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











Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 39(33): 5553-5569, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32655131

RESUMO

Metastasis is the leading cause of cancer-related death. This multistage process involves contribution from both tumour cells and the tumour stroma to release metastatic cells into the circulation. Circulating tumour cells (CTCs) survive circulatory cytotoxicity, extravasate and colonise secondary sites effecting metastatic outcome. Reprogramming the transcriptomic landscape is a metastatic hallmark, but detecting underlying master regulators that drive pathological gene expression is a key challenge, especially in childhood cancer. Here we used whole tumour plus single-cell RNA-sequencing in primary bone cancer and CTCs to perform weighted gene co-expression network analysis to systematically detect coordinated changes in metastatic transcript expression. This approach with comparisons applied to data collected from cell line models, clinical samples and xenograft mouse models revealed mitogen-activated protein kinase 7/matrix metallopeptidase 9 (MAPK7/MMP9) signalling as a driver for primary bone cancer metastasis. RNA interference knockdown of MAPK7 reduces proliferation, colony formation, migration, tumour growth, macrophage residency/polarisation and lung metastasis. Parallel to these observations were reduction of activated interleukins IL1B, IL6, IL8 plus mesenchymal markers VIM and VEGF in response to MAPK7 loss. Our results implicate a newly discovered, multidimensional MAPK7/MMP9 signalling hub in primary bone cancer metastasis that is clinically actionable.


Assuntos
Neoplasias Ósseas/complicações , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Animais , Neoplasias Ósseas/genética , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Metástase Neoplásica
2.
Cell Rep ; 18(2): 482-495, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28076791

RESUMO

Understanding the underlying molecular mechanisms of defined cancers is crucial for effective personalized therapies. Translocations of the mixed-lineage leukemia (MLL) gene produce fusion proteins such as MLL-AF4 that disrupt epigenetic pathways and cause poor-prognosis leukemias. Here, we find that at a subset of gene targets, MLL-AF4 binding spreads into the gene body and is associated with the spreading of Menin binding, increased transcription, increased H3K79 methylation (H3K79me2/3), a disruption of normal H3K36me3 patterns, and unmethylated CpG regions in the gene body. Compared to other H3K79me2/3 marked genes, MLL-AF4 spreading gene expression is downregulated by inhibitors of the H3K79 methyltransferase DOT1L. This sensitivity mediates synergistic interactions with additional targeted drug treatments. Therefore, epigenetic spreading and enhanced susceptibility to epidrugs provides a potential marker for better understanding combination therapies in humans.


Assuntos
Elementos Facilitadores Genéticos/genética , Leucemia/genética , Leucemia/patologia , Metiltransferases/antagonistas & inibidores , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Ilhas de CpG/genética , Metilação de DNA/genética , Regulação Leucêmica da Expressão Gênica , Genoma Humano , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lisina/metabolismo , Metiltransferases/metabolismo , Prognóstico , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo
3.
Blood ; 124(23): 3420-30, 2014 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-25253770

RESUMO

For most children who relapse with acute lymphoblastic leukemia (ALL), the prognosis is poor, and there is a need for novel therapies to improve outcome. We screened samples from children with B-lineage ALL entered into the ALL-REZ BFM 2002 clinical trial (www.clinicaltrials.gov, #NCT00114348) for somatic mutations activating the Ras pathway (KRAS, NRAS, FLT3, and PTPN11) and showed mutation to be highly prevalent (76 from 206). Clinically, they were associated with high-risk features including early relapse, central nervous system (CNS) involvement, and specifically for NRAS/KRAS mutations, chemoresistance. KRAS mutations were associated with a reduced overall survival. Mutation screening of the matched diagnostic samples found many to be wild type (WT); however, by using more sensitive allelic-specific assays, low-level mutated subpopulations were found in many cases, suggesting that they survived up-front therapy and subsequently emerged at relapse. Preclinical evaluation of the mitogen-activated protein kinase kinase 1/2 inhibitor selumetinib (AZD6244, ARRY-142886) showed significant differential sensitivity in Ras pathway-mutated ALL compared with WT cells both in vitro and in an orthotopic xenograft model engrafted with primary ALL; in the latter, reduced RAS-mutated CNS leukemia. Given these data, clinical evaluation of selumetinib may be warranted for Ras pathway-mutated relapsed ALL.


