Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Signal ; 23(4): 630-40, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21138763

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Triazenos/farmacologia , Apoptose/efeitos dos fármacos , Butadienos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , Metilação de DNA , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Sinergismo Farmacológico , Ativação Enzimática , Gefitinibe , Humanos , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Nitrilas/farmacologia , Fosforilação , Temozolomida
2.
J Pharmacol Exp Ther ; 334(1): 9-20, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20348204

RESUMO

Within the context of a new tumor-targeting strategy termed "combi-targeting," we designed RB24 to inhibit epidermal growth factor receptor (EGFR) or Her2 phosphorylation and to further degrade upon hydrolysis to 4-(3'-bromophenylamino)-6-aminoquinazoline (RB10; another EGFR/Her2 inhibitor) plus a strong DNA-alkylating species. 6-(3-Acetoxymethyl-3-methyltriazenyl)-4-(3'-bromophenylamino)quinazoline (RB24) showed significant antiproliferative activity against human breast cancer cells, and transfection of one such cell line, MDA-MB-435, with ErbB1 or ErbB2 (Her2) dramatically enhanced cell death by apoptosis. RB24 was capable of releasing 2- to 3-fold higher levels of RB10 in the transfectants than in their wild-type counterparts. More importantly, RB10 was abundantly distributed in the perinuclear region of the cells, and its elevated levels in the ErbB transfectants were concomitant with increased levels of DNA lesions in the latter cells. This selectivity could be abolished by coincubation of the cells with exogenous RB10, suggesting that the entire combi-molecule may bind primarily to its cognate perinuclear sites before degradation. This localization may exert a bystander effect, allowing the alkylating species to be abundantly propagated into the nucleus. Cell response to this novel targeting mechanism was mediated by 1) activation of c-Jun NH(2)-terminal kinase in response to DNA damage and 2) down-regulation of Bad through blockade of EGFR tyrosine kinase activity: two events that cooperatively converged into enhancement of apoptosis in the oncogene-transfected cells.


Assuntos
Antineoplásicos/farmacologia , Dano ao DNA , Receptores ErbB/antagonistas & inibidores , Quinazolinas/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Triazenos/farmacologia , Antineoplásicos/química , Antineoplásicos/metabolismo , Técnicas de Cultura de Células , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaio Cometa , Receptores ErbB/biossíntese , Citometria de Fluxo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Microscopia de Fluorescência , Estrutura Molecular , Proteína Oncogênica v-akt/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Quinazolinas/química , Quinazolinas/metabolismo , Receptor ErbB-2/biossíntese , Transfecção , Triazenos/química , Triazenos/metabolismo , Proteína de Morte Celular Associada a bcl/metabolismo
3.
Clin Cancer Res ; 13(1): 331-40, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17200372

RESUMO

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.


Assuntos
DNA/química , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/metabolismo , Compostos de Nitrosoureia/farmacologia , Animais , Linhagem Celular Tumoral , Ensaio Cometa , Dano ao DNA , Humanos , Concentração Inibidora 50 , Masculino , Espectrometria de Massas , Camundongos , Microscopia de Fluorescência , Transplante de Neoplasias , Fosforilação
4.
Anticancer Drugs ; 18(2): 171-7, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17159603

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Ensaio Cometa , Dano ao DNA/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Ensaio de Imunoadsorção Enzimática , Fibroblastos/efeitos dos fármacos , Genes erbB-1/efeitos dos fármacos , Genes erbB-1/genética , Genes erbB-2/efeitos dos fármacos , Genes erbB-2/genética , Humanos , Masculino , Camundongos , Células NIH 3T3 , Fosforilação , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia
5.
J Med Chem ; 49(12): 3544-52, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16759097

RESUMO

According to the "combi-targeting" concept, the EGFR tyrosine kinase (TK) inhibitory potency of compounds termed "combi-molecules" is critical for selective growth inhibition of tumor cells with disordered expression of EGFR or its closest family member erbB2. Here we report on the optimization of the EGFR TK inhibitory potency of the combi-molecules of the nitrosourea class by comparison with their aminoquinazoline and ureidoquinazoline precursors. This led to the discovery of a new structural parameter that influences their EGFR TK inhibitory potency, i.e., the torsion angle between the plane of the quinazoline ring and the ureido or the nitrosoureido moiety of the synthesized drugs. Compounds (3'-Cl and Br series) with small angles (0.5-3 degrees ) were generally stronger EGFR TK inhibitors than those with large angles (18-21 degrees ). This was further corroborated by ligand-receptor van der Waals interaction calculations that showed significant binding hindrance imposed by large torsion angles in the narrow ATP cleft of EGFR. Selective antiproliferative studies in a pair of mouse fibroblast NIH3T3 cells, one of which NIH3T3/neu being transfected with the erbB2 oncogene, showed that IC(50) values for inhibition of EGFR TK could be good predictors of their selective potency against the serum-stimulated growth of the erbB2-tranfected cell line (Pearson r = 0.8). On the basis of stability (t(1/2)), EGFR TK inhibitory potency (IC(50)), and selective erbB2 targeting, compound 23, a stable nitrosourea, was considered to have the structural requirements for further development.


Assuntos
Antineoplásicos/síntese química , Receptores ErbB/antagonistas & inibidores , Etilnitrosoureia/análogos & derivados , Compostos de Nitrosoureia/síntese química , Compostos de Anilina/síntese química , Compostos de Anilina/química , Compostos de Anilina/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Estabilidade de Medicamentos , Etilnitrosoureia/síntese química , Etilnitrosoureia/química , Etilnitrosoureia/farmacologia , Ligantes , Camundongos , Modelos Moleculares , Conformação Molecular , Células NIH 3T3 , Compostos de Nitrosoureia/química , Compostos de Nitrosoureia/farmacologia , Receptor ErbB-2/genética , Relação Estrutura-Atividade , Termodinâmica , Transfecção
6.
Prostate ; 59(1): 13-21, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-14991862

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Compostos de Nitrosoureia/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Carmustina/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaio Cometa , Dano ao DNA , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes fos/efeitos dos fármacos , Humanos , Masculino , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , RNA Neoplásico/química , RNA Neoplásico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos
7.
Bull Cancer ; 91(12): 911-5, 2004 Dec.
Artigo em Francês | MEDLINE | ID: mdl-15634632

RESUMO

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".


Assuntos
Antineoplásicos Alquilantes/síntese química , Receptores ErbB/antagonistas & inibidores , Compostos de Nitrosoureia/síntese química , Quinazolinas/síntese química , Antineoplásicos Alquilantes/uso terapêutico , Dano ao DNA , DNA de Neoplasias/efeitos dos fármacos , Compostos de Nitrosoureia/uso terapêutico , Quinazolinas/uso terapêutico
8.
J Med Chem ; 46(25): 5546-51, 2003 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-14640561

RESUMO

The synthesis of a novel acetoxymethyltriazene designed to be a prodrug of multiple inhibitors of the epidermal growth factor receptor (EGFR) and a methyldiazonium species is described. Studies with each of the expected metabolites demonstrated significant EGFR tyrosine kinase inhibitory activities and the released methyldiazonium was trapped with p-nitrobenzylpyridine. Their ability to damage genomic DNA in whole cells was demonstrated by using the single cell microelectrophoresis (comet) assay. The results suggest that this approach may well represent a novel drug combination strategy involving single molecules masking multiple bioactive agents.


Assuntos
Antineoplásicos Alquilantes/síntese química , Inibidores Enzimáticos/administração & dosagem , Receptores ErbB/antagonistas & inibidores , Pró-Fármacos/síntese química , Triazenos/síntese química , Animais , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacologia , Linhagem Celular , Ensaio Cometa , Dano ao DNA , Compostos de Diazônio/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/metabolismo , Humanos , Camundongos , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacologia , Relação Estrutura-Atividade , Triazenos/metabolismo , Triazenos/farmacologia
9.
Cancer Chemother Pharmacol ; 51(1): 1-10, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12497200

RESUMO

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.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Dano ao DNA , Receptores ErbB/antagonistas & inibidores , Compostos de Nitrosoureia/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Divisão Celular/efeitos dos fármacos , Humanos , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Fosforilação , Proteínas Tirosina Quinases , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA