Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Skin Health Dis ; 2(2): e95, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35677930

RESUMO

Background: Renal transplant recipients (RTRs) are at increased risk of keratinocyte cancer (KC), especially cutaneous squamous cell carcinoma (cSCC). Previous studies identified a genetic variant of the Methylenetetrahydrofolate Reductase (MTHFR) gene, C677T, which conferred a risk for diagnosis of cSCC in Irish RTRs. Objective: We sought to find further genetic variation in MTHFR and overlap genes that may be associated with a diagnosis of KC in RTRs. Methods: Genotyping of a combined RTR population (n = 821) from two centres, Ireland (n = 546) and the USA (n = 275), was performed. This included 290 RTRs with KC and 444 without. Eleven single nucleotide polymorphisms (SNPs) in the MTHFR gene and seven in the overlap gene MTHFR Chloride transport protein 6 (CLCN6) were evaluated and association explored by time to event analysis (from transplant to first KC) using Cox proportional hazards model. Results: Polymorphism at MTHFR CLCN6 (rs9651118) was significantly associated with KC in RTRs (HR 1.50, 95% CI 1.17-1.91, p < 0.00061) and cSCC (HR 1.63, 95% CI 1.14-2.34, p = 0.007). A separate SNP, MTHFR C677T, was also significantly associated with KC in the Irish population (HR 1.31, 95% CI 1.05-1.63, p = 0.016), but not American RTRs. Conclusions: We report the association of a SNP in the MTHFR overlap gene, CLCN6 and KC in a combined RTR population. While the exact function of CLCN6 is not known, it is proposed to be involved in folate availability. Future applications could include incorporation in a polygenic risk score for KC in RTRs to help identify those at increased risk beyond traditional risk factor assessment.

2.
Br J Dermatol ; 185(2): 282-293, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34060071

RESUMO

Cancer is caused by the accumulation of pathogenic alterations of the genome and epigenome that result in permanent changes that disrupt cellular homeostasis. The genes that become corrupted in this process vary among different tumour types, reflecting specific vulnerabilities and dependencies of the cell from which the cancer originated. This also applies to 'melanoma', a cancer that constitutes not one, but multiple diseases that can be separated based on their cell of origin, aetiology, clinical appearance and course, and response to treatment. In this article, we review the current classification of melanoma within distinct evolutionary pathways and the associated genetic alterations. In addition, we review the application of molecular diagnostics to the diagnosis of melanocytic tumours in the context of histopathological assessment.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Melanoma/genética , Mutação/genética , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/genética
3.
Ann Oncol ; 28(6): 1380-1387, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28327988

RESUMO

BACKGROUND: The single-arm, phase II Tasigna Efficacy in Advanced Melanoma (TEAM) trial evaluated the KIT-selective tyrosine kinase inhibitor nilotinib in patients with KIT-mutated advanced melanoma without prior KIT inhibitor treatment. PATIENTS AND METHODS: Forty-two patients with KIT-mutated advanced melanoma were enrolled and treated with nilotinib 400 mg twice daily. TEAM originally included a comparator arm of dacarbazine (DTIC)-treated patients; the design was amended to a single-arm trial due to an observed low number of KIT-mutated melanomas. Thirteen patients were randomized to DTIC before the protocol amendment removing this study arm. The primary endpoint was objective response rate (ORR), determined according to Response Evaluation Criteria In Solid Tumors. RESULTS: ORR was 26.2% (n = 11/42; 95% CI, 13.9%-42.0%), sufficient to reject the null hypothesis (ORR ≤10%). All observed responses were partial responses (PRs; median response duration, 7.1 months). Twenty patients (47.6%) had stable disease and 10 (23.8%) had progressive disease; 1 (2.4%) response was unknown. Ten of the 11 responding patients had exon 11 mutations, four with an L576P mutation. The median progression-free survival and overall survival were 4.2 and 18.0 months, respectively. Three of the 13 patients on DTIC achieved a PR, and another patient had a PR following switch to nilotinib. CONCLUSION: Nilotinib activity in patients with advanced KIT-mutated melanoma was similar to historical data from imatinib-treated patients. DTIC treatment showed potential activity, although the low patient number limits interpretation. Similar to previously reported results with imatinib, nilotinib showed greater activity among patients with an exon 11 mutation, including L576P, suggesting that nilotinib may be an effective treatment option for patients with specific KIT mutations. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov, NCT01028222.


Assuntos
Antineoplásicos/uso terapêutico , Mutação , Proteínas Proto-Oncogênicas c-kit/genética , Pirimidinas/uso terapêutico , Idoso , Antineoplásicos/efeitos adversos , Dacarbazina/uso terapêutico , Éxons , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Pirimidinas/efeitos adversos , Análise de Sobrevida
4.
Br J Cancer ; 112(8): 1326-31, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25867272

RESUMO

BACKGROUND: Simultaneous chemotherapy with vascular endothelial growth factor (VEGF) inhibition has not shown additional benefit over chemotherapy alone in advanced melanoma. We tested administration of the potent VEGF inhibitor axitinib followed by paclitaxel/carboplatin to determine whether enhanced tumour proliferation during axitinib withdrawal leads to sustained chemosensitivity. METHODS: We conducted a prospective phase II trial in metastatic melanoma patients with ECOG performance status 0-1 and normal organ function. Axitinib 5 mg PO b.i.d. was taken on days 1-14 of each 21-day treatment cycle, and carboplatin (AUC=5) with paclitaxel (175 mg m(-2)) was administered on day 1 starting with cycle 2. 3'-Deoxy-3'-(18)F-fluorothymidine ((18)F-FLT)-PET scans were performed in five patients to assess tumour proliferation on days 1, 14, 17, and 20 of cycle 1. Molecular profiling for BRAF was performed for all patients with cutaneous, acral, or mucosal melanoma. RESULTS: The treatment was well tolerated. The most common grade 3 AEs were hypertension, neutropenia, and anaemia. Grade 4 non-haematologic AEs were not observed. Four of five patients completing (18)F-FLT-PET scans showed increases (23-92%) in SUV values during the axitinib holiday. Of 36 evaluable patients, there were 8 confirmed PRs by Response Evaluation Criteria in Solid Tumors. Overall, 20 patients had SD and 8 had PD as the best response. The median PFS was 8.7 months and the median overall survival was 14.0 months. Five BRAF(V600E/K) patients had significantly worse PFS than patients without these mutations. CONCLUSIONS: Axitinib followed by carboplatin and paclitaxel was well tolerated and effective in BRAF wild-type metastatic melanoma. 3'-Deoxy-3'-(18)F-fluorothymidine-PET scans showed increased proliferation during axitinib withdrawal.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Imidazóis/administração & dosagem , Indazóis/administração & dosagem , Melanoma/tratamento farmacológico , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Proto-Oncogênicas B-raf/genética , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Axitinibe , Carboplatina/administração & dosagem , Carboplatina/efeitos adversos , Didesoxinucleosídeos , Feminino , Humanos , Imidazóis/efeitos adversos , Indazóis/efeitos adversos , Masculino , Melanoma/diagnóstico por imagem , Melanoma/genética , Pessoa de Meia-Idade , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Tomografia por Emissão de Pósitrons/métodos , Estudos Prospectivos , Inibidores de Proteínas Quinases/efeitos adversos , Radiografia , Resultado do Tratamento
5.
Oncogene ; 33(39): 4724-34, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-24141786

RESUMO

Uveal melanoma (UM) is a genetically and biologically distinct type of melanoma, and once metastatic there is no effective treatment currently available. Eighty percent of UMs harbor mutations in the Gαq family members GNAQ and GNA11. Understanding the effector pathways downstream of these oncoproteins is important to identify opportunities for targeted therapy. We report consistent activation of the protein kinase C (PKC) and MAPK pathways as a consequence of GNAQ or GNA11 mutation. PKC inhibition with AEB071 or AHT956 suppressed PKC and MAPK signalling and induced G1 arrest selectively in melanoma cell lines carrying GNAQ or GNA11 mutations. In contrast, treatment with two different MEK inhibitors, PD0325901 and MEK162, inhibited the proliferation of melanoma cell lines irrespective of their mutation status, indicating that in the context of GNAQ or GNA11 mutation MAPK activation can be attributed to activated PKC. AEB071 significantly slowed the growth of tumors in an allograft model of GNAQ(Q209L)-transduced melanocytes, but did not induce tumor shrinkage. In vivo and in vitro studies showed that PKC inhibitors alone were unable to induce sustained suppression of MAP-kinase signaling. However, combinations of PKC and MEK inhibition, using either PD0325901or MEK162, led to sustained MAP-kinase pathway inhibition and showed a strong synergistic effect in halting proliferation and in inducing apoptosis in vitro. Furthermore, combining PKC and MEK inhibition was efficacious in vivo, causing marked tumor regression in a UM xenograft model. Our data identify PKC as a rational therapeutic target for melanoma patients with GNAQ or GNA11 mutations and demonstrate that combined MEK and PKC inhibition is synergistic, with superior efficacy compared to treatment with either approach alone.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Melanoma/tratamento farmacológico , Neoplasias Uveais/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose , Benzimidazóis/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células , Sinergismo Farmacológico , Feminino , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Humanos , Concentração Inibidora 50 , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Melanoma/genética , Melanoma/secundário , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Mutação de Sentido Incorreto , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Pirróis/administração & dosagem , Quinazolinas/administração & dosagem , Transdução de Sinais , Carga Tumoral/efeitos dos fármacos , Neoplasias Uveais/genética , Neoplasias Uveais/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Pigment Cell Melanoma Res ; 25(2): 182-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22236444

RESUMO

The recent identification of frequent activating mutations in GNAQ or GNA11 in uveal melanoma provides an opportunity to better understand the pathogenesis of this melanoma subtype and to develop rational therapeutics to target the cellular effects mediated by these mutations. Cell lines from uveal melanoma tumors are an essential tool for these types of analyses. We report the mutation status of relevant melanoma genes, expression levels of proteins of interest, and DNA fingerprinting of a panel of uveal melanoma cell lines used in the research community.


Assuntos
Melanoma/genética , Neoplasias Uveais/genética , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Genes Neoplásicos/genética , Humanos , Mutação/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
Oncogene ; 31(13): 1649-60, 2012 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21860424

RESUMO

Erythropoietin (Epo) is widely used clinically to treat anemia associated with various clinical conditions including cancer. Data from several clinical trials suggest significant adverse effect of Epo treatment on cancer patient survival. However, controversy exists whether Epo receptor (EpoR) is functional in cancer cells. In this study, we demonstrated that EpoR mRNA expression was detectable in 90.1% of 65 melanoma cell lines, and increased copy number of the Epo and EpoR loci occurred in 30 and 24.6% of 130 primary melanomas, respectively. EpoR knockdown in melanoma cells resulted in diminished ERK phosphorylation in response to Epo stimulation, decreased cell proliferation and increased response to the inhibitory effect of hypoxia and cisplatin in vitro. EpoR knockdown significantly decreased melanoma xenograft size and tumor invasion in vivo. On the contrary, constitutive activation of EpoR activated cell proliferation pathways in melanoma cells and resulted in increased cell proliferation and resistance to hypoxia and cisplatin treatment in vitro. EpoR activation resulted in significantly larger xenografts with increased tumor invasion of surrounding tissue in vivo. Daily administration of recombinant Epo fails to stimulate melanoma growth in vivo, but the treatment increased vascular size in the xenografts. Increased local recurrence after excision of the primary tumors was observed after Epo treatment. Epo induced angiogenesis in Matrigel plug assays, and neutralization of Epo secreted by melanoma cells results in decreased angiogenesis. These data support that EpoR is functional in melanoma and EpoR activation may promote melanoma progression, and suggest that Epo may stimulate angiogenesis and increase survival of melanoma cells under hypoxic condition in vivo.


Assuntos
Eritropoetina/efeitos adversos , Melanoma/genética , Receptores da Eritropoetina/genética , Neoplasias Cutâneas/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Variações do Número de Cópias de DNA , Progressão da Doença , Epoetina alfa , Eritropoetina/genética , Técnicas de Silenciamento de Genes , Humanos , Melanoma/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neovascularização Patológica , Proteínas Recombinantes/efeitos adversos , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional
8.
Br J Dermatol ; 165(5): 1138-42, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21711345

RESUMO

Congenital malignant melanoma within a pre-existing large congenital melanocytic naevus (CMN) is exceedingly rare. Its incidence is difficult to determine due to the small number of reported cases and because of problems associated with diagnosis. Some benign nodular proliferations (called proliferative nodules) arising in CMN, while rare, are significantly more common and can mimic malignant melanoma clinically or histologically. There are no reported cases of congenital melanoma or benign proliferative nodules in CMN in patients who also had eruptive disseminated Spitz naevi. We describe a girl who was noted to have a dark-brown plaque with several large erythematous nodules affecting the scalp at delivery, in addition to multiple erythematous dome-shaped papules that developed in a disseminated manner over several months, beginning at 10 days of age. It was difficult, not only clinically but also histologically, to determine the benign or malignant nature of all of these lesions. As primary cutaneous melanoma, atypical proliferative nodules in CMN, bland CMN or CMN with foci of increased cellularity and Spitz naevi show clear differences in the genetic aberration patterns, comparative genomic hybridization (CGH) could be a diagnostic help in ambiguous cases such as this. CGH performed on this patient showed multiple DNA copy number changes in the most atypical nodule, but such alterations could not be found in the remainder of the lesions. CGH showed differences between the nodular lesions that occurred in the CMN and helped us in supporting the diagnosis of this unique case of benign proliferative nodules and a possible congenital melanoma arising in a large CMN, associated with multiple widespread eruptive Spitz naevi.


Assuntos
Neoplasias de Cabeça e Pescoço/diagnóstico , Melanoma/diagnóstico , Nevo de Células Epitelioides e Fusiformes/diagnóstico , Nevo Pigmentado/diagnóstico , Couro Cabeludo , Neoplasias Cutâneas/diagnóstico , Hibridização Genômica Comparativa , Diagnóstico Diferencial , Feminino , Neoplasias de Cabeça e Pescoço/congênito , Humanos , Recém-Nascido , Melanoma/congênito , Nevo de Células Epitelioides e Fusiformes/congênito , Nevo Pigmentado/congênito , Neoplasias Cutâneas/congênito
9.
Br J Cancer ; 101(5): 813-5, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19654573

RESUMO

BACKGROUND: Recently, oncogenic G protein alpha subunit q (GNAQ) mutations have been described in about 50% of uveal melanomas and in the blue nevi of the skin. METHODS: GNAQ exon 5 was amplified from 75 ciliary body and choroidal melanoma DNAs and sequenced directly. GNAQ mutation status was correlated with disease-free survival (DFS), as well as other clinical and histopathological factors, and with chromosomal variations detected by FISH and CGH. RESULTS: Of the 75 tumour DNA samples analysed, 40 (53.3%) harboured oncogenic mutations in GNAQ codon 209. Univariate and multivariate analysis showed that GNAQ mutation status was not significantly correlated with DFS. CONCLUSION: The GNAQ mutation status is not suitable to predict DFS. However, the high frequency of GNAQ mutations may render it a promising target for therapeutic intervention.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Melanoma/diagnóstico , Melanoma/genética , Mutação/genética , Neoplasias Uveais/diagnóstico , Neoplasias Uveais/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Hibridização Genômica Comparativa , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Hibridização in Situ Fluorescente , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Análise Multivariada , Neoplasias Uveais/patologia
11.
Pathologe ; 28(6): 464-73, 2007 Nov.
Artigo em Alemão | MEDLINE | ID: mdl-17882420

RESUMO

In the case of many tumors, the development of cancer is associated with a loss of control over genomic integrity, resulting in alterations, determined by selection, of the genome of the cancer cells. Comparative genomic hybridization (CGH) is a method that can be used to assess the entire genome of tumor cells for the presence of changes in DNA copy number. CGH analysis has revealed that melanomas differ from melanocytic nevi in the presence of frequent chromosomal aberrations. CGH analysis of benign melanocytic tumors typically shows no clonally expanded chromosomal aberrations, while in the vast majority of melanomas gains and losses of particular chromosomes are found. As an exception, Spitz nevi show an increased copy number of chromosome 11p in about 20% of cases, something not found in melanoma. These marked differences between the aberration patterns of melanomas and melanocytic nevi can be exploited during differential diagnosis of melanocytic tumors in which histopathologic assessment yields equivocal results. In addition, it has also been shown with the aid of CGH and mutation analysis that melanomas are not a homogenous disease, but rather a group of genetically different tumors. A study checking for correlations between the chromosomal alterations in melanocytic tumors not classified at diagnosis and the course of illness in patients is currently under way.


Assuntos
Aberrações Cromossômicas , DNA de Neoplasias/genética , Melanoma/genética , Neoplasias Cutâneas/genética , DNA/genética , Humanos , Melanócitos , Hibridização de Ácido Nucleico
13.
Cancer Res ; 61(20): 7642-6, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11606406

RESUMO

We investigated the role of alterations of HDM2, the human homologue of murine mdm2, in the tumorigenesis and progression of cutaneous melanoma. A well-characterized cohort of 172 cases representing different points in the spectrum of melanocyte transformation (16 dysplastic nevi, 11 melanomas in situ, 107 invasive primaries, and 38 metastatic lesions), as well as 11 human melanoma cell lines were examined by immunohistochemistry and Western blotting for HDM2 protein expression, and by either Southern blotting (SB) or fluorescence in situ hybridization for HDM2 gene amplification. HDM2 overexpression, defined as >20% tumor cells showing nuclear immunoreactivity, was observed in 1 of 16 (6%) dysplastic nevi, 3 of 11 (27%) melanomas in situ, and 81 of 145 (56%) invasive primary and metastatic melanomas. Comparable frequencies of HDM2 overexpression were observed among invasive primary cases with differing tumor thicknesses as well as among the metastatic cases: 21 of 40 (53%) at < or =1.5 mm; 31 of 50 (62%) at 1.6-3.9 mm; 10 of 17 (58%) at >4 mm; and 19 of 38 (50%) metastases. HDM2 amplification was observed in 1 of 88 (1%) primary cases using fluorescence in situ hybridization, and in 0 of 12 (0%) metastatic cases that overexpressed HDM2 using SB. Melanoma cell lines expressed HDM2 protein, but there was no evidence of amplification by SB. Our data suggest that HDM2 protein overexpression is common in invasive and metastatic melanoma. Observing HDM2 overexpression in noninvasive melanoma suggests that expression of this oncogene may play an early role in melanocyte transformation. HDM2 amplification occurs infrequently, and other mechanisms that up-regulate HDM2 expression are under investigation.


Assuntos
Melanoma/metabolismo , Proteínas Nucleares , Proteínas Proto-Oncogênicas/genética , Neoplasias Cutâneas/genética , Transformação Celular Neoplásica/genética , Estudos de Coortes , Síndrome do Nevo Displásico/genética , Síndrome do Nevo Displásico/metabolismo , Amplificação de Genes , Humanos , Imuno-Histoquímica , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/genética , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas c-mdm2 , Neoplasias Cutâneas/metabolismo , Células Tumorais Cultivadas
14.
Mol Cell Biol ; 21(6): 2144-53, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238948

RESUMO

The frequent loss of both INK4a and ARF in melanoma raises the question of which INK4a-ARF gene product functions to suppress melanoma genesis in vivo. Moreover, the high incidence of INK4a-ARF inactivation in transformed melanocytes, along with the lack of p53 mutation, implies a cell type-specific role for INK4a-ARF that may not be complemented by other lesions of the RB and p53 pathways. A mouse model of cutaneous melanoma has been generated previously through the combined effects of INK4a(Delta2/3) deficiency (null for INK4a and ARF) and melanocyte-specific expression of activated RAS (tyrosinase-driven H-RAS(V12G), Tyr-RAS). In this study, we made use of this Tyr-RAS allele to determine whether activated RAS can cooperate with p53 loss in melanoma genesis, whether such melanomas are biologically comparable to those arising in INK4a(Delta2/3-/-) mice, and whether tumor-associated mutations emerge in the p16(INK4a)-RB pathway in such melanomas. Here, we report that p53 inactivation can cooperate with activated RAS to promote the development of cutaneous melanomas that are clinically indistinguishable from those arisen on the INK4a(Delta2/3) null background. Genomewide analysis of RAS-induced p53 mutant melanomas by comparative genomic hybridization and candidate gene surveys revealed alterations of key components governing RB-regulated G(1)/S transition, including c-Myc, cyclin D1, cdc25a, and p21(CIP1). Consistent with the profile of c-Myc dysregulation, the reintroduction of p16(INK4a) profoundly reduced the growth of Tyr-RAS INK4a(Delta2/3-/-) tumor cells but had no effect on tumor cells derived from Tyr-RAS p53(-/-) melanomas. Together, these data validate a role for p53 inactivation in melanomagenesis and suggest that both the RB and p53 pathways function to suppress melanocyte transformation in vivo in the mouse.


Assuntos
Genes ras , Melanoma/genética , Proteína do Retinoblastoma/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Ciclina D1/genética , Ciclina D1/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ciclinas/metabolismo , Fase G1/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Hibridização In Situ/métodos , Melanoma/metabolismo , Camundongos , Camundongos Mutantes , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína do Retinoblastoma/metabolismo , Fase S/genética , Proteína Supressora de Tumor p14ARF , Proteína Supressora de Tumor p53/genética , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo
15.
Australas Radiol ; 44(3): 321-4, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10974728

RESUMO

Primary tumours of the heart and pericardium are extremely rare. Cardiac lipomas account for only 10% of all primary cardiac tumours. A case of surgically proven pericardial lipoma demonstrated by ultrasound, CT and MRI is presented here.


Assuntos
Ecocardiografia Transesofagiana , Neoplasias Cardíacas/diagnóstico , Lipoma/diagnóstico , Imageamento por Ressonância Magnética , Tomografia Computadorizada por Raios X , Idoso , Diagnóstico Diferencial , Neoplasias Cardíacas/cirurgia , Humanos , Lipoma/cirurgia , Masculino , Pericárdio/diagnóstico por imagem , Pericárdio/patologia
16.
Am J Pathol ; 157(3): 967-72, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10980135

RESUMO

Spitz nevus is a benign melanocytic neoplasm that can be difficult or impossible to histologically distinguish from melanoma. We have recently described copy number increases of chromosome 11p in a subset of Spitz nevi. To study the molecular and histological features of this group, we studied 102 Spitz nevi for 11p copy number increases using fluorescence in situ hybridization (FISH) on tissue arrays. Copy number increases of at least threefold were found in 12 cases (11.8%) and involved the HRAS gene on chromosome 11p. Sequence analysis of HRAS showed frequent oncogenic mutations in cases with copy number increase (8/12 or 67%), contrasting with rare HRAS mutations in cases with normal HRAS copy numbers (1/21 or 5%, P: < 0.0001). Tumors with 11p copy number increases were larger, predominantly intradermal, had marked desmoplasia, characteristic cytological features, and had an infiltrating growth pattern. Proliferation rates in the majority of these cases were low to absent. HRAS activation by either mutation or copy number increase alone could explain several of the histological features that overlap with those of melanoma. We speculate that HRAS activation in the absence of co-operating additional genetic alterations drives the partially transformed melanocytes of these Spitz nevi into senescence or a stable growth arrest. Although there is no data suggesting that Spitz nevi with HRAS activation are at risk for progression to melanoma, future studies are warranted to assess their biological behavior more accurately.


Assuntos
Dosagem de Genes , Genes ras , Mutação , Nevo de Células Epitelioides e Fusiformes/genética , Neoplasias Cutâneas/genética , Cromossomos Humanos Par 11 , Primers do DNA/análise , DNA de Neoplasias/análise , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Nevo de Células Epitelioides e Fusiformes/patologia , Reação em Cadeia da Polimerase , Neoplasias Cutâneas/patologia
17.
Australas Radiol ; 44(2): 208-11, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10849988

RESUMO

A case of atrial myxoma presenting with syncope evaluated by echocardiography and MRI is described. Cine gradient-echo MRI demonstrated atrial myxoma as a very low signal intensity mass indicating the presence of haemosiderin.


Assuntos
Ecocardiografia , Neoplasias Cardíacas/diagnóstico , Imagem Cinética por Ressonância Magnética , Mixoma/diagnóstico , Feminino , Átrios do Coração , Neoplasias Cardíacas/diagnóstico por imagem , Humanos , Pessoa de Meia-Idade , Mixoma/diagnóstico por imagem
18.
Cancer Res ; 60(7): 1968-73, 2000 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10766187

RESUMO

Acral melanoma (AM) is commonly distinguished from superficial spreading melanoma (SSM), the most common type of melanoma, by its clinical presentation as well as its ethnic distribution. However, justification for such a distinction is controversial because of histological overlap and lack of prognostic significance. We analyzed chromosomal aberrations of 15 AMs and 15 SSMs that were comparable for tumor thickness and patient age, using comparative genomic hybridization. All AMs had at least one (mean, 2.0) gene amplification, significantly more than the SSMs, in which only 2 of 15 (13%) had one amplification each (P < 0.0001). At least 15 different genomic regions were amplified in AM. These involved small portions of chromosomal arms, sometimes including known oncogenes implicated in melanoma. The most frequently amplified regions in AMs occurred at 11q13 (47%), 22q11-13 (40%), and 5p15 (20%). Comparison of the amplification levels of invasive and noninvasive portions of the tumors using fluorescence in situ hybridization suggested that amplifications occurred before the formation of the invasive portion. The finding of amplifications of 11q13 in three of five additional cases of AM in situ further supports the notion that amplifications arise early in the progression of AM. Very significantly, we found isolated melanocytes with amplifications in the epidermis up to 3 mm beyond the histologically recognizable extent of the melanomas in 5 of 15 invasive AMs. In conclusion, our data show that AM is a distinct type of melanoma characterized by focused gene amplifications occurring early in tumorigenesis, and that malignant cells are present beyond the histologically detectable boundary, thereby revealing one mechanism of local recurrence.


Assuntos
Mapeamento Cromossômico , Amplificação de Genes , Melanoma/genética , Melanoma/patologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Pele/patologia , Cromossomos Humanos Par 11 , Cromossomos Humanos Par 22 , Cromossomos Humanos Par 5 , Progressão da Doença , Humanos , Hibridização in Situ Fluorescente , Melanoma/classificação , Invasividade Neoplásica , Sensibilidade e Especificidade , Neoplasias Cutâneas/classificação
19.
J Invest Dermatol ; 113(6): 1065-9, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10594753

RESUMO

Spitz nevus is a benign neoplasm of melanocytes that can be difficult or impossible to distinguish from melanoma by clinical and histopathologic examination. We studied genomic DNA from 17 Spitz nevi by comparative genomic hybridization (CGH). Thirteen lesions showed no chromosomal aberrations, three cases had a gain involving the entire p-arm of chromosome 11, and one case showed a gain of chromosome 7q21-qter. Fluorescence in situ hybridization (FISH) on lesional tissue with a probe for the p-arm of chromosome 11 showed 6-10 p-arm signals per nucleus in those cases with a CGH-detected gain of chromosome 11p. One case with a normal CGH profile also showed increased copy number of 11p by FISH. Thus, the majority of Spitz nevi have a normal chromosomal complement at the level of CGH resolution; however some may contain gains, with 11p apparently being the most frequently involved location. These findings differ significantly from the previously reported changes in primary cutaneous melanoma, which show frequent deletions of chromosomes 9p (82%), 10q (63%), 6q (28%), and 8p (22%), as well as gains of chromosomes 7 (50%), 8 (34%), 6p (28%), 1q (25%) by CGH analysis. These clear differences in the location and frequencies of chromosomal aberrations in Spitz nevi and primary cutaneous melanomas could represent a basis for developing adjunctive techniques for refining accuracy in the difficult differential diagnosis of spitzoid melanocytic neoplasms.


Assuntos
Aberrações Cromossômicas , Melanoma/genética , Nevo de Células Epitelioides e Fusiformes/genética , Neoplasias Cutâneas/genética , Adolescente , Adulto , Criança , Análise Citogenética , Feminino , Humanos , Hibridização in Situ Fluorescente , Masculino , Pessoa de Meia-Idade
20.
J Am Acad Dermatol ; 41(4): 606-13, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10495385

RESUMO

BACKGROUND: Signet-ring cells are formed by intracytoplasmic accumulations of various substances that push the nucleus to the cellular border. Signet-ring cells in epithelial neoplasms are often regarded as evidence of adenocarcinoma. OBJECTIVE: The study surveys the rare settings in which signet-ring cells are encountered in dermatopathologic specimens and investigates mechanisms of their formation. METHODS: A total of 23 cutaneous tumors with a significant population of signet-ring cells were studied by immunohistochemistry and electron microscopy. RESULTS: Signet-ring cells were found in a variety of cutaneous neoplasms, including primary cutaneous squamous and basal cell carcinoma and melanoma, as well as in metastatic adenocarcinoma. In all but the metastatic adenocarcinomas the vacuoles were periodic acid Schiff (PAS), PAS-digest, and colloidal iron negative. There was no staining of the vacuoles with antibodies against keratins and vimentin. Electron microscopy showed only empty spaces in all cases. CONCLUSION: The signet-ring like appearance of the cells in most of these conditions is probably the result of coalescence of intracytoplasmic vacuoles and not accumulation of secretory products. Signet-ring formation is not specific for cellular lineage but can occur in a variety of cutaneous neoplasms, analogous to other cellular alterations as rhabdoid, granular, clear, spindle, and balloon cells and oncocytes.


Assuntos
Neoplasias Cutâneas/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Núcleo Celular/ultraestrutura , Criança , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Cutâneas/secundário , Neoplasias Cutâneas/ultraestrutura , Vacúolos/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA