, Burrell RA, Swanton C. Chromosomal instability: a composite phenotype that influences sensitivity to chemotherapy. Cell Cycle. 2009;8(20):3262‐3266. [DOI] [PubMed] [Google Scholar]
  • 30.Su SY, Bell D, Ferrarotto R, et al. Outcomes for olfactory neuroblastoma treated with induction chemotherapy. Head Neck. 2017;39(8):1671‐1679. [DOI] [PubMed] [Google Scholar]
  • 31.Law ME, Corsino PE, Narayan S, Law BK. Cyclin‐dependent kinase inhibitors as anticancer therapeutics. Mol Pharmacol. 2015;88(5):846‐852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lee JH, Kim Y, Choi JW, Kim YS. Correlation of imatinib resistance with the mutational status of KIT and PDGFRA genes in gastrointestinal stromal tumors: a meta‐analysis. J Gastrointestin Liver Dis. 2013;22(4):413‐418. [PubMed] [Google Scholar]
  • 33.Xie J, Wang X, Proud CG. mTOR inhibitors in cancer therapy. F1000Res. 2016;5:1‐11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cha S, Lee SH, Kim JI, Shin JY. Whole exome sequencing of a case of olfactory neuroblastoma. Cancer Res. 2014;74: Abstract nr 4191. [Google Scholar]
  • 35.Kaur RP, Vasudeva K, Kumar R, Munshi A. Role of p53 gene in breast cancer: focus on mutation spectrum and therapeutic strategies. Curr Pharm Des. 2018;24(30):3566‐3575. [DOI] [PubMed] [Google Scholar]
  • 36.Graves B, Thompson T, Xia M, et al. Activation of the p53 pathway by small‐molecule‐induced MDM2 and MDMX dimerization. Proc Natl Acad Sci U S A. 2012;109(29):11788‐11793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bauman JE, Chung CH. CHK it out! Blocking WEE kinase routs TP53 mutant cancer. Clin Cancer Res. 2014;20(16):4173‐4175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Figueroa ME, Abdel‐Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18(6):553‐567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kernytsky A, Wang F, Hansen E, et al. IDH2 mutation‐induced histone and DNA hypermethylation is progressively reversed by small‐molecule inhibition. Blood. 2015;125(2):296‐303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Castaneda VL, Cheah MS, Saldivar VA, Richmond CM, Parmley RT. Cytogenetic and molecular evaluation of clinically aggressive esthesioneuroblastoma. Am J Pediatr Hematol Oncol. 1991;13(1):62‐70. [DOI] [PubMed] [Google Scholar]
  • 41.Holland H, Koschny R, Krupp W, et al. Comprehensive cytogenetic characterization of an esthesioneuroblastoma. Cancer Genet Cytogenet. 2007;173(2):89‐96. [DOI] [PubMed] [Google Scholar]
  • 42.Jin Y, Mertens F, Arheden K, et al. Karyotypic features of malignant tumors of the nasal cavity and paranasal sinuses. Int J Cancer. 1995;60(5):637‐641. [DOI] [PubMed] [Google Scholar]
  • 43.Kristensen M, Quek HH, Chew CT, Chan SH. A cytogenetic study of 74 nasopharyngeal carcinoma biopsies. Ann Acad Med Singapore. 1991;20(5):597‐600. [PubMed] [Google Scholar]
  • 44.Lopez‐Hernandez 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]
  • 45.Riazimand SH, Brieger J, Jacob R, Welkoborsky HJ, Mann WJ. Analysis of cytogenetic aberrations in esthesioneuroblastomas by comparative genomic hybridization. Cancer Genet Cytogenet. 2002;136(1):53‐57. [DOI] [PubMed] [Google Scholar]
  • 46.Szymas J, Wolf G, Kowalczyk D, Nowak S, Petersen I. Olfactory neuroblastoma: detection of genomic imbalances by comparative genomic hybridization. Acta Neurochir. 1997;139(9):839‐844. [DOI] [PubMed] [Google Scholar]
  • 47.VanDevanter DR, George D, McNutt MA, Vogel A, Luthardt F. Trisomy 8 in primary esthesioneuroblastoma. Cancer Genet Cytogenet. 1991;57(1):133‐136. [DOI] [PubMed] [Google Scholar]
  • 48.Weiss GJ, Liang WS, Izatt T, et al. Paired tumor and normal whole genome sequencing of metastatic olfactory neuroblastoma. PLoS One. 2012;7(5):e37029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Whang‐Peng J, Freter CE, Knutsen T, Nanfro JJ, Gazdar A. Translocation t(11;22) in esthesioneuroblastoma. Cancer Genet Cytogenet. 1987;29(1):155‐157. [DOI] [PubMed] [Google Scholar]
  • 50.Kim J, Burke K, Gularte‐Merida R, et al. The landscape of genomic and transcriptomic alterations in esthesioneuroblastoma. Lab Invest. 2017;97:327A. [Google Scholar]
  • 51.Wang L, Ding Y, Wei L, et al. Recurrent olfactory neuroblastoma treated with Cetuximab and Sunitinib: a case report. Medicine (Baltimore). 2016;95(18):e3536. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Laryngoscope Investigative Otolaryngology are provided here courtesy of Wiley

RESOURCES