Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Chem Biol Drug Des ; 101(3): 727-739, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36334047

RESUMEN

The identification of biologically active target compounds and their binding proteins is important in mechanism-of-action studies for drug development. Additionally, the newly discovered binding proteins provide unforeseen ideas for novel drug discovery and for subsequent structural transformation to improve target specificity. Based on the lead and final candidate compounds related to the type 5 phosphodiesterase (PDE5) inhibitor E4021, we designed chemical probes and identified their target proteins by the affinity chromatography approach. Aldehyde dehydrogenase family 1 member A3 (ALDH1A3), currently reported as a cancer stem cell target, was clearly isolated as a binding protein of the lead 'immature' inhibitor probe against PDE5. In the early derivatization to the closely related structure, Compound 5 (ER-001135935) was found to significantly inhibit ALDH1A3 activity. The discovery process of a novel ALDH1A3-selective inhibitor with affinity-based binder identification is described, and the impact of this identification method on novel drug discovery is discussed.


Asunto(s)
Aldehído Oxidorreductasas , Inhibidores de Fosfodiesterasa , Aldehído Oxidorreductasas/metabolismo , Células Madre Neoplásicas/metabolismo , Descubrimiento de Drogas
2.
Nat Chem Biol ; 13(6): 675-680, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28437394

RESUMEN

Target-protein degradation is an emerging field in drug discovery and development. In particular, the substrate-receptor proteins of the cullin-ubiquitin ligase system play a key role in selective protein degradation, which is an essential component of the anti-myeloma activity of immunomodulatory drugs (IMiDs), such as lenalidomide. Here, we demonstrate that a series of anticancer sulfonamides NSC 719239 (E7820), indisulam, and NSC 339004 (chloroquinoxaline sulfonamide, CQS) induce proteasomal degradation of the U2AF-related splicing factor coactivator of activating protein-1 and estrogen receptors (CAPERα) via CRL4DCAF15 mediated ubiquitination in human cancer cell lines. Both CRISPR-Cas9-based knockout of DCAF15 and a single amino acid substitution of CAPERα conferred resistance against sulfonamide-induced CAPERα degradation and cell-growth inhibition. Thus, these sulfonamides represent selective chemical probes for disrupting CAPERα function and designate DCAFs as promising drug targets for promoting selective protein degradation in cancer therapy.


Asunto(s)
Indoles/farmacología , Proteínas Nucleares/metabolismo , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Sulfonamidas/metabolismo , Antineoplásicos/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteolisis/efectos de los fármacos , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Sulfonamidas/farmacología
3.
Mol Cancer Ther ; 15(6): 1208-16, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27196783

RESUMEN

Apratoxin A is a natural product with potent antiproliferative activity against many human cancer cell lines. However, we and other investigators observed that it has a narrow therapeutic window in vivo Previous mechanistic studies have suggested its involvement in the secretory pathway as well as the process of chaperone-mediated autophagy. Still the link between the biologic activities of apratoxin A and its in vivo toxicity has remained largely unknown. A better understanding of this relationship is critically important for any further development of apratoxin A as an anticancer drug. Here, we describe a detailed pathologic analysis that revealed a specific pancreas-targeting activity of apratoxin A, such that severe pancreatic atrophy was observed in apratoxin A-treated animals. Follow-up tissue distribution studies further uncovered a unique drug distribution profile for apratoxin A, showing high drug exposure in pancreas and salivary gland. It has been shown previously that apratoxin A inhibits the protein secretory pathway by preventing cotranslational translocation. However, the molecule targeted by apratoxin A in this pathway has not been well defined. By using a (3)H-labeled apratoxin A probe and specific Sec 61α/ß antibodies, we identified that the Sec 61 complex is the molecular target of apratoxin A. We conclude that apratoxin A in vivo toxicity is likely caused by pancreas atrophy due to high apratoxin A exposure. Mol Cancer Ther; 15(6); 1208-16. ©2016 AACR.


Asunto(s)
Antineoplásicos/toxicidad , Depsipéptidos/toxicidad , Neoplasias/tratamiento farmacológico , Páncreas/efectos de los fármacos , Canales de Translocación SEC/metabolismo , Células A549 , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Depsipéptidos/farmacocinética , Humanos , Células MCF-7 , Dosis Máxima Tolerada , Ratones , Trasplante de Neoplasias , Neoplasias/metabolismo , Especificidad de Órganos , Unión Proteica , Ratas
4.
Cancer Biol Ther ; 16(4): 589-601, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25729885

