Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 295(16): 5496-5508, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32161116

RESUMO

Previous studies have shown that sphingosine kinase interacting protein (SKIP) inhibits sphingosine kinase (SK) function in fibroblasts. SK phosphorylates sphingosine producing the potent signaling molecule sphingosine-1-phosphate (S1P). SKIP gene (SPHKAP) expression is silenced by hypermethylation of its promoter in acute myeloid leukemia (AML). However, why SKIP activity is silenced in primary AML cells is unclear. Here, we investigated the consequences of SKIP down-regulation in AML primary cells and the effects of SKIP re-expression in leukemic cell lines. Using targeted ultra-HPLC-tandem MS (UPLC-MS/MS), we measured sphingolipids (including S1P and ceramides) in AML and control cells. Primary AML cells had significantly lower SK activity and intracellular S1P concentrations than control cells, and SKIP-transfected leukemia cell lines exhibited increased SK activity. These findings show that SKIP re-expression enhances SK activity in leukemia cells. Furthermore, other bioactive sphingolipids such as ceramide were also down-regulated in primary AML cells. Of note, SKIP re-expression in leukemia cells increased ceramide levels 2-fold, inactivated the key signaling protein extracellular signal-regulated kinase, and increased apoptosis following serum deprivation or chemotherapy. These results indicate that SKIP down-regulation in AML reduces SK activity and ceramide levels, an effect that ultimately inhibits apoptosis in leukemia cells. The findings of our study contrast with previous results indicating that SKIP inhibits SK function in fibroblasts and therefore challenge the notion that SKIP always inhibits SK activity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Leucemia Mieloide Aguda/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Ceramidas/metabolismo , Humanos , Células K562 , Células Tumorais Cultivadas
2.
Blood ; 121(12): 2274-84, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23341541

RESUMO

Phosphoinositide-3 kinase (PI3K) pathway activation contributes to mantle cell lymphoma (MCL) pathogenesis, but early-phase studies of the PI3K p110δ inhibitor GS-1101 have reported inferior responses in MCL compared with other non-Hodgkin lymphomas. Because the relative importance of the class IA PI3K isoforms p110α, p110ß, and p110δ in MCL is not clear, we studied expression of these isoforms and assessed their contribution to PI3K signaling in this disease. We found that although p110δ was highly expressed in MCL, p110α showed wide variation and expression increased significantly with relapse. Loss of phosphatase and tensin homolog expression was found in 16% (22/138) of cases, whereas PIK3CA and PIK3R1 mutations were absent. Although p110δ inhibition was sufficient to block B-cell receptor-mediated PI3K activation, combined p110α and p110δ inhibition was necessary to abolish constitutive PI3K activation. In addition, GDC-0941, a predominantly p110α/δ inhibitor, was significantly more active compared with GS-1101 against MCL cell lines and primary samples. We found that a high PIK3CA/PIK3CD ratio identified a subset of primary MCLs resistant to GS-1101 and this ratio increased significantly with relapse. These findings support the use of dual p110α/p110δ inhibitors in MCL and suggest a role for p110α in disease progression.


Assuntos
Linfoma de Célula do Manto/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases , Análise Mutacional de DNA , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Linfoma de Célula do Manto/genética , Linfoma de Célula do Manto/metabolismo , Linfoma de Célula do Manto/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Purinas/administração & dosagem , Purinas/farmacologia , Purinas/uso terapêutico , Quinazolinonas/administração & dosagem , Quinazolinonas/farmacologia , Quinazolinonas/uso terapêutico , Recidiva , Transdução de Sinais/fisiologia , Especificidade por Substrato , Resultado do Tratamento
3.
Brain ; 137(Pt 1): 92-108, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24287115

RESUMO

Progressive multiple sclerosis is associated with metabolic failure of the axon and excitotoxicity that leads to chronic neurodegeneration. Global sodium-channel blockade causes side effects that can limit its use for neuroprotection in multiple sclerosis. Through selective targeting of drugs to lesions we aimed to improve the potential therapeutic window for treatment. This was assessed in the relapsing-progressive experimental autoimmune encephalomyelitis ABH mouse model of multiple sclerosis using conventional sodium channel blockers and a novel central nervous system-excluded sodium channel blocker (CFM6104) that was synthesized with properties that selectively target the inflammatory penumbra in experimental autoimmune encephalomyelitis lesions. Carbamazepine and oxcarbazepine were not immunosuppressive in lymphocyte-driven autoimmunity, but slowed the accumulation of disability in experimental autoimmune encephalomyelitis when administered during periods of the inflammatory penumbra after active lesion formation, and was shown to limit the development of neurodegeneration during optic neuritis in myelin-specific T cell receptor transgenic mice. CFM6104 was shown to be a state-selective, sodium channel blocker and a fluorescent p-glycoprotein substrate that was traceable. This compound was >90% excluded from the central nervous system in normal mice, but entered the central nervous system during the inflammatory phase in experimental autoimmune encephalomyelitis mice. This occurs after the focal and selective downregulation of endothelial p-glycoprotein at the blood-brain barrier that occurs in both experimental autoimmune encephalomyelitis and multiple sclerosis lesions. CFM6104 significantly slowed down the accumulation of disability and nerve loss in experimental autoimmune encephalomyelitis. Therapeutic-targeting of drugs to lesions may reduce the potential side effect profile of neuroprotective agents that can influence neurotransmission. This class of agents inhibit microglial activity and neural sodium loading, which are both thought to contribute to progressive neurodegeneration in multiple sclerosis and possibly other neurodegenerative diseases.


Assuntos
Benzamidas/uso terapêutico , Indazóis/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Oxidiazóis/uso terapêutico , Bloqueadores dos Canais de Sódio/uso terapêutico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Bancos de Espécimes Biológicos , Encéfalo/patologia , Carbamazepina/farmacologia , Proteínas de Transporte/metabolismo , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Sistemas de Liberação de Medicamentos , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Imuno-Histoquímica , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/fisiopatologia , Neurite Óptica/fisiopatologia , Linfócitos T/efeitos dos fármacos , Uveíte/fisiopatologia , Canais de Sódio Disparados por Voltagem/metabolismo
4.
Blood ; 115(19): 3939-48, 2010 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-20190189

RESUMO

GCS-100 is a galectin-3 antagonist with an acceptable human safety profile that has been demonstrated to have an antimyeloma effect in the context of bortezomib resistance. In the present study, the mechanisms of action of GCS-100 are elucidated in myeloma cell lines and primary tumor cells. GCS-100 induced inhibition of proliferation, accumulation of cells in sub-G(1) and G(1) phases, and apoptosis with activation of both caspase-8 and -9 pathways. Dose- and time-dependent decreases in MCL-1 and BCL-X(L) levels also occurred, accompanied by a rapid induction of NOXA protein, whereas BCL-2, BAX, BAK, BIM, BAD, BID, and PUMA remained unchanged. The cell-cycle inhibitor p21(Cip1) was up-regulated by GCS-100, whereas the procycling proteins CYCLIN E2, CYCLIN D2, and CDK6 were all reduced. Reduction in signal transduction was associated with lower levels of activated IkappaBalpha, IkappaB kinase, and AKT as well as lack of IkappaBalpha and AKT activation after appropriate cytokine stimulation (insulin-like growth factor-1, tumor necrosis factor-alpha). Primary myeloma cells showed a direct reduction in proliferation and viability. These data demonstrate that the novel therapeutic molecule, GCS-100, is a potent modifier of myeloma cell biology targeting apoptosis, cell cycle, and intracellular signaling and has potential for myeloma therapy.


Assuntos
Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Galectina 3/antagonistas & inibidores , Mieloma Múltiplo/patologia , Polissacarídeos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Western Blotting , Caspases/metabolismo , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Ativação Enzimática/efeitos dos fármacos , Galectina 3/metabolismo , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides , Células Estromais/efeitos dos fármacos , Células Tumorais Cultivadas
5.
Br J Haematol ; 154(4): 448-56, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21770918

RESUMO

We have previously reported presentation serum selenium level to be predictive of outcome in diffuse large B-cell lymphoma. This has now been studied in a further 430 patients, 163 with acute myeloid leukaemia (AML), 156 with Hodgkin Lymphoma (HL), and 111 with Follicular Lymphoma (FL). Serum selenium was below the UK normal reference range in 45% of patients, and correlated with serum albumin (r=0·24-0·46, P<0·001-0·003) in all tumour types. Independent predictors of presentation selenium were; French-American-British subtype and albumin (P<0·001 for both) in AML, haemoglobin (P=0·002) and B-symptoms (P=0·01) in HL, and albumin (P<0·001) in FL. In AML and HL, response to first line therapy was lower in patients with low serum selenium, but selenium was no longer predictive of response when other variables were entered into a multivariate model. Low selenium was also associated with a worse overall survival in FL [Hazard Ratio (HR) 2·3, 95% confidence interval (CI) 1·4, 4·0] and a trend to a worse overall survival in AML (HR 1·43, 95% CI 0·96, 2·13) by univariate Cox regression analysis, but not by multivariate analysis. In conclusion, low serum selenium is associated with a worse outcome in patients with haematological malignancies, but is not independently predictive, suggesting that it reflects other factors.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias Hematológicas/diagnóstico , Selênio/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Seguimentos , Neoplasias Hematológicas/sangue , Neoplasias Hematológicas/terapia , Doença de Hodgkin/sangue , Doença de Hodgkin/diagnóstico , Doença de Hodgkin/terapia , Humanos , Estimativa de Kaplan-Meier , Leucemia Mieloide Aguda/sangue , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/terapia , Linfoma Folicular/sangue , Linfoma Folicular/diagnóstico , Linfoma Folicular/terapia , Masculino , Pessoa de Meia-Idade , Prognóstico , Albumina Sérica/metabolismo , Resultado do Tratamento , Adulto Jovem
7.
Am J Pathol ; 176(6): 2607-15, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20395447

RESUMO

Development of chemoresistance limits the clinical efficiency of platinum-based therapy. Although many resistance mechanisms have been demonstrated, genetic/molecular alterations responsible for drug resistance in the majority of clinical cases have not been identified. We analyzed three pairs of testicular germ cell tumor cell lines using Affymetrix expression microarrays and revealed a limited number of differentially expressed genes across the cell lines when comparing the parental and resistant cells. Among them, CCND1 was the most significantly differentially expressed gene. Analysis of testicular germ cell tumor clinical samples by quantitative reverse transcription PCR analysis revealed that overall expression of CCND1 was significantly higher in resistant cases compared with sensitive samples (P < 0.0001). We also found that CCND1 was dramatically overexpressed both in induced and intrinsically resistant samples of ovarian and prostate cancer. Finally combined CCND1 knockdown using small-interfering RNA and cisplatin treatment inhibited cell growth in vitro significantly more effectively than any of these single treatments. Therefore, deregulation of CCND1 may be a major cause of cisplatin resistance in testicular germ cell tumors and may also be implicated in ovarian and prostate cancers. CCND1 could be potentially used as a marker for treatment stratification and as a molecular target to improve the treatment of platinum-resistant tumors.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Ciclina D1/genética , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Embrionárias de Células Germinativas , Neoplasias Testiculares , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Hibridização Genômica Comparativa , Ciclina D1/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Análise em Microsséries , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Embrionárias de Células Germinativas/patologia , Neoplasias Embrionárias de Células Germinativas/fisiopatologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/fisiopatologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Neoplasias da Próstata/fisiopatologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/patologia , Neoplasias Testiculares/fisiopatologia
8.
Anal Bioanal Chem ; 399(5): 1789-97, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21140135

RESUMO

The formation of intracellular dimethylselenide (DMSe) as a product of exposure of non-malignant (PBMCs) and lymphoma (RL and DHL-4) cell lines to methylseleninic acid (MSA) at clinical levels is suggested here for the first time. This was achieved by analysis of cell lysates by HPLC coupled to ICP-MS via APEX-Q nebulisation, enabling limits of detection for target methyl-Se species which are up to 12-fold lower than those obtained with conventional nebulisation. Methyl-Se-glutathione (CH3Se-SG), although detected in lysates of cells exposed to MSA, was found to be a reaction product of MSA with glutathione. This was confirmed by HPLC-ESI MS (MS) analysis of lysates of control cells (unexposed to Se) spiked with MSA. The MS/MS data obtained by collision-induced dissociation fragmentation of the ion m/z 402 (for [M+H](+) 8°Se) were consistent with the presence of CH3Se-SG. Formation of DMSe was not detected by HPLC-ICP-MS in these spiked lysates, and it was found to require live cells in cell media containing MSA. Interestingly, the ratio of DMSe to CH3Se-SG was significantly higher in lymphoma cells exposed to MSA in comparison to non-malignant cells. Moreover, maximum Se uptake levels in lymphoma cell lines seemed to be reached much earlier (after 10 min of MSA exposure) than in non-malignant cells. Finally, the GC-TOF-MS speciation data obtained for cell headspace suggested that the major Se species (dimethyldiselenide) appeared to be present in lymphoma cell headspace at significantly higher concentrations than in non-malignant cell headspace after only 10 min of exposure to MSA. Evidence for the presence of dimethylselenidesulfide in lymphoma cell headspace is also provided for the first time.


Assuntos
Linfoma de Células B/química , Selênio/análise , Selênio/farmacocinética , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Humanos , Linfoma de Células B/metabolismo , Espectrometria de Massas , Compostos Organosselênicos/metabolismo , Compostos Organosselênicos/farmacologia , Distribuição Tecidual
9.
Br J Haematol ; 144(6): 887-94, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19183191

RESUMO

This multicenter phase I/II study investigated the maximum tolerated dose (MTD), safety, and efficacy of low dose intravenous (IV) melphalan in combination with bortezomib for patients with relapsed multiple myeloma (MM). Patients received bortezomib 1.3 mg/m(2) on days 1, 4, 8, and 11 and escalating doses of IV melphalan (2.5-10.0 mg/m(2)) on day 2 of a 28-day cycle for a maximum of eight cycles. Dexamethasone 20 mg was added for progressive or stable disease. Fifty-three patients were enrolled. The MTD was defined at melphalan 7.5 mg/m(2) and bortezomib 1.3 mg/m(2). The overall response rate (ORR) was 68% (23% complete or near-complete responses [CR/nCR]) whilst at the MTD (n = 33) the ORR was 76% (34% CR/nCR). After median follow-up of 17 months, the median progression free survival was 10 months, rising to 12 months at the MTD (P < 0.05 vs. non-MTD regimens). The median overall survival was 28 months, but was not yet reached at the MTD. Grade 3/4 adverse events included thrombocytopenia (62%), neutropenia (57%), infection (21%), and neuropathy (15%). Bortezomib and low-dose IV melphalan combination therapy is a safe and highly effective regimen for patients with relapsed MM. These data suggest further investigation of this combination is warranted.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Ácidos Borônicos/administração & dosagem , Ácidos Borônicos/efeitos adversos , Ácidos Borônicos/uso terapêutico , Bortezomib , Dexametasona/administração & dosagem , Dexametasona/efeitos adversos , Dexametasona/uso terapêutico , Intervalo Livre de Doença , Esquema de Medicação , Feminino , Seguimentos , Humanos , Infusões Intravenosas , Estimativa de Kaplan-Meier , Masculino , Dose Máxima Tolerável , Melfalan/administração & dosagem , Melfalan/efeitos adversos , Melfalan/uso terapêutico , Pessoa de Meia-Idade , Mieloma Múltiplo/mortalidade , Pirazinas/administração & dosagem , Pirazinas/efeitos adversos , Pirazinas/uso terapêutico , Recidiva , Estatísticas não Paramétricas , Trombocitopenia/induzido quimicamente
10.
Clin Cancer Res ; 14(22): 7526-34, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19010871

RESUMO

PURPOSE: Copper chelation reduces the secretion of many angiogenic factors and reduces tumor growth and microvascular density in animal models. ATN-224 is a second-generation analogue of ammonium tetrathiomolybdate. The aim of our phase I study was to reduce serum copper levels, as measured by ceruloplasmin, to 5 to 15 mg/dL (normal 16-60) in 14 to 21 days, to determine the pharmacokinetic profile of ATN-224 and to evaluate dose-limiting toxicities. PATIENTS AND METHODS: Cohorts of patients were treated with escalating oral doses of ATN-224 until copper depletion followed by a titrated maintenance dose. RESULTS: Eighteen patients received 78 cycles of ATN-224. Mean baseline ceruloplasmin was 39.6 mg/dL. The maximum administered dose was 330 mg/d where grade 3 fatigue was dose-limiting. At the maximum tolerated dose of 300 mg/d, the median time to achieve target ceruloplasmin was 21 days, and toxicities included grade 3 anemia, grade 3 neutropenia, fatigue, and sulfur eructation. ATN-224 treatment caused a significant reduction (> 90%) in RBC superoxide dismutase 1 activity and circulating endothelial cells. Pharmacokinetic data indicate greater absorption of ATN-224 and more rapid ceruloplasmin reduction when administered with a proton pump inhibitor. Stable disease of > 6 months was observed in 2 patients. CONCLUSIONS: Oral ATN-224 is a well-tolerated therapy and at a loading dose of 300 mg/d leads to a reduction of serum ceruloplasmin levels in 80% patients within 21 days. A loading dose of 300 mg/d for 2 weeks followed by a titrated maintenance dose will be the recommended starting dose for phase II study.


Assuntos
Quelantes/efeitos adversos , Quelantes/farmacocinética , Quelantes/uso terapêutico , Terapia por Quelação , Colina/efeitos adversos , Colina/farmacocinética , Colina/uso terapêutico , Cobre/sangue , Molibdênio/efeitos adversos , Molibdênio/farmacocinética , Molibdênio/uso terapêutico , Neoplasias/tratamento farmacológico , Adulto , Idoso , Ceruloplasmina/efeitos dos fármacos , Citocinas/sangue , Citocinas/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Superóxido Dismutase/sangue , Superóxido Dismutase/efeitos dos fármacos
11.
Genes Chromosomes Cancer ; 47(7): 604-13, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18384131

RESUMO

Since the introduction of cisplatin into the clinic, the treatment of patients with a variety of solid tumors including testicular germ cell tumors, ovarian and lung cancers, has dramatically improved. One of the main causes for therapeutic failure in these malignancies is the development of drug resistance. Testicular germ cell tumors (TGCTs), the most common malignancy in young men, exhibit extreme sensitivity to cisplatin-based chemotherapy, making them an ideal model for investigating the mechanisms of cisplatin chemo-sensitivity and resistance. TGCT development and pathogenesis have been well studied but little is known about the genetic background in chemo-resistant cases. We investigated genomic differences between three TGCT parental cell lines and their cisplatin resistant derivatives. Using 10K single nucleotide polymorphism (SNP) microarray analysis, we identified two small chromosomal regions with consistent copy number changes across all three pairs of resistant cell lines. These were an 8.7 Mb region at 6q26-27, which displayed consistent copy number gain and a 0.3 Mb deletion involving 4 SNPs at 10p14. Both the chromosomal gain and loss were confirmed by fluorescence in situ hybridization. The significance of these regions should be further investigated as they may contain key genes involved in the development of chemo- resistance to cisplatin-based treatment in TGCTs and other cancers.


Assuntos
Antineoplásicos/uso terapêutico , Aberrações Cromossômicas , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/genética , Dosagem de Genes , Humanos , Hibridização in Situ Fluorescente , Masculino , Análise em Microsséries , Polimorfismo de Nucleotídeo Único , Células Tumorais Cultivadas
12.
Cancer Res ; 67(6): 2783-90, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363600

RESUMO

Bortezomib is a proteasome inhibitor with proven efficacy in multiple myeloma and non-Hodgkin's lymphoma. This study reports the effects of bortezomib in B-cell lymphoma cell lines with differing sensitivity to bortezomib to investigate factors that influence sensitivity. Bortezomib induced a time- and concentration-dependent reduction in cell viability in five lymphoma cell lines, with EC(50) values ranging from 6 nmol/L (DHL-7 cells) to 25 nmol/L (DHL-4 cells) after 72 h. Bortezomib cytotoxicity was independent of p53 function, as all cell lines exhibited mutations by sequence analysis. The difference in sensitivity was not explained by proteasome or nuclear factor-kappaB (NF-kappaB) inhibition as these were similar in the most and least sensitive cells. NF-kappaB inhibition was less marked than that of a specific NF-kappaB inhibitor, Bay 11-7082. Cell cycle analysis showed a marked G(2)-arrested population in the least sensitive DHL-4 line only, an effect that was not present with Bay 11-7082 treatment. Conversely, in DHL-7 cells, bortezomib treatment resulted in cells moving into an aberrant mitosis, indicative of mitotic catastrophe that may contribute to increased sensitivity to bortezomib. These studies show that although bortezomib treatment had similar effects on apoptotic and NF-kappaB signaling pathways in these cell lines, different cell cycle effects were observed and induction of a further mechanism of cell death, mitotic catastrophe, was observed in the more sensitive cell line, which may provide some pointers to the difference in sensitivity between cell lines. An improved understanding of how DHL-7 cells abrogate the G(2)-M cell cycle checkpoint may help identify targets to increase the efficacy of bortezomib.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ácidos Borônicos/farmacologia , Linfoma de Células B/tratamento farmacológico , Mitose/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia , Proteína Supressora de Tumor p53/fisiologia , Bortezomib , Caspases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática , Humanos , Linfoma de Células B/patologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Nitrilas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sulfonas/farmacologia
16.
J Med Chem ; 49(2): 800-5, 2006 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-16420064

RESUMO

The synthesis of a novel series of potent inhibitors of histone deacetylases is described, based on arylsulfinyl-2,4-hexadienoic acid hydroxyamides and their derivatives. In vitro IC(50) values down to 40 nM were obtained, and several compounds showed inhibition of CEM (human leukemic) cell viability with IC(50) of approximately 1.5 microM, comparable to or better than that of suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase currently in clinical trials.


Assuntos
Amidas/síntese química , Antineoplásicos/síntese química , Inibidores de Histona Desacetilases , Ácido Sórbico/análogos & derivados , Ácido Sórbico/síntese química , Sulfetos/síntese química , Ácidos Sulfínicos/síntese química , Amidas/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Ácido Sórbico/farmacologia , Relação Estrutura-Atividade , Sulfetos/farmacologia , Ácidos Sulfínicos/farmacologia
17.
Cancer Res ; 62(6): 1876-83, 2002 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11912168

RESUMO

Often the use of cytotoxic drugs in cancer therapy results in stable disease rather than regression of the tumor, and this is typically seen as a failure of treatment. We now show that DNA damage is able to induce senescence in tumor cells expressing wild-type p53. We also show that cytotoxics are capable of inducing senescence in tumor tissue in vivo. Our results suggest that p53 and p21 play a central role in the onset of senescence, whereas p16(INK4a) function may be involved in maintaining senescence. Thus, like apoptosis, senescence appears to be a p53-induced cellular response to DNA damage and an important factor in determining treatment outcome.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Dano ao DNA/fisiologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Ciclo Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/biossíntese , Etoposídeo/farmacologia , Humanos , Irinotecano , Terapia Neoadjuvante , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/fisiologia , beta-Galactosidase/metabolismo
18.
Oncogene ; 23(4): 981-90, 2004 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-14647427

RESUMO

The recombinant growth factors (GFs) erythropoietin (Epo) and granulocyte-macrophage colony stimulating factor (GM-CSF) have important roles in the management of cancer patients. However, the effects of these GFs at a cellular level are not well understood. We examined the effect of GFs alone, and in combination with cytotoxic chemotherapy, in a panel of seven cell lines. Flow cytometric analysis showed varying levels of receptor expression, which correlated with phosphorylated MAPK expression. Additionally, there were also concomitant increases in BCL-2 protein levels in those cells with high levels of MAPK activation. Although culturing cells with Epo or GM-CSF did not alter cell viability by themselves, GF pretreatment in cell lines expressing higher receptor levels resulted in a reduced magnitude of cell kill following exposure to cytotoxic IC50 concentrations of cisplatin. Subsequent co-culture with either the MEK inhibitor U0126 or the GM-CSF antagonist E21R negated this induced resistance to cytotoxic chemotherapy, confirming the importance of the GF receptor as well as MAPK in mediating these effects. These results suggest that the use of GFs during chemotherapy may be detrimental in those cancers expressing higher levels of the specific receptor. Conversely, our results also suggest that GFs are safe to use in chemotherapeutic regimens if the cancer cells do not overexpress the particular receptor.


Assuntos
Antineoplásicos/farmacologia , Eritropoetina/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Ativação Enzimática , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
19.
J Clin Oncol ; 21(6): 1125-32, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12637480

RESUMO

PURPOSE: To assess the safety and toxicity profile of escalating doses of intravenous irinotecan, in combination with a fixed dose of oral ciclosporin (Cs) and to determine the pharmacokinetic profile of irinotecan and its metabolites. PATIENTS AND METHODS: Patients with fluorouracil-refractory metastatic colorectal cancer received escalating doses of intravenous irinotecan from 40 to 125 mg/m(2) every 2 weeks in combination with a fixed dose of oral Cs (5 mg/kg bid for 3 days). Pharmacokinetic analysis of plasma irinotecan and its metabolites SN38 and SN38G was performed during paired cycles with and without Cs. RESULTS: Thirty-seven patients were treated. Dose-limiting toxicity of grade 4 neutropenia was seen at an irinotecan dose of 125 mg/m(2). There was no grade 4 diarrhea, and only one patient experienced grade 3 diarrhea. Toxicities caused by Cs were generally mild. Pharmacokinetic studies demonstrated that irinotecan clearance was reduced from 13.4 to 5.8 L/h/m(2) and area under the curve (AUC)(0-tn) was increased 2.2-fold by the coadministration of Cs. Similar significant increases in AUC(0-24h) were seen for both SN38 and SN38G (2.2-fold and 2.3-fold, respectively) in the presence of Cs. Antitumor activity was seen at every irinotecan dose level. CONCLUSION: The maximum tolerated irinotecan dose and recommended dose for phase II studies is 100 mg/m(2) every 2 weeks. Dose-limiting diarrhea was not seen during this study, supporting the hypothesis that pharmacokinetic modulation of irinotecan by Cs may improve its therapeutic index. Further studies using this combination are warranted.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Camptotecina/análogos & derivados , Neoplasias do Colo/tratamento farmacológico , Administração Oral , Adulto , Idoso , Antimetabólitos Antineoplásicos/farmacologia , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Camptotecina/administração & dosagem , Camptotecina/farmacocinética , Neoplasias do Colo/sangue , Neoplasias do Colo/patologia , Ciclosporina/administração & dosagem , Ciclosporina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Fluoruracila/farmacologia , Humanos , Infusões Intravenosas , Irinotecano , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Resultado do Tratamento
20.
Clin Cancer Res ; 9(13): 4682-8, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-14581337

RESUMO

PURPOSE: There is substantial interpatient variability in etoposide pharmacokinetics. Pharmacokinetic adjustment to specific plasma concentrations may make it possible to define a therapeutic plasma concentration and relate drug target expression in the tumor to response. This study evaluated the combination of cisplatin with a prolonged infusion of etoposide phosphate (EP) in advanced breast cancer and correlated response to topoisomerase II expression. EXPERIMENTAL DESIGN: Eligible patients, previously treated with an anthracycline, received 60 mg/m(2) cisplatin, followed by a 5-day infusion of EP. Plasma etoposide levels were measured on days 2 and 4 of each cycle with adjustment of the infusion rate to achieve an initial target etoposide concentration of 2 micro g/ml or 1.5 micro g/ml. Primary tumor blocks were stained by immunohistochemistry for topoisomerase IIalpha and beta. RESULTS: Thirty-six patients, treated in three consecutive cohorts, received 145 cycles of chemotherapy. Targeting plasma etoposide concentration reduced interpatient pharmacokinetic variability (32% and 62% of patients, respectively, within 10% of target concentration on days 2 and 4; cycle 1). Significant hematological toxicity (89% of patients with at least one episode of grade III/IV neutropenia, 64% of patients with at least one episode of grade III/IV thrombocytopenia) was observed. Thirty-nine percent of patients achieved a partial response, and 19% had stable disease for at least 3 months. The median time to tumor progression was 4 months, with a median survival of 11 months. Topoisomerase IIalpha expression was significantly higher (P < 0.001) in responding patients compared with those with stable or progressive disease. There was no difference in topoisomerase IIbeta expression between groups. CONCLUSION: Cisplatin and infusional EP is an active, but intensive, schedule in heavily pretreated patients with breast cancer. Clinical response correlates with tumor topoisomerase IIalpha expression.


Assuntos
Antineoplásicos/farmacocinética , Neoplasias da Mama/tratamento farmacológico , Cisplatino/farmacocinética , DNA Topoisomerases Tipo II/biossíntese , Etoposídeo/análogos & derivados , Etoposídeo/farmacocinética , Compostos Organofosforados/farmacocinética , Adulto , Idoso , Antígenos de Neoplasias , Neoplasias da Mama/patologia , Estudos de Coortes , Proteínas de Ligação a DNA , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Infusões Intravenosas , Pessoa de Meia-Idade , Metástase Neoplásica , Receptores de Estrogênio/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA