Turkish Journal of Pathology

Türk Patoloji Dergisi

Turkish Journal of Pathology

Turkish Journal of Pathology

2023, Vol 39, Num, 1     (Pages: 023-030)

BRAF, NRAS, KIT, TERT, GNAQ/GNA11 Mutation Profile and Histomorphological Analysis of Anorectal Melanomas: A Clinicopathologic Study

Orhun Cig TASKIN 1, Sule OZTURK SARI 1, Ismail YILMAZ 2, Ozge HURDOGAN 1, Metin KESKIN 3, Nesimi BUYUKBABANI 1, Mine GULLUOGLU 1

1 Department of Pathology, Istanbul University, Istanbul Faculty of Medicine, ISTANBUL, TURKEY
2 Sultan II. Abdulhamid Han Training and Research Hospital, University of Health Sciences, ISTANBUL, TURKEY
3 Department of Surgery, Istanbul University, Istanbul Faculty of Medicine, ISTANBUL, TURKEY

DOI: 10.5146/tjpath.2022.01576
Viewed: 130
 - 
Downloaded : 39

Summary

Objective: Primary anorectal melanomas (AMs) are uncommon neoplasms with aggressive behavior. Molecular profile and clinicopathologic features of AMs are still not well established. In this study, we aimed to investigate BRAF, NRAS, KIT, TERT, and GNAQ/GNA11 mutation status and clinicopathologic features of AMs.

Material and Method: All diagnostic slides of 15 AMs were reviewed. Histopathological and follow-up information were documented. Mutations in exon 15 of the BRAF gene; exons 2 and 3 of the NRAS gene; exons 9, 11, 13, 17, and 18 of the KIT gene; and exons 4 and 5 of the GNAQ/GNA11 genes and mutations in the promoter region of the TERT gene (chr.5, 1,295,228C>T and 1,295,250C>T) were analyzed.

Results: BRAF(V600E) and KIT(V555I and K642E) mutations were observed in one (7%) and two cases (14%), respectively. NRAS, TERT and GNAQ/GNA11 mutations were not detected. The mean age was 65. Patients presented with rectal mass, rectal bleeding, pain, and weight loss. 73% of the lesions were macroscopically polypoid. The most common tumor cell type was epithelioid. Mean tumor thickness was 10.4 mm. One third of the cases lacked pigmentation. In situ melanoma was present in one third of the cases. Among 14 patients with follow-up data, 12 succumbed to disease. The mean overall survival was 36 months.

Conclusion: AMs are uncommon tumors with dismal survival, usually occurring in the elderly in various gross and microscopic appearances. In terms of molecular profile, BRAF and KIT mutations are rarely detected. Profiling of larger cohorts is required to elucidate the pathogenesis and to identify potential molecular indicators that may contribute to the development of individualized targeted therapies.

Introduction

Primary anorectal melanomas (AMs) are uncommon neoplasms that account for about 1% of anal canal tumors [1]. Among mucosal melanomas, which constitute around 1% of all malignant melanomas [2], the anal canal is the second most common site of origin, following the head and neck [3]. AMs are believed to arise from the melanocytes of the anal squamous epithelium and extend towards the anal canal [4,5]; however, cases that originated from the rectal mucosa -without the involvement of the squamous epithelium- have also been reported [6,7].

Patients with AM usually present with rectal bleeding and pain. Tumors often mimic hemorrhoids, anal polyps or rectal carcinoma, forming large, dark-colored masses with expansile and nodular borders, with or without ulceration[8,9]. Microscopically, tumors are often composed of sheets/fascicles of epithelioid or spindled malignant cells with vesicular chromatin and prominent nucleoli, with variable amounts of pigmentation [10]. However, unusual presentations and rare histologic/cytologic patterns often challenge pathologists in the differential diagnosis of AMs, which includes carcinomas, sarcomas and even lymphomas [11,12]. In challenging cases, a panel of immunohistochemical stains, including markers of melanocytic lineage is required to render the accurate diagnosis [13].

Similar to mucosal melanomas of other sites, AMs behave much worse than their cutaneous counterparts. Despite the use of various treatment regimens including extensive surgery, radiotherapy, chemotherapy, and targeted therapies, AMs have an aggressive clinical course with an overall 5-year survival rate of less than 25% [3,14]. Additionally, AMs were associated with the poorest prognosis among mucosal melanomas in a large European cohort [15].

The recent progress in the molecular profiling of cutaneous melanomas has greatly contributed in our understanding of their pathogenesis, as well as their management with the use of targeted therapies and immunotherapy [16]. However, mucosal melanomas tend to differ from their cutaneous counterparts in terms of molecular profiling; albeit showing a heterogeneous molecular profile, they have lower BRAF and TERT, and relatively higher NRAS and KIT mutation frequencies [17-27]. Additionally, GNAQ/GNA11 mutations, which have been reported in uveal melanomas and subjected to targeted therapies [28,29], also occur rarely in AMs [30], but not in other mucosal melanomas [31]. However, molecular profiles of mucosal melanomas are still not well established due to their rareness. In addition to their molecular background, data concerning their clinicopathologic features are highly limited. Accordingly, widely accepted treatment protocols do not exist. In this study, as an extension to our previous work on head and neck mucosal melanomas [31], we aimed to investigate the BRAF, NRAS, KIT, TERT and GNAQ/GNA11 mutation status of 15 AMs, as well as their clinicopathologic features.

Methods

Case Selection, Clinical and Pathological Data Collection
The digital database of the pathology department (Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey) was searched for cases diagnosed as AM between the years 2000 and 2019, including both in-house material and outside consultations. Data on clinical history and physical/ radiologic examination were reviewed for all retrieved cases in order to exclude previous history of cutaneous melanoma and/or the possibility of metastasis. Cases with a suspicion of secondary melanoma were not included.

Diagnostic slides and paraffin blocks were retrieved from the archives. All slides (hematoxylin and eosin and/or immunohistochemically stained) were reviewed. Tumor cell types were classified under four categories (epithelioid, spindle, pleomorphic, and lymphoma-like), as mentioned in the literature [9,12,32,33], and combined morphology was assessed when appropriate. Histopathological information regarding tumor thickness (Breslow), presence of perineural and lymphovascular invasion, pigmentation, ulceration, necrosis, tumor infiltrating lymphocytes, mitotic count (per high power field), and margin status (when applicable) were also documented. Follow-up information was obtained from the clinical files or national database.

Mutation Analysis
Tumor targets (>90% viable tumor) were manually microdissected from 10-mm thick unstained histologic sections for enrichment of tumor cellularity. Deparaffinization of tissue sections was performed. Then, DNA was isolated by using the QIAamp DNA FFPE Tissue Kit (50) (catalog #: 56404) (QIAGEN, Hilden, Germany). DNA concentrations of the samples were assessed spectrophotometrically using a Nanodrop 1000 spectrophotometer (ThermoScientific, USA).

Mutations in exon 15 of BRAF gene; exons 2 and 3 of the NRAS gene; exons 9, 11, 13, 17, and 18 of the KIT gene; and exons 4 and 5 of the GNAQ and GNA11 genes (well-known hotspot regions for oncogenic mutations) and mutations in the promoter region of the TERT gene (chr5, 1,295,228C>T and 1,295,250C>T) were analyzed by validated previously described polymerase chain reaction (PCR)-based direct Sanger sequencing (analytical sensitivity 25%) by using 200 ng of each tumor DNA [31].

Additional Information
The Helsinki principles were respected in this study and patients’ data confidentiality was ensured according to their guidelines. This study was approved by the institutional review board.

Results

Clinical Features
The study was conducted with 15 cases of 15 patients (8 males and 7 females) with a mean age of 65 years (range: 30 - 86 years). The specimens consisted of 8 local excisions, 3 abdominoperineal resections, 1 polypectomy and 3 incisional biopsies. By definition, all tumors originated from the anal canal. Among patients with available information (n=9), presenting symptoms were described as rectal mass, rectal bleeding, pain and weight loss.

Histopathology
Diagnostic slides of the fifteen cases were systematically reviewed. Tumors were polypoid in 73%. The cell type was epithelioid and spindle in 33%, epithelioid and lymphomalike in 27%, spindle in 13%, epithelioid and pleomorphic in 13%, spindle and lymphoma-like in 7%, and lymphoma-like in 7% of cases (Figure 1). Mean tumor thickness in 12 cases was 10.4 mm (range: 1.1-22 mm). Tumor thickness could not be measured in 2 incisional biopsies and 1 polypectomy due to poor orientation. Majority of cases (67%) showed pigmentation, whereas 33% were amelanotic (Figure 2). Ulceration was seen in 80% of cases. Mean mitotic count was 4.9 per 10 high power fields (range 0-10). In situ melanoma was detected in 33% of the cases (Figure 2). Intratumoral lymphocytes were prominent in 53%. Metastasis in lymph nodes was observed in 3 abdominoperineal resections.

Figure 1: Neoplastic cells in anorectal melanoma demonstrate various morphologic appearances: A) Epithelioid melanoma cells with roundish nuclei and wide eosinophilic cytoplasm, B) Lymphoma-like small neoplastic cells, admixed in a fibrous stroma, showing crush artifact, C) Pleomorphic melanoma cells with huge, bizarre nuclei and prominent cytoplasm, D) Spindle cells showing elongated nuclei and sparse cytoplasm (A-D: Hematoxylin&Eosin, x400).

Figure 2: A) Anorectal melanoma cells, which lack melanin pigment (Hematoxylin&Eosin, x200), B) Melanoma in situ, atypical melanocytes showing continuous growth at the basal layer (Hematoxylin&Eosin, x400).

Immunohistochemistry
Among 15 cases, 10 were subjected to immunohistochemical analysis. Five cases that did not require immunohistochemical analysis harbored in situ melanoma component and/or prominent pigmentation.

Among melanocytic markers, S-100 and HMB-45 were positive in all cases (100%; n=9 and 7; respectively). Melan-A was positive in 6/7 (86%) cases. Epithelial markers (Pan-cytokeratin, epithelial membrane antigen, and carcinoembryonic antigen), neuroendocrine markers (chromogranin, synaptophysin), muscle markers (desmin, smooth muscle actin) were all negative when performed, along with leukocyte common antigen, CD30 and CD34. CD117 was positive in 2 of 3 cases performed.

Mutation Analysis
A total of 3 cases (20%) were found to harbor mutations. BRAF (V600E) and KIT (V555I and K642E) mutations were observed in one (7%) and two cases (14%), respectively. NRAS, TERT, and GNAQ/GNA11 mutations were not observed.

Follow-Up and Survival Information
Among 14 patients with available information, 12 died. The mean overall survival was 36 months (range: 0-112 months). The histopathological, clinical, and mutational findings and follow-up information are summarized in Table I.

Table I: Clinicopathologic features of the study group.

Discussion

Mutational profile of mucosal melanomas is known to differ from their cutaneous counterparts, suggesting a different pathway in the pathogenesis: They harbor lower BRAF and TERT, and relatively higher NRAS and KIT mutation frequencies [17-27]. The absence of UV damage is often mentioned to be associated with this disparity. Furthermore, regarding the site of origin, differences also exist in the same subgroup: in an earlier study, we concluded that NRAS and TERT promoter mutation rates were significantly higher in sinonasal than in oral mucosal melanomas of the head and neck [31]. In the current literature, data on AMs’ molecular profile is mostly merged with cutaneous and/or mucosal melanomas, primarily due to their rareness [34-39]. In studies with relatively large cohorts of AMs, KIT mutations were most commonly encountered, followed by mutations in NRAS. The newly introduced NF1 gene also has an important role in the oncogenesis. BRAF mutations were also observed with different frequencies, most likely due to small sample sizes or populational differences of the cohorts [40-44]. In addition, one study showed around 2% GNAQ and 6% GNA11 mutations [39]. In our study group, BRAF and KIT mutations were found in 7% and 14%, respectively. NRAS, TERT, and GNAQ/GNA11 mutations were absent. Together, these supported the low mutation burden of AMs, as stated in the literature [45]. In a large cohort of mucosal melanomas, 3% of AMs showed BRAF, 10% showed NRAS, and 19% showed KIT mutations. This study analyzed a subset of cases by Sanger sequencing, and others by next-generation sequencing (NGS); and proposed NRAS mutation as a predictor of worse survival, independent of stage in all mucosal melanomas [46]. Those being mentioned, as a limitation of this study, we had a limited number of cases, impeding a correlation analysis between mutational status and prognostic data. We also did not have access to NGS techniques. Therefore we were unable to perform a comprehensive genomic analysis including NF1, which was recently integrated in the molecular classification of AMs.

Our findings verified that AMs are highly rare and aggressive neoplasms that generally occur in elder patients, with a mean age of 65 years in the present study. In one study, older age (>70 years) was found to be an independent poor prognostic factor [10]. Although our data did not reveal any significant sex predilection, geographic and populational differences in the relative frequency between two genders have been reported [10,47].

Clinically, recognizing AMs can be challenging for physicians. The symptomatology may include nonspecific rectal bleeding and pain, as well as weight loss in metastatic disease [48]. Endoscopically, tumors can present with various appearances. Polypoid masses are frequently encountered, similar to 73% of our cases. Anal prolapse, and luminal or submucosal masses with or without ulceration or pigmentation can also be seen [48]. This may cause misdiagnosis of AM as hemorrhoids, perianal abscess, anal polyps or other malignancies [49].

The presence of melanin pigmentation can help render the accurate diagnosis. However, it is not always present, with some studies reporting 37% of their cases as amelanotic [12,32]. In addition, in situ melanoma component, or junctional melanocytic activity, which are characteristic in cutaneous melanomas, have been reported in up to 75% of AMs [12,32,49]. However, this feature may be missing due to the absence of adjacent mucosa in incisional biopsies that consist entirely of tumor, and also due to ulceration and fragmentation in excisional biopsies. In our study, a third of the tumors were amelanotic and a third had an in situ component.

Microscopically, epithelioid, spindled, pleomorphic, and lymphoma-like tumor cells may co-exist, with epithelioid being the most frequent with combination of the others [32,40], similar to the present study. Therefore, AMs can mimic a large spectrum of malignancies, making the use of immunohistochemistry crucial in differential diagnosis. Additionally, lack of in situ component and/or lack of pigmentation, also complicate the diagnostic puzzle. At this point, an immunohistochemical panel of commonly used melanocytic markers, S-100 protein/SOX10, Melan-A, HMB-45, can be helpful. Moreover, additional markers may be required to rule out other entities including primary or metastatic carcinomas, neuroendocrine neoplasms, sarcomas, lymphomas, and gastrointestinal stromal tumors. Among those, the use of CD117 requires careful interpretation due to its frequent positivity in AMs (up to 75% in the literature), which can lead to a misdiagnosis of rectal gastrointestinal stromal tumor, if not performed along with other melanocytic markers [12,33]. Additionally, CD117 immunohistochemistry is known not to correlate with KIT status and therefore should not be used with mutation screening purposes [44].

In terms of pathological staging and prognosis, specific guidelines for reporting AMs do not exist. They are usually reported according to the American Joint Commission on Cancer (AJCC) guidelines for cutaneous melanoma [50], which depends mostly on tumor thickness, causing several issues in the daily practice. In the vast majority of cases on reported series [10,40,45,51-53] including ours (10.4 mm), the average tumor thickness was much thicker than the 4 mm threshold used for staging T4 cutaneous melanomas. This threshold inevitably categorizes the bulk of cases as T4, thus diminishing the prognostic stratification of the T classification. Several attempts have been made in order to sharpen the prognostic accuracy, including the implementation of different thickness cut-offs [51], subclassification depending on the localization [52] and metastatic status [40]. Among other histopathologic prognostic factors, presence of metastasis, lymphovascular and perineural invasion, invasion of muscularis propria/ anal sphincter were also reported [10,40,51]. Mitotic rate is a very strong prognostic factor in cutaneous melanomas [54]. Although high mitotic rates are frequently encountered similar to our study, their correlation with the clinical outcome is not well established in AMs [12,33]. Nevertheless, studies on larger cohorts are needed in order to define the relationship between the distinct histopathologic parameters and prognosis.

In terms of treatment, optimal algorithms are lacking and satisfactory results are yet to be achieved [55]. The primary choice of treatment is complete surgical removal of the tumor [8]. Advantages of local approaches (mucosal resection or local excision) over extensive surgery (abdominoperineal resection) have long been discussed; however, literature data lack proof to recommend one modality over the other [56-58]. Moreover, adjuvant or neoadjuvant therapies do not seem to make significant difference on the clinical outcome [55]. The results of recently implemented immunotherapy is yet to be proven [59]. Since our data involved limited information on adjuvant treatment, we were unable to draw any conclusions on this subject.

In conclusion, AMs are uncommon tumors with aggressive behavior and poor survival. They usually occur in the elderly and present in various gross and microscopic appearances, thus involving a wide spectrum of differential diagnoses. For accurate diagnosis, the melanocytic lineage should be demonstrated with immunohistochemistry, especially in the absence of conventional morphological clues such as pigmentation and/or in situ component. In terms of molecular profile, BRAF and KIT mutations rarely occur. Profiling of larger cohorts is required to elucidate the pathogenesis and to identify potential molecular indicators that may contribute in the development of individualized targeted therapies.