Assuntos
Benzimidazóis/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Genes ras , Mutação , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Linhagem Celular Tumoral , Criança , Ensaios Clínicos como Assunto , Frequência do Gene , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/epidemiologia , Recidiva , Transdução de Sinais/genética , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Clin Exp Metastasis ; 31(6): 651-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24859418

RESUMO

COX2 is an inducible cyclooxygenase implicated in the metastasis and migration of tumour cells. In neuroblastoma, COX2 expression has been detected in both cell lines and tumours. The treatment of neuroblastoma cells in vitro with celecoxib, a COX2 inhibitor, induces apoptosis. The aim of this study was to investigate the role of COX2 in neuroblastoma tumour biology by creating a cell line in which COX2 could be conditionally expressed. Xenograft studies showed that the conditional expression of COX2 enhanced tumour growth and malignancy. Elevated COX2 expression enhanced the proliferation and migration of neuroblastoma cells in vitro. However, elevated COX2 expression or variation between cell lines did not affect sensitivity to the COX2 inhibitor celecoxib, indicating that celecoxib does not promote cell death through COX2 inhibition. These data show that increased COX2 expression alone can enhance the tumorigenic properties of neuroblastoma cells; however, high levels of COX2 may not be a valid biomarker of sensitivity to non-steroidal anti-inflammatory drugs such as celecoxib.


Assuntos
Morte Celular , Inibidores de Ciclo-Oxigenase 2/farmacologia , Ciclo-Oxigenase 2/metabolismo , Neuroblastoma/enzimologia , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Animais , Celecoxib , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Nus , Neuroblastoma/patologia
5.
PLoS One ; 8(7): e68859, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874790

RESUMO

Retinoic acid (RA) has paradoxical effects on cancer cells: promoting cell death, differentiation and cell cycle arrest, or cell survival and proliferation. Arachidonic acid (AA) release occurs in response to RA treatment and, therefore, AA and its downstream metabolites may be involved in cell survival signalling. To test this, we inhibited phospholipase A2-mediated AA release, cyclooxygenases and lipoxygenases with small-molecule inhibitors to determine if this would sensitise cells to cell death after RA treatment. The data suggest that, in response to RA, phospholipase A2-mediated release of AA and subsequent metabolism by lipoxygenases is important for cell survival. Evidence from gene expression reporter assays and PPARδ knockdown suggests that lipoxygenase metabolites activate PPARδ. The involvement of PPARδ in cell survival is supported by results of experiments with the PPARδ inhibitor GSK0660 and siRNA-mediated knockdown. Quantitative reverse transcriptase PCR studies demonstrated that inhibition of 5-lipoxygenase after RA treatment resulted in a strong up-regulation of mRNA for PPARδ2, a putative inhibitory PPARδ isoform. Over-expression of PPARδ2 using a tetracycline-inducible system in neuroblastoma cells reduced proliferation and induced cell death. These data provide evidence linking lipoxygenases and PPARδ in a cell survival-signalling mechanism and suggest new drug-development targets for malignant and hyper-proliferative diseases.


Assuntos
Ácido Araquidônico/metabolismo , Neuroblastoma/metabolismo , PPAR delta/metabolismo , Transdução de Sinais , Araquidonato 5-Lipoxigenase/metabolismo , Ácido Araquidônico/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Ativação Enzimática/efeitos dos fármacos , Expressão Gênica , Humanos , Isoenzimas , Neuroblastoma/genética , PPAR delta/genética , Transdução de Sinais/efeitos dos fármacos
6.
Int J Cancer ; 128(7): 1703-11, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20499314

RESUMO

Epidemiological and preclinical studies have revealed that omega-3 fatty acids have anticancer properties. We have previously shown that the omega-3 fatty acid docosahexaenoic acid (DHA) induces apoptosis of neuroblastoma cells in vitro by mechanisms involving intracellular peroxidation of DHA by means of 15-lipoxygenase or autoxidation. In our study, the effects of DHA supplementation on neuroblastoma tumor growth in vivo were investigated using two complementary approaches. For the purpose of prevention, DHA as a dietary supplement was fed to athymic rats before the rats were xenografted with human neuroblastoma cells. For therapeutic purposes, athymic rats with established neuroblastoma xenografts were given DHA daily by gavage and tumor growth was monitored. DHA levels in plasma and tumor tissue were analyzed by gas liquid chromatography. DHA delayed neuroblastoma xenograft development and inhibited the growth of established neuroblastoma xenografts in athymic rats. A revised version of the Pediatric Preclinical Testing Program evaluation scheme used as a measurement of treatment response showed that untreated control animals developed progressive disease, whereas treatment with DHA resulted in stable disease or partial response, depending on the DHA concentration. In conclusion, prophylactic treatment with DHA delayed neuroblastoma development, suggesting that DHA could be a potential agent in the treatment of minimal residual disease and should be considered for prevention in selected cases. Treatment results on established aggressive neuroblastoma tumors suggest further studies aiming at a clinical application in children with high-risk neuroblastoma.


Assuntos
Ácidos Graxos Ômega-3/metabolismo , Neuroblastoma/metabolismo , Administração Oral , Animais , Linhagem Celular Tumoral , Cromatografia Gasosa , Suplementos Nutricionais , Progressão da Doença , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Cromatografia Gasosa-Espectrometria de Massas/métodos , Humanos , Cinética , Transplante de Neoplasias , Ratos , Ratos Nus
7.
Proc Natl Acad Sci U S A ; 106(29): 12043-8, 2009 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-19581587

RESUMO

The t(8:21)(q22;q22) translocation is 1 of the most common chromosomal abnormalities linked to acute myeloid leukemia (AML). AML1-ETO, the product of this translocation, fuses the N-terminal portion of the RUNX transcription factor AML1 (also known as RUNX1), including its DNA-binding domain, to the almost entire transcriptional corepressor ETO (also known as MTG8 or RUNX1T1). This fusion protein acts primarily by interfering with endogenous AML1 function during myeloid differentiation, although relatively few genes are known that participate with AML1-ETO during leukemia progression. Here, we assessed the consequences of expressing this chimera in Drosophila blood cells. Reminiscent of what is observed in AML, AML1-ETO specifically inhibited the differentiation of the blood cell lineage whose development depends on the RUNX factor Lozenge (LZ) and induced increased numbers of LZ(+) progenitors. Using an in vivo RNAi-based screen for suppressors of AML1-ETO, we identified calpainB as required for AML1-ETO-induced blood cell disorders in Drosophila. Remarkably, calpain inhibition triggered AML1-ETO degradation and impaired the clonogenic potential of the human t(8;21) leukemic blood cell line Kasumi-1. Therefore Drosophila provides a promising genetically tractable model to investigate the conserved basis of leukemogenesis and to open avenues in AML therapy.


Assuntos
Calpaína/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Animais , Células Sanguíneas/citologia , Calpaína/antagonistas & inibidores , Contagem de Células , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Ensaio de Unidades Formadoras de Colônias , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/antagonistas & inibidores , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Genes Supressores , Testes Genéticos , Humanos , Modelos Animais , Proteína 1 Parceira de Translocação de RUNX1 , Células-Tronco/citologia , Fatores de Transcrição/metabolismo
8.
FASEB J ; 22(10): 3525-36, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18591367

RESUMO

The metabolism of arachidonic acid by the cyclooxygenase (COX) or lipoxygenase (LO) pathways generates eicosanoids that have been implicated in the pathogenesis of a variety of human diseases, including cancer. In this study, we examined the expression and significance of components within the 5-LO pathway in human neuroblastoma, an embryonal tumor of the sympathetic nervous system. High expression of 5-LO, 5-LO-activating protein (FLAP), leukotriene A(4) hydrolase, leukotriene C(4) synthase, and leukotriene receptors was detected in a majority of primary neuroblastoma tumors and all cell lines investigated. Expression of 5-LO and FLAP was evident in tumor cells but not in nonmalignant adrenal medulla where neuroblastomas typically arise. Moreover, neuroblastoma cells produce leukotrienes, and stimulation of neuroblastoma cells with leukotrienes increased neuroblastoma cell viability. Inhibitors of 5-LO (AA-861), FLAP (MK-886), or the leukotriene receptor antagonist montelukast inhibited neuroblastoma cell growth by induction of G(1)-cell cycle arrest and apoptosis. Similarly, specific 5-LO and leukotriene receptor silencing by small interfering RNA decreased neuroblastoma cell growth. These findings provide new insights into the pathobiology of neuroblastoma, and the use of leukotriene pathway inhibitors as a novel adjuvant therapy for children with neuroblastoma warrants further consideration.


Assuntos
Leucotrienos/biossíntese , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Proteínas Ativadoras de 5-Lipoxigenase , Apoptose , Araquidonato 5-Lipoxigenase/biossíntese , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/biossíntese , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Epóxido Hidrolases/biossíntese , Glutationa Transferase/biossíntese , Humanos , Antagonistas de Leucotrienos/farmacologia , Inibidores de Lipoxigenase , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/biossíntese , Neuroblastoma/tratamento farmacológico , Receptores de Leucotrienos/biossíntese , Receptores de Leucotrienos/efeitos dos fármacos
9.
Mol Cancer Ther ; 6(9): 2409-17, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17876040

RESUMO

Neuroblastoma is the most common extracranial solid tumor of childhood. The activity of J1 (l-melphalanyl-p-l-fluorophenylalanine ethyl ester), an enzymatically activated melphalan prodrug, was evaluated in neuroblastoma models in vitro and in vivo. Seven neuroblastoma cell lines with various levels of drug resistance were screened for cytotoxicity of J1 alone or in combination with standard cytotoxic drugs, using a fluorometric cytotoxicity assay. J1 displayed high cytotoxic activity in vitro against all neuroblastoma cell lines, with IC(50) values in the submicromolar range, significantly more potent than melphalan. The cytotoxicity of J1, but not melphalan, could be significantly inhibited by the aminopeptidase inhibitor bestatin. J1 induced caspase-3 cleavage and apoptotic morphology, had additive effects in combination with doxorubicin, cyclophosphamide, carboplatin, and vincristine, and synergistically killed otherwise drug-resistant cells when combined with etoposide. Athymic rats and mice carrying neuroblastoma xenografts [SH-SY5Y, SK-N-BE(2)] were treated with equimolar doses of melphalan, J1, or no drug, and effects on tumor growth and tissue morphology were analyzed. Tumor growth in vivo was significantly inhibited by J1 compared with untreated controls. Compared with melphalan, J1 more effectively inhibited the growth of mice with SH-SY5Y xenografts, was associated with higher caspase-3 activation, fewer proliferating tumor cells, and significantly decreased mean vascular density. In conclusion, the melphalan prodrug J1 is highly active in models of neuroblastoma in vitro and in vivo, encouraging further clinical development in this patient group.


Assuntos
Dipeptídeos/farmacologia , Neuroblastoma/tratamento farmacológico , Pró-Fármacos/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Carboplatina/administração & dosagem , Linhagem Celular Tumoral/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxorrubicina/administração & dosagem , Ensaios de Seleção de Medicamentos Antitumorais , Etoposídeo/administração & dosagem , Feminino , Humanos , Técnicas Imunoenzimáticas , Masculino , Melfalan/administração & dosagem , Camundongos , Estrutura Molecular , Neovascularização Patológica , Neuroblastoma/irrigação sanguínea , Neuroblastoma/metabolismo , Ratos , Ratos Nus , Células Tumorais Cultivadas , Vincristina/administração & dosagem
10.
Clin Cancer Res ; 13(3): 1036-44, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17289900

RESUMO

PURPOSE: Neuroblastoma is the most common and deadly solid tumor of childhood. Cyclooxygenase-2 is expressed in clinical neuroblastoma tumors and cell lines and inhibitors of this enzyme induce apoptosis in human neuroblastoma cells in vitro and in neuroblastoma xenografts in vivo. We hypothesized that the cyclooxygenase-2-specific inhibitor celecoxib could enhance the cytotoxic effect of chemotherapeutic drugs currently used in neuroblastoma treatment. Furthermore, we investigated if prophylactic treatment with celecoxib could prevent neuroblastoma tumor development in vivo. EXPERIMENTAL DESIGN: Neuroblastoma cell cytotoxicity of chemotherapeutic drugs in combination with celecoxib was examined. In vivo, athymic rats carrying established SH-SY5Y xenografts were treated with celecoxib in combination with irinotecan, doxorubicin or etoposide, or with either drug alone. For prevention studies, rats received celecoxib in the diet, 250 to 2,500 ppm, from the time of tumor cell injection. RESULTS: Celecoxib induced a synergistic or an additive cytotoxic effect in combination with doxorubicin, etoposide, irinotecan or vincristine in vitro. In vivo, treatment with celecoxib in combination with irinotecan or doxorubicin induced a significant growth inhibition of established neuroblastoma tumors. Rats receiving celecoxib in the diet showed a distinct dose-dependent delay in tumor development compared with untreated rats. Plasma levels of celecoxib were comparable with levels obtainable in humans. CONCLUSIONS: Celecoxib potentiates the antitumor effect of chemotherapeutic drugs currently used in neuroblastoma treatment, which argues for clinical trials combining these drugs. Celecoxib could also be a potential drug for treatment of minimal residual disease.


Assuntos
Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ciclo-Oxigenase 2/biossíntese , Neuroblastoma/tratamento farmacológico , Pirazóis/uso terapêutico , Sulfonamidas/uso terapêutico , Animais , Apoptose , Celecoxib , Linhagem Celular Tumoral , Inibidores de Ciclo-Oxigenase/farmacologia , Humanos , Técnicas In Vitro , Antígeno Ki-67/biossíntese , Masculino , Transplante de Neoplasias , Neuroblastoma/metabolismo , Ratos , Ratos Nus
11.
Int J Cancer ; 119(3): 493-500, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16572423

RESUMO

Antimicrobial peptides have been shown to exert cytotoxic activity towards cancer cells through their ability to interact with negatively charged cell membranes. In this study the cytotoxic effect of the antimicrobial peptide, LfcinB was tested in a panel of human neuroblastoma cell lines. LfcinB displayed a selective cytotoxic activity against both MYCN-amplified and non-MYCN-amplified cell lines. Non-transformed fibroblasts were not substantially affected by LfcinB. Treatment of neuroblastoma cells with LfcinB induced rapid destabilization of the cytoplasmic membrane and formation of membrane blebs. Depolarization of the mitochondria membranes and irreversible changes in the mitochondria morphology was also evident. Immuno- and fluorescence-labeled LfcinB revealed that the peptide co-localized with mitochondria. Furthermore, treatment of neuroblastoma cells with LfcinB induced cleavage of caspase-6, -7 and -9 followed by cell death. However, neither addition of the pan-caspase inhibitor, zVAD-fmk, or specific caspase inhibitors could reverse the cytotoxic effect induced by LfcinB. Treatment of established SH-SY-5Y neuroblastoma xenografts with repeated injections of LfcinB resulted in significant tumor growth inhibition. These results revealed a selective destabilizing effect of LfcinB on two important targets in the neuroblastoma cells, the cytoplasmic- and the mitochondria membrane.


Assuntos
Anti-Infecciosos/farmacologia , Lactoferrina/farmacologia , Neuroblastoma/tratamento farmacológico , Animais , Anti-Infecciosos/farmacocinética , Western Blotting , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Lactoferrina/farmacocinética , Potenciais da Membrana/efeitos dos fármacos , Microscopia Eletrônica de Transmissão , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/fisiologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Int J Cancer ; 118(10): 2584-93, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16353135

RESUMO

Docosahexaenoic acid (DHA) is an omega-3 polyunsaturated fatty acid vital for the developing nervous system and significantly decreased in neuroblastoma cells compared to nontransformed nervous tissue. We investigated whether supplementation of DHA affects the susceptibility of neuroblastoma cells to oxidative stress generated endogenously and in response to cytotoxic therapy. DHA, but not the monounsaturated oleic acid (OA), induced dose- and time-dependent neuroblastoma cell death. DHA supplementation was associated with depolarization of the mitochondrial membrane potential, production of reactive oxygen species (ROS) and accumulation of DNA in sub-G1 phase of the cell cycle. The antioxidant, vitamin E, inhibited mitochondrial depolarization and subsequent cell death induced by DHA, whereas, the mitochondrial pore inhibitor, cyclosporin A, partly inhibited DHA-induced neuroblastoma cell death. Depletion of glutathione by L-buthionine-sulfoximine significantly enhanced the cytotoxic effects of DHA. Nontransformed fibroblasts were not substantially affected by DHA. DHA, but not OA, significantly enhanced the cytotoxicity of cisplatin, doxorubicin and irinotecan both in chemosensitive and in multidrug-resistant neuroblastoma cells. DHA potently sensitized neuroblastoma cells to a clinically relevant concentration (1 microM) of arsenic trioxide (As2O3) and enhanced the effect of the nonsteroidal antiinflammatory drug (NSAID), diclofenac. These findings provide experimental evidence that the omega-3 fatty acid, DHA, is cytotoxic to drug-resistant neuroblastoma. The potent action of DHA with arsenic trioxide, NSAID and chemotherapeutic agents suggests clinical testing of this therapeutic concept in children with neuroblastoma.


Assuntos
Arsênio/toxicidade , Ácidos Docosa-Hexaenoicos/farmacologia , Resistência a Múltiplos Medicamentos , Neuroblastoma/patologia , Estresse Oxidativo , Antioxidantes/farmacologia , Apoptose , Relação Dose-Resposta a Droga , Fase G1 , Humanos , Células Tumorais Cultivadas , Vitamina E/farmacologia
13.
Cancer Lett ; 228(1-2): 195-201, 2005 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-15975708

RESUMO

Cyclooxygenases (COX) catalyse the conversion of arachidonic acid to prostaglandins. COX-2 is upregulated in several adult epithelial cancers. In neuroblastoma it has been shown that the majority of primary tumours and cell lines express high levels of COX-2, whereas normal adrenal medullas from children do not express COX-2. Treatment of neuroblastoma cells with nonsteroidal anti-inflammatory drugs (NSAIDs), inhibitors of COX, induces caspase-dependent apoptosis via the intrinsic mitochondrial pathway. Established neuroblastoma xenografts in nude rats treated with the dual COX-1/COX-2 inhibitor, diclofenac, or the COX-2 specific inhibitor, celecoxib significantly inhibits neuroblastoma growth in vivo. In vitro, arachidonic acid and diclofenac synergistically induces neuroblastoma cell death. This effect is further pronounced when lipoxygenases is inhibited simultaneously. Proton MR-spectroscopy (1H MRS) of neuroblastoma cells treated with COX-inhibitors demonstrates accumulation of polyunsaturated fatty acids and depletion of choline compounds. Thus, 1H MRS, which can be performed with clinical MR-scanners, is likely to provide pharmacodynamic markers of neuroblastoma response to COX-inhibition. Taken together, these data suggest the use of NSAIDs as a novel adjuvant therapy for children with neuroblastoma.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Neuroblastoma/tratamento farmacológico , Animais , Apoptose , Inibidores de Ciclo-Oxigenase/uso terapêutico , Espectroscopia de Ressonância Magnética , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Ratos , Ratos Nus
14.
Cancer Lett ; 228(1-2): 247-55, 2005 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-15946794

RESUMO

Non-invasive biological information about residual neuroblastoma tumour tissue could allow treatment monitoring without the need for repeated biopsies. Magnetic resonance spectroscopy (MRS) can be performed with standard MR-scanners, providing specific biochemical information from selected tumour regions. By proton 1H-MRS, lipids, certain amino acids and lactate can be detected and their relative concentrations estimated in vivo. Using experimental models of neuroblastoma, we have described the potential of 1H-MRS for the prediction of tumour tissue viability and treatment response. Whereas viable neuroblastoma tissue is dominated by the choline 1H-MRS resonance, cell death as a consequence of spontaneous necrosis or successful treatment with chemotherapy, angiogenesis inhibitors, or NSAIDs is associated with decreased choline content. Therapy-induced neuroblastoma cell death is also associated with enhanced 1H-MRS resonances from mobile lipids and polyunsaturated fatty acids. The mobile lipid/choline ratio correlates significantly with cell death and based on the dynamics of this ratio tumour regression or continued growth (drug resistance) after chemotherapy can be predicted in vivo. The implications of these findings are discussed with focus on the potentials and limitations of introducing 1H-MRS for clinical assessment of treatment response in children with neuroblastoma. Biochemical monitoring of neuroblastoma with 1H-MRS could enable tailoring of individual therapy as well as provide early pharmacodynamic evaluation of novel therapeutic modalities.


Assuntos
Espectroscopia de Ressonância Magnética , Neuroblastoma/patologia , Humanos , Neuroblastoma/terapia , Prótons , Resultado do Tratamento
15.
Int J Oncol ; 25(6): 1849-57, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15547726

RESUMO

The majority of high-risk neuroblastomas lack the expression of caspase-8 due to gene silencing which suggest a mechanism for the selection of tumour cells that are refractory to multiple cytotoxic drugs including tumour necrosis factor-related apoptosis-inducing ligand (TRAIL). Inhibitors of DNA methyltransferases and IFN-gamma induce expression of caspase-8, and sensitise some neuroblastoma cells to TRAIL-mediated apoptosis. Here we demonstrate that a combination of cytostatic drugs with IFN-gamma and TRAIL synergistically induces neuroblastoma cell death, which may have implications for future therapy of children with neuroblastoma. Treatment of neuroblastoma cells with IFN-gamma induced caspase-8 expression in all cell lines investigated. In five of the neuroblastoma cell lines (SHEP-1, SK-N-AS, SK-N-FI, SH-SY-5Y and Kelly), IFN-gamma promoted TRAIL-mediated cleavage of caspase-8, initiating a caspase cascade involving caspase-7 and PARP followed by apoptosis. IFN-gamma-mediated facilitation of apoptosis was inhibited by the pan-caspase inhibitor zVAD-fmk and the caspase-8 specific inhibitor zIEDT-fmk, indicating an important role of caspase-8 in mediating sensitation by IFN-gamma in neuroblastoma cells. In three of the cell lines [SK-N-BE(2), SK-N-DZ and IMR-32] caspase-8 expression was induced by IFN-gamma, but the cells were still resistant to TRAIL-mediated apoptosis. The pattern of basal TRAIL receptor expression, decoy receptors, FLIP and FADD could not be correlated with resistance or sensitivity to TRAIL-induced apoptosis. Importantly, treatment of neuroblastoma cell lines with cytostatic drugs increased apoptosis in the TRAIL-sensitive cell lines whereas the resistant cell lines were susceptible to TRAIL-mediated apoptosis in the presence of the anticancer drugs. The mechanism of the increased susceptibility to apoptosis might results from drug-mediated up-regulation of the death receptors DR4 and DR5.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspases/farmacologia , Interferon-alfa/farmacologia , Glicoproteínas de Membrana/farmacologia , Neuroblastoma/patologia , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Reguladoras de Apoptose , Caspase 8 , Inibidores de Caspase , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Humanos , Ligantes , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Regulação para Cima
16.
Cancer Res ; 64(20): 7210-5, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15492235

RESUMO

Neuroblastoma is the single most common and deadly tumor of childhood and is often associated with therapy resistance. Cyclooxygenases (COXs) catalyze the conversion of arachidonic acid to prostaglandins. COX-2 is up-regulated in several adult epithelial cancers and is linked to proliferation and resistance to apoptosis. We detected COX-2 expression in neuroblastoma primary tumors and cell lines but not in normal adrenal medullas from children. Treatment of neuroblastoma cells with nonsteroidal anti-inflammatory drugs, inhibitors of COX, induced caspase-dependent apoptosis via the intrinsic mitochondrial pathway. Treatment of established neuroblastoma xenografts in nude rats with the dual COX-1/COX-2 inhibitor diclofenac or the COX-2-specific inhibitor celecoxib significantly inhibited tumor growth in vivo (P < 0.001). In vitro, arachidonic acid and diclofenac synergistically induced neuroblastoma cell death. This effect was further pronounced when lipooxygenases were simultaneously inhibited. Proton magnetic resonance spectroscopy ((1)H MRS) of neuroblastoma cells treated with COX inhibitors demonstrated accumulation of polyunsaturated fatty acids and depletion of choline compounds. Thus, (1)H MRS, which can be performed with clinical magnetic resonance scanners, is likely to provide pharmacodynamic markers of neuroblastoma response to COX inhibition. Taken together, these data suggest the use of nonsteroidal anti-inflammatory drugs as a novel adjuvant therapy for children with neuroblastoma.


Assuntos
Neoplasias das Glândulas Suprarrenais/tratamento farmacológico , Neoplasias das Glândulas Suprarrenais/enzimologia , Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/efeitos dos fármacos , Isoenzimas/biossíntese , Neuroblastoma/tratamento farmacológico , Neuroblastoma/enzimologia , Prostaglandina-Endoperóxido Sintases/biossíntese , Neoplasias das Glândulas Suprarrenais/patologia , Medula Suprarrenal/enzimologia , Medula Suprarrenal/patologia , Animais , Celecoxib , Linhagem Celular Tumoral , Criança , Pré-Escolar , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Diclofenaco/farmacologia , Feminino , Humanos , Lactente , Recém-Nascido , Isoenzimas/antagonistas & inibidores , Masculino , Proteínas de Membrana , Neuroblastoma/patologia , Pirazóis , Ratos , Ratos Nus , Sulfonamidas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Oncol Rep ; 10(5): 1587-92, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12883745

RESUMO

Neuroblastoma, the most common extracranial solid tumour in children, may undergo spontaneous differentiation or regression, but the majority of metastatic neuroblastomas have poor prognosis despite intensive treatment. Retinoic acid and its analogues regulate growth and differentiation of neuroblastoma cells in vitro, and 13-cis retinoic acid has shown activity against human neuroblastomas in vivo. Fenretinide [N-(4-hydroxyphenyl)retinamide] has been identified as a synthetic retinoid able to induce apoptosis of numerous malignant cell lines in vitro, including neuroblastoma. Furthermore, in animal models, fenretinide has shown chemopreventive and therapeutic efficacy against several malignancies without any obvious signs of toxicity. To investigate the anti-neuroblastoma tumour growth effects of oral fenretinide in vivo we used a human neuroblastoma xenograft model. Nude rats with established neuroblastoma xenograft tumours were treated orally with fenretinide for 10 days. Five different doses of fenretinide were used ranging from 2.5 to 75 mg/rat/day (10-300 mg/kg). Tumour volumes and toxic side effects were monitored during treatment and tumour weights were recorded at autopsy. In this study we found no significant anti-tumour growth effects of fenretinide in vivo, when used as oral treatment of rats with established neuroblastoma xenograft tumours. Furthermore, there were no intra tumoural differences in treated compared to untreated tumours. However, because of the promising results of fenretinide on neuroblastoma growth in vitro, further in vivo studies are warranted using other modalities of drug administration.


Assuntos
Fenretinida/administração & dosagem , Fenretinida/uso terapêutico , Neoplasias Experimentais/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Humanos , Masculino , Transplante de Neoplasias , Ratos , Ratos Nus , Fatores de Tempo
18.
Int J Cancer ; 104(4): 418-24, 2003 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-12584737

RESUMO

Retinoids modulate cell proliferation, differentiation and apoptosis in a variety of tumour cells including leukaemia and neuroblastoma, a childhood tumour of the sympathetic nervous system. 13-cis retinoic acid is in clinical use against minimal residual disease in neuroblastoma, where the effect seems to depend on dose, scheduling and tumour mass. Novel retinoids are searched for, to improve potency and lower toxicity. We investigated the effect of the synthetic retinoid Ro 13-6307 on neuroblastoma growth in vitro on SK-N-BE(2) and SH-SY5Y cells. Furthermore, effects on tumour growth and the toxicity profile were investigated in a rat xenograft model. Effects of Ro 13-6307 were compared to 13-cis RA (retinoic acid) in vitro and in vivo. Neuroblastoma cells treated with 1 microM Ro 13-6307 exhibited neuronal differentiation, decreased proliferation and accumulation of cells in G1 phase in at least the same magnitude as 5 microM 13-cis RA. No apoptosis was detected in vitro. Treatment of nude rats with neuroblastoma using Ro 13-6307, 0.12 mg p.o. daily, decreased neuroblastoma growth in vivo, in terms of tumour volume during treatment and tumour weight at sacrifice (p < 0.05). In contrast, Ro 13-6307, 0.08 mg p.o. daily, resulted in no significant reduction in tumour growth. All rats treated with Ro 13-6307 gained less weight than control rats, but they exhibited no other signs of toxicity. The toxicity profile of Ro 13-6307 was similar to what we found with 13-cis RA. Our preclinical results suggest that Ro 13-6307 may be a candidate retinoid for clinical oral therapy of neuroblastoma in children.


Assuntos
Antineoplásicos/farmacologia , Ácidos Graxos Insaturados/farmacologia , Neuroblastoma/tratamento farmacológico , Animais , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Ácidos Graxos Insaturados/toxicidade , Humanos , Isotretinoína/farmacologia , Masculino , Neuroblastoma/patologia , Ratos , Células Tumorais Cultivadas
19.
Int J Oncol ; 22(3): 579-88, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12579311

RESUMO

4-phenylbutyrate (triButyrate trade mark, PB) a derivative of the short-chain fatty acid, butyrate, possesses anti-tumor activity in vitro in different tumor cell lines. Unlike most cytostatic compounds, PB possesses low toxicity. In order to evaluate possible clinical use of PB in cancer therapy, hepatocarcinoma (Hep3B) and hepatoblastoma (HepT1) cell lines, as well as xenografts derived from those in nude rats, were treated with PB in different dose (1-100 mM) and time regimens. Treatment with 10 mM of PB for 24 h (or 5 mM for 48 h) was shown to significantly inhibit Hep3B cell growth in vitro. The HepT1 cell line was more sensitive to PB treatment: already 1 mM of PB for 24 h significantly inhibited the growth of the cells. PB also resulted in regression of xenografts derived from these cell lines in vivo, when administrated by mini-pump with an intratumor catheter, yielding 20 micro mol of PB per cm3 of tumor volume per day. TUNEL assay and caspase-3 activity measurements suggested apoptosis to be the cell death mechanism in both cell lines and xenografts. Increased histones H3 and H4 acetylation was shown in both cells and xenografts, and the inhibition of histone deacetylase is proposed as the main trigger for the anti-tumor action of PB. Concomitant induction of p21Waf1/Cip1 expression was detected by RNase protection assay and Western blotting. Reduction in expression of alpha-fetoprotein was found both in Hep3B cells and xenografts, suggesting also a differentiation effect by PB.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Inibidores Enzimáticos/uso terapêutico , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Hepáticas/patologia , Fenilbutiratos/uso terapêutico , Acetilação , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Caspase 3 , Caspases/análise , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/transplante , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/biossíntese , Ciclinas/genética , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases , Histonas/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Bombas de Infusão , Masculino , Proteínas de Neoplasias/análise , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fenilbutiratos/administração & dosagem , Fenilbutiratos/farmacologia , Ratos , Ratos Nus , Ensaios Antitumorais Modelo de Xenoenxerto , alfa-Fetoproteínas/biossíntese , alfa-Fetoproteínas/genética
20.
Clin Chem ; 49(1): 104-12, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12507966

RESUMO

BACKGROUND: Sensitive monitoring of minimal residual disease may improve the treatment of neuroblastoma in children. To detect and monitor neuroblastoma cells in blood and bone marrow, we developed a quantitative method for the analysis of tyrosine hydroxylase mRNA. METHODS: We used real-time reverse transcription-PCR. The calibrator was constructed from a segment of tyrosine hydroxylase mRNA that included the target. Blood and bone marrow samples from 24 children with neuroblastoma and 1 child with ganglioneuroma were analyzed. Controls were blood samples from the cords of 40 babies, from 58 children 6 months to 15 years of age, and from 34 healthy adults, as well as from 12 children with other diseases. RESULTS: The detection limit was approximately 70 transcripts/mL. All 144 blood controls were below this limit. At diagnosis, blood tyrosine hydroxylase mRNA was higher in children with widespread disease (stage 4/4S; n = 6; range, 203-46,000 transcripts/mL) than in patients with localized disease (stages 1-3; n = 6;

Assuntos
Medula Óssea/enzimologia , Neuroblastoma/diagnóstico , RNA Mensageiro/análise , Tirosina 3-Mono-Oxigenase/sangue , Adolescente , Adulto , Medula Óssea/patologia , Criança , Pré-Escolar , Seguimentos , Humanos , Lactente , Neoplasia Residual , Neuroblastoma/enzimologia , Neuroblastoma/terapia , Reação em Cadeia da Polimerase , RNA Mensageiro/sangue , Sensibilidade e Especificidade , Células Tumorais Cultivadas , Tirosina 3-Mono-Oxigenase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA