Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Pharmacol Rep ; 69(3): 560-565, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28364696

RESUMEN

PURPOSE: To analyze the effect of liver steatosis and obesity on pharmacokinetic profile of two structurally-related liver-selective NO-donors - V-PYRRO/NO and V-PROLI/NO. METHODS: C57BL/6 mice were fed control or high-fat diet for 15 weeks to induced liver steatosis and obesity (HFD mice). Pharmacokinetics and renal elimination studies were conducted in vivo following iv dosing of V-PYRRO/NO and V-PROLI/NO (0.03mmol/kg). Hepatic clearance was evaluated ex vivo in the isolated perfused mice liver and in vitro with the use of liver microsomes. RESULTS: V-PYRRO/NO and V-PROLI/NO, despite similar structure, displayed different pharmacokinetic properties. V-PYRRO/NO was uptaken and metabolized by the liver, while V-PROLI/NO was eliminated unchanged with urine. In HFD mice, despite increased CYP450 metabolism of V-PYRRO/NO the elimination rate was slower most likely due to the impairment of hepatic microcirculation caused by liver fat accumulation. In turn, in HFD mice renal clearence of V-PROLI/NO was accelerated and volume of distribution was increased most likely due to additional intracellular water in HFD mice. CONCLUSIONS: The pharmacokinetics of V-PROLI/NO, the novel proline-based analog of V-PYRRO/NO with additional single carboxylic acid moiety, attached to the molecule of V-PYRRO/NO to improve the water solubility, was differently affected by liver steatosis and obesity as compared with the parent compound V-PYRRO/NO.


Asunto(s)
Hígado Graso/fisiopatología , Donantes de Óxido Nítrico/farmacocinética , Obesidad/complicaciones , Pirrolidinas/farmacocinética , Triazenos/farmacocinética , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Modelos Animales de Enfermedad , Hígado/metabolismo , Hígado/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Microcirculación , Microsomas Hepáticos/metabolismo , Donantes de Óxido Nítrico/química , Pirrolidinas/química , Solubilidad , Distribución Tisular , Triazenos/química
2.
Drug Metab Lett ; 5(2): 141-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21457144

RESUMEN

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


Asunto(s)
Carbamatos/farmacocinética , Receptores ErbB/antagonistas & inhibidores , Profármacos/farmacocinética , Quinazolinas/farmacocinética , Triazenos/farmacocinética , Animales , Agregación Celular , Línea Celular Tumoral , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Distribución Tisular
3.
Anticancer Drugs ; 20(8): 659-67, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19581798

RESUMEN

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.


Asunto(s)
Quinazolinas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Triazenos/farmacología , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Antineoplásicos/efectos de la radiación , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/efectos de la radiación , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/efectos de la radiación , Receptores ErbB/antagonistas & inhibidores , Gefitinib , Humanos , Microscopía Fluorescente , Quinazolinas/metabolismo , Quinazolinas/farmacocinética , Quinazolinas/efectos de la radiación , Fármacos Sensibilizantes a Radiaciones/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Fármacos Sensibilizantes a Radiaciones/efectos de la radiación , Triazenos/metabolismo , Triazenos/farmacocinética , Triazenos/efectos de la radiación , Ensayo de Tumor de Célula Madre , Rayos X
4.
Drug Metab Lett ; 3(1): 1-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19356110

RESUMEN

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


Asunto(s)
Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacocinética , Receptores ErbB/metabolismo , Triazenos/metabolismo , Triazenos/farmacocinética , Animales , Antineoplásicos Alquilantes/uso terapéutico , Biotransformación , Cromatografía Líquida de Alta Presión , Receptores ErbB/antagonistas & inhibidores , Humanos , Masculino , Espectrometría de Masas , Ratones , Profármacos/química , Profármacos/metabolismo , Profármacos/farmacocinética , Espectrofotometría Ultravioleta , Distribución Tisular , Triazenos/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Eur J Med Chem ; 44(3): 1049-56, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18687506

RESUMEN

A series of 3-[alpha-(acylamino)acyl]-1-aryl-3-methyltriazenes 6a-l, potential cytotoxic triazene prodrugs, were synthesised by coupling 1-aryl-3-methyltriazenes to N-acylamino acids. Their hydrolysis was studied in isotonic pH 7.4 phosphate buffer and in human plasma, while hydrolysis of the derivative 6a was studied in more depth across a range of pH values. Prodrugs 6a-l hydrolyse by cleavage of the triazene acyl group to afford the corresponding monomethyltriazenes. Studies in human plasma demonstrate that acylation of the alpha-amino group of the amino acid carrier is an effective means of reducing the chemical reactivity of the alpha-aminoacyl derivatives while retaining a rapid rate of enzymatic hydrolysis. These derivatives displayed logP values that suggest they should be well absorbed through biological membranes.


Asunto(s)
Alquilantes/síntesis química , Profármacos/síntesis química , Triazenos/síntesis química , Alquilantes/sangre , Alquilantes/química , Humanos , Cinética , Profármacos/farmacocinética , Espectrometría de Masa por Ionización de Electrospray , Triazenos/sangre , Triazenos/química , Triazenos/farmacocinética
6.
J Med Chem ; 51(4): 1043-7, 2008 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-18237109
7.
Pharmacol Res ; 56(4): 275-87, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17897837

RESUMEN

Triazene compounds of clinical interest (i.e. dacarbazine and temozolomide) are a group of alkylating agents with similar chemical, physical, antitumour and mutagenic properties. Their mechanism of action is mainly related to methylation of O(6)-guanine, mediated by methyldiazonium ion, a highly reactive derivative of the two compounds. The cytotoxic/mutagenic effects of these drugs are based on the presence of DNA O(6)-methylguanine adducts that generate base/base mismatches with cytosine and with thymine. These adducts lead to cell death, or if the cell survives, provoke somatic point mutations represented by C:G-->T:A transition in DNA helix. Triazene compounds have excellent pharmacokinetic properties and limited toxicity. Dacarbazine requires hepatic activation whereas temozolomide is spontaneously converted into active metabolite in aqueous solution at physiological pH. Moreover, temozolomide is fully active when administrated orally (100% bioavailability). The biological effects of triazene compounds and cell resistance to them depend on at least three DNA repair systems, (a) O(6)-alkylguanine-DNA-alkyltransferase, called also methyl-guanine methyl-transferase (MGMT); (b) mismatch repair (MMR), and (c) base excision repair (BER). MGMT is a small enzyme-like protein that removes small alkyl adducts from the O(6) position of DNA guanine through a stoichiometric and auto-inactivating reaction. This reaction consists in a covalent transfer of the alkyl group from the alkylated site in DNA to an internal cysteine residue of MGMT protein. High levels of MGMT are responsible for normal and tumour cell resistance to triazenes. Therefore, pre-treatment with MGMT inhibitors - i.e. O(6)-benzylguanine or O(6)-(4-bromotenyl)guanine (Lomeguatrib) - is followed by a great increase in the activity of triazenes against target cells expressing high MGMT levels. MMR is represented by a protein complex dedicated to the repair of biosynthetic errors generated during DNA replication. The MMR system recognizes base mismatches and insertion-deletion loops, cuts the nucleotide sequence containing the lesion, and restores the correct base sequence. Therefore, not only MGMT but also MMR is involved in target cell susceptibility to triazenes. However, the system does not suppress, but instead promotes the cytotoxic effects of triazenes. In fact, MMR is not able to repair the incorrect base pairing determined by treatment with triazenes and, according to a predominant hypothesis, it causes reiterated "futile" attempts of damage repair leading to the activation of cell cycle arrest and apoptosis. BER removes lesions due to cellular metabolism, or to physical or chemical agents. BER is able to repair N(7)-methylguanine and N(3)-methyladenine determined by treatment with triazenes. Therefore, triazene compounds can also kill tumour cells by a N(3)-methyladenine-mediated mechanism if BER activity is inhibited by chemical agents (i.e. PARP inhibitors). In conclusion, in selected cases, triazenes can represent a therapeutic alternative to treatment of neoplastic diseases including haematological malignancies. Moreover, the susceptibility of neoplastic cells to these compounds can be substantially increased through pharmacological modulation of the expression level and functional activity of DNA repair enzymes.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Reparación del ADN , Triazenos/farmacología , Antineoplásicos Alquilantes/efectos adversos , Antineoplásicos Alquilantes/farmacocinética , Disparidad de Par Base , Dacarbazina/efectos adversos , Dacarbazina/análogos & derivados , Dacarbazina/farmacocinética , Dacarbazina/farmacología , Humanos , Temozolomida , Triazenos/efectos adversos , Triazenos/farmacocinética
8.
Anticancer Drugs ; 17(2): 165-71, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16428934

RESUMEN

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


Asunto(s)
Quinazolinas/farmacocinética , Triazenos/farmacocinética , Acetilación , Animales , Proliferación Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Receptores ErbB/antagonistas & inhibidores , Humanos , Masculino , Espectrometría de Masas , Ratones , Neoplasias de la Próstata/metabolismo , Quinazolinas/síntesis química , Triazenos/síntesis química , Células Tumorales Cultivadas
9.
J Med Chem ; 46(20): 4313-21, 2003 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-13678409

RESUMEN

The combi-targeting concept postulates that a molecule termed a "combi-molecule" designed to interact with an oncoreceptor on its own and allowed to further degrade to another more stable inhibitor of the latter receptor + a DNA-damaging species should be more potent than the individual combination of the same inhibitor with a DNA-damaging agent in cells expressing the targeted receptor. Recently, using the epidermal growth factor receptor (EGFR) as a target, we demonstrated the feasibility of combi-molecules with dual EGFR/DNA-targeting properties and with the ability to degrade to another potent inhibitor of EGFR. However, despite a clear demonstration of their superior potency when compared with classical combinations in EGFR-expressing cells, the true contribution of each fragment of the combi-molecules to their overall antiproliferative activity remained elusive. Here, we report a structure-function approach whereby a series of quinazoline-based "combi-triazenes" were altered to either abrogate the affinity of the EGFR-targeting quinazoline head or to suppress the DNA-damaging property of the triazene tail. The results showed that (a) inactivation of the quinazoline head by appending an N-methylaniline group to its 4-position reduced EGFR tyrosine kinase (TK) inhibitory activity by ca. 200-fold and decreased the ability of the combi-molecule to block serum-induced growth stimulation in c-erbB2 transfected NIH3T3 cells by ca. 10-fold, (b) abrogation of the alkylating activity or the DNA-damaging potential of the triazene tail by forming 3,3-dimethyltriazenes did not suppress EGFR TK inhibitory affinity but decreased the antiproliferative activity in basal growth assays, and (c) the antiproliferative activities of the monoalkyltriazenes that possessed binary EGFR TK inhibitory and alkylating activities were superior to those of their monotargeted counterparts. The results in toto suggest that each component of the dual targeting property of combi-triazenes plays a critical role in their overall antiproliferative activity.


Asunto(s)
Daño del ADN , Receptores ErbB/antagonistas & inhibidores , Quinazolinas/química , Quinazolinas/farmacología , Triazenos/química , Triazenos/farmacología , Células 3T3/citología , Células 3T3/efectos de los fármacos , Animales , Biotransformación , División Celular/efectos de los fármacos , Metilación de ADN , Inhibidores Enzimáticos/farmacología , Receptores ErbB/metabolismo , Humanos , Concentración 50 Inhibidora , Ratones , Quinazolinas/farmacocinética , Espectrometría de Fluorescencia/métodos , Relación Estructura-Actividad , Triazenos/farmacocinética , Células Tumorales Cultivadas
10.
Int J Pharm ; 247(1-2): 39-45, 2002 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-12429483

RESUMEN

Biochemical and biological activities of two recently synthesized spin labeled triazenes, containing the nitroxyl free radical moiety at different places of the triazene structure have been studied and compared with those of the antitumor drug Dacarbazine (DTIC). Tissue distribution of the triazenes was investigated in vitro in organ homogenates, tumor (B16 melanoma) and blood of C57BL mice using the electron paramagnetic resonance (EPR) method. The spin labeled triazenes were mainly localized in the tumor and in the brain. Normal leucocites, YAC-1 mNK target Moloney lymphoma cells and B16 melanoma cells were treated with spin labeled triazenes in vitro and the effects on cell viability were compared. Spin labeled 3,3-dimethyl triazene with nitroxyl radical as a substituent in the benzen ring was more cytotoxic to B16 melanoma cells than to YAC-1 Moloney lymphoma cells and normal leucocites in comparison to the spin labeled monomethyl triazene. The spin labeled derivatives were assessed with low toxicity for BDF1 mice hybrids in vivo. These results could be interpreted in terms of a possible correlation between tissue distribution and the selective antimelanoma activity of the spin labeled triazenes.


Asunto(s)
Marcadores de Spin/síntesis química , Triazenos/química , Triazenos/farmacocinética , Animales , Células Cultivadas , Humanos , Leucemia Linfoide/metabolismo , Masculino , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Distribución Tisular/fisiología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
11.
Toxic Rep Ser ; (73): 1-23, A1-C6, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12370695

RESUMEN

Diazoaminobenzene is used as an intermediate, complexing agent, and polymer additive. It is also an impurity in certain color additives used in cosmetics, food products, and pharmaceuticals. Diazoaminobenzene was selected for metabolism and toxicity studies based on the potential for worker exposure from its use in laboratories, positive Salmonella typhimurium gene mutation data, its presence as an impurity in foods and cosmetics, and the lack of adequate toxicity data. Several structural analogues and presumed metabolites of diazoaminobenzene are carcinogenic, providing evidence for the possible carcinogenicity of diazoaminobenzene. The chemical structure of diazoaminobenzene suggested that it would be metabolized into aniline and benzene; therefore, metabolism and disposition studies were performed in male and female F344/N rats and male B6C3F1 mice administered a single oral, dermal, or intravenous dose of diazoaminobenzene. Electron spin resonance (ESR) studies were conducted to assess the possible formation of a phenyl radical from the reduction of diazoaminobenzene by components of the cytochrome P450 mixed-function oxidase (P450) system in microsomes or by gut microflora in anaerobic cecal incubations. Bile duct-cannulated male F344/N rats were administered diazoaminobenzene and 5,5-dimethyl-1- pyrroline-N-oxide (DMPO) for in vivo determination of the DMPO-phenyl radical. 16-Day toxicity studies were performed to identify target organs of diazoaminobenzene following dermal application to male and female F344/N rats and B6C3F1 mice. In the disposition and metabolism studies, oral doses of 20 mg/kg to male and female rats and male mice were readily absorbed and excreted mainly in the urine, with exhalation of volatile organics accounting for about 1% of the dose. The only volatile metabolite detected in the breath was benzene, and all the metabolites in the urine were those previously shown to result from the metabolism of benzene and aniline in rats and mice. While dermal doses to rats and mice (2 and 20 mg/cm2) were only slightly absorbed, benzene and aniline metabolites were nonetheless detected in the urine. High circulating levels of benzene, aniline, and their metabolites were detected in the blood of rats administered 20 mg/kg diazoaminobenzene as early as 15 minutes after exposure. At 24 hours after dosing, diazoaminobenzene was detected at low levels (<1%) in the adipose tissue, blood, kidney, liver, muscle, skin, and spleen. Metabolites of benzene and aniline were also formed in an in vitro study using human liver slices. In the ESR spin-trapping experiments, the ESR spectrum of the DMPO-phenyl radical was detected when diazoaminobenzene was incubated with microsomes or P450 reductase, DMPO, and NADPH, or when incubated with cecal contents and DMPO. The DMPO-phenyl radical spectrum was not attenuated by the P450 inhibitor, 1-aminobenzotriazole, or carbon monoxide suggesting that P450s were not required. In in vivo experiments in which rats were administered diazoaminobenzene and DMPO, the DMPO-phenyl radical adduct was detected in bile within 1 hour after treatment. In the 16-day toxicity studies, groups of five male and five female F344/N rats and B6C3F1 mice received dermal applications of 0, 12.5, 25, 50, 100, or 200 mg diazoaminobenzene/kg body weight. Animals were evaluated for absolute and relative organ weights, for hematological effects, and for gross and microscopic lesions. No mortality occurred in rats. However, most male mice exposed to concentrations of 50 mg/kg or greater and female mice exposed to 200 mg/kg died. Body weights of male and female rats and female mice were less than those of the vehicle controls. Similar chemical-related toxicities were observed in both species. Clinical pathology data indicated a chemical-related methemoglobinemia and Heinz body formation in male and female rats and mice. Analysis of organ weights indicated possible chemical-related effects in the thymus, heart, spleen, kidney, and liver of rats and/or mice. Increases in the incidences of several skin lesionseral skin lesions, including hyperplasia of the epidermis and hair follicles, and inflammation in rats and mice and ulceration in female mice were observed. Other nonneoplastic lesions that were considered to be related to diazoaminobenzene administration were atrophy of the thymus, mandibular and/or mesenteric lymph nodes, and white pulp of the spleen, as well as splenic hematopoietic cell proliferation in rats and mice. In mice, there were increased incidences of atrial thrombosis, and necrosis was observed in the renal tubules and liver. Diazoaminobenzene was mutagenic in S. typhimurium strains TA98, TA100, and TA1537 with induced rat or hamster liver S9 enzymes; no activity was noted in strain TA1535, with or without S9. In vivo, two gavage administrations of either diazoaminobenzene or benzene induced highly significant increases in micronucleated polychromatic erythrocytes in bone marrow of male B6C3F1 mice at all doses tested. Diazoaminobenzene is metabolized to the known carcinogens benzene and aniline. Further evidence of this metabolism is that some toxic effects associated with aniline (methemoglobinemia) and benzene (atrophy of the lymphoid tissue) were identified. Based on these results, it is predicted that diazoaminobenzene is a carcinogen.


Asunto(s)
Carcinógenos/toxicidad , Animales , Cricetinae , Femenino , Humanos , Absorción Intestinal , Ratones , Pruebas de Mutagenicidad , Embarazo , Ratas , Ratas Endogámicas F344 , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/genética , Distribución Tisular , Triazenos/farmacocinética , Triazenos/toxicidad
12.
J Pharmacol Exp Ther ; 303(1): 238-46, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12235257

RESUMEN

The Combi-Targeting concept postulates that a molecule termed combi-molecule (C-molecule) with binary epidermal growth factor receptor (EGFR) targeting/DNA-damaging properties and with the ability to be hydrolyzed to another EGFR inhibitor should induce sustained antiproliferative activity in cells overexpressing EGFR. Because we postulate that the EGFR affinity of the C-molecule and that of its hydrolytic metabolites are critical parameters for sustained potency against EGFR-overexpressing cells, we synthesized BJ2000 (IC(50) = 0.1 microM, competitive binding at ATP site), a novel C-molecule that can decompose into a 6-amino-4-anilinoquinazoline FD105 (IC(50) = 0.2 microM). Studies using the EGFR-overexpressing A431 cells revealed that BJ2000 could damage DNA and block epidermal growth factor-stimulated EGFR autophosphorylation by a partially irreversible mechanism. Blockade of EGFR autophosphorylation subsequently induced inhibition of mitogen-activated protein kinase activation and c-fos gene expression. Enzyme-linked immunosorbent assay and growth factor-mediated stimulation of proliferation assays in the EGFR-expressing NIH3T3HER14 demonstrated the preferential EGFR-targeting properties of BJ2000, and more importantly suggest that blockade of EGFR phosphorylation by this drug translate into significant growth inhibitory effects. These properties culminated into irreversible antiproliferative effects as confirmed by a sulforhodamine B assay. Five days after a 2-h treatment, BJ2000 retained significant antiproliferative effect in A431 cells, whereas its reversible metabolite FD105 almost completely lost its activity. This result in toto lend support to the Combi-Targeting concept according to which a molecular conjugate kept small enough to interact with EGFR and designed to degrade into another inhibitor of the same target plus a DNA-damaging species may induce sustained growth inhibitory effect in EGFR-overexpressing cells.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Triazenos/farmacología , Células 3T3 , Animales , Biotransformación , Daño del ADN , Relación Dosis-Respuesta a Droga , Receptores ErbB/fisiología , Humanos , Ratones , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Quinazolinas/síntesis química , Quinazolinas/farmacocinética , Transducción de Señal/fisiología , Triazenos/síntesis química , Triazenos/farmacocinética , Células Tumorales Cultivadas
13.
Biomed Chromatogr ; 15(2): 75-8, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11268045

RESUMEN

A capillary gas chromatographic method was developed for determining 1-p-(3.3-dimethyl-1-triazeno) benzoic acid in the plasma and urine of cancer patients under pharmacokinetic study. The drug was extracted with ethyl acetate and methylated with diazomethane. Octadelane (10 microg/ml) was added as internal standard. The separation was carried out on an OV-1 quartz capillary column, 15 m x 0.32 mm (0.52 microm), with high-purity nitrogen as carrier gas and flame ionization detector (FID) as detector. The column temperature was held at 130 degrees C for 9 min and then programmed to 240 degrees C, at a rate of 35 degrees C/min. The temperature of both injector and detector was 260 degrees C. The standard curve was linear from 0.4 to 40 microg/mL in plasma, and from 0.8 to 20 microg/mL in urine, with correlation coefficients of 0.9979 and 0.9932. The relative standard deviations (RSD) were less than 9.7%. The minimum recovery of this method was 81.8%. This method was applied to the pharmacokinetic studies of 1-p-(3.3-dimethyl-1-triazeno) benzoic acid in cancer patients after a single dose (i.v.) of 160, 420 or 760 mg/m(2) was administered. They all conformed to the two-compartment open model and showed linear pharmacokinetics. The excretion of this drug in the urine was minimal.


Asunto(s)
Antineoplásicos/farmacocinética , Cromatografía de Gases/métodos , Neoplasias/metabolismo , Triazenos/farmacocinética , Adulto , Humanos , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/orina , Reproducibilidad de los Resultados , Triazenos/sangre , Triazenos/orina
14.
Nitric Oxide ; 5(1): 77-81, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11178940

RESUMEN

Endothelial cells secrete large amounts of 5,6,7,8-tetrahydrobiopterin (BH(4)) in septic conditions. BH(4) is a cofactor for nitric oxide (NO) synthase and an essential regulator of its activity. We recently showed that NO can be a modulator of both platelet 12-lipoxygenase and cyclooxygenase activities. In the present study, we investigated the effect of BH(4) on the activities of 12-lipoxygenase and cyclooxygenase in rabbit platelets. The influence of BH(4) on NO-induced modulation of these enzyme activities was investigated. Exogenous BH(4) did not affect platelet 12-lipoxygenase and cyclooxygenase activities. The modulatory effects of NO on the two enzymatic pathways were reversed by addition of BH(4) but not by reduced glutathione. These results suggest that exogenous BH(4) is not essential for NO synthase activity of platelets, but that it is an important regulator of the action of NO released from other sources on platelet 12-lipoxygenase and cyclooxygenase activities.


Asunto(s)
Araquidonato 12-Lipooxigenasa/sangre , Biopterinas/análogos & derivados , Biopterinas/farmacología , Plaquetas/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Inhibidores de la Lipooxigenasa/farmacología , Óxido Nítrico/farmacología , Prostaglandina-Endoperóxido Sintasas/sangre , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/sangre , Animales , Ácidos Araquidónicos/sangre , Plaquetas/enzimología , Ácidos Grasos Insaturados/sangre , Glutatión/sangre , Donantes de Óxido Nítrico/farmacología , Conejos , Tromboxano B2/sangre , Triazenos/farmacocinética
15.
Anticancer Drugs ; 11(7): 515-33, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11036954

RESUMEN

Alkylating agents are widely used in high-dose chemotherapy regimens in combination with hematological support. Knowledge about the pharmacokinetics and pharmacodynamics of these agents administered in high doses is critical for the safe and efficient use of these regimens. The aim of this review is to summarize the clinical pharmacology of the alkylating agents (including the platinum compounds) in high-dose chemotherapy. Differences between conventional and high doses will be discussed.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/farmacocinética , Alcanosulfonatos/administración & dosificación , Alcanosulfonatos/efectos adversos , Alcanosulfonatos/farmacocinética , Alcanosulfonatos/farmacología , Antineoplásicos Alquilantes/administración & dosificación , Antineoplásicos Alquilantes/efectos adversos , Aziridinas/administración & dosificación , Aziridinas/efectos adversos , Aziridinas/farmacocinética , Aziridinas/farmacología , Humanos , Compuestos de Mostaza Nitrogenada/administración & dosificación , Compuestos de Mostaza Nitrogenada/efectos adversos , Compuestos de Mostaza Nitrogenada/farmacocinética , Compuestos de Mostaza Nitrogenada/farmacología , Compuestos de Nitrosourea/administración & dosificación , Compuestos de Nitrosourea/efectos adversos , Compuestos de Nitrosourea/farmacocinética , Compuestos de Nitrosourea/farmacología , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/efectos adversos , Compuestos Organoplatinos/farmacocinética , Compuestos Organoplatinos/farmacología , Triazenos/administración & dosificación , Triazenos/efectos adversos , Triazenos/farmacocinética , Triazenos/farmacología
16.
Bioorg Med Chem ; 8(7): 1719-25, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10976519

RESUMEN

A series of 3-acyloxymethyloxycarbonyl-1-aryl-3-methyltriazenes 5 was synthesised by the sequential reaction of 1-aryl-3-methyltriazenes with (i) chloromethyl chloroformate, (ii) NaI in dry acetone, and (iii) either the silver carboxylate or the carboxylic acids in the presence of silver carbonate. The hydrolysis of these compounds was studied in pH 7.7 isotonic phosphate buffer and in human plasma. Triazene acyloxycarbamates demonstrated their ability to act as substrates for plasma enzymes. For compound 5f, a pH-rate profile was obtained which showed the hydrolysis to involve acid-base catalysis. The reaction is also buffer catalysed. Thus, at pH 7.7, pH-independent, base-catalysed and buffer-catalysed processes all contribute to the hydrolysis reaction. The sensitivity of the hydrolysis reaction to various structural parameters in the substrates indicates that hydrolysis occurs at the ester rather than the carbamate functionality. In plasma, the rates of hydrolysis correlate with partition coefficients, the most lipophilic compounds being the most stable. An aspirin derivative suffers two consecutive enzymatic reactions, the scission of the aspirin acetyl group being followed by the scission of the acyloxy ester group. These results indicate that triazene acyloxymethyl carbamates are prodrugs of the antitumour monomethyltriazenes. They combine chemical stability with a rapid enzymatic hydrolysis, and are consequently good candidates for further prodrug development. Moreover, this type of derivative allowed the synthesis of mutual prodrugs, associating the antitumour monomethyltriazenes with anti-inflammatory NSAIDs as well as with the anticancer agent butyric acid.


Asunto(s)
Carbamatos/síntesis química , Carbamatos/metabolismo , Triazenos/síntesis química , Triazenos/metabolismo , Triazenos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Aspirina/metabolismo , Aspirina/farmacología , Carbamatos/química , Carbamatos/farmacología , Cromatografía Líquida de Alta Presión , Estabilidad de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Plasma/enzimología , Profármacos/síntesis química , Profármacos/metabolismo , Profármacos/farmacocinética , Relación Estructura-Actividad , Triazenos/farmacocinética
17.
Drug Metab Dispos ; 27(12): 1499-504, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10570033

RESUMEN

1,3-Diphenyl-1-triazene (DPT) is used in the synthesis of polymers and dyes, and has been found as an impurity in the color additives D&C Red 33 and FD&C Yellow 5. [(14)C]DPT, randomly labeled in the phenyl rings, was used to investigate its disposition in rodents. Dermal doses to rats and mice (2 and 20 mg/cm(2)) were poorly absorbed (

Asunto(s)
Aditivos Alimentarios/farmacocinética , Triazenos/farmacocinética , Administración Cutánea , Administración Oral , Animales , Femenino , Aditivos Alimentarios/metabolismo , Humanos , Técnicas In Vitro , Inyecciones Intravenosas , Hígado/metabolismo , Tasa de Depuración Metabólica , Ratones , Ratas , Ratas Endogámicas F344 , Triazenos/metabolismo
18.
FEBS Lett ; 398(1): 53-6, 1996 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-8946952

RESUMEN

The effects of nitric oxide (NO) released from zwitterionic polyamine/NO adducts on Cu2+-induced low density lipoprotein (LDL) oxidation were studied. When each of the two kinds of NO releasing zwitterionic polyamine/NO adducts (NOC5 and NOC7) was incubated at 5 microM with isolated human LDL (0.25 mg/ml) and Cu2+, the formation of thiobarbituric acid reactive substances (TBARS) was inhibited. The duration of inhibition by NOC7 (20 min) and NOC5 (100 min) corresponded to the NO generation lives of respective zwitterionic polyamine/NO adducts. The duration of inhibition was dependent on the amount of NOC5 added (2.5-20 microM). Repeated additions of 5 microM NOC5 at 100 min intervals worked as inhibitor in the same manner. NOC5 broke to inhibit at any process of the Cu2+-induced LDL oxidation reaction. Fragmentation of apolipoprotein B derived from Cu2+-induced LDL oxidation was also prevented by the addition of NOC5. These results clearly indicate that NO inhibits the oxidative modification of LDL induced by Cu2+. NO releasing zwitterionic polyamine/NO adducts are good reagents for NO studies.


Asunto(s)
Cobre/metabolismo , Lipoproteínas LDL/metabolismo , Óxido Nítrico/metabolismo , Triazenos/farmacocinética , Antioxidantes/farmacocinética , Apolipoproteínas/efectos de los fármacos , Apolipoproteínas/metabolismo , Cobre/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Lipoproteínas LDL/efectos de los fármacos , Óxido Nítrico/farmacocinética , Oxidación-Reducción , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Factores de Tiempo
19.
Br J Cancer ; 67(2): 362-8, 1993 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8431367

RESUMEN

Decarbazine is an imidazole dimethyltriazene with reproducible activity in patients with metastatic melanoma. CB10-277 is a phenyl dimethyltriazene which, like dacarbazine, requires metabolic activation to its corresponding monomethyl species for antitumour activity. In preclinical models (human melanoma xenografts and transplantable rodent tumours) CB10-277 showed a similar spectrum and level of activity when compared to dacarbazine. Pharmacokinetic studies were performed with CB10-277 in mice treated i.v. at the LD10 (750 mg m-2) and plasma analysed by HPLC. The parent drug area under the plasma concentration vs time curve (AUC) was 142 mM x minutes. Drug metabolism occurred as evidenced by the HPLC identification of the monomethyl species (AUC = 8 mM x minutes) as well as other metabolites. A Phase I trial using a short infusion with doses repeated every 21 days has been performed. Thirty-six patients received 80 courses over a dose range of 80-6,000 mg m-2. The dose limiting toxicity was nausea and vomiting which occurred in 80% of the evaluable courses > or = 900 mg m-2. The only other common side effect was a flushing or warm sensation, which occurred in over 75% of courses at > or = 1,350 mg m-2. There were no hemodynamic consequences. Responses occurred in patients with melanoma (one complete, two partial, one mixed/11), sarcoma (one mixed/6) and carcinoid (one partial/l). Pharmacokinetics were performed in 46 courses. The CB10-277 AUC increased linearly with dose (r = 0.9203, P < 0.001) up to 700 mM x minutes at 6,000 mg m-2). Evidence of CB10-277 metabolism was observed, as in mice, by detection of the monomethyl species and other metabolites. However, the plasma levels of the monomethyl species in patients (1.8 and 3.7 mM x minutes at 6,000 mg m-2) were less than those predicted from studies in mice. Despite this, antitumour activity in dacarbazine sensitive histologies was observed and additional studies with CB10-277 are recommended.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Neoplasias/tratamiento farmacológico , Triazenos/farmacología , Triazenos/farmacocinética , Adulto , Anciano , Animales , Antineoplásicos/efectos adversos , Dacarbazina/uso terapéutico , Modelos Animales de Enfermedad , Esquema de Medicación , Femenino , Humanos , Infusiones Intravenosas , Masculino , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Neoplasias/metabolismo , Especificidad de la Especie , Trasplante Heterólogo , Triazenos/efectos adversos
20.
Br J Cancer ; 67(2): 369-73, 1993 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8431368

RESUMEN

The dose limiting toxicities of the short infusion trial of the dacarbazine analog, CB10-277, were nausea and vomiting which appeared to be related to the peak plasma level of the parent drug. In addition, based on mouse studies, these dose limiting toxicities occurred at a less than optimal level of the monomethyl metabolite, the presumed species required for antitumour activity. An alternative schedule that would avoid the parent drug peak plasma levels of short infusion, while possibly allowing an increase in the amount of monomethyl metabolite produced was considered. Thus, a 24 h continuous infusion schedule, repeated every 21 days was explored. Twenty-two patients received 42 courses with a dose range of 4,700-15,000 mg m-2. The dose limiting toxicity was myelosuppression (leucopenia and thrombocytopenia). Although nausea and vomiting also occurred, it was manageable with routine antiemetic therapy. Other toxicities included diarrhoea, hallucinations, malaise, muscle ache, headache and flushing and all were < or = WHO grade 2. Pharmacokinetic studies were performed with 13 courses which included all dose levels. The mean t1/2 of the parent drug was 178 min. Area under the concentration x time curve (AUC) at the highest dose for the parent drug and the monomethyl metabolite were 2,350 and 9 mM x minutes, respectively. This monomethyl metabolite AUC and the associated myelosuppression showed a more favourable comparison to the preclinical data determined in mice than the results from the short infusion trial of CB10-277. Therefore, the recommended Phase II dose and schedule of this drug was 12,000 mg m-2 given by 24 h continuous infusion.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Neoplasias/tratamiento farmacológico , Triazenos/efectos adversos , Triazenos/farmacocinética , Adulto , Anciano , Antineoplásicos/uso terapéutico , Esquema de Medicación , Femenino , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Triazenos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA