Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biomed Res Int ; 2020: 1520651, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32149078

RESUMEN

Acute T lymphocytic leukemia (T-ALL) is an aggressive hematologic resulting from the malignant transformation of T-cell progenitors. Drug resistance and relapse are major difficulties in the treatment of T-ALL. Here, we report the antitumor potency of NL-101, a compound that combines the nitrogen mustard group of bendamustine with the hydroxamic acid group of vorinostat. We found NL-101 exhibited efficient antiproliferative activity in T-ALL cell lines (IC50 1.59-1.89 µM), accompanied by cell cycle arrest and apoptosis, as evidenced by the increased expression of Cyclin E1, CDK2, and CDK4 proteins and cleavage of PARP. In addition, this bendamustine-derived drug showed both a HDACi effect as demonstrated by histone hyperacetylation and p21 transcription and a DNA-damaging effect as shown by an increase in γ-H2AX. Intriguingly, we found that NL-101-induced autophagy in T-ALL cells through inhibiting Akt-mTOR signaling pathway, as indicated by an increase in LC3-I to LC3-II conversion and decrease of p62. Furthermore, inhibition of autophagy by 3-methyladenine increased apoptotic cell death by NL-101, suggesting a prosurvival role of autophagy. In summary, our finding provides rationale for investigation of NL-101 as a DNA/HDAC dual targeting drug in T-ALL, either as a single agent or in combination with autophagy inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Clorhidrato de Bendamustina , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Apoptosis/efectos de los fármacos , Clorhidrato de Bendamustina/análogos & derivados , Clorhidrato de Bendamustina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos
2.
J Pharm Biomed Anal ; 174: 168-174, 2019 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-31170630

RESUMEN

A selective, sensitive and rapid mice dried blood spot (DBS) method has been developed and validated for the simultaneous quantification of bendamustine (BM) and γ-hydroxy-bendamustine (HBM) as per regulatory guidelines using an LC-MS/MS. Quality control, calibration curve and study sample DBS cards were sonicated with 5% formic acid in water before extraction with ethyl acetate enriched with internal standard (I.S.). The organic layer was evaporated and residue was reconstituted in 0.1% formic acid in acetonitrile for LC-MS/MS analysis. Chromatographic resolution of both analytes (BM and HBM) and the I.S. (loperamide) was achieved on an Atlantis dC18 column using 0.2% formic acid:acetonitrile (25:75, v/v) as an eluant delivered at a constant flow-rate of 0.5 mL/min. The total chromatographic run time was 3.2 min. The MS/MS ion transitions monitored were m/z 358.0 → 228.0, 374.0 → 338.0 and 477.0 → 210.0 for BM, HBM and the I.S, respectively. The assay was linear in the range of 5.65-2544 ng/mL for both BM and HBM. The within-run and between-run accuracy and within-run and between-run precision were in the range of 0.96-1.00 and 1.36-9.94%, respectively for BM; 0.88-1.03 and 4.57-11.7%, respectively for HBM on mice DBS cards. Stability studies showed that both analytes were stable at room temperature for 7 days and at -80 °C for 55 days on DBS cards. The validated DBS method has been applied to a pharmacokinetic study in mice.


Asunto(s)
Clorhidrato de Bendamustina/análogos & derivados , Clorhidrato de Bendamustina/sangre , Clorhidrato de Bendamustina/farmacocinética , Pruebas con Sangre Seca/métodos , Animales , Calibración , Cromatografía Líquida de Alta Presión , Estabilidad de Medicamentos , Hematócrito , Humanos , Límite de Detección , Modelos Lineales , Masculino , Ratones , Ratones Endogámicos BALB C , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem , Temperatura
3.
Drug Res (Stuttg) ; 67(9): 497-508, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28561238

RESUMEN

A highly sensitive, specific and rapid LC-ESI-MS/MS method has been developed and validated for the simultaneous quantification of bendamustine (BM) and γ-hydroxybendamustine (HBM) in small volume (20 µL) mice and dog plasma using phenacetin as an internal standard (IS) as per regulatory guidelines. Both the analytes and IS were extracted from mice and dog plasma using a liquid-liquid extraction method. Chromatography was achieved on Atlantis dC18 column using an isocratic mobile phase (0.2% formic acid:acetonitrile, 25:75) at a flow rate of 0.40 mL/min. The total chromatographic run time was 3.0 min and the elution of BM, HBM and IS occurred at ~1.2, 1.2 and 2.0 min, respectively. A linear response function was established 0.11-518 ng/mL for both the analytes in mice and dog plasma. The intra- and inter-day accuracy and precisions were in the range of 3.46-12.9 and 3.63-8.23%; 1.15-9.00 and 7.86-9.49% for BM and HBM, respectively in mice plasma and 2.15-6.49 and 1.73-13.1%; 4.35-13.9 and 4.33-10.5% for BM and HBM, respectively in dog plasma. This novel method has been applied to a pharmacokinetic study in mice and dogs.


Asunto(s)
Antineoplásicos Alquilantes/sangre , Clorhidrato de Bendamustina/análogos & derivados , Cromatografía Liquida/métodos , Espectrometría de Masas en Tándem/métodos , Animales , Clorhidrato de Bendamustina/sangre , Perros , Extracción Líquido-Líquido , Masculino , Ratones , Ratones Endogámicos BALB C , Reproducibilidad de los Resultados
4.
J Hematol Oncol ; 10(1): 127, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28633670

RESUMEN

BACKGROUND: Despite recent advances in the treatment of multiple myeloma (MM), the prognosis of most patients remains poor, and resistance to traditional and new drugs frequently occurs. EDO-S101 is a novel therapeutic agent conceived as the fusion of a histone deacetylase inhibitor radical to bendamustine, with the aim of potentiating its alkylating activity. METHODS: The efficacy of EDO-S101 was evaluated in vitro, ex vivo and in vivo, alone, and in combination with standard anti-myeloma agents. The underlying mechanisms of action were also evaluated on MM cell lines, patient samples, and different murine models. RESULTS: EDO-S101 displayed potent activity in vitro in MM cell lines (IC50 1.6-4.8 µM) and ex vivo in cells isolated from MM patients, which was higher than that of bendamustine and independent of the p53 status and previous melphalan resistance. This activity was confirmed in vivo, in a CB17-SCID murine plasmacytoma model and in de novo Vk*MYC mice, leading to a significant survival improvement in both models. In addition, EDO-S101 was the only drug with single-agent activity in the multidrug resistant Vk12653 murine model. Attending to its mechanism of action, the molecule showed both, a HDACi effect (demonstrated by α-tubulin and histone hyperacetylation) and a DNA-damaging effect (shown by an increase in γH2AX); the latter being again clearly more potent than that of bendamustine. Using a reporter plasmid integrated into the genome of some MM cell lines, we demonstrate that, apart from inducing a potent DNA damage, EDO-S101 specifically inhibited the double strand break repair by the homologous recombination pathway. Moreover, EDO-S101 treatment reduced the recruitment of repair proteins such as RAD51 to DNA-damage sites identified as γH2AX foci. Finally, EDO-S101 preclinically synergized with bortezomib, both in vitro and in vivo. CONCLUSION: These findings provide rationale for the clinical investigation of EDO-S101 in MM, either as a single agent or in combination with other anti-MM drugs, particularly proteasome inhibitors.


Asunto(s)
Antineoplásicos/uso terapéutico , Bencimidazoles/uso terapéutico , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Clorhidrato de Bendamustina/análogos & derivados , Clorhidrato de Bendamustina/farmacología , Clorhidrato de Bendamustina/uso terapéutico , Bencimidazoles/química , Bencimidazoles/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones SCID , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología
5.
PLoS One ; 12(1): e0170709, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28125678

RESUMEN

Bendamustine (BENDA), which bears the bis(2-chloroethyl)amino moiety, is an alkylating agent that stops the growth of cancer cells by binding to DNA and interfering with its replication. However, the mechanism of action underlying its excellent clinical efficacy remains unclear. In this work, we report that BENDA inhibits signal transducer and activator of transcription 3 (STAT3). In an AlphaScreen-based biochemical assay using recombinant human STAT3, binding of STAT3-Src homology 2 (SH2) to the phosphotyrosine (pTyr, pY) peptide was inhibited by BENDA but not by the inactive metabolite dihydroxy bendamustine (HP2). When a single point mutation of C550A or C712A was introduced into recombinant human STAT3, its sensitivity to BENDA was substantially reduced, suggesting that these cysteine residues are important for BENDA to inhibit STAT3. Furthermore, BENDA suppressed the function of cellular STAT3 as a transcriptional activator in a human breast cancer cell line, MDA-MB-468, with constitutively activated STAT3. A competitive pull-down assay using biotinylated BENDA (Bio-BENDA) revealed that BENDA bound tightly to cellular STAT3, presumably through covalent bonds. Therefore, our results suggest that the anticancer effects of BENDA may be associated, at least in part, with its inhibitory effect on the SH2 domain of STAT3.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Clorhidrato de Bendamustina/farmacología , Cisteína/química , Fosfotirosina/química , Mutación Puntual , Factor de Transcripción STAT3/antagonistas & inhibidores , Alanina/química , Alanina/metabolismo , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/metabolismo , Clorhidrato de Bendamustina/análogos & derivados , Clorhidrato de Bendamustina/metabolismo , Sitios de Unión , Línea Celular Tumoral , Cisteína/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Expresión Génica , Humanos , Péptidos/antagonistas & inhibidores , Péptidos/síntesis química , Péptidos/metabolismo , Fosfotirosina/metabolismo , Unión Proteica , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Relación Estructura-Actividad , Dominios Homologos src
6.
Drug Res (Stuttg) ; 66(7): 351-6, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27120083

RESUMEN

Bendamustine is an alkylating agent administered as 1 h intravenous infusion in the clinic for the treatment of malignant haematological cancers. The aim of the study was to evaluate the pharmacokinetics of bendamustine and its key cytochrome P 450 (CYP) 1A2 mediated γ-hydroxybendamustine (M3) metabolite after 30- and 60-min intravenous infusion of bendamustine in rats. 2 groups were assigned to receive bendamustine either as 30- or 60-min infusion and doses were normalized to 15 mg/kg for the sake of statistical evaluation. Serial pharmacokinetic samples were collected and were analysed for the circulatory levels of bendamustine and its M3 metabolite. Standard pharmacokinetic parameters were generated for bendamustine and its M3 metabolite. Regardless of the intravenous regimens, Cmax coincided with end of infusion for both bendamustine and its M3 metabolite. Immediately after stoppage of infusion, a rapid decline in the plasma levels occurred for both bendamustine and M3 metabolite. The Cmax and AUC0-∞ parameters for bendamustine after 60-min infusion were 1.90 and 1.34-fold higher; while CL was lower by 1.32-fold as compared to the 30-min infusion. In contrast, the Cmax and AUC0-∞ after 30-min infusion for the M3 metabolite was 2.15- and 2.78-fold greater; while CL was 2.32-fold lower when compared to the 60-min infusion. However, T1/2 and Vz values were similar between the 2 intravenous treatments for bendamustine or the M3 metabolite. The data unequivocally confirmed the existence of differential pharmacokinetics of bendamustine and its M3 metabolite as the function of the duration of intravenous infusion.


Asunto(s)
Clorhidrato de Bendamustina/análogos & derivados , Clorhidrato de Bendamustina/administración & dosificación , Clorhidrato de Bendamustina/farmacocinética , Animales , Clorhidrato de Bendamustina/sangre , Clorhidrato de Bendamustina/metabolismo , Infusiones Intravenosas , Masculino , Ratas , Factores de Tiempo
7.
PLoS One ; 10(7): e0133743, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26196503

RESUMEN

The alkylating agent bendamustine is approved for the treatment of hematopoietic malignancies such as non-Hodgkin lymphoma, chronic lymphocytic leukemia and multiple myeloma. As preliminary data on recently disclosed bendamustine esters suggested increased cytotoxicity, we investigated representative derivatives in more detail. Especially basic esters, which are positively charged under physiological conditions, were in the crystal violet and the MTT assay up to approximately 100 times more effective than bendamustine, paralleled by a higher fraction of early apoptotic cancer cells and increased expression of p53. Analytical studies performed with bendamustine and representative esters revealed pronounced cellular accumulation of the derivatives compared to the parent compound. In particular, the pyrrolidinoethyl ester showed a high enrichment in tumor cells and inhibition of OCT1- and OCT3-mediated transport processes, suggesting organic cation transporters to be involved. However, this hypothesis was not supported by the differential expression of OCT1 (SLC22A1) and OCT3 (SLC22A3), comparing a panel of human cancer cells. Bendamustine esters proved to be considerably more potent cytotoxic agents than the parent compound against a broad panel of human cancer cell types, including hematologic and solid malignancies (e.g. malignant melanoma, colorectal carcinoma and lung cancer), which are resistant to bendamustine. Interestingly, spontaneously immortalized human keratinocytes, as a model of "normal" cells, were by far less sensitive than tumor cells against the most potent bendamustine esters.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Clorhidrato de Bendamustina/análogos & derivados , Carcinoma/metabolismo , Sarcoma/metabolismo , Clorhidrato de Bendamustina/farmacología , Línea Celular Tumoral , Ésteres/química , Células HEK293 , Humanos , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 1 de Catión Orgánico/metabolismo
8.
Cancer Chemother Pharmacol ; 75(6): 1143-54, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25829094

RESUMEN

PURPOSE: Bendamustine is a unique alkylating agent indicated for the treatment of chronic lymphocytic leukemia and rituximab-refractory, indolent B cell non-Hodgkin's lymphoma. Despite the extensive experience with bendamustine, its pharmacokinetic profile has only recently been described. This overview summarizes the pharmacokinetics, pharmacokinetic/pharmacodynamic relationships, and drug-drug interactions of bendamustine in adult and pediatric patients with hematologic malignancies. METHODS: A literature search and data on file (including a human mass balance study, pharmacokinetic population analyses in adult and pediatric patients, and modeling analyses) were evaluated for inclusion. RESULTS: Bendamustine concentrations peak at end of intravenous infusion (~1 h). Subsequent elimination is triphasic, with the intermediate t 1/2 (~40 min) as the effective t 1/2 since the final phase represents <1 % of the area under the curve. Bendamustine is rapidly hydrolyzed to monohydroxy-bendamustine and dihydroxy-bendamustine, which have little or no activity. Cytochrome P450 (CYP) 1A2 oxidation yields the active metabolites γ-hydroxybendamustine and N-desmethyl-bendamustine, at low concentrations, which contribute minimally to cytotoxicity. Minor involvement of CYP1A2 in bendamustine elimination suggests a low likelihood of drug-drug interactions with CYP1A2 inhibitors. Systemic exposure to bendamustine 120 mg/m(2) is comparable between adult and pediatric patients; age, race, and sex have been shown to have no significant effect on systemic exposure in either population. The effect of hepatic/renal impairment on bendamustine pharmacokinetics remains to be elucidated. Higher bendamustine concentrations may be associated with increased probability of nausea or infection. No clear exposure-efficacy response relationship has been observed. CONCLUSIONS: Altogether, the findings support dosing based on body surface area for most patient populations.


Asunto(s)
Clorhidrato de Bendamustina/análogos & derivados , Neoplasias Hematológicas/tratamiento farmacológico , Compuestos de Mostaza Nitrogenada/farmacocinética , Compuestos de Mostaza Nitrogenada/uso terapéutico , Área Bajo la Curva , Clorhidrato de Bendamustina/farmacocinética , Clorhidrato de Bendamustina/uso terapéutico , Citocromo P-450 CYP1A2/metabolismo , Interacciones Farmacológicas/fisiología , Neoplasias Hematológicas/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA