Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Radiat Oncol Biol Phys ; 92(2): 399-406, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25823448

RESUMO

PURPOSE: ZRBA1 is a combi-molecule designed to induce DNA alkylating lesions and to block epidermal growth factor receptor (EGFR) TK domain. Inasmuch as ZRBA1 downregulates the EGFR TK-mediated antisurvival signaling and induces DNA damage, we postulated that it might be a radiosensitizer. The aim of this study was to further investigate the potentiating effect of ZRBA1 in combination with radiation and to elucidate the possible mechanisms of interaction between these 2 treatment modalities. METHODS AND MATERIALS: The triple negative human breast MDA-MB-468 cancer cell line and mouse mammary cancer 4T1 cell line were used in this study. Clonogenic assay, Western blot analysis, and DNA damage analysis were performed at multiple time points after treatment. To confirm our in vitro findings, in vivo tumor growth delay assay was performed. RESULTS: Our results show that a combination of ZRBA1 and radiation increases the radiation sensitivity of both cell lines significantly with a dose enhancement factor of 1.56, induces significant numbers of DNA strand breaks, prolongs higher DNA damage up to 24 hours after treatment, and significantly increases tumor growth delay in a syngeneic mouse model. CONCLUSIONS: Our data suggest that the higher efficacy of this combination could be partially due to increased DNA damage and delayed DNA repair process and to the inhibition of EGFR. The encouraging results of this combination demonstrated a significant improvement in treatment efficiency and therefore could be applicable in early clinical trial settings.


Assuntos
Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Terapia de Alvo Molecular , Quinazolinas/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Tolerância a Radiação/efeitos da radiação , Triazenos/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/radioterapia , Animais , Linhagem Celular Tumoral , Terapia Combinada/métodos , Quebras de DNA de Cadeia Dupla , Quebras de DNA de Cadeia Simples , Dano ao DNA/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Fatores de Tempo , Neoplasias de Mama Triplo Negativas/genética , Ensaio Tumoral de Célula-Tronco
2.
PLoS One ; 10(2): e0117215, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25658745

RESUMO

Cancer cells are characterized by a complex network of interrelated and compensatory signaling driven by multiple kinases that reduce their sensitivity to targeted therapy. Therefore, strategies directed at inhibiting two or more kinases are required to robustly block the growth of refractory tumour cells. Here we report on a novel strategy to promote sustained inhibition of two oncogenic kinases (Kin-1 and Kin-2) by designing a molecule K1-K2, termed "combi-molecule", to induce a tandem blockade of Kin-1 and Kin-2, as an intact structure and to be further hydrolyzed to two inhibitors K1 and K2 directed at Kin-1 and Kin-2, respectively. We chose to target EGFR (Kin-1) and c-Src (Kin-2), two tyrosine kinases known to synergize to promote tumour growth and progression. Variation of K1-K2 linkers led to AL776, our first optimized EGFR-c-Src targeting prototype. Here we showed that: (a) AL776 blocked EGFR and c-Src as an intact structure using an in vitro kinase assay (IC50 EGFR = 0.12 µM and IC50 c-Src = 3 nM), (b) it could release K1 (AL621, a nanomolar EGFR inhibitor) and K2 (dasatinib, a clinically approved Abl/c-Src inhibitor) by hydrolytic cleavage both in vitro and in vivo, (c) it could robustly inhibit phosphorylation of EGFR and c-Src (0.25-1 µM) in cells, (d) it induced 2-4 fold stronger growth inhibition than gefitinib or dasatinib and apoptosis at concentrations as low as 1 µM, and, (e) blocked motility and invasion at sub-micromolar doses in the highly invasive 4T1 and MDA-MB-231 cells. Despite its size (MW = 1032), AL776 blocked phosphorylation of EGFR and c-Src in 4T1 tumours in vivo. We now term this new targeting model consisting of designing a kinase inhibitor K1-K2 to target Kin-1 and Kin-2, and to further release two inhibitors K1 and K2 of the latter kinases, "type III combi-targeting".


Assuntos
Apoptose/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Inibidores de Proteínas Quinases/toxicidade , Quinazolinas/toxicidade , Tiazóis/toxicidade , Quinases da Família src/antagonistas & inibidores , Animais , Sítios de Ligação , Proteína Tirosina Quinase CSK , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dasatinibe/toxicidade , Desenho de Fármacos , Receptores ErbB/metabolismo , Feminino , Gefitinibe , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Simulação de Dinâmica Molecular , Células NIH 3T3 , Neoplasias/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/uso terapêutico , Estrutura Terciária de Proteína , Quinazolinas/síntese química , Quinazolinas/uso terapêutico , Tiazóis/síntese química , Tiazóis/uso terapêutico , Transplante Heterólogo , Quinases da Família src/metabolismo
3.
Chem Biol Drug Des ; 85(2): 153-62, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25092264

RESUMO

To potentiate the quinazoline-based inhibitor of the epidermal growth factor receptor (EGFR), a chloroethyl alkylating moiety was appended to its 6-position. This led to molecules with extremely strong EGFR inhibitory potency and anomalously strong DNA-damaging potential. To assess the role of the chloroethyl group on potency, we designed a molecule in which it is shifted to the 7-position where it would be less reactive and away from the cys773 of the EGFR ATP site. The results showed that (i) ZR2009 was 10-fold less potent than its positional isomer ZR2003 in EGFR tyrosine kinase inhibition, (ii) it consistently exhibited significantly weaker antiproliferative potency than ZR2003, (iii) in reversibility assays, while ZR2003 induced sustained inhibition of EGFR phosphorylation, ZR2009 inhibitory activity was partially reversed, and (iv) likewise, ZR2009 significantly lost its activity in short exposure growth inhibitory assays and induced lower levels of DNA damage than ZR2003. Molecular modeling suggested that while the chloroethylamino group in ZR2003 was at 3.5 Å away from Cys773, that of ZR2009 was at 6.3 Å. The results in toto suggest that, while the chloroethyl is a strong alkylating group, its appendage to the 6-position is optimal for DNA damage, sustained EGFR, and growth inhibition.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Dano ao DNA/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Compostos de Mostarda Nitrogenada/química , Compostos de Mostarda Nitrogenada/farmacologia , Quinazolinas/química , Quinazolinas/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/metabolismo , Humanos , Isomerismo , Camundongos , Modelos Moleculares , Células NIH 3T3 , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais/efeitos dos fármacos
4.
Invest New Drugs ; 31(6): 1409-23, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23959266

RESUMO

Capecitabine, an orally available prodrug of 5-FU, requires activation by carboxylesterase (CES) enzymes present in the liver to generate 5'-deoxy-5-flurocytidine ribose (5'-DFCR). The deamination of the latter by cytidine deaminase gives 5'-deoxy-5-fluorouridine ribose (5'-DFUR). Finally, the conversion of 5'-DFUR to the cytotoxic drug 5-FU, occurs primarily in the tumour and is catalyzed by thymidine phosphorylase (TP). Accordingly, it was surmised that events associated with an increase of TP levels should enhance the potency of capecitabine and its metabolites. EGFR inhibition was found to be one such event. The observed synergy between gefitinib and 5'-DFUR has inspired the design of single molecules capable of acting as prodrugs of both an EGFR inhibitor and 5-FU. Here, we report on the synthesis and characterization of one such molecule, ZRX1, that consists of an acetylated 5'-DFCR moiety linked to a quinazoline inhibitor of EGFR through an alkyl dicarbamate spacer that requires CES activation to generate the two active metabolites. Our results showed that ZRX1 was ineffective as an intact molecule. However, when CES was present, ZRX1 induced an increase in EGFR inhibition, TP expression, DNA damage and apoptosis. ZRX1 was, at least, 3-fold more potent than capecitabine and 5'-DFUR and recapitulated the effects of the combination treatments. LC-MS analysis showed that in the presence of CES, ZRX1 is metabolized into a mixture of bioactive quinazoline derivatives and 5'-DFCR derived metabolites. Our results in toto, suggest that capecitabine-based EGFR targeting combi-molecules of the same type than ZRX1, have the potential to induce stronger growth inhibitory potency than capecitabine, 5'-DFUR or single EGFR inhibitors and equivalent potency when compared with combinations of EGFR inhibitors + 5'-DFUR.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carbamatos/farmacologia , Carboxilesterase/metabolismo , Desoxicitidina/análogos & derivados , Receptores ErbB/antagonistas & inibidores , Neoplasias/metabolismo , Quinazolinas/farmacologia , Animais , Antineoplásicos/administração & dosagem , Capecitabina , Linhagem Celular Tumoral , Desoxicitidina/administração & dosagem , Desoxicitidina/farmacologia , Receptores ErbB/metabolismo , Fluoruracila/administração & dosagem , Fluoruracila/análogos & derivados , Humanos , Hidrólise , Camundongos , Camundongos Endogâmicos BALB C , Timidina Fosforilase/metabolismo
5.
J Signal Transduct ; 2012: 282050, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22523681

RESUMO

Recently, we reported the combination of multitargeted ErbB1 inhibitor-DNA damage combi-molecules with OCT in order to downregulate ErbB1 and activate SSTRs. Absence of translation to cell kill was believed to be partially due to insufficient ErbB1 blockage and DNA damage. In this study, we evaluated cell response to molecules that damage DNA more aggressively and induce stronger attenuation of ErbB1 phosphorylation. We used three cell lines expressing low levels (U87MG) or transfected to overexpress wildtype (U87/EGFR) or a variant (U87/EGFRvIII) of ErbB1. The results showed that Iressa ± HN2 and the combi-molecules, ZRBA4 and ZR2003, significantly blocked ErbB1 phosphorylation in U87MG cells. Addition of OCT significantly altered cell cycle distribution. Analysis of the DNA damage response pathway revealed strong upregulation of p53 by HN2 and the combi-molecules. Apoptosis was only induced by a 48 h exposure to HN2. All other treatments resulted in cell necrosis. This is in agreement with Akt-Bad pathway activation and survivin upregulation. Despite strong DNA damaging properties and downregulation of ErbB1 phosphorylation by these molecules, the strongest effect of SSTR activation was on cell cycle distribution. Therefore, any enhanced antiproliferative effects of combining ErbB1 inhibition with SSTR activation must be addressed in the context of cell cycle arrest.

6.
Prostate ; 72(12): 1273-85, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22290742

RESUMO

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.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , DNA/antagonistas & inibidores , Sistemas de Liberação de Medicamentos/métodos , Compostos de Mostarda Nitrogenada/administração & dosagem , Neoplasias da Próstata/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , DNA/metabolismo , Humanos , Masculino , Camundongos , Células NIH 3T3 , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais/fisiologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
7.
Breast Cancer Res Treat ; 133(1): 217-26, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21915635

RESUMO

Capecitabine (Xeloda) is a prodrug of 5-FU used in the clinical management of advanced breast cancer. It is metabolized first in the liver by carboxylesterases to generate 5'-deoxy-5-flurocytidine ribose (5'-DFCR), which is subsequently converted to 5'-deoxy-5-fluorouridine ribose (5'-DFUR) by cytidine deaminase in tumour and normal tissues. The conversion of 5'-DFUR to the cytotoxic 5-FU, occurs primarily in the tumour and is catalyzed by thymidine phosphorylase (TP). Prior work in head and neck cancer showed that cell treatment with an inhibitor of the epidermal growth receptor (EGFR) gefitinib led to an increase in TP expression and sensitized them to 5'-DFUR. This work seeks to investigate the factors influencing the potency of gefitinib + 5'-DFUR combination. Here, we studied these factors in a panel of six human breast cancer cell lines, with varied levels of sensitivity to gefitinib. Our results first confirmed that 5'-DFUR potency linearly correlates with TP basal levels in the panel of cell lines. In contrast, the strength of the synergistic effect of the gefitinib + 5'-DFUR combination, as measured by their combination indices (CI) correlates with pEGFR percent inhibition and with the modulation of TP expression by gefitinib (as quantitated by TP fold change) rather than TP basal levels. The results, in toto, suggest that the extent of modulation of TP by gefitinib may be used as a predictor of tumour sensitivity to gefitinib + capecitabine/5'-DFUR combinations.


Assuntos
Antineoplásicos/farmacologia , Desoxicitidina/análogos & derivados , Receptores ErbB/metabolismo , Floxuridina/farmacologia , Fluoruracila/análogos & derivados , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Quinazolinas/farmacologia , Neoplasias da Mama , Capecitabina , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/farmacologia , Sinergismo Farmacológico , Feminino , Fluoruracila/farmacologia , Gefitinibe , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Metotrexato/farmacologia , Fosforilação , Timidina Fosforilase/genética , Timidina Fosforilase/metabolismo , Timidilato Sintase/antagonistas & inibidores , Timidilato Sintase/genética , Timidilato Sintase/metabolismo
8.
J Pharm Biomed Anal ; 56(3): 592-9, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21764236

RESUMO

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.


Assuntos
Antinematódeos/farmacologia , DNA/antagonistas & inibidores , DNA/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Neoplasias/tratamento farmacológico , Compostos de Mostarda Nitrogenada/farmacologia , Quinazolinas/farmacologia , Triazenos/farmacologia , Animais , Antinematódeos/farmacocinética , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Masculino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Compostos de Mostarda Nitrogenada/farmacocinética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/genética , Quinazolinas/farmacocinética , Neoplasias Vulvares/tratamento farmacológico , Neoplasias Vulvares/enzimologia , Neoplasias Vulvares/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
9.
Drug Metab Lett ; 5(2): 141-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21457144

RESUMO

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.


Assuntos
Carbamatos/farmacocinética , Receptores ErbB/antagonistas & inibidores , Pró-Fármacos/farmacocinética , Quinazolinas/farmacocinética , Triazenos/farmacocinética , Animais , Agregação Celular , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Distribuição Tecidual
10.
Mol Cancer Res ; 9(3): 320-31, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21263032

RESUMO

To enhance the potency of current EGFR inhibitors, we developed a novel strategy that seeks to confer them an additional DNA damaging function, leading to the design of drugs termed combi-molecules. ZRS1 is a novel combi-molecule that contains an EGFR tyrosine kinase targeting quinazoline arm and a methyltriazene-based DNA damaging one. We examined its effect on human tumor cell lines with varied levels of EGFR and O6-methylguanine DNA methyltransferase (MGMT). ZRS1 was more potent than the clinical methylating agent temozolomide in all cell lines, regardless of their MGMT status. However, its potency was in the same range as or less than that of Iressa, an EGFR inhibitor, against MGMT-proficient cells. In the MGMT-deficient or in MGMT-proficient cells exposed to the MGMT inhibitor O6-benzylguanine, its potency was superior to that of Iressa and temozolomide or a temozolomide+Iressa combination. Cell signaling analysis in A549 (MGMT(+)) and A427 (MGMT(-)) showed that ZRS1 strongly inhibited EGFR phosphorylation and related signaling pathways. In addition, the p53 pathway was activated by DNA damage in both cell lines, but apoptosis was significantly more pronounced in A427 cells. Using MGMT shRNA to block endogenous MGMT protein expression in A549 resulted in significant sensitization to ZRS1. Furthermore, transfection of MGMT into A427 greatly decreased the potency of ZRS1. These results conclusively show that MGMT is a critical molecular determinant for the full-blown potency of the dual EGFR-DNA targeting combi-molecule.


Assuntos
Antineoplásicos/farmacologia , Metilação de DNA/efeitos dos fármacos , Dacarbazina/análogos & derivados , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Quinazolinas/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dacarbazina/síntese química , Dacarbazina/farmacologia , Compostos de Diazônio/síntese química , Compostos de Diazônio/farmacologia , Inibidores Enzimáticos/farmacologia , Gefitinibe , Humanos , Fosforilação/efeitos dos fármacos , Quinazolinas/síntese química , Transdução de Sinais/efeitos dos fármacos , Temozolomida , Proteína Supressora de Tumor p53/metabolismo
11.
Invest New Drugs ; 29(5): 833-45, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20428924

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Carbamatos/química , Carbamatos/farmacologia , DNA de Neoplasias/metabolismo , Receptores ErbB/antagonistas & inibidores , Triazenos/química , Triazenos/farmacologia , Animais , Antineoplásicos/química , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Ensaios Enzimáticos , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Citometria de Fluxo , Humanos , Hidrólise/efeitos dos fármacos , Camundongos , Microscopia de Fluorescência , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia
13.
Anticancer Drugs ; 20(8): 659-67, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19581798

RESUMO

ZRBA1 is a molecule termed 'combi-molecule' designed to induce DNA-alkylating lesions and to block epidermal growth factor receptor (EGFR) tyrosine kinase. Owing to its ability to downregulate the EGFR tyrosine kinase-mediated antiapoptotic signaling and DNA repair proteins, we inferred that it could significantly sensitize cells to ionizing radiation. Using the MDA-MB-468 human breast cancer cell line in which ZRBA1 has already been reported to induce significant EGFR/DNA-targeting potency, the results showed that: (i) concurrent administration of ZRBA1 and 4 Gy radiation led to a significant decrease in cell viability, (ii) the greater efficacy of the combination was sequential, being limited to conditions wherein the drug was administered concurrently with radiation or before radiation, and (iii) the efficacy enhancement of the combination was further confirmed by clonogenic assays from which a dose enhancement factor of 1.34 could be observed at survival fraction of 0.01. Flow cytometric analysis showed significant enhancement of cell cycle arrest in G2/M (P<0.046, irradiated cells vs. cells treated with ZRBA1 and radiation) and increased apoptosis when ZRBA1 was combined with radiation. Likewise, significant levels of double-strand breaks were observed for the combination, as determined by neutral comet assay (P<0.045, irradiated cells vs. cells treated with ZRBA1 and radiation). These results in toto suggest that the superior efficacy of the ZRBA1 plus radiation combination may be secondary to the ability of ZRBA1 to arrest the cells in G2/M, a cell cycle phase in which tumor cells are sensitive to radiation. Furthermore, the increased levels of DNA damage, combined with the concomitant downregulation of EGFR-mediated signaling by ZRBA1, may account for the significant levels of cell killing induced by the combination.


Assuntos
Quinazolinas/farmacologia , Radiossensibilizantes/farmacologia , Triazenos/farmacologia , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos/efeitos da radiação , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/efeitos da radiação , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/efeitos da radiação , Receptores ErbB/antagonistas & inibidores , Gefitinibe , Humanos , Microscopia de Fluorescência , Quinazolinas/metabolismo , Quinazolinas/farmacocinética , Quinazolinas/efeitos da radiação , Radiossensibilizantes/metabolismo , Radiossensibilizantes/farmacocinética , Radiossensibilizantes/efeitos da radiação , Triazenos/metabolismo , Triazenos/farmacocinética , Triazenos/efeitos da radiação , Ensaio Tumoral de Célula-Tronco , Raios X
14.
Drug Metab Lett ; 3(1): 1-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19356110

RESUMO

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.


Assuntos
Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacocinética , Receptores ErbB/metabolismo , Triazenos/metabolismo , Triazenos/farmacocinética , Animais , Antineoplásicos Alquilantes/uso terapêutico , Biotransformação , Cromatografia Líquida de Alta Pressão , Receptores ErbB/antagonistas & inibidores , Humanos , Masculino , Espectrometria de Massas , Camundongos , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacocinética , Espectrofotometria Ultravioleta , Distribuição Tecidual , Triazenos/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Mol Cancer Ther ; 7(5): 1033-43, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18483293

RESUMO

Blockade of Bcr-Abl by the inhibitor Imatinib has proven efficacious in the therapy of chronic myelogenous leukemia (CML). However resistance to the drug emerges at the advanced phases of the disease. Therefore, novel therapy models remained to be designed. We have developed a novel dual targeted agent termed "combi-molecule" designed to not only block Bcr-Abl but also damage DNA. ZRF1, the first optimized prototype of the approach, was "programmed" to degrade into another inhibitor ZRF0 plus a methyl diazonium species. It was approximately 2-fold stronger Abl tyrosine kinase inhibitor than Imatinib and a more potent DNA-damaging agent than Temodal. In the p53 wild-type Mo7p210 cells, the potency of ZRF1 was approximately 1,000-fold superior to that of the equieffective combinations of Imatinib plus Temodal. More importantly, its superior potency over Imatinib was more pronounced in Bcr-Abl-positive cells coexpressing wild-type p53. Studies to rationalize these results showed that, through its Bcr-Abl inhibitory function, it down-regulated p53. However, sufficient level of the latter protein was available for transactivating p21 and Bax, which are required for cell cycle arrest and apoptosis. The results suggest that, in p53 wild-type cells, apoptosis is induced not only through Bcr-Abl inhibition but also through the p53-controlled DNA-damaging pathway, leading to an additive effect that translates into enhanced cell death. The study conclusively showed that p53 is a major determinant for the cytotoxic advantages of the novel combi-molecular approach in CML, a disease in which 70% to 85% of all the cases express wild-type p53.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ligação a DNA/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Proteínas Oncogênicas/farmacologia , Antineoplásicos/uso terapêutico , Ciclo Celular , Proliferação de Células , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Genes abl , Humanos , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Modelos Biológicos , Chaperonas Moleculares , Proteínas Oncogênicas/metabolismo , Proteínas de Ligação a RNA , Temozolomida , Proteína Supressora de Tumor p53/metabolismo
16.
Chem Biol Drug Des ; 71(4): 374-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18324926

RESUMO

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.


Assuntos
Receptores ErbB/química , Quinazolinas/síntese química , Triazenos/síntese química , Receptores ErbB/metabolismo , Concentração Inibidora 50 , Modelos Moleculares , Quinazolinas/química , Solubilidade , Triazenos/química , Água
17.
Bioorg Med Chem Lett ; 17(15): 4248-53, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17572088

RESUMO

Steps toward the identification of combi-molecules with strong abl tyrosine kinase (TK) inhibitory property and significant DNA damaging potential are described. The optimized combi-molecule 13a was shown to induce approximately twofold stronger abl TK inhibitory activity than Gleevec and high levels of DNA damage in chronic myelogenous leukemic cells.


Assuntos
DNA/efeitos dos fármacos , Proteínas de Fusão bcr-abl/metabolismo , Linhagem Celular Tumoral , DNA/metabolismo , Dano ao DNA , Humanos , Modelos Moleculares
18.
J Med Chem ; 50(11): 2605-8, 2007 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-17472358

RESUMO

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.


Assuntos
Antineoplásicos Alquilantes/síntese química , Receptores ErbB/antagonistas & inibidores , Compostos de Mostarda Nitrogenada/síntese química , Triazenos/síntese química , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Reagentes de Ligações Cruzadas/síntese química , Reagentes de Ligações Cruzadas/química , Reagentes de Ligações Cruzadas/farmacologia , DNA/metabolismo , Dano ao DNA , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/biossíntese , Receptores ErbB/genética , Humanos , Masculino , Modelos Moleculares , Compostos de Mostarda Nitrogenada/química , Compostos de Mostarda Nitrogenada/farmacologia , Relação Estrutura-Atividade , Transfecção , Triazenos/química , Triazenos/farmacologia
19.
Anticancer Drugs ; 17(2): 165-71, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16428934

RESUMO

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.


Assuntos
Quinazolinas/farmacocinética , Triazenos/farmacocinética , Acetilação , Animais , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Cromatografia Líquida , Receptores ErbB/antagonistas & inibidores , Humanos , Masculino , Espectrometria de Massas , Camundongos , Neoplasias da Próstata/metabolismo , Quinazolinas/síntese química , Triazenos/síntese química , Células Tumorais Cultivadas
20.
Biochem Pharmacol ; 70(4): 511-9, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15982640

RESUMO

Previous studies showed that SMA41, a 3-methyltriazene termed "combi-molecules" possessing a dual epidermal growth factor receptor (EGFR)/DNA targeting properties induced potent antiproliferative activity against alkylating-agent-resistant cells expressing EGFR in vitro. However, despite its marked potency, its antitumour activity in vivo was significantly hampered by its poor hydrosolubility and the moderate reactivity of its alkylating moiety. To circumvent this problem, we designed the quinazolinotriazene ZRBA1 to contain a N,N-dimethylaminoethyl group grafted to the 3-position of the triazene chain where it could serve both as a water soluble and a more potent alkylating moiety. ZRBA1 exhibited five-fold stronger EGFR tyrosine kinase (TK) inhibitory activity (IC(50)=37nM) than SMA41, decomposed into a 6-amino-quinazoline FD105 (IC(50)=200nM) and preferentially blocked EGF- over platelet-derived growth factor (PDGF)-or serum-induced cell growth. ZRBA1 induced DNA damage, concomitantly blocked EGF-stimulated EGFR phosphorylation by a partially irreversible mechanism in MDA-MB-468 breast cancer cells, and induced partially irreversible antiproliferative activity. It also prevented EGFR-mediated MAP kinase activation and, in contrast to FD105 and SMA41, induced high levels of apoptosis. Furthermore, ZRBA1 showed significantly greater antitumor activity (p<0.05) than SMA41 in the human MDA-MB-468 breast cancer xenograft model. The results in toto indicate that the appendage of N,N-dimethylaminoethyl to combi-triazenes may be an alternative to the reduced hydrosolubility and also to the lack of potency of monofunctional combi-triazenes against resistant tumours.


Assuntos
Receptores ErbB/antagonistas & inibidores , Quinazolinas/farmacologia , Triazenos/farmacologia , Apoptose , Linhagem Celular Tumoral , Ensaio Cometa , Dano ao DNA , Humanos , Hidrólise , Fosforilação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Quinazolinas/química , Solubilidade , Triazenos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...