Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 66(4): 2477-2497, 2023 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-36780426

RESUMEN

Phenyl 4-(2-oxo-3-alkylimidazolidin-1-yl)benzenesulfonates (PAIB-SOs) are a new family of antimitotic prodrugs bioactivated in breast cancer cells expressing CYP1A1. In this study, we report that the 14C-labeled prototypical PAIB-SO [14C]CEU-818 and its antimitotic counterpart [14C]CEU-602 are distributed in whole mouse body and they show a short half-life in mice. To circumvent this limitation, we evaluated the effect of the homologation of the alkyl side chain of the imidazolidin-2-one moiety of PAIB-SOs. Our studies evidence that PAIB-SOs bearing an n-pentyl side chain exhibit antiproliferative activity in the nanomolar-to-low-micromolar range and a high selectivity toward CYP1A1-positive breast cancer cells. Moreover, the most potent n-pentyl PAIB-SOs were significantly more stable toward rodent liver microsomes. In addition, PAIB-SOs 10 and 14 show significant antitumor activity and low toxicity in chorioallantoic membrane (CAM) assay. Our study confirms that homologation is a suitable approach to improve the rodent hepatic stability of PAIB-SOs.


Asunto(s)
Antimitóticos , Neoplasias , Profármacos , Ratones , Animales , Antimitóticos/química , Profármacos/química , Citocromo P-450 CYP1A1 , Roedores , Microsomas Hepáticos , Bencenosulfonatos/química
2.
Eur J Med Chem ; 229: 114003, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-34839998

RESUMEN

We recently discovered a new family of prodrugs deriving from phenyl 4-(2-oxo-3-imidazolidin-1-yl)benzenesulfonates (PIB-SOs) bioactivatable by cytochrome P450 1A1 (CYP1A1) into potent antimitotics referred to as phenyl 4-(2-oxo-3-alkylimidazolidin-1-yl)benzenesulfonates (PAIB-SOs). PAIB-SOs display significant selectivity toward human breast cancer cells based on the N-dealkylation of PAIB-SOs into their corresponding PIB-SOs by CYP1A1. In this study, we have evaluated the molecular mechanism of the bioactivation of PAIB-SOs into PIB-SOs by branching the linear alkyl chain on the imidazolidin-2-one (IMZ) moiety of PAIB-SOs by branched alkyl groups such as isopropyl, isobutyl and sec-butyl. Our results show that PAIB-SOs bearing an isobutyl group on the IMZ moiety and either a methoxy, a chloro or a bromo group at positions 3, 3,5 or 3,4,5 on the aromatic ring B exhibit antiproliferative activity ranging from 0.13 to 6.9 µM and selectivity toward MCF7 and MDA-MB-468 mammary cancer cells comparatively to other cell lines tested. Moreover, the most potent and selective PAIB-SOs bearing an isobutyl group and either a 3,5-Cl (44), 3,5-Br (45) or a 3,4,5-OMe (46) on the IMZ moiety exhibit antiproliferative activity in the sub-micromolar range and high selectivity ratios toward mammary cancer cells. They stop the cell cycle of MCF7 cells in the G2/M phase and disrupt their cytoskeleton. Furthermore, our studies evidenced that PAIB-SOs bearing either an isopropyl, a sec-butyl or an isobutyl group are hydroxylated on the carbon atom adjacent to the IMZ (Cα-OH) but only PAIB-SOs bearing an isobutyl group are bioactivated into PIB-SOs. Finally, PAIB-SOs 45 and 46 exhibit low toxicity toward normal cells and chick embryos and are thus promising antimitotic prodrugs highly selective toward CYP1A1-expressing breast cancer cells.


Asunto(s)
Antimitóticos/química , Bencenosulfonatos/química , Citocromo P-450 CYP1A1/metabolismo , Profármacos/química , Animales , Antimitóticos/síntesis química , Antimitóticos/farmacología , Bencenosulfonatos/síntesis química , Bencenosulfonatos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Pollos , Citocromo P-450 CYP1A1/química , Ensayos de Selección de Medicamentos Antitumorales , Estabilidad de Medicamentos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Semivida , Humanos , Microsomas Hepáticos/metabolismo , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Profármacos/farmacocinética , Profármacos/farmacología , Relación Estructura-Actividad , Especificidad por Sustrato
3.
Cancers (Basel) ; 13(14)2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34298771

RESUMEN

Triple-negative breast cancer (TNBC) is a major concern among the different subtypes of breast cancer (BC) due to the lack of effective treatment. In a previous study by our group aimed at understanding the difference between TNBC and non-TNBC tumors, we identified the gene TBC1 domain family member 9 (TBC1D9), the expression of which was lower in TNBC as compared to non-TNBC tumors. In the present study, analysis of TBC1D9 expression in TNBC (n = 58) and non-TNBC (n = 25) patient tumor samples validated that TBC1D9 expression can differentiate TNBC (low) from non-TNBC (high) samples and that expression of TBC1D9 was inversely correlated with grade and proliferative index. Moreover, we found that downregulation of the TBC1D9 gene decreases the proliferation marginally in non-TNBC and was associated with increased migratory and tumorigenic potential in both TNBC and luminal BC cell lines. This increase was mediated by the upregulation of ARL8A, ARL8B, PLK1, HIF1α, STAT3, and SPP1 expression in TBC1D9 knockdown cells. Our results suggest that TBC1D9 expression might limit tumor aggressiveness and that it has a differential expression in TNBC vs. non-TNBC tumors.

4.
J Pharm Pharmacol ; 72(2): 249-258, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31729035

RESUMEN

OBJECTIVES: In this study, the antiproliferative activity of 3 phenyl 4-(2-oxo-3-alkylimidazolidin-1-yl)benzenesulfonates (PAIB-SOs) was assessed in a time-dependent manner together with their hepatic stability and metabolism using human, mouse and rat liver microsomes. METHODS: CEU-818, -820 and -913 were selected as promising hit compounds. Their antiproliferative activity on human breast carcinoma MCF-7 cells was evaluated using escalating concentrations of drugs at 24, 36 and 48 h and the sulforhodamine B assay. Their hepatic stability was evaluated by HPLC-UV of extracts obtained from human, mouse and rat liver microsomes. KEY FINDINGS: The antiproliferative activity of PAIB-SOs is concentration and time-dependent and requires between 24 and 36 h of contact with MCF-7 cells to detect a significant antiproliferative activity. PAIB-SOs stability in microsomes usually decreases following this order: human ≈ (rat > mouse). The CEU-913 exhibits the longest half-life in rat and human liver microsomes while the CEU-820 exhibits the longest half-life in mouse liver microsomes. CONCLUSIONS: Our in vitro results suggest that PAIB-SOs should have a minimum contact time of 24 h with the tumour to trigger significant antitumoural activity. The activity of mouse liver microsomes towards PAIB-SOs is higher than rat microsomes and tends to be higher than human liver microsomes.


Asunto(s)
Antineoplásicos/farmacología , Bencenosulfonatos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Microsomas Hepáticos/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Bencenosulfonatos/administración & dosificación , Bencenosulfonatos/química , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Citocromo P-450 CYP1A1/metabolismo , Femenino , Semivida , Humanos , Células MCF-7 , Ratones , Profármacos , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Factores de Tiempo
5.
J Med Chem ; 60(12): 4963-4982, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28535350

RESUMEN

Prodrug-mediated utilization of the cytochrome P450 (CYP) 1A1 to obtain the selective release of potent anticancer products within cancer tissues is a promising approach in chemotherapy. We herein report the rationale, preparation, biological evaluation, and mechanism of action of phenyl 4-(2-oxo-3-alkylimidazolidin-1-yl)benzenesulfonates (PAIB-SOs) that are antimicrotubule prodrugs activated by CYP1A1. Although PAIB-SOs are inert in most cells tested, they are highly cytocidal toward several human breast cancer cells, including hormone-independent and chemoresistant types. PAIB-SOs are N-dealkylated into cytotoxic phenyl 4-(2-oxo-3-imidazolidin-1-yl)benzenesulfonates (PIB-SOs) in CYP1A1-positive cancer cells, both in vitro and in vivo. In conclusion, PAIB-SOs are novel chemotherapeutic prodrugs with no equivalent among current antineoplastics and whose selective action toward breast cancer is tailored to the characteristic pattern of CYP1A1 expression observed in a large percentage of human breast tumors.


Asunto(s)
Antimitóticos/farmacología , Bencenosulfonatos/química , Neoplasias de la Mama/tratamiento farmacológico , Citocromo P-450 CYP1A1/metabolismo , Profármacos/farmacología , Animales , Antimitóticos/farmacocinética , Bencenosulfonatos/síntesis química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Técnicas de Química Sintética , Embrión de Pollo , Citocromo P-450 CYP1A1/genética , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Humanos , Profármacos/farmacocinética
6.
Bioorg Med Chem Lett ; 27(2): 299-302, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27903409

RESUMEN

The synthesis of two series of novel substituted pyrimidine derivatives bearing a sulfamide group have been described and their in vitro cancer growth inhibition activities have been evaluated against three human tumour cell lines (HT-29, M21, and MCF7). In general, growth inhibition activity has been enhanced by the introduction of a bulky substituent on the aromatic ring with the best compound having GI50<6µM for all the human tumour cell lines. The MCF7 selective compounds were evaluated on four additional human invasive breast ductal carcinoma cell lines (MDA-MB-231, MDA-MB-468, SKBR3, and T47D) and were selective against T47D cell line in all cases except one, suggesting a potential antiestrogen activity.


Asunto(s)
Antineoplásicos/farmacología , Pirimidinas/farmacología , Sulfonamidas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Pirimidinas/síntesis química , Pirimidinas/química , Relación Estructura-Actividad , Sulfonamidas/química
7.
Steroids ; 115: 98-104, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27553724

RESUMEN

A series of 7α-linked testosterone dimers were made and tested for biological activity on both androgen-dependent (LNCaP) and androgen-independent (DU-145 and PC3) prostate cancer cell lines. The synthesis proceeds through the formation of a trans-4-(17ß-acetoxy-4-androsten-3-one-7α-yl)-but-2-enoic acid 4-hydroxy-alkyl ester intermediate of various length (7a-d) followed by the final dimerization step. The dimers showed interesting biological activity in comparison to the ω-hydroxyalkyl ester intermediates 7a-d. The most active dimer 8a (n=1) showed IC50 of 3.8, 1.4 and 1.8µM, respectively on LNCaP, DU-145 and PC3 cancer cell lines. On these cell lines, this dimer is about 12, 70 and 47 times more powerful than cyproterone acetate (CPA) the reference antiandrogen. Furthermore, dimers 8b-d (n=2, 3, 4) were less active than 8a but showed selective activity on androgen-dependent LNCaP prostate cancer cells. This indicates possible application for the treatment of androgen-dependent prostate cancer.


Asunto(s)
Andrógenos/metabolismo , Neoplasias de la Próstata/metabolismo , Testosterona/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Espectroscopía de Resonancia Magnética , Masculino , Estructura Molecular , Testosterona/química
8.
Can J Physiol Pharmacol ; 94(3): 341-6, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26679837

RESUMEN

Islet amyloid polypeptide (IAPP) has been shown to form amyloid deposits in pancreatic islets, thereby furthering type 2 diabetes disease progression. Further discovery of new molecules is needed to create a diverse set of molecules that impede pancreatic amyloidosis. We have recently designed and synthesized N-phenyl-N'-(2-ethyl)ureas (EU) that are non-cytotoxic small molecules, to evaluate the role of the aryl-substituted moiety on the inhibition of hIAPP fibrillization. Several EUs were tested in vitro for their anti-amyloidogenic activity using the fluorometric ThT assay, the photo-induced cross-linking (PIUCP) assay, and cell survival assay in pancreatic MIN-6 cells. EU-362 and EU-418 were able to significantly inhibit the formation of hIAPP fibrils and protected cells from amyloid cytotoxic effects. Our results suggest that increasing the nucleophilic potency of the aryl moiety significantly enhances the anti-amyloidogenic activity of the molecules.


Asunto(s)
Polipéptido Amiloide de los Islotes Pancreáticos/antagonistas & inhibidores , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Urea/análogos & derivados , Amiloide/metabolismo , Amiloidosis/tratamiento farmacológico , Amiloidosis/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/efectos adversos , Islotes Pancreáticos/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Urea/farmacología
9.
Eur J Med Chem ; 100: 34-43, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26069928

RESUMEN

Combretastatin A-4 (CA-4) is a well-studied and attractive molecular template to develop new antimitotics. Several thousand of modifications were performed on the ring B and the ethenyl bridge of CA-4 but only a few involved the trimethoxyphenyl moiety (TMP, ring A) often considered essential to the antiproliferative and antimicrotubule activities. In this study, we described the design, the preparation, the characterization and the biological evaluation of three new series of CA-4 analogs namely styryl-N-phenyl-N'-ethylureas (SEUs), styryl-N-phenyl-N'-(2-chloroethyl)ureas (SCEUs) and styrylphenylimidazolidin-2-ones (SIMZs) bearing a 3-Cl (series a), 3,5-Me (series b) and TMP (series c) substituents, respectively. All SCEU and SIMZ Z-isomers were active in the high and the low nanomolar range, respectively. Conversely to SEUs and their E-isomers that were significantly less active or inactive. Interestingly, the TMP moiety is giving rise to derivatives exhibiting the lowest antiproliferative activity in the SCEU series (10c) and the most active compound in the SIMZ series (12c). Moreover, SIMZ Z-isomers bearing either a 3-Cl (12a) or a 3,5-Me (12b) exhibited antiproliferative activities that are also in the same order of magnitude as 12c. All SCEU and SIMZ Z-isomers also arrested the cell cycle progression in G2/M phase, bound to the colchicine-binding site and disrupted the cytoskeleton of cancer cells. In addition to the promising and innovative microtubule-disrupting properties of SCEUs and SIMZs, these results show that the TMP moiety is not essential for the cytocidal activity of these new CA-4 analogs.


Asunto(s)
Antineoplásicos/farmacología , Imidazolidinas/farmacología , Microtúbulos/efectos de los fármacos , Estilbenos/farmacología , Urea/análogos & derivados , Antineoplásicos/síntesis química , Antineoplásicos/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HT29 , Humanos , Imidazolidinas/síntesis química , Imidazolidinas/química , Células MCF-7 , Estructura Molecular , Estilbenos/química , Relación Estructura-Actividad , Urea/síntesis química , Urea/química , Urea/farmacología
10.
J Med Chem ; 55(13): 6194-208, 2012 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-22694057

RESUMEN

Twenty-eight new substituted N-phenyl ureidobenzenesulfonate (PUB-SO) and 18 N-phenylureidobenzenesulfonamide (PUB-SA) derivatives were prepared. Several PUB-SOs exhibited antiproliferative activity at the micromolar level against the HT-29, M21, and MCF-7 cell lines and blocked cell cycle progression in S-phase similarly to cisplatin. In addition, PUB-SOs induced histone H2AX (γH2AX) phosphorylation, indicating that these molecules induce DNA double-strand breaks. In contrast, PUB-SAs were less active than PUB-SOs and did not block cell cycle progression in S-phase. Finally, PUB-SOs 4 and 46 exhibited potent antitumor activity in HT-1080 fibrosarcoma cells grafted onto chick chorioallantoic membranes, which was similar to cisplatin and combretastatin A-4 and without significant toxicity toward chick embryos. These new compounds are members of a promising new class of anticancer agents.


Asunto(s)
Anticarcinógenos/química , Anticarcinógenos/farmacología , Bencenosulfonatos/química , Bencenosulfonatos/farmacología , Proliferación Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena , Compuestos de Fenilurea/química , Compuestos de Fenilurea/farmacología , Fase S/efectos de los fármacos , Animales , Anticarcinógenos/síntesis química , Bencenosulfonatos/síntesis química , Ciclo Celular , Línea Celular Tumoral , Embrión de Pollo , Membrana Corioalantoides/efectos de los fármacos , Cisplatino/farmacología , Células HT29 , Histonas/metabolismo , Humanos , Imidazolidinas/síntesis química , Imidazolidinas/química , Imidazolidinas/farmacología , Células Jurkat , Neoplasias/tratamiento farmacológico , Compuestos de Fenilurea/síntesis química , Fosforilación , Estilbenos/farmacología , Relación Estructura-Actividad
11.
J Med Chem ; 54(13): 4559-80, 2011 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-21604746

RESUMEN

Sixty-one phenyl 4-(2-oxoimidazolidin-1-yl)benzenesulfonates (PIB-SOs) and 13 of their tetrahydro-2-oxopyrimidin-1(2H)-yl analogues (PPB-SOs) were prepared and biologically evaluated. The antiproliferative activities of PIB-SOs on 16 cancer cell lines are in the nanomolar range and unaffected in cancer cells resistant to colchicine, paclitaxel, and vinblastine or overexpressing the P-glycoprotein. None of the PPB-SOs exhibit significant antiproliferative activity. PIB-SOs block the cell cycle progression in the G(2)/M phase and bind to the colchicine-binding site on ß-tubulin leading to cytoskeleton disruption and cell death. Chick chorioallantoic membrane tumor assays show that compounds 36, 44, and 45 efficiently block angiogenesis and tumor growth at least at similar levels as combretastatin A-4 (CA-4) and exhibit low to very low toxicity on the chick embryos. PIB-SOs were subjected to CoMFA and CoMSIA analyses to establish quantitative structure-activity relationships.


Asunto(s)
Arilsulfonatos/síntesis química , Derivados del Benceno/síntesis química , Imidazolidinas/síntesis química , Estilbenos/química , Moduladores de Tubulina/síntesis química , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Animales , Arilsulfonatos/química , Arilsulfonatos/farmacología , Derivados del Benceno/química , Derivados del Benceno/farmacología , Sitios de Unión , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/efectos de los fármacos , Colchicina/metabolismo , Diseño de Fármacos , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Imidazolidinas/química , Imidazolidinas/farmacología , Modelos Moleculares , Imitación Molecular , Neovascularización Fisiológica/efectos de los fármacos , Relación Estructura-Actividad Cuantitativa , Estilbenos/farmacología , Trasplante Heterólogo , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología
12.
Biochem Pharmacol ; 81(9): 1116-23, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21371445

RESUMEN

The cyclization of anticancer drugs into active intermediates has been reported mainly for DNA alkylating molecules including nitrosoureas. We previously defined the original cytotoxic mechanism of anticancerous N-phenyl-N'-(2-chloroethyl)ureas (CEUs) that involves their reactivity towards cellular proteins and not against DNA; two CEU subsets have been shown to alkylate ß-tubulin and prohibitin leading to inhibition of cell proliferation by G2/M or G1/S cell cycle arrest. In this study, we demonstrated that cyclic derivatives of CEUs, N-phenyl-4,5-dihydrooxazol-2-amines (Oxas) are two- to threefold more active than CEUs and share the same cytotoxic properties in B16F0 melanoma cells. Moreover, the CEU original covalent binding by an ester linkage on ß-tubulin Glu198 and prohibitin Asp40 was maintained with Oxas. Surprisingly, we observed that Oxas were spontaneously formed from CEUs in the cell culture medium and were also detected within the cells. Our results suggest that the intramolecular cyclization of CEUs leads to active Oxas that should then be considered as the key intermediates for protein alkylation. These results will be useful for the design of new prodrugs for cancer chemotherapy.


Asunto(s)
Alquilantes/farmacología , Aminas/metabolismo , Ácido Aspártico/metabolismo , Ácido Glutámico/metabolismo , Proteínas Represoras/metabolismo , Tubulina (Proteína)/metabolismo , Urea/metabolismo , Aminas/química , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Ciclización , Electroforesis en Gel Bidimensional , Técnica del Anticuerpo Fluorescente , Melanoma Experimental/patología , Ratones , Prohibitinas , Proteínas Represoras/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tubulina (Proteína)/química , Urea/química
13.
J Pharmacol Exp Ther ; 336(2): 460-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20978170

RESUMEN

N-Phenyl-N'-(2-chloroethyl)ureas (CEUs) are antimicrotubule agents interacting covalently with ß-tubulin near the colchicine-binding site (C-BS). Glutamyl 198 residue in ß-tubulin (Glu198), which is adjacent to the C-BS behind the two potent nucleophilic residues, Cys239 and Cys354, has been shown to covalently react with 1-(2-chloroethyl)-3-(4-iodophenyl)urea (ICEU). By use of mass spectrometry, we have now identified residues in ß-tubulin that have become modified irreversibly by 1-(2-chloroethyl)-3-[3-(5-hydroxypentyl)phenyl]urea (HPCEU), 1-[4-(3-hydroxy-4-methoxystyryl)phenyl]-3-(2-chloroethyl)urea (4ZCombCEU), and N,N'-ethylenebis(iodoacetamide) (EBI). The binding of HPCEU and 4ZCombCEU to ß-tubulin resulted in the acylation of Glu198, a protein modification of uncommon occurrence in living cells. Prototypical CEUs then were used as molecular probes to assess, in mouse B16F0 and human MDA-MB-231 cells, the role of Glu198 in microtubule stability. For that purpose, we studied the effect of Glu198 modification by ICEU, HPCEU, and 4ZCombCEU on the acetylation of Lys40 on α-tubulin, a key indicator of microtubule stability. We show that modification of Glu198 by prototypical CEUs correlates with a decrease in Lys40 acetylation, as observed also with other microtubule depolymerizing agents. Therefore, CEU affects the stability and the dynamics of microtubule, likewise a E198G mutation, which is unusual for xenobiotics. We demonstrate for the first time that EBI forms an intramolecular cross-link between Cys239 and Cys354 of ß-tubulin in living cells. This work establishes a novel basis for the development of future chemotherapeutic agents and provides a framework for the design of molecules useful for studying the role of Asp and Glu residues in the structure/function and the biological activity of several cellular proteins under physiological conditions.


Asunto(s)
Antineoplásicos/metabolismo , Microtúbulos/química , Moduladores de Tubulina/metabolismo , Tubulina (Proteína)/metabolismo , Urea/análogos & derivados , Acetilación , Acilación , Animales , Células Cultivadas , Humanos , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Relación Estructura-Actividad , Tubulina (Proteína)/química
14.
Can J Physiol Pharmacol ; 88(11): 1102-14, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21076498

RESUMEN

Aryl chloroethyl ureas (CEUs) are new protein alkylating agents exhibiting anticancer activity both in vitro and in vivo. We report herein that 14C-labeled CEU derivatives, designated CEU-025 and CEU-027, covalently bind to thioredoxin-1 (TRX1). Covalent binding of these molecules slightly decreases the disulfide-reducing activity of recombinant TRX1, when compared with the effect of strong thioalkylating agents such as N-ethylmaleimide. Moreover, site-directed mutagenesis and diamide competition assays demonstrated that TRX1 cysteinyl residues are not the prime targets of CEUs. CEU-025 abrogates the nuclear translocation of TRX1 in human cancer cells. In addition, we show that CEU-025 can block TRX1 nuclear translocation induced by cisplatin. Unexpectedly, pretreatment with sublethal CEU-025 concentrations that block TRX1 nuclear translocation protected the cells against cisplatin cytotoxicity. Overexpression of TRX1 in HT1080 fibrosarcoma cells attenuated CEU-025 cytotoxicity, while its suppression using TRX1-specific siRNA increased the effects of CEU-025, suggesting that loss of function of TRX1 is involved, at least in part, in the cytotoxic activity of CEU-025. These results suggest that CEU-025 and CEU-027 exhibit anticancer activity through a novel, unique mechanism of action. The importance of TRX1 and the dependence of the cytotoxicity of CEU-025 and CEU-027 on TRX1 intracellular localization are also discussed.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Tiorredoxinas/metabolismo , Urea/análogos & derivados , Urea/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Catálisis , Línea Celular Tumoral , Cisplatino/farmacología , Humanos , Transporte de Proteínas/efectos de los fármacos , Relación Estructura-Actividad
15.
J Pharm Pharm Sci ; 13(2): 175-90, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20816004

RESUMEN

PURPOSE: We investigated the involvement of MAPK signaling in the cell death mechanisms of classical microtubule interfering agents (MIA) and aryl-3-(2-chloroethyl)ureas (CEU) acting as antimitotics, along with CEU that don't affect directly microtubules (non-MIA CEU). METHODS: To ascertain the activated signaling pathway profile of MIA and non-MIA CEU, Western blot, immunoprecipitation and transfection experiments were performed. RESULTS: Non-MIA CEU do not activate p38, as opposed to MIA, and the extent of ERK and JNK activation is lower than in response to MIA. The effect of MIA and non-MIA CEU on focal adhesion associated protein was also studied; MIA were shown to induce focal adhesion dismantlement associated with a sustained increase in paxillin phosphorylation and FAK cleavage, as opposed to non-MIA CEU. In addition, bcl-2 phosphorylation and AKT cleavage, induced by all MIA tested, was not observed in response to non-MIA CEU further emphasizing the differential cell death mechanisms induced by MIA and non-MIA CEU. Pharmacologic and genetic approaches emphasize that the ASK1-p38 pathway activation contributes to the cytotoxic mechanism of MIA, in contrast to non-MIA CEU. ASK1-p38 is important for increased paxillin phosphorylation and FAK cleavage, suggesting that ASK-1-p38 is an upstream event of FA structure dismantlement induced by MIA. Moreover, the endogen inhibitor of ASK-1, thioredoxin, is released from ASK-1 in response to MIA as opposed to non-MIA CEU. CONCLUSION: Our study supports that ASK1-p38 activation is an important signaling event, induced by MIA, which impairs focal adhesion structure and induces anchorage-dependent apoptosis or anoikis.


Asunto(s)
Antineoplásicos/farmacología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Moduladores de Tubulina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Anoicis/efectos de los fármacos , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular , Línea Celular Tumoral , Adhesiones Focales/metabolismo , Humanos , Ratones , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transfección , Urea/análogos & derivados , Urea/farmacología
16.
Eur J Med Chem ; 45(7): 2928-37, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20400211

RESUMEN

Seven subsets of aromatic urea and amide analogues of N-phenyl-N'-(2-chloroethyl)ureas (CEU) have been synthesized by nucleophilic addition of 3-chloropropylisocyanate, 2-chloroacetylisocyanate, ethylisocyanate, 2-chloroacetyl chloride, 3-chloropropanoyl chloride, 4-chlorobutanoyl chloride, and acryloyl chloride, respectively, to selected anilines or benzylamines to afford 3-chloropropylureas (1, CPU), 2-chloroacetylureas (2, CAU), ethylureas (3, EU), 2-chloroacetamides (4, CA), 3-chloropropionamides (5, CPA), 4-chlorobutyramides (6, CBA) and acrylamides (7, Acr). The molecular structure of these compounds has been confirmed by IR, (1)H and (13)C NMR, and MS spectra and their purity also confirmed by HPLC. The CEU analogues were evaluated for their antiproliferative activity against three human tumor cell lines, namely human colon carcinoma HT-29, human skin melanoma M21, and human breast carcinoma MCF-7. CAU (2c to 2g), CA (4a to 4d, 4f and 4 g), CPA (5a) and Acr (7a and 7b) had IC(50) ranging from 1.4 to 25 microM. CAU, CA, CPA and Acr exhibited interesting antiproliferative activity through mechanism(s) of action unrelated to the acylation of glutamic acid at position 198 on beta-tubulin that is characterizing CEU.


Asunto(s)
Amidas/química , Amidas/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Urea/química , Urea/farmacología , Amidas/síntesis química , Amidas/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colchicina/metabolismo , Humanos , Relación Estructura-Actividad , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Urea/síntesis química , Urea/metabolismo
17.
Biol Proced Online ; 12(1): 113-7, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21406120

RESUMEN

Development of antimitotic binding to the colchicine-binding site for the treatment of cancer is rapidly expanding. Numerous antimicrotubule agents are prepared every year, and the determination of their binding affinity to tubulin requires the use of purified tubulins and radiolabeled ligands. Such a procedure is costly and time-consuming and therefore is limited to the most promising candidates. Here, we report a quick and inexpensive method that requires only usual laboratory resources to assess the binding of antimicrotubules to colchicine-binding site. The method is based on the ability of N,N'-ethylene-bis(iodoacetamide) (EBI) to crosslink in living cells the cysteine residues at position 239 and 354 of ß-tubulin, residues which are involved in the colchicine-binding site. The ß-tubulin adduct formed by EBI is easily detectable by Western blot as a second immunoreacting band of ß-tubulin that migrates faster than ß-tubulin. The occupancy of colchicine-binding site by pertinent antimitotics inhibits the formation of the EBI: ß-tubulin adduct, resulting in an assay that allows the screening of new molecules targeting this binding site.

18.
Br J Pharmacol ; 158(5): 1375-86, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19785654

RESUMEN

BACKGROUND AND PURPOSE: A peptide bradykinin (BK) B(2) receptor agonist partially resistant to degradation, B-9972, down-regulates this receptor subtype. We have used another recently described non-peptide agonist, compound 47a, as a tool to study further the effects of metabolically more stable and thus persistent, agonists of the BK B(2) receptor on signalling, desensitization and down-regulation of this receptor. EXPERIMENTAL APPROACH AND KEY RESULTS: Compound 47a was a partial agonist at the B(2) receptor in the human umbilical vein, where it shared with B-9972 a very slow relaxation on washout, and in HEK 293 cell lines expressing tagged forms [myc, green fluorescent protein (GFP)] of the rabbit B(2) receptor. Compound 47a desensitized the umbilical vein to BK. In the cellular systems, the inactivation-resistant agonists induced [Ca(2+)](i) transients as brief as those of BK but affected other functions with a longer duration than BK [12 h; receptor endocytosis, endosomal beta-arrestin(1/2) translocation, protein kinase C-dependent extracellular signal-regulated kinases (ERK)1/2 phosphorylation and c-Fos expression]. The B(2) receptor-GFP was degraded in cells exposed to B-9972 or compound 47a for 12 h. The non-peptide B(2) receptor antagonist LF 16-0687 prevented all effects of compound 47a, which were also absent in cells lacking recombinant B(2) receptors. CONCLUSION AND IMPLICATIONS: Inactivation-resistant agonists revealed a long-lasting assembly of the agonist-B(2) receptor-beta-arrestin complexes in endosomal structures and induce 'biased signalling' (in terms of activation of ERK and c-Fos) as a function of time. Further, B-9972 and compound 47a, unlike BK, efficiently down-regulated BK B(2) receptors.


Asunto(s)
Aminopiridinas/farmacología , Bencimidazoles/farmacología , Receptor de Bradiquinina B2/agonistas , Receptor de Bradiquinina B2/fisiología , Animales , Arrestinas/metabolismo , Unión Competitiva , Antagonistas del Receptor de Bradiquinina B2 , Calcio/metabolismo , Línea Celular , Regulación hacia Abajo , Humanos , Técnicas In Vitro , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Oligopéptidos/farmacología , Quinolinas/farmacología , Conejos , Ensayo de Unión Radioligante , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/antagonistas & inhibidores , Transducción de Señal , Venas Umbilicales/efectos de los fármacos , Venas Umbilicales/metabolismo , beta-Arrestina 1 , beta-Arrestinas
19.
Chem Phys Lipids ; 158(2): 91-101, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19428353

RESUMEN

Gene and synthetic drug-delivery vectors have been developed and characterized to treat several genetic diseases and cancers. Our study aims at characterizing cationic liposomes containing the zwitterionic phospholipid DMPC and the cationic lipid DOTAP as well as their interactions with two types of DNA and a new class of antineoplastic agents derived from arylchloroethylureas (CEU). Results obtained using FTIR spectroscopy as well as (31)P and (2)H NMR indicate that DMPC and DOTAP form cationic liposomes in a highly disordered fluid phase at a molar ratio of 1:1. In addition, the FTIR results indicate that the presence of DNA or CEUs within the liposomes does not significantly affect the conformational order of both the DMPC and DOTAP acyl chains. Our results therefore provide a detailed characterization of complexes between cationic liposomes and both DNA and drugs and indicate that these complexes are stable and fluid assemblies.


Asunto(s)
ADN/metabolismo , Dimiristoilfosfatidilcolina/química , Ácidos Grasos Monoinsaturados/química , Liposomas/química , Liposomas/metabolismo , Compuestos de Fenilurea/metabolismo , Compuestos de Amonio Cuaternario/química , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Cationes/química , Bovinos , ADN/química , Dimiristoilfosfatidilcolina/metabolismo , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Resonancia Magnética Nuclear Biomolecular , Compuestos de Fenilurea/química , Plásmidos , Compuestos de Amonio Cuaternario/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier , Temperatura , Timo/química
20.
Bioorg Med Chem ; 17(10): 3690-7, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19398206

RESUMEN

Computational tools such as CoMSIA and CoMFA models reported in a recent study revealed the structure-activity relationships ruling the interactions occurring between hydrophobic N-phenyl-N'-(2-chloroethyl)ureas (CEU) and the colchicine-binding site (C-BS) on beta(II)-tubulin. Here, we describe the mechanisms involved in the covalent binding of three subsets of CEU derivatives to the C-BS. The FlexiDock experiments confirmed that the interaction of non-covalent portions of the CEU auxophore moiety of CEU is involved in the binding of the drug to the C-BS facilitate the nucleophilic attack of Glu-beta198 rather than Cys-beta239. In addition, these studies suggest that Cys-beta239 together with Asn-alpha99, Ser-alpha176, Thr-alpha177, Leu-beta246, Asn-beta247, Ala-beta248, Lys-beta252 and Asn-beta256 are implicated in the stabilization of a C-BS-CEU complex prior to the acylation of Glu-beta198 by CEU. Our molecular models propose the formation of a stabilized C-BS-CEU complex before the completion of the Glu-beta198 acylation; acylation triggering conformational changes of beta-tubulin, microtubule depolymerization and anoikis. The computational models presented here might be useful to the design of selective and more potent C-BS inhibitors. Of interest, in vivo acylation of acidic amino acid residues by xenobiotics is an unusual reaction and may open new approaches for the design of irreversible protein inhibitors such as tubulin.


Asunto(s)
Colchicina/metabolismo , Moduladores de Tubulina/química , Tubulina (Proteína)/metabolismo , Urea/análogos & derivados , Sitios de Unión , Línea Celular Tumoral , Colchicina/química , Simulación por Computador , Humanos , Modelos Químicos , Modelos Moleculares , Relación Estructura-Actividad , Tubulina (Proteína)/química , Moduladores de Tubulina/metabolismo , Moduladores de Tubulina/farmacología , Urea/química , Urea/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...