Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Surg Res ; 166(2): 247-54, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19691974

RESUMEN

BACKGROUND: Prediction of chemosensitivity is a major goal of modern oncology. The aim of this study was to establish a simple and effective model of primary culture of colorectal cancer fragments and to test whether it allows prediction of chemosensitivity. METHODS: Colorectal cancer fragments (primary tumors or liver metastases) of 94 consecutive and previously untreated patients were obtained, prepared, and cultured in polyHEMA. For each fragment cultured, a proliferative index (PI) was calculated after immunostaining at d 0 and after 7 d in culture with media alone or supplemented for 24h with the topoisomerase I inhibitor metabolite SN-38. The correlation between in vitro response (decrease in PI after exposure to the drug) and in vivo response (RECIST criteria) was studied in a subset of patients who had measurable metastases and were treated with a topoisomerase I inhibitor. RESULTS: PolyHEMA allowed three-dimensional culture of tumor fragments up to 7 d without fibroblastic invasion and with a slight but significant decrease of PI (59% at d 0 versus 51% after 7 d in culture, P < 0.001). In vitro drug efficacy was tested in 67 fragments, the mean PI after culture with SN-38 dropped to 22% (P < 0.001). In a subset of 12 patients, there was no statistically significant correlation between in vitro and in vivo response (P = 0.13). CONCLUSION: Primary culture in polyHEMA was easy to perform successfully in 71% of cases. On this model, the antiproliferative effect of SN-38 could be measured and results correlated to clinical data.


Asunto(s)
Adenocarcinoma , Camptotecina/análogos & derivados , Técnicas de Cultivo de Célula/métodos , Neoplasias Colorrectales/patología , Neoplasias Hepáticas , Adenocarcinoma/diagnóstico por imagen , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/secundario , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Camptotecina/administración & dosificación , Camptotecina/farmacología , División Celular/efectos de los fármacos , Monitoreo de Drogas/métodos , Resistencia a Antineoplásicos , Células HT29 , Humanos , Irinotecán , Antígeno Ki-67/metabolismo , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/secundario , Polihidroxietil Metacrilato/farmacología , Radiografía , Inhibidores de Topoisomerasa I/administración & dosificación , Inhibidores de Topoisomerasa I/farmacología
2.
J Biol Chem ; 284(42): 29136-45, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19648112

RESUMEN

The simultaneous activation of many distinct G protein-coupled receptors (GPCRs) and heterotrimeric G proteins play a major role in various pathological conditions. Pan-inhibition of GPCR signaling by small molecules thus represents a novel strategy to treat various diseases. To better understand such therapeutic approach, we have characterized the biomolecular target of BIM-46187, a small molecule pan-inhibitor of GPCR signaling. Combining bioluminescence and fluorescence resonance energy transfer techniques in living cells as well as in reconstituted receptor-G protein complexes, we observed that, by direct binding to the Galpha subunit, BIM-46187 prevents the conformational changes of the receptor-G protein complex associated with GPCR activation. Such a binding prevents the proper interaction of receptors with the G protein heterotrimer and inhibits the agonist-promoted GDP/GTP exchange. These observations bring further evidence that inhibiting G protein activation through direct binding to the Galpha subunit is feasible and should constitute a new strategy for therapeutic intervention.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Animales , Células COS , Calcio/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , AMP Cíclico/metabolismo , Ciclohexanos/farmacología , ADN Complementario/metabolismo , Humanos , Modelos Biológicos , Plásmidos/metabolismo , Pirazinas/farmacología , Transducción de Señal
3.
Int J Cancer ; 127(5): 1028-37, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20017140