Conflict of Interest and Funding Statement
Authors have no conflicts of interest to declare. All authors have read and contributed to the final manuscript and confirm that this is an original work that has not been previously published, nor has it been submitted to another journal for simultaneous review. This study is supported by the Scientific Research Project Fund of Istanbul University (Project number: 51524). This study was partially presented in 28th Congress of the European Society of Pathology, 25- 29 September 2016, Cologne, Germany.

Authorship Contributions
Concept: SOS, OT, Design: SOS, OT, Data collection or processing: OS, OT, IY, OH, Analysis or Interpretation: SOS, OT, IY, OH, Literature search: OT, OH, Writing: SOS, OT, OH, Approval: NB, MG.

Reference

1) Belbaraka R, Elharroudi T, Ismaili N, Fetohi M, Tijami F, Jalil A,Errihani H. Management of anorectal melanoma: Report of 17cases and literature review. J Gastrointest Cancer. 2012;43:31-5.

2) Chen H, Cai Y, Liu Y, He J, Hu Y, Xiao Q, Hu W, Ding K.Incidence, surgical treatment, and prognosis of anorectalmelanoma from 1973 to 2011 a population-based SEER analysis.Med. (United States) 2016;95:e2770.

3) Chang AE, Karnell LH, Menck HR. The national cancer data basereport on cutaneous and noncutaneous melanoma: A summaryof 84,836 cases from the past decade. Cancer. 1998;83:1664-78.

4) Ackermann DM, Polk HC, Schrodt GR. Desmoplastic melanomaof the anus. Hum Pathol. 1985;16:1277-9.

5) Morson BC, Volkstädt H. Malignant melanoma of the anal canal.J Clin Pathol. 1963;16:126-32.

6) Nicholson AG, Cox PM, Marks CG, Cook MG. Primary malignantmelanoma of the rectum. Histopathology. 1993;22:261-4.

7) Werdin C, Limas C, Knodell RG. Primary malignant melanomaof the rectum. Evidence for origination from rectal mucosalmelanocytes. Cancer. 1988;61:1364-70.

8) Malaguarnera G, Madeddu R, Catania VE, Bertino G, MorelliL, Perrotta RE, Drago F, Malaguarnera M, Latteri S. Anorectalmucosal melanoma. Oncotarget. 2018;9:8785-800.

9) da Cruz GMG, Andrade Filho J de S, Patrus G, Leite SM de O,da Silva IG, Teixeira RG, Braga ÁCG, Ferreira RMRS. Anorectalmelanoma-histopathological and immunohistochemical featuresand treatment. J Coloproctology. 2014;34:95-103.

10) Ren M, Lu Y, Lv J, Shen X, Kong J, Dai B, Kong Y. Prognosticfactors in primary anorectal melanoma: A clinicopathologicalstudy of 60 cases in China. Hum Pathol. 2018;79:77-85.

11) Nakhleh RE, Wick MR, Rocamora A, Swanson PE, Dehner LP.Morphologic diversity in malignant melanomas. Am J ClinPathol. 1990;93:731-40.

12) Tariq MU, Ud Din N, Ud Din NF, Fatima S, Ahmad Z. Malignantmelanoma of anorectal region: A clinicopathologic study of 61cases. Ann Diagn Pathol. 2014;18:275-81.

13) Prieto VG, Shea CR. Immunohistochemistry of melanocyticproliferations. Arch Pathol Lab. Med 2011;135:853-9.

14) Singer M, Mutch MG. Anal melanoma. Clin Colon Rectal Surg.2006;19:78-87.

15) Heppt MV, Roesch A, Weide B, Gutzmer R, Meier F, Loquai C,Kähler KC, Gesierich A, Meissner M, von Bubnoff D, Göppner D,Schlaak M, Pföhler C, Utikal J, Heinzerling L, Cosgarea I, EngelJ, Eckel R, Martens A, Mirlach L, Satzger I, Schubert-FritschleG, Tietze JK, Berking C. Prognostic factors and treatmentoutcomes in 444 patients with mucosal melanoma. Eur J Cancer.2017;81:36-44.

16) Melis C, Rogiers A, Bechter O, van den Oord JJ. Moleculargenetic and immunotherapeutic targets in metastatic melanoma.Virchows Arch. 2017;471:281-93.

17) Tacastacas JD, Bray J, Cohen YK, Arbesman J, Kim J, Koon HB,Honda K, Cooper KD, Gerstenblith MR. Update on primarymucosal melanoma. J Am Acad Dermatol. 2014;71:366-75.

18) Guo J, Si L, Kong Y, Flaherty KT, Xu X, Zhu Y, Corless CL, Li L,Li H, Sheng X, Cui C, Chi Z, Li S, Han M, Mao L, Lin X, Du N,Zhang X, Li J, Wang B, Qin S. Phase II, open-label, single-armtrial of imatinib mesylate in patients with metastatic melanomaharboring c-Kit mutation or amplification. J Clin Oncol.2011;29:2904-9.

19) Miao Y, Wang R, Ju H, Ren G, Guo W. TERT promoter mutationis absent in oral mucosal melanoma. Oral Oncol. 2015;51:e65-6.

20) Hodi FS, Corless CL, Giobbie-Hurder A, Fletcher JA, Zhu M,Marino-Enriquez A, Friedlander P, Gonzalez R, Weber JS,Gajewski TF, O'Day SJ, Kim KB, Lawrence D, Flaherty KT,Luke JJ, Collichio FA, Ernstoff MS, Heinrich MC, Beadling C,Zukotynski KA, Yap JT, Van den Abbeele AD, Demetri GD, FisherDE. Imatinib for melanomas harboring mutationally activated oramplified KIT arising on mucosal, acral, and chronically sundamagedskin. J Clin Oncol. 2013;31:3182-90.

21) Carvajal RD, Antonescu CR, Wolchok JD, Chapman PB, RomanRA, Teitcher J, Panageas KS, Busam KJ, Chmielowski B, LutzkyJ, Pavlick AC, Fusco A, Cane L, Takebe N, Vemula S, BouvierN, Bastian BC, Schwartz GK. KIT as a therapeutic target inmetastatic melanoma. JAMA. 2011;305:2327-34.

22) Beadling C, Jacobson-Dunlop E, Hodi FS, Le C, Warrick A,Patterson J, Town A, Harlow A, Cruz F, Azar S, Rubin BP,Muller S, West R, Heinrich MC, Corless CL. KIT gene mutationsand copy number in melanoma subtypes. Clin Cancer Res.2008;14:6821-8.

23) Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activationof KIT in distinct subtypes of melanoma. J Clin Oncol.2006;24:4340-6.

24) Minor DR, Kashani-Sabet M, Garrido M, O’Day SJ, Hamid O,Bastian BC. Sunitinib therapy for melanoma patients with KITmutations. Clin Cancer Res. 2012;18:1457-63.

25) Griewank KG, Murali R, Puig-Butille JA, Schilling B, LivingstoneE, Potrony M, Carrera C, Schimming T, Möller I, SchwambornM, Sucker A, Hillen U, Badenas C, Malvehy J, Zimmer L, ScheragA, Puig S, Schadendorf D. TERT promoter mutation status asan independent prognostic factor in cutaneous melanoma. J NatlCancer Inst. 2014;106:dju246.

26) Egberts F, Krüger S, Behrens HM, Bergner I, PapaspyrouG, Werner JA, Alkatout I, Haag J, Hauschild A, Röcken C.Melanomas of unknown primary frequently harbor TERTpromotermutations. Melanoma Res. 2014;24:131-6.

27) Jangard M, Zebary A, Ragnarsson-Olding B, Hansson J. TERTpromoter mutations in sinonasal malignant melanoma: A studyof 49 cases. Melanoma Res. 2014;25:185-8.

28) Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, GauglerL, O’Brien JM, Simpson EM, Barsh GS, Bastian BC. Frequentsomatic mutations of GNAQ in uveal melanoma and blue naevi.Nature. 2009;457:599-602.

29) Chen X, Wu Q, Tan L, Porter D, Jager MJ, Emery C, Bastian BC.Combined PKC and MEK inhibition in uveal melanoma withGNAQ and GNA11 mutations. Oncogene. 2014;33:4724-34.

30) Kim CY, Kim DW, Kim K, Curry J, Torres-Cabala C, Patel S.GNAQ mutation in a patient with metastatic mucosal melanoma.BMC Cancer. 2014;14:516.

31) Öztürk Sari Ş, Yilmaz İ, Taşkin OÇ, Narli G, Şen F, ÇomoğluŞ, Firat P, Bilgiç B, Yilmazbayhan D, Özlük Y, Büyükbabani N.BRAF, NRAS, KIT, TERT, GNAQ/GNA11 mutation profileanalysis of head and neck mucosal melanomas: A study of 42cases. Pathology. 2017;49:55-61.

32) Charifa A, Zhang X. Morphologic and immunohistochemicalcharacteristics of anorectal melanoma. Int J Surg Pathol.2018;26:725-9.

33) Chute DJ, Cousar JB, Mills SE. Anorectal malignant melanoma:Morphologic and immunohistochemical features. Am J ClinPathol. 2006;126:93-100.

34) Omholt K, Grafström E, Kanter-Lewensohn L, Hansson J,Ragnarsson-Olding BK. KIT pathway alterations in mucosalmelanomas of the vulva and other sites. Clin Cancer Res.2011;17:3933-42.

35) Quek C, Rawson RV, Ferguson PM, Shang P, Silva I, Saw RPM,Shannon K, Thompson JF, Hayward NK, Long GV, Mann GJ,Scolyer RA, Wilmott JS. Recurrent hotspot SF3B1 mutationsat codon 625 in vulvovaginal mucosal melanoma identifiedin a study of 27 Australian mucosal melanomas. Oncotarget.2019;10:930-41.

36) Schaefer T, Satzger I, Gutzmer R. Clinics, prognosis and newtherapeutic options in patients with mucosal melanoma: Aretrospective analysis of 75 patients. Med. (United States)2017;96:e5753.

37) Edwards RH, Ward MR, Wu H, Medina CA, Brose MS, VolpeP, Nussen-Lee S, Haupt HM, Martin AM, Herlyn M, LessinSR, Weber BL. Absence of BRAF mutations in UV-protectedmucosal melanomas. J Med Genet. 2004;41:270-2.

38) Newell F, Kong Y, Wilmott JS, Johansson PA, Ferguson PM, CuiC, Li Z, Kazakoff SH, Burke H, Dodds TJ, Patch AM, NonesK, Tembe V, Shang P, van der Weyden L, Wong K, Holmes O,Lo S, Leonard C, Wood S, Xu Q, Rawson RV, MukhopadhyayP, Dummer R, Levesque MP, Jönsson G, Wang X, Yeh I, WuH, Joseph N, Bastian BC, Long GV, Spillane AJ, Shannon KF,Thompson JF, Saw RPM, Adams DJ, Si L, Pearson JV, HaywardNK, Waddell N, Mann GJ, Guo J, Scolyer RA. Whole-genomelandscape of mucosal melanoma reveals diverse drivers andtherapeutic targets. Nat Commun. 2019;10:3163.

39) Sheng X, Kong Y, Li Y, Zhang Q, Si L, Cui C, Chi Z, Tang B,Mao L, Lian B, Wang X, Yan X, Li S, Dai J, Guo J. GNAQ andGNA11 mutations occur in 9.5% of mucosal melanoma and areassociated with poor prognosis. Eur J Cancer. 2016;65:156-63.

40) Nagarajan P, Piao J, Ning J, Noordenbos LE, Curry JL, Torres-Cabala CA, Diwan AH, Ivan D, Aung PP, Ross MI, Royal RE,Wargo JA, Wang WL, Samdani R, Lazar AJ, Rashid A, Davies MA,Prieto VG, Gershenwald JE, Tetzlaff MT. Prognostic model forpatient survival in primary anorectal mucosal melanoma: Stageat presentation determines relevance of histopathologic features.Mod Pathol. 2020;33:496-513.

41) Yang HM, Hsiao SJ, Schaeffer DF, Lai C, Remotti HE, HorstD, Mansukhani MM, Horst BA. Identification of recurrentmutational events in anorectal melanoma. Mod Pathol.2017;30:286-96.

42) Ni S, Huang D, Chen X, Huang J, Kong Y, Xu Y, Du X, Sheng W.C-kit gene mutation and CD117 expression in human anorectalmelanomas. Hum Pathol. 2012;43:801-7.

43) Hintzsche JD, Gorden NT, Amato CM, Kim J, Wuensch KE,Robinson SE, Applegate AJ, Couts KL, Medina TM, Wells KR,Wisell JA, McCarter MD, Box NF, Shellman YG, Gonzalez RC,Lewis KD, Tentler JJ, Tan AC, Robinson WA. Whole-exomesequencing identifies recurrent SF3B1 R625 mutation andcomutation of NF1 and KIT in mucosal melanoma. MelanomaRes. 2017;27:189-99.

