Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mod Pathol ; 37(5): 100469, 2024 May.
Article in English | MEDLINE | ID: mdl-38467248

ABSTRACT

NRAS activating mutations are prevalent in melanocytic neoplasia, occurring in a subset of common acquired melanocytic nevi and ∼30% of cutaneous melanomas. In this study, we described a cohort of 7 distinctive melanocytic tumors characterized by activating point mutations in codon 61 of NRAS with amplification of the mutant NRAS allele and shared clinicopathologic features. These tumors occurred predominantly in younger patients, with a median age of 20 years (range, 6-56 years). They presented as papules on the helix of the ear (4 cases) or extremities (3 cases). Microscopically, the tumors were cellular, relatively well-circumscribed, compound, or intradermal proliferations. The tumor cells often extended into the deep reticular dermis and involved the superficial subcutaneous fat in some cases. The melanocytes were epithelioid to spindled with moderate amounts of cytoplasm and conspicuous nucleoli. They were arranged in short plexiform fascicles, nests, and cords. Some cases had occasional pleomorphic and multinucleated melanocytes. Rare dermal mitotic figures were present in all cases. The dermis contained thick collagen bundles and minimal solar elastosis. Follow-up data were available for 5 patients, with a median period of 4.2 years (range, 1-9 years), during which no recurrences or metastases were reported. Our series highlights a clinicopathologically and molecularly distinctive subset of NRAS-mutated tumors with amplification of the mutant NRAS allele.


Subject(s)
GTP Phosphohydrolases , Membrane Proteins , Nevus, Epithelioid and Spindle Cell , Skin Neoplasms , Humans , GTP Phosphohydrolases/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Male , Female , Middle Aged , Membrane Proteins/genetics , Adult , Adolescent , Child , Young Adult , Nevus, Epithelioid and Spindle Cell/genetics , Nevus, Epithelioid and Spindle Cell/pathology , Melanoma/genetics , Melanoma/pathology , Gene Amplification , Melanocytes/pathology , Mutation , Nevus, Pigmented/genetics , Nevus, Pigmented/pathology , Point Mutation
2.
Pigment Cell Melanoma Res ; 34(5): 987-993, 2021 09.
Article in English | MEDLINE | ID: mdl-33522711

ABSTRACT

Patients with non-supernumerary ring chromosome 7 syndrome have an increased incidence of hemangiomas, café-au-lait spots, and melanocytic nevi. The mechanism for the increased incidence of these benign neoplasms is unknown. We present the case of a 22-year-old man with ring chromosome 7 and multiple melanocytic nevi. Two nevi, one on the right ear and the other on the right knee, were biopsied and diagnosed as desmoplastic Spitz nevi. Upon targeted next-generation DNA sequencing, both harbored BRAF fusions. Copy number alterations and fluorescence in situ hybridization (FISH) for BRAF suggested that the fusions arose on the ring chromosome 7. Hence, one reason for increased numbers of nevi in patients with non-supernumerary ring chromosome 7 syndrome may be increased likelihood of BRAF fusions, due to the instability of the ring chromosome.


Subject(s)
Chromosome Disorders , Ear Neoplasms , Nevus, Epithelioid and Spindle Cell , Oncogene Proteins, Fusion , Proto-Oncogene Proteins B-raf , Ring Chromosomes , Skin Neoplasms , Adult , Chromosome Disorders/genetics , Chromosome Disorders/metabolism , Chromosomes, Human, Pair 7/genetics , Chromosomes, Human, Pair 7/metabolism , Chromosomes, Human, Pair 7/physiology , Ear Neoplasms/genetics , Ear Neoplasms/metabolism , Ear Neoplasms/pathology , Humans , Male , Nevus, Epithelioid and Spindle Cell/genetics , Nevus, Epithelioid and Spindle Cell/metabolism , Nevus, Epithelioid and Spindle Cell/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
3.
J Pathol ; 240(3): 282-290, 2016 11.
Article in English | MEDLINE | ID: mdl-27477320

ABSTRACT

Oncogenic fusions in TRK family receptor tyrosine kinases have been identified in several cancers and can serve as therapeutic targets. We identified ETV6-NTRK3, MYO5A-NTRK3 and MYH9-NTRK3 fusions in Spitz tumours, and demonstrated that NTRK3 fusions constitutively activate the mitogen-activated protein kinase, phosphoinositide 3-kinase and phospholipase Cγ1 pathways in melanocytes. This signalling was inhibited by DS-6051a, a small-molecule inhibitor of NTRK1/2/3 and ROS1. NTRK3 fusions expand the range of oncogenic kinase fusions in melanocytic neoplasms and offer targets for a small subset of melanomas for which no targeted options currently exist. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Discoidin Domain Receptor 2/genetics , Molecular Motor Proteins/genetics , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Nevus, Epithelioid and Spindle Cell/enzymology , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Skin Neoplasms/enzymology , Adolescent , Adult , Aged , Child , Child, Preschool , Comparative Genomic Hybridization , Discoidin Domain Receptor 2/metabolism , Female , Humans , Male , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Molecular Motor Proteins/metabolism , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Nevus, Epithelioid and Spindle Cell/genetics , Nevus, Epithelioid and Spindle Cell/pathology , Oncogene Fusion , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/metabolism , Sequence Analysis, DNA , Sequence Analysis, RNA , Skin Neoplasms/genetics , Skin Neoplasms/pathology , ETS Translocation Variant 6 Protein
4.
Nat Commun ; 6: 7174, 2015 May 27.
Article in English | MEDLINE | ID: mdl-26013381

ABSTRACT

Oncogenic gene fusions have been identified in many cancers and many serve as biomarkers or targets for therapy. Here we identify six different melanocytic tumours with genomic rearrangements of MET fusing the kinase domain of MET in-frame to six different N-terminal partners. These tumours lack activating mutations in other established melanoma oncogenes. We functionally characterize two of the identified fusion proteins (TRIM4-MET and ZKSCAN1-MET) and find that they constitutively activate the mitogen-activated protein kinase (MAPK), phosphoinositol-3 kinase (PI3K) and phospholipase C gamma 1 (PLCγ1) pathways. The MET inhibitors cabozantinib (FDA-approved for progressive medullary thyroid cancer) and PF-04217903 block their activity at nanomolar concentrations. MET fusion kinases thus provide a potential therapeutic target for a rare subset of melanoma for which currently no targeted therapeutic options currently exist.


Subject(s)
Gene Rearrangement , Melanoma, Experimental/genetics , Nevus, Epithelioid and Spindle Cell/genetics , Oncogene Fusion , Proto-Oncogene Proteins c-met/genetics , Adult , Animals , Cell Line , Female , Humans , Male , Mice , Middle Aged
5.
Am J Surg Pathol ; 39(5): 581-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25602801

ABSTRACT

Activating kinase fusions have recently been described as early oncogenic events that are mutually exclusive with HRAS and BRAF mutations in Spitz tumors. Here, we report a series of 32 Spitz tumors with ALK fusions (6 Spitz nevi, 22 atypical Spitz tumors, and 4 spitzoid melanomas) in patients ranging from 5 months to 64 years (median=12 y) of age. The tumors typically presented as exophytic papules on the extremities and were occasionally darkly pigmented. In addition to ALK fusions previously described in other tumor types (NPM1-ALK, TPR-ALK), we identified 2 novel ALK fusions (CLIP1-ALK and GTF3C2-ALK) in our cohort of Spitz tumors. Array comparative genomic hybridization of 19 of these tumors demonstrated a high frequency of chromosome 2 aberrations (where ALK resides, 63%) and chromosome 1p loss in 37% of the cases. Spitz tumors with ALK fusions demonstrated unique histopathologic features. Clefts and small vesicle-like spaces were arrayed between plump spindled melanocytes with fibrillar cytoplasm and enlarged nuclei. These melanocytes were typically arrayed in elongated and fusiform nests with radial orientation. The tumors often had extension into the dermis or subcutis with a wedge-shaped or bulbous lower border (45% and 17%, respectively). An infiltrative growth pattern was often present at the periphery of the tumor and was highlighted by ALK immunohistochemistry. In conclusion, Spitz tumors with ALK rearrangement show distinct histopathologic features that should aid in improving classification of these diagnostically challenging tumors.


Subject(s)
Nevus, Epithelioid and Spindle Cell/genetics , Nevus, Epithelioid and Spindle Cell/pathology , Receptor Protein-Tyrosine Kinases/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Adolescent , Anaplastic Lymphoma Kinase , Child , Child, Preschool , Comparative Genomic Hybridization , Female , Genomics , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Middle Aged , Nucleophosmin , Oncogene Proteins, Fusion/genetics , Young Adult
6.
Cell Rep ; 9(4): 1228-34, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25456125

ABSTRACT

Somatic mutations in cancer are more frequent in heterochromatic and late-replicating regions of the genome. We report that regional disparities in mutation density are virtually abolished within transcriptionally silent genomic regions of cutaneous squamous cell carcinomas (cSCCs) arising in an XPC(-/-) background. XPC(-/-) cells lack global genome nucleotide excision repair (GG-NER), thus establishing differential access of DNA repair machinery within chromatin-rich regions of the genome as the primary cause for the regional disparity. Strikingly, we find that increasing levels of transcription reduce mutation prevalence on both strands of gene bodies embedded within H3K9me3-dense regions, and only to those levels observed in H3K9me3-sparse regions, also in an XPC-dependent manner. Therefore, transcription appears to reduce mutation prevalence specifically by relieving the constraints imposed by chromatin structure on DNA repair. We model this relationship among transcription, chromatin state, and DNA repair, revealing a new, personalized determinant of cancer risk.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA Repair/genetics , Genome, Human/genetics , Heterochromatin/genetics , Mutation Rate , Skin Neoplasms/genetics , Transcription, Genetic , DNA Packaging/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Germ Cells/metabolism , Humans , Proto-Oncogene Proteins/genetics
7.
Am J Surg Pathol ; 38(8): 1088-95, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24705312

ABSTRACT

Germline loss-of-function mutations in BAP1 are associated with the development of cutaneous melanocytic tumors with some histopathologic characteristics seen in Spitz nevi. Similar melanocytic tumors occurring in a sporadic setting have been demonstrated to have biallelic loss of BAP1. In some of these sporadic tumors, loss of BAP1 occurs through mutation of 1 allele and genomic loss of the other. We screened our database of comparative genomic hybridization profiles of ambiguous melanocytic tumors to identify cases with a single genomic event involving loss of the BAP1 locus. The prevalence of tumors with a single genomic event involving loss of BAP1 was 6.7% in our study population. We further characterized the BAP1 status in 17 of these tumors with available additional material, confirming loss of BAP1 in all cases. We describe BAP1 loss in a blue nevus-like melanoma and further expand the histopathologic spectrum of spitzoid melanocytic neoplasms with BAP1 loss.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Melanoma/genetics , Skin Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Adolescent , Adult , Aged , Child , Comparative Genomic Hybridization , Female , Germ-Line Mutation , Humans , Male , Middle Aged , Young Adult
8.
Methods Mol Biol ; 1102: 199-226, 2014.
Article in English | MEDLINE | ID: mdl-24258981

ABSTRACT

The majority of melanocytic neoplasms can be correctly diagnosed using routine histopathologic analysis. However, a significant minority of tumors have ambiguous histopathologic attributes that overlap between melanocytic nevi and melanoma. Ancillary tests that assist in distinguishing potentially lethal melanomas from benign melanocytic nevi with atypical histopathologic features are available, but still need refining.Most melanomas have chromosomal copy number aberrations, frequently involving multiple chromosomes. With rare exceptions, such anomalies are not found in melanocytic nevi. This difference formed the basis to develop assays that can help distinguish melanoma from nevi by fluorescence in situ hybridization (FISH) and comparative genomic hybridization (CGH). FISH can detect chromosomal copy number changes of a limited number of loci within individual cells. By contrast, CGH assesses copy number across the entire genome, but typically is performed on bulk cell populations so that copy number changes in individual cells or subpopulations of cells can go undetected. Both FISH and CGH have been used to provide genomic information in histopathologically ambiguous melanocytic tumors that can assist pathologists make correct diagnoses.


Subject(s)
Chromosomes, Human/genetics , DNA Copy Number Variations/genetics , Melanoma/diagnosis , Melanoma/genetics , Pathology, Molecular/methods , Comparative Genomic Hybridization , Humans , In Situ Hybridization, Fluorescence , Nucleic Acid Denaturation , Paraffin Embedding , Tissue Fixation
9.
Pigment Cell Melanoma Res ; 26(6): 845-51, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23890088

ABSTRACT

BRAF is the most prevalent oncogene and an important therapeutic target in melanoma. In some cancers, BRAF is activated by rearrangements that fuse its kinase domain to 5' partner genes. We examined 848 comparative genomic hybridization profiles of melanocytic tumors and found copy number transitions within BRAF in 10 tumors, of which six could be further characterized by sequencing. In all, the BRAF kinase domain was fused in-frame to six N-terminal partners. No other mutations were identified in melanoma oncogenes. One of the seven melanoma cell lines without known oncogenic mutations harbored a similar BRAF fusion, which constitutively activated the MAP kinase pathway. Sorafenib, but not vemurafenib, could block MAP kinase pathway activation and proliferation of the cell line at clinically relevant concentrations, whereas BRAF(V) (600E) mutant melanoma cell lines were significantly more sensitive to vemurafenib. The patient from whom the cell line was derived showed a durable clinical response to sorafenib.


Subject(s)
Melanocytes/pathology , Melanoma/drug therapy , Melanoma/enzymology , Molecular Targeted Therapy , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/enzymology , Adolescent , Adult , Child, Preschool , Enzyme Activation/drug effects , Female , Gene Rearrangement/drug effects , Humans , Indoles/pharmacology , Indoles/therapeutic use , MAP Kinase Signaling System/drug effects , Male , Melanocytes/drug effects , Melanocytes/enzymology , Melanoma/pathology , Middle Aged , Nevus, Epithelioid and Spindle Cell/pathology , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Sorafenib , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Vemurafenib , Young Adult
10.
J Cutan Pathol ; 39(5): 521-5, 2012 May.
Article in English | MEDLINE | ID: mdl-22416678

ABSTRACT

We present a unique dermal tumor for which we propose the term plexiform melanocytic schwanomma. The proliferation consisted of lobules of epithelioid and spindled cells with S100, Melan-A and HMB-45 positivity but without obvious melanin pigmentation. The nuclei were moderately pleomorphic in some areas, and in a few areas the mitotic index was elevated. Schwannian differentiation was inferred from the presence of areas with nuclear palisading resembling Verocay bodies, from plexiform architecture and from the presence of a thin rim of EMA positivity around the tumor. Array-based comparative genomic hybridization showed genomic losses that overlap with those seen in sporadic schwanomma. The differential diagnosis included melanoma, melanotic schwannoma and cutaneous melanocytoneuroma, and we compare and contrast our case with these entities.


Subject(s)
Dermis , Melanoma , Neurilemmoma , Skin Neoplasms , Adult , Cell Proliferation , Dermis/metabolism , Dermis/pathology , Female , Humans , MART-1 Antigen/metabolism , Melanoma/metabolism , Melanoma/pathology , Melanoma-Specific Antigens/metabolism , Neurilemmoma/metabolism , Neurilemmoma/pathology , S100 Proteins/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Pigmentation , gp100 Melanoma Antigen
11.
Am J Dermatopathol ; 34(6): 668-70, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22456515

ABSTRACT

We present a case that highlights the use of two maneuvers useful in the diagnosis of spindle cell melanoma. A shave biopsy from the cheek of a 58-year-old man demonstrated a thin invasive melanoma of 0.3 mm thickness with a less than 2-mm-wide intra-epidermal component. Below this melanocytic lesion, but not contiguous with it, there was a transected S-100-positive and Melan-A-negative spindle cell proliferation. Upon re-excision, no residuum of conventional melanoma was identified, but a residual spindle cell neoplasm that was 4 mm in diameter, nodular, well-circumscribed, cytologically bland, and S-100 positive was noted. At our consensus conference, our group favored neurofibroma but agreed that spindle cell melanoma could not be excluded based on histopathologic features alone. To further address the differential diagnosis, we performed CD34 staining that demonstrated lack of a CD34 fingerprint. We also completed array-based comparative genomic hybridization, which demonstrated gain of chromosome 6p, loss of 6p and gain of 7. These two methods of analysis support a diagnosis of spindle cell melanoma.


Subject(s)
Antigens, CD34/genetics , Comparative Genomic Hybridization/methods , DNA Fingerprinting/methods , Melanoma/genetics , Neurofibroma/genetics , Skin Neoplasms/genetics , Antigens, CD34/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Biopsy , Cheek , Humans , Male , Melanoma/diagnosis , Melanoma/pathology , Middle Aged , Neurofibroma/diagnosis , Neurofibroma/pathology , Skin/metabolism , Skin/pathology , Skin Neoplasms/diagnosis , Skin Neoplasms/pathology
12.
Proc Natl Acad Sci U S A ; 108(43): 17761-6, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-22006338

ABSTRACT

Squamous cell carcinomas (SCCs) are one of the most frequent forms of human malignancy, but, other than TP53 mutations, few causative somatic aberrations have been identified. We identified NOTCH1 or NOTCH2 mutations in ~75% of cutaneous SCCs and in a lesser fraction of lung SCCs, defining a spectrum for the most prevalent tumor suppressor specific to these epithelial malignancies. Notch receptors normally transduce signals in response to ligands on neighboring cells, regulating metazoan lineage selection and developmental patterning. Our findings therefore illustrate a central role for disruption of microenvironmental communication in cancer progression. NOTCH aberrations include frameshift and nonsense mutations leading to receptor truncations as well as point substitutions in key functional domains that abrogate signaling in cell-based assays. Oncogenic gain-of-function mutations in NOTCH1 commonly occur in human T-cell lymphoblastic leukemia/lymphoma and B-cell chronic lymphocytic leukemia. The bifunctional role of Notch in human cancer thus emphasizes the context dependency of signaling outcomes and suggests that targeted inhibition of the Notch pathway may induce squamous epithelial malignancies.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Communication/genetics , Lung Neoplasms/genetics , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Signal Transduction/genetics , Skin Neoplasms/genetics , Base Sequence , Codon, Nonsense/genetics , Electrophoretic Mobility Shift Assay , Humans , Lod Score , Molecular Sequence Data , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...