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.
Eur J Med Chem ; 240: 114573, 2022 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-35797900

RESUMEN

A series of quinoline and quinazoline analogs were designed and synthesized as new tubulin polymerization (TP) and histone deacetylases (HDAC) inhibitors. Compounds 12a and 12d showed the best cytotoxicity activities against a panel of human cancer cell lines with an averaged IC50 value of 0.6 and 0.7 nM, respectively. Furthermore, these lead compounds showed good activities against CA-4-resistant colon-carcinoma and multidrug-resistant leukemia cells. In addition, compounds 12a and 12d induced HT29 cell cycle arrest in the G2/M phase and produced caspase-induced apoptosis of HT29 cells through mitochondrial dysfunction. Also, 12a and 12d inhibited HDAC8, 6, and 11 activities. Furthermore, lead compound 12a exhibited higher metabolic stability than isoCA-4 and was highly potent in suppressing tumor growth in the fibrosarcoma MCA205 tumor model. Collectively, these studies suggest that 12a represents a new dual inhibitor of TP and HDAC activities, which makes it a suitable candidate for further investigations in clinical development.


Asunto(s)
Antineoplásicos , Quinolinas , Línea Celular Tumoral , Proliferación Celular , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Polimerizacion , Quinolinas/farmacología , Proteínas Represoras , Tubulina (Proteína)/metabolismo
2.
Eur J Med Chem ; 223: 113656, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34171660

RESUMEN

In this study, a variety of original ligands related to Combretastatin A-4 and isoCombretastatin A-4, able to inhibit the tubulin polymerization into microtubules, was designed, synthesized, and evaluated. Our lead compound 15d having a quinazoline as A-ring and a 2-substituted indole as B-ring separated by a N-methyl linker displayed a remarkable sub-nanomolar level of cytotoxicity (IC50 < 1 nM) against 9 human cancer cell lines.


Asunto(s)
Antineoplásicos/farmacología , Indoles/química , Estilbenos/química , Moduladores de Tubulina/química , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Sitios de Unión , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Estabilidad de Medicamentos , Humanos , Indoles/metabolismo , Indoles/farmacología , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Ratas , Estilbenos/metabolismo , Estilbenos/farmacología , Relación Estructura-Actividad , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/metabolismo , Moduladores de Tubulina/farmacología
3.
J Med Chem ; 62(4): 1902-1916, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30525602

RESUMEN

We report the synthesis and metabolic and biological evaluation of a series of 17 novel heterocyclic derivatives of isocombretastatin-A4 (iso-CA-4) and their structure-activity relationships. Among these derivatives, the most active compound, 4f, inhibited the growth of a panel of seven cancer cell lines with an IC50 in the low nanomolar range. In addition, 4f showed interesting activity against CA-4-resistant colon-carcinoma cells and multidrug-resistant leukemia cells. It also induced G2/M cell-cycle arrest. Structural data indicated binding of 4f to the colchicine site of tubulin, likely preventing the curved-to-straight tubulin structural changes that occur during microtubule assembly. Also, 4f disrupted the blood-vessel-like assembly formed by human umbilical-vein endothelial cells in vitro, suggesting its function as a vascular-disrupting agent. An in vitro metabolism study of 4f showed its high human-microsomal stability in comparison with that of iso-CA-4. The physicochemical properties of 4f may be conducive to CNS permeability, suggesting that this compound may be a possible candidate for the treatment of glioblastoma.


Asunto(s)
Carbazoles/farmacología , Quinaldinas/farmacología , Moduladores de Tubulina/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Carbazoles/síntesis química , Carbazoles/metabolismo , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Microsomas Hepáticos/metabolismo , Estructura Molecular , Polimerizacion/efectos de los fármacos , Unión Proteica , Quinaldinas/síntesis química , Quinaldinas/metabolismo , Ratas , Relación Estructura-Actividad , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/metabolismo
4.
Int J Pharm ; 532(2): 748-756, 2017 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-28546071

RESUMEN

Oxazaphosphorines including cyclophosphamide, trofosfamide and ifosfamide (IFO) belong to the alkylating agent class and are indicated in the treatment of numerous cancers. However, IFO is subject to limiting side-effects in high-dose protocols. To circumvent IFO drawbacks in clinical practices, preactivated IFO analogs were designed to by-pass the toxic metabolic pathway. Among these IFO analogs, some of them showed the ability to self-assemble due to the use of a poly-isoprenyloxy chain as preactivating moiety. We present here, the in vitro activity of the nanoassembly formulations of preactivated IFO derivatives with a C-4 geranyloxy, farnesyloxy and squalenoxy substituent on a large panel of tumor cell lines. The chemical and colloidal stabilities of the geranyloxy-IFO (G-IFO), farnesyloxy-IFO (F-IFO) and squalenoxy-IFO (SQ-IFO) NAs were further evaluated in comparison to their free formulation. Finally, pharmacokinetic parameters and maximal tolerated dose of the most potent preactivated IFO analog (G-IFO) were determined and compared to IFO, paving the way to in vivo studies.


Asunto(s)
Antineoplásicos Alquilantes/administración & dosificación , Ifosfamida/análogos & derivados , Ifosfamida/administración & dosificación , Nanoestructuras/administración & dosificación , Animales , Antineoplásicos Alquilantes/farmacocinética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Ifosfamida/química , Ifosfamida/farmacocinética , Masculino , Dosis Máxima Tolerada , Ratones Desnudos , Nanoestructuras/química , Prenilación
5.
Pharm Res ; 33(8): 1913-22, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27091031

RESUMEN

PURPOSE: Busulfan-melphalan high-dose chemotherapy followed by autologous stem cell transplantation is an essential consolidation treatment of high-risk neuroblastoma in children. Main treatment limitation is hepatic veno-occlusive disease, the most severe and frequent extra-hematological toxicity. This life threatening toxicity has been related to a drug interaction between busulfan and melphalan which might be increased by prior disturbance of iron homeostasis, i.e. an increased plasma ferritin level. METHODS: We performed an experimental study of busulfan and melphalan pharmacodynamic and pharmacokinetics in iron overloaded mice. RESULTS: Iron excess dramatically increased the toxicity of melphalan or busulfan melphalan combination in mice but it did not modify the clearance of either busulfan or melphalan. We show that prior busulfan treatment impairs the clearance of melphalan. This clearance alteration was exacerbated in iron overloaded mice demonstrating a pharmacokinetic interaction. Additionally, iron overload increased melphalan toxicity without altering its pharmacokinetics, suggesting a pharmacodynamic interaction between iron and melphalan. Based on iron homeostasis disturbance, we postulated that prior induction of ferritin, through Nrf2 activation after oxidative stress, may be associated with the alteration of melphalan metabolism. CONCLUSION: Iron overload increases melphalan and busulfan-melphalan toxicity through a pharmacodynamic interaction and reveals a pharmacokinetic drug interaction between busulfan and melphalan.


Asunto(s)
Busulfano/metabolismo , Busulfano/toxicidad , Sobrecarga de Hierro/metabolismo , Melfalán/metabolismo , Melfalán/toxicidad , Animales , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/toxicidad , Interacciones Farmacológicas/fisiología , Sobrecarga de Hierro/patología , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Artículo en Inglés | MEDLINE | ID: mdl-25939095

RESUMEN

The antitumor drug, ifosfamide (IFO), requires activation by cytochrome P450 (CYP) to form the active metabolite, 4-hydroxyisfosfamide (4-OHIFO), leading to toxic by-products at high dose. In order to overcome these drawbacks, preactivated ifosfamide derivatives (RXIFO) were designed to release 4-OHIFO without CYP involvement. A high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) method was developed for the simultaneous quantification of 4-OHIFO, IFO and four derivatives RXIFO in mouse plasma using multiple reaction monitoring. Because of its instability in plasma, 4-OHIFO was immediately converted to the semi-carbazone derivative, 4-OHIFO-SCZ. For the six analytes, the calibration curves were linear from 20 to 5000ng/mL in 50µL plasma and the lower limit of quantitation was determined at 20ng/mL with accuracies within ±10% of nominal and precisions less than 12%. Their recoveries ranged from 62 to 96% by using liquid-liquid extraction. With an improved assay sensitivity compared to analogues, the derivative 4-OHIFO-SCZ was stable in plasma at 4°C for 24h and at -20°C for three months. For all compounds, the assay was validated with accuracies within ±13% and precisions less than 15%. This method was applied to a comparative pharmacokinetic study of 4-OHIFO from IFO and three derivatives RXIFO in mice. This active metabolite was produced by some of the novel conjugates with good pharmacokinetic properties.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Ifosfamida/análogos & derivados , Ifosfamida/sangre , Espectrometría de Masas en Tándem/métodos , Animales , Estabilidad de Medicamentos , Femenino , Ifosfamida/química , Ifosfamida/farmacocinética , Modelos Lineales , Ratones , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
7.
J Med Chem ; 58(2): 705-17, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25494842

RESUMEN

Oxazaphosphorines are alkylating agents used in routine clinical practices for treatment of cancer for many years. They are antitumor prodrugs that require cytochrome P450 bioactivation leading to 4-hydroxy derivatives. In the case of ifosfamide (IFO), the bioactivation produces two toxic metabolites: acrolein, a urotoxic compound, concomitantly generated with the isophosphoramide mustard; and chloroacetaldehyde, a neurotoxic and nephrotoxic compound, arising from the oxidation of the side chains. To improve the therapeutic index of IFO, we have designed preactivated IFO derivatives with the covalent binding of several O- and S-alkyl moieties including polyisoprenoid groups at the C-4 position of the oxazaphosphorine ring to avoid cytochrome bioactivation favoring the release of the active entity and limiting the chloroacetaldehyde release. Thanks to the grafted terpene moieties, some of these new conjugates demonstrated spontaneous self-assembling properties into nanoassemblies when dispersed in water. The cytotoxic activities on a panel of human tumor cell lines of these novel oxazaphosphorines, in bulk form or as nanoassemblies, and the release of 4-hydroxy-IFO from these preactivated IFO analogues in plasma are reported.


Asunto(s)
Antineoplásicos Alquilantes/síntesis química , Ifosfamida/análogos & derivados , Mostazas de Fosforamida/metabolismo , Profármacos/síntesis química , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacología , Línea Celular Tumoral , Diseño de Fármacos , Humanos , Ifosfamida/metabolismo , Profármacos/metabolismo , Profármacos/farmacología
8.
J Clin Endocrinol Metab ; 98(6): 2401-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23543666

RESUMEN

OBJECTIVES: Vandetanib was approved by the U.S. Food and Drug Association for the treatment of advanced medullary thyroid cancer (MTC). Because body weight (BW) loss is observed in MTC and because low skeletal muscle mass (SM) is associated with drug toxicity, this study assessed effects of vandetanib on SM and adipose tissue (AT) and explored the association between SM, toxicity, and serum concentration of vandetanib. METHODS: Thirty-three patients with MTC received vandetanib (n = 23) or placebo (n = 10) in the ZETA study. Visceral AT (VAT), sc AT (SAT), and SM were assessed with computed tomography imaging by measuring tissue cross-sectional areas (square centimers per square meter). Dose-limiting toxicities (DLTs) were prospectively recorded. RESULTS: Early at 3 months, compared with placebo group who lost BW, muscle, and SAT, patients treated with vandetanib gained 1.5 kg BW (P = 0.02), 1.3 cm(2)/m(2) (∼0.7 kg) of SM (P = 0.009), and 4.5 cm(2)/m(2) (∼0.5 kg) of SAT (P = 0.004) and gained more VAT, 5.1 cm(2)/m(2) (∼0.7 kg) (P = 0.02). Patients with DLT had lower SM index (37.2 vs 44.3 cm(2)/m(2), P = 0.003) and a higher vandetanib serum concentration (1091 vs 739 ng/mL, P = 0.03). Patients with SM index <43.1 cm(2)/m(2) had a higher probability of DLT (73% vs 14%, P = 0.004) and a higher vandetanib serum concentration (1037 vs 745 ng/mL, P = 0.04). Patients with the highest compared with the intermediate and lower levels of vandetanib serum concentration experienced more DLT, respectively, 78% vs 40% vs 20% (P = 0.04). CONCLUSIONS: Muscle and adipose tissues are restored after only 3 months of vandetanib treatment. Patients with low muscle mass had high vandetanib serum concentration and high incidence of toxicities.


Asunto(s)
Composición Corporal , Músculo Esquelético/metabolismo , Piperidinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/uso terapéutico , Neoplasias de la Tiroides/tratamiento farmacológico , Adulto , Anciano , Carcinoma Neuroendocrino , Estudios Cruzados , Femenino , Humanos , Masculino , Persona de Mediana Edad , Piperidinas/efectos adversos , Piperidinas/sangre , Quinazolinas/efectos adversos , Quinazolinas/sangre , Neoplasias de la Tiroides/metabolismo
9.
Oncoimmunology ; 2(2): e23079, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23525192

RESUMEN

Imatinib mesylate (IM) is a small molecule inhibitor of protein tyrosine kinases. In addition to its direct effect on malignant cells, it has been suggested IM may activate of natural killer (NK) cells, hence exerting immunomodulatory functions. In preclinical settings, improved antitumor responses have been observed when IM and interleukin-2 (IL-2), a cytokine that enhances NK cells functions, were combined. The goals of this study were to determine the maximum tolerated dose (MTD) of IL-2 combined with IM at a constant dose of 400 mg, the pharmacokinetics of IM and IL-2, as well as toxicity and clinical efficacy of this immunotherapeutic regimen in patients affected by advanced tumors. The treatment consisted in 50 mg/day cyclophosphamide from 21 d before the initiation of IM throughout the first IM cycle (from D-21 to D14), 400 mg/day IM for 14 d (D1 to D14) combined with escalating doses of IL-2 (3, 6, 9 and 12 MIU/day) from days 10 to 14. This treatment was administered at three week intervals to 17 patients. Common side effects of the combination were mild to moderate, including fever, chills, fatigue, nausea and hepatic enzyme elevation. IL-2 dose level II, 6 MIU/day, was determined as the MTD with the following dose-limiting toxicities: systemic capillary leak syndrome, fatigue and anorexia. Pharmacokinetic studies revealed that the area under the curve and the maximum concentration of IM and its main metabolite CGP74588 increased significantly when IM was concomitantly administered with IL-2. In contrast, IM did not modulate IL-2 pharmacokinetics. No objective responses were observed. The best response obtained was stable disease in 8/17 (median duration: 12 weeks). Finally, IL-2 augmented the impregnation of IM and its metabolite. The combination of IM (400 mg/day) and IL-2 (6 MIU/day) in tumors that express IM targets warrants further investigation.

10.
Am J Obstet Gynecol ; 207(6): 514.e1-7, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23174392

RESUMEN

OBJECTIVES: The use of taxanes (paclitaxel and docetaxel) in pregnant cancer patients is increasing. We aimed to compare their transplacental transfer using the gold standard human placental perfusion model, to guide drug selection. STUDY DESIGN: Term placentas were perfused with paclitaxel or docetaxel and 2 different albumin concentrations. Main transfer parameters such as fetal transfer rate (FTR), clearance index, and placental uptake of taxanes were assessed. RESULTS: Twelve placentas were perfused, 6 with paclitaxel and 6 with docetaxel. Mean FTR of paclitaxel decreased significantly from 5.67 ± 0.02% in low albumin conditions to 1.72 ± 0.09% in physiological albumin conditions. Similarly, mean clearance index decreased significantly from 0.22 ± 0.02 to 0.09 ± 0.01. Regarding docetaxel, mean FTR were similar in low albumin and physiological conditions (5.03 ± 0.60% and 4.04 ± 0.22%, respectively) while mean clearance index decreased significantly from 0.18 ± 0.02 to 0.13 ± 0.01. Taxanes accumulation in cotyledon was similar for docetaxel and paclitaxel: 4.54 ± 1.84% vs 3.31 ± 1.88%, respectively. CONCLUSION: Transplacental transfer and placental accumulation of paclitaxel and docetaxel were low and similar, especially in physiological conditions of albumin. Further studies are warranted to optimize the selection of a taxane in pregnant cancer patients.


Asunto(s)
Antineoplásicos/farmacocinética , Intercambio Materno-Fetal , Paclitaxel/farmacocinética , Placenta/fisiología , Taxoides/farmacocinética , Adulto , Antineoplásicos/uso terapéutico , Docetaxel , Femenino , Humanos , Neoplasias/tratamiento farmacológico , Paclitaxel/uso terapéutico , Embarazo , Taxoides/uso terapéutico
11.
J Chromatogr B Analyt Technol Biomed Life Sci ; 879(11-12): 743-50, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21377940

RESUMEN

Among antitumor oxazaphosphorine drugs, the prodrug ifosfamide (IFO) and its analogs require metabolic activation by specific liver cytochrome P450 (CYP) enzymes to become therapeutically active. New 7,9-dimethyl-ifosfamide analogs have shown greater cytotoxic activity than IFO, whereas side-chain oxidation still occurred leading to monochloroacetone after N-dechloropropylation. A sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay was developed and validated for the simultaneous quantitation of the prodrug 7S,9S-dimethyl-ifosfamide (diMeIFO) and its two inactive metabolites, N(2)- and N(3)-deschloropropyl-dimethylifosfamide (N(2)-DCP-diMeIFO and N(3)-DCP-diMeIFO) in mouse plasma. After protein precipitation with methanol, the analytes were separated by isocratic reversed-phase chromatography with (methanol/ammonium formate pH 5.5, 60:40, v/v) and detected by tandem mass spectrometry using multiple reaction monitoring of transitions ions m/z 289→168 for diMeIFO, m/z 213→168 for N(2)-DCP-diMeIFO, m/z 213→92 for N(3)-DCP-diMeIFO and m/z 261→154 for IFO (internal standard). The calibration curves were linear over the concentration range of 20-10,000ng/mL for the three analytes. Mean extraction recoveries from mouse plasma were 99, 96, 99 and 100% for diMeIFO, N(2)-DCP-diMeIFO, N(3)-DCP-diMeIFO and IFO, respectively. The lower limit of quantitation for diMeIFO and its metabolites was 20 ng/mL in 50 µL plasma. The method was accurate with calculated bias from -5.8 to 4.0% for diMeIFO, from -1.1 to 10.6% for N(2)-DCP-diMeIFO and from -6.9 to 9.8% for N(3)-DCP-diMeIFO, and precise with coefficients of variation lower than 6.8%, 7.8% and 14.3%, respectively. The assay was successfully applied to a preliminary pharmacokinetic study of diMeIFO and of its metabolites in mice.


Asunto(s)
Cromatografía Liquida/métodos , Ifosfamida/análogos & derivados , Ifosfamida/sangre , Espectrometría de Masas en Tándem/métodos , Animales , Estabilidad de Medicamentos , Femenino , Humanos , Ifosfamida/farmacocinética , Análisis de los Mínimos Cuadrados , Ratones , Ratones Desnudos , Reproducibilidad de los Resultados , Rabdomiosarcoma/sangre , Rabdomiosarcoma/metabolismo , Sensibilidad y Especificidad
12.
Expert Opin Drug Metab Toxicol ; 6(8): 919-38, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20446865

RESUMEN

IMPORTANCE OF THE FIELD: The oxazaphosphorines (cyclophosphamide, ifosfamide and trofosfamide) are widely used in clinical practice for their antitumor and immunomodulatory activities. However, their use is associated with toxicities. The metabolism of oxazaphosphorines involves cytochrome P450 biotransformations, leading to highly reactive metabolites such as acrolein and chloroacetaldehyde responsible for urotoxicity, neurotoxicity and nephrotoxicity. While the mechanisms behind these toxicities remain under investigation, some advances have been made, as exemplified by the use of mesna to limit acrolein related urotoxicity. AREAS COVERED IN THIS REVIEW: This review highlights potential strategies for limiting side effects commonly associated with the oxazaphosphorine drugs, through pharmacological or medicinal chemistry-based approaches. WHAT THE READER WILL GAIN: The readers will gain a comprehensive review of these approaches to treatment in terms of: i) pharmacology: use of antidotes and modification of metabolism through inhibition/induction of CYP enzymes or use of gene therapy; and ii) medicinal chemistry: the design of new drugs to target cancer cells and avoid CYP biotransformation with pre-activated prodrugs or with side-chain substituted analogues. TAKE HOME MESSAGE: An increased knowledge of oxazaphosphorines' metabolism and toxicity may allow the development of new anticancer drugs combined with drug delivery systems to circumvent drug toxicity, providing increased tumoral specificity and greater anticancer activity.


Asunto(s)
Ciclofosfamida/farmacología , Diseño de Fármacos , Ifosfamida/farmacología , Animales , Antineoplásicos Alquilantes/efectos adversos , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacología , Ciclofosfamida/efectos adversos , Ciclofosfamida/análogos & derivados , Ciclofosfamida/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Sistemas de Liberación de Medicamentos , Terapia Genética/métodos , Humanos , Ifosfamida/efectos adversos , Ifosfamida/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/fisiopatología
13.
J Pharm Biomed Anal ; 52(3): 391-7, 2010 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-19804952

RESUMEN

The quaternary benzo[c]phenanthridine alkaloid, sanguinarine (SA), has been detected in the mustard oil contaminated with Argemone mexicana, which produced severe human intoxications during epidemic dropsy in India. Today, SA metabolism in human and in rat has not yet been fully elucidated. The goal of this study is to investigate the oxidative metabolites of SA formed during incubations with rat liver microsomes (RLM) and recombinant human cytochrome P450 (CYP) and to tentatively identify the CYP isoforms involved in SA detoxification. Metabolites were analyzed by liquid chromatography combined with electrospray ionization-tandem mass spectrometry. Up to six metabolites were formed by RLM and their modified structure has been proposed using their mass spectra and mass shifts from SA (m/z 332). The main metabolite M2 (m/z 320) resulted from ring-cleavage of SA followed by demethylation, whereas M4 (m/z 348) is oxidized by CYP in the presence of NADPH. The diol-sanguinarine metabolite M6 (m/z 366) formed by RLM might derive from a putative epoxy-sanguinarine metabolite M5 (m/z 348). M4 and M6 could be detected in rat urine as their respective glucuronides. 5,6-Dihydrosanguinarine is the prominent derivative formed from SA in cells expressing no CYP. Oxidative biotransformation of SA was investigated using eight human CYPs: only CYP1A1 and CYP1A2 displayed activity.


Asunto(s)
Benzofenantridinas/metabolismo , Cromatografía Liquida/métodos , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Isoquinolinas/metabolismo , Microsomas Hepáticos/metabolismo , Espectrometría de Masas en Tándem/métodos , Animales , Biotransformación , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Ionización de Electrospray/métodos
14.
Int J Pharm ; 379(2): 235-43, 2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19467308

RESUMEN

Glycerolipidic prodrug is an interesting concept to enhance lymphatic absorption of polar drugs intended to oral delivery such as didanosine (ddI). In order to improve ddI bioavailability, two didanosine glycerolipidic prodrugs, the phosphorylated (ProddIP) and the non-phosphorylated derivatives (ProddINP) were synthesized to follow triglyceride metabolism. The biomimetism approach of these prodrugs has been studied in vitro at two steps. First, liposomal formulation of each prodrug was incubated with a lipolysis model based on pancreatin and analysed using liquid chromatography combined with tandem mass spectrometry (LC-MS/MS). These experiments evidenced that both didanosine prodrugs were recognized by the lipases; as expected, they were cleaved at both positions sn-1 and sn-3 of glycerol. ProddIP was metabolised twice more rapidly than ProddINP suggesting an implication of some phospholipases in ProddIP degradation. Secondly, the detection of dideoxyadenosine triphosphate (ddA-TP) into HIV-1 infected cells after their incubation with ProddINP loaded liposomes evidenced their ability to release ddI that could penetrate into the cells and be metabolised by intracellular kinases. These results confirmed that the synthesized glycerolipidic prodrugs of didanosine could be investigated for a biomimetic approach with final aiming of increasing the drug oral bioavailability by enhancing intestinal absorption.


Asunto(s)
Materiales Biomiméticos/metabolismo , Química Farmacéutica/métodos , Modelos Biológicos , Profármacos/metabolismo , Animales , Materiales Biomiméticos/química , Humanos , Espectrometría de Masas/métodos , Redes y Vías Metabólicas/fisiología , Profármacos/química
15.
J Pharmacol Exp Ther ; 328(2): 598-609, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19017849

RESUMEN

Ifosfamide is a well known prodrug for cancer treatment with cytochrome P450 metabolism. It is associated with both antitumor activity and toxicities. Isophosphoramide mustard is the bisalkylating active metabolite, and acrolein is a urotoxic side product. Because acrolein toxicity is limited by coadministration of sodium mercaptoethanesulfonate, the incidence of urotoxicity has been lowered. Current evidence suggests that chloroacetaldehyde, a side-chain oxidation metabolite, is responsible for neurotoxicity and nephrotoxicity. The aim of our research is to prevent chloroacetaldehyde formation using new enantioselectively synthesized ifosfamide analogs, i.e., C7,C9-dimethyl-ifosfamide. We hypothesize that reduced toxicogenic catabolism may induce less toxicity without changing anticancer activity. Metabolite determinations of the dimethyl-ifosfamide analogs were performed using liquid chromatography and tandem mass spectrometry after in vitro biotransformation by drug-induced rat liver microsomes and human microsomes expressing the main CYP3A4 and minor CYP2B6 enzymes. Both human and rat microsomes incubations produced the same N-deschloroalkylated and 4-hydroxylated metabolites. A coculture assay of 9L rat glioblastoma cells and rat microsomes was performed to evaluate their cytotoxicity. Finally, a mechanistic study using (31)P NMR kinetics allowed estimating the alkylating activity of the modified mustards. The results showed that C7,C9-dimethyl-ifosfamide exhibited increased activities, although they were still metabolized through the same N-deschloroalkylation pathway. Analogs were 4 to 6 times more cytotoxic than ifosfamide on 9L cells, and the generated dimethylated mustards were 28 times faster alkylating agents than ifosfamide mustards. Among these new ifosfamide analogs, the 7S,9R-enantiomer will be assessed for further in vivo investigations for its anticancer activity and its toxicological profile.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Ifosfamida/efectos adversos , Síndrome Nefrótico/inducido químicamente , Síndromes de Neurotoxicidad , Animales , Antineoplásicos Alquilantes/uso terapéutico , Biotransformación , Células Cultivadas , Humanos , Ifosfamida/análogos & derivados , Ifosfamida/uso terapéutico , Cinética , Masculino , Profármacos/efectos adversos , Profármacos/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Drug Metab Dispos ; 36(8): 1570-7, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18474674

RESUMEN

Gemcitabine (2',2'-difluorodeoxyribofuranosylcytosine; dFdC) is an anticancer nucleoside analog active against wide variety of solid tumors. However, this compound is rapidly inactivated by enzymatic deamination and can also induce drug resistance. To overcome the above drawbacks, we recently designed a new squalenoyl nanomedicine of dFdC [4-N-trisnorsqualenoyl-gemcitabine (SQdFdC)] by covalently coupling gemcitabine with the 1,1',2-trisnorsqualenic acid; the resultant nanomedicine displayed impressively greater anticancer activity compared with the parent drug in an experimental murine model. In the present study, we report that SQdFdC nanoassemblies triggered controlled and prolonged release of dFdC and displayed considerably greater t(1/2) (approximately 3.9-fold), mean residence time (approximately 7.5-fold) compared with the dFdC administered as a free drug in mice. It was also observed that the linkage of gemcitabine to the 1,1',2-trisnorsqualenic acid noticeably delayed the metabolism of dFdC into its inactive difluorodeoxyuridine (dFdU) metabolite, compared with dFdC. Additionally, the elimination of SQdFdC nanoassemblies was considerably lower compared with free dFdC, as indicated by lower radioactivity found in urine and kidneys, in accordance with the plasmatic concentrations of dFdU. SQdFdC nanoassemblies also underwent considerably higher distribution to the organs of the reticuloendothelial system, such as spleen and liver (p < 0.05), both after single- or multiple-dose administration schedule. Herein, this paper brings comprehensive pharmacokinetic and biodistribution insights that may explain the previously observed greater efficacy of SQdFdC nanoassemblies against experimental leukemia.


Asunto(s)
Antineoplásicos/farmacocinética , Desoxicitidina/análogos & derivados , Escualeno/metabolismo , Animales , Antineoplásicos/sangre , Antineoplásicos/metabolismo , Cromatografía Liquida , Desoxicitidina/sangre , Desoxicitidina/metabolismo , Desoxicitidina/farmacocinética , Femenino , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos DBA , Espectrometría de Masas en Tándem , Distribución Tisular , Gemcitabina
17.
J Microencapsul ; 24(8): 715-30, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17926165

RESUMEN

Hepatic veino-occlusive disease (HVOD) is the most severe and frequent busulphan high risk injury. The development of busulphan-loaded'stealth'nanospheres which avoid liver accumulation should minimize busulphan toxicity. To reach this important goal, this study has attempted to develop poly(ethylene glycol) (PEG)-coated nanospheres using polyester-PEG diblock copolymers and studied the busulphan physico-chemical characteristics related to its encapsulation. Nanospheres were prepared by nanoprecipitation and by emulsion-solvent evaporation techniques. They were characterized by microscopy and dynamic light-scattering. Busulphan interactions with adjuvants were studied using gas chromatography and mass spectrometry, high performance thin-layer chromatography and dynamic scanning calorimetry. These investigations explain both the low busulphan loadings obtained ( approximately 1% w/w) and its rapid release from nanoparticles. These experiments constituted an essential step for the development of busulphan-loaded long-circulating nanospheres.


Asunto(s)
Antineoplásicos Alquilantes/química , Busulfano/química , Nanocápsulas/química , Nanosferas/química , Polietilenglicoles/química , Antineoplásicos Alquilantes/efectos adversos , Antineoplásicos Alquilantes/farmacología , Busulfano/efectos adversos , Busulfano/farmacología , Rastreo Diferencial de Calorimetría , Preparaciones de Acción Retardada/efectos adversos , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Enfermedad Veno-Oclusiva Hepática/inducido químicamente , Nanocápsulas/ultraestructura , Nanosferas/ultraestructura
18.
Artículo en Inglés | MEDLINE | ID: mdl-17851141

RESUMEN

Gemcitabine-squalene is a new prodrug that self-organizes in water forming nanoassemblies. It exhibits better anti-cancer properties in vitro and in vivo than gemcitabine. A liquid chromatography/tandem mass spectrometry assay of gemcitabine-squalene and gemcitabine was developed in human plasma in order to quantitate gemcitabine and its squalene conjugate. After protein precipitation with acetonitrile/methanol (90/10, v/v), the compounds were analyzed by reversed-phase high performance liquid chromatography and detected by tandem mass spectrometry using multiple reaction monitoring. The method was linear over the concentration range of 10-10,000 ng/ml of human plasma for both compounds with an accuracy lower than 10.4% and a precision below 14.8%. The method showed a lower limit of quantitation of 10 ng/ml of human plasma for dFdC and dFdC-SQ. A preliminary in vivo study in mice was shown as application of the method as no significant difference between human and mice plasma for the analysis of dFdC and dFdC-SQ was demonstrated.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Desoxicitidina/análogos & derivados , Escualeno/sangre , Espectrometría de Masas en Tándem/métodos , Desoxicitidina/sangre , Desoxicitidina/química , Humanos , Estructura Molecular , Reproducibilidad de los Resultados , Escualeno/química , Gemcitabina
19.
Bioorg Med Chem Lett ; 17(8): 2237-40, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17276686

RESUMEN

Novel glycerolipidic prodrugs of didanosine and didanosine monophosphate designed to by-pass the hepatic first pass metabolism were synthesized and tested for their cytotoxicity and anti-HIV-1 activity. Formulation as liposomes of dipalmitoylphosphatidylcholine was elaborated. A simple quantitative HPLC-UV method was developed and validated, and ESI-MS was used for qualitative purpose. These two prodrugs exhibited promising biological activities against HIV-1 in in vitro infected cell culture.


Asunto(s)
Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/farmacocinética , Didanosina/síntesis química , Didanosina/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Sistema Linfático/metabolismo , Profármacos/síntesis química , Profármacos/farmacocinética , Triglicéridos/síntesis química , Triglicéridos/farmacocinética , Fármacos Anti-VIH/administración & dosificación , Disponibilidad Biológica , Células Cultivadas , Didanosina/administración & dosificación , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/virología , Liposomas , Sistema Linfático/virología , Nucleósidos de Purina/administración & dosificación , Nucleósidos de Purina/uso terapéutico , Replicación Viral/efectos de los fármacos
20.
Drug Metab Dispos ; 35(2): 306-14, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17132762

RESUMEN

Busulfan is an example of a drug eliminated through glutathione S-transferase (GST)-catalyzed conjugation with reduced glutathione (GSH). We studied the pharmacokinetics and toxicity of busulfan in C57BL6 mice in correlation with liver GST activity and GSH synthesis by accurate determination of precursors, namely, gamma-glutamyl-cysteine and cysteine. A significantly lower incidence of acute toxicity was observed in mice receiving busulfan 16.5 mg/kg twice a day compared with animals receiving 33 mg/kg once a day. In both cases, a total dose of 132 mg/kg was administered over 4 days. The difference in toxicity was explained by pharmacokinetics since a strong induction of clearance was observed only in animals treated twice daily. Induction of metabolism was correlated with an increase in liver cysteine content and enhanced glutathione synthesis rate, whereas GST activity was unchanged. To our knowledge, this is the first time that in vivo flux of GSH synthesis has been shown to be closely related to a drug plasma clearance and toxicity. These results allow hypothesizing that GSH liver synthesis may directly influence busulfan clearance in humans with possible implications in the occurrence of hepatic veno-occlusive disease.


Asunto(s)
Busulfano/toxicidad , Glutatión/biosíntesis , Animales , Trasplante de Médula Ósea , Busulfano/farmacocinética , Interacciones Farmacológicas , Glutatión Transferasa/metabolismo , Enfermedad Veno-Oclusiva Hepática/inducido químicamente , Hígado/metabolismo , Masculino , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA