Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Eur J Clin Pharmacol ; 80(9): 1399-1407, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38904799

RESUMEN

OBJECTIVE: This study is aimed at investigating the pharmacokinetic (PK) characteristics of pegylated liposomal mitoxantrone (PLM) in patients with relapsed/refractory lymphoma or small cell lung cancer (SCLC) by constructing population pharmacokinetic (popPK) models for both liposome-encapsulated mitoxantrone and free mitoxantrone. METHODS: A total of 23 patients with relapsed/refractory lymphoma and 42 patients with SCLC were included. A popPK model was simultaneously developed utilizing a non-linear mixed effects model (NONMEM) to explore the PK profiles of liposome-encapsulated mitoxantrone and free mitoxantrone. Clearance (CL) and distribution volume (V) were calculated, and covariate analysis was employed to evaluate the influence of patient disease type, demographic information, and biochemical indicators of liver and kidney function on PK parameters. RESULTS: The concentration-time profiles for both liposome-encapsulated mitoxantrone and free mitoxantrone were described by a one-compartment model. The release (Rel) of liposome-encapsulated mitoxantrone to free mitoxantrone was determined to be 0.0191 L/h, and the V of liposome-encapsulated mitoxantrone was 2.32 L. The apparent CL of free mitoxantrone was estimated at 1.66 L/h. The apparent V of free mitoxantrone was 35.8 L in patients with relapsed/refractory lymphoma and 22.2 L for patients with SCLC. In patients with relapsed/refractory lymphoma, lower maximum concentration (Cmax) and higher apparent V of free mitoxantrone were observed compared with patients with SCLC. CONCLUSION: The popPK characteristics of both liposome-encapsulated and free mitoxantrone in patients with relapsed/refractory lymphoma or SCLC were effectively described by a one-compartment model.


Asunto(s)
Liposomas , Neoplasias Pulmonares , Mitoxantrona , Modelos Biológicos , Carcinoma Pulmonar de Células Pequeñas , Humanos , Mitoxantrona/farmacocinética , Mitoxantrona/administración & dosificación , Masculino , Persona de Mediana Edad , Femenino , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Anciano , Adulto , Linfoma/tratamiento farmacológico , Antineoplásicos/farmacocinética , Antineoplásicos/administración & dosificación , Anciano de 80 o más Años , Polietilenglicoles/farmacocinética , Polietilenglicoles/administración & dosificación
2.
Drug Metab Dispos ; 45(11): 1166-1177, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28904007

RESUMEN

Multidrug resistance (MDR) caused by the overexpression of ATP-binding cassette (ABC) transporters in cancer cells is a major obstacle in cancer chemotherapy. Previous studies have shown that curcumin, a natural product and a dietary constituent of turmeric, inhibits the function of MDR-related ABC transporters, including ABCB1, ABCC1, and especially ABCG2. However, the limited bioavailability of curcumin prevents its use for modulation of the function of these transporters in the clinical setting. In this study, we investigated the effects of 24 synthetic curcumin analogs with increased bioavailability on the transport function of ABCG2. The screening of the 24 synthetic analogs by means of flow cytometry revealed that four of the curcumin analogs (GO-Y030, GO-Y078, GO-Y168, and GO-Y172) significantly inhibited the efflux of the ABCG2 substrates, mitoxantrone and pheophorbide A, from ABCG2-overexpressing K562/breast cancer resistance protein (BCRP) cells. Biochemical analyses showed that GO-Y030, GO-Y078, and GO-Y172 stimulated the ATPase activity of ABCG2 at nanomolar concentrations and inhibited the photolabeling of ABCG2 with iodoarylazidoprazosin, suggesting that these analogs interact with the substrate-binding sites of ABCG2. In addition, when used in cytotoxicity assays, GO-Y030 and GO-Y078 were found to improve the sensitivity of the anticancer drug, SN-38, in K562/BCRP cells. Taken together, these results suggest that nontoxic synthetic curcumin analogs with increased bioavailability, especially GO-Y030 and GO-Y078, inhibit the function of ABCG2 by directly interacting at the substrate-binding site. These synthetic curcumin analogs could therefore be developed as potent modulators to overcome ABCG2-mediated MDR in cancer cells.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Antineoplásicos/farmacología , Curcumina/análogos & derivados , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/química , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Azidas/química , Derivados del Benceno , Disponibilidad Biológica , Transporte Biológico , Camptotecina/análogos & derivados , Camptotecina/farmacología , Línea Celular Tumoral , Curcumina/farmacología , Sinergismo Farmacológico , Citometría de Flujo , Humanos , Irinotecán , Cetonas , Mitoxantrona/farmacocinética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Prazosina/análogos & derivados , Prazosina/química
3.
Med Res Rev ; 36(2): 248-99, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26286294

RESUMEN

Mitoxantrone is a synthetic anthracenedione originally developed to improve the therapeutic profile of the anthracyclines and is commonly applied in the treatment of breast and prostate cancers, lymphomas, and leukemias. A comprehensive overview of the drug's molecular, biochemical, and cellular pharmacology is presented here, beginning with the cardiotoxic nature of its predecessor doxorubicin and how these properties shaped the pharmacology of mitoxantrone itself. Although mitoxantrone is firmly established as a DNA topoisomerase II poison within mammalian cells, it is now clear that the drug interacts with a much broader range of biological macromolecules both covalently and noncovalently. Here, we consider each of these interactions in the context of their wider biological relevance to cancer therapy and highlight how they may be exploited to further enhance the therapeutic value of mitoxantrone. In doing so, it is now clear that mitoxantrone is more than just another topoisomerase II poison.


Asunto(s)
Mitoxantrona/farmacología , Inhibidores de Topoisomerasa II/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Ensayos Clínicos como Asunto , Descubrimiento de Drogas , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Mitoxantrona/administración & dosificación , Mitoxantrona/química , Mitoxantrona/farmacocinética , Inhibidores de Topoisomerasa II/química
4.
Drug Dev Ind Pharm ; 42(8): 1351-9, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26754913

RESUMEN

Novel nanostructured lipid-carrageenan hybrid carriers (NLCCs) were exploited for controlled delivery of water soluble chemotherapeutic agent mitoxantrone hydrochloride (MTO) with high loading capacity, sustained release property, and potential for improving oral bioavailability and antitumor efficacy. By introducing the negative polymer of carrageenan, MTO was highly incorporated into NLCCs with encapsulation efficiency of 95.8% by electrostatic interaction. In vivo pharmacokinetics of MTO solution (MTO-Sol) and MTO-NLCCs in rats demonstrated that the apparent bioavailability of MTO-NLCCs was increased to approximate 3.5-fold compared to that of MTO-Sol. The cytotoxicity investigations by MTT method indicated that NLCCs could significantly enhanced the antitumor efficacy against resistant MCF-7/MX cells. The relative cellular association of MTO-NLCCs was 9.2-fold higher than that of MTO-Sol in breast cancer resistance protein (BCRP) over-expressing MCF-7/MX cells, implying that BCRP-mediated drug efflux was diminished by the introduction of NLCCs. The endocytosis inhibition study implied that the NLCCs entered the MCF-7/MX cells by clathrin-mediated endocytosis process, which can bypass the efflux of MTO mediated by BCRP. The new developed NLCCs provide an effective strategy for oral delivery of water-soluble MTO with improved encapsulation efficiency, oral bioavailability, and cytotoxicity against resistant breast cancer cells.


Asunto(s)
Antineoplásicos/química , Carragenina/química , Preparaciones de Acción Retardada/química , Portadores de Fármacos/química , Lípidos/química , Mitoxantrona/química , Nanoestructuras/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Disponibilidad Biológica , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Masculino , Mitoxantrona/administración & dosificación , Mitoxantrona/farmacocinética , Ratas , Ratas Wistar
5.
Neurobiol Dis ; 74: 144-57, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25173807

RESUMEN

Aß accumulation plays a central role in the pathogenesis of Alzheimer's disease (AD). Recent studies suggest that the process of Aß nucleated polymerization is essential for Aß fibril formation, pathology spreading and toxicity. Therefore, targeting this process represents an effective therapeutic strategy to slow or block disease progression. To discover compounds that might interfere with the Aß seeding capacity, toxicity and pathology spreading, we screened a focused library of FDA-approved drugs in vitro using a seeding polymerization assay and identified small molecule inhibitors that specifically interfered with Aß seeding-mediated fibril growth and toxicity. Mitoxantrone, bithionol and hexachlorophene were found to be the strongest inhibitors of fibril growth and protected primary cortical neuronal cultures against Aß-induced toxicity. Next, we assessed the effects of these three inhibitors in vivo in the mThy1-APPtg mouse model of AD (8-month-old mice). We found that mitoxantrone and bithionol, but not hexachlorophene, stabilized diffuse amyloid plaques, reduced the levels of Aß42 oligomers and ameliorated synapse loss, neuronal damage and astrogliosis. Together, our findings suggest that targeting fibril growth and Aß seeding capacity constitutes a viable and effective strategy for protecting against neurodegeneration and disease progression in AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/efectos de los fármacos , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/fisiopatología , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/efectos de los fármacos , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/toxicidad , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Bitionol/farmacocinética , Bitionol/farmacología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Gliosis/tratamiento farmacológico , Gliosis/patología , Gliosis/fisiopatología , Hexaclorofeno/farmacocinética , Hexaclorofeno/farmacología , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitoxantrona/farmacocinética , Mitoxantrona/farmacología , Degeneración Nerviosa/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/fisiología , Fármacos Neuroprotectores/farmacocinética , Fragmentos de Péptidos/toxicidad , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/patología , Placa Amiloide/fisiopatología , Ratas
6.
Pharm Res ; 32(4): 1354-67, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25319103

RESUMEN

PURPOSE: To improve therapeutic activity of mitoxantrone (MTO)-based chemotherapy by reducing toxicity through encapsulation in nanoliposomes and enhancing intracellular drug delivery using short-chain sphingolipid (SCS) mediated tumor cell membrane permeabilization. METHODS: Standard (MTOL) and nanoliposomes enriched with the SCS, C8-Glucosylceramide or C8-Galactosylceramide (SCS-MTOL) were loaded by a transmembrane ammonium sulphate gradient and characterized by DLS and cryo-TEM. Intracellular MTO delivery was measured by flow cytometry and imaged by fluorescence microscopy. In vitro cytotoxicity was studied in breast carcinoma cell lines. Additionally, live cell confocal microscopy addressed the drug delivery mechanism by following the intracellular fate of the nanoliposomes, the SCS and MTO. Intratumoral MTO localization in relation to CD31-positive tumor vessels and CD11b positive cells was studied in an orthotopic MCF-7 breast cancer xenograft. RESULTS: Stable SCS-MTOL were developed increasing MTO delivery and cytotoxicity to tumor cells compared to standard MTOL. This effect was much less pronounced in normal cells. The drug delivery mechanism involved a transfer of SCS to the cell membrane, independently of drug transfer and not involving nanoliposome internalization. MTO was detected intratumorally upon MTOL and SCS-MTOL treatment, but not after free MTO, suggesting an important improvement in tumor drug delivery by nanoliposomal formulation. Nanoliposomal MTO delivery and cellular uptake was heterogeneous throughout the tumor and clearly correlated with CD31-positive tumor vessels. Yet, MTO uptake by CD11b positive cells in tumor stroma was minor. CONCLUSIONS: Nanoliposomal encapsulation improves intratumoral MTO delivery over free drug. Liposome bilayer-incorporated SCS preferentially permeabilize tumor cell membranes enhancing intracellular MTO delivery.


Asunto(s)
Antineoplásicos/administración & dosificación , Galactosilceramidas/química , Glucosilceramidas/química , Mitoxantrona/administración & dosificación , Nanopartículas/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Humanos , Liposomas , Células MCF-7 , Mitoxantrona/farmacocinética , Mitoxantrona/farmacología
7.
Drug Metab Dispos ; 42(5): 943-6, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24568887

RESUMEN

Lignans are phytoestrogens that are metabolized by the gut microbiota to enterodiol and enterolactone, the main biologically active enterolignans. Substantial interindividual variation in plasma concentration and urinary excretion of enterolignans has been reported, this being determined, at least in part, by the intake of lignan precursors, the gut microbiota, and the host's phase 2 conjugating enzyme activity. However, the role of ATP-binding cassette (ABC) transporters in the transport and disposition of enterolactone has not been reported so far. Active transport assays using parental and Madin-Darby canine kidney epithelial cells transduced with murine and human ABCG2 showed a significant increase in apically directed translocation of enterolactone in transduced cells, which was confirmed by using the selective ABCG2 inhibitor Ko143. In addition, enterolactone also inhibited transport of the antineoplastic agent mitoxantrone as a model substrate, with inhibition percentages of almost 40% at 200 µM for human ABCG2. Furthermore, the endogenous levels in plasma and milk of enterolactone in wild-type and Abcg2((-/-)) knockout female mice were analyzed. The milk/plasma ratio decreased significantly in the Abcg2((-/-)) phenotype, as compared with the wild-type mouse group (0.4 ± 0.1 as against 6.4 ± 2.6). This paper is the first to report that enterolactone is a transported substrate and therefore most probably a competitive inhibitor of ABCG2, which suggests it has a role in the interindividual variations in the disposition of enterolactone and its secretion into milk. The inhibitory activity identified provides a solid basis for further investigation in possible food-drug interactions.


Asunto(s)
4-Butirolactona/análogos & derivados , Transportadoras de Casetes de Unión a ATP/fisiología , Lignanos/farmacocinética , Leche/química , Proteínas de Neoplasias/fisiología , 4-Butirolactona/sangre , 4-Butirolactona/metabolismo , 4-Butirolactona/farmacocinética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/genética , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Transporte Biológico , Dicetopiperazinas , Perros , Femenino , Compuestos Heterocíclicos de 4 o más Anillos , Lignanos/sangre , Células de Riñón Canino Madin Darby , Ratones , Ratones Noqueados , Mitoxantrona/metabolismo , Mitoxantrona/farmacocinética , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Especificidad por Sustrato
8.
Yao Xue Xue Bao ; 49(2): 256-9, 2014 Feb.
Artículo en Zh | MEDLINE | ID: mdl-24761619

RESUMEN

To investigate whether accelerated blood clearance (ABC) phenomenon could be induced after repeated injection of mitoxantrone thermosensitive liposomes, LC-MS/MS and enzyme linked immunosorbent assay (ELISA) were used to measure the concentration of mitoxantrone and the anti-polyethylene glycol (PEG) IgM levels in rat plasma, separately. The drug was rapidly cleared away after the second administration. The anti-PEG IgM was detected after the first dose which was neutralized quickly after the second dose. It is proved that repeated administration of mitoxantrone thermosensitive liposomes in rat caused the ABC phenomenon.


Asunto(s)
Antineoplásicos/farmacocinética , Inmunoglobulina M/sangre , Liposomas/farmacocinética , Mitoxantrona/farmacocinética , Polietilenglicoles/farmacocinética , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Cromatografía Líquida de Alta Presión , Liposomas/administración & dosificación , Liposomas/sangre , Masculino , Tasa de Depuración Metabólica , Mitoxantrona/administración & dosificación , Mitoxantrona/sangre , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Ratas , Ratas Wistar , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
9.
Adv Healthc Mater ; 13(12): e2303631, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38278138

RESUMEN

Currently, the secondary development and modification of clinical drugs has become one of the research priorities. Researchers have developed a variety of TME-responsive nanomedicine carriers to solve certain clinical problems. Unfortunately, endogenous stimuli such as reactive oxygen species (ROS), as an important prerequisite for effective therapeutic efficacy, are not enough to achieve the expected drug release process, therefore, it is difficult to achieve a continuous and efficient treatment process. Herein, a self-supply ROS-responsive cascade polyprodrug (PMTO) is designed. The encapsulation of the chemotherapy drug mitoxantrone (MTO) in a polymer backbone could effectively reduce systemic toxicity when transported in vivo. After PMTO is degraded by endogenous ROS of the TME, another part of the polyprodrug backbone becomes cinnamaldehyde (CA), which can further enhance intracellular ROS, thereby achieving a sustained drug release process. Meanwhile, due to the disruption of the intracellular redox environment, the efficacy of chemotherapy drugs is enhanced. Finally, the anticancer treatment efficacy is further enhanced due to the mild hyperthermia effect of PMTO. In conclusion, the designed PMTO demonstrates remarkable antitumor efficacy, effectively addressing the limitations associated with MTO.


Asunto(s)
Acroleína/análogos & derivados , Mitoxantrona , Especies Reactivas de Oxígeno , Mitoxantrona/química , Mitoxantrona/farmacología , Mitoxantrona/farmacocinética , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos , Ratones , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Hipertermia Inducida/métodos , Profármacos/química , Profármacos/farmacología , Acroleína/química , Acroleína/farmacología , Ratones Endogámicos BALB C , Liberación de Fármacos , Femenino , Ratones Desnudos , Portadores de Fármacos/química , Polímeros/química
10.
ACS Sens ; 9(5): 2567-2574, 2024 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-38696667

RESUMEN

In vivo drug monitoring is crucial for evaluating the effectiveness and safety of drug treatment. Blood sampling and analysis is the current gold standard but needs professional skills and cannot meet the requirements of point-of-care testing. Dermal interstitial fluid (ISF) showed great potential to replace blood for in vivo drug monitoring; however, the detection was challenging, and the drug distribution behavior in ISF was still unclear until now. In this study, we proposed surface-enhanced Raman spectroscopy (SERS) microneedles (MNs) for the painless and real-time analysis of drugs in ISF after intravenous injection. Using methylene blue (MB) and mitoxantrone (MTO) as model drugs, the innovative core-satellite structured Au@Ag SERS substrate, hydrogel coating over the MNs, rendered sensitive and quantitative drug detection in ISF of mice within 10 min. Based on this technique, the pharmacokinetics of the two drugs in ISF was investigated and compared with those in blood, where the drugs were analyzed via liquid chromatography-mass spectrometry. It was found that the MB concentration in ISF and blood was comparable, whereas the concentration of MTO in ISF was 2-3 orders of magnitude lower than in blood. This work proposed an efficient tool for ISF drug monitoring. More importantly, it experimentally proved that the penetration ratio of blood to ISF was drug-dependent, providing insightful information into the potential of ISF as a blood alternative for in vivo drug detection.


Asunto(s)
Monitoreo de Drogas , Líquido Extracelular , Hidrogeles , Azul de Metileno , Agujas , Espectrometría Raman , Animales , Espectrometría Raman/métodos , Líquido Extracelular/química , Azul de Metileno/química , Ratones , Hidrogeles/química , Monitoreo de Drogas/métodos , Monitoreo de Drogas/instrumentación , Plata/química , Mitoxantrona/sangre , Mitoxantrona/análisis , Mitoxantrona/farmacocinética , Oro/química , Piel/metabolismo , Piel/química
11.
J Colloid Interface Sci ; 669: 731-739, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38735255

RESUMEN

HYPOTHESIS: Hydrophilic cationic drugs such as mitoxantrone hydrochloride (MTO) pose a significant delivery challenge to the development of nanodrug systems. Herein, we report the use of a hydrophobic ion-pairing strategy to enhance the nano-assembly of MTO. EXPERIMENTS: We employed biocompatible sodium cholesteryl sulfate (SCS) as a modification module to form stable ion pairs with MTO, which balanced the intermolecular forces and facilitated nano-assembly. PEGylated MTO-SCS nanoassemblies (pMS NAs) were prepared via nanoprecipitation. We systematically evaluated the effect of the ratio of the drug module (MTO) to the modification module (SCS) on the nanoassemblies. FINDINGS: The increased lipophilicity of MTO-SCS ion pair could significantly improve the encapsulation efficiency (∼97 %) and cellular uptake efficiency of MTO. The pMS NAs showed prolonged blood circulation, maintained the same level of tumor antiproliferative activity, and exhibited reduced toxicity compared with the free MTO solution. It is noteworthy that the stability, cellular uptake, cytotoxicity, and in vivo pharmacokinetic behavior of the pMS NAs increased in proportion to the molar ratio of SCS to MTO. This study presents a self-assembly strategy mediated by ion pairing to overcome the challenges commonly associated with the poor assembly ability of hydrophilic cationic drugs.


Asunto(s)
Antineoplásicos , Ésteres del Colesterol , Interacciones Hidrofóbicas e Hidrofílicas , Mitoxantrona , Mitoxantrona/química , Mitoxantrona/farmacología , Mitoxantrona/farmacocinética , Humanos , Animales , Ésteres del Colesterol/química , Antineoplásicos/química , Antineoplásicos/farmacología , Ratones , Proliferación Celular/efectos de los fármacos , Cationes/química , Supervivencia Celular/efectos de los fármacos , Tamaño de la Partícula , Nanopartículas/química , Propiedades de Superficie , Portadores de Fármacos/química , Ensayos de Selección de Medicamentos Antitumorales , Línea Celular Tumoral , Polietilenglicoles/química
12.
J Pharmacol Exp Ther ; 344(2): 467-78, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23192654

RESUMEN

Cardiotoxicity from the antitumor anthracycline doxorubicin correlates with doxorubicin cardiac levels, redox activation to superoxide anion (O(2)(.-)) and hydrogen peroxide (H(2)O(2)), and formation of the long-lived secondary alcohol metabolite doxorubicinol. Cardiotoxicity may first manifest during salvage therapy with other drugs, such as the anthracenedione mitoxantrone. Minimal evidence for cardiotoxicity in anthracycline-pretreated patients with refractory-relapsed non-Hodgkin lymphoma was observed with the novel anthracenedione pixantrone. We characterized whether pixantrone and mitoxantrone caused different effects on doxorubicin levels, redox activation, and doxorubicinol formation. Pixantrone and mitoxantrone were probed in a validated ex vivo human myocardial strip model that was either doxorubicin-naïve or preliminarily subjected to doxorubicin loading and washouts to mimic doxorubicin treatment and elimination in the clinical setting. In doxorubicin-naïve strips, pixantrone showed higher uptake than mitoxantrone; however, neither drug formed O(2)(.-) or H(2)O(2). In doxorubicin-pretreated strips, neither pixantrone nor mitoxantrone altered the distribution and clearance of residual doxorubicin. Mitoxantrone showed an unchanged uptake and lacked effects on doxorubicin levels, but synergized with doxorubicin to form more O(2)(.-) and H(2)O(2), as evidenced by O(2)(.-)-dependent inactivation of mitochondrial aconitase or mitoxantrone oxidation by H(2)O(2)-activated peroxidases. In contrast, pixantrone uptake was reduced by prior doxorubicin exposure; moreover, pixantrone lacked redox synergism with doxorubicin, and formed an N-dealkylated product that inhibited metabolism of residual doxorubicin to doxorubicinol. Redox inactivity and inhibition of doxorubicinol formation correlate with the cardiac safety of pixantrone in doxorubicin-pretreated patients. Redox inactivity in the face of high cardiac uptake suggests that pixantrone might also be safe in doxorubicin-naïve patients.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Doxorrubicina/análogos & derivados , Corazón/efectos de los fármacos , Isoquinolinas/farmacología , Mitoxantrona/farmacología , Miocardio/metabolismo , Anciano , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/uso terapéutico , Biotransformación , Cromatografía Líquida de Alta Presión , Doxorrubicina/administración & dosificación , Doxorrubicina/efectos adversos , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapéutico , Sinergismo Farmacológico , Femenino , Humanos , Peróxido de Hidrógeno/metabolismo , Técnicas In Vitro , Isoquinolinas/administración & dosificación , Isoquinolinas/farmacocinética , Masculino , Mitoxantrona/administración & dosificación , Mitoxantrona/farmacocinética , Estructura Molecular , Oxidación-Reducción , Superóxidos/metabolismo
13.
Blood ; 117(12): 3302-10, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21239698

RESUMEN

Flavopiridol is a protein bound, cytotoxic, cyclin-dependent kinase inhibitor. Flavopiridol given by 1-hour bolus at 50 mg/m(2) daily 3 times followed by cytosine arabinoside and mitoxantrone (FLAM) is active in adults with poor-risk acute leukemias. A pharmacologically derived "hybrid" schedule (30-minute bolus followed by 4-hour infusion) of flavopiridol was more effective than bolus administration in refractory chronic lymphocytic leukemia. Our phase 1 trial "hybrid FLAM" in 55 adults with relapsed/refractory acute leukemias began at a total flavopiridol dose of 50 mg/m(2) per day 3 times (20-mg/m(2) bolus, 30-mg/m(2) infusion). Dose-limiting toxicity occurred at level 6 (30-mg/m(2) bolus, 70-mg/m(2) infusion) with tumor lysis, hyperbilirubinemia, and mucositis. Death occurred in 5 patients (9%). Complete remission occurred in 22 (40%) across all doses. Overall and disease-free survivals for complete remission patients are more than 60% at more than 2 years. Pharmacokinetics demonstrated a dose-response for total and unbound plasma flavopiridol unrelated to total protein, albumin, peripheral blast count, or toxicity. Pharmacodynamically, flavopiridol inhibited mRNAs of multiple cell cycle regulators, but with uniform increases in bcl-2. "Hybrid FLAM" is active in relapsed/refractory acute leukemias, with a recommended "hybrid" dose of bolus 30 mg/m(2) followed by infusion of 60 mg/m(2) daily for 3 days. This clinical trial is registered at www.clinicaltrials.gov as #NCT00470197.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Citarabina/administración & dosificación , Flavonoides/farmacocinética , Leucemia/tratamiento farmacológico , Leucemia/metabolismo , Mitoxantrona/administración & dosificación , Piperidinas/farmacocinética , Enfermedad Aguda , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Quimioterapia Adyuvante , Citarabina/farmacocinética , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Flavonoides/administración & dosificación , Humanos , Bombas de Infusión , Masculino , Persona de Mediana Edad , Mitoxantrona/farmacocinética , Piperidinas/administración & dosificación , Adulto Joven
14.
J Surg Res ; 175(1): 35-43, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21470623

RESUMEN

BACKGROUND: Magnetic drug targeting is a new treatment principle for tumors using cytostatics coupled to ferromagnetic nanoparticles and extracorporeal magnets. Higher concentrations in tumor tissue with lower systemic concentrations and without damage of healthy organs should be achieved. MATERIALS AND METHODS: n = 42 adult Wag/Rij rats were transfected with rhabdomyosarcoma R(1)H in their right gastrocnemius muscle. In the biodistribution trial (n = 36) concentrations of mitoxantrone-iron oxide with and without an extracorporeal 0.6 tesla magnet and regular mitoxantrone were measured in plasma and tumor tissue for one- and two-dose administration. In the plasma iron trial (n = 6) iron concentrations were measured in plasma before, during, and up to 30 min after drug administration. Seven days after the trial liver, spleen and tumor samples were obtained and histologically assessed. RESULTS: Mitoxantrone iron-oxide concentration in plasma was significantly (P < 0.05) lower when a magnet was placed over the tumor area and as low as uncoupled mitoxantrone. Mitoxantrone concentration in tumor tissue was always significantly higher with magnetic drug targeting when compared with uncoupled mitoxantrone. Two doses resulted in drug accumulation in tumor tissue. Plasma iron concentrations rose when the drug was first administered. Plasma levels fell below the starting level with a magnet applied. A rebound phenomenon with rising iron concentrations was observed after the magnet was removed. Tumors showed fresh necrosis and liver and spleen had detectable iron depositions but no necrosis 7 d after treatment. No allergies or toxic reactions were observed. CONCLUSIONS: We showed that magnetic drug targeting achieves higher concentrations of cytostatics in tumor tissue compared with blood. During magnetic drug targeting, iron particles are quickly sliced and kept in the tumor area. Organs of the reticuloendothelial system are not affected by cytostatic damage.


Asunto(s)
Antineoplásicos/farmacocinética , Compuestos Férricos/farmacocinética , Nanopartículas de Magnetita , Mitoxantrona/farmacocinética , Rabdomiosarcoma/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Compuestos Férricos/administración & dosificación , Hígado/fisiopatología , Mitoxantrona/administración & dosificación , Plasma/fisiología , Ratas , Rabdomiosarcoma/fisiopatología , Bazo/fisiopatología
15.
Pharm Res ; 29(9): 2407-18, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22555380

RESUMEN

PURPOSE: To investigate the effect of surface functionalization of mesoporous silica nanoparticles (MSN) on crystallization, loading, release and activity of mitoxantrone (MTX). METHODS: Thiol-, amine-, and mixed thiol/amine-functionalized MSN were synthesized and characterized by electron microscopy, thermogravimetry, surface area analysis, elemental analysis and zeta potential. MTX loading and release kinetics were determined in phosphate and acetate buffers (pH 7.4 and 4.5). The crystalline state of MTX in MSN was determined by differential scanning calorimetry and X-ray diffraction. Cytotoxicity and activity of MTX loaded MSN were determined by MTS assay in MDA-MB-231 cells. RESULTS: Our results demonstrate that loading of MTX depends strongly on the type of surface functional groups in MSN. Thiol-MSN showed the highest MTX loading (18 % w/w) when compared with thiol/amine-MSN (6 % w/w) and amine-MSN (1 % w/w). MTX release was strongly dependent on the pH of the release medium and the type of surface functional group. MTX was found in the amorphous form when loaded in thiol-functionalized MSN. No significant effect of surface modification of MSN on particle toxicity was observed. MTX loaded in MSN exhibited comparable anticancer activity in vitro as free MTX. CONCLUSION: Surface modifications of MSN have significant effect on MTX crystallization and release behavior.


Asunto(s)
Antineoplásicos/farmacocinética , Mitoxantrona/farmacocinética , Nanopartículas , Dióxido de Silicio , Rastreo Diferencial de Calorimetría , Línea Celular Tumoral , Humanos , Concentración de Iones de Hidrógeno , Microscopía Electrónica de Transmisión , Propiedades de Superficie
16.
Pharm Res ; 29(7): 1949-59, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22399388

RESUMEN

PURPOSE: To test targeted liposomes in an effort to improve drug transport across cellular barriers into the brain. METHODS: Therefore we prepared Mitoxantrone (MTO) entrapping, rigid and fluid liposomes, equipped with a 19-mer angiopeptide as ligand for LDL lipoprotein receptor related protein (LRP) targeting. RESULTS: Fluid, ligand bearing liposomes showed in vitro the highest cellular uptake and transcytosis and were significantly better than the corresponding ligand-free liposomes and rigid, ligand-bearing vesicles. Treatment of mice, transplanted with human breast cancer cells subcutaneously and into the brain, with fluid membrane liposomes resulted in a significant reduction in the tumor volume by more than 80% and in a clear reduction in drug toxicity. The improvement was mainly depended on liposome fluidity while the targeting contributed only to a minor degree. Pharmacokinetic parameters were also improved for liposomal MTO formulations in comparison to the free drug. So the area under the curve was increased and t(1/2) was extended for liposomes. CONCLUSION: Our data show that it is possible to significantly improve the therapy of brain metastases if MTO-encapsulating, fluid membrane liposomes are used instead of free MTO. This effect could be further enhanced by fluid, ligand bearing liposomes.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/secundario , Encéfalo/efectos de los fármacos , Proteínas Relacionadas con Receptor de LDL/metabolismo , Liposomas/metabolismo , Mitoxantrona/uso terapéutico , Secuencia de Aminoácidos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Liposomas/química , Ratones , Mitoxantrona/administración & dosificación , Mitoxantrona/farmacocinética , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo
17.
Future Oncol ; 8(7): 829-37, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22830403

RESUMEN

Electrochemotherapy is a tumor treatment that adapts the systemic or local delivery of anticancer drugs by the application of permeabilizing electric pulses with appropriate amplitude and waveforms. This allows the use of lipophobic drugs, which frequently have a narrow therapeutic index, with a decreased morbidity for the patient, while maintaining appropriate anticancer efficacy. Electrochemotherapy is used in humans for the treatment of cutaneous neoplasms or the palliation of skin tumor metastases, and a standard operating procedure has been devised. In veterinary oncology, the electrochemotherapy approach is gaining popularity, becoming a first-line treatment in consideration of its high efficacy and low toxicity. This review summarizes the state of the art in veterinary oncology as a preclinical model.


Asunto(s)
Antineoplásicos/administración & dosificación , Electroquimioterapia/métodos , Neoplasias/veterinaria , Animales , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica , Bleomicina/farmacocinética , Bleomicina/farmacología , Carcinoma/tratamiento farmacológico , Enfermedades de los Gatos/tratamiento farmacológico , Gatos , Cisplatino/farmacocinética , Cisplatino/farmacología , Enfermedades de los Perros/tratamiento farmacológico , Perros , Evaluación Preclínica de Medicamentos , Electroquimioterapia/veterinaria , Epirrubicina , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Melanoma/veterinaria , Mitoxantrona/farmacocinética , Mitoxantrona/farmacología , Neoplasias/tratamiento farmacológico , Quinazolinas , Sarcoma/tratamiento farmacológico , Tiofenos
18.
Pharm Res ; 28(5): 1090-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21279423

RESUMEN

PURPOSE: The objective of our study was to investigate the effect of 5,7-DMF on the accumulation of mitoxantrone (MX) in BCRP-expressing normal cells and to investigate its impact on the PK and tissue distribution of MX in mice. METHODS: The in vitro effect of 5,7-DMF on MX accumulation was examined in MDCK cells transfected with BCRP. The pharmacokinetic and tissue distribution of mitoxantrone, with and without co-administration of 5,7-DMF or multiple flavonoid combinations, were determined in mice. RESULTS: In the presence of 2.5 µM or 25 µM of 5,7-DMF, the intracellular concentration of MX was significantly increased in MDCK/Bcrp1 and MDCK/BCRP cells, but not in MDCK/Mock cells. The AUC values of MX in several tissues were significantly increased when MX was co-administered with 5,7-DMF. The most substantial elevations of MX AUC in the presence of 5,7-DMF occurred in the liver (94.5%) and kidneys (61.9%), which is in apparent agreement with the relatively high levels of mouse Bcrp1 expression in these two tissues. CONCLUSIONS: Bcrp1-mediated DMF-MX interactions occur both in vitro and in vivo. 5,7-DMF represents a novel and very promising chemosensitizing agent for the BCRP-mediated MDR due to its low toxicity and potent BCRP inhibition.


Asunto(s)
Antineoplásicos/farmacocinética , Flavonoides/farmacología , Mitoxantrona/farmacocinética , Proteínas de Neoplasias/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Perros , Interacciones Farmacológicas , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Proteínas de Neoplasias/genética , Neoplasias/tratamiento farmacológico
19.
Eur Rev Med Pharmacol Sci ; 25(24): 7806-7822, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34982442

RESUMEN

OBJECTIVE: Mitoxantrone (MTX)- induced cardiotoxicity is a clinical concern that is limiting its use. The aim of this paper, therefore, was to investigate the subchronic administration of MTX plus nonspecific/specific inhibitors of CYP450/2E1, to assess the extent of oxidative-induced injury by measuring levels of oxidative cardiac and injury biomarkers in mice and to evaluate the effects of CYP2E1 on caspase 3 activity and nuclear factor erythroid 2-related factor-2 (NRF-2). MATERIALS AND METHODS: Mice (n = 32) were divided into four treatment groups of eight: control, MTX, MTX + 4-methlypyrazole (4MP) and MTX + disulfiram (Disf). After 6 weeks of treatments, blood and heart samples were collected. RESULTS: Liquid chromatography-mass spectrometry (LCMS) analysis of MTX-treated plasma samples revealed several metabolites with different retention times. Cardiac antioxidant enzymes and creatine kinase (CK) levels were not significantly different among the groups. However, cardiac troponin and caspase 3 activity were significantly raised, with increased CYP2E1 expressions and reduced NRF-2 expression. Tissue damage was observed in all the treatment groups, including MTX, leading to the conclusion that MTX-induced cardiotoxicity was mediated by CYP2E1 activity, which initiated caspase 3 production, and decreased NRF-2 expression. CONCLUSIONS: Therefore, agents that inhibit CPY2E1 expression might attenuate MTX-induced cardiotoxicity by increasing NRF-2 expression.


Asunto(s)
Antineoplásicos/toxicidad , Cardiotoxicidad/tratamiento farmacológico , Inhibidores del Citocromo P-450 CYP2E1/uso terapéutico , Disulfiram/uso terapéutico , Fomepizol/uso terapéutico , Mitoxantrona/toxicidad , Animales , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Cardiotoxicidad/sangre , Cardiotoxicidad/metabolismo , Cardiotoxicidad/patología , Caspasa 3/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Inhibidores del Citocromo P-450 CYP2E1/farmacología , Disulfiram/farmacología , Femenino , Fomepizol/farmacología , Masculino , Ratones Endogámicos BALB C , Mitoxantrona/sangre , Mitoxantrona/farmacocinética , Miocardio/metabolismo , Miocardio/patología , Factor 2 Relacionado con NF-E2/metabolismo , Troponina I/metabolismo
20.
J Pharmacol Exp Ther ; 333(3): 788-96, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20304939

RESUMEN

A synergistic effect of P-glycoprotein (P-gp)/Abcb1a and breast cancer resistance protein (Bcrp)/Abcg2 was reported to limit the brain penetration of their common substrates. This study investigated this based on pharmacokinetics using Mdr1a/1b(-/-), Bcrp(-/-), and Mdr1a/1b(-/-)/Bcrp(-/-) mice. Comparison of the brain- and testis-to-plasma ratios (C(brain)/C(plasma) and C(testis)/C(plasma), respectively) of the reference compounds quinidine and dantrolene for P-gp and Bcrp, respectively, indicates that impairment of either P-gp and Bcrp did not cause any change in the efflux activities of Bcrp or P-gp, respectively, at both the blood-brain barrier (BBB) and blood-testis barrier (BTB). The C(brain)/C(plasma) and C(testis)/C(plasma) of the common substrates erlotinib, flavopiridol, and mitoxantrone were markedly increased in Mdr1a/1b(-/-)/Bcrp(-/-) mice even compared with Mdr1a/1b(-/-) and Bcrp(-/-) mice. Efflux activities by P-gp and Bcrp relative to passive diffusion at the BBB and BTB were separately evaluated based on the C(brain)/C(plasma) and C(testis)/C(plasma) in the knockout strains to the wild-type strain. P-gp made a larger contribution than Bcrp to the net efflux of the common substrates, but Bcrp activities were also significantly larger than passive diffusion. These parameters could reasonably account for the marked increase in C(brain)/C(plasma) and C(testis)/C(plasma) in the Mdr1a/1b(-/-)/Bcrp(-/-) mice. In conclusion, the synergistic effect of P-gp and Bcrp on C(brain)/C(plasma) and C(testis)/C(plasma) can be explained by their contribution to the net efflux at the BBB and BTB without any interaction between P-gp and Bcrp.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacocinética , Encéfalo/metabolismo , Flavonoides/farmacocinética , Mitoxantrona/farmacocinética , Proteínas de Neoplasias/metabolismo , Piperidinas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Quinazolinas/farmacocinética , Testículo/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Algoritmos , Animales , Antimaláricos/farmacocinética , Barrera Hematoencefálica/efectos de los fármacos , Dantroleno/farmacocinética , Clorhidrato de Erlotinib , Cinética , Masculino , Ratones , Ratones Noqueados , Relajantes Musculares Centrales/farmacocinética , Proteínas de Neoplasias/genética , Quinidina/farmacocinética , Distribución Tisular , Xenobióticos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA