Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Cancer Ther ; 16(1): 228-238, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27811012

RESUMO

HER2/ERBB2 status is used to select patients for HER2-targeted therapy. HER2/ERBB2 amplification/overexpression of upper gastrointestinal (UGI) adenocarcinomas was determined locally or in two central laboratories to select patients for the TRIO-013/LOGiC trial of chemotherapy with or without lapatinib. Patients selected locally had central laboratory confirmation of HER2 amplification for inclusion in the primary efficacy population. HER2 was assessed with PathVysion or IQ PharmDx FISH and HercepTest immunohistochemistry assays. Associations with outcomes were retrospectively evaluated. Overall, HER2 status was determined in UGI cancers from 4,674 patients in a central laboratory for eligibility (1,995 cases) and for confirmation of local HER2 results (333 cases). Of 1,995 adenocarcinomas screened centrally, 322 (16.1%) had HER2-amplified disease with 29 (1.5%) showing HER2 genomic heterogeneity. Men and older patients had higher rates of amplification. Of 545 patients accrued to the trial (gastric, 87.3%; GEJ, 8.3% and esophageal cancer, 4.4%), 487 patients (89%) were centrally confirmed as having HER2-amplified disease. Concordance between central and local HER2 testing was 83%. Concordance between PathVysion and IQ PharmDx FISH assays was 99% and FISH in the two central laboratories was 95%. Lapatinib-treated Asian participants and those less than 60 years had significant improvement in progression-free survival (PFS), particularly among those whose cancers had 5.01-10.0 and >10.0-fold amplification of HER2 In conclusion, HER2 is commonly amplified in UGI adenocarcinomas with amplification highly correlated to overexpression, and HER2 amplification levels correlated with PFS. While HER2 genomic heterogeneity occurs, its prevalence is low. Mol Cancer Ther; 16(1); 228-38. ©2016 AACR.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico , Quinazolinas/uso terapêutico , Receptor ErbB-2/antagonistas & inibidores , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Adenocarcinoma/mortalidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidade , Feminino , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Lapatinib , Masculino , Pessoa de Meia-Idade , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidade , Resultado do Tratamento , Adulto Jovem
2.
J Craniomaxillofac Surg ; 44(8): 1054-60, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27316856

RESUMO

Giant cell granuloma (GCG) of the jaw is a rare, well-known feature of neurofibromatosis type 1 (NF1), an inborn multisystem disorder. Recently, the development of GCG in NF1 was attributed to second hit mutations in the NF1 gene. The treatment of GCG is pragmatic with a preference for local curettage of lytic osseous areas. This report describes the surgical therapy of an NF1-affected female with multilocular mandibular GCG and hypodontia who additionally suffered from a brain tumour and Hashimoto's thyroiditis. Although local recurrence of GCG was noted, augmentation of the curetted cavities with a bone substitute in successive interventions successfully restored the extensive periradicular local defects and stabilised the teeth. A meticulous in vitro study of the GCG specimen revealed a second hit mutation in the NF1 gene in the GCG spindle-cells. This study contributes to the increasing knowledge of the molecular basis for GCG in the jaw of NF1 patients, indicating that it is a neoplasm.


Assuntos
Genes da Neurofibromatose 1 , Granuloma de Células Gigantes/genética , Doenças Mandibulares/genética , Mutação , Adolescente , Substitutos Ósseos , Neoplasias Encefálicas/complicações , Tomografia Computadorizada de Feixe Cônico , Feminino , Granuloma de Células Gigantes/diagnóstico por imagem , Granuloma de Células Gigantes/cirurgia , Humanos , Processamento de Imagem Assistida por Computador , Doenças Mandibulares/diagnóstico por imagem , Doenças Mandibulares/cirurgia , Neurofibromatose 1/complicações , Neurofibromatose 1/genética , Radiografia Panorâmica , Recidiva
3.
Oncotarget ; 6(32): 33867-77, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26418954

RESUMO

BACKGROUND: Glioblastomas (GBM) are often characterized by an elevated expression of the epidermal growth factor receptor variant III (EGFRvIII). We used GBM cell lines with native EGFRvIII expression to determine whether this EGFR variant affects radiosensitivity with or without EGFR targeting. METHODS: Experiments were performed with GBM cell lines lacking (LN229, U87MG, U251, CAS-1) or endogenously expressing EGFRvIII (BS153, DKMG). The two latter cell lines were also used to establish sublines with a low (-) or a high proportion (+) of cells expressing EGFRvIII. EGFR signaling and the cell cycle were analyzed using Western blot and flow cytometry; cell survival was assessed by colony forming assay and double-strand break repair capacity by immunofluorescence. RESULTS: DKMG and BS153 parental cells with heterogeneous EGFRvIII expression were clearly more radiosensitive compared to other GBM cell lines without EGFRvIII expression. However, no significant difference was observed in cell proliferation, clonogenicity or radiosensitivity between the EGFRvIII- and + sublines derived from DKMG and BS153 parental cells. Expression of EGFRvIII was associated with decreased DSB repair capacity for BS153 but not for DKMG cells. The effects of EGFR targeting by gefitinib alone or in combination with irradiation were also found not to depend on EGFRvIII expression. Gefitinib was only observed to influence the proliferation of EGFRvIII- BS153 cells. CONCLUSION: The data indicate that EGFRvIII does not alter radiosensitivity with or without anti-EGFR treatment.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Quinazolinas/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Separação Celular , Sobrevivência Celular , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Citometria de Fluxo , Gefitinibe , Glioblastoma/metabolismo , Humanos , Microscopia de Fluorescência , Tolerância a Radiação , Transdução de Sinais , Raios X
4.
Radiother Oncol ; 115(1): 120-7, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25796091

RESUMO

PURPOSE: How EGF receptor (EGFR) inhibition induces cellular radiosensitization and with that increase in tumor control is still a matter of discussion. Since EGFR predominantly regulates cell cycle and proliferation, we studied whether a G1-arrest caused by EGFR inhibition may contribute to these effects. MATERIALS AND METHODS: We analyzed human non-small cell lung cancer (NSCLC) cell lines either wild type (wt) or mutated in p53 (A549, H460, vs. H1299, H3122) and HCT116 cells (p21 wt and negative). EGFR was inhibited by BIBX1382BS, erlotinib or cetuximab; p21 was knocked down by siRNA. Functional endpoints analyzed were cell signaling, proliferation, G1-arrest, cell survival as well as tumor control using an A549 tumor model. RESULTS: When combined with IR, EGFR inhibition enhances the radiation-induced permanent G1 arrest, though solely in cells with intact p53/p21 signaling. This increase in G1-arrest was always associated with enhanced cellular radiosensitivity. Strikingly, this effect was abrogated when cells were re-stimulated, suggesting the initiation of dormancy. In line with this, only a small non-significant increase in tumor control was observed for A549 tumors treated with fractionated RT and EGFR inhibition. CONCLUSION: For NSCLC cells increase in radiosensitivity by EGFR inhibition results from enhanced G1-arrest. However, this effect does not lead to improved tumor control because cells can be released from this arrest by re-stimulation.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Receptores ErbB/antagonistas & inibidores , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Neoplasias Pulmonares/patologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Divisão Celular , Linhagem Celular Tumoral , Cetuximab , Cloridrato de Erlotinib , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos da radiação , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/radioterapia , Camundongos , Camundongos Nus , Quinazolinas/farmacologia , RNA Interferente Pequeno/genética , Tolerância a Radiação/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Virchows Arch ; 465(6): 673-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25267585

RESUMO

Transcriptional regulation of the TERT gene is a major cause of the cancer-specific increase in telomerase activity. Recently, frequent somatic mutations in the TERT promoter have been described in several tumor entities such as melanoma, glioblastoma, bladder cancer, and hepatocellular carcinoma. By generating a putative consensus binding site for ETS transcription factors within the TERT promoter, these mutations are predicted to increase promoter activity and TERT transcription. In order to improve the understanding of the role of TERT promoter mutation in liver tumorigenesis, the mutational status of the TERT promoter was analyzed in 78 hepatocellular carcinomas, 15 hepatocellular adenomas, and 52 intrahepatic cholangiocarciomas. The promoter region of TERT was screened for the two hotspot mutations using PCR and restriction fragment length analysis, utilizing the introduction of novel restriction sites by the somatic mutations. TERT promoter mutation was found in 37 of 78 hepatocellular carcinomas (47 %) and was restricted to the -124C>T mutation. Frequency of mutations was associated with grade of differentiation ranging from 39 % in well-differentiated tumors to 73 % in high-grade hepatocellular carcinomas. TERT promoter mutations were not found in 15 hepatocellular adenomas and 52 intrahepatic cholangiocarcinomas. These data show that TERT promoter mutation is the most frequent genetic alteration in hepatocellular carcinoma known at this time. The striking predominance of the -124C>T mutation compared with other tumor entities suggest a biological difference of the two hotspot mutations. Analysis of TERT promoter mutation might become a diagnostic tool distinguishing hepatocellular adenoma from well-differentiated hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Regiões Promotoras Genéticas/genética , Telomerase/genética , Adenoma/genética , Adolescente , Adulto , Idoso , Neoplasias dos Ductos Biliares/genética , Ductos Biliares Intra-Hepáticos , Criança , Colangiocarcinoma/genética , Análise Mutacional de DNA , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Adulto Jovem
6.
Radiother Oncol ; 109(2): 286-92, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23953412

RESUMO

BACKGROUND AND PURPOSE: There is a great need to improve the outcome of locoregionally advanced squamous cell carcinomas of the head and neck (HNSCC). Standard treatment includes a combination of surgery, radio- and chemotherapy. The addition of molecular targeting agents to conventional treatment may improve outcomes. In this study the Raf inhibitor sorafenib was used to increase the radiosensitivity of HNSCC cell lines. MATERIAL AND METHODS: In a panel of six cell lines (A549, FaDu, UTSCC 60A, UTSCC 42A, UTSCC 42B, UTSCC 29) radiosensitivity was measured by colony formation assay and apoptosis and cell cycle analysis were performed by flow cytometry. DNA repair was analyzed by 53BP1 immunohistochemistry. RESULTS: Sorafenib added prior to irradiation resulted in an increased cellular radiosensitivity (DEF0.5=1.11-1.84). Radiosensitization was not caused by an enhanced rate of apoptosis or cell cycle effects. In contrast, sorafenib was shown for the first time to block the repair of DNA double-strand breaks (DSB). CONCLUSION: Our data suggest that sorafenib may be used to overcome the radioresistance of HNSCC through the inhibition of DSB repair.


Assuntos
Carcinoma de Células Escamosas/radioterapia , Neoplasias de Cabeça e Pescoço/radioterapia , Niacinamida/análogos & derivados , Compostos de Fenilureia/uso terapêutico , Radiossensibilizantes/uso terapêutico , Quinases raf/antagonistas & inibidores , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Reparo do DNA , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Niacinamida/uso terapêutico , Sorafenibe , Carcinoma de Células Escamosas de Cabeça e Pescoço
7.
J Thorac Cardiovasc Surg ; 146(4): 781-7, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23778087

RESUMO

OBJECTIVE: The aim of this study was to assess the influence of lymphatic and vascular invasion on overall survival in patients with surgically resected non-small cell lung cancer (NSCLC) without lymph node and distant metastases. METHODS: From January 1999 to December 2009, a total of 190 NSCLC patients with node-negative pT1-pT4 disease underwent radical resection with lymphadenectomy. Pathologic reports were reclassified to the TNM-7 version, and the influence of lymphatic and vascular invasion on overall survival was examined using Kaplan-Meier and adjusted Cox proportional hazards analyses. RESULTS: Lymphatic invasion was present in 34 (17.9%) and vascular invasion in 28 (14.7%) of 190 cases. Lymphatic and vascular invasions were correlated with higher Union for International Cancer Control stages (P = .056 and P = .011, respectively) and poor differentiated tumors (P = .051 and P = .012, respectively). There was no difference between pT1a and pT1b tumors in the presence of lymphatic (P = .912) or vascular (P = .134) invasion. Survival analyses revealed lymphatic (P < .001) and vascular (P = .008) invasion as statistically significant for the entire study population. Multivariable Cox analysis adjusted for age, Union for International Cancer Control stage, and lymphatic and vascular invasion confirmed lymphatic, but not vascular, invasion as an independent prognostic factor (P < .001; hazard ratio, 3.002; 95% confidence interval, 1.780-5.061). Especially in early stages, lymphatic invasion was associated with poorer overall survival in pT1a (P < .001), pT1b (P = .019), and pT2a (P = .028) tumors. CONCLUSIONS: Lymphatic invasion represents an independent risk factor for node-negative NSCLC. Its implications on therapy decision making should be further evaluated, especially in early stages.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/cirurgia , Neoplasias Pulmonares/cirurgia , Excisão de Linfonodo , Pneumonectomia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/secundário , Feminino , Humanos , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Excisão de Linfonodo/efeitos adversos , Excisão de Linfonodo/mortalidade , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Invasividade Neoplásica , Estadiamento de Neoplasias , Pneumonectomia/efeitos adversos , Pneumonectomia/mortalidade , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
8.
Radiother Oncol ; 107(2): 242-6, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23602369

RESUMO

BACKGROUND AND PURPOSE: When treated by radiotherapy, patients with squamous cell carcinomas of the head and neck (HNSCC) positive for HPV and p16(INK4a) possess a clearly favorable prognosis as compared to those with HPV-negative HNSCC. The aim of this work was to study whether the better outcomes might be caused by an enhanced cellular radiosensitivity. MATERIALS AND METHODS: The radiation response of five HPV/p16(INK4a)-positive and five HPV-negative cell lines was characterized with regard to cellular radiosensitivity by colony formation assay. Furthermore G1- and G2-arrest, apoptosis and residual DNA double-strand breaks (DSB) were analyzed by the colcemid-based G1-efflux assay, propidium iodide staining, the detection of PARP cleavage, the fluorescence-based detection of caspase activity and the immunofluorescence staining of γH2AX and 53BP1 foci. RESULTS: On average, the cellular radiosensitivity of the HNSCC cell lines positive for HPV and p16(INK4a) was higher as compared to the sensitivity of a panel of five HPV-negative HNSCC cell lines (SF3=0.2827 vs. 0.4455). The higher sensitivity does not result from increased apoptosis or the execution of a permanent G1-arrest, but is rather associated with both, elevated levels of residual DSBs and extensive G2-arrest. CONCLUSIONS: Increased cellular radiosensitivity due to compromised DNA repair capacity is likely to contribute to the improved outcome of patients with HPV/p16(INK4a)-positive tumors when treated by radiotherapy.


Assuntos
Carcinoma de Células Escamosas/radioterapia , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Neoplasias de Cabeça e Pescoço/radioterapia , Proteínas de Neoplasias/análise , Papillomaviridae/isolamento & purificação , Tolerância a Radiação , Apoptose , Carcinoma de Células Escamosas/química , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/virologia , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina , Fase G1 , Neoplasias de Cabeça e Pescoço/química , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/virologia , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço
9.
Exp Mol Pathol ; 94(1): 155-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23022742

RESUMO

BACKGROUND: Several lines of evidence indicate that mutational activation of KRAS is an early event in the carcinogenesis of non-small cell lung cancer (NSCLC). Nonetheless, previous studies report high frequencies of divergent KRAS mutational status between primary NSCLC and corresponding metastases. This suggests heterogeneity of the primary tumor in respect to its KRAS status. We therefore aimed to examine the frequency and the extent of such intratumoral heterogeneity. METHODS: 40 NSCLC were examined for intratumoral heterogeneity of KRAS mutation (20 adenocarcinomas, 10 squamous cell carcinomas and 10 large cell carcinomas). Three to eight different tumor areas were analyzed for KRAS mutation and up to four corresponding lymph node metastases were included for analysis in nineteen cases. A combination of different methods for screening of heterogeneity and its validation were used including direct sequencing, laser-capture microdissection for tumor cell enrichment and the very sensitive ARMS/S method. RESULTS: Mutations of KRAS were found in 13/30 adenocarcinomas and large cell carcinomas. No mutations were detected in 10 squamous cell carcinomas. Four cases showed heterogeneous KRAS results by direct sequencing. More sensitive methods for KRAS mutation analysis revealed false negative results due to admixture of non-neoplastic cells in all of these samples. Intratumoral heterogeneity of KRAS mutational status was therefore confirmed in none of the analyzed cases. In addition, identical KRAS mutations were present in the primary tumor and the corresponding lymph node metastases in 19 cases examined. CONCLUSIONS: Intratumoral heterogeneity of KRAS mutational status is rare in NSCLC but highly sensitive tools are required to reliably identify these mutations. This finding is in line with the hypothesis that oncogenic activation of KRAS is an early event and a bona fide "driver mutation" in NSCLC. Furthermore, future therapies targeting KRAS will not be limited by intratumoral heterogeneity.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/genética , Metástase Linfática/genética , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Carcinoma Pulmonar de Células não Pequenas/secundário , Genes ras , Variação Genética , Humanos , Neoplasias Pulmonares/patologia , Mutação , Proteínas Proto-Oncogênicas p21(ras) , Análise de Sequência de DNA
10.
Lung Cancer ; 79(3): 221-7, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23238037

RESUMO

Next to EGFR mutation, EGFR gene copy number evaluated by fluorescence in situ hybridization (FISH) emerged as a potential predictive marker for sensitivity to EGFR tyrosine kinase inhibitors, although controversial data exist. As the diagnostic accuracy of predictive biomarkers can be substantially limited by regional differences within tumors, heterogeneity of EGFR gene copy gain in NSCLC was assessed in this study. For this purpose, a novel tissue microarray (TMA) based analysis platform was developed. TMAs were constructed containing 8 different tissue cylinders from 144 primary NSCLCs. From 62 of these patients additional nodal metastases were sampled. EGFR gene copy number and EGFR expression was analyzed by FISH and immunohistochemistry according to the suggested guidelines. 13 (9.0%) of the 144 evaluated tumors showed EGFR amplification and 37 (25.7%) tumors high polysomy in at least one tumor area. In 7 (53.8%) of 13 amplified cases the analysis of different tumor areas revealed subclones without EGFR gene copy gain next to subclones with amplification. All of the 36 evaluable tumors with high polysomy showed heterogeneity of EGFR gene copy number with areas negative for gene copy gain within the individual tumors. Heterogeneity of EGFR gene copy gain in lung cancer challenges the concept of using small biopsies for the analysis of EGFR FISH status. EGFR gene copy number is highly heterogeneous within individual NSCLCs and this finding might well be a reason for the controversial clinical data existing regarding responsiveness to anti-EGFR therapy.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Receptores ErbB/genética , Dosagem de Genes/genética , Neoplasias Pulmonares/diagnóstico , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/terapia , Feminino , Humanos , Hibridização in Situ Fluorescente , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Prognóstico , Análise Serial de Tecidos
11.
Mod Pathol ; 25(12): 1566-73, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22899293

RESUMO

The HER2 protein, encoded by the ERBB2 gene, is a molecular target for the treatment of breast and gastric cancer by monoclonal antibodies or tyrosine kinase inhibitors. While intratumoral heterogeneity of ERBB2 amplification is rare in breast cancer it is reported to be frequent in bladder and colorectal cancer. To address the potential heterogeneity of the HER2 status in adenocarcinomas, squamous cell carcinomas and large cell undifferentiated carcinomas of the lung, 590 tumors were analyzed for HER2 overexpression and ERBB2 amplification using FDA-approved reagents for immunohistochemistry and fluorescence in-situ hybridization (FISH). Moderate and strong immunostaining (2+, 3+) was seen in 10% of the tumors. ERBB2 amplification was found in 17 (3%) lung cancer patients including 10 cases (2%) with high-level amplification forming gene clusters. ERBB2 amplification was significantly related to histologic subtype and tumor grade, resulting in 12% ERBB2 amplified tumors in the subgroup of high-grade adenocarcinomas. Heterogeneity was analyzed in all highly amplified tumors. For this purpose, all available tumor tissue blocks from these patients were evaluated. Heterogeneity of ERBB2 amplification was found in 4 of 10 tumors as assessed by FISH. These included two tumors with a mixture of low-level and high-level amplification and two tumors with non-amplified tumor areas next to regions with high-level ERBB2 amplification. High-level ERBB2 amplification occurs in a small fraction of lung cancers with a strong propensity to high-grade adenocarcinomas. Heterogeneity of amplification may limit the utility of anti-HER2 therapy in some of these tumors. Further attempts to assess the utility of HER2-targeting therapy in homogeneously amplified lung cancers appear to be justified.


Assuntos
Adenocarcinoma/genética , Carcinoma de Células Escamosas/genética , Amplificação de Genes , Neoplasias Pulmonares/genética , Receptor ErbB-2/genética , Adenocarcinoma/mortalidade , Adenocarcinoma/secundário , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/secundário , DNA de Neoplasias/análise , Alemanha/epidemiologia , Humanos , Hibridização in Situ Fluorescente , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Linfonodos/patologia , Pessoa de Meia-Idade , Taxa de Sobrevida , Análise Serial de Tecidos
12.
Oral Oncol ; 48(10): 991-996, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22694907

RESUMO

AIMS: Epidermal growth factor (EGFR) is involved in angiogenesis, cell differentiation, proliferation and progression of many cancers and is an important therapy target in lung and colorectal cancer. To determine the potential applicability of EGFR targeted therapies, EGFR status of over 800 salivary gland tumors of different entities were analyzed on DNA and protein level by FISH and IHC. MATERIALS AND METHODS: A tissue microarray was constructed from 721 carcinomas and 205 adenomas of the salivary gland. EGFR expression and EGFR gene copy number was assessed by means of immunohistochemistry and fluorescence in situ hybridization (FISH). EGFR mutation analysis of exon 19 and 21 was performed in a subset of 107 carcinomas. RESULTS: Positive immunohistochemical staining (definition?) for EGFR was shown in 324 of 663 (48.9%) salivary gland carcinomas. The frequency was dependent on the tumor entity and ranged from 17.9% (30 of 168 cases) positive immunostaining in acinic cell adenocarcinomas to 85.7% (42 of 49 cases) in Warthin tumors. No EGFR amplification was found by FISH. EGFR mutation analysis of Exon 19 and 21 in 107 salivary gland carcinomas revealed mutations in two acinic cell adenocarcinomas. CONCLUSION: EGFR protein expression is common in salivary gland tumors but is not associated with gene amplification. Activating mutations of EGFR are rare. Nonetheless, selected cases of patients with salivary gland carcinomas might potentially benefit of anti-EGFR therapy.


Assuntos
Adenoma/metabolismo , Carcinoma/metabolismo , Receptores ErbB/metabolismo , Neoplasias das Glândulas Salivares/metabolismo , Adenoma/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma/genética , Receptores ErbB/genética , Éxons , Feminino , Amplificação de Genes , Humanos , Hibridização in Situ Fluorescente , Masculino , Mutação , Neoplasias das Glândulas Salivares/genética
13.
Breast Cancer Res Treat ; 134(2): 561-7, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22610646

RESUMO

Women with triple-negative breast cancer (TNBC) do not benefit from endocrine therapy or trastuzumab. Chemotherapy is the only systemic therapy currently available. To reduce the elevated risk of disease progression in these patients, better treatment options are needed, which are less toxic and more targeted to this patient population. We performed a comprehensive analysis of potential targetable genetic aberrations affecting the receptor tyrosine kinase/RAS/MAPK pathway, which are observed at higher frequencies in adenocarcinomas of other organs. Sixty-five individual TNBCs were studied by sequence analysis for HER2 (exon 18-23), EGFR (exon 18-21), KRAS (exon 2), and BRAF (exon 15) mutations. In addition, a tissue microarray was constructed to screen for EGFR gene copy gain and EML4-ALK fusion by FISH. Triple-negative status was confirmed by immunohistochemistry and FISH on tissue microarray sections. EGFR and CK5/6 immunohistochemical analyses were performed for identification of the basal-like phenotype. In addition, mutation analysis of TP53 (exon 5-8) was included. Sequence analysis revealed HER2 gene mutation in only one patient (heterozygous missense mutation in exon 19: p.L755S). No mutations were found in EGFR, KRAS, and BRAF. High polysomy of EGFR was detected in 5 of the 62 informative cases by FISH. True EGFR gene amplification accompanied by strong membranous EGFR protein expression was observed in only one case. No rearrangement of the ALK gene was detected. Basal-like phenotype was identified in 38 of the 65 TNBCs (58.5 %). TP53 gene mutation was found in 36/63 (57.1 %) tumors. We conclude that targetable genetic aberrations in the receptor tyrosine kinase/RAS/MAPK pathway occur rarely in TNBC.


Assuntos
Neoplasias da Mama/genética , Carcinoma Ductal de Mama/genética , Mutação , Oncogenes , Adulto , Idoso , Idoso de 80 Anos ou mais , Quinase do Linfoma Anaplásico , Sequência de Bases , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/tratamento farmacológico , Carcinoma Ductal de Mama/metabolismo , Análise Mutacional de DNA , Receptores ErbB/genética , Feminino , Amplificação de Genes , Dosagem de Genes , Humanos , Hibridização in Situ Fluorescente , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras) , Receptores Proteína Tirosina Quinases/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Análise Serial de Tecidos , Proteína Supressora de Tumor p53/genética , Proteínas ras/genética
14.
J Clin Oncol ; 28(20): 3264-70, 2010 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-20498397

RESUMO

PURPOSE: Core needle biopsies (CNBs) are widely used to determine human epidermal growth factor receptor 2 (HER2) status in breast cancer. Recent publications reported up to 20% false-positive results on CNBs if immunohistochemistry (IHC) is compared with fluorescent in situ hybridization (FISH). To clarify, if confirmation of IHC positivity by FISH is generally required, we analyzed the reliability of IHC positivity on CNBs versus surgical specimens in a multi-institutional study. PATIENTS AND METHODS: Five pathologic laboratories contributed to this study by performing IHC on 500 CNBs and the corresponding surgical specimens overall. If IHC revealed score 2+ or 3+, HER2 status was confirmed by FISH in a central laboratory. We compared evaluation according to US Food and Drug Administration-approved scoring criteria and recently published American Society of Clinical Oncology (ASCO)-College of American Pathologists (CAP) guidelines. RESULTS: CNBs scored 3+ revealed five false-positive results if scoring followed the US Food and Drug Administration criteria (five of 40; 12.5%) and two false-positives in terms of the ASCO-CAP criteria (two of 33; 6.1%). IHC was false negative in one CNB only. By contrast, IHC on surgical specimens revealed five false-negative results, but only one false-positive result (one of 35; 2.9%) if scored following US Food and Drug Administration-approved criteria. With the aid of the ASCO-CAP criteria, false-positive IHC results were obtained in only one of the five participating institutions. CONCLUSION: IHC 3+ scores on CNBs proved to be reliable in four of the five participating institutions if scoring followed the ASCO-CAP criteria. Therefore, accurate determination of HER2 status in breast cancer is possible on CNB using the common strategy to screen all cases by IHC and retest only 2+ scores by FISH. Prerequisites are quality assurance and the application of the new ASCO-CAP criteria.


Assuntos
Neoplasias da Mama/metabolismo , Imuno-Histoquímica , Receptor ErbB-2/metabolismo , Biópsia por Agulha , Neoplasias da Mama/patologia , Reações Falso-Negativas , Reações Falso-Positivas , Feminino , Guias como Assunto , Humanos , Hibridização In Situ , Reprodutibilidade dos Testes
15.
J Leukoc Biol ; 81(6): 1599-608, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17347302

RESUMO

The death-associated protein kinase 2 (DAPK2) belongs to a family of Ca(2+)/calmodulin-regulated serine/threonine kinases involved in apoptosis. During investigation of candidate genes operative in granulopoiesis, we identified DAPK2 as highly expressed. Subsequent investigations demonstrated particularly high DAPK2 expression in normal granulocytes compared with monocytes/macrophages and CD34(+) progenitor cells. Moreover, significantly increased DAPK2 mRNA levels were seen when cord blood CD34(+) cells were induced to differentiate toward neutrophils in tissue culture. In addition, all-trans retinoic acid (ATRA)-induced neutrophil differentiation of two leukemic cell lines, NB4 and U937, revealed significantly higher DAPK2 mRNA expression paralleled by protein induction. In contrast, during differentiation of CD34(+) and U937 cells toward monocytes/macrophages, DAPK2 mRNA levels remained low. In primary leukemia, low expression of DAPK2 was seen in acute myeloid leukemia samples, whereas chronic myeloid leukemia samples in chronic phase showed intermediate expression levels. Lentiviral vector-mediated expression of DAPK2 in NB4 cells enhanced, whereas small interfering RNA-mediated DAPK2 knockdown reduced ATRA-induced granulocytic differentiation, as evidenced by morphology and neutrophil stage-specific maturation genes, such as CD11b, G-CSF receptor, C/EBPepsilon, and lactoferrin. In summary, our findings implicate a role for DAPK2 in granulocyte maturation.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/biossíntese , Leucemia Mieloide/patologia , Células Mieloides/citologia , Mielopoese/fisiologia , Neutrófilos/citologia , Doença Aguda , Antígenos CD34/metabolismo , Antígenos de Diferenciação/metabolismo , Proteínas Reguladoras de Apoptose , Diferenciação Celular , Linhagem Celular Tumoral , Doença Crônica , Proteínas Quinases Associadas com Morte Celular , Perfilação da Expressão Gênica , Granulócitos/citologia , Granulócitos/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Leucemia Mieloide/metabolismo , Células Mieloides/fisiologia , Neutrófilos/fisiologia , RNA Interferente Pequeno/biossíntese , Tretinoína/farmacologia , Regulação para Cima
16.
Br J Haematol ; 135(4): 520-3, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17061979

RESUMO

Microarray gene expression profiles of fresh clinical samples of chronic myeloid leukaemia in chronic phase, acute promyelocytic leukaemia and acute monocytic leukaemia were compared with profiles from cell lines representing the corresponding types of leukaemia (K562, NB4, HL60). In a hierarchical clustering analysis, all clinical samples clustered separately from the cell lines, regardless of leukaemic subtype. Gene ontology analysis showed that cell lines chiefly overexpressed genes related to macromolecular metabolism, whereas in clinical samples genes related to the immune response were abundantly expressed. These findings must be taken into consideration when conclusions from cell line-based studies are extrapolated to patients.


Assuntos
Leucemia/genética , Células Tumorais Cultivadas/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica/métodos , Humanos , Leucemia/metabolismo , Leucemia Monocítica Aguda/genética , Leucemia Monocítica Aguda/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mieloide/genética , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Regulação para Cima
17.
Leuk Lymphoma ; 45(6): 1205-7, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15360002

RESUMO

B-cell chronic lymphocytic leukemia is a heterogenous disease with disturbed apoptosis in which the precise molecular defects leading to this pathogenesis are still unclear. The p73 gene (a p53 homologue) encodes 2 proteins with opposing functions. TAp73 induces cell cycle arrest and apoptosis, whilst the oncogenic deltaNp73 inhibits both TAp73 and p53 induced apoptosis. Microsatellite analysis was performed to investigate the p73 gene locus in B-CLL. Moreover, we investigated the expression of the TAp73 and deltaNp73 variant by measuring the mRNA transcripts in 51 B-CLL patients by real-time RT-PCR. And in addition, protein expression was analyzed by Western blotting technique in 20 B-CLL patients. There was no evidence of clonal loss of heterozygosity at 1p36, the p73 gene locus in B-CLL patients. The real time RT-PCR analysis showed that the expression of both p73 gene variants was much higher in leukemic cells compared to controls. In 17/20 (85%) patients deltaNp73 and TAp73 protein were present. The observed increase of expression of the antiapoptotic deltaNp73 variant in neoplastic cells may lead to a functional p53 inactivation. This mechanism might be relevant in malignancies with an intact p53 gene but disturbed apoptosis mechanisms such as in B-CLL.


Assuntos
Biomarcadores Tumorais/genética , Proteínas de Ligação a DNA/genética , Leucemia Linfocítica Crônica de Células B/genética , Proteínas Nucleares/genética , Western Blotting , Genes Supressores de Tumor , Humanos , Leucemia Linfocítica Crônica de Células B/metabolismo , Perda de Heterozigosidade , Repetições de Microssatélites , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Tumoral p73 , Proteínas Supressoras de Tumor , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...