Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mod Pathol ; 37(5): 100469, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38467248

RESUMO

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.


Assuntos
GTP Fosfo-Hidrolases , Proteínas de Membrana , Nevo de Células Epitelioides e Fusiformes , Neoplasias Cutâneas , Humanos , GTP Fosfo-Hidrolases/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Masculino , Feminino , Pessoa de Meia-Idade , Proteínas de Membrana/genética , Adulto , Adolescente , Criança , Adulto Jovem , Nevo de Células Epitelioides e Fusiformes/genética , Nevo de Células Epitelioides e Fusiformes/patologia , Melanoma/genética , Melanoma/patologia , Amplificação de Genes , Melanócitos/patologia , Mutação , Nevo Pigmentado/genética , Nevo Pigmentado/patologia , Mutação Puntual
2.
J Pathol ; 240(3): 282-290, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27477320

RESUMO

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.


Assuntos
Receptor com Domínio Discoidina 2/genética , Proteínas Motores Moleculares/genética , Cadeias Pesadas de Miosina/genética , Miosina Tipo V/genética , Nevo de Células Epitelioides e Fusiformes/enzimologia , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Repressoras/genética , Neoplasias Cutâneas/enzimologia , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Hibridização Genômica Comparativa , Receptor com Domínio Discoidina 2/metabolismo , Feminino , Humanos , Masculino , Melanoma/enzimologia , Melanoma/genética , Melanoma/patologia , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Motores Moleculares/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Nevo de Células Epitelioides e Fusiformes/genética , Nevo de Células Epitelioides e Fusiformes/patologia , Fusão Oncogênica , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Repressoras/metabolismo , Análise de Sequência de DNA , Análise de Sequência de RNA , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Variante 6 da Proteína do Fator de Translocação ETS
3.
Proc Natl Acad Sci U S A ; 108(43): 17761-6, 2011 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-22006338

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/genética , Comunicação Celular/genética , Neoplasias Pulmonares/genética , Receptor Notch1/genética , Receptor Notch2/genética , Transdução de Sinais/genética , Neoplasias Cutâneas/genética , Sequência de Bases , Códon sem Sentido/genética , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Escore Lod , Dados de Sequência Molecular , Análise de Sequência de DNA
4.
J Cutan Pathol ; 39(5): 521-5, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22416678

RESUMO

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.


Assuntos
Derme , Melanoma , Neurilemoma , Neoplasias Cutâneas , Adulto , Proliferação de Células , Derme/metabolismo , Derme/patologia , Feminino , Humanos , Antígeno MART-1/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Antígenos Específicos de Melanoma/metabolismo , Neurilemoma/metabolismo , Neurilemoma/patologia , Proteínas S100/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Pigmentação da Pele , Antígeno gp100 de Melanoma
5.
Am J Dermatopathol ; 34(6): 668-70, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22456515

RESUMO

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.


Assuntos
Antígenos CD34/genética , Hibridização Genômica Comparativa/métodos , Impressões Digitais de DNA/métodos , Melanoma/genética , Neurofibroma/genética , Neoplasias Cutâneas/genética , Antígenos CD34/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Biópsia , Bochecha , Humanos , Masculino , Melanoma/diagnóstico , Melanoma/patologia , Pessoa de Meia-Idade , Neurofibroma/diagnóstico , Neurofibroma/patologia , Pele/metabolismo , Pele/patologia , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/patologia
6.
Pigment Cell Melanoma Res ; 34(5): 987-993, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33522711

RESUMO

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.


Assuntos
Transtornos Cromossômicos , Neoplasias da Orelha , Nevo de Células Epitelioides e Fusiformes , Proteínas de Fusão Oncogênica , Proteínas Proto-Oncogênicas B-raf , Cromossomos em Anel , Neoplasias Cutâneas , Adulto , Transtornos Cromossômicos/genética , Transtornos Cromossômicos/metabolismo , Cromossomos Humanos Par 7/genética , Cromossomos Humanos Par 7/metabolismo , Cromossomos Humanos Par 7/fisiologia , Neoplasias da Orelha/genética , Neoplasias da Orelha/metabolismo , Neoplasias da Orelha/patologia , Humanos , Masculino , Nevo de Células Epitelioides e Fusiformes/genética , Nevo de Células Epitelioides e Fusiformes/metabolismo , Nevo de Células Epitelioides e Fusiformes/patologia , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
7.
Am J Surg Pathol ; 39(5): 581-91, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25602801

RESUMO

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.


Assuntos
Nevo de Células Epitelioides e Fusiformes/genética , Nevo de Células Epitelioides e Fusiformes/patologia , Receptores Proteína Tirosina Quinases/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Adolescente , Quinase do Linfoma Anaplásico , Criança , Pré-Escolar , Hibridização Genômica Comparativa , Feminino , Genômica , Humanos , Hibridização in Situ Fluorescente , Lactente , Masculino , Pessoa de Meia-Idade , Nucleofosmina , Proteínas de Fusão Oncogênica/genética , Adulto Jovem
8.
Nat Commun ; 6: 7174, 2015 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-26013381

RESUMO

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.


Assuntos
Rearranjo Gênico , Melanoma Experimental/genética , Nevo de Células Epitelioides e Fusiformes/genética , Fusão Oncogênica , Proteínas Proto-Oncogênicas c-met/genética , Adulto , Animais , Linhagem Celular , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade
9.
Methods Mol Biol ; 1102: 199-226, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24258981

RESUMO

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.


Assuntos
Cromossomos Humanos/genética , Variações do Número de Cópias de DNA/genética , Melanoma/diagnóstico , Melanoma/genética , Patologia Molecular/métodos , Hibridização Genômica Comparativa , Humanos , Hibridização in Situ Fluorescente , Desnaturação de Ácido Nucleico , Inclusão em Parafina , Fixação de Tecidos
10.
Am J Surg Pathol ; 38(8): 1088-95, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24705312

RESUMO

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.


Assuntos
Cromossomos Humanos Par 3/genética , Melanoma/genética , Neoplasias Cutâneas/genética , Proteínas Supressoras de Tumor/genética , Ubiquitina Tiolesterase/genética , Adolescente , Adulto , Idoso , Criança , Hibridização Genômica Comparativa , Feminino , Mutação em Linhagem Germinativa , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
11.
Cell Rep ; 9(4): 1228-34, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25456125

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/genética , Reparo do DNA/genética , Genoma Humano/genética , Heterocromatina/genética , Taxa de Mutação , Neoplasias Cutâneas/genética , Transcrição Gênica , Empacotamento do DNA/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Células Germinativas/metabolismo , Humanos , Proteínas Proto-Oncogênicas/genética
12.
Pigment Cell Melanoma Res ; 26(6): 845-51, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23890088

RESUMO

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.


Assuntos
Melanócitos/patologia , Melanoma/tratamento farmacológico , Melanoma/enzimologia , Terapia de Alvo Molecular , Proteínas de Fusão Oncogênica/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Neoplasias Cutâneas/enzimologia , Adolescente , Adulto , Pré-Escolar , Ativação Enzimática/efeitos dos fármacos , Feminino , Rearranjo Gênico/efeitos dos fármacos , Humanos , Indóis/farmacologia , Indóis/uso terapêutico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Melanócitos/efeitos dos fármacos , Melanócitos/enzimologia , Melanoma/patologia , Pessoa de Meia-Idade , Nevo de Células Epitelioides e Fusiformes/patologia , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Compostos de Fenilureia/farmacologia , Compostos de Fenilureia/uso terapêutico , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Sorafenibe , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Vemurafenib , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA