Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Cancer Res ; 5(8): 2387-95, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26396914

RESUMEN

OBJECTIVE: This study aimed to investigate the biological effects of "combi-targeting" JDF12 and its effect on the DNA repair pathway in hormone-refractory prostate cancer (HRPC). METHODS: HRPC cell lines (PC3 cells and VCap cells) were treated with JDF12 at different concentrations, and SRB method was employed to detect the proliferation of HRPC cells; Annexin V-FITC kit was used to detect the apoptosis of PC3 cells; Alkaline comet assay was performed to detect DNA damage; Western blot assay was done to detect the expressions of autophosphorylated EGFR, XRCC1 and ERCC1 (later two are proteins in DNA repair pathway); the anti-tumor effect was evaluated in nude mice inoculated with PC3 cells. RESULTS: JDF12 could inhibit the proliferation of PC3 cells and VCap cells in a concentration dependent manner (IC50: 14.04 ± 1.22 for PC3 and 15.57 ± 1.13 for VCap) and significantly increase the apoptotic cells as compared to those treated with mitozolomide or iressa alone. In PC3 cells, JDF12 induced DNA damage and also inhibited the expressions of phosphorylated EGFR, XRCC1 and ERCC1 in a concentration dependent manner. Moreover, JDF12 markedly inhibited tumor growth in nude mice. CONCLUSION: The novel "combi-targeting" JDF12 may exert more potent anti-proliferative effect as compared to mitozolomide or iressa alone, and the inhibitory effect on the EGFR signaling pathway and down-regulated XRCC1 and ERCC1 expressions may be ascribed to the JDF12 induced DNA damage.

2.
Prostate ; 72(12): 1273-85, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22290742

RESUMEN

PURPOSE: At the preclinical stage, mitozolomide (MTZ) showed exciting preclinical activity but failed later in clinical trial due to toxic side effects. We surmised that by targeting MTZ to epidermal growth factor receptor (EGFR), we may not only alter its toxicity profile, but also enhance its potency in EGFR-overexpressing tumors. To test this hypothesis, we designed JDF12, studied its mechanism of action in human prostate cancer (PCa) cells and determined its potency in vivo. EXPERIMENTAL DESIGN: To analyze its mixed EGFR-DNA targeting potential, we performed an enzyme linked immunosorbent assay (ELISA) and western blotting analysis of EGFR phosphorylation in cells stimulated with EGF. DNA damage was analyzed using the comet assay, and apoptosis quantitated by annexin V binding assay. Growth inhibition in vitro was determined by the sulforhodamine B (SRB) assay and in vivo efficacy analyzed in male CD-1 nude mice. RESULTS: The results showed that: Under physiological conditions, JDF12 was hydrolyzed to JDF04R and both agents were capable of inhibiting isolated EGFR tyrosine kinase (TK) and EGFR phosphorylation in EGF-stimulated cells. JDF12 significantly damaged DNA, induced apoptosis in DU145 cells and was up to 2-10-fold more potent than equieffective combinations of MTZ and JDF04R or Iressa in a panel that also included LNCaP and its EGFR and ErbB2 transfectants. In vivo, it induced significant antitumor activity in a DU145 xenograft model. CONCLUSIONS: The results suggest that the superior cytotoxicity of JDF12 when compared with MTZ and JDF04R may be imputed to its potent EGFR-DNA targeting properties and confirm the ability of this novel strategy to confer EGFR targeting properties to a classical alkylator.


Asunto(s)
Antineoplásicos Alquilantes/administración & dosificación , ADN/antagonistas & inhibidores , Sistemas de Liberación de Medicamentos/métodos , Compuestos de Mostaza Nitrogenada/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , ADN/metabolismo , Humanos , Masculino , Ratones , Células 3T3 NIH , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal/fisiología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
3.
J Pharm Biomed Anal ; 56(3): 592-9, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21764236

RESUMEN

ZR2003 is a type II of combi-molecule designed to target DNA and the epidermal growth factor receptor (EGFR) without requirement for hydrolysis. In human tumour cell lines cultured as monolayers, it showed 6.5-35 fold greater activity than Iressa. Further evaluation in 3D organ-like multilayer aggregates showed that it could block proliferation at submicromolar level. However, despite the superior potency of ZR2003 over Iressa in vitro, its activity xenograft models was not significantly different from that of Iressa. To rationalize these results, we determined the tumour concentration of both ZR2003 and Iressa in vivo and more importantly in vitro in multicellular aggregates. The results showed that in A431 and 4T1 xenografts, the level of ZR2003 absorbed in the tumours were consistently 2-fold less than those generated by Iressa. Likewise, in the multicellular aggregates model, the penetration of ZR2003 was consistently lower than Iressa. In serum containing media, the level of extractable or free ZR2003 was also inferior to those of Iressa. The results from this bioanalytical study, suggest that the discrepancy between the in vitro and in vivo potency of ZR2003 when compared with Iressa, may be imputed to its significantly lower tumour concentration.


Asunto(s)
Antinematodos/farmacología , ADN/antagonistas & inhibidores , ADN/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Neoplasias/tratamiento farmacológico , Compuestos de Mostaza Nitrogenada/farmacología , Quinazolinas/farmacología , Triazenos/farmacología , Animales , Antinematodos/farmacocinética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Daño del ADN , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Masculino , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/patología , Compuestos de Mostaza Nitrogenada/farmacocinética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Quinazolinas/farmacocinética , Neoplasias de la Vulva/tratamiento farmacológico , Neoplasias de la Vulva/enzimología , Neoplasias de la Vulva/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
4.
Drug Metab Lett ; 5(2): 141-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21457144

RESUMEN

Combi-molecules are agents designed to block receptors on their own and to further degrade to bioactive agents. Here we studied the fate of a novel combi-molecule of triazene class termed "ZRS1" in biological medium using multilayer aggregates and mouse tumour models. ZRS1 is a second generation derivative of RB107, a prodrug designed to release an EGFR inhibitor FD105 plus a methyl diazonium species. RB107 contains an acetoxymethyl function that is hydrolyzed too rapidly to generate BJ2000, a monoalkyltriazene that further degrades to FD105 and DNA alkylating methyldiazonium species. Recently, in order to prevent rapid hydrolysis of the acetoxymethylene function in the absence of cells and to delay the release of BJ2000, we designed ZRS1 that contains a more stable acetoxymethyl carbamate function. The results showed that ZRS1 was more stable than RB107 in cell culture medium supplemented with serum, with a rather long half life (>2 h). However, in an experiment where it was allowed to degrade in multilayer aggregates of ovarian cancer cells OV90, it rapidly released BJ2000 and its corresponding metabolite FD105, both in the medium and the multilayer aggregates. Interestingly, the intact ZRS1 could be detected in the multilayer aggregates with a T(max) around 10 min. Studies in vivo, in human DU145 prostate cancer xenograft model, revealed that ZRS1 blocked tumour growth and released FD105 and its acetylated metabolite FD105Ac, the latter being the major metabolite. Likewise, time course analysis in 4T1 mouse syngeneic breast cancer model showed a rapid release of FD105 and FD105Ac in the plasma and in the tumours. In summary, ZRS1 appeared as a good prodrug of the stable EGFR inhibitory metabolites FD105 and FD105Ac. Its ability to generate high concentrations of FD105Ac, a more potent EGFR inhibitor as is its major metabolite, is significant over previous methylating combi-molecules. Furthermore, this study showed that multilayer OV90 aggregates could be developed as an effective model to predict the stability and degradation of ZRS1 in vivo.


Asunto(s)
Carbamatos/farmacocinética , Receptores ErbB/antagonistas & inhibidores , Profármacos/farmacocinética , Quinazolinas/farmacocinética , Triazenos/farmacocinética , Animales , Agregación Celular , Línea Celular Tumoral , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Distribución Tisular
5.
Invest New Drugs ; 29(5): 833-45, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20428924

RESUMEN

Previous strategies for stabilizing combi-triazenes were based on masking the 1,2,3-triazene chain with a 3-acetoxymethylene group. The half-lives of the latter molecules were only ca 5 min longer than those of their parent 1,2,3-triazenes. The novel combi-molecules described herein contain a hydrolysable carbamate group that modulates their kinetics of degradation. Their half-lives were prolonged by ca 20-55 min when compared with their acetoxymethyltriazene counterparts. While they decomposed slowly in serum-containing medium, their intracellular decomposition was extremely rapid. They blocked EGFR tyrosine kinase in an isolated enzyme assay and in MDA-MB-468 breast cancer cells. Similarly, they all induced a dose-dependent DNA damage and G2/M cell cycle arrest in MDA-MB-468 cells, except the most stable compound ZRL2 (a 3-vinyl carbamate). ZRL4 (a chloromethyl carbamate) was the most potent and ZRL2 was the least active of the series against MDA-MB-468 cells. In selectivity assay with NIH-3T3 and NIH-3T3/HER-14, all compounds selectively blocked proliferation of NIH-3T3/HER-14. ZRS1 exerted the strongest growth inhibitory potency of the series. The results in toto suggest that ZRL2, despite being the most stable compound, could not hydrolyze at a rate that permitted the generation of DNA damaging species, thereby behaving primarily as an EGFR inhibitor. Thus the study permitted the definition of an optimized combi-molecule as one that decomposes at a rate that is slower than that of acetoxymethyltriazenes, but rapid enough to generate strong EGFR-DNA targeting potential and growth inhibition. Based on the latter criteria, ZRS1 and ZRL4 were tested in vivo and ZRS1 has proven the more effective.


Asunto(s)
Antineoplásicos/farmacología , Carbamatos/química , Carbamatos/farmacología , ADN de Neoplasias/metabolismo , Receptores ErbB/antagonistas & inhibidores , Triazenos/química , Triazenos/farmacología , Animales , Antineoplásicos/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Daño del ADN , Relación Dosis-Respuesta a Droga , Estabilidad de Medicamentos , Pruebas de Enzimas , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Citometría de Flujo , Humanos , Hidrólisis/efectos de los fármacos , Ratones , Microscopía Fluorescente , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología
6.
Cell Signal ; 23(4): 630-40, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21138763

RESUMEN

RB24 (NSC 741279), a 3-methyltriazene termed "combi-molecule" designed to possess mixed epidermal growth factor receptor (EGFR) targeting and DNA methylating properties showed over a 100-fold greater antiproliferative activity than Temodal(®) (TEM), a 4-fold greater potency than gefitinib and a 5-fold stronger activity than an equi-effective combination of gefitinib+TEM against the O(6)-alkylguanine transferase (AGT)-proficient DU145 cell line that co-expresses EGFR. Investigation of the mechanisms underlying the unique potency of RB24 revealed that cell exposure to TEM was accompanied by activation of p38MAPK and concomitant elevation of the levels of X-ray repair cross-complementing group 1 (XRCC1) protein. Levels of phospho-p38MAPK and XRCC1 were increased by 2-fold in EGF-stimulated cells. In contrast, EGF-stimulation did not alter the status of these proteins in RB24-treated cells and this translated into a 2-fold lower level of XRCC1 when compared with those exposed to TEM+EGF. These effects correlated with significantly delayed DNA repair activity in combi-molecule-treated cells when compared with TEM-exposed ones. Further analysis demonstrated that in contrast to TEM, RB24 could block Bad phosphorylation at serine 136 in a dose-dependent manner and induced significantly higher levels of apoptosis than the former molecule. Tandem depletion of XRCC1 and Bad activation through alternative pathways using the MEK1 inhibitor, PD98059, led to substantial levels of apoptosis in RB24-treated cells. The results in toto indicate that the superior activity of the combi-molecule may be attributed to its ability to down-regulate DNA repair proteins such as XRCC1 and to alleviate anti-apoptotic signaling through blockade of EGFR-mediated signaling while inflicting high levels of DNA lesions to the cells.


Asunto(s)
Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Triazenos/farmacología , Apoptosis/efectos de los fármacos , Butadienos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Daño del ADN , Metilación de ADN , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Sinergismo Farmacológico , Activación Enzimática , Gefitinib , Humanos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Nitrilos/farmacología , Fosforilación , Temozolomida
7.
Drug Metab Lett ; 3(1): 1-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19356110

RESUMEN

Combi-molecules are novel agents designed to be hydrolyzed into two bioactive species: an epidermal growth factor receptor (EGFR) tyrosine kinase (TK) inhibitor + a DNA alkylating agent. With the purpose of enhancing the tumour concentration of the bioactive species, we synthesized and compared the activities of RB107, a quinazolinotriazene designed to generate the bioactive BJ2000 upon hydrolysis, ZRDM and RB107ZR that require metabolic activation to generate BJ2000. The results showed that RB107 released the highest level of BJ2000 and its degradation product FD105 in vivo and high levels of the DNA alkylating methyl diazonium ion in the brain, kidney, liver and the DU145 tumours as confirmed by (14)C-labeling. The results in toto suggest that RB107 was stable enough to deliver the bioactive species to the tumour site and for optimal tumour distribution of the bioactive species, combi-molecules of the triazene class must be designed to be primarily degraded by hydrolytic cleavage and not by metabolic activation.


Asunto(s)
Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacocinética , Receptores ErbB/metabolismo , Triazenos/metabolismo , Triazenos/farmacocinética , Animales , Antineoplásicos Alquilantes/uso terapéutico , Biotransformación , Cromatografía Líquida de Alta Presión , Receptores ErbB/antagonistas & inhibidores , Humanos , Masculino , Espectrometría de Masas , Ratones , Profármacos/química , Profármacos/metabolismo , Profármacos/farmacocinética , Espectrofotometría Ultravioleta , Distribución Tisular , Triazenos/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Chem Biol Drug Des ; 71(4): 374-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18324926

RESUMEN

A novel type of 3,3-disubstituted bis-triazenes containing an ethylaminoethyl linker flanked by two identical anilinoquinazoline ring was synthesized. These model molecules contained an N-ethylaminomorpholine moiety designed to enhance water solubility. Despite their significant bulkiness, they blocked epidermal growth factor receptor (EGFR) tyrosine kinase in a dose-dependent manner with IC(50) values in low micromolar range. Molecular modeling to predict the interactions of the molecule with the ATP binding site of EGFR suggests that the N-ethylaminomorpholine side chain plays a binding role.


Asunto(s)
Receptores ErbB/química , Quinazolinas/síntesis química , Triazenos/síntesis química , Receptores ErbB/metabolismo , Concentración 50 Inhibidora , Modelos Moleculares , Quinazolinas/química , Solubilidad , Triazenos/química , Agua
9.
J Med Chem ; 50(11): 2605-8, 2007 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-17472358

RESUMEN

To enhance the potency of "combi-molecules", we designed 6a-d and 18 to release an inhibitor of EGFR TK and a bifunctional alkylator. The combi-molecules blocked EGFR TK with potency increasing with the basicity of the mustard moiety. They selectively killed cells transfected with EGFR and were potent against the DU145 prostate cancer cells. Combi-molecule 6a blocked EGFR phosphorylation in an irreversible manner, induced DNA-cross-links, and arrested the cells in mid-S.


Asunto(s)
Antineoplásicos Alquilantes/síntesis química , Receptores ErbB/antagonistas & inhibidores , Compuestos de Mostaza Nitrogenada/síntesis química , Triazenos/síntesis química , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Reactivos de Enlaces Cruzados/síntesis química , Reactivos de Enlaces Cruzados/química , Reactivos de Enlaces Cruzados/farmacología , ADN/metabolismo , Daño del ADN , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Humanos , Masculino , Modelos Moleculares , Compuestos de Mostaza Nitrogenada/química , Compuestos de Mostaza Nitrogenada/farmacología , Relación Estructura-Actividad , Transfección , Triazenos/química , Triazenos/farmacología
10.
Clin Cancer Res ; 13(1): 331-40, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17200372

RESUMEN

PURPOSE: JDA58 (NSC 741282), a "combi-molecule" optimized in the context of the "combi-targeting concept," is a nitrosourea moiety tethered to an anilinoquinazoline. Here, we sought to show its binary epidermal growth factor receptor (EGFR)/DNA targeting property and to study its fragmentation in vitro and in vivo. EXPERIMENTAL DESIGN: The fragmentation of JDA58 was detected in cells in vitro and in vivo by fluorescence microscopy and tandem mass spectrometry. EGFR phosphorylation and DNA damage were determined by Western blotting and comet assay, respectively. Tumor data were examined for statistical significance using the Student's t test. RESULTS: JDA58 inhibited EGFR tyrosine kinase (IC(50), 0.2 micromol/L) and blocked EGFR phosphorylation in human DU145 prostate cancer cells. It induced significant levels of DNA damage in DU145 cells in vitro or in vivo and showed potent antiproliferative activity both in vitro and in a DU145 xenograft model. In cell-free medium, JDA58 was hydrolyzed to JDA35, a fluorescent amine that could be observed in tumor cells both in vitro and in vivo. In tumor cells in vitro or in vivo, or in plasma collected from mice, the denitrosated species JDA41 was the predominant metabolite. However, mass spectrometric analysis revealed detectable levels of the hydrolytic product JDA35 in tumor cells both in vitro and in vivo. CONCLUSIONS: The results in toto suggest that growth inhibition in vitro and in vivo may be sustained by the intact combi-molecule plus JDA35 plus JDA41, three inhibitors of EGFR, and the concomitantly released DNA-damaging species. This leads to a model wherein a single molecule carries a complex multitargeted-multidrug combination.


Asunto(s)
ADN/química , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/metabolismo , Compuestos de Nitrosourea/farmacología , Animales , Línea Celular Tumoral , Ensayo Cometa , Daño del ADN , Humanos , Concentración 50 Inhibidora , Masculino , Espectrometría de Masas , Ratones , Microscopía Fluorescente , Trasplante de Neoplasias , Fosforilación
11.
Anticancer Drugs ; 18(2): 171-7, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17159603

RESUMEN

We recently designed molecules termed "type II combi-molecules" to block the epidermal growth factor receptor and to damage DNA without the requirement for hydrolytic cleavage. Here, we studied two such combi-molecules (JDD36 and JDE05), containing a novel quinazoline-linked chloroethyltriazolinium system. The epidermal growth factor receptor-targeting potential of these novel structures was studied by ELISA for isolated epidermal growth factor receptor and by Western blotting for whole-cell assays. DNA damage was analyzed using the single-cell microelectrophoresis comet assay. Antiproliferative effects were determined by the sulforhodamine B assay. JDD36 showed an IC50 of 0.6 micromol/l in the ELISA for epidermal growth factor receptor tyrosine kinase, a dose-dependent inhibition of epidermal growth factor receptor phosphorylation and significant levels of DNA damage in the human DU145 prostate cancer cell line. JDD36 was an overall 2- to 15-fold stronger antiproliferative agent than JDE05 that showed potent epidermal growth factor receptor inhibitory activity (IC50 epidermal growth factor receptor, 0.035 micromol/l) but weak DNA-damaging potential. In a panel of LNCaP erbB transfectants, in contrast to JDE05, JDD36 showed remarkable and selective potency against the LNCaPerbB2 transfectant. The results in toto suggest that the overall superior potency of JDD36 when compared with JDE05 may be imputed to its balanced binary epidermal growth factor receptor-DNA-targeting properties that may induce a tandem blockade of epidermal growth factor receptor-mediated mitogenic signaling while depleting alternative survival mechanism by damaging DNA.


Asunto(s)
Antineoplásicos/farmacología , Animales , División Celular/efectos de los fármacos , Ensayo Cometa , Daño del ADN/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/efectos de los fármacos , Genes erbB-1/efectos de los fármacos , Genes erbB-1/genética , Genes erbB-2/efectos de los fármacos , Genes erbB-2/genética , Humanos , Masculino , Ratones , Células 3T3 NIH , Fosforilación , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología
12.
Anticancer Drugs ; 17(2): 165-71, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16428934

RESUMEN

With the purpose of developing drugs that can block multiple targets in tumor cells, molecules termed combi-molecules or TZ-I have been designed to be hydrolyzed in vitro to TZ+I, where TZ is a DNA-damaging species and I is an inhibitor of the epidermal growth factor receptor (EGFR). Using HPLC and liquid chromatography-mass spectrometry (LC-MS), we investigated the mechanism of in vivo degradation of a prototype of one such combi-molecule, ZRBA1, which when administered i.p. rapidly degraded into FD105 (Cmax=50 micromol/l, after 30 min), a 6-aminoquinazoline that was N-acetylated to give FD105Ac (IAc) (Cmax=18 micromol/l, after 4 h). A similar rate of acetylation was observed when independently synthesized FD105 was administered i.p. More importantly, the EGFR binding affinity of IAc was 3-fold greater than that of I, indicating that the latter is converted in vivo into an even more potent EGFR inhibitor. The results in toto suggest that while in vitro TZ-I is only hydrolyzed to I+TZ, further acetylation of I in vivo leads to a third component--a highly potent EGFR inhibitor with a delayed Cmax.


Asunto(s)
Quinazolinas/farmacocinética , Triazenos/farmacocinética , Acetilación , Animales , Proliferación Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Receptores ErbB/antagonistas & inhibidores , Humanos , Masculino , Espectrometría de Masas , Ratones , Neoplasias de la Próstata/metabolismo , Quinazolinas/síntesis química , Triazenos/síntesis química , Células Tumorales Cultivadas
13.
Chin J Integr Med ; 11(1): 49-53, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15975309

RESUMEN

OBJECTIVE: To study the therapeutic effects and mechanism of Jiechangning (JCN) decoction on carrageenan induced experimental ulcerative colitis (UC). METHODS: After sensitizing guinea pigs with carrageenan, we established UC animal models by free drinking water containing 2% acid degraded carrageenan (ADC). JCN decoction was orally administered once a day for 2 weeks after carrageenan treatment. Salicylazosulfapyridine (SASP) and normal saline were given to the other two groups as control. The levels of colon lipid peroxide (LPO), acid phosphatase (ACP) activity and tumor necrosis factor-alpha (TNF-alpha) were measured; colitis activity score (CAS) was carried out for assessment of the degree of tissue inflammation and injury; the colonic pathological changes were examined simultaneously with hematoxylin and eosin (HE) and toluidine blue staining used to evaluate the therapeutic effects of JCN decoction and SASP. RESULTS: Experimental colitis models resembling human UC were successfully induced. The levels of tissue LPO, ACP activity and the content of tissue TNF-alpha were markedly increased in the model group as compared with the normal control group (P < 0.01) and were positively correlated with CAS. JCN decoction could reverse these changes like SASP. HE staining showed that JCN decoction and SASP could reduce CAS and the degree of tissue injury, toluidine blue staining revealed that mucosa and submucosa red metachromasia pellets in JCN group and SASP group were markedly fewer than those in the model group. CONCLUSION: JCN decoction is effective in treating experimental UC, which provides theoretical basis for its clinical application.


Asunto(s)
Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/patología , Medicina Tradicional China , Preparaciones de Plantas/farmacología , Fosfatasa Ácida/metabolismo , Animales , Carragenina , Colitis Ulcerosa/inducido químicamente , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Fármacos Gastrointestinales/farmacología , Cobayas , Peróxidos Lipídicos/metabolismo , Masculino , Sulfasalazina/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
14.
Int J Cancer ; 112(3): 484-91, 2004 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-15382076

RESUMEN

The mechanism of action of ZR2002, a chimeric amino quinazoline designed to possess mixed EGFR tyrosine kinase (TK) inhibitory and DNA targeting properties, was compared to those of ZR01, a reversible inhibitor of the same class and PD168393, a known irreversible inhibitor of EGFR. ZR2002 exhibited 4-fold stronger EGFR TK inhibitory activity than its structural homologue ZR01 but was approximately 3-fold less active than the 6-acrylamidoquinazoline PD168393. It preferentially blocked EGF and TGFalpha-induced cell growth over PDGF and serum. It also inhibited signal transduction in heregulin-stimulated breast tumour cells, indicating that it does not only block EGFR but also its closely related erbB2 gene product. In contrast to its structural homologues, ZR2002 was capable of inducing significant levels of DNA strand breaks in MDA-MB-468 cells after a short 2 hr drug exposure at a concentration as low as 10 microM. Reversibility studies using whole cell autophosphorylation and growth assays in human breast cell lines showed that in contrast to its reversible inhibitor counterpart ZR01, ZR2002 induced irreversible inhibition of EGF-stimulated autophosphorylation in MDA-MB-468 cells and irreversible inhibition of cell growth. Moreover despite possessing a weaker binding affinity than PD168393, it induced a significantly more sustained antiproliferative effect than the latter after a pulse 2 hr exposure. More importantly, in contrast to ZR01 and PD168393, ZR2002 was capable of inducing significant levels of cell death by apoptosis in MDA-MB-468 cells. The results in toto suggest that the superior antiproliferative potency of ZR2002 may be due to its ability to induce a protracted blockade of receptor tyrosine kinase-mediated signaling while damaging cellular DNA, a combination of events that may trigger cell-killing by apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Proteínas Oncogénicas v-erbB/antagonistas & inhibidores , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Femenino , Humanos , Estructura Molecular , Fosforilación/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Factor de Crecimiento Transformador alfa/farmacología , Células Tumorales Cultivadas
15.
Prostate ; 59(1): 13-21, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-14991862

RESUMEN

BACKGROUND: FD137, a nitrosourea appended to a quinazoline ring, was designed to simultaneously block epidermal growth factor receptor (EGFR)-mediated signaling and damage genomic DNA in refractory EGF-dependent prostate tumors. METHODS: The mixed inhibition of cell signaling and DNA damage by FD137 were determined by Western blotting, RT-PCR, flow cytometry, sulforhodamine B (SRB), and comet assay. RESULTS: FD137 and its metabolite FD110 induced a dose-dependent increase in inhibition of EGF-stimulated EGFR autophosphorylation and this translated into blockade of c-fos gene expression in DU145 cells. FD137 induced significant levels of DNA damage and showed 150-fold greater anti-proliferative activity than BCNU, a classical nitrosourea. In contrast to BCNU, complete inhibition of EGF-induced cell transition to S-phase was observed at concentrations of FD137 as low as 3 microM. CONCLUSION: FD137 could not only damage DNA, but also significantly block downstream EGFR-mediated signaling. The superior activity of FD137 may be imputable to the combined effect of its mixed EGFR/DNA targeting properties. This novel strategy may well represent a new approach to target nitrosoureas to EGFR-overexpressing carcinomas of the prostate.


Asunto(s)
Antineoplásicos/farmacología , Compuestos de Nitrosourea/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Carmustina/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayo Cometa , Daño del ADN , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes fos/efectos de los fármacos , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Neoplásico/química , ARN Neoplásico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
16.
Bull Cancer ; 91(12): 911-5, 2004 Dec.
Artículo en Francés | MEDLINE | ID: mdl-15634632

RESUMEN

It is now known that tumour cells possess many signaling pathways to repair damage inflicted by alkylating agents. However, most of these cytotoxic agents only target DNA and this does not suffice to induce sustained antiproliferative activity. Furthermore, the efficacy of antitumour alkylating agents is hampered by a lack of selectivity for tumour tissues. To circumvent these problems, we recently designed a novel strategy termed combi-targeting that sought to synthesize compounds capable of not only damaging DNA, but also blocking signaling associated with aggressive proliferation. The first prototypes described herein can block signaling associated with the epidermal growth factor receptor (EGFR) and significantly damage DNA. In addition to their binary EGFR/DNA targeting properties, we demonstrated that their effects are selective for cells to which EGFR has conferred a proliferative advantage. These novel agents with mixed targeting properties are termed "combi-molecules".


Asunto(s)
Antineoplásicos Alquilantes/síntesis química , Receptores ErbB/antagonistas & inhibidores , Compuestos de Nitrosourea/síntesis química , Quinazolinas/síntesis química , Antineoplásicos Alquilantes/uso terapéutico , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , Compuestos de Nitrosourea/uso terapéutico , Quinazolinas/uso terapéutico
17.
Cancer Chemother Pharmacol ; 51(1): 1-10, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12497200

RESUMEN

PURPOSE: To study the dual mechanism of action of FD137, a 3,3-disubstituted nitrosourea designed to block signaling mediated by the epidermal growth factor receptor (EGFR) on its own and to be hydrolyzed to an inhibitor of EGFR plus a DNA-damaging species. MATERIALS AND METHODS: HPLC was used to determine the half-life (t(1/2)) of FD137 and to characterize its derived metabolite FD110. The dual mechanisms of DNA damaging and EGFR tyrosine kinase (TK) targeting were ascertained by the comet assay for DNA damage and by inmunodetection of phosphotyrosine in an ELISA and a whole-cell assay for EGFR-mediated signaling. The antiproliferative effects of the different drugs and their combinations were determined by the sulforhodamine B (SRB) assay. RESULTS: In contrast to BCNU, FD137 significantly blocked EGF-induced EGFR autophosphorylation (IC(50) 4 micro M) in the human solid tumor cell line A431. DNA damage induced by FD137 could only be observed after 24 h exposure, but the level of DNA damage remained 3.6-fold lower than that induced by BCNU. This difference was rationalized by the 160-fold greater stability of FD137 when compared with BCNU in serum-containing medium. Further, degradation of FD137 was accompanied by the slow release of FD110, an extremely potent inhibitor of EGFR TK [IC(50) (EGFR autophosphorylation) <0.3 micro M]. The complex properties of FD137 translated into a 55-fold greater antiproliferative activity than BCNU against the EGFR-overexpressing A431 cells that coexpresses the O(6)-alkylguanine transferase (AGT). Depletion of AGT in these cells by the use of O(6)-benzylguanine (O(6)-BG) enhanced their sensitivity to BCNU by 8-fold, but only by 3-fold to FD137. CONCLUSIONS: The results overall suggest that the superior antiproliferative activity of FD137 when compared with BCNU may be associated with its ability to behave as a combination of many species with different mechanisms of action. However, the enhancement of its potency by O(6)-BG suggests that its antiproliferative effect was at least partially mitigated by AGT and perhaps it may be largely dominated by its signal transduction inhibitory component.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Daño del ADN , Receptores ErbB/antagonistas & inhibidores , Compuestos de Nitrosourea/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , División Celular/efectos de los fármacos , Humanos , O(6)-Metilguanina-ADN Metiltransferasa/fisiología , Fosforilación , Proteínas Tirosina Quinasas , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...