Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Lancet Oncol ; 17(12): e529-e541, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27924751

RESUMO

Neurological complications of cancer and of anticancer treatments can be substantially disabling to patients, especially with classic chemotherapies. As a rare but important complication, targeted therapies might also result in similar unwanted effects, partly because inhibition of VEGF is a common downstream effect. Therapeutic antibodies, such as the CD20-depleting antibody rituximab, and underlying haematological malignancies, can induce long-lasting cellular immunosuppression, predisposing patients to opportunistic CNS infections, such as progressive multifocal leukoencephalopathy, where treatment-induced recovery can result in severe reconstitution of immune inflammatory syndromes of the central nervous system. Immune-related neurological adverse events, particularly from immune-activating checkpoint inhibitors, occur as a result of immune activation, resulting in organ-specific autoimmune-like disease. The prevalence of immune-related neurological adverse events might only be about 1%-a low prevalence compared with toxicities in other organs-but it constitutes new patterns of neurological toxic forms, which could result in considerable morbidity and fatal outcomes. Clinicians should be aware of treatment-associated neurotoxicity, and consider discontinuation of the drug with parallel supportive measures to help patients.


Assuntos
Imunoterapia/efeitos adversos , Terapia de Alvo Molecular/efeitos adversos , Neoplasias/terapia , Doenças do Sistema Nervoso/etiologia , Encefalopatias/etiologia , Doenças Cerebelares/etiologia , Humanos , Leucoencefalopatias/etiologia , Convulsões/etiologia
2.
J Neurooncol ; 129(3): 533-539, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27422128

RESUMO

Bevacizumab is frequently used in patients with progressive glioblastoma raising questions regarding frequency of treatments, dosage, duration of therapy and the possibility of tapering and discontinuation for selected patient groups. We retrospectively assessed the safety and outcome of tapering and discontinuation of bevacizumab therapy for reasons other than disease progression and toxicity in 19 patients with progressive glioblastoma receiving bevacizumab for at least 6 months. In 10 of the 19 patients tapering bevacizumab resulted in complete discontinuation and reinitiation after disease progression during halted treatment. As a comparison group 33 patients with bevacizumab for at least 6 months continuously dosed at 10 mg/kg every 2 weeks were selected. Age and Karnofsky performance status at start of bevacizumab were similar in both groups. Influenced by the selection process, progression-free survival (PFS) and overall survival (OS) were longer in the group receiving a tapered and discontinued bevacizumab regimen (PFS 22.7 versus 11.2 months, HR 0.33, p-value = 0.01; OS 29.9 versus 15.5 months, HR 0.22, p-value = 0.001) with a median time of discontinuation of 4.5 months (range: 1.9-44.2 months). Stable disease or partial response according to RANO at ≥3 months was achieved in 89 % of patients with reinitiated bevacizumab therapy after discontinuation. These data indicate that tapering and discontinuation of bevacizumab therapy for other reasons than progression is feasible without an increased risk for tumor rebound or unresponsiveness to reinitiated bevacizumab therapy.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Bevacizumab/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Adulto , Idoso , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/mortalidade , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Progressão da Doença , Esquema de Medicação , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/mortalidade , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Análise de Regressão , Estudos Retrospectivos , Análise de Sobrevida , Temozolomida , Resultado do Tratamento , Adulto Jovem
3.
Neuro Oncol ; 20(3): 367-379, 2018 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-29016808

RESUMO

Background: O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation status is a predictive biomarker in glioblastoma. We investigated whether this marker furthermore defines a molecularly distinct tumor subtype with clinically different outcome. Methods: We analyzed copy number variation (CNV) and methylation profiles of 1095 primary and 92 progressive isocitrate dehydrogenase wildtype glioblastomas, including paired samples from 49 patients. DNA mutation data from 182 glioblastoma samples of The Cancer Genome Atlas (TCGA) and RNA expression from 107 TCGA and 55 Chinese Glioma Genome Atlas samples were analyzed. Results: Among untreated glioblastomas, MGMT promoter methylated (mMGMT) and unmethylated (uMGMT) tumors did not show different CNV or specific gene mutations, but a higher mutation count in mMGMT tumors. We identified 3 methylation clusters. Cluster 1 showed the highest average methylation and was enriched for mMGMT tumors. Seventeen genes including gastrulation brain homeobox 2 (GBX2) were found to be hypermethylated and downregulated on the mRNA level in mMGMT tumors. In progressive glioblastomas, platelet derived growth factor receptor alpha (PDGFRA) and GLI2 amplifications were enriched in mMGMT tumors. Methylated MGMT tumors gain PDGFRA amplification of PDGFRA, whereas uMGMT tumors with amplified PDGFRA frequently lose this amplification upon progression. Glioblastoma patients surviving <6 months and with mMGMT harbored less frequent epidermal growth factor receptor (EGFR) amplifications, more frequent TP53 mutations, and a higher tumor necrosis factor-nuclear factor-kappaB (TNF-NFκB) pathway activation compared with patients surviving >12 months. Conclusions: MGMT promoter methylation status does not define a molecularly distinct glioblastoma subpopulation among untreated tumors. Progressive mMGMT glioblastomas and mMGMT tumors of patients with short survival tend to have more unfavorable molecular profiles.


Assuntos
Metilação de DNA , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Glioblastoma/genética , Isocitrato Desidrogenase/genética , Mutação , Regiões Promotoras Genéticas , Proteínas Supressoras de Tumor/genética , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Variações do Número de Cópias de DNA , Perfilação da Expressão Gênica , Glioblastoma/patologia , Humanos , Prognóstico , Taxa de Sobrevida
4.
Neuro Oncol ; 18(3): 315-28, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26459812

RESUMO

Molecular targets for the pathological vasculature are the vascular endothelial growth factor (VEGF)/VEGF receptor axis, integrins, angiopoietins, and platelet-derived growth factor receptor (PDGFR), as well as several intracellular or downstream effectors like protein kinase C beta and mammalian target of rapamycin (mTOR). Besides hypoxic damage or tumor cell starvation, preclinical models imply vessel independent tumor regression and suggest differential effects of anti-angiogenic treatments on tumorous and nontumorous precursor cells or the immune system. Despite compelling preclinical data and positive data in other cancers, the outcomes of clinical trials with anti-angiogenic agents in gliomas by and large have been disappointing and include VEGF blockage with bevacizumab, integrin inhibition with cilengitide, VEGF receptor inhibition with sunitinib or cediranib, PDGFR inhibition with imatinib or dasatinib, protein kinase C inhibition with enzastaurin, and mTOR inhibition with sirolimus, everolimus, or temsirolimus. Importantly, there is a lack of real understanding for this negative data. Anti-angiogenic therapies have stimulated the development of standardized imaging assessment and the integration of functional MRI sequences into daily practice. Here, we delineate directions in the identification of molecularly or image-based defined subgroups, anti-angiogenic cotreatment for immunotherapy, and the potential of ongoing trials or modified targets to change the game.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Glioma/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Terapia Combinada/métodos , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Neuro Oncol ; 18(12): 1673-1679, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27298312

RESUMO

BACKGROUND: The purpose of this study was to determine the relevance of clinical data, apparent diffusion coefficient (ADC), and relative cerebral blood volume (rCBV) from dynamic susceptibility contrast (DSC) perfusion and the volume transfer constant (ktrans) from dynamic contrast-enhanced (DCE) perfusion for predicting overall survival (OS) and progression-free survival (PFS) in newly diagnosed treatment-naïve glioblastoma patients. METHODS: Preoperative MR scans including standardized contrast-enhanced T1 (cT1), T2 - fluid-attenuated inversion recovery (FLAIR), ADC, DSC, and DCE of 125 patients with subsequent histopathologically confirmed glioblastoma were performed on a 3 Tesla MRI scanner. ADC, DSC, and DCE parameters were analyzed in semiautomatically segmented tumor volumes on contrast-enhanced (CE) cT1 and hyperintense signal changes on T2 FLAIR (ED). Univariate and multivariable Cox regression analyses including age, sex, extent of resection (EOR), and KPS were performed to assess the influence of each parameter on OS and PFS. RESULTS: Univariate Cox regression analysis demonstrated a significant association of age, KPS, and EOR with PFS and age, KPS, EOR, lower ADC, and higher rCBV with OS. Multivariable analysis showed independent significance of male sex, KPS, EOR, and increased rCBVCE for PFS, and age, sex, KPS, and EOR for OS. CONCLUSIONS: MRI parameters help to predict OS in a univariate Cox regression analysis, and increased rCBVCE is associated with shorter PFS in the multivariable model. In summary, however, our findings suggest that the relevance of MRI parameters is outperformed by clinical parameters in a multivariable analysis, which limits their prognostic value for survival prediction at the time of initial diagnosis.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/mortalidade , Imagem de Difusão por Ressonância Magnética , Glioblastoma/diagnóstico por imagem , Glioblastoma/mortalidade , Angiografia por Ressonância Magnética , Adulto , Idoso , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Volume Sanguíneo Cerebral , Meios de Contraste , Intervalo Livre de Doença , Feminino , Glioblastoma/patologia , Humanos , Aumento da Imagem , Interpretação de Imagem Assistida por Computador , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Análise de Sobrevida
6.
Oncotarget ; 7(35): 56713-56725, 2016 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-27447560

RESUMO

Influencing cancer metabolism by lifestyle changes is an attractive strategy as - if effective - exercise-induced problems may be less severe than those induced by classical anti-cancer therapies. Pursuing this idea, clinical trials evaluated the benefit of e.g. different diets such as the ketogenic diet, intermittent caloric restriction and physical exercise (PE) in the primary and secondary prevention of different cancer types. PE proved to be beneficial in the context of breast and colon cancer.Glioblastoma has a dismal prognosis, with an average overall survival of about one year despite maximal safe resection, concomitant radiochemotherapy with temozolomide followed by adjuvant temozolomide therapy. Here, we focused on the influence of PE as an isolated and adjuvant treatment in murine GB therapy.PE did not reduce toxic side effects of chemotherapy in mice administered in a dose escalating scheme as shown before for starvation. Although regular treadmill training on its own had no obvious beneficial effects, its combination with temozolomide was beneficial in the treatment of glioblastoma-bearing mice. As PE might partly act through the induction of reactive oxygen species, dihydroartemisinin - an approved anti-malarial drug which induces oxidative stress in glioma cells - was further evaluated in vitro and in vivo. Dihydroartemisinin showed anti-glioma activity by promoting autophagy, reduced the clonogenic survival and proliferation capacity of glioma cells, and prolonged the survival of tumor bearing mice. Using the reactive oxygen species scavenger n-acetyl-cysteine these effects were in part reversible, suggesting that dihydroartemisinin partly acts through the generation of reactive oxygen species.


Assuntos
Antimaláricos/administração & dosagem , Artemisininas/administração & dosagem , Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Glioblastoma/terapia , Estresse Oxidativo , Condicionamento Físico Animal , Animais , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Terapia Combinada , Dacarbazina/administração & dosagem , Progressão da Doença , Feminino , Glioblastoma/tratamento farmacológico , Humanos , Camundongos , Camundongos Nus , Espécies Reativas de Oxigênio/metabolismo , Temozolomida
7.
Oncotarget ; 7(50): 82028-82045, 2016 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-27880937

RESUMO

The outcome of patients with anaplastic gliomas varies considerably depending on single molecular markers, such as mutations of the isocitrate dehydrogenase (IDH) genes, as well as molecular classifications based on epigenetic or genetic profiles. Remarkably, 98% of the RNA within a cell is not translated into proteins. Of those, especially microRNAs (miRNAs) have been shown not only to have a major influence on physiologic processes but also to be deregulated and prognostic in malignancies.To find novel survival markers and treatment options we performed unbiased DNA methylation screens that revealed 12 putative miRNA promoter regions with differential DNA methylation in anaplastic gliomas. Methylation of these candidate regions was validated in different independent patient cohorts revealing a set of miRNA promoter regions with prognostic relevance across data sets. Of those, miR-155 promoter methylation and miR-155 expression were negatively correlated and especially the methylation showed superior correlation with patient survival compared to established biomarkers.Functional examinations in malignant glioma cells further cemented the relevance of miR-155 for tumor cell viability with transient and stable modifications indicating an onco-miRNA activity. MiR-155 also conferred resistance towards alkylating temozolomide and radiotherapy as consequence of nuclear factor (NF)κB activation.Preconditioning glioma cells with an NFκB inhibitor reduced therapy resistance of miR-155 overexpressing cells. These cells resembled tumors with a low methylation of the miR-155 promoter and thus mir-155 or NFκB inhibition may provide treatment options with a special focus on patients with IDH wild type tumors.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Metilação de DNA , Epigênese Genética , Glioma/genética , MicroRNAs/genética , Antineoplásicos Alquilantes/farmacologia , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Glioma/mortalidade , Glioma/patologia , Glioma/terapia , Humanos , Estimativa de Kaplan-Meier , MicroRNAs/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Fenótipo , Prognóstico , Regiões Promotoras Genéticas , Modelos de Riscos Proporcionais , Interferência de RNA , Tolerância a Radiação/genética , Temozolomida , Fatores de Tempo , Transcrição Gênica , Transfecção
8.
Oncotarget ; 6(31): 31050-68, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-25682871

RESUMO

Loss of the tumor suppressor phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a prerequisite for tumor cell-specific expression of vascular endothelial growth factor receptor (VEGFR)-2 in glioblastoma defining a subgroup prone to develop evasive resistance towards antiangiogenic treatments. Immunohistochemical analysis of human tumor tissues showed VEGFR-2 expression in glioma cells in 19% of specimens examined, mainly in the infiltration zone. Glioma cell VEGFR-2 positivity was restricted to PTEN-deficient tumor specimens. PTEN overexpression reduced VEGFR-2 expression in vitro, as well as knock-down of raptor or rictor. Genetic interference with VEGFR-2 revealed proproliferative, antiinvasive and chemoprotective functions for VEGFR-2 in glioma cells. VEGFR-2-dependent cellular effects were concomitant with activation of 'kappa-light-chain-enhancer' of activated B-cells, protein kinase B, and N-myc downstream regulated gene 1. Two-photon in vivo microscopy revealed that expression of VEGFR-2 in glioma cells hampers antiangiogenesis. Bevacizumab induces a proinvasive response in VEGFR-2-positive glioma cells. Patients with PTEN-negative glioblastomas had a shorter survival after initiation of bevacizumab therapy compared with PTEN-positive glioblastomas. Conclusively, expression of VEGFR-2 in glioma cells indicates an aggressive glioblastoma subgroup developing early resistance to temozolomide or bevacizumab. Loss of PTEN may serve as a biomarker identifying those tumors upfront by routine neuropathological methods.


Assuntos
Inibidores da Angiogênese/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Glioma/tratamento farmacológico , Neovascularização Patológica , PTEN Fosfo-Hidrolase/deficiência , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Bevacizumab/farmacologia , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioma/enzimologia , Glioma/genética , Glioma/mortalidade , Glioma/patologia , Humanos , Estimativa de Kaplan-Meier , Camundongos Nus , Invasividade Neoplásica , PTEN Fosfo-Hidrolase/genética , Transdução de Sinais/efeitos dos fármacos , Temozolomida , Fatores de Tempo , Transfecção , Carga Tumoral/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Biochem Pharmacol ; 82(6): 632-41, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21703247

RESUMO

3,4-dimethoxycinnamonyl-anthranilic acid (tranilast) is an orally available anti-allergic drug with structural and functional homologies to immunosuppressive catabolites of the essential amino acid tryptophan and broad anti-inflammatory properties. It has recently been shown to be effective in animal models of multiple sclerosis and rheumatoid arthritis, two autoimmune diseases that are mediated by auto-aggressive Th1-polarized CD4+ T lymphocytes. Here we demonstrate potent suppressive effects of tranilast on the function of naïve human CD4+ T cells. Tranilast inhibited inhibits activation and proliferation of purified CD4+ T cells stimulated through the T cell receptor with an EC50 of less than 10 µM, a concentration that is well below plasma levels achieved after oral administration of approved doses of 200-600 mg in humans. The antiproliferative effects were less potent on naïve CD8+ T cells. Suppression of CD4+ and CD8+ T cell proliferation was associated with an inhibition of T cell activation. Cytokine analyses of naïve CD4+ T cells revealed that tranilast interferes with the production of cyto- and chemokines driven by signal transducer and activator of transcription 1 (STAT1), notably chemokine (C-X-C motif) ligands (CXCL) 9 and 10. Tranilast limited STAT1 phosphorylation in activated T cells and supplementation of CXCL9 or CXCL10 reversed the anti-proliferative effects of tranilast. These data imply CXCL9 and CXCL10 as novel therapeutic targets of tranilast in Th1-mediated autoimmune diseases and identify phospho-STAT1 and its target chemokines CXCL9 and CXCL10 as potential markers for monitoring the bioactivity of tranilast in humans.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Quimiocina CXCL10/imunologia , Quimiocina CXCL9/imunologia , Ativação Linfocitária/efeitos dos fármacos , ortoaminobenzoatos/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Western Blotting , Linfócitos T CD4-Positivos/imunologia , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/metabolismo , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Células Jurkat , Ativação Linfocitária/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA