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










Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 13: 508-517, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35284037

RESUMO

Gastrointestinal stromal tumor (GIST) is a mesenchymal neoplasm with variable behavior. An increased understanding of the tumor pathogenesis may improve clinical decision-making. Our aim was to obtain more data about the overall chromosome aberrations and intratumor cytogenetic heterogeneity in GIST. We analyzed 306 GIST samples from 291 patients using G-banding, direct sequencing, and statistics. Clonal chromosome aberrations were found in 81% of samples, with 34% of 226 primary tumors demonstrating extensive cytogenetic heterogeneity. 135 tumors had simple (≤5 changes) and 91 had complex (>5 changes) karyotypes. The karyotypically complex tumors more often were non-gastric (P < 0.001), larger (P < 0.001), more mitotically active (P = 0.009) and had a higher risk of rupture (P < 0.001) and recurrence (P < 0.001). Significant differences between gastric and non-gastric tumors were found also in the frequency of main chromosome losses: of 14q (79% vs. 63%), 22q (38% vs. 67%), 1p (23% vs. 88%), and 15q (18% vs. 77%). Gastric PDGFRA-mutated tumors, compared with gastric KIT-mutated, had a lower incidence of 22q losses (18% vs. 43%) but a higher rate of 1p losses (42% vs. 22%). The present, largest by far karyotypic study of GISTs provides further evidence for the existence of variable pathogenetic pathways operating in these tumors' development.


Assuntos
Neoplasias Gastrointestinais , Tumores do Estroma Gastrointestinal , Neoplasias Gástricas , Aberrações Cromossômicas , Bandeamento Cromossômico , Citogenética , Neoplasias Gastrointestinais/genética , Tumores do Estroma Gastrointestinal/patologia , Humanos , Cariotipagem , Mutação , Proteínas Proto-Oncogênicas c-kit/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética
2.
Ann Surg Oncol ; 28(11): 6837-6845, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33651216

RESUMO

BACKGROUND: Adjuvant imatinib for 3 years is recommended to patients with high-risk gastrointestinal stromal tumor (GIST). Risk stratification is inaccurate, and risk assessments are further complicated by the increased use of neoadjuvant treatment. Anatomical criteria for prognostication have not been investigated. METHODS: Clinical, molecular, and anatomical variables were retrospectively studied in a population-based cohort of 295 patients with gastric GIST resected between 2000 and 2018. Gastric subsite was divided into the upper, middle, and lower thirds. Growth pattern was classified as luminal, exophytic, or transmural based on imaging and surgical reports. RESULTS: Of 113 tumors in the upper third of the stomach, 103 (91.2%) were KIT mutated, 7 (6.2%) were PDGFRA mutated, and 104 (92.0%) harbored genotypes sensitive to imatinib. Transmural tumors were strongly associated with a high mitotic index. Five-year recurrence-free survival (RFS) was 71% for patients with transmural tumors versus 96% with luminal or exophytic tumors (hazard ratio [HR] 8.45, 95% confidence interval [CI] 3.69-19.36; p < 0.001), and, in high-risk patients, 5-year RFS was 46% for patients with transmural tumors versus 83% with luminal or exophytic tumors (HR 4.47, 95% CI 1.71-11.66; p = 0.001). Among 134 patients with tumors > 5 cm, there were 29 recurrences. Only five patients with exophytic or luminal tumors had recurrent disease, of whom four had tumor rupture. Five-year RFS for patients with exophytic/luminal tumors >5 cm without rupture was 98%. CONCLUSIONS: In the upper third, over 90% of tumors were sensitive to imatinib. Patients with exophytic or luminal tumors without rupture, irrespective of size, had an excellent prognosis and may not benefit from adjuvant therapy.


Assuntos
Antineoplásicos , Neoplasias Gastrointestinais , Tumores do Estroma Gastrointestinal , Antineoplásicos/uso terapêutico , Neoplasias Gastrointestinais/tratamento farmacológico , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/cirurgia , Humanos , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/cirurgia , Proteínas Proto-Oncogênicas c-kit , Estudos Retrospectivos , Fatores de Risco , Estômago
4.
PLoS One ; 13(5): e0197456, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29771965

RESUMO

Nucleic acid material of adequate quality is crucial for successful high-throughput sequencing (HTS) analysis. DNA and RNA isolated from archival FFPE material are frequently degraded and not readily amplifiable due to chemical damage introduced during fixation. To identify optimal nucleic acid extraction kits, DNA and RNA quantity, quality and performance in HTS applications were evaluated. DNA and RNA were isolated from five sarcoma archival FFPE blocks, using eight extraction protocols from seven kits from three different commercial vendors. For DNA extraction, the truXTRAC FFPE DNA kit from Covaris gave higher yields and better amplifiable DNA, but all protocols gave comparable HTS library yields using Agilent SureSelect XT and performed well in downstream variant calling. For RNA extraction, all protocols gave comparable yields and amplifiable RNA. However, for fusion gene detection using the Archer FusionPlex Sarcoma Assay, the truXTRAC FFPE RNA kit from Covaris and Agencourt FormaPure kit from Beckman Coulter showed the highest percentage of unique read-pairs, providing higher complexity of HTS data and more frequent detection of recurrent fusion genes. truXTRAC simultaneous DNA and RNA extraction gave similar outputs as individual protocols. These findings show that although successful HTS libraries could be generated in most cases, the different protocols gave variable quantity and quality for FFPE nucleic acid extraction. Selecting the optimal procedure is highly valuable and may generate results in borderline quality specimens.


Assuntos
DNA/isolamento & purificação , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Inclusão em Parafina , RNA/isolamento & purificação , Animais , Humanos
5.
Int J Oncol ; 45(5): 1829-36, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25176350

RESUMO

Spindle cell tumors are clinically heterogeneous but morphologically similar neoplasms that can occur anywhere, mostly in adult patients. They are treated primarily with surgery to which is often added adjuvant or neoadjuvant radiation. Sub-classification of spindle cell sarcomas requires integration of histology, clinicopathological parameters, immunohistochemistry, cytogenetics (including fluorescence in situ hybridization) and/or molecular genetics. Some of the tumor subtypes are characterized by the presence of distinct chromosomal translocations and fusion genes. When no signs of differentiation are seen, the diagnosis by exclusion becomes undifferentiated spindle cell sarcoma. Cytogenetic, RNA sequencing and RT-PCR analyses were performed on a case of spindle cell sarcoma. The karyotype of the primary tumor was 46,X,del(X)(p?11p?22), der(12)(12pter→12q?22::12q?15→q?22::16p11→16pter),-16,+r(12). MDM2 was found amplified in both the primary tumor and a meta-stasis. RNA-Seq of the primary tumor identified four fusion genes, PTGES3-PTPRB, HMGA2-DYRK2, TMBIM4-MSRB3 and USP15-CNTN1, in which all the partner genes map to the q arm of chromosome 12. In material from the metastasis, RT-PCR detected the PTGES3-PTPRB, HMGA2-DYRK2 and TMBIM4-MSRB3 whereas no USP15-CNTN1 fusion transcript was found. Because MDM2 amplification and the fusion transcripts PTGES3-PTPRB, HMGA2-DYRK2 and TMBIM4-MSRB3 were found both in the primary tumor and in the metastasis, they are components of the same clone and may be involved both in initiation and progression of the tumor. Which of them is pathogenetically primary remains unknown.


Assuntos
Proteínas de Fusão Oncogênica/genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Sarcoma/genética , Transcriptoma/genética , Adulto , Carcinogênese , Cromossomos Humanos Par 12/genética , Proteína HMGA2/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Cariotipagem , Metástase Neoplásica , Proteínas de Fusão Oncogênica/isolamento & purificação , Translocação Genética/genética
6.
Acta Oncol ; 52(4): 776-82, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23480638

RESUMO

BACKGROUND: Acquired resistance to imatinib is frequently caused by secondary KIT mutations. We have investigated the effects of imatinib in mice with human gastrointestinal stromal tumour (GIST) xenograft which harbours a primary exon 11 deletion mutation and a secondary imatinib resistance mutation D816H in exon 17. Such mutations are commonly present in imatinib-resistant GIST in humans. MATERIAL AND METHODS: The mice were randomly allocated to receive imatinib either continuously or intermittently. Dynamic (18)F-FDG PET was performed and blood volume fraction (vB), rate transfer constants (k1, k2, k3) and metabolic rate of (18)F-FDG (MRFDG) were computed using a three-compartment model. Tumours were evaluated for the mitotic rate and the expression of HIF-1α , caspase-3 and glucose transporters (GLUTs). RESULTS: Both intermittent and continuous imatinib delayed tumour growth significantly compared to controls, significantly in favour of the latter. k1 (representing perfusion, vascular permeability and binding of (18)F-FDG to the GLUTs) was significantly higher in the intermittent group compared to the continuous group, as was tumour GLUT-3 expression. k3 (representing internalisation of (18)F-FDG to the cells) and MR(FDG) were significantly lower. CONCLUSION: Imatinib delays GIST xenograft growth despite the presence of the D816H resistance mutation. The schedule of imatinib administration may influence tumour glucose uptake rate and metabolic rate.


Assuntos
Antineoplásicos/administração & dosagem , Benzamidas/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Gastrointestinais/tratamento farmacológico , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Mutação de Sentido Incorreto , Piperazinas/administração & dosagem , Proteínas Proto-Oncogênicas c-kit/genética , Pirimidinas/administração & dosagem , Substituição de Aminoácidos/genética , Animais , Ácido Aspártico/genética , Esquema de Medicação , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/patologia , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/patologia , Histidina/genética , Humanos , Mesilato de Imatinib , Camundongos , Camundongos Nus , Mutação de Sentido Incorreto/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Anticancer Res ; 29(11): 4331-6, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20032375

RESUMO

BACKGROUND: The majority of gastrointestinal stromal tumours (GISTs) contain oncogenic KIT (v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog) or platelet-derived growth factor-alpha (PDGFRA) receptor tyrosine kinase (TK) mutations and are initially, but only temporarily sensitive to TK inhibitors. The aim of this study was to establish and characterize a human GIST xenograft that could be used for evaluating various molecularly targeted therapies. MATERIALS AND METHODS: GIST tissue from four patients was implanted under the skin of athymic nude mice. In one case a tumour line was established. RESULTS: The xenograft showed characteristic GIST morphology and exhibited the same mutation profile as that of the patient. CONCLUSION: A human GIST xenograft with mutation in KIT exons 11 and 17 has been established and maintained in nude mice for 3 years (13 passages). This model will enable further studies on mechanisms of resistance, combination therapies and allow testing of novel targeted therapies.


Assuntos
Tumores do Estroma Gastrointestinal/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Adulto , Animais , Benzamidas , Resistencia a Medicamentos Antineoplásicos , Feminino , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/terapia , Humanos , Mesilato de Imatinib , Imuno-Histoquímica , Cariotipagem , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Mutação , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-kit/genética , Pirimidinas/farmacologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética
8.
Leuk Res ; 33(10): 1379-85, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19394083

RESUMO

The transmembrane transporter P-glycoprotein (P-gp) encoded by ABCB1, is one major cause for multidrug resistance (MDR). We compared the genomic profile and gene expression pattern of the P-gp positive K562VCR cells with parental P-gp negative K562wt cells. In K562VCR array CGH revealed amplification of ABCB1, ABCB4, ABCB5 and SEMA3D, whereas expression microarrays demonstrated upregulation of stem cell genes (e.g. KIT and HOXB4), anti-apoptotic genes (e.g. IGF1R and CCNG1), and downregulation of pro-apoptotic genes (e.g. CASP4, 6 and 7). Thus, K562VCR cells disclose stem cell characteristics including a range of drug resistance mechanisms possibly attained as a stem cell program switched on en bloc.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Células K562/patologia , Leucemia Eritroblástica Aguda/genética , Mapeamento Cromossômico , Cromossomos Humanos , Citometria de Fluxo , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Células-Tronco/genética , Células-Tronco/citologia , Células-Tronco/fisiologia , Fatores de Transcrição/genética , Regulação para Cima
9.
BMC Genomics ; 8: 73, 2007 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-17359542

RESUMO

BACKGROUND: Soft tissue sarcoma (STS) diagnosis is challenging because of a multitude of histopathological subtypes, different genetic characteristics, and frequent intratumoral pleomorphism. One-third of STS metastasize and current risk-stratification is suboptimal, therefore, novel diagnostic and prognostic markers would be clinically valuable. We assessed the diagnostic and prognostic value of array-based gene expression profiles using 27 k cDNA microarrays in 177, mainly high-grade, STS of 13 histopathological subtypes. RESULTS: Unsupervised analysis resulted in two major clusters--one mainly containing STS characterized by type-specific genetic alterations and the other with a predominance of genetically complex and pleomorphic STS. Synovial sarcomas, myxoid/round-cell liposarcomas, and gastrointestinal stromal tumors clustered tightly within the former cluster and discriminatory signatures for these were characterized by developmental genes from the EGFR, FGFR, Wnt, Notch, Hedgehog, RAR and KIT signaling pathways. The more pleomorphic STS subtypes, e.g. leiomyosarcoma, malignant fibrous histiocytoma/undifferentiated pleomorphic sarcoma and dedifferentiated/pleomorphic liposarcoma, were part of the latter cluster and were characterized by relatively heterogeneous profiles, although subclusters herein were identified. A prognostic signature partly characterized by hypoxia-related genes was identified among 89 genetically complex pleomorphic primary STS and could, in a multivariate analysis including established prognostic markers, independently predict the risk of metastasis with a hazard ratio of 2.2 (P = 0.04). CONCLUSION: Diagnostic gene expression profiles linking signaling pathways to the different STS subtypes were demonstrated and a hypoxia-induced metastatic profile was identified in the pleomorphic, high-grade STS. These findings verify diagnostic utility and application of expression data for improved selection of high-risk STS patients.


Assuntos
Hipóxia Celular/genética , Perfilação da Expressão Gênica , Técnicas de Diagnóstico Molecular , Metástase Neoplásica/diagnóstico , Sarcoma/diagnóstico , Sarcoma/genética , Análise Serial de Tecidos/métodos , Análise por Conglomerados , Intervalo Livre de Doença , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/patologia , Histiocitoma Fibroso Maligno/genética , Histiocitoma Fibroso Maligno/patologia , Humanos , Leiomiossarcoma/genética , Leiomiossarcoma/patologia , Lipossarcoma Mixoide/genética , Lipossarcoma Mixoide/patologia , Neoplasias de Bainha Neural/genética , Neoplasias de Bainha Neural/patologia , Prognóstico , Sarcoma/patologia , Sarcoma/terapia , Sarcoma Sinovial/genética , Sarcoma Sinovial/patologia
10.
J Pathol ; 208(3): 431-8, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16353171

RESUMO

The expression of fibroblast growth factors (FGF1 and FGF2) and their receptors has been reported in a variety of human neoplasms, including haematological neoplasia. Fibroblast growth factors can promote tumour growth directly, or indirectly through promoting the growth of vessels. In the present study, we evaluated the expression of FGF1 and FGF2 as well as FGF receptors 1-4 (FGFR1-FGFR4) in 39 cases of Hodgkin's lymphoma, including all subtypes, as well as Hodgkin's lymphoma cell lines. FGF1 and FGF2 and their receptors FGFR2-FGFR4, but not FGFR1, were found to be expressed by the malignant cells in the majority of cases. Interestingly, only FGFR3, but none of the FGFs or the other FGFRs, was expressed by the Hodgkin's lymphoma cell lines. This indicates that only FGFR3 is constitutively expressed by Hodgkin's lymphoma cells, whereas FGFs and the other FGFRs are induced in vivo. Culture of the Hodgkin's cell lines under conditions of hypoxic stress could induce vascular endothelial growth factor (VEGF) but not FGF expression. FGFs, in contrast to VEGF, might be expressed in response to paracrine stimuli. In situ hybridization did not reveal FGFR3 gene amplification or translocation as the cause of constitutive FGFR3 expression, as has been shown in a subset of multiple myeloma. FGFR3 might be expressed as part of the Hodgkin's cell phenotype. The demonstration of widespread expression of FGFs and some of their receptors in Hodgkin's lymphoma suggests that FGFs are important for sustaining growth of the lymphoma and suggests that anti-FGF antibodies could be used as an adjuvant treatment.


Assuntos
Fatores de Crescimento de Fibroblastos/análise , Doença de Hodgkin/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/análise , Hipóxia Celular , Linhagem Celular Tumoral , Fator 1 de Crescimento de Fibroblastos/análise , Fator 2 de Crescimento de Fibroblastos/análise , Expressão Gênica , Humanos , Immunoblotting , Imuno-Histoquímica/métodos , Hibridização in Situ Fluorescente , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/análise , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/análise , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/análise , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Mol Cancer ; 4: 39, 2005 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-16274472

RESUMO

BACKGROUND: Amplification of the q21-q23 region on chromosome 1 is frequently found in sarcomas and a variety of other solid tumours. Previous analyses of sarcomas have indicated the presence of at least two separate amplicons within this region, one located in 1q21 and one located near the apolipoprotein A-II (APOA2) gene in 1q23. In this study we have mapped and characterized the amplicon in 1q23 in more detail. RESULTS: We have used fluorescence in situ hybridisation (FISH) and microarray-based comparative genomic hybridisation (array CGH) to map and define the borders of the amplicon in 10 sarcomas. A subregion of approximately 800 kb was identified as the core of the amplicon. The amplification patterns of nine possible candidate target genes located to this subregion were determined by Southern blot analysis. The genes activating transcription factor 6 (ATF6) and dual specificity phosphatase 12 (DUSP12) showed the highest level of amplification, and they were also shown to be over-expressed by quantitative real-time reverse transcription PCR (RT-PCR). In general, the level of expression reflected the level of amplification in the different tumours. DUSP12 was expressed significantly higher than ATF6 in a subset of the tumours. In addition, two genes known to be transcriptionally activated by ATF6, glucose-regulated protein 78 kDa and -94 kDa (GRP78 and GRP94), were shown to be over-expressed in the tumours that showed over-expression of ATF6. CONCLUSION: ATF6 and DUSP12 seem to be the most likely candidate target genes for the 1q23 amplification in sarcomas. Both genes have possible roles in promoting cell growth, which makes them interesting candidate targets.


Assuntos
Apolipoproteína A-II/genética , Cromossomos Humanos Par 1/genética , Genoma Humano/genética , Hibridização in Situ Fluorescente , Análise de Sequência com Séries de Oligonucleotídeos , Sarcoma/genética , Chaperona BiP do Retículo Endoplasmático , Amplificação de Genes , Dosagem de Genes , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Hibridização de Ácido Nucleico , Mapeamento Físico do Cromossomo , RNA Mensageiro/genética
12.
J Neuropathol Exp Neurol ; 64(1): 74-81, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15715087

RESUMO

The molecular biology underlying the development of highly malignant peripheral nerve sheath tumors (MPNSTs) remains mostly unknown. In the present study, the expression pattern of 10 selected cell cycle components is investigated in a series of 15 MPNSTs from patients with (n = 9) or without (n = 5) neurofibromatosis type 1 (NF1). Thirteen tumors did not express the cyclin-dependent kinase inhibitor, p16(INK4A), an observation that was related to homozygote gene deletions in three tumors, heterozygote deletions in five, and gross gene rearrangements in five. The absence of protein expression in the tumors with one seemingly intact allele was not caused by promoter hypermethylation of p16(INK4A) or p14(ARF). All tumor samples expressed normal sized RB1, cyclin D3, CDK2, CDK4, p21(CIP1), and p27(KlP1) proteins, and only a single tumor showed an aberrant protein band for one of these proteins, p21(CIP1). Cyclin D1 was absent in four tumors; all except one tumor showed expression of TP53 protein, and three of nine MPNSTs had expression of normal-sized MDM2. In conclusion, this study shows that the vast majority of MPNSTs had gross rearrangements of the p16(INK4A) gene, explaining the absence of the encoded protein in the same tumors. The level of expression was equally distributed between the familial (NF1) and sporadic cases, although it should be noted that the 2 cases with p16(INK4A) expression were sporadic. The data imply that the complete absence of p16(INK4A) is sufficient for activation of the cell cycle in most MPNSTs; thus, it is not necessary for tumor proliferation to further stimulate the cycle through alteration of other central components.


Assuntos
Proteínas de Ciclo Celular/biossíntese , Ciclo Celular , Regulação Neoplásica da Expressão Gênica , Neoplasias de Bainha Neural/genética , Neoplasias de Bainha Neural/metabolismo , Neurofibromatose 1/genética , Neurofibromatose 1/metabolismo , Adulto , Idoso , Western Blotting , Proteínas de Ciclo Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Inibidor p16 de Quinase Dependente de Ciclina/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias de Bainha Neural/patologia , Neurofibromatose 1/patologia , Proteína do Retinoblastoma/biossíntese , Proteína do Retinoblastoma/genética , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...