Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pediatr Hematol Oncol ; 33(1): 25-30, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21063221

RESUMEN

The primary aim of this phase 1 study was to determine the maximum tolerated dose (MTD) and evaluate the safety of nifurtimox alone and in combination with cyclophosphamide and topotecan in multiple relapsed/refractory neuroblastoma pediatric patients. The secondary aim was to evaluate the pharmacokinetics of nifurtimox and the treatment response. To these ends, we performed a phase 1 dose escalation trial of daily oral nifurtimox with toxicity monitoring to determine the MTD, followed by 3 cycles of nifurtimox in combination with cyclophosphamide and topotecan. Samples were collected to determine the pharmacokinetic parameters maximum concentration, time at which maximum concentration is reached, and area under the curve between 0 and 8 hours. Treatment response was evaluated by radiographic and radionuclide (I-metaiodobenzylguanidine) imaging, measurement of urinary catecholamines, and clearance of bone marrow disease. We determined the MTD of nifurtimox to be 30 mg/kg/d. The non-dose-limiting toxicities were mainly nausea and neuropathy. The dose-limiting toxicities of 2 patients at 40 mg/kg/d were a grade 3 pulmonary hemorrhage and a grade 3 neuropathy (reversible). Overall, nifurtimox was well tolerated by pediatric patients at a dose of 30 mg/kg/d, and tumor responses were seen both as a single agent and in combination with chemotherapy. A Phase 2 study to determine the antitumor efficacy of nifurtimox is currently underway.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neuroblastoma/tratamiento farmacológico , Nifurtimox/efectos adversos , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Preescolar , Ciclofosfamida/efectos adversos , Ciclofosfamida/farmacocinética , Ciclofosfamida/uso terapéutico , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Neuroblastoma/prevención & control , Nifurtimox/farmacocinética , Nifurtimox/uso terapéutico , Recurrencia , Topotecan/efectos adversos , Topotecan/farmacocinética , Topotecan/uso terapéutico , Resultado del Tratamiento
2.
Biochemistry ; 48(34): 8249-60, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19639961

RESUMEN

The glycosyl-phosphatidylinositol anchored folate receptor (FR) mediates selective delivery of a broad range of experimental drugs to the receptor-rich tumors, but molecular mechanisms controlling FR internalization have not been adequately studied. FR quantitatively recycles between the cell surface and endocytic compartments via a Cdc42-dependent pinocytic pathway. Protein kinase C (PKC) activators including diacylglycerol and phorbol ester have previously been reported to increase the proportion of FR on the cell surface. Here we identify the alpha-subtype of PKC as the mediator of phorbol ester action on FR recycling and provide evidence that activated PKCalpha is recruited to FR-rich membrane microdomains where, in association with its receptor RACK1, it inhibits FR internalization; the activation state of Cdc42 remains unaltered. We also show that the PKC substrate, annexin II, is required for FR internalization. The studies clarify a molecular mechanism for the regulation of FR recycling through PKC which could potentially be exploited for effective drug delivery.


Asunto(s)
Proteínas Portadoras/metabolismo , Endocitosis , Proteína Quinasa C-alfa/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Anexina A2/metabolismo , Secuencia de Bases , Línea Celular , Detergentes/farmacología , Endocitosis/efectos de los fármacos , Receptores de Folato Anclados a GPI , Humanos , Ligandos , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Ésteres del Forbol/farmacología , Proteína Quinasa C-alfa/genética , Transporte de Proteínas/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología , Proteína de Unión al GTP cdc42/metabolismo
3.
Exp Cell Res ; 314(17): 3107-17, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18722367

RESUMEN

Bone marrow-derived mesenchymal stromal cells (MSCs) localize to solid tumors. Defining the signaling mechanisms that regulate this process is important in understanding the role of MSCs in tumor growth. Using a combination of chromatography and electrospray tandem mass spectrometry we have identified novel soluble signaling molecules that induce MSC chemotaxis present in conditioned medium of the breast carcinoma cell line MDA-MB231. Previous work has employed survey strategies using ELISA assay to identify known chemokines that promote MSC chemotaxis. While these studies provide valuable insights into the intercellular signals that impact MSC behavior, many less well-described, but potentially important soluble signaling molecules could be overlooked using these methods. Through the less directed method of column chromatography we have identified novel candidate MSC chemotactic peptides. Two proteins, cyclophilin B and hepatoma-derived growth factor were then further characterized and shown to promote MSC chemotaxis.


Asunto(s)
Células de la Médula Ósea/metabolismo , Factores Quimiotácticos/química , Medios de Cultivo Condicionados/química , Células Madre Mesenquimatosas/metabolismo , Células del Estroma/metabolismo , Células Tumorales Cultivadas/metabolismo , Secuencia de Aminoácidos , Animales , Células de la Médula Ósea/citología , Neoplasias de la Mama , Factores Quimiotácticos/metabolismo , Quimiotaxis/fisiología , Cromatografía de Afinidad/métodos , Ciclofilinas/genética , Ciclofilinas/metabolismo , Citoesqueleto , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Madre Mesenquimatosas/citología , Datos de Secuencia Molecular , Péptidos/genética , Péptidos/metabolismo , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células del Estroma/citología , Células Tumorales Cultivadas/química
4.
Pediatr Neurol ; 40(1): 34-41, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19068252

RESUMEN

The neurotoxic effects of therapy for childhood acute lymphoblastic leukemia can result in leukoencephalopathy or measurable deficits in cognitive function. However, there are no validated biomarkers that allow the identification of those patients at greatest risk. With the objective of identifying such predictors, cerebrospinal fluid collected from 53 patients over 2.5 years of therapy for childhood acute lymphoblastic leukemia was retrospectively studied. Cerebrospinal fluid folate, concentrated relative to serum folate prior to therapy, dropped during the first month of therapy and remained below baseline throughout treatment. Cerebrospinal fluid homocysteine was inversely related to cognitive function prior to treatment. Oral methotrexate was associated with decreased cerebrospinal fluid folate and increased cerebrospinal fluid homocysteine, but these changes were not seen with oral aminopterin. Of 36 patients who had imaging after completion of therapy, 9 had periventricular or subcortical white matter abnormalities consistent with leukoencephalopathy. Peak cerebrospinal fluid tau concentrations during therapy were higher among patients who had leukoencephalopathy after completion of therapy than among those with normal imaging studies at the end of therapy. If confirmed prospectively, these markers may allow the identification of those patients at greatest risk of developing treatment-induced neurocognitive dysfunction, thus guiding preventive interventions.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Biomarcadores de Tumor/líquido cefalorraquídeo , Cognición/efectos de los fármacos , Ácido Fólico/líquido cefalorraquídeo , Homeostasis , Leucemia-Linfoma Linfoblástico de Células Precursoras/líquido cefalorraquídeo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Administración Oral , Adolescente , Aminopterina/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Biomarcadores de Tumor/sangre , Encéfalo/patología , Niño , Preescolar , Cromatografía Líquida de Alta Presión , Demencia Vascular/etiología , Demencia Vascular/patología , Demencia Vascular/fisiopatología , Ácido Fólico/sangre , Antagonistas del Ácido Fólico/efectos adversos , Homocisteína/líquido cefalorraquídeo , Humanos , Lactante , Imagen por Resonancia Magnética , Metotrexato/efectos adversos , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/patología , Síndromes de Neurotoxicidad/fisiopatología , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangre , Estudios Retrospectivos , Adulto Joven , Proteínas tau/líquido cefalorraquídeo
5.
J Neurochem ; 104(6): 1494-503, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18086128

RESUMEN

Reduced derivatives of folic acid (folates) play a critical role in the development, function and repair of the CNS. However, the molecular systems regulating folate uptake and homeostasis in the central nervous system remain incompletely defined. Choroid plexus epithelial cells express high levels of folate receptor alpha (FRalpha) suggesting that the choroid plays an important role in CNS folate trafficking and maintenance of CSF folate levels. We have characterized 5-methyltetrahydrofolate (5-MTHF) uptake and metabolism by primary rat choroid plexus epithelial cells in vitro. Two distinct processes are apparent; one that is FRalpha dependent and one that is independent of the receptor. FRalpha binds 5-MTHF with high affinity and facilitates efficient uptake of 5-MTHF at low extracellular folate concentrations; a lower affinity FRalpha independent system accounts for increased folate uptake at higher concentrations. Cellular metabolism of 5-MTHF depends on the route of folate entry into the cell. 5-MTHF taken up via a non-FRalpha -mediated process is rapidly metabolized to folylpolyglutamates, whereas 5-MTHF that accumulates via FRalpha remains non-metabolized, supporting the hypothesis that FRalpha may be part of a pathway for transcellular movement of the vitamin. The proton-coupled folate transporter, proton-coupled folate transporter (PCFT), mRNA was also shown to be expressed in choroid plexus epithelial cells. This is consistent with the role we have proposed for proton-coupled folate transporter in FRalpha-mediated transport as the mechanism of export of folates from the endocytic compartment containing FRalpha.


Asunto(s)
Plexo Coroideo/citología , Células Epiteliales/metabolismo , Ácido Fólico/farmacocinética , Complejo Vitamínico B/farmacocinética , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Endodesoxirribonucleasas/metabolismo , Células Epiteliales/citología , Técnica del Anticuerpo Fluorescente , Receptores de Folato Anclados a GPI , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Tetrahidrofolatos/farmacocinética , Vesículas Transportadoras/metabolismo , Tritio
6.
Cancer Chemother Pharmacol ; 62(1): 65-75, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17768625

RESUMEN

PURPOSE: Aminopterin offers advantages over the related antifolate, methotrexate, including greater potency, complete bioavailability, and more consistent accumulation and metabolism by patients' blasts. This current trial was done to document the toxicity of the aminopterin within a multiagent therapeutic regimen for children with newly diagnosed ALL. EXPERIMENTAL DESIGN: Patients at high risk of relapse were non-randomly assigned to therapy including oral aminopterin 4 mg/m(2), in two doses 12 h apart, in place of methotrexate 100 mg/m(2) in four divided doses. RESULTS: Thirty-two patients, 22 with pre-B ALL and ten with T-lineage ALL, have been treated with aminopterin, with median follow up of 40 months. Hematologic, mucosal and hepatic toxicity has been tolerable and reversible. There have been no toxic deaths among patients in remission. During weekly AMT therapy, higher mean neutrophil counts were observed among patients who were wild type for polymorphisms in methylene tetrahydrofolate reductase and methionine synthase reductase. CONCLUSIONS: Aminopterin can be safely incorporated in multiagent therapy for patients with ALL, in place of systemic methotrexate, without causing excessive toxicity. These results support a larger trial comparing the efficacy and toxicity of aminopterin and methotrexate in therapy for patients with ALL.


Asunto(s)
Aminopterina/uso terapéutico , Antagonistas del Ácido Fólico/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Adolescente , Aminopterina/efectos adversos , Aminopterina/farmacocinética , Antídotos/uso terapéutico , Antimetabolitos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Enfermedades Óseas/inducido químicamente , Niño , Preescolar , Sobredosis de Droga , Eritrocitos/metabolismo , Femenino , Fiebre/complicaciones , Fiebre/etiología , Antagonistas del Ácido Fólico/efectos adversos , Antagonistas del Ácido Fólico/farmacocinética , Enfermedades Gastrointestinales/inducido químicamente , Enfermedades Gastrointestinales/complicaciones , Humanos , Leucovorina/uso terapéutico , Masculino , Metotrexato/uso terapéutico , Síndromes de Neurotoxicidad/complicaciones , Proyectos Piloto , Polimorfismo de Nucleótido Simple , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Recurrencia , Resultado del Tratamiento
8.
Mol Cancer Ther ; 6(11): 2909-20, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18025275

RESUMEN

Folylpolyglutamyl synthase (FPGS) converts intracellular folates and antifolates to polyglutamates. Polyglutamylated folates and antifolates are retained in cells longer and are better substrates than their monoglutamate counterparts for enzymes involved in one-carbon transfer. FPGS modulation affects the chemosensitivity of cancer cells to antifolates, such as methotrexate, and 5-fluorouracil (5FU) by altering polyglutamylation of antifolates and specific target intracellular folate cofactors. However, this effect may be counterbalanced by FPGS modulation-induced changes in polyglutamylation of other intracellular folate cofactors and total intracellular folate pools. We generated an in vitro model of FPGS overexpression and inhibition in breast cancer cells by stably transfecting human MDA-MB-435 breast cancer cells with the sense FPGS cDNA or FPGS-targeted small interfering RNA, respectively, and investigated the effects of FPGS modulation on chemosensitivity to 5FU and methotrexate. FPGS modulation-induced changes in polyglutamylation of both antifolates and folate cofactors and in intracellular folate pools affected chemosensitivity of breast cancer cells to pemetrexed and trimetrexate whose cytotoxic effects do or do not depend on polyglutamylation, respectively, in a predictable manner. However, the effects of FPGS modulation on the chemosensitivity of breast cancer cells to 5FU and methotrexate seem to be highly complex and depend not only on polyglutamylation of a specific target intracellular folate cofactor or methotrexate, respectively, but also on total intracellular folate pools and polyglutamylation of other intracellular folate cofactors. Whether or not FPGS modulation may be an important clinical determinant of chemosensitivity of breast cancer cells to 5FU and methotrexate-based chemotherapy needs further exploration.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Ensayos de Selección de Medicamentos Antitumorales , Ácido Fólico/farmacología , Péptido Sintasas/metabolismo , Línea Celular Tumoral , Glutamatos/farmacología , Guanina/análogos & derivados , Guanina/farmacología , Humanos , Concentración 50 Inhibidora , Metotrexato/farmacología , Pemetrexed , Ácido Poliglutámico/metabolismo , ARN Interferente Pequeño , Transfección
9.
Cancer Chemother Pharmacol ; 60(6): 883-9, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17333191

RESUMEN

PURPOSE: The DNA repair protein, O (6)-alkylguanine-DNA alkyltransferase (AGT), is a primary source of tumor resistance to agents such as temozolomide and chloroethylnitrosoureas that form DNA lesions at the O (6)-position of guanines. To increase the efficacy of these drugs, pseudosubstrate inactivators of AGT such as O (6)-benzylguanine have been developed. A novel inactivator of AGT, O (4)-benzylfolic acid (O(4)-BFA), has been reported which is more potent and water soluble than O (6)-benzylguanine. Previous studies have suggested that uptake of O(4)-BFA is mediated by the folate receptor (FR), and, thus, its use may be limited to cells expressing FR. METHODS: We measured AGT activity in cell extracts from a panel of brain tumor cells exposed to O(4)-BFA. Inactivation of AGT by O(4)-BFA was measured in cells grown without folic acid as well as in cells grown in folic acid-containing media. Competitive binding studies were performed using purified FR to determine its affinity for O(4)-BFA. RESULTS: The observed IC(50) for O(4)-BFA in brain tumor cell lines ranged from 0.2 to 1.3 microM for cells grown in media containing 2.3 microM folic acid. At this concentration, folic acid would saturate the FR and the FR would be unable to take up O(4)-BFA. When cells were grown in folic acid free media, there was at most a 50% decrease in the observed IC(50)s, indicating that the FR was not essential for O(4)-BFA uptake. Competitive binding studies using purified FR confirmed that the IC(50) for O(4)-BFA is approximately 180 times greater than folic acid, i.e., it has a very weak affinity for FR. CONCLUSION: These results indicate that O(4)-BFA has potentially broad use as an inactivator of AGT as its use is not limited to tumors expressing high levels of FR.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Proteínas Portadoras/metabolismo , Ácido Fólico/análogos & derivados , O(6)-Metilguanina-ADN Metiltransferasa/efectos de los fármacos , Receptores de Superficie Celular/metabolismo , Antineoplásicos Alquilantes/administración & dosificación , Unión Competitiva , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Receptores de Folato Anclados a GPI , Ácido Fólico/administración & dosificación , Ácido Fólico/metabolismo , Ácido Fólico/farmacología , Humanos , Concentración 50 Inhibidora , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Temozolomida
11.
Cancer Chemother Pharmacol ; 57(6): 826-34, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16170572

RESUMEN

4-Amino-pteroyl-glutamic acid (Aminotrexate; AMT) has several advantages over the related antifolate methotrexate (MTX), including greater potency, complete oral bioavailability, and greater accumulation by leukemic blasts in vitro. We compared the pharmacodynamic properties of AMT (given orally at 4 mg/m2 in two divided doses per week) and MTX (100 mg/m2 in four divided doses per week) among children with acute lymphoblastic leukemia. We find AMT and MTX to have equivalent penetration into the bone marrow compartment of these patients, as indicated by the steady-state concentrations within mature red blood cells (RBCs). However, MTX concentrations in the cerebrospinal fluid after oral dosage are significantly greater than AMT. To confirm these clinical observations, mice were treated four weekly injections of AMT or MTX, at a 1:20 dosage ratio, and tissue antifolate content was then determined over the subsequent 22 days. We confirm the selective exclusion of AMT from the CNS compartment, while showing equivalent accumulation of AMT and MTX in the RBCs, liver, spleen, kidneys and testes. Finally, we demonstrate that AMT, MTX, and their predominant polyglutamate species are equipotent inhibitors of their target intracellular enzyme dihydrofolate reductase, emphasizing the critical nature of steady-state tissue accumulation in determining the relative cytotoxic potency of these two antifolates.


Asunto(s)
Aminopterina/farmacocinética , Antimetabolitos Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Antagonistas del Ácido Fólico/farmacocinética , Metotrexato/farmacocinética , Aminopterina/administración & dosificación , Aminopterina/líquido cefalorraquídeo , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/líquido cefalorraquídeo , Protocolos de Quimioterapia Combinada Antineoplásica/líquido cefalorraquídeo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Niño , Preescolar , Eritrocitos/metabolismo , Humanos , Lactante , Masculino , Metotrexato/administración & dosificación , Metotrexato/líquido cefalorraquídeo , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Tetrahidrofolato Deshidrogenasa/metabolismo , Distribución Tisular
12.
Clin Cancer Res ; 11(5): 1884-9, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15756014

RESUMEN

PURPOSE: To evaluate the toxicity, antileukemic activity, and pharmacology of raltitrexed administered weekly for 3 weeks to patients with refractory or recurrent leukemia. EXPERIMENTAL DESIGN: Raltitrexed was administered as a 15-minute infusion for 3 consecutive weeks every 5 weeks, at doses ranging from 1.3 to 2.8 mg/m(2). The first course was used to determine the dose-limiting toxicities and maximum tolerated dose. Correlative studies included an assessment of raltitrexed pharmacokinetics and measurement of plasma 2'-deoxyuridine concentrations, a surrogate measure of thymidylate synthase inhibition. RESULTS: Twenty-one children (18 evaluable) with refractory leukemia received 25 courses of raltitrexed. The dose-limiting toxicity was reversible elevation in liver transaminases at the 2.8-mg/m(2) dose level and the maximum tolerated dose was 2.1 mg/m(2) per dose. Pharmacokinetics were best characterized by a two-compartment model with a clearance of 139 mL/min/m(2) (8.3 L/h/m(2)), a 2.4-L volume of distribution, an initial half-life (t(1/2alpha)) of 6 minutes, and a terminal half-life (t(1/2beta)) of 45 minutes. There were three objective responses. CONCLUSIONS: Raltitrexed was well tolerated when administered as a single agent to children with recurrent or refractory leukemia. We observed preliminary evidence of antileukemia activity using this weekly dosing schedule and these observations support further evaluation of raltitrexed in this population.


Asunto(s)
Antimetabolitos Antineoplásicos/efectos adversos , Antimetabolitos Antineoplásicos/farmacocinética , Leucemia/tratamiento farmacológico , Quinazolinas/efectos adversos , Quinazolinas/farmacocinética , Tiofenos/efectos adversos , Tiofenos/farmacocinética , Adolescente , Adulto , Antimetabolitos Antineoplásicos/administración & dosificación , Niño , Preescolar , Resistencia a Antineoplásicos , Femenino , Humanos , Lactante , Infusiones Intravenosas , Masculino , Dosis Máxima Tolerada , Quinazolinas/administración & dosificación , Tiofenos/administración & dosificación , Resultado del Tratamiento
13.
Clin Cancer Res ; 11(22): 8089-96, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16299240

RESUMEN

PURPOSE: To determine the antileukemic activity of weekly oral aminopterin in patients with refractory acute leukemia; to describe the pharmacodynamic properties of aminopterin; and to contrast the intracellular metabolism of aminopterin and methotrexate by patients' blasts in vitro. EXPERIMENTAL DESIGN: Forty-six patients were enrolled in three strata: children with acute lymphoblastic leukemia (ALL), adults with ALL, and patients with acute myeloid leukemia (AML). Aminopterin was given weekly, in two doses of 2 mg/m(2), 12 hours apart. Limited sampling pharmacokinetic analysis was done during the first week of therapy. Accumulation of [(3)H]aminopterin and [(3)H]methotrexate by leukemic blasts was studied in vitro. RESULTS: Six of 22 children with ALL (27%; 95% confidence interval, 8-47%) had clinically significant responses. None of those with AML and only two of 11 adults with ALL had responses meeting protocol definitions, although peripheral blast counts tended to decrease with therapy in all groups. Mucosal toxicity was minimal, even with limited use of leucovorin rescue. Complete bioavailability of aminopterin was confirmed, with a mean area under the curve of 0.52 +/- 0.03 micromol hour/L after oral dosing. No relationship between aminopterin pharmacokinetics and response was seen. In vitro, aminopterin showed more consistent metabolism by leukemic blasts to polyglutamates than methotrexate. Lineage-specific differences in the pattern of intracellular antifolylpolyglutamates were observed. CONCLUSIONS: Weekly oral aminopterin has significant activity among children with refractory ALL. With greater cellular accumulation and metabolism, more reliable bioavailability than methotrexate, and tolerable toxicity at this dose and schedule, aminopterin deserves further study as a potent alternative to methotrexate.


Asunto(s)
Aminopterina/uso terapéutico , Antagonistas del Ácido Fólico/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Administración Oral , Adolescente , Adulto , Anciano , Aminopterina/sangre , Aminopterina/farmacocinética , Área Bajo la Curva , Niño , Preescolar , Resistencia a Antineoplásicos , Femenino , Antagonistas del Ácido Fólico/farmacocinética , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/etnología , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/etnología , Resultado del Tratamiento
17.
Pharmacotherapy ; 25(5): 748-55, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15899736

RESUMEN

High-dose intravenous methotrexate is an important component of many effective chemotherapeutic regimens for childhood acute lymphoblastic leukemia (ALL). Its use has a strong pharmacologic rationale: to overcome mechanisms of resistance of the malignant cells and to achieve cytotoxic concentrations in sanctuary sites for lymphoblasts. Although therapeutic progress in ALL during the past 4 decades has been closely associated with more widespread use of intravenous methotrexate and in progressively larger doses, little data exist to clearly support the use of high-dose intravenous methotrexate over a regimen of prolonged administration of low-dose methotrexate. The implied superiority of intravenous methotrexate mainly stems from studies that used identical leucovorin rescue with low-dose methotrexate or from studies of upfront window therapy in untreated children with ALL in which single standard doses of oral methotrexate were compared with high-dose intravenous methotrexate with leucovorin rescue. The evidence favoring administration of intravenous methotrexate for children with ALL is critically reviewed. Despite its extensive use, high-dose intravenous methotrexate has not been proved conclusively to be more effective than less toxic, less labor intensive, and less costly methods of methotrexate administration.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Antagonistas del Ácido Fólico/administración & dosificación , Metotrexato/administración & dosificación , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Animales , Antimetabolitos Antineoplásicos/efectos adversos , Antimetabolitos Antineoplásicos/uso terapéutico , Niño , Ensayos Clínicos como Asunto , Esquema de Medicación , Quimioterapia Combinada , Antagonistas del Ácido Fólico/farmacocinética , Antagonistas del Ácido Fólico/uso terapéutico , Humanos , Infusiones Intravenosas , Leucovorina/uso terapéutico , Metotrexato/efectos adversos , Metotrexato/uso terapéutico
20.
Adv Drug Deliv Rev ; 56(8): 1085-97, 2004 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-15094208

RESUMEN

Folate receptor alpha (FRalpha), a glycosyl phosphatidylinositol linked protein with a great affinity for folic acid and some reduced folates such as 5-methyltetrahydrofolate and tetrahydrofolate is present on a limited number of epithelial cells, especially the kidney, placenta and choroid plexus. It is also over-expressed in many carcinomas. The receptor appears to remain membrane bound but cycles between the cell surface and an internal compartment. Its localization to detergent resistant membranes (lipid rafts) may be important to its cycling and indeed putative function to conserve folate in selected body compartments. Agents that disrupt the actin cytoskeleton such as cytochalasin D as well as phorbol myristic acid effect cycling of the receptor. The monkey kidney cell line, MA104, has proved to be a useful model for studying FRalpha cycling and 5-methyltetrahydrofolate accumulation. This chapter reviews much of this work and compares and contrasts it to studies of other cells, both normal and malignant.


Asunto(s)
Proteínas Portadoras/metabolismo , Citoplasma/metabolismo , Ácido Fólico/química , Ácido Fólico/metabolismo , Receptores de Superficie Celular/metabolismo , Tetrahidrofolatos/metabolismo , Animales , Transporte Biológico/fisiología , Proteínas Portadoras/química , Línea Celular , Receptores de Folato Anclados a GPI , Humanos , Receptores de Superficie Celular/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA