Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 24(17): 4256-4270, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29844128

RESUMO

Purpose: The DEAD-box RNA helicase eIF4A1 carries out the key enzymatic step of cap-dependent translation initiation and is a well-established target for cancer therapy, but no drug against it has entered evaluation in patients. We identified and characterized a natural compound with broad antitumor activities that emerged from the first target-based screen to identify novel eIF4A1 inhibitors.Experimental Design: We tested potency and specificity of the marine compound elatol versus eIF4A1 ATPase activity. We also assessed eIF4A1 helicase inhibition, binding between the compound and the target including binding site mutagenesis, and extensive mechanistic studies in cells. Finally, we determined maximum tolerated dosing in vivo and assessed activity against xenografted tumors.Results: We found elatol is a specific inhibitor of ATP hydrolysis by eIF4A1 in vitro with broad activity against multiple tumor types. The compound inhibits eIF4A1 helicase activity and binds the target with unexpected 2:1 stoichiometry at key sites in its helicase core. Sensitive tumor cells suffer acute loss of translationally regulated proteins, leading to growth arrest and apoptosis. In contrast to other eIF4A1 inhibitors, elatol induces markers of an integrated stress response, likely an off-target effect, but these effects do not mediate its cytotoxic activities. Elatol is less potent in vitro than the well-studied eIF4A1 inhibitor silvestrol but is tolerated in vivo at approximately 100× relative dosing, leading to significant activity against lymphoma xenografts.Conclusions: Elatol's identification as an eIF4A1 inhibitor with in vivo antitumor activities provides proof of principle for target-based screening against this highly promising target for cancer therapy. Clin Cancer Res; 24(17); 4256-70. ©2018 AACR.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Produtos Biológicos/farmacologia , Fator de Iniciação 4A em Eucariotos/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Compostos de Espiro/farmacologia , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Animais , Apoptose/efeitos dos fármacos , Organismos Aquáticos/química , Produtos Biológicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Fator de Iniciação 4A em Eucariotos/química , Fator de Iniciação 4A em Eucariotos/genética , Fibroblastos/efeitos dos fármacos , Xenoenxertos , Humanos , Camundongos , Modelos Moleculares , Neoplasias/genética , Biossíntese de Proteínas/efeitos dos fármacos , Proteômica , Compostos de Espiro/química
2.
J Med Chem ; 58(17): 6984-93, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-26305181

RESUMO

Prostate cancer (PC) is the second most prevalent cancer among men in Western societies, and those who develop metastatic castration-resistant PC (CRPC) invariably succumb to the disease. The need for effective treatments for CRPC is a pressing concern, especially due to limited durable responses with currently employed therapies. Here, we demonstrate the successful application of a high-throughput gene-expression profiling assay directly targeting genes of the androgen receptor pathway to screen a natural products library leading to the identification of 17ß-hydroxywithanolides 1-5, of which physachenolide D (5) exhibited potent and selective in vitro activity against two PC cell lines, LNCaP and PC-3. Epoxidation of 5 afforded physachenolide C (6) with higher potency and stability. Structure-activity relationships for withanolides as potential anti-PC agents are presented together with in vivo efficacy studies on compound 6, suggesting that 17ß-hydroxywithanolides are promising candidates for further development as CRPC therapeutics.


Assuntos
Androgênios/metabolismo , Antineoplásicos/química , Produtos Biológicos/química , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Vitanolídeos/química , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Xenoenxertos , Ensaios de Triagem em Larga Escala , Humanos , Calicreínas/genética , Calicreínas/metabolismo , Masculino , Camundongos SCID , Transplante de Neoplasias , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/metabolismo , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Receptores Androgênicos/metabolismo , Relação Estrutura-Atividade , Vitanolídeos/síntese química , Vitanolídeos/farmacologia
3.
Mol Cancer Ther ; 4(9): 1349-57, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16170026

RESUMO

Epidermal growth factor receptor (EGFR) inhibitors such as gefitinib show antitumor activity in a subset of non-small cell lung cancer (NSCLC) patients having mutated EGFR. Recent work shows that phosphatidylinositol-3-kinase (PI3-K) is coupled to the EGFR only in NSCLC cell lines expressing ErbB-3 and that EGFR inhibitors do not inhibit PI3-K signaling in these cells. The central role PI3-K plays in cell survival suggests that a PI3-K inhibitor offers a strategy to increase the antitumor activity of EGFR inhibitors in resistant NSCL tumors that do not express ErbB-3. We show that PX-866, a PI3-K inhibitor with selectivity for p110alpha, potentiates the antitumor activity of gefitinib against even large A-549 NSCL xenografts giving complete tumor growth control in the early stages of treatment. A-549 xenograft phospho-Akt was inhibited by PX-866 but not by gefitinib. A major toxicity of PX-866 administration was hyperglycemia with decreased glucose tolerance, which was reversed upon cessation of treatment. The decreased glucose tolerance caused by PX-866 was insensitive to the AMP-activated protein kinase inhibitor metformin but reversed by insulin and by the peroxisome proliferator-activated receptor-gamma activator pioglitazone. Prolonged PX-866 administration also caused increased neutrophil counts. Thus, PX-866, by inhibiting PI3-K signaling, may have clinical use in increasing the response to EGFR inhibitors such as gefitinib in patients with NSCLC and possibly in other cancers who do not respond to EGFR inhibition.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/antagonistas & inibidores , Gonanos/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Quinazolinas/farmacologia , Animais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Inibidores Enzimáticos/farmacologia , Gefitinibe , Teste de Tolerância a Glucose , Humanos , Hiperglicemia/etiologia , Hipoglicemiantes/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Metformina/farmacologia , Camundongos , Camundongos SCID , Neutrófilos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Pioglitazona , Tiazolidinedionas/farmacologia , Transplante Heterólogo , Células Tumorais Cultivadas
4.
Mol Cancer Ther ; 3(7): 763-72, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15252137

RESUMO

We have developed biologically stable semisynthetic viridins as inhibitors of phosphoinositide (PtdIns)-3-kinases. The most active compound was PX-866 (acetic acid (1S,4E,10R,11R,13S,14R)-[4-diallylaminomethylene-6-hydroxy-1-methoxymethyl-10,13-dimethyl-3,7,17-trioxo-1,3,4,7,10,11,12,13,14,15,16,17-dodecahydro-2-oxa-cyclopenta[a]phenanthren-11-yl ester), which inhibited purified PtdIns-3-kinase with an IC50 of 0.1 nmol/L and PtdIns-3-kinase signaling measured by phospho-Ser473-Akt levels in HT-29 colon cancer cells with an IC50 of 20 nmol/L. PX-866 administered to mice at 10 mg/kg inhibited phospho-Ser473-Akt in HT-29 colon tumor xenografts up to 80% with recovery taking >48 hours after p.o. administration but more rapidly after i.v. or i.p. administration. PX-866 was eliminated from mouse plasma with a half-life of 18 minutes and a clearance of 360 mL/min/kg following i.v. administration and, when administered i.p. or p.o., showed first-pass metabolism with sequential N-deallylation. Synthetic standards of the N-deallylated metabolites of PX-866 inhibited PtdIns-3-kinase at low nanomolar per liter concentrations. PX-866 exhibited in vivo antitumor activity against s.c. OvCar-3 human ovarian cancer and A-549 human lung cancer xenografts in immunodeficient mice with log cell kills up to 1.2. PX-866 also increased the antitumor activity of cisplatin against A-549 xenografts and radiation treatment against OvCar-3 xenografts. The results show that PX-866 is a biologically stable broad-spectrum PtdIns-3-kinase inhibitor with good pharmacokinetics that causes prolonged inhibition of PtdIns-3-kinase signaling in human tumor xenografts. PX-866 exhibits single agent in vivo antitumor activity and increases the antitumor effects of cisplatin and radiation treatment.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Gonanos/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Androstadienos/sangue , Androstadienos/farmacologia , Androstadienos/toxicidade , Androstenos/sangue , Androstenos/farmacologia , Androstenos/toxicidade , Animais , Anticorpos Fosfo-Específicos/imunologia , Antineoplásicos/química , Bacteriocinas/sangue , Bacteriocinas/farmacologia , Bacteriocinas/toxicidade , Linhagem Celular Tumoral , Cisplatino/farmacologia , Neoplasias do Colo/enzimologia , Inibidores Enzimáticos/química , Feminino , Gonanos/química , Humanos , Neoplasias Pulmonares/enzimologia , Camundongos , Camundongos SCID , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/radioterapia , Proteínas Serina-Treonina Quinases/análise , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/imunologia , Proteínas Proto-Oncogênicas c-akt , Wortmanina , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Mol Cancer Ther ; 3(3): 233-44, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15026543

RESUMO

The hypoxia-inducible factor-1 (HIF-1) transcription factor is an important regulator of tumor response to hypoxia that include increased angiogenesis, glycolytic metabolism, and resistance to apoptosis. HIF-1 activity is regulated by the availability of the HIF-1alpha subunit, the levels of which increase under hypoxic conditions. PX-478 (S-2-amino-3-[4'-N,N,-bis(2-chloroethyl)amino]phenyl propionic acid N-oxide dihydrochloride) is an inhibitor of constitutive and hypoxia-induced HIF-1alpha levels and thus HIF-1 activity. We report that PX-478 given to mice suppresses HIF-1alpha levels in HT-29 human colon cancer xenografts and inhibits the expression of HIF-1 target genes including vascular endothelial growth factor and the glucose transporter-1. PX-478 shows antitumor activity against established (0.15-0.40 cm(3)) human tumor xenografts with cures of SHP-77 small cell lung cancer and log cell kills up to 3.0 for other tumors including HT-29 colon, PC-3 prostate, DU-145 prostate, MCF-7 breast, Caki-1 renal, and Panc-1 pancreatic cancers. Large (0.83 cm(3)) PC-3 prostate tumors showed 64% regression, which was greater than for smaller tumors. The antitumor response to PX-478 was positively correlated with tumor HIF-1alpha levels (P < 0.02) and was accompanied by massive apoptosis. The results show that PX-478 is an inhibitor of HIF-1alpha and HIF-1 transcription factor activity in human tumor xenografts and has marked antitumor activity against even large tumor xenografts, which correlates positively with HIF-1alpha levels.


Assuntos
Antineoplásicos/farmacocinética , Compostos de Mostarda/farmacocinética , Neoplasias/tratamento farmacológico , Fenilpropionatos/farmacocinética , Fatores de Transcrição/antagonistas & inibidores , Animais , Apoptose , Área Sob a Curva , Western Blotting , Linhagem Celular Tumoral , Eritropoetina/sangue , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Proteínas de Transporte de Monossacarídeos/metabolismo , Compostos de Mostarda/química , Transplante de Neoplasias , Fenilpropionatos/química , Fatores de Tempo , Fatores de Transcrição/química , Fator A de Crescimento do Endotélio Vascular/sangue , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Clin Cancer Res ; 9(13): 4961-71, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-14581371

RESUMO

PURPOSE: The antiestrogen tamoxifen (Tam) has been used as therapy against estrogen receptor (ER)-positive breast cancer for decades. Most tumors respond initially, but resistance frequently develops. The ER exists in a multiprotein complex containing the molecular chaperone heat shock protein (Hsp) 90, which is known to regulate the stability and activity of this receptor. Therefore, we investigated a ligand-independent approach to hormonal therapy that depletes cellular levels of the receptor by inhibiting the function of Hsp90. EXPERIMENTAL DESIGN: The activity of the Hsp90 inhibitor geldanamycin (GA) and its clinically relevant derivative, 17-allylamino-17-demethoxygeldanamycin (17AAG), was examined at the molecular and cellular levels using Tam-resistant MCF-7 breast cancer cells both in vitro and in tumor xenografts. RESULTS: The ER was depleted by GA in several Tam-resistant cell lines, as were other Hsp90 client proteins such as Akt and Raf-1. Unexpectedly, Tam inhibited ER depletion by GA but had no effect on destabilization of Akt or Raf-1. When SCID mice supplemented with Tam were treated with 17AAG, their tumors also showed no decrease in ER levels as measured by immunofluorescent staining and laser scanning cytometry. In these same tumors, however, decreased Akt and Raf-1 levels were observed. Drug administration also led to inhibition of tumor xenograft growth. The mechanism by which Tam inhibits GA-mediated ER depletion is unclear, but immunoprecipitation experiments showed that Tam does not inhibit the ability of GA to alter the ER-chaperone complex. CONCLUSIONS: Based on its ability to deplete the ER as well as other critical signaling molecules in Tam-resistant breast cancer, 17AAG may provide a useful alternative treatment for patients with recurrent, hormone-refractory breast cancer that should be explored further in Phase II trials. In this context, combined treatment with 17AAG and Tam should be avoided because Tam may inhibit the ability of 17AAG to deplete the ER, potentially reducing its anticancer activity.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Rifabutina/análogos & derivados , Animais , Antibióticos Antineoplásicos/farmacologia , Benzoquinonas , Linhagem Celular Tumoral , Colágeno/farmacologia , Combinação de Medicamentos , Estrogênios/metabolismo , Genes Reporter , Proteínas de Choque Térmico HSP90/metabolismo , Hormônios/metabolismo , Humanos , Immunoblotting , Lactamas Macrocíclicas , Laminina/farmacologia , Lasers , Ligantes , Camundongos , Camundongos SCID , Microscopia Confocal , Microscopia de Fluorescência , Transplante de Neoplasias , Testes de Precipitina , Proteoglicanas/farmacologia , Quinonas/farmacologia , Receptores de Estrogênio/metabolismo , Rifabutina/farmacologia , Transdução de Sinais , Fatores de Tempo , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...