Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Med Chem ; 124: 840-851, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-27643641

RESUMO

A series of [1,2]Oxazolo [5,4-e]isoindoles has been synthesized through a versatile and high yielding sequence. All the new structures showed in the 1HNMR spectra, the typical signal in the 8.34-8.47 ppm attributable to the H-3 of the [1,2]oxazole moiety. Among all derivatives, methoxy benzyl substituents at positions 3 and 4 or/and 5 were very effective in reducing the growth of different tumor cell lines, including diffuse malignant peritoneal mesothelioma (DMPM), an uncommon and rapidly malignancy poorly responsive to available therapeutic options. The most active compound 6j was found to impair tubulin polymerization, cause cell cycle arrest at G2/M phase and induce apoptosis in DMPM cells, making it as a new lead for the discovery of new potent antimitotic drugs.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Isoindóis/química , Isoindóis/farmacologia , Multimerização Proteica/efeitos dos fármacos , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacologia , Antineoplásicos/síntese química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Isoindóis/síntese química , Isomerismo , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Estrutura Quaternária de Proteína , Tubulina (Proteína)/química , Moduladores de Tubulina/síntese química
2.
J Med Chem ; 59(15): 7223-38, 2016 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-27428868

RESUMO

A series of 22 derivatives of the [1,2]oxazolo[5,4-e]isoindole system were synthesized through an efficient and versatile procedure that involves the annelation of the [1,2]oxazole moiety to the isoindole ring, producing derivatives with a wide substitution pattern. The structure-activity relationship indicates that the N-4-methoxybenzyl group appears crucial for potent activity. In addition, the presence of a 6-phenyl moiety is important and the best activity is reached with a 3,4,5-trimethoxy substituent. The most active compound, bearing both the structural features, was able to inhibit tumor cell proliferation at nanomolar concentrations when tested against the full NCI human tumor cell line panel. Interestingly, this compound was effective in reducing in vitro and in vivo cell growth, impairing cell cycle progression and inducing apoptosis, as a consequence of the inhibition of tubulin polymerization, in experimental models of diffuse malignant peritoneal mesothelioma (DMPM), a rapidly lethal disease, poorly responsive to conventional therapeutic strategies.


Assuntos
Antineoplásicos/farmacologia , Isoindóis/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Mesotelioma/tratamento farmacológico , Neoplasias Peritoneais/tratamento farmacológico , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Isoindóis/síntese química , Isoindóis/química , Neoplasias Pulmonares/patologia , Mesotelioma/patologia , Mesotelioma Maligno , Camundongos , Camundongos Nus , Estrutura Molecular , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Neoplasias Peritoneais/patologia , Relação Estrutura-Atividade , Células Tumorais Cultivadas
3.
Oncotarget ; 7(31): 49649-49663, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27351133

RESUMO

Medullary thyroid cancer (MTC) relies on the aberrant activation of RET proto-oncogene. Though targeted approaches (i.e., tyrosine kinase inhibitors) are available, the absence of complete responses and the onset of resistance mechanisms indicate the need for novel therapeutic interventions. Due to their role in regulation of gene expression, G-quadruplexes (G4) represent attractive targets amenable to be recognized or stabilized by small molecules. Here, we report that exposure of MTC cells to a tri-substituted naphthalene diimide (NDI) resulted in a significant antiproliferative activity paralleled by inhibition of RET expression. Biophysical analysis and gene reporter assays showed that impairment of RET expression was consequent to the NDI-mediated stabilization of the G4 forming within the gene promoter. We also showed for the first time that systemic administration of the NDI in mice xenotransplanted with MTC cells resulted in a remarkable inhibition of tumor growth in vivo. Overall, our findings indicate that NDI-dependent RET G4 stabilization represents a suitable approach to control RET transcription and delineate the rationale for the development of G4 stabilizing-based treatments for MTC as well as for other tumors in which RET may have functional and therapeutic implications.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Neuroendócrino/terapia , Imidas/química , Naftalenos/química , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-ret/metabolismo , Neoplasias da Glândula Tireoide/terapia , Animais , Carcinoma Neuroendócrino/genética , Carcinoma Neuroendócrino/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Quadruplex G , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Ligantes , Camundongos , Camundongos SCID , Mutação , Transplante de Neoplasias , Proto-Oncogene Mas , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/metabolismo
4.
Chemistry ; 21(39): 13598-608, 2015 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-26286886

RESUMO

Hsp90 is a molecular chaperone of pivotal importance for multiple cell pathways. ATP-regulated internal dynamics are critical for its function and current pharmacological approaches block the chaperone with ATP-competitive inhibitors. Herein, a general approach to perturb Hsp90 through design of new allosteric ligands aimed at modulating its functional dynamics is proposed. Based on the characterization of a first set of 2-phenylbenzofurans showing stimulatory effects on Hsp90 ATPase and conformational dynamics, new ligands were developed that activate Hsp90 by targeting an allosteric site, located 65 Šfrom the active site. Specifically, analysis of protein responses to first-generation activators was exploited to guide the design of novel derivatives with improved ability to stimulate ATP hydrolysis. The molecules' effects on Hsp90 enzymatic, conformational, co-chaperone and client-binding properties were characterized through biochemical, biophysical and cellular approaches. These designed probes act as allosteric activators of the chaperone and affect the viability of cancer cell lines for which proper functioning of Hsp90 is necessary.


Assuntos
Adenosina Trifosfatases/química , Trifosfato de Adenosina/química , Benzofuranos/química , Chaperoninas/química , Proteínas de Choque Térmico HSP90/química , Adenosina Trifosfatases/metabolismo , Sítio Alostérico , Fenômenos Bioquímicos , Linhagem Celular Tumoral , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Hidrólise , Ligantes , Ligação Proteica , Conformação Proteica
5.
Oncotarget ; 6(15): 13119-32, 2015 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-25948791

RESUMO

Survivin, which is highly expressed and promotes cell survival in diffuse malignant peritoneal mesothelioma (DMPM), exclusively relies on exportin 1 (XPO1/CRM1) to be shuttled into the cytoplasm and perform its anti-apoptotic function. Here, we explored the efficacy of Selective Inhibitors of Nuclear Export (SINE), KPT-251, KPT-276 and the orally available, clinical stage KPT-330 (selinexor), in DMPM preclinical models. Exposure to SINE induced dose-dependent inhibition of cell growth, cell cycle arrest at G1-phase and caspase-dependent apoptosis, which were consequent to a decrease of XPO1/CRM1 protein levels and the concomitant nuclear accumulation of its cargo proteins p53 and CDKN1a. Cell exposure to SINE led to a time-dependent reduction of cytoplasmic survivin levels. In addition, after an initial accumulation, the nuclear protein abundance progressively decreased, as a consequence of an enhanced ubiquitination and proteasome-dependent degradation. SINE and the survivin inhibitor YM155 synergistically cooperated in reducing DMPM cell proliferation. Most importantly, orally administered SINE caused a significant anti-tumor effect in subcutaneous and orthotopic DMPM xenografts without appreciable toxicity. Overall, we have demonstrated a marked efficacy of SINE in DMPM preclinical models that may relay on the interference with survivin intracellular distribution and function. Our study suggests SINE-mediated XPO1/CRM1 inhibition as a novel therapeutic option for DMPM.


Assuntos
Antineoplásicos/farmacologia , Proteínas Inibidoras de Apoptose/metabolismo , Carioferinas/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Mesotelioma/tratamento farmacológico , Neoplasias Peritoneais/tratamento farmacológico , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Acrilamidas/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Humanos , Hidrazinas/farmacologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Mesotelioma/metabolismo , Mesotelioma/patologia , Mesotelioma Maligno , Camundongos , Camundongos SCID , Proteínas de Neoplasias/metabolismo , Oxidiazóis/farmacologia , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/patologia , Reação em Cadeia da Polimerase em Tempo Real , Survivina , Tiazóis/farmacologia , Triazóis/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Proteína Exportina 1
6.
Int J Cancer ; 136(2): 299-309, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24866585

RESUMO

Because available treatments have limited efficacy in triple-negative breast cancer (TNBC), the identification of new therapeutic strategies to improve patients' outcome is urgently needed. In our study, we investigated the effects of the administration of the small molecule selective survivin suppressant YM155, alone or in association with CD34+ cells transduced with a replication-deficient adenovirus encoding the human tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene (CD34-TRAIL+ cells), in three TNBC cell models. YM155 exposure significantly impaired TNBC cell growth and selectively modulated survivin expression at both mRNA and protein level. In addition, co-culturing YM155-treated TNBC cells with CD34-TRAIL+ cells resulted in markedly increased cytotoxic effect and apoptotic response in comparison with single treatments. Such a chemosensitizing effect was observed only in TNBC cells inherently expressing DR5 and relied on the ability of YM155 to upregulate DR5 expression through a p38 MAPK- and CHOP-dependent mechanism. YM155/CD34-TRAIL+ combination also showed a significant inhibitory effect on the growth of DR5-expressing TNBC cells following xenotransplantation into NOD/SCID mice, in the absence of toxicity. Overall, our data (i) provide, for the first time, evidence that YM155 sensitizes TNBC cells to CD34-TRAIL+ cells-induced apoptosis by a mechanism involving the downregulation of survivin and the simultaneous p38 MAPK- and CHOP-mediated upregulation of DR5, and (ii) suggest the combination of YM155 with TRAIL-armed CD34+ progenitor cells as a promising therapeutic option for patients with TNBC expressing DR5.


Assuntos
Imidazóis/farmacologia , Naftoquinonas/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fator de Transcrição CHOP/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Membrana Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Feminino , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Proteínas Inibidoras de Apoptose/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Espécies Reativas de Oxigênio/metabolismo , Survivina , Neoplasias de Mama Triplo Negativas/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Breast Cancer Res ; 16(3): R55, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24886669

RESUMO

INTRODUCTION: The molecular determinants of breast cancer resistance to first-line anthracycline-containing chemotherapy are unknown. METHODS: We examined the response to doxorubicin of organotypic cultures of primary human breast tumors ex vivo with respect to cell proliferation, DNA damage and modulation of apoptosis. Samples were analyzed for genome-wide modulation of cell death pathways, differential activation of p53, and the role of survivin family molecules in drug resistance. Rational drug combination regimens were explored by high-throughput screening, and validated in model breast cancer cell types. RESULTS: Doxorubicin treatment segregated organotypic human breast tumors into distinct Responder or Non Responder groups, characterized by differential proliferative index, stabilization of p53, and induction of apoptosis. Conversely, tumor histotype, hormone receptor or human epidermal growth factor receptor-2 (HER2) status did not influence chemotherapy sensitivity. Global analysis of cell death pathways identified survivin and its alternatively spliced form, survivin-ΔEx3 as uniquely overexpressed in Non Responder breast tumors. Forced expression of survivin-ΔEx3 preserved cell viability and prevented doxorubicin-induced apoptosis in breast cancer cell types. High-throughput pharmacologic targeting of survivin family proteins with a small-molecule survivin suppressant currently in the clinic (YM155) selectively potentiated the effect of doxorubicin, but not other chemotherapeutics in breast cancer cell types, and induced tumor cell apoptosis. CONCLUSIONS: Survivin family proteins are novel effectors of doxorubicin resistance in chemotherapy-naive breast cancer. The incorporation of survivin antagonist(s) in anthracycline-containing regimens may have improved clinical activity in these patients.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Imidazóis/farmacologia , Proteínas Inibidoras de Apoptose/genética , Naftoquinonas/farmacologia , Processamento Alternativo , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/genética , Camptotecina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Dano ao DNA/efeitos dos fármacos , Doxorrubicina/farmacologia , Etoposídeo/farmacologia , Feminino , Ensaios de Triagem em Larga Escala , Humanos , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Células MCF-7 , Paclitaxel/farmacologia , Receptor ErbB-2/metabolismo , Survivina , Proteína Supressora de Tumor p53/genética
8.
J Clin Endocrinol Metab ; 99(2): 498-509, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24276455

RESUMO

CONTEXT: Tyrosine kinase inhibitors represent a new treatment option for patients with advanced medullary thyroid cancer (MTC). However, cures have not been achieved with current available agents used in monotherapy. OBJECTIVE: Because RET has been shown to negatively regulate CD95 death receptor activation in preclinical models of RET-dependent MTC, we investigated the potential of the combination approach with the RET-targeting tyrosine kinase inhibitor sunitinib and cisplatin to enhance apoptosis activation through the extrinsic pathway. DESIGN: The effects of sunitinib and cisplatin were examined in human MTC cell lines harboring oncogenic RET mutations. Experiments were designed to determine drug effects on RET signaling, cell growth, apoptosis, autophagy, and tumor growth in mice and to investigate the mechanisms of the drug interaction. RESULTS: Sunitinib and cisplatin synergistically inhibited the growth of MZ-CRC-1 cells harboring the RET M918T activating mutation. The combination enhanced apoptosis activation through CD95-mediated, caspase-8-dependent pathway. Moreover, sunitinib induced a severe perturbation of the autophagic flux characterized by autophagosome accumulation and a remarkable lysosomal dysfunction, which was further enhanced, with lysosomal leakage induction, by cisplatin. Administration of the drug combination to mice xenografted with MZ-CRC-1 cells improved the antitumor efficacy, as compared with single-agent treatments, inducing complete responses in 30% of the treated mice, a significant increase in caspase-3 activation (P < .01 vs cisplatin, and P < .0005 vs sunitinib) and apoptosis in tumor cells. CONCLUSIONS: Addition of cisplatin to sunitinib potentiates apoptotic cell death and has promising preclinical activity in MTCs harboring the RET M918T oncogene.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Medular/tratamento farmacológico , Cisplatino/farmacologia , Indóis/farmacologia , Pirróis/farmacologia , Neoplasias da Glândula Tireoide/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma Medular/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/uso terapêutico , Sinergismo Farmacológico , Humanos , Indóis/uso terapêutico , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Pirróis/uso terapêutico , Sunitinibe , Neoplasias da Glândula Tireoide/patologia
9.
Biochem Pharmacol ; 87(4): 579-97, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24370341

RESUMO

Compelling evidence suggests that epithelial-to-mesenchymal transition is involved in the resistance of human cancer cells to chemotherapy. We previously reported that the expression of miR-205, a miRNA down-regulated in prostate cancer, is further repressed in prostate cancer cells undergoing epithelial-to-mesenchymal transition, suggesting a possible involvement of the miRNA in the acquisition of the chemoresistant phenotype. In the present study, we show that miR-205 replacement in castration-resistant mesenchymal prostate cancer cells caused an enhancement of cisplatin cytotoxic activity in vitro and in vivo, as a consequence of autophagy impairment. Specifically, the constraints on the autophagic flux were associated to the miRNA-dependent down-regulation of the lysosome-associated proteins RAB27A and LAMP3. These findings suggest that miR-205-mediated impairment of the autophagic pathway may interfere with the detoxifying capabilities of prostate cancer cells in their attempt to cope with cisplatin-induced detrimental effects. Overall, our data indicate that (i) loss of miR-205 may indeed contribute to acquire mesenchymal tracts and concomitantly establish a permissive autophagic milieu that confers a chemotherapy resistant phenotype to prostate cancer cells, and (ii) strategies aimed at restoring miR-205 expression levels may represent a successful approach to overcome resistance of prostate cancer to platinum compounds.


Assuntos
Antineoplásicos/farmacologia , Autofagia/genética , Cisplatino/farmacologia , Citotoxinas/farmacologia , MicroRNAs/genética , Neoplasias da Próstata/genética , Animais , Antineoplásicos/uso terapêutico , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Citotoxinas/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Masculino , Camundongos , Camundongos SCID , Neoplasias da Próstata/tratamento farmacológico
10.
J Med Chem ; 56(17): 7060-72, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23919303

RESUMO

In this study, we describe the synthesis of new nortopsentin analogues, 1H-pyrrolo[2,3-b]pyridine derivatives and their biological effects in experimental models of diffuse malignant peritoneal mesothelioma (DMPM), a rare and rapidly fatal disease, poorly responsive to conventional therapies. The three most active compounds, 1f (3-[2-(5-fluoro-1-methyl-1H-indol-3-yl)-1,3-thiazol-4-yl]-1H-pyrrolo[2,3-b]pyridine), 3f (3-[2-(1H-indol-3-yl)-1,3-thiazol-4-yl]-1-methyl-1H-pyrrolo[2,3-b]pyridine), and 1l (3-[2-(5-fluoro-1-methyl-1H-indol-3-yl)-1,3-thiazol-4-yl]-1-methyl-1H-pyrrolo[2,3-b] pyridine), which were shown to act as cyclin-dependent kinase 1 inhibitors, consistently reduced DMPM cell proliferation and induced a caspase-dependent apoptotic response, with a concomitant reduction of the expression of the active Thr(34)-phosphorylated form of the antiapoptotic protein survivin. Moreover, the combined treatment of DMPM cells with 3f derivative and paclitaxel produced a synergistic cytotoxic effect, which was paralleled by an enhanced apoptotic response. In the mouse model, i.p. administration of 1f, 3f, and 1l derivatives was effective, resulting in a significant tumor volume inhibition of DMPM xenografts (range, 58-75%) at well-tolerated doses, and two complete responses were observed in each treatment group.


Assuntos
Antineoplásicos/farmacologia , Mesotelioma/tratamento farmacológico , Neoplasias Peritoneais/tratamento farmacológico , Piridinas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/uso terapêutico , Linhagem Celular , Humanos , Espectroscopia de Ressonância Magnética , Piridinas/síntese química , Piridinas/uso terapêutico , Espectrofotometria Ultravioleta
11.
Cancer Res ; 72(13): 3251-9, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22586065

RESUMO

Survivin is an oncogene that functions in cancer cell cytoprotection and mitosis. Here we report that differential expression in cancer cells of a C-terminal splice variant of survivin, termed survivin-ΔEx3, is tightly associated with aggressive disease and markers of unfavorable prognosis. In contrast to other survivin variants, survivin-ΔEx3 localized exclusively to nuclei in tumor cells and was phosphorylated at multiple residues by the checkpoint kinase Chk2 during DNA damage. Mutagenesis of the Chk2 phosphorylation sites enhanced the stability of survivin-ΔEx3 in tumor cells, inhibited the expression of phosphorylated H2AX (γH2AX) in response to double-strand DNA breaks, and impaired growth after DNA damage. DNA damage induced Chk2 phosphorylation, stabilization of p53, induction of the cyclin-dependent kinase inhibitor p21, and homologous recombination-induced repair were not affected. In vivo, active Chk2 was detected at the earliest stages of the colorectal adenoma-to-carcinoma transition, persisted in advanced tumors, and correlated with increased survivin expression. Together, our findings suggest that Chk2-mediated phosphorylation of survivin-ΔEx3 contributes to a DNA damage-sensing checkpoint that may affect cancer cell sensitivity to genotoxic therapies.


Assuntos
Dano ao DNA , Proteínas Inibidoras de Apoptose/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Quinase do Ponto de Checagem 2 , Primers do DNA , Humanos , Mutagênese , Neoplasias/patologia , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Frações Subcelulares/metabolismo , Survivina
12.
ChemMedChem ; 6(7): 1300-9, 2011 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-21523912

RESUMO

Given the potent antimicrobial, antiviral, and antitumor activities of many natural products, there is an increasing interest in the synthesis of new molecules based on natural compound scaffolds. Based on a 2,4-bis(3'-indolyl)imidazole skeleton, two new series of phenylthiazolylindoles and phenylthiazolyl-7-azaindoles were obtained by Hantzsch reaction between substituted phenylthioamides and the α-bromoacetyl derivatives. Some azaindole derivatives, tested at the National Cancer Institute against a panel of ∼60 tumor cell lines derived from nine human cancer cell types, showed inhibitory effects against all cell lines investigated at micromolar to nanomolar concentrations. Two of them exhibited a high affinity for CDK1, with IC(50) values of 0.41 and 0.85 µM. These promising results will set the foundation for future investigations into the development of anticancer therapies.


Assuntos
Antineoplásicos/síntese química , Indóis/química , Tiazóis/química , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Proteína Quinase CDC2/química , Proteína Quinase CDC2/metabolismo , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Indóis/síntese química , Indóis/uso terapêutico , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/uso terapêutico
13.
Cancer ; 115(13 Suppl): 3058-67, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19544547

RESUMO

Prostate cancer has a variable clinical outcome and, therefore, there is a clear need for novel molecular markers that are specifically associated with biologically aggressive disease to improve staging and prognostication and also to provide mechanistic information to facilitate treatment selection. Different candidate biomarkers have been identified that are linked to patient prognosis and/or response to specific treatments. Such molecules are involved in diverse cellular processes (including cell cycle regulation, cell death and apoptosis, signal transduction, cell adhesion, and angiogenesis) within which aberrant activity of several regulatory pathways has been seen in prostate cancer. Although the number of molecular markers continues to grow, mainly because of the advent of high-throughput methods, more work needs to be done to develop uniform standards for their characterization to enable comparison of markers across studies. Moreover, a rate-limiting step in the development of molecular markers is large-scale clinical assessment and their evaluation in the context of prediction model improvement. In fact, thus far, only a few studies have tested and demonstrated whether the addition of new biological markers improves the accuracy of standard clinical models (nomograms) in predicting biochemical progression in patients with clinically localized prostate cancer who underwent radical prostatectomy. Cancer 2009;115(13 suppl):3058-67. (c) 2009 American Cancer Society.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/terapia , Apoptose , Adesão Celular , Ciclo Celular , Epigênese Genética , Previsões , Humanos , Masculino , Neovascularização Patológica , Nomogramas , Prognóstico , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...