RESUMEN

Natural compound schweinfurthins are of considerable interest for novel therapy development because of their selective anti-proliferative activity against human cancer cells. We previously reported the isolation of highly active schweinfurthins E-H, and in the present study, mechanisms of the potent and selective anti-proliferation were investigated. We found that schweinfurthins preferentially inhibited the proliferation of PTEN deficient cancer cells by indirect inhibition of AKT phosphorylation. Mechanistically, schweinfurthins and their analogs arrested trans-Golgi-network trafficking, an intracellular vesicular trafficking system, resulting in the induction of endoplasmic reticulum stress and the suppression of both lipid raft-mediated PI3K activation and mTOR/RheB complex formation, which collectively led to an effective inhibition of mTOR/AKT signaling. The trans-Golgi-network traffic arresting effect of schweinfurthins was associated with their in vitro binding activity to oxysterol-binding proteins that are known to regulate intracellular vesicular trafficking. Moreover, schweinfurthins were found to be highly toxic toward PTEN-deficient B cell lymphoma cells, and displayed 2 orders of magnitude lower activity toward normal human peripheral blood mononuclear cells and primary fibroblasts in vitro. These results revealed a previously unrecognized role of schweinfurthins in regulating trans-Golgi-network trafficking, and linked mechanistically this cellular effect with mTOR/AKT signaling and with cancer cell survival and growth. Our findings suggest the schweinfurthin class of compounds as a novel approach to modulate oncogenic mTOR/AKT signaling for cancer treatment.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Estilbenos/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Red trans-Golgi/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Linfoma de Células B/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
5.
Bioorg Med Chem ; 22(19): 5513-29, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25139751

RESUMEN

Hypoxia-inducible factor-1 (HIF-1) is the chief transcription factor regulating hypoxia-driven gene expression. HIF-1 overexpression is associated with poor prognosis in several cancers and therefore represents an attractive target for novel antitumor agents. We explored small molecule inhibitors of the HIF-1 pathway. Using high-throughput-screening, we identified benzanilide compound 1 (IC50=560 nM) as a seed. Subsequent extensive derivatization led to the discovery of compounds 43a and 51d, with anti-HIF-1 activities in vitro (IC50=21 and 0.47 nM, respectively), and in vivo. Additionally, 43a (12.5-100mg/kg) also displayed in vivo anti-tumor efficacy, without influencing body weight.


Asunto(s)
Anilidas/química , Anilidas/farmacología , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Anilidas/administración & dosificación , Anilidas/síntesis química , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Ensayos Analíticos de Alto Rendimiento , Humanos , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/síntesis química , Relación Estructura-Actividad
6.
Int J Mol Med ; 29(4): 541-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22211243

RESUMEN

Hypoxia-inducible factor 1 (HIF-1) activates the transcription of genes that play crucial roles in the adaptation of cancer cells to hypoxia. HIF-1α overexpression has been associated with poor prognosis in patients with various types of cancer. Here, we describe ER-400583-00 as a novel HIF-1 inhibitor. ER-400583-00 suppressed the production of HIF-1α protein in response to hypoxia, with a half-maximal inhibitory concentration value of 3.7 nM in human U251 glioma cells. The oral administration of 100 mg/kg ER-400583-00 to mice bearing U251 tumor xenografts resulted in a rapid suppression of HIF-1α that persisted for 24 h. Immunohistochemical analysis revealed that ER-400583-00 suppressed the proliferation of cancer cells most prominently in areas distal to the region of blood perfusion, where HIF-1α-expressing hypoxic cancer cells were located. These hypoxic cancer cells were resistant to radiation therapy. ER-400583-00 showed a synergistic interaction with radiation therapy in terms of antitumor activity. These data suggest that HIF-1 blockade by small compounds may have therapeutic value in cancer, especially in combination with radiation therapy.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Glioma/tratamiento farmacológico , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Piperazinas/farmacología , Administración Oral , Animales , Antineoplásicos/uso terapéutico , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Glioma/radioterapia , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto
7.
FEBS J ; 278(24): 4870-80, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21981285

RESUMEN

Pladienolide is a naturally occurring macrolide that binds to the SF3b complex to inhibit mRNA splicing. It has not been fully validated whether the splicing impairment is a relevant mechanism for the potent antitumor activity of pladienolide. We established pladienolide-resistant clones from WiDr and DLD1 colorectal cancer cells that were insensitive to the inhibitory action of pladienolide on cell proliferation and splicing. An mRNA-Seq differential analysis revealed that these two cell lines have an identical mutation at Arg1074 in the gene for SF3B1, which encodes a subunit of the SF3b complex. Reverse expression of the mutant protein transferred pladienolide resistance to WiDr cells. Furthermore, immunoprecipitation analysis using a radiolabeled probe showed that the mutation impaired the binding affinity of paldienolide to its target. These results clearly demonstrate that pladienolide exerts its potent activity by targeting SF3b and also suggest that inhibition of SF3b is a promising drug target for anticancer therapy.


Asunto(s)
Compuestos Epoxi/farmacología , Macrólidos/farmacología , Fosfoproteínas/efectos de los fármacos , Ribonucleoproteína Nuclear Pequeña U2/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Compuestos Epoxi/metabolismo , Humanos , Macrólidos/metabolismo , Fosfoproteínas/metabolismo , Empalme del ARN/efectos de los fármacos , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2/metabolismo
9.
Nat Chem Biol ; 3(9): 570-5, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17643112

RESUMEN

Pladienolide is a naturally occurring antitumor macrolide that was discovered by using a cell-based reporter gene expression assay controlled by the human vascular endothelial growth factor promoter. Despite the unique mechanisms of action and prominent antitumor activities of pladienolides B and D in diverse in vitro and in vivo systems, their target protein has remained unclear. We used 3H-labeled, fluorescence-tagged and photoaffinity/biotin (PB)-tagged 'chemical probes' to identify a 140-kDa protein in splicing factor SF3b as the binding target of pladienolide. Immunoblotting of an enhanced green fluorescent protein fusion protein of SF3b subunit 3 (SAP130) revealed direct interaction between the PB probe and SAP130. The binding affinities of pladienolide derivatives to the SF3b complex were highly correlated with their inhibitory activities against reporter gene expression and cell proliferation. Furthermore, pladienolide B impaired in vivo splicing in a dose-dependent manner. Our results demonstrate that the SF3b complex is a pharmacologically relevant protein target of pladienolide and suggest that this splicing factor is a potential antitumor drug target.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Epoxi/farmacología , Macrólidos/farmacología , Proteínas de Unión al ARN/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Genes Reporteros , Humanos , Unión Proteica , Empalme del ARN/efectos de los fármacos , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2
10.
J Antibiot (Tokyo) ; 60(6): 364-9, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17617693

RESUMEN

Pladienolide B is a 12-membered macrolide isolated from Streptomyces platensis Mer-11107. It showed potent in vitro and in vivo antitumor activities and is a potential lead for novel antitumor agents. The absolute configurations at ten chiral centers were determined on the basis of spectral data of pladienolide B and its chemical transformation products.


Asunto(s)
Compuestos Epoxi/química , Macrólidos/química , Antineoplásicos/química , Modelos Moleculares , Conformación Molecular , Resonancia Magnética Nuclear Biomolecular , Espectrometría de Masa por Ionización de Electrospray , Estereoisomerismo , Streptomyces/química
12.
Chem Pharm Bull (Tokyo) ; 52(9): 1071-81, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15340192

RESUMEN

As part of a series of studies to discover new topoisomerase II inhibitors, novel pyrimidoacridones, pyrimidophenoxadines, and pyrimidocarbazoles were synthesized, and in vitro and in vivo antitumor activities and DNA-protein and/or DNA-topoisomerase II cross-linking activity as an indicator of topoisomerase II-DNA cleavable complex formation were evaluated. The pyrimidocarbazoles possessed high in vitro and in vivo potencies. Compound 26 (ER-37326), 8-acetyl-2-[2-(dimethylamino)ethyl]-1H-pyrimido[5,6,1-jk]carbazole-1,3(2H)-dione, showed in vitro growth inhibitory activity with respective IC(50) values of 0.049 microM and 0.35 microM against mouse leukemia P388 and human oral cancer KB. In vivo, this compound inhibited the tumor growth of mouse sarcoma M5076 implanted into mice with T/C values of 42% and 13% at 3.13 and 6.25 mg/kg/d respectively without significantly affecting the body weight. In addition, compound 26 (ER-37326) increased the formation of DNA-topoisomerase II cross-linking in P388 cells.


Asunto(s)
Acridinas/síntesis química , Antineoplásicos/síntesis química , Carbazoles/síntesis química , Pirimidinonas/síntesis química , Inhibidores de Topoisomerasa II , Acridinas/farmacología , Animales , Antineoplásicos/farmacología , Carbazoles/farmacología , Línea Celular Tumoral , ADN-Topoisomerasas de Tipo II/química , Relación Dosis-Respuesta a Droga , Humanos , Células KB , Leucemia P388/tratamiento farmacológico , Ratones , Estructura Molecular , Pirimidinas/síntesis química , Pirimidinas/farmacología , Pirimidinonas/farmacología
13.
Cancer Sci ; 94(1): 119-24, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12708485

RESUMEN

We have discovered a novel topoisomerase II (topo II) poison, ER-37328 (12,13-dihydro-5-[2-(dimethylamino)ethyl]-4H-benzo[c]pyrimido[5,6,1-jk]carbazole-4,6,10(5H,11H)-trione hydrochloride), which shows potent tumor regression activity against Colon 38 cancer inoculated s.c. Here, we describe studies on the cell-killing activity against a panel of human cancer cell lines and the antitumor activity of ER-37328 against human tumor xenografts. In a cell-killing assay involving 1-h drug treatment, ER-37328 showed more potent cell-killing activity (50% lethal concentrations (LC50s) ranging from 2.9 to 20 microM) than etoposide (LC50s>60 microM) against a panel of human cancer cell lines. ER-37328 induced double-stranded DNA cleavage, an indicator of topo II-DNA cleavable complex formation, within 1 h in MX-1 cells, and the extent of cleavage showed a bell-shaped relationship to drug concentration, with the maximum at 2.5 microM. After removal of the drug (2.5 microM) at 1 h, incubation was continued in drug-free medium, and the amount of cleaved DNA decreased. However, at 10 microM, which is close to the LC50s against MX-1 cells, DNA cleavage was not detected immediately after 1-h treatment, but appeared and increased after drug removal. This result may explain the potent cell-killing activity of ER-37328 in the 1-h treatment. In vivo, ER-37328 showed potent tumor regression activity against MX-1 and NS-3 tumors. Moreover, ER-37328 had a different antitumor spectrum from irinotecan or cisplatin against human tumor xenografts. In conclusion, ER-37328 is a promising topo II poison with strong cell killing activity in vitro and tumor regression activity in vivo, and is a candidate for the clinical treatment of malignant solid tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Carbazoles/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Proteínas de Neoplasias/antagonistas & inhibidores , Pirimidinas/uso terapéutico , Inhibidores de Topoisomerasa II , Adenocarcinoma/patología , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Carbazoles/farmacología , Neoplasias del Colon/patología , Daño del ADN , ADN de Neoplasias/análisis , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Etopósido/uso terapéutico , Femenino , Humanos , Neoplasias Pulmonares/patología , Sustancias Macromoleculares , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Unión Proteica/efectos de los fármacos , Pirimidinas/farmacología , Neoplasias Gástricas/patología , Células Tumorales Cultivadas/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Mol Cancer Ther ; 1(3): 169-75, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12467211

RESUMEN

DNA topoisomerase II has been shown to be an important therapeutic target in cancer chemotherapy. Here, we describe studies on the antitumor activity of a novel topoisomerase II inhibitor, ER-37328 [12,13-dihydro-5-[2-(dimethylamino)ethyl]-4H-benzo[c]pyrimido[5,6,1- jk]carbazole-4,6,10(5H,11H)-trione hydrochloride]. ER-37328 inhibited topoisomerase II activity at 10 times lower concentration than etoposide in relaxation assay and induced double-strand DNA cleavage within 1 h in murine leukemia P388 cells, in a bell-shaped manner with respect to drug concentration. The maximum amount of DNA cleavage was obtained at 2 microM. Like etoposide, ER-37328 (2 microM) induced topoisomerase II-DNA cross-linking in P388 cells. A spectroscopic study of ER-37328 mixed with DNA demonstrated that ER-37328 has apparent binding activity to DNA. ER-37328 showed potent growth-inhibitory activity against a panel of 21 human cancer cell lines [mean (50% growth-inhibitory concentration) GI50 = 59 nM]. COMPARE analysis according to the National Cancer Institute screening protocol showed that the pattern of the growth-inhibitory effect of ER-37328 was similar to that of etoposide, but different from that of doxorubicin. Studies on etoposide-, amsacrine [4'-(9-acridinylamino)methanesulfon-m-anisidide (m-AMSA)]-, and camptothecin-resistant P388 cell lines showed that: (a) etoposide- and m-AMSA-resistant P388 cell lines were partially resistant to ER-37328 compared with the parental cell line; and (b) a camptothecin-resistant cell line showed no cross-resistance to ER-37328. In addition, ER-37328 overcame P-glycoprotein-mediated resistance. In vivo, ER-37328 produced potent tumor regression of Colon 38 carcinoma inoculated s.c., and its activity was superior to that of etoposide or doxorubicin. These results indicate that ER-37328 inhibits topoisomerase II activity through the formation of topoisomerase II-DNA cleavable complex and has potent antitumor activity both in vitro and in vivo.


Asunto(s)
Antineoplásicos/farmacología , Carbazoles/farmacología , Neoplasias del Colon/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Pirimidinas/farmacología , Inhibidores de Topoisomerasa II , Amsacrina/farmacología , Animales , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Reactivos de Enlaces Cruzados , ADN de Neoplasias/efectos de los fármacos , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Etopósido/farmacología , Femenino , Humanos , Técnicas In Vitro , Leucemia P388/tratamiento farmacológico , Leucemia P388/enzimología , Leucemia P388/patología , Ratones , Ratones Endogámicos C57BL , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/enzimología , Células Tumorales Cultivadas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...