44) Santi R, Simi L, Fucci R, Paglierani M, Pepi M, Pinzani P, MerelliB, Santucci M, Botti G, Urso C, Massi D. KIT genetic alterationsin anorectal melanomas. J Clin Pathol. 2015;68:130-4.

45) Dodds TJ, Wilmott JS, Jackett LA, Lo SN, Long GV, ThompsonJF, Scolyer RA. Primary anorectal melanoma: Clinical,immunohistology and DNA analysis of 43 cases. Pathology.2019;51:39-45.

46) Wróblewska JP, Dias-Santagata D, Ustaszewski A, Wu CL,Fujimoto M, Selim MA, Biernat W, Ryś J, Marszalek A, HoangMP. Prognostic roles of BRAF, KIT, NRAS, IGF2R and SF3B1mutations in mucosal melanomas. Cells. 2021;10:2216.

47) Callahan A, Anderson WF, Patel S, Barnholtz-Sloan JS, BordeauxJS, Tucker MA, Gerstenblith MR. Epidemiology of anorectalmelanoma in the United States: 1992 to 2011. Dermatologic Surg.2016;42:94-9.

48) La Selva D, Kozarek RA, Dorer RK, Rocha FG, Gluck M. Primaryand metastatic melanoma of the GI tract: Clinical presentation,endoscopic findings, and patient outcomes. Surg Endosc.2020;34:4456-62.

49) Cooper PH, Mills SE, Allen MS. Malignant melanoma of the anus- Report of 12 patients and analysis of 255 additional cases. DisColon Rectum. 1982;25:693-703.

50) Amin MB, Edge S, Greene F, Byrd DR, Brookland RK, WashingtonMK, Gershenwald JE, Compton CC, Hess KR, Sullivan DC,Jessup JM, Brierley JD, Gaspar LE, Schilsky RL, Balch CM.AJCC Cancer Staging Manual. 8th Ed. Springer InternationalPublishing: American Joint Commission on Cancer; 2017.

51) Yeh JJ, Shia J, Hwu WJ, Busam KJ, Paty PB, Guillem JG, CoitDG, Wong WD, Weiser MR. The role of abdominoperinealresection as surgical therapy for anorectal melanoma. Ann Surg.2006;244:1012-7.

52) Bello DM, Smyth E, Perez D, Khan S, Temple LK, Ariyan CE,Weiser MR, Carvajal RD. Anal versus rectal melanoma: Does siteof origin predict outcome? Dis Colon Rectum. 2013;56:150-7.

53) Ben-Izhak O, Levy R, Weill S, Groisman G, Cohen H, StajermanS, Misselevich I, Nitecky S, Eidelman S, Kerner H. Anorectalmalignant melanoma: A clinicopathologic study, includingimmunohistochemistry and DNA flow cytometry. Cancer.1997;79:18-25.

54) Piñero-Madrona A, Ruiz-Merino G, Cerezuela Fuentes P,Martínez-Barba E, Rodríguez-López JN, Cabezas-Herrera J.Mitotic rate as an important prognostic factor in cutaneousmalignant melanoma. Clin Transl Oncol. 2019;21:1348-56.

55) Kirchoff DD, Deutsch GB, Foshag LJ, Lee JH, Sim MS, Faries MB.Evolving therapeutic strategies in mucosal melanoma have notimproved survival over five decades. Am Surg. 2016;82:1-5.

56) Kelly P, Zagars GK, Cormier JN, Ross MI, Guadagnolo BA.Sphincter-sparing local excision and hypofractionated radiationtherapy for anorectal melanoma: A 20-year experience. Cancer.2011;117:4747-55.

57) Ross M, Pezzi C, Pezzi T, Meurer D, Hickey R, Balch C. Patternsof failure in anorectal melanoma: A guide to surgical therapy.Arch Surg. 1990;125:313-6.

58) Che X, Zhao DB, Wu YK, Wang CF, Cai JQ, Shao YF, Zhao P.Anorectal malignant melanomas: Retrospective experience withsurgical management. World J Gastroenterol. 2011;17:534-9.

59) Taylor JP, Stem M, Yu D, Chen SY, Fang SH, Gearhart SL, SafarB, Efron JE. Treatment strategies and survival trends for anorectalmelanoma: Is it time for a change? World J Surg. 2019;43:1809-19.

Keywords : Anorectal melanoma, BRAF, NRAS, KIT, TERT, GNA