RESUMEN

A goal of oncology is to predict chemosensitivity of tumors. This approach assumes that in a patient all tumor deposits are homogeneous. We have tested the heterogeneity between several samples of the same liver metastasis (LM; intrametastatic heterogeneity) or between multiple LM (intermetastatic heterogeneity) from colorectal cancer in a single patient. In 16 untreated patients, several fragments of LM and nontumorous liver were collected. Heterogeneity to anticancer drug treatment was assessed in vitro on primary tissue cultures on poly-HEMA-coated surface with or without the topoisomerase-I inhibitor metabolite SN-38. Heterogeneity of response to SN-38 was observed in 55% of cases from one fragment to another in the same LM and in 64% of cases from one LM to another in the same patient. Allelic losses were characterized on 5q, 8p, 17p, 18q, 22q using 29 microsatellites markers. Seven patients (58%) had a perfect homogeneity for allelic losses in their LM whereas 3 (21%) had intrametastatic and 2 (18%) had intermetastatic heterogeneity. The analysis of gene expression was carried out by real time RT-PCR quantification using specific probes for TS, TOPO1, ERCC1, and CES2. Level expression of genes tested appeared heterogeneous with average variations of 57(+ or - 23)%, 52(+ or - 18)%, 53(+ or - 18)%, 56(+ or - 16)% for TS, TOPO1, ERCC1, and CES2 respectively for intermetastatic variability and 47(+ or - 26)%, 36(+ or - 14)%, 38(+ or - 19)%, and 56(+ or - 29)%, respectively for intrametastatic variability. Our results demonstrate intermetastatic and intrametastatic heterogeneity suggesting that pretherapeutic analysis of a single tumor biopsy is likely to lead to a misinterpretation of sensitivity to anticancer treatment.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Camptotecina/análogos & derivados , Neoplasias Colorrectales/genética , Variación Genética , Neoplasias Hepáticas/genética , Pérdida de Heterocigocidad/genética , ARN Mensajero/genética , Camptotecina/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Humanos , Técnicas In Vitro , Irinotecán , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/secundario , Polihidroxietil Metacrilato/química , Polihidroxietil Metacrilato/metabolismo , Pronóstico , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Inhibidores de Topoisomerasa I
4.
Int J Cancer ; 124(6): 1449-56, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19065668

RESUMEN

CDC25 phosphatases are key actors in cyclin-dependent kinases activation whose role is essential at various stages of the cell cycle. CDC25 expression is upregulated in a number of human cancers. CDC25 phosphatases are therefore thought to represent promising novel targets in cancer therapy. Here, we report the identification and the characterization of IRC-083864, an original bis-quinone moiety that is a potent and selective inhibitor of CDC25 phosphatases in the low nanomolar range. IRC-083864 inhibits cell proliferation of a number of cell lines, regardless of their resistance to other drugs. It irreversibly inhibits cell proliferation and cell cycle progression and prevents entry into mitosis. In addition, it inhibits the growth of HCT-116 tumor spheroids with induction of p21 and apoptosis. Finally, IRC-083864 reduced tumor growth in mice with established human prostatic and pancreatic tumor xenografts. This study describes a novel compound, which merits further study as a potential anticancer agent.


Asunto(s)
Benzotiazoles/uso terapéutico , Benzoxazoles/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Quinonas/uso terapéutico , Fosfatasas cdc25/antagonistas & inhibidores , Animales , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Quinasas Ciclina-Dependientes/metabolismo , Citometría de Flujo , Humanos , Ratones , Ratones Desnudos , Trasplante Heterólogo
5.
Clin Cancer Res ; 14(2): 597-606, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18223236

RESUMEN

PURPOSE: The aim of these studies was to characterize the action of STX140 in a P-glycoprotein-overexpressing tumor cell line both in vitro and in vivo. In addition, its efficacy was determined against xenografts derived from patients who failed docetaxel therapy. EXPERIMENTAL DESIGN: The effects of STX140, Taxol, and 2-methoxyestradiol (2-MeOE2) on cell proliferation, cell cycle, and apoptosis were assessed in vitro in drug-resistant cells (MCF-7(DOX)) and the parental cell line (MCF-7(WT)). Mice bearing an MCF-7(DOX) tumor on one flank and an MCF-7(WT) tumor on the other flank were used to assess the in vivo efficacy. Furthermore, the responses to STX140 of three xenografts, derived from drug-resistant patients, were assessed. RESULTS: In this study, STX140 caused cell cycle arrest, cyclin B1 induction, and subsequent apoptosis of both MCF-7(DOX) and MCF-7(WT) cells. Taxol and 2-MeOE2 were only active in the MCF-7(WT) parental cell line. Although both STX140 and Taxol inhibited the growth of xenografts derived from MCF-7(WT) cells, only STX140 inhibited the growth of tumors derived from MCF-7(DOX) cells. 2-MeOE2 was ineffective at the dose tested against both tumor types. Two out of the three newly derived docetaxel-resistant xenografts, including a metastatic triple-negative tumor, responded to STX140 but not to docetaxel treatment. CONCLUSIONS: STX140 shows excellent efficacy in both MCF-7(WT) and MCF-7(DOX) breast cancer xenograft models, in contrast to Taxol and 2-MeOE2. The clinical potential of STX140 was further highlighted by the efficacy seen in xenografts recently derived from patients who had failed on taxane therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Estrenos/uso terapéutico , Paclitaxel/uso terapéutico , 2-Metoxiestradiol , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Estradiol/análogos & derivados , Estradiol/uso terapéutico , Humanos , Ratones , Ratones Desnudos , Moduladores de Tubulina/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Drug Resist Updat ; 11(4-5): 123-51, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18718806

RESUMEN

The mechanisms involved in the epithelial to mesenchymal transition (EMT) are integrated in concert with master developmental and oncogenic pathways regulating in tumor growth, angiogenesis, metastasis, as well as the reprogrammation of specific gene repertoires ascribed to both epithelial and mesenchymal cells. Consequently, it is not unexpected that EMT has profound impacts on the neoplastic progression, patient survival, as well as the resistance of cancers to therapeutics (taxol, vincristine, oxaliplatin, EGF-R targeted therapy and radiotherapy), independent of the "classical" resistance mechanisms linked to genotoxic drugs. New therapeutic combinations using genotoxic agents and/or EMT signaling inhibitors are therefore expected to circumvent the chemotherapeutic resistance of cancers characterized by transient or sustained EMT signatures. Thus, targeting critical orchestrators at the convergence of several EMT pathways, such as the transcription pathways NF-kappaB, AKT/mTOR axis, MAPK, beta-catenin, PKC and the AP-1/SMAD factors provide a realistic strategy to control EMT and the progression of human epithelial cancers. Several inhibitors targeting these signaling platforms are already tested in preclinical and clinical oncology. In addition, upstream EMT signaling pathways induced by receptor and nonreceptor tyrosine kinases (e.g. EGF-R, IGF-R, VEGF-R, integrins/FAK, Src) and G-protein-coupled receptors (GPCR) constitute practical options under preclinical research, clinical trials or are currently used in the clinic for cancer treatment: e.g. small molecule inhibitors (Iressa: targeting selectively the EGF-R; CP-751,871, AMG479, NVP-AEW541, BMS-536924, PQIP, AG1024: IGF-R; AZD2171, ZD6474: VEGF-R; AZD0530, BMS-354825, SKI606: Src; BIM-46174: GPCR; rapamycin, CCI-779, RAD-001: mTOR) and humanized function blocking antibodies (Herceptin: ErbB2; Avastin: VEGF-A; Erbitux: EGF-R; Abegrin: alphavbeta3 integrins). We can assume that silencing RNA and adenovirus-based gene transfer of therapeutic miR and dominant interferring expression vectors targeting EMT pathways and signaling elements will bring additional ways for the treatment of epithelial cancers. Identification of the factors that initiate, modulate and effectuate EMT signatures and their underlying upstream oncogenic pathways should provide the basis of more efficient strategies to fight cancer progression as well as genetic and epigenetic forms of drug resistance. This goal can be accomplished using global screening of human clinical tumors by EMT-associated cDNA, proteome, miRome, and tissue arrays.


Asunto(s)
Transdiferenciación Celular , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Mesodermo/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Animales , Antineoplásicos/uso terapéutico , Supervivencia Celular , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Resistencia a Antineoplásicos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Terapia Genética , Humanos , Mesodermo/efectos de los fármacos , Mesodermo/patología , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
7.
Mol Cancer Ther ; 7(8): 2426-34, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18723488

RESUMEN

Tubulin is a validated target for antitumor drugs. However, the effectiveness of these microtubule-interacting agents is limited by the fact that they are substrates for drug efflux pumps (P-glycoprotein) and/or by the acquisition of point mutations in tubulin residues important for drug-tubulin binding. To bypass these resistance systems, we have identified and characterized a novel synthetic imidazole derivative IRC-083927, which inhibits the tubulin polymerization by a binding to the colchicine site. IRC-083927 inhibits in vitro cell growth of human cancer cell lines in the low nanomolar range. More interesting, it remains highly active against cell lines resistant to microtubule-interacting agents (taxanes, Vinca alkaloids, or epothilones). Such resistances are due to the presence of efflux pumps (NCI-H69/LX4 resistant to navelbine and paclitaxel) and/or the presence of mutations on beta-tubulin and on alpha-tubulin and beta-tubulin (A549.EpoB40/A549.EpoB480 resistant to epothilone B or paclitaxel). IRC-083927 displayed cell cycle arrest in G(2)-M phase in tumor cells, including in the drug-resistant cells. In addition, IRC-083927 inhibited endothelial cell proliferation in vitro and vessel formation in the low nanomolar range supporting an antiangiogenic behavior. Finally, chronic oral treatment with IRC-083927 (5 mg/kg) inhibits the growth of two human tumor xenografts in nude mice (C33-A, human cervical cancer and MDA-MB-231, human hormone-independent breast cancer). Together, the antitumor effects induced by IRC-083927 on tumor models resistant to tubulin agents support further investigations to fully evaluate its potential for the treatment of advanced cancers, particularly those resistant to current clinically available drugs.


Asunto(s)
Antineoplásicos/farmacología , División Celular/efectos de los fármacos , Imidazoles/farmacología , Sulfonamidas/farmacología , Tubulina (Proteína)/metabolismo , Animales , Antineoplásicos/farmacocinética , Disponibilidad Biológica , Ciclo Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Humanos , Ratones , Neovascularización Patológica , Trasplante Heterólogo
8.
Pharmacol Ther ; 115(1): 1-12, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17531323

RESUMEN

The CDC25 phosphatases are key regulators of cell cycle progression and play a central role in the checkpoint response to DNA damage. Their inhibition may therefore represent a promising therapeutic approach in oncology, and small molecule design strategies are currently leading to the identification of various classes of CDC25 inhibitors. Most structures developed so far are quinonoid-based compounds, but also phosphate surrogates or electrophilic entities. Considering the characteristics of the highly conserved active sites of the enzymes, many mechanisms of action have been proposed for these inhibitors. Quinonoid compounds may oxidize the catalytic site cysteine, but can also be considered as Michaël acceptors capable of reacting with the activated thiolate or other electrophilic entities. Phosphate surrogates are thought to interfere with the arginine residue, leading to reversible enzyme inhibition. But some inhibitors can combine in the same molecule several of these mechanisms, thus by fitting into the active site of the enzyme through one part of the molecule and bringing the reactive moiety in close proximity to the catalytic cysteine. This review summarizes novel classes of inhibitors that show specificity for the CDC25s over other phosphatases, cause cell proliferation inhibition and cell cycle arrest in vitro but also, for several of them, inhibition of xenografted tumoral cell growth in vivo. These promising results confirm the interest of the inhibition of CDC25 phosphatases as an anticancer therapeutic strategy.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Animales , Inhibidores Enzimáticos/química , Humanos
9.
Eur J Pharmacol ; 594(1-3): 70-6, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18664366

RESUMEN

BIM-46187 (7-[2-amino-1-oxo-3-thio-propyl]-8-cyclohexylmethyl-2-phenyl-5,6,7,8-tetrahydro-imidazo-[1,2a]-pyrazine dimer, hydrochloride) is an inhibitor of the heterotrimeric G-protein complex signalling. Since many mediators of pain act through G-protein coupled receptors, the anti-hyperalgesic effects of BIM-46187 were assessed on experimental models of pain. In addition since opioids are widely used in pain management and act through specific G-protein-coupled receptors, the effects of BIM-46187 on the analgesic properties of morphine have also been investigated. BIM-46187 elicited a dose dependent analgesic effect in the models of carrageenan-induced hyperalgesia (0.1-1 mg/kg; i.v.) and chronic constriction injury (0.3-3 mg/kg; i.v.) in rats. BIM-46187, however, up to 10 mg/kg did not modify the paw oedema induced by carrageenan excluding an anti-inflammatory effect. In addition, at these doses, the compound was not sedative as shown by the lack of effect on the motor performance in the rotarod test. The combination of BIM-46187 and morphine (ratio 1/1) resulted in an unexpected synergistic effect in the model of carrageenan-induced hyperalgesia and in the chronic constriction injury model in rats when evaluated by isobolographic analysis. This synergy allowed a reduction of at least 20 fold in the dose of each compound. Conversely, the drug combination did not increase the side effects of morphine as assessed in the rotarod test. In conclusion, BIM-46187 elicits a potent anti-hyperalgesic effect and strongly synergizes with morphine. This work highlights the role of heterotrimeric G-protein complexes in pain and supports further investigations of the use of BIM-46187 alone, or in combination with low doses of morphine, in the management of pain.


Asunto(s)
Analgésicos no Narcóticos/farmacología , Analgésicos Opioides/farmacología , Ciclohexanos/farmacología , Proteínas de Unión al GTP Heterotriméricas/antagonistas & inhibidores , Hiperalgesia/tratamiento farmacológico , Morfina/farmacología , Pirazinas/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Carragenina , Constricción Patológica/patología , Sinergismo Farmacológico , Edema/inducido químicamente , Edema/patología , Hiperalgesia/inducido químicamente , Masculino , Dimensión del Dolor/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/patología , Enfermedades del Sistema Nervioso Periférico/psicología , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
11.
Cancer Res ; 66(14): 7128-35, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16854822

RESUMEN

The effects of cell adhesion on leukemia cell proliferation remain poorly documented and somehow controversial. In this work, we investigated the effect of adhesion to fibronectin on the proliferation of acute myeloid leukemia (AML) cell lines (U937 and KG1a) and CD34+ normal or leukemic primary cells. We observed an increased rate of proliferation of AML cells when adhered to fibronectin, concomitant with accelerated S-phase entry and accumulation of CDC25A. Conversely, normal CD34+ cell proliferation was decreased by adhesion to fibronectin with a concomitant drop in CDC25A expression. Importantly, we showed that both small interfering RNA (siRNA)-mediated CDC25A down-regulation and a recently developed CDC25 pharmacologic inhibitor impaired this adhesion-dependent proliferation, establishing a functional link between CDC25A accumulation and adhesion-dependent proliferation in leukemic cells. CDC25A accumulation was found only slightly dependent on transcriptional regulation and essentially due to modifications of the proteasomal degradation of the protein as shown using proteasome inhibitors and reverse transcription-PCR. Interestingly, CDC25A regulation was Chk1 dependent in these cells as suggested by siRNA-mediated down-regulation of this protein. Finally, we identified activation of the phosphatidylinositol 3-kinase/Akt pathway as an adhesion-dependent regulation mechanism of CDC25A protein expression. Altogether, our data show that in leukemic cells adhesion to fibronectin increases CDC25A expression through proteasome- and Chk1-dependent mechanisms, resulting in enhanced proliferation. They also suggest that these adhesion-dependent proliferation properties of hematopoietic cells may be modified during leukemogenesis.


Asunto(s)
Fosfatasas cdc25/biosíntesis , Enfermedad Aguda , Adhesión Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Fibronectinas/metabolismo , Células HL-60 , Humanos , Células Jurkat , Leucemia Mieloide/enzimología , Leucemia Mieloide/genética , Leucemia Mieloide/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Quinasas/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Serina-Treonina Quinasas TOR , Células U937 , Regulación hacia Arriba , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
12.
Cancer Res ; 66(18): 9227-34, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16982767

RESUMEN

A large number of hormones and local agonists activating guanine-binding protein-coupled receptors (GPCR) play a major role in cancer progression. Here, we characterize the new imidazo-pyrazine derivative BIM-46174, which acts as a selective inhibitor of heterotrimeric G-protein complex. BIM-46174 prevents the heterotrimeric G-protein signaling linked to several GPCRs mediating (a) cyclic AMP generation (Galphas), (b) calcium release (Galphaq), and (c) cancer cell invasion by Wnt-2 frizzled receptors and high-affinity neurotensin receptors (Galphao/i and Galphaq). BIM-46174 inhibits the growth of a large panel of human cancer cell lines, including anticancer drug-resistant cells. Exposure of cancer cells to BIM-46174 leads to caspase-3-dependent apoptosis and poly(ADP-ribose) polymerase cleavage. National Cancer Institute COMPARE analysis for BIM-46174 supports its novel pharmacologic profile compared with 12,000 anticancer agents. The growth rate of human tumor xenografts in athymic mice is significantly reduced after administration of BIM-46174 combined with either cisplatin, farnesyltransferase inhibitor, or topoisomerase inhibitors. Our data validate the feasibility of targeting heterotrimeric G-protein functions downstream the GPCRs to improve anticancer chemotherapy.


Asunto(s)
Cisteína/análogos & derivados , Proteínas de Unión al GTP Heterotriméricas/antagonistas & inhibidores , Imidazoles/farmacología , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Cisteína/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Subunidades alfa de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Subunidades beta de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Células HL-60 , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Ratones , Invasividad Neoplásica , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Mol Cancer Ther ; 6(1): 318-25, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17237290

RESUMEN

The CDC25 cell cycle regulators are promising targets for new pharmacologic approaches in cancer therapy. Inhibitory compounds such as BN82685 have proven to be effective in specifically targeting CDC25 in cultured cells and in inhibiting tumor cell growth. Here, we report that BN82685 impairs microtubule dynamic instability and alters microtubule organization and assembly at the centrosome in interphase cells. Treatment of mitotic cells with BN82685 delays mitotic spindle assembly, chromosome capture, and metaphase plate formation. Furthermore, we show that combining low concentrations of both BN82685 and paclitaxel inhibits the proliferation of HT29 human colon cancer cells. Our results show a role for CDC25 phosphatases in regulating microtubule dynamics throughout the cell cycle and suggest that combinations of CDC25 inhibitors with microtubule-targeting agents may be of therapeutic value.


Asunto(s)
Benzoquinonas/farmacología , Inhibidores Enzimáticos/farmacología , Interfase/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Huso Acromático/efectos de los fármacos , Tiazoles/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Cromosomas Humanos/efectos de los fármacos , Sinergismo Farmacológico , Células HT29 , Células HeLa , Humanos , Metafase/efectos de los fármacos , Prometafase/efectos de los fármacos
14.
Mol Cancer Ther ; 5(6): 1446-51, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16818502

RESUMEN

Cell cycle arrest at the G2-M checkpoint is an essential feature of the mechanisms that preserve genomic integrity. CDC25 phosphatases control cell cycle progression by dephosphorylating and activating cyclin-dependent kinase/cyclin complexes. Their activities are, therefore, tightly regulated to modulate cell cycle arrest in response to DNA damage exposure. Here, we report that overexpression of CDC25B affects viability, reduces clonogenic efficiency, and increases sensitivity of cancer cells to a genotoxic agent. We show that ectopic expression of CDC25B results in bypass of a genotoxic-induced G2-M checkpoint. In addition, cancer cells constitutively expressing high level of CDC25B are shown to be prone to exit prematurely from the G2-M checkpoint arrest and to enter mitosis. Finally, we show that this exit is dependent on CDC25B expression. Together with previous results, our data strongly support a model in which CDC25B is the key phosphatase that controls entry into mitosis after DNA damage, thus emphasizing the relevance of its overexpression in many human tumors.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Fosfatasas cdc25/metabolismo , Antineoplásicos Fitogénicos/farmacología , Western Blotting , Proteínas de Ciclo Celular/genética , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Daño del ADN/efectos de los fármacos , Daño del ADN/fisiología , Etopósido/farmacología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Fase G2/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Tetraciclina/farmacología , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre , Fosfatasas cdc25/genética
15.
Cancer Res ; 64(14): 4942-9, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15256467

RESUMEN

BN80927 belongs to a novel family of camptothecin analogs, the homocamptothecins, developed on the concept of topoisomerase I (Topo I) inhibition and characterized by a stable seven-membered beta-hydroxylactone ring. Preclinical data reported here show that BN80927 retains Topo I poisoning activity in cell-free assay (DNA relaxation) as well as in living cells, in which in vivo complexes of topoisomerase experiments and quantification of DNA-protein-complexes stabilization, have confirmed the higher potency of BN80927 as compared with the Topo I inhibitor SN38. In addition, BN80927 inhibits Topo II-mediated DNA relaxation in vitro but without cleavable-complex stabilization, thus indicating catalytic inhibition. Moreover, a Topo I-altered cell line (KBSTP2), resistant to SN38, remains sensitive to BN80927, suggesting that a part of the antiproliferative effects of BN80927 are mediated by a Topo I-independent pathway. This hypothesis is also supported by in vitro data showing an antiproliferative activity of BN80927 on a model of resistance related to the noncycling state of cells (G(0)-G(1) synchronized). In cell growth assays, BN80927 is a very potent antiproliferative agent as shown by IC(50) values consistently lower than those of SN38 in tumor cell lines as well as in their related drug-resistant lines. BN80927 shows high efficiency in vivo in tumor xenograft studies using human androgen-independent prostate tumors PC3 and DU145. Altogether, these data strongly support the clinical development of BN80927.


Asunto(s)
Antineoplásicos/farmacología , Camptotecina/análogos & derivados , Camptotecina/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Adenocarcinoma/sangre , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Animales , Camptotecina/sangre , División Celular/efectos de los fármacos , Línea Celular Tumoral , ADN-Topoisomerasas de Tipo I/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , ADN Superhelicoidal/efectos de los fármacos , ADN Superhelicoidal/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Estabilidad de Medicamentos , Inhibidores Enzimáticos/farmacología , Células HL-60 , Humanos , Células K562 , Masculino , Ratones , Ratones Desnudos , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/patología , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Inhibidores de Topoisomerasa I , Inhibidores de Topoisomerasa II , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Res ; 64(9): 3320-5, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15126376

RESUMEN

CDC25 dual-specificity phosphatases are essential regulators that dephosphorylate and activate cyclin-dependent kinase/cyclin complexes at key transitions of the cell cycle. CDC25 activity is currently considered to be an interesting target for the development of new antiproliferative agents. Here we report the identification of a new CDC25 inhibitor and the characterization of its effects at the molecular and cellular levels, and in animal models. BN82002 inhibits the phosphatase activity of recombinant human CDC25A, B, and C in vitro. It impairs the proliferation of tumoral cell lines and increases cyclin-dependent kinase 1 inhibitory tyrosine phosphorylation. In synchronized HeLa cells, BN82002 delays cell cycle progression at G1-S, in S phase and at the G2-M transition. In contrast, BN82002 arrests U2OS cell cycle mostly in the G1 phase. Selectivity of this inhibitor is demonstrated: (a) by the reversion of the mitotic-inducing effect observed in HeLa cells upon CDC25B overexpression; and (b) by the partial reversion of cell cycle arrest in U2OS expressing CDC25. We also show that BN82002 reduces growth rate of human tumor xenografts in athymic nude mice. BN82002 is a original CDC25 inhibitor that is active both in cell and animal models. This greatly reinforces the interest in CDC25 as an anticancer target.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , División Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Etilaminas , Femenino , Células HeLa , Humanos , Ratones , Ratones Desnudos , Mitosis/efectos de los fármacos , Nitrocompuestos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Fosfatasas cdc25/biosíntesis , Fosfatasas cdc25/genética
17.
Mol Cancer Ther ; 4(9): 1378-87, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16170030

RESUMEN

Cell cycle regulators, such as the CDC25 phosphatases, are potential targets for the development of new anticancer drugs. Here we report the identification and the characterization of BN82685, a quinone-based CDC25 inhibitor that is active in vitro and in vivo. BN82685 inhibits recombinant CDC25A, B, and C phosphatases in vitro. It inhibits the growth of human tumor cell lines with an IC(50) in the submicromolar range, independently of their resistance to chemotherapeutic agents. This inhibitory effect is irreversible on both the purified CDC25 enzyme in vitro and on tumor cell proliferation. The specificity of BN82685 towards the CDC25 phosphatases is shown by an increase in cyclin-dependent kinase 1 tyrosine 15 phosphorylation, by the reversion of the mitosis-inducing effect of CDC25B overexpression in HeLa cells, and by the lack of a growth inhibitory effect in an assay based on the use of a CDC25-independent fission yeast model. Finally, when administered p.o., BN82685 is shown to inhibit the growth of the human pancreatic tumor Mia PaCa-2 xenografted in athymic nude mice. BN82685 is therefore a promising new compound targeting CDC25, which confirms the interest of the inhibition of these enzymes as an anticancer therapeutic strategy.


Asunto(s)
Benzoquinonas/farmacología , Inhibidores Enzimáticos/farmacología , Neoplasias Pancreáticas/patología , Tiazoles/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Administración Oral , Animales , Benzoquinonas/administración & dosificación , Benzoquinonas/síntesis química , Disponibilidad Biológica , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Células HeLa , Humanos , Ratones , Ratones Desnudos , Mitosis/efectos de los fármacos , Neoplasias Pancreáticas/enzimología , Schizosaccharomyces/genética , Schizosaccharomyces/crecimiento & desarrollo , Schizosaccharomyces/metabolismo , Tiazoles/administración & dosificación , Tiazoles/síntesis química , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Biochem Pharmacol ; 65(3): 423-33, 2003 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-12527335

RESUMEN

Epstein-Barr virus (EBV)-associated nasopharyngeal carcinomas (NPC) are much more sensitive to chemotherapy than other head and neck carcinomas. Spectacular regressions are frequently observed after induction chemotherapy. However, these favorable responses are difficult to predict and often of short duration. So far there have been only few experiments to investigate the mechanisms which underline the cytotoxic effects of anti-neoplastic drugs against NPC cells. In addition, these studies were performed almost entirely on EBV-negative cell lines therefore not truly representative of NPC cells. For the first time, we have used two EBV-positive NPC tumor lines derived from a North African (C15) and a Chinese (C666-1) patient as in vitro targets for a panel of anti-neoplastic agents. Doxorubicin, taxol and in a lesser extent cis-platinum efficiently inhibited NPC cell proliferation at clinically relevant concentrations, but all three agents failed to induce apoptosis. However, massive apoptosis of C15 cells was achieved when doxorubicin (1 microM) was combined with a farnesyl-transferase inhibitor, BIM 2001 (5 microM). Moreover, this apoptotic process was associated with a caspase-dependent early cleavage of the TNF-receptor associated factor 1 (TRAF-1) molecule, a signaling adaptor which is specifically expressed in latently EBV-infected cells. TRAF-1 cleavage might become a useful indicator of chemo-induced apoptosis in EBV-associated NPCs.


Asunto(s)
Apoptosis , Doxorrubicina/farmacología , Inhibidores Enzimáticos/farmacología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Neoplasias Nasofaríngeas/patología , Nitrilos/farmacología , Proteínas/metabolismo , Transferasas Alquil y Aril/antagonistas & inhibidores , División Celular/efectos de los fármacos , Combinación de Medicamentos , Farnesiltransferasa , Femenino , Herpesvirus Humano 4/aislamiento & purificación , Humanos , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/virología , Factor 1 Asociado a Receptor de TNF , Células Tumorales Cultivadas
19.
Oncotarget ; 5(16): 6687-700, 2014 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-25149532

RESUMEN

Tumor-angiogenesis is the multi-factorial process of sprouting of endothelial cells (EC) into micro-vessels to provide tumor cells with nutrients and oxygen. To explore miRNAs as therapeutic angiogenesis-inhibitors, we performed a functional screen to identify miRNAs that are able to decrease EC viability. We identified miRNA-7 (miR-7) as a potent negative regulator of angiogenesis. Introduction of miR-7 in EC resulted in strongly reduced cell viability, tube formation, sprouting and migration. Application of miR-7 in the chick chorioallantoic membrane assay led to a profound reduction of vascularization, similar to anti-angiogenic drug sunitinib. Local administration of miR-7 in an in vivo murine neuroblastoma tumor model significantly inhibited angiogenesis and tumor growth. Finally, systemic administration of miR-7 using a novel integrin-targeted biodegradable polymeric nanoparticles that targets both EC and tumor cells, strongly reduced angiogenesis and tumor proliferation in mice with human glioblastoma xenografts. Transcriptome analysis of miR-7 transfected EC in combination with in silico target prediction resulted in the identification of OGT as novel target gene of miR-7. Our study provides a comprehensive validation of miR-7 as novel anti-angiogenic therapeutic miRNA that can be systemically delivered to both EC and tumor cells and offers promise for miR-7 as novel anti-tumor therapeutic.


Asunto(s)
Glioblastoma/terapia , MicroARNs/administración & dosificación , Animales , Proliferación Celular/genética , Embrión de Pollo , Femenino , Terapia Genética/métodos , Glioblastoma/irrigación sanguínea , Glioblastoma/genética , Glioblastoma/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos A , Ratones Desnudos , MicroARNs/genética , Neovascularización Patológica/genética , Neovascularización Patológica/terapia , Distribución Aleatoria , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA