Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Pharmacogenomics J ; 18(1): 56-63, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-27958379

RESUMO

This study was designed to investigate the mode of action of trabectedin in myelomonocytic leukemia cells by applying systems biology approaches to mine gene expression profiling data and pharmacological assessment of the cellular effects. Significant enrichment was found in regulons of target genes inferred for specific transcription factors, among which MAFB was the most upregulated after treatment and was central in the transcriptional network likely to be relevant for the specific therapeutic effects of trabectedin against myelomonocytic cells. Using the Connectivity Map, similarity among transcriptional signatures elicited by treatment with different compounds was investigated, showing a high degree of similarity between transcriptional signatures of trabectedin and those of the topoisomerase I inhibitor, irinotecan, and an anti-dopaminergic antagonist, thioridazine. The study highlights the potential importance of systems biology approaches to generate new hypotheses that are experimentally testable to define the specificity of the mechanism of action of drugs.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Leucemia Mieloide/tratamento farmacológico , Trabectedina/farmacologia , Linhagem Celular Tumoral , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes/efeitos dos fármacos , Humanos , Biologia de Sistemas/métodos , Transcrição Gênica/efeitos dos fármacos
2.
Br J Cancer ; 113(12): 1687-93, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26633559

RESUMO

BACKGROUND: In order to investigate the mechanisms of acquired resistance to trabectedin, trabectedin-resistant human myxoid liposarcoma (402-91/T) and ovarian carcinoma (A2780/T) cell lines were derived and characterised in vitro and in vivo. METHODS: Resistant cell lines were obtained by repeated exposures to trabectedin. Characterisation was performed by evaluating drug sensitivity, cell cycle perturbations, DNA damage and DNA repair protein expression. In vivo experiments were performed on A2780 and A2780/T xenografts. RESULTS: 402-91/T and A2780/T cells were six-fold resistant to trabectedin compared with parental cells. Resistant cells were found to be hypersensitive to UV light and did not express specific proteins involved in the nucleotide excision repair (NER) pathway: XPF and ERCC1 in 402-91/T and XPG in A2780/T. NER deficiency in trabectedin-resistant cells was associated with the absence of a G2/M arrest induced by trabectedin and with enhanced sensitivity (two-fold) to platinum drugs. In A2780/T, this collateral sensitivity, confirmed in vivo, was associated with an increased formation of DNA interstrand crosslinks. CONCLUSIONS: Our finding that resistance to trabectedin is associated with the loss of NER function, with a consequent increased sensitivity to platinum drugs, provides the rational for sequential use of these drugs in patients who have acquired resistance to trabectedin.


Assuntos
Antineoplásicos/farmacologia , Dioxóis/farmacologia , Compostos Organoplatínicos/farmacologia , Tetra-Hidroisoquinolinas/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Feminino , Histonas/metabolismo , Humanos , Camundongos , Camundongos Nus , Trabectedina , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Int J Cancer ; 131(1): 59-69, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21805478

RESUMO

Myxoid Liposarcomas (MLS), characterized by the expression of FUS-CHOP fusion gene are clinically very sensitive to the DNA binding antitumor agent, trabectedin. However, resistance eventually occurs, preventing disease eradication. To investigate the mechanisms of resistance, a trabectedin resistant cell line, 402-91/ET, was developed. The resistance to trabectedin was not related to the expression of MDR related proteins, uptake/efflux of trabectedin or GSH levels that were similar in parental and resistant cells. The 402-91/ET cells were hypersensitive to UV light because of a nucleotide excision repair defect: XPG complementation decreased sensitivity to UV rays, but only partially to trabectedin. 402-91/ET cells showed collateral sensitivity to temozolomide due to the lack of O(6) -methylguanine-DNA-methyltransferase (MGMT) activity, related to the hypermethylation of MGMT promoter. In 402-91 cells chromatin immunoprecipitation (ChIP) assays showed that FUS-CHOP was bound to the PTX3 and FN1 gene promoters, as previously described, and trabectedin caused FUS-CHOP detachment from DNA. Here we report that, in contrast, in 402-91/ET cells, FUS-CHOP was not bound to these promoters. Differences in the modulation of transcription of genes involved in different pathways including signal transduction, apoptosis and stress response between the two cell lines were found. Trabectedin activates the transcription of genes involved in the adipogenic-program such as c/EBPα and ß, in 402-91 but not in 402-91/ET cell lines. The collateral sensitivity of 402-91/ET to temozolomide provides the rationale to investigate the potential use of methylating agents in MLS patients resistant to trabectedin.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Linhagem Celular Tumoral , Dioxóis/farmacologia , Lipossarcoma Mixoide/genética , Lipossarcoma Mixoide/metabolismo , Tetra-Hidroisoquinolinas/farmacologia , Apoptose , Proteína C-Reativa/genética , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/genética , Metilação de DNA , Metilases de Modificação do DNA/deficiência , Metilases de Modificação do DNA/genética , Metilases de Modificação do DNA/metabolismo , Reparo do DNA , Enzimas Reparadoras do DNA/deficiência , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Fibronectinas/genética , Humanos , Lipossarcoma Mixoide/tratamento farmacológico , Lipossarcoma Mixoide/patologia , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Regiões Promotoras Genéticas , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/metabolismo , Componente Amiloide P Sérico/genética , Transdução de Sinais , Temozolomida , Trabectedina , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Raios Ultravioleta
4.
Cell Mol Immunol ; 18(3): 711-722, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32728200

RESUMO

In cancer, myeloid cells have tumor-supporting roles. We reported that the protein GPNMB (glycoprotein nonmetastatic B) was profoundly upregulated in macrophages interacting with tumor cells. Here, using mouse tumor models, we show that macrophage-derived soluble GPNMB increases tumor growth and metastasis in Gpnmb-mutant mice (DBA/2J). GPNMB triggers in the cancer cells the formation of self-renewing spheroids, which are characterized by the expression of cancer stem cell markers, prolonged cell survival and increased tumor-forming ability. Through the CD44 receptor, GPNMB mechanistically activates tumor cells to express the cytokine IL-33 and its receptor IL-1R1L. We also determined that recombinant IL-33 binding to IL-1R1L is sufficient to induce tumor spheroid formation with features of cancer stem cells. Overall, our results reveal a new paracrine axis, GPNMB and IL-33, which is activated during the cross talk of macrophages with tumor cells and eventually promotes cancer cell survival, the expansion of cancer stem cells and the acquisition of a metastatic phenotype.


Assuntos
Fibrossarcoma/patologia , Receptores de Hialuronatos/metabolismo , Interleucina-33/metabolismo , Neoplasias Pulmonares/patologia , Macrófagos/imunologia , Glicoproteínas de Membrana/metabolismo , Células-Tronco Neoplásicas/patologia , Animais , Apoptose , Proliferação de Células , Fibrossarcoma/etiologia , Fibrossarcoma/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Receptores de Hialuronatos/genética , Interleucina-33/genética , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos DBA , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Sarcoma Experimental/etiologia , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Eur J Cancer ; 44(4): 609-18, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18243687

RESUMO

Trabectedin (ET-743, Yondelis) is a natural marine compound with antitumour activity currently undergoing phase II/III clinical trials. The mechanism of the drug's action is still to be defined, even though it has been clearly demonstrated the key role of Nucleotide Excision Repair (NER). To get further insights into the drug's mode of action, we studied the involvement of the DNA-double strand break repair (DNA-DSB) pathways: homologous and non-homologous recombination, both in budding yeasts and in mammalian cells and the possible cross-talk between NER and these repair pathways. Budding yeasts and mammalian cells deficient in the non-homologous end-joining pathway were moderately sensitive to trabectedin, while systems deficient in the homologous recombination pathway were extremely sensitive to the drug, with a 100-fold decrease in the IC50, suggesting that trabectedin-induced lesions are repaired by this pathway. The induction of Rad51 foci and the appearance of gamma-H2AX were chosen as putative markers for DNA-DSBs and were studied at different time points after trabectedin treatment in NER proficient and deficient systems. Both were clearly detected only in the presence of an active NER, suggesting that the DSBs are not directly caused by the drug, but are formed during the processing/repair of the drug- induced lesions.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Dioxóis/farmacologia , Tetra-Hidroisoquinolinas/farmacologia , Animais , Antineoplásicos/farmacologia , Biomarcadores/metabolismo , Células CHO , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Cisplatino/farmacologia , Cricetinae , Cricetulus , Reparo do DNA/genética , Relação Dose-Resposta a Droga , Fibroblastos/efeitos dos fármacos , Citometria de Fluxo , Histonas/efeitos dos fármacos , Histonas/metabolismo , Humanos , Imuno-Histoquímica , Metanossulfonato de Metila/farmacologia , Mutação , Fosforilação , Saccharomyces cerevisiae/genética , Trabectedina
6.
Leuk Res ; 32(5): 791-7, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17964649

RESUMO

T acute lymphoblastic leukemia cell lines treated with hexamethylene bisacetamide (HMBA) undergo a delay in cell cycle progression and increase susceptibility to apoptosis, although they never overcome the differentiation block. In accordance with changes in cell cycle and apoptosis, transitory p53 pathway activation commonly occurs. Bcl-2 inhibition further favours the pro-apoptotic effect of HMBA. Notch1 expression is down regulated by reduction of its transcription level. Accordingly, Notch1 protein and transcriptional activity were affected. Even if HMBA generally reduces Notch1 level in T acute lymphoblastic leukemia (T-ALL) cell lines, this does not commonly influence the biological response; in fact all the analysed cell lines, except CEM cells, display no biological effect following DAPT-induced Notch inhibition.


Assuntos
Acetamidas/farmacologia , Antineoplásicos/farmacologia , Leucemia-Linfoma de Células T do Adulto/tratamento farmacológico , Linhagem Celular Tumoral , Quinase 4 Dependente de Ciclina/análise , Humanos , Leucemia-Linfoma de Células T do Adulto/patologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Receptor Notch1/fisiologia , Transdução de Sinais , Triglicerídeos/farmacologia , Proteína Supressora de Tumor p53/fisiologia , Ácido gama-Aminobutírico/análogos & derivados , Ácido gama-Aminobutírico/farmacologia
7.
J Control Release ; 278: 49-56, 2018 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-29621597

RESUMO

Spinal cord injury (SCI) is an acute neurodegenerative disorder caused by traumatic damage of the spinal cord. The neuropathological evolution of the primary trauma involves multifactorial processes that exacerbate the pathology, worsening the neurodegeneration and limiting neuroregeneration. This complexity suggests that multi-therapeutic approaches, rather than any single treatment, might be more effective. Encouraging preclinical results indicate that stem cell-based treatments may improve the disease outcome due to their multi-therapeutic ability. Mesenchymal Stem Cells (MSCs) are currently considered one of the most promising approaches. Significant improvement in the behavioral outcome after MSC treatment sustained by hydrogel has been demonstrated. However, it is still not known how hydrogel contribute to the delivery of factors secreted from MSCs and what factors are released in situ. Among different mediators secreted by MSCs after seeding into hydrogel, we have found CCL2 chemokine, which could account for the neuroprotective mechanisms of these cells. CCL2 secreted from human MSCs is delivered efficaciously in the lesioned spinal cord acting not only on recruitment of macrophages, but driving also their conversion to an M2 neuroprotective phenotype. Surprisingly, human CCL2 delivered also plays a key role in preventing motor neuron degeneration in vitro and after spinal cord trauma in vivo, with a significant improvement of the motor performance of the rodent SCI models.


Assuntos
Biomimética , Quimiocina CCL2/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Traumatismos da Medula Espinal/terapia , Animais , Quimiocina CCL2/administração & dosagem , Modelos Animais de Doenças , Humanos , Hidrogéis , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Motores/patologia , Recuperação de Função Fisiológica , Resultado do Tratamento
8.
Chem Sci ; 8(2): 1295-1302, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-28451272

RESUMO

Alzheimer's disease is a neurodegenerative disorder linked to oligomerization and fibrillization of amyloid ß peptides, with Aß1-42 being the most aggregative and neurotoxic one. We report herein the synthesis and conformational analysis of Aß1-42-amyloid related ß-hairpin peptidomimetics, built on a piperidine-pyrrolidine semi rigid ß-turn inducer and bearing two small recognition peptide sequences, designed on oligomeric and fibril structures of Aß1-42. According to these peptide sequences, a stable ß-hairpin or a dynamic equilibrium between two possible architectures was observed. These original constructs are able to greatly delay the kinetics of Aß1-42 aggregation process as demonstrated by thioflavin-T fluorescence, and transmission electron microscopy. Capillary electrophoresis indicates their ability to preserve the monomer species, inhibiting the formation of toxic oligomers. Furthermore, compounds protect against toxic effects of Aß on neuroblastoma cells even at substoichiometric concentrations. This study is the first example of acyclic small ß-hairpin mimics possessing such a highly efficient anti-aggregation activity. The protective effect is more pronounced than that observed with molecules which have undergone clinical trials. The structural elements made in this study provide valuable insights in the understanding of the aggregation process and insights to explore the design of novel acyclic ß-hairpin targeting other types of amyloid-forming proteins.

9.
Biochem Pharmacol ; 144: 52-62, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28782526

RESUMO

Trabectedin and its analogue lurbinectedin are effective drugs used in the treatment of ovarian cancer. Since the presence of ascites is a frequent event in advanced ovarian cancer we asked the question whether ascites could modify the activity of these compounds against ovarian cancer cells. The cytotoxicity induced by trabectedin or lurbinectedin against A2780, OVCAR-5 cell lines or primary culture of human ovarian cancer cells was compared by performing treatment in regular medium or in ascites taken from either nude mice or ovarian cancer patients. Ascites completely abolished the activity of lurbinectedin at up to 10nM (in regular medium corresponds to the IC90), strongly reduced that of trabectedin, inhibited the cellular uptake of lurbinectedin and, to a lesser extent, that of trabectedin. Since α1-acid glycoprotein (AGP) is present in ascites at relatively high concentrations, we tested if the binding of the drugs to this protein could be responsible for the reduction of their activity. Adding AGP to the medium at concentration range of those found in ascites, we reproduced the anticytotoxic effect of ascites. Erythromycin partially restored the activity of the drugs, presumably by displacing them from AGP. Equilibrium dialysis experiments showed that both drugs bind AGP, but the affinity of binding of lurbinectedin was much greater than that of trabectedin. KD values are 8±1.7 and 87±14nM for lurbinectedin and trabectedin, respectively. The studies intimate the possibility that AGP present in ascites might reduce the activity of lurbinectedin and to a lesser extent of trabectedin against ovarian cancer cells present in ascites. AGP plasma levels could influence the distribution of these drugs and thus they should be monitored in patients receiving these compounds.


Assuntos
Ascite/metabolismo , Carbolinas/metabolismo , Dioxóis/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/metabolismo , Orosomucoide/metabolismo , Neoplasias Ovarianas/metabolismo , Tetra-Hidroisoquinolinas/metabolismo , Animais , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Carbolinas/farmacologia , Carbolinas/uso terapêutico , Linhagem Celular Tumoral , Dioxóis/farmacologia , Dioxóis/uso terapêutico , Relação Dose-Resposta a Droga , Feminino , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/uso terapêutico , Humanos , Camundongos , Camundongos Nus , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/cirurgia , Ligação Proteica/fisiologia , Tetra-Hidroisoquinolinas/farmacologia , Tetra-Hidroisoquinolinas/uso terapêutico , Trabectedina , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
10.
Cancer Res ; 47(10): 2547-52, 1987 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-3032408

RESUMO

Six short term-cultured melanoma cell lines and one small cell lung cancer cell line were treated in vitro with the alkylating agent mafosfamide. The sensitivity of the surviving cells to in vitro lysis by recombinant interleukin 2-activated autologous and allogeneic lymphocytes was then investigated. In no case did chemo-surviving tumor cells appear less sensitive to lymphocyte-mediated lysis than untreated counterparts. In three of seven cases (two of which were derived from the same patient), chemo-selected cells were even more sensitive to cytotoxic lymphocytes, a difference not explained by a different distribution of neoplastic cells in the various cell cycle phases. We also studied the inhibitory activity of activated lymphocytes on the clonogenic potential of chemo-surviving tumor cells by the human tumor clonogenic assay. Inhibitions of tumor cell growth in the two patients tested were 100 and 94%, respectively; the activity of lymphocytes was dependent on the coculture time and the effector/target cell ratio. These data indicate that in vitro treatment with mafosfamide does not select cells resistant to the action of activated lymphocytes and that, given the right experimental conditions, these immune effectors can completely lyse tumor cells.


Assuntos
Carcinoma de Células Pequenas/patologia , Ciclofosfamida/análogos & derivados , Neoplasias Pulmonares/patologia , Linfócitos/imunologia , Melanoma/patologia , Carcinoma de Células Pequenas/tratamento farmacológico , Carcinoma de Células Pequenas/terapia , Linhagem Celular , Células Clonais/análise , Ciclofosfamida/uso terapêutico , Citometria de Fluxo , Fluorometria , Humanos , Imunoterapia , Técnicas In Vitro , Interleucina-2/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/terapia , Ativação Linfocitária , Melanoma/tratamento farmacológico , Melanoma/terapia
11.
Cancer Res ; 51(1): 199-204, 1991 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-1859575

RESUMO

N-Deformyl-N-(4-N-N,N-bis(2-chloroethylamino)benzoyl)distamy cin A (FCE 24517) is a novel cytotoxic and antitumor agent shortly to be investigated in phase I clinical trials. It was equally effective in inhibiting the growth of the murine L1210 line and of a subline (L1210/PAM) resistant to nitrogen mustards, whereas distamycin A was virtually inactive. The cellular uptake and retention of FCE 24517 and distamycin A were similar, thus excluding the possibility that this marked variation in cytotoxic activity was due to different intracellular concentrations of the two compounds. FCE 24517 did not appear to act as an inhibitor of macromolecule synthesis. As shown by radioactively labeled precursor incorporation only 24 h after drug treatment a significant inhibition of DNA synthesis was observed in L1210 or in L1210/PAM, when a marked proportion of cells was arrested in premitotic phase. FCE 24517 did not cause DNA breaks, DNA interstrand cross-links, or DNA-protein cross-links in L1210 cells exposed to active drug concentrations. A very low amount of radioactivity was found to be bound irreversibly to DNA in L1210 cells exposed for 1 h to [14C]FCE 24517. Using plasmid pBr322 DNA fragments in a modified version of the Maxam and Gilbert DNA sequencing technique we found no detectable binding of FCE 24517 to N-7-guanine (the major site of alkylation for classical alkylating agents), whereas some alkylations to adenine (presumably to N-3-adenine) were demonstrated. Thus it appears that FCE 24517 is a novel antitumor agent with a mode of action different from that of the drugs currently used in the clinic. In summary it is suggested that FCE 24517 acts by causing a few selective alkylations to adenines in the minor groove of DNA, although the precise base sequence necessary has yet to be elucidated.


Assuntos
Alquilantes/química , Antineoplásicos/química , Dano ao DNA , DNA de Neoplasias/química , Distamicinas/química , Compostos de Mostarda Nitrogenada/química , Animais , Sequência de Bases , Sítios de Ligação , Transporte Biológico , Divisão Celular/efeitos dos fármacos , DNA de Neoplasias/biossíntese , Distamicinas/metabolismo , Inibidores do Crescimento , Camundongos , Mitose , Dados de Sequência Molecular , Compostos de Mostarda Nitrogenada/metabolismo , Células Tumorais Cultivadas
12.
Cancer Res ; 47(18): 4884-9, 1987 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-3621181

RESUMO

DNA damage caused by methazolastone [an analogue of 5-(3,3-dimethyl-1-triazeno)imidazole-4-carboxamide which does not require metabolic activation] was investigated in L-1210 leukemia which is sensitive to this drug and in a L-1210 subline [L-1210/N,N-bis(2-chloroethyl)-N-nitrosourea (BCNU)] which is resistant to both chloroethylnitrosoureas and methyltriazenes. Both in vitro and in vivo metazolastone caused formation of DNA alkali-labile sites (assessed by alkaline elution techniques) which were present in similar amounts and repaired at a similar rate in L-1210 and L-1210/BCNU. This suggests that these lesions are not crucial to methyltriazenes activity. DNA alkali-labile sites may be due to the removal of 7-methylguanine by 7-methylguanine-DNA glycosylase which showed the same activity in L-1210 and L-1210/BCNU. Flow cytometry studies revealed that in L-1210 but not in L-1210/BCNU methazolastone induced an arrest of cells in SL-G2-M phases. This blockade was delayed, occurring after at least two cell divisions after drug treatment and therefore appeared temporally unrelated to the presence of DNA alkali-labile sites. There was three times more O6-methylguanine-DNA methyltransferase in L-1210/BCNU than in L-1210 suggesting that methylation of O6-guanine is an important lesion for methyltriazenes activity and resistance to this drug may be linked to its repair.


Assuntos
Antineoplásicos/farmacologia , Carmustina/farmacologia , Dano ao DNA , Reparo do DNA , Dacarbazina/análogos & derivados , Leucemia L1210/tratamento farmacológico , Animais , DNA de Neoplasias/análise , Dacarbazina/farmacologia , Resistência a Medicamentos , Citometria de Fluxo , Metilação , Metiltransferases/análise , Camundongos , O(6)-Metilguanina-DNA Metiltransferase , Temozolomida
13.
Cancer Res ; 48(8): 2036-41, 1988 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-3349474

RESUMO

N10-Propargyl-5,8-dideazafolic acid (CB 3717), a new antifolate which directly inhibits thymidylate synthase and which is now under early clinical investigation, was compared with methotrexate (MTX) for its antiproliferative activity and mode of action on M14 human melanoma cell line and NIH/3T3 murine fibroblasts transfected with human c-Ha-ras oncogene (NIH/3T3R). CB 3717 was as active as MTX on both cell lines in inhibiting colony formation, but 20-100 times less potent. After 24 h of exposure both drugs caused an accumulation of cells in the G1 phase of the cell cycle, probably because of inhibition of DNA synthesis and blockage at the G1-S boundary. In NIH/3T3R treated for 16 h with 2 microM MTX or 200 microM CB 3717, we found DNA single-strand breaks amounting to approximately 130 and 140 rad equivalents, respectively, and a considerable number of DNA double-strand breaks, far more than expected if they had been the result of the proximity of single-strand breaks on the two complementary DNA strands. No DNA-protein cross-links were detected. When cells were incubated in drug-free medium for 8 h, there was a further accumulation of single-strand breaks, possibly due to the effects of the drug retained intracellularly as polyglutamyl derivative. Simultaneous treatment with 1.77 microM cycloheximide prevented DNA damage produced by both drugs. Thymidine (10 microM), renewed in the culture medium every 24 h, also prevented DNA damage and cytotoxicity. Since after 16 h treatment with MTX or CB 3717 cells were completely viable, as assessed by [3H]thymidine release, trypan blue exclusion test, and 51Cr release, DNA damage appears to be an early event preceding cell death and may be a feature of the killing ability of the drugs. The involvement of a protein in the formation of DNA breaks is suggested by the fact that when protein synthesis was inhibited with cycloheximide DNA damage was no longer seen.


Assuntos
Dano ao DNA , DNA/efeitos dos fármacos , Antagonistas do Ácido Fólico/farmacologia , Ácido Fólico/análogos & derivados , Metotrexato/farmacologia , Quinazolinas/farmacologia , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cicloeximida/farmacologia , Ácido Fólico/farmacologia , Humanos , Proteínas/metabolismo , Timidina/metabolismo , Timidina/farmacologia
14.
Cancer Res ; 43(12 Pt 1): 5674-80, 1983 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-6357429

RESUMO

Resistance of mouse M5076 (M5) ovarian reticular cell sarcoma to cyclophosphamide (CTX) was obtained in vivo by repeated drug treatment followed by transplantation of the regrowing tumor. After 16 passages, we obtained an M5 subline resistant to CTX (M5-CTX-16R). Median survival times were approximately 29 and 39 days for M5 and M5-CTX-16R, respectively. Survival of M5-bearing mice given a single i.p. dose of 200 or 300 mg/kg was 160 and 168% of controls, respectively, whereas in M5-CTX-16R it ws 103 and 123%, respectively. The resistance was not reversible after 14 additional passages with no further CTX treatment. M5 and M5-CTX-16R appear similar in histological features, pattern of metastasis formation, and DNA content, as assessed by flow cytometry (hypotetraploid). Metastases of M5-CTX-16R were also resistant to CTX. Flow cytometry studies 12 and 24 hr after CTX treatment revealed a block in S and G2-M phases in both tumors. After 48 hr and at subsequent times, no cytokinetic pertubation was evident in M5-CTX-16R, whereas in M5 marked accumulation of cells in G2-M was observed at 48, 72, 96, and 120 hr. Cross-resistance was found between CTX, L-phenylalanine mustard, chlorambucil, and hexamethylmelamine. M5-CTX-16R was sensitive, but less so than M5, to cis-platinum, 1,3-bis(2-chloroethyl)-1-nitrosourea, and imidazole-4-carboxamide,5-(3,3-dimethyl-1-triazene). Adriamycin was equally active on M5 and M5-CTX-16R, while 4'-demethylepipodophyllotoxin-9-(4,6-O-ethylidine-beta-D-glucopyranoside) was inactive. This model appears to be suitable for studies on the mechanism of resistance to CTX and alkylating agents and for screening new, non-cross-resistant drugs.


Assuntos
Antineoplásicos/uso terapêutico , Ciclofosfamida/uso terapêutico , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Animais , Carmustina/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Dacarbazina/uso terapêutico , Resistência a Medicamentos , Feminino , Linfoma Difuso de Grandes Células B/fisiopatologia , Melfalan/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/fisiopatologia
15.
Leukemia ; 17(1): 52-9, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12529660

RESUMO

The mechanism by which aplidine, a marine natural product in early clinical development as an anticancer agent, induces cell growth inhibition and apoptosis has been investigated in the human leukemia cell line MOLT-4. This cell line is characterized not only by the ability to secrete VEGF, but also for the presence on its surface of the VEGF receptor-1 (VEGFR-1). Previous studies from our laboratory concerned with evaluating early changes in gene expression induced by aplidine in MOLT-4 cells have shown that the drug decreases the expression of VEGFR-1 (Marchini et al. Proc Am Assoc Cancer Res 2000; 41: 833). Here, we report the ability of aplidine to block the VEGF/VEGFR-1 loop. We found that aplidine blocked VEGF secretion that was temporally followed by a decrease in both VEGF and VEGFR-1 production. Aplidine did not directly affect either VEGF transcription or stabilization of its mRNA. Transfection of MOLT-4 cells with an antisense VEGF cDNA construct, resulted in inhibition of colony formations. One clone, transfected with sense VEGF cDNA, secreting 8-10 times more VEGF than parental cells, was less sensitive to aplidine-induced cytotoxicity and apoptosis than control cells. Moreover, addition of VEGF in the medium decreased the activity of aplidine in MOLT-4 cells. These data demonstrate that aplidine inhibits the growth and induces apoptosis in MOLT-4 cells through the inhibition of VEGF secretion which blocks the VEGF/VEGFR-1 autocrine loop necessary for the growth of these cells.


Assuntos
Antineoplásicos/farmacologia , Depsipeptídeos , Fatores de Crescimento Endotelial/antagonistas & inibidores , Leucemia de Células T/tratamento farmacológico , Linfocinas/antagonistas & inibidores , Peptídeos Cíclicos/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Comunicação Autócrina , Divisão Celular/efeitos dos fármacos , Primers do DNA/química , Dactinomicina/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Fatores de Crescimento Endotelial/genética , Fatores de Crescimento Endotelial/metabolismo , Meia-Vida , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Leucemia de Células T/genética , Leucemia de Células T/metabolismo , Luciferases/metabolismo , Linfocinas/genética , Linfocinas/metabolismo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/metabolismo , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fatores de Crescimento do Endotélio Vascular
16.
Metallomics ; 7(4): 613-21, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25611161

RESUMO

Staphylococcus aureus possesses two canonical ABC-importers dedicated to nickel acquisition: the NikABCDE and the CntABCDF systems, active under different growth conditions. This study reports on the extracytoplasmic nickel-binding components SaNikA and SaCntA. We showed by protein crystallography that SaNikA is able to bind either a Ni-(l-His)2 complex or a Ni-(l-His) (2-methyl-thiazolidine dicarboxylate) complex, depending on their availability in culture supernatants. Native mass spectrometry experiments on SaCntA revealed that it binds the Ni(ii) ion via a different histidine-dependent chelator but it cannot bind Ni-(l-His)2. In vitro experiments are consistent with in vivo nickel content measurements that showed that l-histidine has a high positive impact on nickel import via the Cnt system. These results suggest that although both systems may require free histidine, they use different strategies to import nickel.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Histidina/química , Níquel/química , Staphylococcus aureus/metabolismo , Tiazolidinas/química , Proteínas de Bactérias/química , Quelantes/química , Cristalografia , Citoplasma/metabolismo , Escherichia coli/metabolismo , Espectrometria de Massas , Conformação Proteica
17.
Eur J Cancer ; 28(1): 66-71, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1314631

RESUMO

Exposure of human ovarian cancer SW626 cell line to 0.08 mumol/l methotrexate or 25 mumol/l aphidicolin for 24 h caused no cytotoxicity but enhanced etoposide cytotoxicity. Methotrexate or aphidicolin treatment induced a reversible blockade at the beginning of S phase which was reversed upon drug removal with a consequent wave of synchronisation. The enhancement of etoposide cytotoxicity was not due to higher etoposide intracellular uptake in the methotrexate or aphidicolin-pretreated cells. The topoisomerase II content in methotrexate or aphidicolin pretreated SW626 cells was higher than in control cells assessed by western blotting or flow cytometry. The higher etoposide cytotoxicity observed after synchronization with methotrexate or aphidicolin was apparently unrelated to the number of drug-induced DNA-topoisomerase II complexes evaluated as DNA double strand breaks or DNA-protein crosslinks. These data support the view that etoposide-induced DNA-topoisomerase II complexes are more cytotoxic in cells which are in S-phase.


Assuntos
Afidicolina/farmacologia , Etoposídeo/farmacologia , Metotrexato/farmacologia , Neoplasias Ovarianas/patologia , Sobrevivência Celular/efeitos dos fármacos , DNA Topoisomerases Tipo I/metabolismo , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Sinergismo Farmacológico , Feminino , Humanos , Mitose/efeitos dos fármacos , Neoplasias Ovarianas/metabolismo , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco
18.
Eur J Cancer ; 27(11): 1406-11, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1660292

RESUMO

Camptothecin, a specific inhibitor of topoisomerase I, caused erythroid differentiation of the human leukaemia cell-line K562, as assessed by benzidine staining at 70 h recovery following a 60 min treatment of the cells. Differentiation was confirmed by increased levels of epsilon-globin and gamma-globin mRNA in the treated cells and was accompanied by down-regulation of c-myb mRNA. Synchronisation of K562 cells by non-cytotoxic doses of methotrexate increased the differentiation induced by camptothecin, without affecting the camptothecin-induced inhibition of cellular proliferation. Camptothecin induction of differentiation and inhibition of proliferation may occur by independent mechanisms.


Assuntos
Camptotecina/farmacologia , Leucemia/patologia , Diferenciação Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo , Globinas/análise , Humanos , Linfócitos Nulos/química , Metotrexato/farmacologia , Mitose/efeitos dos fármacos , RNA Mensageiro/análise , Inibidores da Topoisomerase I , Células Tumorais Cultivadas/efeitos dos fármacos
19.
Eur J Cancer ; 37(1): 97-105, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11165136

RESUMO

The mode of action of Ecteinascidin-743 (ET-743), a marine tetrahydroisoquinoline alkaloid isolated from Ecteinascidia turbinata, which has shown very potent antitumour activity in preclinical systems and encouraging results in Phase I clinical trials was investigated at a cellular level. Both SW620 and LoVo human intestinal carcinoma cell lines exposed for 1 h to ET-743 progress through S phase more slowly than control cells and then accumulate in the G2M phase. The sensitivity to ET-743 of G1 synchronised cells was much higher than that of cells synchronised in S phase and even higher than that of cells synchronised in G2M. ET-743 concentrations up to four times higher than the IC(50) value caused no detectable DNA breaks or DNA-protein cross-links as assessed by alkaline elution techniques. ET-743 induced a significant increase in p53 levels in cell lines expressing wild-type (wt) (p53). However, the p53 status does not appear to be related to the ET-743 cytotoxic activity as demonstrated by comparing the drug sensitivity in p53 (-/-) or (+/+) mouse embryo fibroblasts and in A2780 ovarian cancer cells or the A2780/CX3 sub-line transfected with a dominant-negative mutant TP53. The cytotoxic potency of ET-743 was comparatively evaluated in CHO cell lines proficient or deficient in nucleotide excision repair (NER), and it was found that ET-743 was approximately 7-8 times less active in ERCC3/XPB and ERCC1-deficient cells than control cells. The findings that G1 phase cells are hypersensitive and that NER-deficient cells are resistant to ET-743 indicate that the mode of action of ET-743 is unique and different from that of other DNA-interacting drugs.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Dioxóis/uso terapêutico , Isoquinolinas/uso terapêutico , Antineoplásicos Alquilantes/farmacologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular , Neoplasias do Colo/patologia , Ciclinas/metabolismo , Dano ao DNA , DNA de Neoplasias/análise , Dioxóis/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Isoquinolinas/farmacologia , Tetra-Hidroisoquinolinas , Trabectedina , Células Tumorais Cultivadas/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo
20.
Eur J Cancer ; 39(13): 1920-6, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12932672

RESUMO

Yondelis (trabectidin, ET-743) is a marine natural product that has shown activity both in preclinical systems and in human malignancies such as soft tissue sarcoma and ovarian cancers that are resistant to previous chemotherapies. Molecular pharmacological studies indicated that Yondelis interacts with DNA and DNA repair systems in a way that is different from Cisplatin (DDP). The current study was designed to investigate the effects of the combination of Yondelis and DDP in human cancer cell lines and in xenografts derived from different tumours. The in vitro studies performed in human TE-671 rhabdomyosarcoma, Igrov-1 and 1A9 human ovarian carcinoma cell lines showed additive effects or slight synergism. Several human tumour xenografts, such as TE-671 rhabdomyosarcoma, SK-N-DX neuroblastoma, FADU head and neck, LX-1 non-small cell lung cancer (NSCLC), H-187 melanoma and SKOV HOC 8 ovarian carcinoma, showed an antitumour effect for the combination that was greater than that of each drug when given as a single agent. No consistent changes in the activity were observed if Yondelis and DDP were given 1 h apart in sequence or simultaneously. An orthotopically transplanted human ovarian cancer HOC 8 growing in the peritoneal cavity of nude mice was used that is insensitive to Yondelis alone and only moderately sensitive to DDP alone. The combination of the two drugs produced a dramatic increase of survival lasting several months. In conclusion, the combination of Yondelis and DDP is synergistic in vivo (i.e. the antitumour effect is greater than that of each drug used as a single agent at the maximum tolerated dose (MTD)) in different human tumour xenografts. The two drugs can be combined at the MTD of each drug, thus indicating there are no overlapping toxicities. These results provide a rationale for testing the combination of Yondelis and DDP in the clinic.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Rabdomiossarcoma/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Dioxóis/administração & dosagem , Dioxóis/efeitos adversos , Sinergismo Farmacológico , Feminino , Humanos , Isoquinolinas/administração & dosagem , Isoquinolinas/efeitos adversos , Camundongos , Transplante de Neoplasias , Tetra-Hidroisoquinolinas , Trabectedina , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA