Skip to main content
Melanoma Management logoLink to Melanoma Management
. 2015 May 18;2(2):121–125. doi: 10.2217/mmt.15.6

Spitzoid melanoma of childhood: a case series and review

Sandeep Batra 1,1,2,3,*
PMCID: PMC6094708  PMID: 30190841

SUMMARY 

Spitzoid melanomas (SM) and atypical Spitz tumors (AST) are rare pediatric neoplasms. We performed a retrospective, single-institution review and report our institutional experience. We identified 10 patients (median age: 12.5 years). A sentinel node biopsy (SNB) was performed in 8/10 (80%) patients, and interestingly 7/8 (87.5%) were found to be positive for malignant cells. A complete regional lymphadenectomy was performed in all SNB-positive patients, but only 2/8 (25%) were found to have additional lymph node spread. Adjuvant therapy was administered in 5/8 SLNB-positive and 2/2 (100%) regional LN-positive cases. All patients had excellent long-term outcomes (100% survival). This report highlights the excellent outcomes associated with SNB + pediatric SM and AST.

KEYWORDS : long-term survival, lymph node metastasis, sentinel lymph node biopsy, Spitzoid melanoma, variants


Practice points.

  • Atypical Spitz tumors (AST) and Spitzoid melanomas (SM) are biologically distinct from adult type melanomas (MM).

  • AST and SM are usually treated as per current MM management guidelines.

  • Existing data suggest that adjuvant therapy may not be necessary in fully resected nonmetastatic sentinel lymph node-positive (SLB+) SM/AST and that these patients could be observed as they are unlikely to recur.

  • A great degree of variability exists in the management of these tumors amongst pediatric oncologists.

  • Further molecular studies are needed to understand the unique biological behavior of AST/SM. These studies may ultimately lead to important insights into transformation of benign nevi into melanomas and identify new therapeutic targets in advanced melanoma.

Spitzoid melanoma (SM) and atypical Spitz tumors (AST) are rare neoplasms that affect children. SM and AST often pose a diagnostic challenge, as these lesions are often hard to distinguish from adult type malignant melanoma (MM) based on pathologic criteria [1,2]. The optimal management of SM/AST remains controversial [3]. Management usually consists of surgical removal of large, rapidly changing lesions with atypical dermoscopic or histological features [4,5]. Complete lymphadenectomy is usually performed in patients with sentinel lymph node positivity [5–8].

This report summarizes our institutional experience in treating these rare pediatric tumors.

Methods

We retrospectively collected clinical data from the melanoma database in patients ≤21 years of age diagnosed with SM/AST from 1980–2014, at Riley Hospital for Children, Indianapolis, after approval by our Institutional review Board. Every year, approximately two to four pediatric patients with newly diagnosed melanoma are diagnosed and treated at Riley Hospital.

The tumors were classified as AST or SM if they displayed all or some of the following features: asymmetry, epidermal effacement, high cellular density, cytological atypia, high nuclear/cytoplasmic ration, mitotic rate >2/mm2 (AST and SM) and a high degree of pleomorphism (SM) [9,10]. In all patients, a second opinion from an external expert pathologist was obtained, to confirm the diagnosis. However, some of the cases were diagnosed and managed in the 1980s and 1990s, and the pathological criteria at that time might have been less defined than the current criteria. Thus a limitation of this study is observer bias.

Results

Ten cases of SM/AST were diagnosed and managed at our institution from 1980–2014. The histopathological features and patient demographics, treatment and outcomes are summarized in Tables 1 & 2, respectively.

Table 1. . Histopathologic features.

Patient Histology Breslow thickness Clark level Ulcer V/L invasion Mitosis/mm2 Margins Associated nevus Cytogenetics
1 SM NA NA NA NA NA Negative No NA

2 SM Intermediate (1–3.99 mm) IV No No 3 Positive No 6q-, 9-, 10-

3 SM Intermediate IV No NA 12 Positive No 1q-, 2p-, 2q-, 9- (total loss), 19-

4 SM Intermediate NA NA NA NA Negative Yes 3q-, 6p-

5 SM Intermediate NA No NA NA Positive No 2p-, 18-, 16q-, 12q-

6 SM Intermediate III No No NA Negative Yes NA

7 SM Higher risk (>4 mm) IV Yes No NA Negative NA 11p+, 15+, 18+, 22+, 3q-, 13q-

8 AST Intermediate IV No NA NA Negative Yes NA

9 AST NA NA No NA NA NA Yes NA

10 AST Higher risk NA No NA NA NA Yes NA

AST: Atypical Spitz tumors; SM: Spitzoid melanoma; NA: Information not available.

Table 2. . Patient demographics and clinical follow-up.

Patient Age (years) Sex Site Wide excision Sentinel LN Regional LND Stage Adjuvant therapy Status Follow-up (RFS)
1 14 Male Extremity Yes No No II No Alive 24

2 6 Female Extremity Yes Yes(+) Yes (-) IIIA No Alive 17

3 16 Female Extremity Yes Yes (+) Yes (-) IIIIA IFN × 2 yrs Alive 27

4 10 Male Extremity Yes Yes (+) Yes (-) IIIA IFN × 1 year Alive 34

5 2 Female Head/neck Yes Yes (+) Yes (-) IIIC No Alive 50

6 13 Male Head/neck Yes Yes (+) Yes (+) IIIC IFN × 1 year, Local RT Alive 34

7 4 Female Head/neck Yes No No IIC No Alive 84

8 14 Male Head/neck Yes Yes (+) Yes (-) III IFN × 48 weeks Alive 12

9 15 Male Head/neck Yes Yes (-) Yes (-) III No Alive 84

10 12 Male Head/neck Yes Yes (+) Yes (+) III IFN Alive 60

Patient 2, 3, 5 and 6 underwent a re-excision after partial resection.

Patient 6 had multiple regional LN that were positive and had extra capsular local spread.

LN: Lymph node; LND: Lymph node dissection; RFS: Relapse-free survival; RT: Radiotherapy.

The median age of the ten patients was 12.5 years. The median follow-up was for 34 months. All patients were alive, with no local or distant disease recurrence. Six out of ten patients had a complete and wide excision of the primary lesion with negative margins. Four patients (patient # 2, 3, 5 and 6) underwent a re-excision with negative margins. A SLB was performed for all stage III lesion (8/10 = 80%) and was found to positive (SLB+) for atypical malignant cells in 7/8 (87.5%, 6/8 of these had isolated tumor cells and 1/8 [patient #6] had extensive involvement) samples. All SLB+ patients underwent a regional lymph node dissection (LND) but only 2/8 (25%) of the dissected lymph nodes were positive (LND+) for malignant cells.

Two patients (# 2 and 5), who were SLB+/LND-, were observed and received no further therapy. Three patients (patient numbers 3, 4 and 8, SLB+, LND-) received adjuvant immunotherapy (interferon [IFN]) (Table 2). Both patients with regional nodal disease (patient numbers 6 and 10) received IFN and local radiotherapy (RT) or IFN only respectively (Table 2). Patient numbers 1 and 7 did not undergo a SLB and were presumed to be stage II.

Abnormal and varied cytogenetics were documented in four patients (patient # 2, 3, 5 and 7). Melanoma specific FISH probes, BRAF and RAS mutations were not tested in these patients. Interestingly, the two SLB+ patients, who were observed, were found to have atypical cytogenetic findings of 6q-, total loss of chromosome 9 and 10, and 1q-, 2p-, loss of chromosome 9 and 19, respectively, remained relapse free 17 months and 50 months postresection.

Discussion

SM/AST are rare pediatric tumors that are a distinct clinicopathologic entity, first described by Spitz in 1948. Consistent with our results, a higher incidence of sentinel node positivity (SLB+) has been reported in SM/AST compared with malignant melanoma [6–7,11]. The higher incidence of SLB+ is usually not associated with either extensive nodal involvement or distant metastasis, especially in prepubescent children (only 25% in our case series) and importantly doesn't usually translate into inferior outcomes [7,12–14].

The increased propensity to metastasize to local lymph nodes but not distant sites could be explained by a vigorous immune reaction, better microfiltration, natural involution of lymph nodes in the pediatric population [15]. One could also hypothesize that the malignant cells that are identified in the lymph nodes are nodal melanophages that have been over interpreted as aggressive melanoma cells [16].

Whether or not SM/AST represent a continuum between benign nevi and aggressive adult type malignant melanoma remains a mystery. Majority of malignant melanomas demonstrate chromosomal aberrations and gene amplifications [17,18], and loss of replicative senescence by telomerase overexpression [19] and angiotropism [20]. While the majority of conventional pediatric melanomas demonstrate a bigger array of chromosomal copy number aberrations, they may also demonstrate 6p25, 8q24 and 11q13 gains, deletions of 6q23 and 9p21, like their Spitzoid counterparts [21–25].

Benign Nevi such as blue or congenital nevi usually do not exhibit similar cytogenetic abnormalities or telomerase overexpression but may exhibit angiotropism [26–28].

Using comparative genomic hybridization (CGH) and FISH, gains in 6p25 or 11q13 and homozygous deletions in 9p21 have been identified in SM/AST and were found to be associated with a more aggressive clinical behavior and higher propensity towards metastatic spread beyond the sentinel lymph node [22,29].

Based on this observations, one could speculate that unique chromosomal aberrations and dysregulation of the telomere checkpoint may play a role in determining the phenotype of a SM/AST [24–25,30–32].

SM also have a unique spectral signature further suggesting that these neoplasms are biologically unique and differ in pathogenesis from MM, especially in the prepubertal pediatric age group [18,33]. A small subset of Spitz nevi demonstrate an isolated gain of chromosome 11p and HRAS mutations [32,34], a finding not seen in adult type melanoma. HRAS mutations seem to confer a benign clinical behavior. Unlike conventional melanomas, NRAS mutations are rare in Spitzoid lesions [21,35–36]. More recent studies have demonstrated the presence of additional kinase fusions (ROS1, NTRK1, ALK and RET oncogenes) in adult Spitzoid neoplasms [30], but little is known of the precise role of these oncogenes in the transformation of Spitz nevi into AST and SM.

Activating mutations of BRAF kinase are common in MM [37]. However, BRAF mutations have also been found in benign nevi, suggesting that BRAF mutation is an early mutagenic event that by itself may not confer malignant potential [35,36]. The heterogeneity of BRAF mutations while documented in adult type melanoma and other tumor types, have not been elucidated in pediatric AST or SMs but warrants further investigation [2,21]. Like their adult counterparts, the heterogeneity of BRAF mutations, may ultimately dictate clinical response to BRAF inhibitors in this patient population, and may ultimately provide the foundation for the use of kinase inhibitors for the treatment of refractory SM and ASTs [2,21].

Another biological feature of aggressive MM is the ability to evade effective immune response possibly by inhibiting T-cell activation [38–40]. The PD-1 receptor has been shown to attenuate activated T cells to mount an immune response towards melanoma cells [41–43]. Melanoma cells produce the ligand (PD-L1) which binds PD-1 on the T-cell surface. Recent trials have demonstrated that Ipilimumab, a monoclonal antibody to CTLA-4 receptor and PD-1 is efficacious in advanced melanoma [43].

One could speculate that one of the reasons, majority of ASTs and SMs may not be able to evade host immune response and tend to be limited to local spread, is that they do note attenuate activated T cells via the PD-1 pathway. It is plausible that the ASTs or SMs that are fatal and metastasize may have effectively evaded host response in a similar manner to MM.

Conclusion

A great degree of variability still exists in the management of AST and SM among pediatric oncologists especially regarding the use of adjuvant therapy in these patients.

In the past, metastatic or aggressive AST or SM have been treated as per current MM management guidelines. However, as our case series demonstrates, clinicians should elect to observe fully resected nonmetastatic SLB+ SM/AST, based on the notion that these are biologically a different entity than adult type malignant melanoma. While postpubescent state, presence of homozygous 9p21 deletion or a high number of chromosomal aberrations may confer a higher risk of advanced local-regional disease, it does not necessarily translate into inferior outcomes [13,44]. Our experience further supports the notion that the aggressive clinical approach used in adult melanoma may not be applicable to sentinel node-positive SM/AST, as the relapse or recurrence rate is low with or without adjuvant therapy.

Future perspective

AST and SM are clinically and biologically different from MM. Based on our observations and emerging clinical data these rare pediatric neoplasms may have been overtreated in the past, especially if the treating physician followed current MM management guidelines. The majority of these lesions can simply be excised or re-excised, followed by careful physical, radiological or echotomographic monitoring of the regional nodes.

Little is known about what determines the unique clinical behavior of pediatric AST and SMs, and why these tumors are associated with better outcome even if they metastasize locally. Further molecular studies are needed to understand the unique biological behavior of these rare pediatric tumors. These studies may ultimately lead to important insights into transformation of benign nevi into melanomas and identify new therapeutic targets in advanced melanoma.

Footnotes

Financial & competing interests disclosure

The author has no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

Informed consent disclosure

The author states that they have obtained verbal and written informed consent from the patient/patients for the inclusion of their medical and treatment history within this case report.

References

Papers of special note have been highlighted as: • of interest; •• of considerable interest

  • 1.Paradela S, Fonseca E, Prieto VG. Melanoma in children. Arch. Pathol. Lab. Med. 2011;135(3):307–316. doi: 10.5858/2009-0503-RA.1. [DOI] [PubMed] [Google Scholar]
  • 2.Pappo AS. Pediatric melanoma: the whole (genome) story. Am. Soc. Clin. Oncol. Educ. Book Meeting. 2014:e432–e435. doi: 10.14694/EdBook_AM.2014.34.e432. [DOI] [PubMed] [Google Scholar]; •• Excellent reviews for atypical melanomas in the pediatric age.
  • 3.Reed D, Kudchadkar R, Zager JS, Sondak VK, Messina JL. Controversies in the evaluation and management of atypical melanocytic proliferations in children, adolescents, and young adults. J. Natl Compr. Canc. Netw. 2013;11(6):679–686. doi: 10.6004/jnccn.2013.0087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Haliasos EC, Kerner M, Jaimes N, et al. Dermoscopy for the pediatric dermatologist part III: dermoscopy of melanocytic lesions. Pediatr. Dermatol. 2013;30(3):281–293. doi: 10.1111/pde.12041. [DOI] [PubMed] [Google Scholar]
  • 5.Lallas A, Kyrgidis A, Ferrara G, et al. Atypical Spitz tumours and sentinel lymph node biopsy: a systematic review. Lancet Oncol. 2014;15(4):e178–e183. doi: 10.1016/S1470-2045(13)70608-9. [DOI] [PubMed] [Google Scholar]; • Underscore the importance of observation in atypical pediatric melanomas.
  • 6.Howman-Giles R, Uren RF, Thompson J. Sentinel lymph node biopsy in pediatric and adolescent patients: a proven technique. Ann. Surg. 2014;259(6):e86. doi: 10.1097/SLA.0000000000000350. [DOI] [PubMed] [Google Scholar]; •• Excellent reviews for atypical melanomas in the pediatric age.
  • 7.Mills OL, Marzban S, Zager JS, Sondak VK, Messina JL. Sentinel node biopsy in atypical melanocytic neoplasms in childhood: a single institution experience in 24 patients. J. Cutan. Pathol. 2012;39(3):331–336. doi: 10.1111/j.1600-0560.2011.01853.x. [DOI] [PubMed] [Google Scholar]
  • 8.Berk DR, Labuz E, Dadras SS, Johnson DL, Swetter SM. Melanoma and melanocytic tumors of uncertain malignant potential in children, adolescents and young adults – the Stanford experience 1995–2008. Pediatr. Dermatol. 2010;27(3):244–254. doi: 10.1111/j.1525-1470.2009.01078.x. [DOI] [PubMed] [Google Scholar]; •• Excellent reviews for atypical melanomas in the pediatric age.
  • 9.Mooi WJ, Krausz T. Spitz nevus versus Spitzoid melanoma: diagnostic difficulties, conceptual controversies. Adv. Anat. Pathol. 2006;13(4):147–156. doi: 10.1097/00125480-200607000-00001. [DOI] [PubMed] [Google Scholar]
  • 10.Barnhill RL. The Spitzoid lesion: rethinking Spitz tumors, atypical variants, ‘Spitzoid melanoma’ and risk assessment. Mod. Pathol. Inc. 2006;19(Suppl. 2):S21–S33. doi: 10.1038/modpathol.3800519. [DOI] [PubMed] [Google Scholar]
  • 11.Mu E, Lange JR, Strouse JJ. Comparison of the use and results of sentinel lymph node biopsy in children and young adults with melanoma. Cancer. 2012;118(10):2700–2707. doi: 10.1002/cncr.26578. [DOI] [PubMed] [Google Scholar]
  • 12.Cerrato F, Wallins JS, Webb ML, Mccarty ER, Schmidt BA, Labow BI. Outcomes in pediatric atypical Spitz tumors treated without sentinel lymph node biopsy. Pediatr. Dermatol. 2012;29(4):448–453. doi: 10.1111/j.1525-1470.2011.01699.x. [DOI] [PubMed] [Google Scholar]; •• Excellent reviews for atypical melanomas in the pediatric age.
  • 13.Moore-Olufemi S, Herzog C, Warneke C, et al. Outcomes in pediatric melanoma: comparing prepubertal to adolescent pediatric patients. Ann. Surg. 2011;253(6):1211–1215. doi: 10.1097/SLA.0b013e318217e852. [DOI] [PubMed] [Google Scholar]
  • 14.Aldrink JH, Selim MA, Diesen DL, et al. Pediatric melanoma: a single-institution experience of 150 patients. J. Pediatr. Surg. 2009;44(8):1514–1521. doi: 10.1016/j.jpedsurg.2008.12.003. [DOI] [PubMed] [Google Scholar]
  • 15.Coit DG, Ernstoff MS, Busam KJ. Is pediatric melanoma always malignant? Cancer. 2013;119(22):3910–3913. doi: 10.1002/cncr.28291. [DOI] [PubMed] [Google Scholar]; • Underscore the importance of observation in atypical pediatric melanomas
  • 16.Guitera P, Li LX, Scolyer RA, Menzies SW. Morphologic features of melanophages under in vivo reflectance confocal microscopy. Arch. Dermatol. 2010;146(5):492–498. doi: 10.1001/archdermatol.2009.388. [DOI] [PubMed] [Google Scholar]
  • 17.Demarchis EH, Swetter SM, Jennings CD, Kim J. Fluorescence in situ hybridization analysis of atypical melanocytic proliferations and melanoma in young patients. Pediatr. Dermatol. 2014;31(5):561–569. doi: 10.1111/pde.12382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.North JP, Vemula SS, Bastian BC. Chromosomal copy number analysis in melanoma diagnostics. Methods Mol. Biol. 2014;1102:199–226. doi: 10.1007/978-1-62703-727-3_12. [DOI] [PubMed] [Google Scholar]
  • 19.Miracco C, Margherita De Santi M, Schurfeld K, et al. Quantitative in situ evaluation of telomeres in fluorescence in situ hybridization-processed sections of cutaneous melanocytic lesions and correlation with telomerase activity. Br. J. Dermatol. 2002;146(3):399–408. doi: 10.1046/j.1365-2133.2002.04600.x. [DOI] [PubMed] [Google Scholar]
  • 20.Lugassy C, Barnhill RL. Angiotropic melanoma and extravascular migratory metastasis: a review. Adv. Anat. Pathol. 2007;14(3):195–201. doi: 10.1097/PAP.0b013e31805048d9. [DOI] [PubMed] [Google Scholar]
  • 21.Lu C, Zhang J, Nagahawatte P, et al. The genomic landscape of childhood and adolescent melanoma. J. Invest. Dermatol. 2014;135(3):816–823. doi: 10.1038/jid.2014.425. [DOI] [PMC free article] [PubMed] [Google Scholar]; • Underscore the importance of observation in atypical pediatric melanomas.
  • 22.Gerami P, Scolyer RA, Xu X, et al. Risk assessment for atypical Spitzoid melanocytic neoplasms using FISH to identify chromosomal copy number aberrations. Am. J. Surg. Pathol. 2013;37(5):676–684. doi: 10.1097/PAS.0b013e3182753de6. [DOI] [PubMed] [Google Scholar]; • Underscore the importance of observation in atypical pediatric melanomas.
  • 23.Gerami P, Li G, Pouryazdanparast P, et al. A highly specific and discriminatory FISH assay for distinguishing between benign and malignant melanocytic neoplasms. Am. J. Surg. Pathol. 2012;36(6):808–817. doi: 10.1097/PAS.0b013e31824b1efd. [DOI] [PubMed] [Google Scholar]
  • 24.Gammon B, Beilfuss B, Guitart J, Gerami P. Enhanced detection of Spitzoid melanomas using fluorescence in situ hybridization with 9p21 as an adjunctive probe. Am. J. Surg. Pathol. 2012;36(1):81–88. doi: 10.1097/PAS.0b013e31822d5ff8. [DOI] [PubMed] [Google Scholar]
  • 25.Raskin L, Ludgate M, Iyer RK, et al. Copy number variations and clinical outcome in atypical Spitz tumors. Am. J. Surg. Pathol. 2011;35(2):243–252. doi: 10.1097/PAS.0b013e31820393ee. [DOI] [PubMed] [Google Scholar]; • Underscore the importance of observation in atypical pediatric melanomas.
  • 26.Busam KJ. Molecular pathology of melanocytic tumors. Semin. Diagn. Pathol. 2013;30(4):362–374. doi: 10.1053/j.semdp.2013.11.002. [DOI] [PubMed] [Google Scholar]
  • 27.Kokta V, Hung T, Al Dhaybi R, Lugassy C, Barnhill RL. High prevalence of angiotropism in congenital melanocytic nevi: an analysis of 53 cases. Am. J. Dermatopathol. 2013;35(2):180–183. doi: 10.1097/DAD.0b013e318260908c. [DOI] [PubMed] [Google Scholar]
  • 28.Diaz A, Puig-Butille JA, Valera A, et al. TERT and AURKA gene copy number gains enhance the detection of acral lentiginous melanomas by fluorescence in situ hybridization. J. Mol. Diagn. 2014;16(2):198–206. doi: 10.1016/j.jmoldx.2013.10.009. [DOI] [PubMed] [Google Scholar]
  • 29.Gammon B, Gerami P. Fluorescence in situ hybridization for ambiguous melanocytic tumors. Histol. Histopathol. 2012;27(12):1539–1542. doi: 10.14670/HH-27.1539. [DOI] [PubMed] [Google Scholar]
  • 30.Busam KJ, Kutzner H, Cerroni L, Wiesner T. Clinical and pathologic findings of Spitz nevi and atypical Spitz tumors with ALK fusions. Am. J. Surg. Pathol. 2014;38(7):925–933. doi: 10.1097/PAS.0000000000000187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Barnhill RL, Kutzner H, Schmidt B, et al. Atypical Spitzoid melanocytic neoplasms with angiotropism: a potential mechanism of locoregional involvement. Am. J. Dermatopathol. 2011;33(3):236–243. doi: 10.1097/DAD.0b013e3181f8c0e1. [DOI] [PubMed] [Google Scholar]
  • 32.Bastian BC, Wesselmann U, Pinkel D, Leboit PE. Molecular cytogenetic analysis of Spitz nevi shows clear differences to melanoma. J. Invest. Dermatol. 1999;113(6):1065–1069. doi: 10.1046/j.1523-1747.1999.00787.x. [DOI] [PubMed] [Google Scholar]
  • 33.Gaudi S, Meyer R, Ranka J, et al. Hyperspectral imaging of melanocytic lesions. Am. J. Dermatopathol. 2014;36(2):131–136. doi: 10.1097/DAD.0b013e31829bad10. [DOI] [PubMed] [Google Scholar]
  • 34.Bastian BC, Leboit PE, Pinkel D. Mutations and copy number increase of HRAS in Spitz nevi with distinctive histopathological features. Am. J. Pathol. 2000;157(3):967–972. doi: 10.1016/S0002-9440(10)64609-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Da Forno PD, Pringle JH, Fletcher A, et al. BRAF, NRAS and HRAS mutations in Spitzoid tumours and their possible pathogenetic significance. Br. J. Dermatol. 2009;161(2):364–372. doi: 10.1111/j.1365-2133.2009.09181.x. [DOI] [PubMed] [Google Scholar]
  • 36.Fullen DR, Poynter JN, Lowe L, et al. BRAF and NRAS mutations in Spitzoid melanocytic lesions. Mod. Pathol. Inc. 2006;19(10):1324–1332. doi: 10.1038/modpathol.3800653. [DOI] [PubMed] [Google Scholar]
  • 37.Villanueva MT. Melanoma: blocking BRAF to the BRIM. Nat. Rev. Clin. Oncol. 2014;11(4):179. doi: 10.1038/nrclinonc.2014.32. [DOI] [PubMed] [Google Scholar]
  • 38.Tucci M, Stucci S, Passarelli A, Giudice G, Dammacco F, Silvestris F. The immune escape in melanoma: role of the impaired dendritic cell function. Exp. Rev. Clin. Immunol. 2014;10(10):1395–1404. doi: 10.1586/1744666X.2014.955851. [DOI] [PubMed] [Google Scholar]
  • 39.Tomei S, Bedognetti D, De Giorgi V, et al. The immune-related role of BRAF in melanoma. Mol. Oncol. 2014;9(1):93–104. doi: 10.1016/j.molonc.2014.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sullivan RJ, Fisher DE. Understanding the biology of melanoma and therapeutic implications. Hematol. Oncol. Clin. North Am. 2014;28(3):437–453. doi: 10.1016/j.hoc.2014.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Rodic N, Anders RA, Eshleman JR, et al. PD-L1 expression in melanocytic lesions does not correlate with the BRAF V600E mutation. Cancer Immunol. Res. 2014;3(2):110–115. doi: 10.1158/2326-6066.CIR-14-0145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Massi D, Brusa D, Merelli B, et al. PD-L1 marks a subset of melanomas with a shorter overall survival and distinct genetic and morphological characteristics. Ann. Oncol. 2014;25(12):2433–2442. doi: 10.1093/annonc/mdu452. [DOI] [PubMed] [Google Scholar]
  • 43.Coit DG, Thompson JA, Andtbacka R, et al. Melanoma, version 4.2014. J. Natl Compr. Canc. Netw. 2014;12(5):621–629. doi: 10.6004/jnccn.2014.0066. [DOI] [PubMed] [Google Scholar]
  • 44.Cesinaro AM, Schirosi L, Bettelli S, Migaldi M, Maiorana A. Alterations of 9p21 analysed by FISH and MLPA distinguish atypical Spitzoid melanocytic tumours from conventional Spitz's nevi but do not predict their biological behaviour. Histopathology. 2010;57(4):515–527. doi: 10.1111/j.1365-2559.2010.03653.x. [DOI] [PubMed] [Google Scholar]

Articles from Melanoma Management are provided here courtesy of Taylor & Francis

RESOURCES