Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Clin Pharmacol ; 88(4): 1722-1734, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34519068

RESUMEN

AIMS: The aim of this work is the development of a mechanistic physiologically-based pharmacokinetic (PBPK) model using in vitro to in vivo extrapolation to conduct a drug-drug interaction (DDI) assessment of treosulfan against two cytochrome p450 (CYP) isoenzymes and P-glycoprotein (P-gp) substrates. METHODS: A PBPK model for treosulfan was developed de novo based on literature and unpublished clinical data. The PBPK DDI analysis was conducted using the U.S. Food and Drug Administration (FDA) DDI index drugs (probe substrates) midazolam, omeprazole and digoxin for CYP3A4, CYP2C19 and P-gp, respectively. Qualified and documented PBPK models of the probe substrates have been adopted from an open-source online model database. RESULTS: The PBPK model for treosulfan, based on both in vitro and in vivo data, was able to predict the plasma concentration-time profiles and exposure levels of treosulfan applied for a standard conditioning treatment. Medium and low potentials for DDI on CYP3A4 (maximum area under the concentration-time curve ratio (AUCRmax = 2.23) and CYP2C19 (AUCRmax = 1.6) were predicted, respectively, using probe substrates midazolam and omeprazole. Treosulfan was not predicted to cause a DDI on P-gp. CONCLUSION: Medicinal products with a narrow therapeutic index (eg, digoxin) that are substrates for CYP3A4, CYP2C19 or P-gp should not be given during treatment with treosulfan. However, considering the comprehensive treosulfan-based conditioning treatment schedule and the respective pharmacokinetic properties of the concomitantly used drugs (eg, half-life), the potential for interaction on all evaluated mechanisms would be low (AUCR < 1.25), if concomitantly administered drugs are dosed either 2 hours before or 8 hours after the 2-hour intravenous infusion of treosulfan.


Asunto(s)
Citocromo P-450 CYP3A , Midazolam , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Busulfano/análogos & derivados , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP3A/metabolismo , Digoxina , Interacciones Farmacológicas , Humanos , Midazolam/farmacocinética , Modelos Biológicos , Omeprazol , Preparaciones Farmacéuticas
2.
Biol Blood Marrow Transplant ; 24(8): 1651-1656, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29753157

RESUMEN

This multicenter study evaluated a treosulfan-based regimen in children and young adults with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) undergoing allogeneic hematopoietic cell transplant (HCT). Forty patients with median age 11 years (range, 1 to 19) underwent allogeneic HCT for AML in first (n = 18), second (n = 11), and third or greater remission (n = 3) or MDS (n = 8) using bone marrow (n = 25), peripheral blood stem cells (n = 5), or cord blood (n = 9). The regimen consisted of body surface area (BSA)-based treosulfan 10 g/m2/day (BSA ≤ .5 m2), 12 g/m2/day (BSA > .5 to 1.0 m2), or 14 g/m2/day (BSA > 1.0 m2) on days -6 to -4; fludarabine 30 mg/m2/day on days -6 to -2; and a single fraction of 200 cGy total body irradiation on day -1. Graft-versus-host disease (GVHD) prophylaxis included tacrolimus and methotrexate for marrow and peripheral blood stem cell and cyclosporine/mycophenolate mofetil for cord blood. One-year overall survival, disease-free survival, and nonrelapse mortality were 80%, 73%, and 3%, respectively. One-year relapse was 38% for AML and 13% for MDS. No serious organ toxicities were observed. All 37 assessable patients engrafted. Cumulative incidences of grades II to IV acute GVHD and chronic GVHD were 22% and 40%, respectively. BSA-based treosulfan dosing resulted in predictable area under the curve and maximum concentration, which is required for dosing without measuring individual pharmacokinetic parameters. Observed differences in pharmacokinetics did not impact disease control or regimen toxicity. This BSA-based treosulfan regimen resulted in excellent engraftment and disease-free survival and minimal toxicity and transplant-related mortality (3%) in children and young adults with AML and MDS.


Asunto(s)
Busulfano/análogos & derivados , Trasplante de Células Madre Hematopoyéticas/métodos , Leucemia Mieloide Aguda/terapia , Síndromes Mielodisplásicos/terapia , Irradiación Corporal Total , Adolescente , Busulfano/administración & dosificación , Busulfano/uso terapéutico , Niño , Preescolar , Femenino , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/mortalidad , Humanos , Lactante , Leucemia Mieloide Aguda/mortalidad , Masculino , Síndromes Mielodisplásicos/mortalidad , Análisis de Supervivencia , Trasplante Homólogo , Vidarabina/administración & dosificación , Vidarabina/análogos & derivados , Vidarabina/uso terapéutico , Adulto Joven
3.
Ther Drug Monit ; 39(6): 617-624, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28937535

RESUMEN

BACKGROUND: The value of therapeutic drug monitoring (TDM) for paclitaxel (PTX) was recently demonstrated in the largest TDM trial ever conducted in oncology. The trial demonstrated significant reduction in neuropathy when using TDM. Dose adjustment for PTX was based on time above a threshold concentration (Tc>0.05). Tc>0.05 must be calculated with a pharmacokinetic model and complex nonlinear mixed-effects software. The use of the software and chromatographic methods to measure PTX requires specialized expertise. User-friendly methods to quantitate PTX and calculate Tc>0.05 could simplify the introduction of TDM into routine clinical practice. METHODS: The immunoassay (MyPaclitaxel) was used to quantitate PTX in samples from the clinical trial; the results were used to calculate Tc>0.05 using a stand-alone computer program with a simple, friendly graphical user interface for nonlinear mixed-effects pharmacokinetic calculations (MyCare Drug Exposure Calculator). The resulting dose recommendations from the calculated Tc>0.05 were compared with those using liquid chromatography-ultraviolet detection and NONMEM to examine the efficacy of the simpler tools for TDM. RESULTS: There was a good agreement between the immunoassay and liquid chromatography-ultraviolet detection: Passing-Bablok regression slope was 1.045 and intercept was -6.00, R was 0.9757, and mean bias was -1.77 ng/mL (-2.07 nmol/L). Dosing recommendations were identical for 70% of the cycles and within 10% for 89% of the samples. All Tc>0.05 values were at the same or adjacent medical decision points. CONCLUSIONS: MyPaclitaxel assay and MyCare Drug Exposure Calculator are convenient, user-friendly tools that may be suitable for routine TDM of PTX in clinical care.


Asunto(s)
Antineoplásicos Fitogénicos/sangre , Antineoplásicos Fitogénicos/farmacocinética , Técnicas de Apoyo para la Decisión , Monitoreo de Drogas/métodos , Paclitaxel/sangre , Paclitaxel/farmacocinética , Humanos , Inmunoensayo/métodos , Reproducibilidad de los Resultados , Programas Informáticos
4.
Graefes Arch Clin Exp Ophthalmol ; 251(1): 279-84, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22782301

RESUMEN

OBJECTIVE: In conjunctival melanoma, local chemotherapy has been based so far on clinical evidence and limited to the therapy of melanoma in situ. Our aim was to define substances that may have the potential to add to therapeutic options in extended local growth and metastatic disease. Two conjunctival cell lines (CRMM-1 and CRMM-2) have been established from recurrent conjunctival melanoma. In this study, we examined the chemosensitivity of these cell lines to different cytotoxic substances. MATERIALS AND METHODS: The cell lines CRMM-1 and CRMM-2 were exposed to chemotherapeutics for 24 h and the IC50 was generated. Sulforhodamin-B assays were used for quantification of in vitro efficacy. Time of exposure and escalating concentrations of the substances were adapted to the experimental setting. RESULTS: Bortezomib, clusianone 502 (nemorosone), ranpirnase, and sorafenib were efficient in inhibiting the growth of conjunctival melanoma cell lines. The IC50 achieved concentrations below or around 10 µM for these substances. CONCLUSIONS: Bortezomib, clusianone 502, ranpirnase, and sorafenib inhibited growth in conjunctival melanoma cell lines efficiently. The new substances may be a suitable alternative for local therapy. New therapeutic options with highly specific targeted agents for metastatic disease have to be evaluated in further experiments.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Conjuntiva/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Benzofenonas/uso terapéutico , Ácidos Borónicos/uso terapéutico , Bortezomib , Proliferación Celular/efectos de los fármacos , Neoplasias de la Conjuntiva/patología , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Melanoma/patología , Niacinamida/análogos & derivados , Niacinamida/uso terapéutico , Compuestos de Fenilurea/uso terapéutico , Pirazinas/uso terapéutico , Ribonucleasas/uso terapéutico , Sorafenib , Células Tumorales Cultivadas
5.
Drug Metab Pharmacokinet ; 52: 100515, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37481830

RESUMEN

Intravenously infused treosulfan was evaluated in adult and pediatric patients for conditioning regimen prior to allogeneic hematopoietic stem cell transplantation. A population pharmacokinetic (PK) model was initially developed on 116 adult and pediatric PK profiles from historical trials, to support treosulfan dose recommendations for children in 2 prospective trials. The aim was to assess and update the initial population PK model by inclusion of additional 83 pediatric PK profiles from these 2 trials. The final population PK model was 2-compartmental with dosing in the central compartment, linear elimination, and inter-compartmental clearance. Inter-individual variability was included on clearance (CL), central volume (V1), peripheral volume (V2), and inter-compartmental clearance (Q). The final model described an effect of the body surface area (BSA) on CL, V1, V2, and Q. The final model resulted in a modified dose recommendation for children and advises treosulfan doses of 10 g/m2, 12 g/m2, and 14 g/m2 for BSAs of <0.4 m2, ≥0.4 to <0.9 m2, and ≥0.9 m2, respectively. This simplified BSA-dependent dose recommendation was developed for children, ensuring a well comparable treosulfan exposure as a dose of 14 g/m2 in adults - irrespective of their age and without applying individual therapeutic drug monitoring.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Acondicionamiento Pretrasplante , Adulto , Humanos , Niño , Estudios Prospectivos , Acondicionamiento Pretrasplante/métodos , Busulfano/farmacocinética , Busulfano/uso terapéutico
6.
Biol Blood Marrow Transplant ; 17(3): 341-50, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20685259

RESUMEN

In this prospective study 60 patients of median age 46 (range: 5-60 years), with acute myelogenous leukemia (AML; n = 44), acute lymphoblastic leukemia (ALL; n = 3), or myelodysplastic syndrome (MDS; n = 13) were conditioned for allogeneic hematopoietic cell transplantation with a treosulfan/fludarabine (Flu) combination. Most patients were considered at high risk for relapse or nonrelapse mortality (NRM). Patients received intravenous treosulfan, 12 g/m(2)/day (n = 5) or 14 g/m(2)/day (n = 55) on days -6 to -4, and Flu (30 mg/m(2)/day) on days -6 to -2, followed by infusion of marrow (n = 7) or peripheral blood stem cells (n = 53) from HLA-identical siblings (n = 30) or unrelated donors (n = 30). All patients engrafted. NRM was 5% at day 100, and 8% at 2 years. With a median follow-up of 22 months, the 2-year relapse-free survival (RFS) for all patients was 58% and 88% for patients without high-risk cytogenetics. The 2-year cumulative incidence of relapse was 33% (15% for patients with MDS, 34% for AML in first remission, 50% for AML or ALL beyond first remission and 63% for AML in refractory relapse). Thus, a treosulfan/Flu regimen was well tolerated and yielded encouraging survival and disease control with minimal NRM. Further trials are warranted to compare treosulfan/Flu to other widely used regimens, and to study the impact of using this regimen in more narrowly defined groups of patients.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Busulfano/análogos & derivados , Neoplasias Hematológicas/terapia , Acondicionamiento Pretrasplante/métodos , Vidarabina/análogos & derivados , Adolescente , Adulto , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Antineoplásicos Alquilantes/administración & dosificación , Antineoplásicos Alquilantes/efectos adversos , Antineoplásicos Alquilantes/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Busulfano/administración & dosificación , Busulfano/efectos adversos , Busulfano/farmacocinética , Busulfano/uso terapéutico , Niño , Preescolar , Femenino , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Recurrencia , Factores de Riesgo , Análisis de Supervivencia , Vidarabina/administración & dosificación , Vidarabina/efectos adversos , Vidarabina/farmacocinética , Vidarabina/uso terapéutico , Adulto Joven
8.
Anticancer Drugs ; 20(2): 97-103, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19209025

RESUMEN

A phase I and pharmacokinetic study was carried out with the new ruthenium complex indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019, FFC14A). Seven patients with various types of solid tumours refractory to standard therapy were treated with escalating doses of KP1019 (25-600 mg) twice weekly for 3 weeks. No dose-limiting toxicity occurred. Ruthenium plasma concentration-time profiles after the first dose and under multiple-dose conditions were analysed using a compartmental approach. The pharmacokinetic disposition was characterised by a small volume of distribution, low clearance and long half-life. Only a small fraction of ruthenium was excreted renally. The area under the curve values increased proportionally with dose indicating linear pharmacokinetics.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Indazoles/administración & dosificación , Indazoles/farmacocinética , Neoplasias/tratamiento farmacológico , Compuestos de Rutenio/administración & dosificación , Compuestos de Rutenio/farmacocinética , Adulto , Anciano , Algoritmos , Antineoplásicos/efectos adversos , Área Bajo la Curva , Femenino , Humanos , Indazoles/efectos adversos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Modelos Biológicos , Compuestos Organometálicos , Compuestos de Rutenio/efectos adversos
9.
Cancer Chemother Pharmacol ; 62(1): 19-31, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17823799

RESUMEN

OBJECTIVES: High-dose chemotherapy with the alkylating agent busulfan has been widely used in the treatment of patients with high-risk Ewing's sarcoma. Because of risks for toxicity, busulfan and radiotherapy can not be applied together, leading to the omission of one effective therapy component. Treosulfan is a derivative of busulfan which has a lower side effect profile than busulfan and which can be used together with radiotherapy. We investigated the effect of treosulfan in a panel of Ewing's sarcoma cell lines on cell survival, cell cycle and apoptosis in vitro and compared it to busulfan. Furthermore, the anti-tumor effect of treosulfan was studied in an orthotopic Ewing's sarcoma mouse xenograft model. METHODS: Cell survival was measured by MTT assay and cell cycle analysis by flow cytometry. Apoptosis was analyzed via detection of DNA fragmentation, Hoechst 33258 staining, Annexin V, and cleavage of caspases-3 and 9. The effect of treosulfan and busulfan on primary tumor growth was assessed in Ewing's sarcoma xenografts in NOD/SCID mice (10 mice per group), pharmacokinetics of treosulfan were analyzed in nude mice. RESULTS: Treosulfan inhibited cell growth to at least 70% in all cell lines at concentrations achievable in vivo. Treosulfan had a greater effect on the inhibition of cell growth at equivalent concentrations compared to busulfan. The growth inhibitory effect of treosulfan at low concentrations was mainly due to a G2 cell cycle arrest, whereas at higher concentrations it was due to apoptosis. Apoptosis was induced at lower concentrations compared to busulfan. In contrast to busulfan, treosulfan induced cell death in an apoptosis-deficient cell line at concentrations achievable in vivo. In mice, treosulfan suppressed tumor growth at dosages of 2,500 and 3,000 mg/kg. Pharmacokinetic exposures of treosulfan in mice were similar to previous reports in human patients. At maximal tolerated dosages treosulfan had a higher anti-tumor activity than busulfan. CONCLUSIONS: Our results suggest that treosulfan has efficacy against Ewing's sarcoma cells in vitro and in mice. Therefore, controlled trials examining the role of treosulfan in patients with Ewing's sarcoma are warranted.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Busulfano/análogos & derivados , Sarcoma de Ewing/tratamiento farmacológico , Animales , Anexina A5/fisiología , Antineoplásicos Alquilantes/farmacocinética , Apoptosis/efectos de los fármacos , Bisbenzimidazol , Western Blotting , Busulfano/farmacocinética , Busulfano/uso terapéutico , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Colorantes , Fase G1/efectos de los fármacos , Indicadores y Reactivos , Inyecciones Intraperitoneales , Ratones , Ratones Desnudos , Sales de Tetrazolio , Tiazoles , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Clin Oncol ; 23(5): 965-72, 2005 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-15613696

RESUMEN

PURPOSE: BAY 43-9006 is a novel dual-action Raf kinase and vascular endothelial growth factor receptor inhibitor that inhibits tumor cell proliferation and angiogenesis. This study established the safety and pharmacokinetics of BAY 43-9006 in 69 patients with advanced refractory solid tumors. PATIENTS AND METHODS: BAY 43-9006 (50 to 800 mg) was administered once or twice daily on a varying weekly schedule. Pharmacokinetic sampling was performed in all patients; preliminary tumor response was also assessed. The effect of BAY 43-9006 on phorbol myristate acetate-stimulated ERK phosphorylation in peripheral blood lymphocytes was studied using flow cytometry. RESULTS: Mild to moderate diarrhea was the most common (55%) treatment-related adverse event. The maximum-tolerated dose was 400 mg bid continuous. Dose-limiting toxicities were grade 3 diarrhea and fatigue at 800 mg bid, and grade 3 skin toxicity at 600 mg bid. BAY 43-9006 pharmacokinetics were highly variable for single and multiple dosing, and toxicity did not appear to be dose dependent. Significant decreases of phorbol myristate acetate-stimulated ERK phosphorylation (P < .01) were identified at doses >/= 200 mg bid continuous. Forty-five patients were assessable for efficacy; one patient had a partial response (hepatocellular carcinoma at 400 mg bid continuous), 25 patients had stable disease, with eight lasting > 6 months and five for >12 months. Eighteen patients had progressive disease, and tumor response could not be evaluated in one patient. CONCLUSION: Oral BAY 43-9006 was well tolerated and appeared to provide some clinical benefits. Based on the results of this study, BAY 43-9006 at 400 mg bid continuous is recommended for ongoing and future studies.


Asunto(s)
Antineoplásicos/administración & dosificación , Bencenosulfonatos/administración & dosificación , Proteínas de Unión a Fosfatidiletanolamina/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Piridinas/administración & dosificación , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Bencenosulfonatos/farmacocinética , Carcinoma Hepatocelular/tratamiento farmacológico , Estudios de Cohortes , Neoplasias del Colon/tratamiento farmacológico , Diarrea/inducido químicamente , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Fatiga/inducido químicamente , Femenino , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Linfocitos/efectos de los fármacos , Linfocitos/enzimología , Masculino , Persona de Mediana Edad , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Proteínas de Unión a Fosfatidiletanolamina/efectos adversos , Proteínas de Unión a Fosfatidiletanolamina/farmacocinética , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacocinética , Piridinas/farmacocinética , Neoplasias del Recto/tratamiento farmacológico , Seguridad , Sorafenib
11.
Clin Colorectal Cancer ; 5(3): 188-96, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16197622

RESUMEN

BACKGROUND: Sorafenib (BAY 43-9006), a multiple kinase inhibitor, has been shown to inhibit tumor growth and tumor angiogenesis by targeting Raf kinase, vascular endothelial growth factor receptor, and platelet-derived growth factor receptor. In phase I studies, sorafenib demonstrated single-agent activity in patients with advanced solid tumors and was successfully combined with oxaliplatin in preclinical studies. This phase I study investigated the safety, pharmacokinetics, and efficacy of sorafenib in combination with oxaliplatin. PATIENTS AND METHODS: Twenty-seven patients with refractory solid tumors were enrolled in the initial dose-escalation part (cohorts 1, 2A, and 2B) and 10 additional patients with oxaliplatin-refractory colorectal cancer were subsequently enrolled in an extension part (cohort 3). Oxaliplatin 130 mg/m2 was given on day 1 of a 3-week cycle and oral sorafenib was administered continuously from day 4 of cycle 1 at 200 mg twice daily (cohort 1) or 400 mg twice daily (cohorts 2A, 2B, and 3). RESULTS: Adverse events were generally mild to moderate and the maximum tolerated dose was not reached. Common adverse events were diarrhea (52% of patients in the dose-escalation part and 20% in the extension part), sensory neuropathy (44% and 20%), and dermatologic toxicities (41% and 80%). No pharmacokinetic interaction between sorafenib and oxaliplatin was detectable. Two patients with gastric cancer had a partial response. Forty-three percent of patients in cohorts 1 and 2A/B and 78% of patients in cohort 3 exhibited stable disease for >or=10 weeks. CONCLUSION: Continuous oral sorafenib 400 mg twice daily was safely combined with oxaliplatin without detectable drug interactions and showed preliminary antitumor activity in this phase I study. This dose is recommended for phase II studies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Bencenosulfonatos/administración & dosificación , Neoplasias Colorrectales/patología , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Niacinamida/análogos & derivados , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino , Compuestos de Fenilurea , Piridinas/administración & dosificación , Sorafenib
12.
J Inorg Biochem ; 153: 247-252, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26364260

RESUMEN

Intracellular binding of cisplatin to non-DNA partners, such as proteins, has received increasing attention as an additional mode of action and as mechanism of resistance. We investigated two cisplatin-interacting isoforms of protein disulfide isomerase regarding their contribution to acquired cisplatin resistance using sensitive and resistant A2780/A2780cis ovarian cancer cells. Cisplatin cytotoxicity was assessed after knockdown of either protein disulfide isomerase family A member 1 (PDIA1) or protein disulfide isomerase family A member 3 (PDIA3). Whereas PDIA1 knockdown led to increased cytotoxicity in resistant A2780cis cells, PDIA3 knockdown showed no influence on cytotoxicity. Coincubation with propynoic acid carbamoyl methyl amide 31 (PACMA31), a PDIA1 inhibitor, resensitized A2780cis cells to cisplatin treatment. Determination of the combination index revealed that the combination of cisplatin and PACMA31 acts synergistically. Our results warrant further evaluation of PDIA1 as promising target for chemotherapy, and its inhibition by PACMA31 as a new therapeutic approach.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos , Procolágeno-Prolina Dioxigenasa/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Antineoplásicos/química , Línea Celular Tumoral , Cisplatino/química , Inhibidores Enzimáticos/farmacología , Humanos , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Procolágeno-Prolina Dioxigenasa/genética , Unión Proteica , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/genética
13.
Semin Oncol ; 31(6 Suppl 13): 16-35, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15717736

RESUMEN

Encapsulation of doxorubicin in polyethylene glycol-coated liposomes (Doxil/Caelyx [PLD]), was developed to enhance the safety and efficacy of conventional doxorubicin. The liposomes alter pharmacologic and pharmacokinetic parameters of conventional doxorubicin so that drug delivery to the tumor is enhanced while toxicity normally associated with conventional doxorubicin is decreased. In animals and humans, pharmacokinetic advantages of PLD include an increased area under the plasma concentration-time curve, longer distribution half-life, smaller volume of distribution, and reduced clearance. In preclinical models, PLD produced remission and cure against many cancers including tumors of the breast, lung, ovaries, prostate, colon, bladder, and pancreas, as well as lymphoma, sarcoma, and myeloma. It was also found to be effective as adjuvant therapy. In addition, it was found to cross the blood-brain barrier and induce remission in tumors of the central nervous system. Increased potency over conventional doxorubicin was observed and, in contrast to conventional doxorubicin, PLD was equally effective against low- and high-growth fraction tumors. The combination of PLD with vincristine or trastuzumab resulted in additive effects and possible synergy. PLD appeared to overcome multidrug resistance, possibly as the result of increased intracellular concentrations and an interaction between the liposome and P-glycoprotein function. On the basis of pharmacokinetic and preclinical studies, PLD, either alone or as part of combination therapy, has potential applications to treat a variety of cancers.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Animales , Antibióticos Antineoplásicos/farmacocinética , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Cetuximab , Doxorrubicina/farmacocinética , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Liposomas/farmacocinética , Modelos Animales , Trastuzumab
14.
PLoS One ; 8(9): e74555, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24040280

RESUMEN

Pancreatic cancer is one of the leading cancer-related causes of death in the western world with an urgent need for new treatment strategies. Recently, hyperforin and nemorosone have been described as promising anti-cancer lead compounds. While hyperforin has been thoroughly investigated in vitro and in vivo, in vivo data for nemorosone are still missing. Thus, we investigated the growth-inhibitory potential of nemorosone on pancreatic cancer xenografts in NMRI nu/nu mice and determined basic pharmacokinetic parameters. Xenograft tumors were treated with nemorosone and gemcitabine, the current standard of care. Tumor sections were subjected to H&E as well as caspase 3 and Ki-67 staining. Nemorosone plasma kinetics were determined by HPLC and mass spectrometry. Induction of CYP3A4 and other metabolizing enzymes by nemorosone and hyperforin was tested on primary hepatocytes using qRT-PCR. At a dose of 50 mg/kg nemorosone per day, a significant growth-inhibitory effect was observed in pancreatic cancer xenografts. The compound was well tolerated and rapidly absorbed into the bloodstream with a half-life of approximately 30 min. Different metabolites were detected, possibly resembling CYP3A4-independent oxidation products. It is concluded that nemorosone is a potential anti-cancer lead compound with good bioavailability, little side-effects and promising growth-inhibitory effects, thus representing a valuable compound for a combination therapy approach.


Asunto(s)
Antineoplásicos/farmacocinética , Benzofenonas/farmacocinética , Carcinoma/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Benzofenonas/administración & dosificación , Benzofenonas/sangre , Disponibilidad Biológica , Biotransformación , Carcinoma/sangre , Carcinoma/patología , Caspasa 3/genética , Caspasa 3/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Esquema de Medicación , Femenino , Expresión Génica , Semivida , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Inyecciones Subcutáneas , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/patología , Floroglucinol/administración & dosificación , Floroglucinol/análogos & derivados , Cultivo Primario de Células , Terpenos/administración & dosificación , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
15.
Oncotarget ; 4(5): 677-90, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23633472

RESUMEN

Cyclic AMP (cAMP) regulates a number of cellular processes and modulates cell death induction. cAMP levels are altered upon stimulation of specific G-protein-coupled receptors inhibiting or activating adenylyl cyclases. Opioid receptor stimulation can activate inhibitory Gi-proteins which in turn block adenylyl cyclase activity reducing cAMP. Opioids such as D,L-methadone induce cell death in leukemia cells. However, the mechanism how opioids trigger apoptosis and activate caspases in leukemia cells is not understood. In this study, we demonstrate that downregulation of cAMP induced by opioid receptor activation using the opioid D,L-methadone kills and sensitizes leukemia cells for doxorubicin treatment. Enhancing cAMP levels by blocking opioid-receptor signaling strongly reduced D,L-methadone-induced apoptosis, caspase activation and doxorubicin-sensitivity. Induction of cell death in leukemia cells by activation of opioid receptors using the opioid D,L-methadone depends on critical levels of opioid receptor expression on the cell surface. Doxorubicin increased opioid receptor expression in leukemia cells. In addition, the opioid D,L-methadone increased doxorubicin uptake and decreased doxorubicin efflux in leukemia cells, suggesting that the opioid D,L-methadone as well as doxorubicin mutually increase their cytotoxic potential. Furthermore, we found that opioid receptor activation using D,L-methadone alone or in addition to doxorubicin inhibits tumor growth significantly in vivo. These results demonstrate that opioid receptor activation via triggering the downregulation of cAMP induces apoptosis, activates caspases and sensitizes leukemia cells for doxorubicin treatment. Hence, opioid receptor activation seems to be a promising strategy to improve anticancer therapies.


Asunto(s)
Adenilil Ciclasas/metabolismo , Apoptosis , AMP Cíclico/metabolismo , Leucemia de Células B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Receptores Opioides/metabolismo , Inhibidores de Adenilato Ciclasa , Analgésicos Opioides/farmacología , Animales , Antibióticos Antineoplásicos/farmacología , Caspasas/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Doxorrubicina/sangre , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Humanos , Masculino , Metadona/sangre , Metadona/farmacología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Receptores Opioides/biosíntesis , Transducción de Señal/efectos de los fármacos
16.
PLoS One ; 8(1): e54193, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23349823

RESUMEN

INTRODUCTION: Inherent and acquired cisplatin resistance reduces the effectiveness of this agent in the management of non-small cell lung cancer (NSCLC). Understanding the molecular mechanisms underlying this process may result in the development of novel agents to enhance the sensitivity of cisplatin. METHODS: An isogenic model of cisplatin resistance was generated in a panel of NSCLC cell lines (A549, SKMES-1, MOR, H460). Over a period of twelve months, cisplatin resistant (CisR) cell lines were derived from original, age-matched parent cells (PT) and subsequently characterized. Proliferation (MTT) and clonogenic survival assays (crystal violet) were carried out between PT and CisR cells. Cellular response to cisplatin-induced apoptosis and cell cycle distribution were examined by FACS analysis. A panel of cancer stem cell and pluripotent markers was examined in addition to the EMT proteins, c-Met and ß-catenin. Cisplatin-DNA adduct formation, DNA damage (γH2AX) and cellular platinum uptake (ICP-MS) was also assessed. RESULTS: Characterisation studies demonstrated a decreased proliferative capacity of lung tumour cells in response to cisplatin, increased resistance to cisplatin-induced cell death, accumulation of resistant cells in the G0/G1 phase of the cell cycle and enhanced clonogenic survival ability. Moreover, resistant cells displayed a putative stem-like signature with increased expression of CD133+/CD44+cells and increased ALDH activity relative to their corresponding parental cells. The stem cell markers, Nanog, Oct-4 and SOX-2, were significantly upregulated as were the EMT markers, c-Met and ß-catenin. While resistant sublines demonstrated decreased uptake of cisplatin in response to treatment, reduced cisplatin-GpG DNA adduct formation and significantly decreased γH2AX foci were observed compared to parental cell lines. CONCLUSION: Our results identified cisplatin resistant subpopulations of NSCLC cells with a putative stem-like signature, providing a further understanding of the cellular events associated with the cisplatin resistance phenotype in lung cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Antígeno AC133 , Aldehído Deshidrogenasa/metabolismo , Antígenos CD/metabolismo , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Aductos de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Glicoproteínas/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Factores de Transcripción SOXB1/metabolismo , beta Catenina/metabolismo
17.
Cancer Chemother Pharmacol ; 69(3): 753-61, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22020317

RESUMEN

PURPOSE: S-1 is a novel oral fluoropyrimidine comprised of FT and two modulators, gimeracil (CDHP) and oteracil potassium (Oxo). This study investigated the food effects on the pharmacokinetics (PK) of Oxo, other components of S-1, and their metabolites at different gastric pH adjusted by proton pump inhibitor (PPI). METHODS: Patients with and without PPI were treated with S-1 at 30 mg/m(2) twice daily orally on days 1-7 under either fed or fasting condition, and then were crossed over to fasting/fed conditions on days 15-21 with washout on days 8-14 and 22-28. RESULTS: The study enrolled 55 patients including 27 PK-evaluable patients. For the single-dose and multiple-dose pharmacokinetics, the administration of S-1 under fed conditions resulted in decreased exposure to Oxo relative to fasting administration. There was a marginal decrease in exposure to CDHP and 5-FU under fed versus fasting conditions, although FT exposure was not altered by food, which demonstrated lack of food effect. PPI administration together with S-1 did not significantly change its bioavailability. CONCLUSIONS: Oxo exposure was reduced under fed compared to fasting condition. To increase the bioavailability of S-1, the administration of S-1 under fasting condition was more effective in the western countries.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacocinética , Interacciones Alimento-Droga , Neoplasias/tratamiento farmacológico , Ácido Oxónico/farmacocinética , Inhibidores de la Bomba de Protones/farmacología , Tegafur/farmacocinética , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/efectos adversos , Antimetabolitos Antineoplásicos/uso terapéutico , Disponibilidad Biológica , Estudios Cruzados , Esquema de Medicación , Combinación de Medicamentos , Ayuno , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ácido Oxónico/administración & dosificación , Ácido Oxónico/efectos adversos , Ácido Oxónico/uso terapéutico , Inhibidores de la Bomba de Protones/administración & dosificación , Inhibidores de la Bomba de Protones/uso terapéutico , Tegafur/administración & dosificación , Tegafur/efectos adversos , Tegafur/uso terapéutico , Resultado del Tratamiento
18.
Br J Ophthalmol ; 96(4): 591-6, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22275346

RESUMEN

OBJECTIVE: Two conjunctival cell lines (CRMM-1 and CRMM-2) have been established from recurrent conjunctival melanoma. The authors examined the chemosensitivity of these cell lines to cytotoxic substances and combinations to identify substances that inhibit cell growth efficiently in vitro. MATERIAL AND METHODS: CRMM-1 and CRMM-2 were exposed to cisplatin, mitomycin C (MMC), all-trans-retinoic-acid (ATRA), fotemustine or imatinib for 24 h. Sulforhodamine-B assays were used to assess the IC(50). Isobolograms were performed to test possible synergism and antagonism with ATRA or imatinib. RESULTS: Cisplatin and MMC were efficient to inhibit the growth of CRMM-1 and CRMM-2. Combination of imatinib with MMC showed additive antitumoral effect on both cell lines. Combined treatment of imatinib with fotemustine or cisplatin resulted in antagonism. Strong antagonisms were also obtained with ATRA and fotemustine or cisplatin in both cell lines. A synergism was found for ATRA and mitomycin or imatinib in CRMM-2, in contrast to CRMM-1, where antagonism was obtained. CONCLUSIONS: Cisplatin and MMC inhibit cell growth in conjunctival melanoma cell lines. The potential of ATRA was evident only in combination with MMC or imatinib in CRMM-2 cells. Imatinib and mitomycin increased their efficiency under combination therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Conjuntiva/patología , Melanoma/patología , Línea Celular Tumoral , Proliferación Celular , Neoplasias de la Conjuntiva/tratamiento farmacológico , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Melanoma/tratamiento farmacológico
19.
J Anal Toxicol ; 35(5): 302-11, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21619725

RESUMEN

Intoxications with succinylcholine (SUX) lead to a potentially lethal respiratory paralysis, and forensic cases involving accidental or deliberate SUX-application have been reported. Detection of SUX as well as its metabolite succinylmonocholine (SMC) is difficult: both substances are analytically challenging, and the extremely short plasma half-life of SUX additionally hampers detection of the parent compound. Pharmacokinetic data are scarce on SUX and non-existent on SMC. To enhance forensic knowledge concerning SUX intoxications, plasma pharmacokinetics of SMC were investigated in anesthetized patients. Fifteen subjects scheduled for a surgical procedure were included in this study. Muscle-relaxation was initialized with a bolus injection of 80-100 mg SUX. Blood sampling was performed within 6 h after SUX application using paraoxonized tubes. Collected plasma was processed according to a validated isotope dilution high-performance liquid chromatography-tandem mass spectrometry method using ion-pair solid-phase extraction. Pharmacokinetic parameters were derived from a user-defined as well as a three-compartment model. For SMC, peak plasma concentrations were reached after 0.03-2.0 min. In contrast to SUX, SMC was more slowly and more extensively distributed, featuring triphasic plasma concentration time profiles. Pharmacokinetic key parameters were subject to interindividual variation of potential forensic importance, with terminal half-lives of 1-3 h indicating a detection interval of 8-24 h for SMC in plasma. SMC was proven to be the only realistic SUX marker in a forensic context. The present work defines meaningful detection windows for plasmatic SMC after SUX application and offers guideline values for forensic toxicological casework.


Asunto(s)
Fármacos Neuromusculares Despolarizantes/farmacocinética , Succinilcolina/análogos & derivados , Adulto , Anciano , Análisis Químico de la Sangre , Femenino , Toxicología Forense , Semivida , Humanos , Límite de Detección , Masculino , Persona de Mediana Edad , Fármacos Neuromusculares Despolarizantes/efectos adversos , Intoxicación/sangre , Intoxicación/diagnóstico , Succinilcolina/efectos adversos , Succinilcolina/farmacocinética , Adulto Joven
20.
Br J Pharmacol ; 162(5): 1045-59, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21091652

RESUMEN

BACKGROUND AND PURPOSE: Pancreatic cancer is one of the leading cancer-related causes of death due to high chemo-resistance and fast metastasation. Nemorosone, a polycyclic polyprenylated acylphloroglucinol, has recently been identified as a promising anticancer agent. Here, we examine its growth-inhibitory effects on pancreatic cancer cells. Based on transcription profiling, a molecular mode of action is proposed. EXPERIMENTAL APPROACH: Nemorosone cytotoxicity was assessed by the resazurin proliferation assay on pancreatic cancer cells and fibroblasts. Apoptosis was determined by Annexin V/propidium iodide staining as well as cytochrome c and caspase activation assays. Staining with the voltage-dependent dye JC-1 and fluorescence microscopy were used to detect effects on mitochondrial membrane potential. Total RNA was isolated from treated cell lines and subjected to microarray analysis, subsequent pathway identification and modelling. Gene expression data were validated by quantitative polymerase chain reaction and siRNA-mediated gene knock-down. KEY RESULTS: Nemorosone significantly inhibited cancer cell growth, induced cytochrome c release and subsequent caspase-dependent apoptosis, rapidly abolished mitochondrial membrane potential and elevated cytosolic calcium levels, while fibroblasts were largely unaffected. Expression profiling revealed 336 genes to be affected by nemorosone. A total of 75 genes were altered in all three cell lines, many of which were within the unfolded protein response (UPR) network. DNA damage inducible transcript 3 was identified as a key regulator in UPR-mediated cell death. CONCLUSIONS AND IMPLICATIONS: Nemorosone could be a lead compound for the development of novel anticancer drugs amplifying the already elevated UPR level in solid tumours, thus driving them into apoptosis. This study forms the basis for further investigations identifying nemorosone's direct molecular target(s).


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Benzofenonas/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Apoptosis/efectos de los fármacos , Secuencia de Bases , Calcio/metabolismo , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocromos c/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Perfilación de la Expresión Génica , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Interferencia de ARN , ARN Interferente Pequeño/genética , Estrés Fisiológico/efectos de los fármacos , Factor de Transcripción CHOP/antagonistas & inhibidores , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Respuesta de Proteína Desplegada/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA