Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Rev. esp. med. nucl. imagen mol. (Ed. impr.) ; 36(2): 103-109, mar.-abr. 2017. ilus
Artigo em Inglês | IBECS | ID: ibc-160782

RESUMO

The aim of this review was to evaluate the potential advantages of SPECT/CT hybrid imaging in the management of neuroendocrine tumors, adrenal tumors, pheochromocytomas and paragangliomas. From the collected data, the superiority of fused images was observed as providing both functional/molecular and morphological imaging compared to planar imaging. This provided an improvement in diagnostic imaging, with significant advantages as regards: (1) precise locating of the lesions; (2) an improvement in characterization of the findings, resulting higher specificity, improved sensitivity, and overall greater accuracy, (3) additional anatomical information derived from the CT component; (4) CT-based attenuation correction and potential for volumetric dosimetry calculations, and (5) improvement on the impact on patient management (e.g. in better defining treatment plans, in shortening surgical operating times). It can be concluded that SPECT/CT hybrid imaging provides the nuclear medicine physician with a powerful imaging modality in comparison to planar imaging, providing essential information about the location of lesions, and high quality homogeneous images (AU)


El objetivo de esta revisión es evaluar las ventajas que puede ofrecer la imagen híbrida mediante SPECT/TC en el manejo de los tumores neuroendocrinos, tumores adrenales, feocromocitomas y paragangliomas. A partir de los datos recogidos de la literatura se pone de manifiesto la superioridad de las imágenes de fusión que proporcionan información morfológica y funcional/molecular con respecto a la imagen planar, permitiendo una mejora diagnóstica con importantes ventajas: 1) localización precisa de las lesiones; 2) mejora en la caracterización de los hallazgos, lo que supone una mayor especificidad y sensibilidad y por lo tanto una mayor exactitud diagnóstica; 3) información anatómica adicional obtenida con la TC; 4) corrección de la atenuación que permite realizar cálculos dosimétricos volumétricos basados en la TC; 5) mejora en el impacto del manejo del paciente como afinar el tipo de tratamiento o la reducción del tiempo quirúrgico. En resumen, la SPECT/TC es una herramienta importante para el manejo de este tipo de tumores ya que en comparación con la imagen planar da información esencial sobre la localización y la caracterización de las lesiones (AU)


Assuntos
Humanos , Masculino , Feminino , Tumores Neuroendócrinos , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Feocromocitoma/patologia , Paraganglioma , Tomografia Computadorizada de Emissão/instrumentação , Tomografia Computadorizada de Emissão/métodos , Tomografia Computadorizada de Emissão , Tecnécio/administração & dosagem , Glândulas Suprarrenais/patologia , Glândulas Suprarrenais
2.
Oncogene ; 36(28): 4037-4046, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28288138

RESUMO

Immunotherapeutic approaches, particularly programmed death 1/programmed death ligand 1 (PD-1/PD-L1) blockade, have improved the treatment of non-small-cell lung cancer (NSCLC), supporting the premise that evasion of immune destruction is of importance for NSCLC progression. However, the signals responsible for upregulation of PD-L1 in NSCLC cells and whether they are integrated with the regulation of other immune-related genes are not known. Mucin 1 (MUC1) is aberrantly overexpressed in NSCLC, activates the nuclear factor-κB (NF-κB) p65→︀ZEB1 pathway and confers a poor prognosis. The present studies demonstrate that MUC1-C activates PD-L1 expression in NSCLC cells. We show that MUC1-C increases NF-κB p65 occupancy on the CD274/PD-L1 promoter and thereby drives CD274 transcription. Moreover, we demonstrate that MUC1-C-induced activation of NF-κB→︀ZEB1 signaling represses the TLR9 (toll-like receptor 9), IFNG, MCP-1 (monocyte chemoattractant protein-1) and GM-CSF genes, and that this signature is associated with decreases in overall survival. In concert with these results, targeting MUC1-C in NSCLC tumors suppresses PD-L1 and induces these effectors of innate and adaptive immunity. These findings support a previously unrecognized central role for MUC1-C in integrating PD-L1 activation with suppression of immune effectors and poor clinical outcome.


Assuntos
Antígeno B7-H1/genética , Carcinoma Pulmonar de Células não Pequenas , Imunidade Celular/genética , Neoplasias Pulmonares , Mucina-1/fisiologia , Evasão Tumoral/genética , Células A549 , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Transdução de Sinais/genética , Transdução de Sinais/imunologia
3.
Rev. esp. med. nucl. imagen mol. (Ed. impr.) ; 35(6): 385-390, nov.-dic. 2016. ilus
Artigo em Inglês | IBECS | ID: ibc-157475

RESUMO

Nuclear medicine traditionally employs planar and single photon emission computed tomography (SPECT) imaging techniques to depict the biodistribution of radiotracers for the diagnostic investigation of a range of disorders of endocrine gland function. The usefulness of combining functional information with anatomy derived from computed tomography (CT), magnetic resonance imaging (MRI), and high resolution ultrasound (US), has long been appreciated, either using visual side-by-side correlation, or software-based co-registration. The emergence of hybrid SPECT/CT camera technology now allows the simultaneous acquisition of combined multi-modality imaging, with seamless fusion of 3D volume datasets. Thus, it is not surprising that there is growing literature describing the many advantages that contemporary SPECT/CT technology brings to radionuclide investigation of endocrine disorders, showing potential advantages for the pre-operative locating of the parathyroid adenoma using a minimally invasive surgical approach, especially in the presence of ectopic glands and in multiglandular disease. In conclusion, hybrid SPECT/CT imaging has become an essential tool to ensure the most accurate diagnostic in the management of patients with hyperparathyroidism (AU)


La medicina nuclear emplea tradicionalmente imágenes estáticas 2D, aunque en los últimos años se ha incrementado la imagen 3D tomográfica SPECT. Estas técnicas de imagen permiten la observación de la biodistribución de los radiotrazadores, destinados a la investigación diagnóstica de trastornos endocrinos que cursan con afectación glandular. La utilidad de combinar la información funcional con la anatómica tomando como referencia la tomografía computarizada (TC), la resonancia magnética (RM) y los ultrasonidos de alta resolución (US) ha sido estudiada mediante una correlación visual primero y posteriormente con el soporte de un software de co-registro de imágenes, sin obtener resultados satisfactorios. Los equipos híbridos SPECT/TC permiten la adquisición simultánea de diferentes modalidades de imagen, consiguiendo una fusión adecuada de ellas y su posterior reconstrucción volumétrica. Así pues, no es de extrañar que cada vez observemos mayor evidencia en la literatura que describe las múltiples ventajas de la tecnología SPECT/TC en la localización preoperatoria del adenoma de paratiroides con cirugía mínimamente invasiva, especialmente en presencia de patología multiglandular o de glándulas ectópicas. En resumen, la imagen híbrida se ha convertido en una herramienta esencial que garantiza la mayor exactitud diagnóstica en el manejo de pacientes con hiperparatiroidismo (AU)


Assuntos
Humanos , Masculino , Feminino , Tomografia Computadorizada de Emissão de Fóton Único/instrumentação , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Tomografia Computadorizada de Emissão de Fóton Único , Neoplasias das Paratireoides , Glândulas Paratireoides/patologia , Glândulas Paratireoides , Medicina Nuclear/métodos , Medicina Nuclear/normas , Hiperparatireoidismo/patologia , Hiperparatireoidismo
4.
Ann Oncol ; 27 Suppl 1: i16-i19, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-27141064

RESUMO

Molecular studies in ovarian serous borderline tumors (OSBTs) have been used to understand different aspects of this neoplasm. (i) Pathogenesis, Kras and Braf mutations represent very early events in the tumorigenesis of OSBT as both are detected in serous cystadenomas associated with OSBTs. In contrast, serous cystadenomas without OSBTs do not show Kras or Braf mutations. In OSBTs, Kras mutations range from 17% to 39.5%, while Braf mutations range from 23% to 48%. The former is comparable with the range of Kras mutations in ovarian low-grade serous carcinomas (OLGSCa), 19%-54.5%. In contrast, Braf mutations in OLGSCa range from 0% to 33%. Serous cystadenomas appear to progress to OSBT due to a Braf mutation, but this mutation is rarely involved in the progression to OLGSCa. OSBTs with Braf mutation are associated with cellular senescence and up-regulation of tumor suppressor genes. In contrast, OSBTs without a Braf mutation may progress to OLGSCa due to Kras mutation or some other genetic alterations. (ii) The relationship between OSBTs and the extraovarian disease, a monoclonal versus mutifocal origin? This is still matter of debate as studies using different techniques have failed to settle this controversy. (iii) Biological behavior, Braf mutations appear to have a protective role against the progression of OSBT to OLGSCa, while Kras mutations are commonly seen in cases of OSBT that recurred as LGSCa. Nevertheless, LGSCa as a recurrence of an OSBT can originate from OSBTs with or without detectable Kras mutations. Also, it appears to be an association between Kras G12v mutation and a more aggressive phenotype of OSBT that recurred as LGSCa. (iv) Actionable targets, currently there are limited data. It has been reported that cancer cell lines with Kras G12v mutation are more sensitive to selumetinib than cell lines with wild-type Kras.


Assuntos
Cistadenocarcinoma Seroso/genética , Neoplasias Ovarianas/genética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Transformação Celular Neoplásica/genética , Cistadenocarcinoma Seroso/tratamento farmacológico , Cistadenocarcinoma Seroso/patologia , Feminino , Genes ras , Humanos , Terapia de Alvo Molecular , Mutação , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Patologia Molecular , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética
5.
Oncogene ; 34(18): 2347-59, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-24931169

RESUMO

Notch activity regulates tumor biology in a context-dependent and complex manner. Notch may act as an oncogene or a tumor-suppressor gene even within the same tumor type. Recently, Notch signaling has been implicated in cellular senescence. Yet, it remains unclear as to how cellular senescence checkpoint functions may interact with Notch-mediated oncogenic and tumor-suppressor activities. Herein, we used genetically engineered human esophageal keratinocytes and esophageal squamous cell carcinoma cells to delineate the functional consequences of Notch activation and inhibition along with pharmacological intervention and RNA interference experiments. When expressed in a tetracycline-inducible manner, the ectopically expressed activated form of Notch1 (ICN1) displayed oncogene-like characteristics inducing cellular senescence corroborated by the induction of G0/G1 cell-cycle arrest, Rb dephosphorylation, flat and enlarged cell morphology and senescence-associated ß-galactosidase activity. Notch-induced senescence involves canonical CSL/RBPJ-dependent transcriptional activity and the p16(INK4A)-Rb pathway. Loss of p16(INK4A) or the presence of human papilloma virus (HPV) E6/E7 oncogene products not only prevented ICN1 from inducing senescence but permitted ICN1 to facilitate anchorage-independent colony formation and xenograft tumor growth with increased cell proliferation and reduced squamous-cell differentiation. Moreover, Notch1 appears to mediate replicative senescence as well as transforming growth factor-ß-induced cellular senescence in non-transformed cells and that HPV E6/E7 targets Notch1 for inactivation to prevent senescence, revealing a tumor-suppressor attribute of endogenous Notch1. In aggregate, cellular senescence checkpoint functions may influence dichotomous Notch activities in the neoplastic context.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Receptor Notch1/metabolismo , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Pontos de Checagem do Ciclo Celular , Transformação Celular Viral , Células Cultivadas , Senescência Celular , Carcinoma de Células Escamosas do Esôfago , Esôfago/citologia , Esôfago/metabolismo , Humanos , Queratinócitos/metabolismo , Fosforilação , Fator de Crescimento Transformador beta/metabolismo , Proteínas Virais/metabolismo
6.
Oncogene ; 34(17): 2167-77, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-24909170

RESUMO

Somatic alterations of fibroblast growth factor receptors (FGFRs) have been described in a wide range of malignancies. A number of anti-FGFR therapies are currently under investigation in clinical trials for subjects with FGFR gene amplifications, mutations and translocations. Here, we develop cell line models of acquired resistance to FGFR inhibition by exposure of cell lines harboring FGFR3 gene amplification and translocation to the selective FGFR inhibitor BGJ398 and multitargeted FGFR inhibitor ponatinib. We show that the acquisition of resistance is rapid, reversible and characterized by an epithelial to mesenchymal transition and a switch from dependency on FGFR3 to ERBB family members. Acquired resistance was associated with demonstrable changes in gene expression including increased production of ERBB2/3 ligands, which were sufficient to drive resistance in the setting of FGFR3 dependency but not dependency on other FGFR family members. These data support the concept that activation of ERBB family members is sufficient to bypass dependency on FGFR3 and suggest that concurrent inhibition of these two pathways may be desirable when targeting FGFR3-dependent cancers.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Transição Epitelial-Mesenquimal , Imidazóis/farmacologia , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridazinas/farmacologia , Pirimidinas/farmacologia , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Humanos , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética
7.
Oncogene ; 30(32): 3522-36, 2011 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-21423204

RESUMO

Ovarian cancer is a complex and deadly disease that remains difficult to detect at an early curable stage. Furthermore, although some oncogenic (Kras, Pten/PI3K and Trp53) pathways that are frequently mutated, deleted or amplified in ovarian cancer are known, how these pathways initiate and drive specific morphological phenotypes and tumor outcomes remain unclear. We recently generated Pten(fl/fl); Kras(G12D); Amhr2-Cre mice to disrupt the Pten gene and express a stable mutant form of Kras(G12D) in ovarian surface epithelial (OSE) cells. On the basis of histopathologic criteria, the mutant mice developed low-grade ovarian serous papillary adenocarcinomas at an early age and with 100% penetrance. This highly reproducible phenotype provides the first mouse model in which to study this ovarian cancer subtype. OSE cells isolated from ovaries of mutant mice at 5 and 10 weeks of age exhibit temporal changes in the expression of specific Mullerian epithelial marker genes, grow in soft agar and develop ectopic invasive tumors in recipient mice, indicating that the cells are transformed. Gene profiling identified specific mRNAs and microRNAs differentially expressed in purified OSE cells derived from tumors of the mutant mice compared with wild-type OSE cells. Mapping of transcripts or genes between the mouse OSE mutant data sets, the Kras signature from human cancer cell lines and the human ovarian tumor array data sets, documented significant overlap, indicating that KRAS is a key driver of OSE transformation in this context. Two key hallmarks of the mutant OSE cells in these mice are the elevated expression of the tumor-suppressor Trp53 (p53) and its microRNA target, miR-34a-c. We propose that elevated TRP53 and miR-34a-c may exert negatively regulatory effects that reduce the proliferative potential of OSE cells leading to the low-grade serous adenocarcinoma phenotype.


Assuntos
Transformação Celular Neoplásica/genética , Células Epiteliais/metabolismo , PTEN Fosfo-Hidrolase/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Carcinoma Epitelial do Ovário , Linhagem Celular Transformada , Células Cultivadas , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patologia , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , MicroRNAs/genética , Transplante de Neoplasias , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ovário/citologia , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/genética
8.
Clin Exp Rheumatol ; 28(5): 679-85, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20822708

RESUMO

OBJECTIVES: To elucidate the incidence rate and relative risk of tuberculosis (TB) in patients with rheumatoid arthritis (RA) compared to the general population in Hong Kong between 2004 and 2008, and to assess whether this risk is associated with exposure to tumour necrosis factor (TNF) blockers after adjusting for other known risk factors. METHODS: We reviewed all the medical records of RA patients to determine the standardised incidence ratio (SIR) of TB in RA patients. Independent explanatory variables associated with active TB in RA were ascertained using the Cox regression model. RESULTS: A total of 2441 RA patients followed at the 5 centres were recruited. The mean age at the start of follow up was 56±14 years. The median follow-up duration was 6,616 and 185 patient-years for the TNF naive and TNF treated groups, respectively. Compared to age- and sex-matched population controls, the SIR of active TB in RA was significantly increased (SIR for TNF naïve RA: 2.35, 95% CI 1.17-4.67, p=0.013, SIR for TNF treated RA: 34.92, 95% CI 8.89-137.20, p<0.001). Independent explanatory variables associated with an increase risk of active TB included older age at study entry (RR 1.05, p=0.013) a past history of pulmonary TB (RR 5.48, p=0.001), extra-pulmonary TB (RR 16.45, p<0.001), Felty's syndrome (RR 43.84, p=0.005), prednisolone>10mg daily (RR 4.44, p=0.009) and the use of TNF blockers (RR 12.48, p<0.001). CONCLUSIONS: Exposure to TNF blockers remained to be an independent risk factor for TB in RA after adjusting for other known risk factors.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Antirreumáticos/efeitos adversos , Artrite Reumatoide/epidemiologia , Imunossupressores/efeitos adversos , Tuberculose/epidemiologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Anticorpos Monoclonais/imunologia , Antirreumáticos/imunologia , Artrite Reumatoide/tratamento farmacológico , Comorbidade , Feminino , Hong Kong/epidemiologia , Humanos , Hospedeiro Imunocomprometido , Terapia de Imunossupressão , Imunossupressores/imunologia , Incidência , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Risco , Tuberculose/etiologia
9.
Oncogene ; 29(18): 2616-27, 2010 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-20154724

RESUMO

Recent studies have established that amplification of the MET proto-oncogene can cause resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) in non-small cell lung cancer (NSCLC) cell lines with EGFR-activating mutations. The role of non-amplified MET in EGFR-dependent signaling before TKI resistance, however, is not well understood. Using NSCLC cell lines and transgenic models, we demonstrate here that EGFR activation by either mutation or ligand binding increases MET gene expression and protein levels. Our analysis of 202 NSCLC patient specimens was consistent with these observations: levels of MET were significantly higher in NSCLC with EGFR mutations than in NSCLC with wild-type EGFR. EGFR regulation of MET levels in cell lines occurred through the hypoxia-inducible factor (HIF)-1alpha pathway in a hypoxia-independent manner. This regulation was lost, however, after MET gene amplification or overexpression of a constitutively active form of HIF-1alpha. EGFR- and hypoxia-induced invasiveness of NSCLC cells, but not cell survival, were found to be MET dependent. These findings establish that, absent MET amplification, EGFR signaling can regulate MET levels through HIF-1alpha and that MET is a key downstream mediator of EGFR-induced invasiveness in EGFR-dependent NSCLC cells.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Receptores ErbB/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas c-met/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Animais , Hipóxia Celular , Linhagem Celular Tumoral , Receptores ErbB/antagonistas & inibidores , Amplificação de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/análise , Camundongos , Invasividade Neoplásica , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-met/análise , Proteínas Proto-Oncogênicas c-met/genética , Receptores de Fatores de Crescimento/análise , Receptores de Fatores de Crescimento/genética
10.
Oncogene ; 29(11): 1588-97, 2010 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-19966866

RESUMO

Germline TSC1 or TSC2 mutations cause tuberous sclerosis complex (TSC), a hamartoma syndrome with lung involvement. To explore the potential interaction between TSC1 and KRAS activation in lung cancer, mice in which Tsc1 loss and Kras(G12D) expression occur in a small fraction of lung epithelial cells were generated. Mice with a combined Tsc1-Kras(G12D) mutation had dramatically reduced tumor latency (median survival: 11.6-15.6 weeks) in comparison with Kras(G12D) alone mutant mice (median survival: 27.5 weeks). Tsc1-Kras(G12D) tumors showed consistent activation of mTOR (mammalian target of rapamycin)C1 and responded to treatment with rapamycin, leading to significantly improved survival, whereas rapamycin had minor effects on cancers in Kras(G12D) alone mice. Loss of heterozygosity for TSC1 or TSC2 was found in 22% of 86 human lung cancer specimens. However, none of the 80 lung cancer lines studied showed evidence of the lack of expression of either TSC1 or TSC2 or a signaling pattern corresponding to complete loss. These data indicate that Tsc1 loss synergizes with the Kras mutation to enhance lung tumorigenesis in the mouse, but that this is a rare event in human lung cancer. Rapamycin may have unique benefit for patients with lung cancer, for whom the TSC1/TSC2 function is limited.


Assuntos
Neoplasias Pulmonares/prevenção & controle , Proteínas Proto-Oncogênicas p21(ras)/genética , Sirolimo/farmacologia , Proteínas Supressoras de Tumor/genética , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Brônquios/patologia , Linhagem Celular , Ativação Enzimática , Humanos , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Estimativa de Kaplan-Meier , Perda de Heterozigosidade , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos , Mutação , Proteínas , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Serina-Treonina Quinases TOR , Fatores de Transcrição/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo
11.
Br J Cancer ; 99(2): 245-52, 2008 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-18594528

RESUMO

Somatic mutations of LKB1 tumour suppressor gene have been detected in human cancers including non-small cell lung cancer (NSCLC). The relationship between LKB1 mutations and clinicopathological characteristics and other common oncogene mutations in NSCLC is inadequately described. In this study we evaluated tumour specimens from 310 patients with NSCLC including those with adenocarcinoma, adenosquamous carcinoma, and squamous cell carcinoma histologies. Tumours were obtained from patients of US (n=143) and Korean (n=167) origin and screened for LKB1, KRAS, BRAF, and EGFR mutations using RT-PCR-based SURVEYOR-WAVE method followed by Sanger sequencing. We detected mutations in the LKB1 gene in 34 tumours (11%). LKB1 mutation frequency was higher in NSCLC tumours of US origin (17%) compared with 5% in NSCLCs of Korean origin (P=0.001). They tended to occur more commonly in adenocarcinomas (13%) than in squamous cell carcinomas (5%) (P=0.066). LKB1 mutations associated with smoking history (P=0.007) and KRAS mutations (P=0.042) were almost mutually exclusive with EGFR mutations (P=0.002). The outcome of stages I and II NSCLC patients treated with surgery alone did not significantly differ based on LKB1 mutation status. Our study provides clinical and molecular characteristics of NSCLC, which harbour LKB1 mutations.


Assuntos
Povo Asiático/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Genes Supressores de Tumor , Neoplasias Pulmonares/genética , Mutação , Proteínas Serina-Treonina Quinases/genética , População Branca/genética , Quinases Proteína-Quinases Ativadas por AMP , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/etnologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Receptores ErbB/genética , Genes ras , Predisposição Genética para Doença , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/etnologia , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas B-raf/genética
12.
Oncogene ; 27(34): 4702-11, 2008 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-18408761

RESUMO

Genetic alterations in the kinase domain of the epidermal growth factor receptor (EGFR) in non-small cell lung cancer (NSCLC) patients are associated with sensitivity to treatment with small molecule tyrosine kinase inhibitors. Although first-generation reversible, ATP-competitive inhibitors showed encouraging clinical responses in lung adenocarcinoma tumors harboring such EGFR mutations, almost all patients developed resistance to these inhibitors over time. Such resistance to first-generation EGFR inhibitors was frequently linked to an acquired T790M point mutation in the kinase domain of EGFR, or upregulation of signaling pathways downstream of HER3. Overcoming these mechanisms of resistance, as well as primary resistance to reversible EGFR inhibitors driven by a subset of EGFR mutations, will be necessary for development of an effective targeted therapy regimen. Here, we show that BIBW2992, an anilino-quinazoline designed to irreversibly bind EGFR and HER2, potently suppresses the kinase activity of wild-type and activated EGFR and HER2 mutants, including erlotinib-resistant isoforms. Consistent with this activity, BIBW2992 suppresses transformation in isogenic cell-based assays, inhibits survival of cancer cell lines and induces tumor regression in xenograft and transgenic lung cancer models, with superior activity over erlotinib. These findings encourage further testing of BIBW2992 in lung cancer patients harboring EGFR or HER2 oncogenes.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Quinazolinas/uso terapêutico , Receptor ErbB-2/antagonistas & inibidores , Afatinib , Animais , Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/patologia , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Camundongos Transgênicos , Células NIH 3T3 , Fosforilação/efeitos dos fármacos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Resultado do Tratamento , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Oncogene ; 26(34): 5023-7, 2007 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-17311002

RESUMO

Mutations in the ERBB2 gene were recently found in approximately 2% of primary non-small cell lung cancer (NSCLC) specimens; however, little is known about the functional consequences and the relevance to responsiveness to targeted drugs for most of these mutations. Here, we show that the major lung cancer-derived ERBB2 mutants, including the most frequent mutation, A775insYVMA, lead to oncogenic transformation in a cellular assay. Murine cells transformed with these mutants were relatively resistant to the reversible epidermal growth factor receptor (EGFR) inhibitor erlotinib, resembling the resistant phenotype found in cells carrying the homologous mutations in exon 20 of EGFR. However, the same cells were highly sensitive to the irreversible dual-specificity EGFR/ERBB2 kinase inhibitor HKI-272, as were those overexpressing wild-type ERBB2. Finally, the NSCLC cell line, Calu-3, overexpressing wild-type ERBB2 owing to a high-level amplification of the ERBB2 gene were highly sensitive to HKI-272. These results provide a rationale for treatment of patients with ERBB2-mutant or ERBB2-amplified lung tumors with HKI-272.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/genética , Genes erbB-2 , Neoplasias Pulmonares/genética , Mutação , Inibidores de Proteínas Quinases/farmacologia , Quinolinas/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Receptores ErbB/antagonistas & inibidores , Amplificação de Genes , Humanos , Neoplasias Pulmonares/enzimologia
14.
Oncogene ; 26(20): 2815-21, 2007 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-17072335

RESUMO

An adequate and appropriate response to physiological and pathophysiological stresses is critical for long-term homeostasis and viability of the aging organism. Previous work has pointed to the immune system, telomeres and DNA repair pathways as important and distinct determinants of a normal healthy lifespan. In this study, we explored the genetic interactions of telomeres and DNA-PKcs, a protein involved in non-homologous end-joining (NHEJ) and immune responses, in the context of a key aspect of aging and lifespan--the capacity to mount an acute and appropriate immune-mediated stress response. We observed that the combination of DNA-PKcs deficiency and telomere dysfunction resulted in a shortened lifespan that was reduced further following viral infection or experimental activation of the innate immune response. Analysis of the innate immune response in the DNA-PKcs-deficient mice with short dysfunctional telomeres revealed high basal serum levels of tumor necrosis factor alpha (TNFalpha) and hyper-active cytokine responses upon challenge with polyinosinic-polycytidylic acid (poly-IC). We further show that serum cytokine levels become elevated in telomere dysfunctional mice as a function of age. These results raise speculation that these genetic factors may contribute to misdirected immune responses of the aged under conditions of acute and chronic stress.


Assuntos
Proteína Quinase Ativada por DNA/genética , Proteínas de Ligação a DNA/genética , Longevidade/genética , Proteínas Nucleares/genética , Estresse Fisiológico/genética , Estresse Fisiológico/mortalidade , Telômero/metabolismo , Animais , Cruzamentos Genéticos , Hepatite Animal/sangue , Hepatite Animal/genética , Hepatite Animal/imunologia , Interleucina-1beta/sangue , Interleucina-6/sangue , Fígado/patologia , Camundongos , Camundongos Transgênicos , Vírus da Hepatite Murina/imunologia , RNA/genética , Estresse Fisiológico/patologia , Telomerase/genética , Telômero/fisiologia , Fator de Necrose Tumoral alfa/sangue
15.
Oncogene ; 25(14): 2105-12, 2006 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-16288213

RESUMO

Activating mutations in K-ras are one of the most common genetic alterations in human lung cancer. To dissect the role of K-ras activation in bronchial epithelial cells during lung tumorigenesis, we created a model of lung adenocarcinoma by generating a conditional mutant mouse with both Clara cell secretory protein (CC10)-Cre recombinase and the Lox-Stop-Lox K-ras(G12D) alleles. The activation of K-ras mutant allele in CC10 positive cells resulted in a progressive phenotype characterized by cellular atypia, adenoma and ultimately adenocarcinoma. Surprisingly, K-ras activation in the bronchiolar epithelium is associated with a robust inflammatory response characterized by an abundant infiltration of alveolar macrophages and neutrophils. These mice displayed early mortality in the setting of this pulmonary inflammatory response with a median survival of 8 weeks. Bronchoalveolar lavage fluid from these mutant mice contained the MIP-2, KC, MCP-1 and LIX chemokines that increased significantly with age. Cell lines derived from these tumors directly produced MIP-2, LIX and KC. This model demonstrates that K-ras activation in the lung induces the elaboration of inflammatory chemokines and provides an excellent means to further study the complex interactions between inflammatory cells, chemokines and tumor progression.


Assuntos
Genes ras , Neoplasias Pulmonares/genética , Pneumonia/genética , Animais , Sequência de Bases , Líquido da Lavagem Broncoalveolar , Linhagem Celular Tumoral , Primers do DNA , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/complicações , Neoplasias Pulmonares/fisiopatologia , Macrófagos Alveolares/patologia , Camundongos , Camundongos Mutantes , Pneumonia/complicações , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Artigo em Inglês | MEDLINE | ID: mdl-16869734

RESUMO

Lung cancer is the leading cause of cancer mortality worldwide. With the recent success of molecularly targeted therapies in this disease, a detailed knowledge of the spectrum of genetic lesions in lung cancer represents a critical step in the development of additional effective agents. An integrated high-resolution survey of regional amplifications and deletions and gene expression profiling of non-small-cell lung cancers (NSCLC) identified 93 focal high-confidence copy number alterations (CNAs), with 21 spanning less than 0.5 Mb with a median of five genes. Most CNAs were novel and included high-amplitude amplification and homozygous deletion events. Pathogenic relevance of these genomic alterations was further reinforced by their recurrence and overlap with focal alterations of other tumor types. Additionally, the comparison of the genomic profiles of the two major subtypes of NSCLC, adenocarcinoma (AC) and squamous cell carcinoma (SCC), showed an almost complete overlap with the exception of one amplified region on chromosome 3, specific for SCC. Among the few genes overexpressed within this amplicon was p63, a known regulator of squamous cell differentiation. These findings suggest that the AC and SCC subtypes may arise from a common cell of origin and they are driven to their distinct phenotypic end points by altered expression of a limited number of key genes such as p63.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Perfilação da Expressão Gênica , Neoplasias Pulmonares/genética , Adenocarcinoma/classificação , Adenocarcinoma/genética , Carcinoma Pulmonar de Células não Pequenas/classificação , Carcinoma de Células Escamosas/classificação , Carcinoma de Células Escamosas/genética , Cromossomos Humanos Par 3/genética , Citogenética , Dosagem de Genes , Humanos , Hibridização in Situ Fluorescente , Neoplasias Pulmonares/classificação , Proteínas de Membrana/genética , Oncogenes
17.
J Inorg Biochem ; 76(3-4): 187-95, 1999 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-10605836

RESUMO

Alkylated derivatives of the iron storage protein, ferritin, have been prepared by carbodiimide-activated coupling of long chain (C9, C12, C14) primary amines to surface carboxylic acid residues. In the case of a nonyl-derivatized protein, alkylation results in covalent modification of approximately 400 of the 520 amino acid carboxyl groups in the protein molecule. The hydrophobic proteins have a net positive charge in water and can be transferred from THF/water mixtures into dichloromethane, ethyl acetate and toluene by addition of small amounts of NaCl. Transmission electron microscopy, analytical ultracentrifugation, and polyacrylamide gel electrophoresis indicate that the hydrophobic proteins dissolve in the organic solvents as structurally intact, non-aggregated macromolecules which can be subsequently back-extracted into water.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA