Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 14(3): 3773-3783, 2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-35014790

RESUMEN

Folic acid was reported to significantly improve chondrogenic differentiation of mesenchymal stem cells. In a similar mechanism of action, we investigated clinically approved antifolates by the U.S. Food and Drug Administration as chondrogenic-promoting compounds for tonsil-derived mesenchymal stem cells. A poly(ethylene glycol)-poly(l-alanine) thermogelling system was used as a three-dimensional cell culture matrix, where stem cells and antifolates could be incorporated simultaneously during a heat-induced in situ sol-to-gel transition. The antifolates could be supplied over several days by the sustained release of the drug from the thermogel. Initially, seven antifolates were prescreened based on cell viability and expression of a typical chondrogenic biomarker of type II collagen (COL II) at the mRNA level. Then, dapsone, pralatrexate, and trimethoprim were selected as candidate compounds in the second round screening, and detailed studies were carried out on the mRNA and protein expression of various chondrogenic biomarkers including COL II, SRY box transcription factor 9, and aggrecan. Three-dimensional cultures of stem cells in the thermogel in the absence of a chondrogenic promoter compound and in the presence of kartogenin (KGN) were performed as a negative control and positive control, respectively. The chondrogenic biomarkers were significantly increased in the selected antifolate-incorporating systems compared to the negative control system, without an increase in type I collagen (an osteogenic biomarker) expression. Pralatrexate was the best compound for inducing chondrogenic differentiation of the stem cells, even better than the positive control (KGN). Nuclear translocation of the core-binding factor ß subunit (CBFß) and enhanced nuclear runt-related transcription factor 1 (RUNX1) by antifolate treatment suggested that the chondrogenesis-enhancing mechanism is mediated by CBFß and RUNX1. An in silico modeling study confirmed the mechanism by proving the high binding affinity of pralatrexate to a target protein of filamin A compared with other antifolate candidates. To conclude, pralatrexate was rediscovered as a lead compound, and the polypeptide thermogel incorporating pralatrexate and mesenchymal stem cells can be a very effective system in promoting chondrogenic differentiation of stem cells and might be used in injectable tissue engineering for cartilage repair.


Asunto(s)
Aminopterina/análogos & derivados , Materiales Biocompatibles/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Péptidos/química , Temperatura , Aminopterina/química , Aminopterina/farmacología , Materiales Biocompatibles/química , Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Geles/química , Humanos , Ensayo de Materiales
2.
Cells ; 10(8)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34440885

RESUMEN

Since activated macrophages express a functional folate receptor ß (FRß), targeting this macrophage population with folate-linked drugs could increase selectivity to treat inflammatory diseases. Using a macrophage-mediated anti-glomerular basement membrane (anti-GBM) glomerulonephritis (GN) in WKY rats, we investigated the effect of a novel folic acid-aminopterin (AMT) conjugate (EC2319) designed to intracellularly deliver AMT via the FR. We found that treatment with EC2319 significantly attenuated kidney injury and preserved renal function. Kidney protection with EC2319 was blocked by a folate competitor, indicating that its mechanism of action was specifically FRß-mediated. Notably, treatment with methotrexate (MTX), another folic acid antagonist related to AMT, did not protect from kidney damage. EC2319 reduced glomerular and interstitial macrophage infiltration and decreased M1 macrophage recruitment but not M2 macrophages. The expression of CCL2 and the pro-fibrotic cytokine TGF-ß were also reduced in nephritic glomeruli with EC2319 treatment. In EC2319-treated rats, there was a significant decrease in the deposition of collagens. In nephritic kidneys, FRß was expressed on periglomerular macrophages and macrophages present in the crescents, but its expression was not observed in normal kidneys. These data indicate that selectively targeting the activated macrophage population could represent a novel means for treating anti-GBM GN and other acute crescentic glomerulonephritis.


Asunto(s)
Receptor 2 de Folato/metabolismo , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Macrófagos/metabolismo , Aminopterina/química , Aminopterina/uso terapéutico , Animales , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Ácido Fólico/química , Ácido Fólico/uso terapéutico , Macrófagos/efectos de los fármacos , Metotrexato/uso terapéutico , Ratas
3.
PLoS Comput Biol ; 16(12): e1008489, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33382685

RESUMEN

The spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus poses serious threats to the global public health and leads to worldwide crisis. No effective drug or vaccine is readily available. The viral RNA-dependent RNA polymerase (RdRp) is a promising therapeutic target. A hybrid drug screening procedure was proposed and applied to identify potential drug candidates targeting RdRp from 1906 approved drugs. Among the four selected market available drug candidates, Pralatrexate and Azithromycin were confirmed to effectively inhibit SARS-CoV-2 replication in vitro with EC50 values of 0.008µM and 9.453 µM, respectively. For the first time, our study discovered that Pralatrexate is able to potently inhibit SARS-CoV-2 replication with a stronger inhibitory activity than Remdesivir within the same experimental conditions. The paper demonstrates the feasibility of fast and accurate anti-viral drug screening for inhibitors of SARS-CoV-2 and provides potential therapeutic agents against COVID-19.


Asunto(s)
Aminopterina/análogos & derivados , Antivirales/farmacología , Evaluación Preclínica de Medicamentos/métodos , Reposicionamiento de Medicamentos , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , SARS-CoV-2/fisiología , Aminopterina/química , Aminopterina/farmacología , Animales , Azitromicina/química , Azitromicina/farmacología , Chlorocebus aethiops , Simulación por Computador , Aprendizaje Profundo , Simulación de Dinámica Molecular , ARN Polimerasa Dependiente del ARN/química , Células Vero , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
4.
Bioorg Med Chem ; 28(2): 115247, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31843461

RESUMEN

Rheumatoid arthritis (RA) is a chronic inflammatory disease that causes severe joints damage and other extra-articular alterations. Despite the efficacy of low-dose methotrexate (LD-MTX) in RA treatment, adverse effects are the predominant reasons for discontinuation of therapy. As a therapeutic targeting strategy, the presence of increased concentrations of reactive oxygen species (ROS) in the inflammatory environment can serve as the stimulus for prodrug activation in site-selective drug delivery systems. Our group has previously reported novel ROS sensitive prodrugs (1-3) of MTX and aminopterin (AMT) for site-selective delivery to inflammatory tissue associated with RA, with the aim of reducing side effects in RA therapy. Herein, we investigate the effect and toxicity of the same prodrugs in a rat CIA (collagen-induced arthritis) model of RA. We find that prodrug 1, an arylboronic acid ROS-sensitive MTX-prodrug, displays similar in vivo efficacy as MTX at an equimolar dose, while avoiding adverse effects known to restrict MTX treatment. To further characterize prodrug 1 and its ROS mediated activation, we synthesized compound 4, a negative control lacking the boronic acid moiety. We then investigated the effect of molecules on cell proliferation and cytotoxicity in the presence of the ROS scavenger pyruvate, as well as their stability in buffer and cell media, demonstrating a direct correlation between ROS concentration and the prodrug activity. Moreover, the in vitro ADME properties were investigated, including permeability, rat plasma and microsomal stability.


Asunto(s)
Aminopterina/farmacología , Antirreumáticos/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Metotrexato/farmacología , Profármacos/farmacología , Aminopterina/administración & dosificación , Aminopterina/química , Animales , Antirreumáticos/administración & dosificación , Antirreumáticos/química , Apoptosis/efectos de los fármacos , Artritis Experimental/inducido químicamente , Artritis Experimental/metabolismo , Artritis Reumatoide/inducido químicamente , Artritis Reumatoide/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/metabolismo , Inyecciones Intraperitoneales , Metotrexato/administración & dosificación , Metotrexato/química , Estructura Molecular , Profármacos/administración & dosificación , Profármacos/química , Ratas , Especies Reactivas de Oxígeno/análisis , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad
5.
Nanoscale ; 11(32): 15224-15233, 2019 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-31385577

RESUMEN

One of the frontiers of nanomedicine is the rational design of theranostic nanovectors. These are nanosized materials combining diagnostic and therapeutic capabilities, i.e. capable of tracking cancer cells and tissues in complex environments, and of selectively acting against them. We herein report on the preparation and application of antifolate plasmonic nanovectors, made of functionalized gold nanoparticles conjugated with the folic acid competitors aminopterin and methotrexate. Due to the overexpression of folate binding proteins on many types of cancer cells, these nanosystems can be exploited for selective cancer cell targeting. The strong surface enhanced Raman scattering (SERS) signature of these nanovectors acts as a diagnostic tool, not only for tracing their presence in biological samples, but also, through a careful spectral analysis, to precisely quantify the amount of drug loaded on a single nanoparticle, and therefore delivered to the cells. Meanwhile, the therapeutic action is implemented based on the strong toxicity of antifolate drugs. Remarkably, supplying the drug in the nanostructured form, rather than as a free molecule, enhances its specific toxicity. The selectivity of the antifolate nanovectors can be optimized by the design of a hybrid folate/antifolate coloaded nanovector for the specific targeting of folate receptor α, which is overexpressed on numerous cancer cell types.


Asunto(s)
Antagonistas del Ácido Fólico/química , Nanoestructuras/química , Espectrometría Raman , Nanomedicina Teranóstica , Aminopterina/química , Aminopterina/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ácido Fólico/química , Ácido Fólico/farmacología , Antagonistas del Ácido Fólico/farmacología , Oro/química , Humanos , Nanopartículas del Metal/química , Metotrexato/química , Metotrexato/farmacología
6.
Future Oncol ; 9(1): 21-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23252560

RESUMEN

Pralatrexate is a folate analogue metabolic inhibitor manufactured by Allos Therapeutics, Inc., a wholly-owned subsidiary of Spectrum Pharmaceuticals, Inc. In both preclinical and clinical studies, pralatrexate demonstrated activity in lymphoma. Pralatrexate was US FDA approved for the treatment of relapsed/refractory peripheral T-cell lymphoma in 2009. Approval was based on data from the PROPEL trial that demonstrated an overall response rate of 29% in a heavily pretreated patient population. The dose and schedule of pralatrexate is 30-mg/m(2) weekly for 6 weeks, given in 7-week cycles. Folate and vitamin B12 supplementation are required to minimize toxicity. The most common toxicities are mucositis, thrombocytopenia, nausea and fatigue.


Asunto(s)
Aminopterina/análogos & derivados , Antineoplásicos/uso terapéutico , Linfoma de Células T/tratamiento farmacológico , Aminopterina/efectos adversos , Aminopterina/química , Aminopterina/farmacocinética , Aminopterina/uso terapéutico , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/química , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Recurrencia
7.
Bioorg Med Chem Lett ; 21(4): 1202-5, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21236665

RESUMEN

Efficient syntheses of folate receptor (FR) targeting conjugates of the anti-inflammatory, aminopterin hydrazide, are described. 2-{4-Benzoylamino}-5-oxo-5-{N'-[2-(pyridin-2-yldisulfanyl)-ethoxycarbonyl]-hydrazino}-pentanoic acid is synthesized from commercially available 4-[(2-amino-4-imino-3,4-dihydro-pteridin-6-yl-methyl)-amino]-benzoic acid. Conjugation of this novel, activated aminopterin hydrazide to folic acid through cysteine-terminating (C-terminus), peptide/carbohydrate spacers results in highly water soluble conjugates which allow for the release of free aminopterin hydrazide within the endosomes of targeted cells.


Asunto(s)
Aminopterina/química , Antiinflamatorios/química , Ácido Fólico/análogos & derivados , Aminopterina/síntesis química , Aminopterina/uso terapéutico , Antiinflamatorios/síntesis química , Antiinflamatorios/uso terapéutico , Diseño de Fármacos , Ácido Fólico/síntesis química , Ácido Fólico/química , Ácido Fólico/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Estereoisomerismo
8.
Clin Cancer Res ; 16(20): 4921-7, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20739433

RESUMEN

PURPOSE: On September 24, 2009, the U.S. Food and Drug Administration granted accelerated approval for Folotyn (pralatrexate injection, Allos Therapeutics, Inc.) as a single agent for the treatment of patients with relapsed or refractory peripheral T-cell lymphoma (PTCL); it is the first drug approved for this indication. EXPERIMENTAL DESIGN: This review was based on study PDX-008, a phase II, single-arm, nonrandomized, open-label, international, multicenter trial, designed to evaluate the safety and efficacy of pralatrexate when administered concurrently with vitamin B(12) and folic acid supplementation in patients with relapsed or refractory PTCL. RESULTS: The overall response rate was 27% in 109 evaluable patients [95% confidence interval (CI), 19-36%]. Twelve percent of 109 evaluable patients (95% CI, 7-20%)] had a response duration of ≥14 weeks. Six of these 13 patients achieved a complete response, and one patient had complete response unconfirmed. The most common grade 3 and 4 toxicities were thrombocytopenia, mucositis, and neutropenia. CONCLUSION: This accelerated approval was based on a response rate that is reasonably likely to predict clinical benefit in this heavily pretreated patient population with this rare disease. The applicant has committed to conducting postmarketing clinical trials to assess clinical benefit. The recommended starting dose of pralatrexate in patients with relapsed or refractory PTCL is 30 mg/m(2) via intravenous push over 3 to 5 min weekly for 6 weeks followed by a one-week rest (one cycle). Intramuscular injection of 1 mg vitamin B(12) should be administered every 8 to 10 weeks along with 1.0 mg folic acid given orally once a day.


Asunto(s)
Aminopterina/análogos & derivados , Linfoma de Células T/tratamiento farmacológico , Anciano , Aminopterina/efectos adversos , Aminopterina/química , Aminopterina/uso terapéutico , Aprobación de Drogas , Femenino , Antagonistas del Ácido Fólico/efectos adversos , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/uso terapéutico , Humanos , Masculino , Persona de Mediana Edad , Estados Unidos , United States Food and Drug Administration
9.
Cancer Chemother Pharmacol ; 65(6): 1125-30, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19784838

RESUMEN

PURPOSE: Renewed interest in antifols for the treatment of childhood cancers has resulted from identification of novel antifols with broad spectrums of anti-cancer activity and from re-evaluation of the original clinical antifol, aminopterin. In this pre-clinical study we evaluated the in vitro activity of both traditional antifols (methotrexate, aminopterin) and novel antifols (pemetrexed, talotrexin) in childhood acute leukemias and lymphomas. METHODS: We compared the in vitro cytotoxicity of methotrexate, aminopterin, pemetrexed, and talotrexin in a panel of six pediatric leukemia and lymphoma cell lines using the sulforhodamine B assay. In addition to defining a 50% growth inhibitory concentration (IC50) for a 120-h drug exposure, we contrasted the activity of the drugs in the context of clinically achievable (tolerable) drug exposures using the area under the plasma concentration-time curve (AUC). We defined each agent's clinical potency index (CPI) as the AUC achieved with standard pediatric dosing regimens divided by the in vitro IC50. RESULTS: Across all cell lines, talotrexin (median IC50 7 nM) and aminopterin (median IC50 17 nM) had lower IC50's than methotrexate (median IC50 78 nM) and pemetrexed (median IC50 155 nM). However, the CPI for methotrexate (median 0.9) was significantly greater than that for aminopterin (median 0.4). In contrast, pemetrexed had a significantly better CPI (median 13) than the traditional antifols. CONCLUSIONS: Aminopterin does not appear to offer any advantage over methotrexate for the treatment of childhood ALL. Further study of pemetrexed in childhood leukemias is warranted.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Antagonistas del Ácido Fólico/farmacología , Aminopterina/química , Aminopterina/farmacología , Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Niño , Relación Dosis-Respuesta a Droga , Antagonistas del Ácido Fólico/química , Glutamatos/química , Glutamatos/farmacología , Guanina/análogos & derivados , Guanina/química , Guanina/farmacología , Humanos , Concentración 50 Inhibidora , Células Jurkat , Leucemia/patología , Linfoma/patología , Metotrexato/química , Metotrexato/farmacología , Estructura Molecular , Ornitina/análogos & derivados , Ornitina/química , Ornitina/farmacología , Pemetrexed , Pterinas/química , Pterinas/farmacología
10.
Biochem Pharmacol ; 77(7): 1161-72, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19174154

RESUMEN

4-Amino-4-deoxy-5,8,10-trideazapteroyl-d,l-4'-methyleneglutamic acid (CH-1504) is the prototype of a potentially therapeutically more selective class of antifolates for rheumatoid arthritis treatment. This class is characterized by retention of dihydrofolate reductase (DHFR; EC 1.5.1.3) as their locus of action and transport by the reduced folate carrier (RFC; SLC19A1), but their lack of metabolism by known pathways of antifolate (e.g., methotrexate (MTX)) metabolism. Five new CH-1504 analogs (CHL-001-CHL-005) were synthesized and diastereomers of CH-1504 itself were obtained by preparative chiral HPLC; all were characterized biochemically. The analogs are not metabolized by aldehyde oxidase (EC 1.2.3.1), carboxypeptidase G2 (EC 3.4.17.11), or (excepting CHL-003) folylpolyglutamate synthetase (EC 6.3.2.17) and thus, unlike MTX, are "metabolism-blocked". All analogs are potent DHFR inhibitors; several are nearly as potent as MTX or CH-1504. Each analog uses the RFC for transport, although with varying apparent affinities. In contrast, each weakly inhibits other enzymes of folate metabolism relevant to rheumatoid arthritis therapy (thymidylate synthase (EC 2.1.1.45), two formyltransferases of purine biosynthesis (EC 2.1.2.2 and EC 2.1.2.3), and 5,10-methylenetetrahydrofolate reductase (EC 1.5.1.20)). Biochemical characterization showed one 4'-diastereomer of racemic CH-1504 was significantly more active than the other. Based on literature data concerning the effect of d- and l-glutamic acid substitution on antifolate activity, it is likely that the diastereomer containing l-4'-methylene-glutamic acid is the more active. Because of concern about potential pharmacokinetic and biochemical effects of d-4'-methylene-glutamic acid-containing species, these data suggest that future analogs should contain only l-4'-methylene-glutamic acid. Overall, these data provide several interesting new leads for preclinical development.


Asunto(s)
Aminopterina/análogos & derivados , Antirreumáticos/uso terapéutico , Antagonistas del Ácido Fólico/uso terapéutico , Redes y Vías Metabólicas/fisiología , Aminopterina/química , Aminopterina/farmacología , Aminopterina/uso terapéutico , Animales , Antirreumáticos/química , Antirreumáticos/farmacología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Línea Celular Tumoral , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/farmacología , Humanos , Insectos , Redes y Vías Metabólicas/efectos de los fármacos , Conejos
11.
J Med Chem ; 49(2): 770-88, 2006 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-16420062

RESUMEN

Ester prodrugs of the phosphinate pseudopeptide N-[(4-deoxy-4-amino-10-methyl)pteroyl]glutamate-gamma-[psiP(O)(OH)]-glutarate (1a) were synthesized. H-phosphinic acids derived from N-Cbz vinyl glycine esters were converted to the desired pseudopeptides by Michael addition to alpha-methyleneglutarate esters. Pivaloyloxymethyl (POM) ester moieties were incorporated in both the N-terminal and C-terminal fragments prior to formation of either C-P bond. N-Alkylation of the corresponding amides derived from p-(N-methyl)aminobenzoic acid with 2,4-diamino-6-(bromomethyl)pteridine gave the target compounds. POM esters of methotrexate and the corresponding gamma-glutamyl conjugate were also synthesized using the same strategy. All prodrugs were evaluated in Chinese hamster ovary cells. Although the pseudopeptide prodrugs were ineffective, prodrugs of methotrexate and the corresponding gamma-glutamyl conjugate were equipotent with the parent compounds. Stability of the prodrugs was investigated in both phosphate buffer and cell line medium to provide a rationale for the observed biological data.


Asunto(s)
Aminopterina/análogos & derivados , Antineoplásicos/síntesis química , Péptido Sintasas/antagonistas & inhibidores , Ácidos Fosfínicos/síntesis química , Profármacos/síntesis química , Aminopterina/síntesis química , Aminopterina/química , Aminopterina/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Células CHO , Cricetinae , Cricetulus , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Ésteres/síntesis química , Ésteres/química , Ésteres/farmacología , Hidrólisis , Cinética , Metotrexato/administración & dosificación , Metotrexato/química , Metotrexato/farmacología , Ácidos Pentanoicos/química , Ácidos Fosfínicos/química , Ácidos Fosfínicos/farmacología , Profármacos/química , Profármacos/farmacología , Relación Estructura-Actividad
12.
J Biol Chem ; 280(46): 38457-63, 2005 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-16162503

RESUMEN

Cyanobacterial and plant genomes encode proteins with some similarity to the folate and biopterin transporters of the trypanosomatid parasite Leishmania. The Synechocystis slr0642 gene product and its closest Arabidopsis homolog, the At2g32040 gene product, are representative examples. Both have 12 probable transmembrane domains, and the At2g32040 protein has a predicted chloroplast transit peptide. When expressed in Escherichia coli pabA pabB or folE, mutants, which are unable to produce or take up folates, the slr0642 protein and a modified At2g32040 protein (truncated and fused to the N terminus of slr0642) enabled growth on 5-formyltetrahydrofolate or folic acid but not on 5-formyltetrahydrofolate triglutamate, demonstrating that both proteins mediate folate monoglutamate transport. Both proteins also mediate transport of the antifolate analogs methotrexate and aminopterin, as evidenced by their ability to greatly increase the sensitivity of E. coli to these inhibitors. The full-length At2g32040 polypeptide was translocated into isolated pea chloroplasts and, when fused to green fluorescent protein, directed the passenger protein to the envelope of Arabidopsis chloroplasts in transient expression experiments. At2g32040 transcripts were present at similar levels in roots and aerial organs, indicating that the protein occurs in non-green plastids as well as chloroplasts. Insertional inactivation of At2g32040 significantly raised the total folate content of chloroplasts and lowered the proportion of 5-methyltetrahydrofolate but did not discernibly affect growth. These findings establish conservation of function among folate and biopterin transporter family proteins from three kingdoms of life.


Asunto(s)
Cianobacterias/metabolismo , Plastidios/metabolismo , Trypanosoma/metabolismo , Aminopterina/química , Animales , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/fisiología , Transporte Biológico , Biopterinas/metabolismo , Membrana Celular/metabolismo , Clorofila/química , Cloroplastos/metabolismo , Clonación Molecular , Secuencia Conservada , Cianobacterias/química , Transportadores de Ácidos Dicarboxílicos/fisiología , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Escherichia coli/metabolismo , Ácido Fólico/química , Ácido Fólico/metabolismo , Genoma de Planta , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/metabolismo , Leucovorina/química , Metotrexato/farmacología , Modelos Biológicos , Modelos Químicos , Mutación , Pisum sativum/metabolismo , Péptidos/química , Estructura Terciaria de Proteína , Transporte de Proteínas , ARN Mensajero/metabolismo , Synechocystis/metabolismo
13.
Biochem Pharmacol ; 65(9): 1427-33, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12732354

RESUMEN

Thirteen structural analogs of the potent nonpolyglutamatable dihydrofolate reductase inhibitor N(alpha)-(4-amino-4-deoxypteroyl)-N(delta)-hemiphthaloyl-L-ornithine (PT523) with modifications in the side chain, the para-aminobenzoyl moiety, or the 9,10-bridge were evaluated for the ability to inhibit human recombinant dihydrofolate reductase (DHFR), to utilize the reduced folate carrier (RFC) for influx, and to inhibit the growth of CCRF-CEM human leukemia cells in culture. In spectrophotometric assays of the kinetics of the reduction of dihydrofolate by DHFR in the presence of NADPH, these compounds had K(i) values ranging from 0.2 to 1.3pM, and thus were not greatly different in potency from the parent drug PT523. By comparison, the K(i) values of aminopterin (AMT), methotrexate (MTX), and 10-ethyl-10-deazaaminopterin (EDX) were 3.7, 4.8, and 11pM. In assays of competitive inhibition of [3H]MTX influx into CCRF-CEM cells, the K(i) values ranged from 0.21 to 7.3 micro M, as compared with 0.71, 5.4, and 1.1 micro M for PT523, AMT, and EDX. The K(t) for MTX was also re-analyzed and found to be 4.7 micro M, in better agreement with the literature than our previously reported value of 7.1 micro M. The IC(50) values of these compounds as inhibitors of the growth of CCRF-CEM cells after 72hr of drug exposure ranged from 0.53 to 55nM, and were qualitatively consistent with the other results.


Asunto(s)
Aminopterina/farmacología , Antineoplásicos/farmacología , Proteínas Portadoras/metabolismo , Proteínas de Transporte de Membrana , Ornitina/análogos & derivados , Tetrahidrofolato Deshidrogenasa/metabolismo , Aminoácidos/química , Aminopterina/análogos & derivados , Aminopterina/química , Antineoplásicos/química , Ácidos Carboxílicos/química , Proteínas Portadoras/antagonistas & inhibidores , Humanos , Ornitina/química , Ornitina/farmacología , Pterinas/química , Pterinas/farmacología , Proteína Portadora de Folato Reducido , Células Tumorales Cultivadas
14.
Anticancer Drugs ; 13(6): 615-23, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12172507

RESUMEN

The antifolate aminopterin (AMPT) was developed before methotrexate (MTX), but was not clinically established or generally used due its increased toxicity compared to MTX. Recently, we reported on the increased metabolism of albumin conjugates such as methotrexate-albumin (MTX-SA) in malignant tumors and the feasibility of using albumin as a carrier for drug targeting. Consequently, AMPT was covalently bound to serum albumin (AMPT-SA) at a 1:1 molar ratio. Biodistribution, tolerability and efficacy of this novel conjugate were studied in Walker-256 (W-256) carcinoma-bearing rats. As compared to native albumin, the same biodistribution and plasma clearance were found for AMPT-SA, which achieved 20.1% tumor uptake (estimated uptake per g tumor 6.4%) within 24 h after i.v. administration in rats. In a randomized study, AMPT-SA, repeatedly i.v. injected, was compared with low-molecular-weight AMPT. Depending on the molar concentration, the maximum tolerated dose (MTD) of AMPT covalently bound to SA was twice that of unbound AMPT (three repeated injections of 1.0 mg AMPT-SA/kg body weight versus three repeated injections of 0.5 mg AMPT/kg body weight; p=0.0006). Efficacy was studied at the level of the MTD and MTD/2, and demonstrated that AMPT-SA was significantly more active. At the MTD/2 in W-256 carcinoma-bearing rats, AMPT-SA achieved a 100% volume reduction and an optimal volume reduction during treatment/control (T/C) of 8.3% compared to a 53% volume reduction of AMPT and a T/C of 16.5% (p=0.032). Tumor relapses were reduced and occurred later in the AMPT-SA group (two tumor recurrences for AMPT-SA versus seven for AMPT; p=0.05). In this comparative study, the AMPT-SA conjugate showed high antitumor activity in vivo and a favorable toxicity compared to low-molecular-weight AMPT. These effects are attributed to the albumin carrier which seems to be an effective tool for selective tumor drug targeting.


Asunto(s)
Aminopterina/uso terapéutico , Antimetabolitos Antineoplásicos/uso terapéutico , Carcinoma 256 de Walker/tratamiento farmacológico , Antagonistas del Ácido Fólico/uso terapéutico , Aminopterina/química , Aminopterina/toxicidad , Animales , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/toxicidad , Carcinoma 256 de Walker/diagnóstico por imagen , Quelantes , Sistemas de Liberación de Medicamentos , Femenino , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/toxicidad , Trasplante de Neoplasias , Ácido Pentético , Cintigrafía , Ratas , Ratas Sprague-Dawley , Albúmina Sérica/química , Distribución Tisular , Células Tumorales Cultivadas
15.
Eur J Med Chem ; 36(3): 237-42, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11337102

RESUMEN

N-[4-[[2,4-diamino-6-pteridinyl)methyl]amino]bicyclo[2.2.2]octane-1-carbonyl]-L-glutamic acid (1) was synthesized and tested for antifolate activity. N-(4-Aminobicyclo[2.2.2]octane-1-carbonyl-L-glutamic acid dimethyl ester (6), the side chain precursor to subject compound 1, was synthesized readily via reported bicyclo[2.2.2]octane-1,4-dicarboxylic acid monoethyl ester (2). The side chain precursor 6 was alkylated by 6-(bromomethyl)-2,4-pteridinediamine (7). Subsequent ester hydrolysis then afforded 1. Antifolate and antitumor evaluation of 1 verses L1210 dihydrofolate reductase (DHFR) and three tumor cell lines (L1210, S180, and HL60) showed it to be ineffective. Although compound 1 was very similar to aminopterin structurally, the bicyclo[2.2.2]octane ring system in place of the phenyl ring in the p-aminobenzoate moiety effectively negates the stoichiometric binding displayed by many classical DHFR inhibitors bearing appropriate aromatic ring systems in the side chain.


Asunto(s)
Aminopterina/química , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/farmacología , Aminopterina/farmacología , División Celular/efectos de los fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Antagonistas del Ácido Fólico/síntesis química , Humanos , Relación Estructura-Actividad , Tetrahidrofolato Deshidrogenasa/efectos de los fármacos , Células Tumorales Cultivadas
16.
Pharmacol Ther ; 85(3): 191-205, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10739874

RESUMEN

N(alpha)-(4-Amino-4-deoxypteroyl)-N(delta)-hemiphthaloyl-L-o rnithine (PT523) is an unusually tight-binding dihydrofolate reductase (DHFR) inhibitor and is efficiently taken up into cells via the reduced folate carrier (RFC). Unlike classical DHFR inhibitors with a glutamate side chain, such as methotrexate and aminopterin, PT523 cannot form polyglutamates. Thus, it resembles lipophilic antifolates such as trimetrexate in not requiring metabolic activation by folylpolyglutamate synthetase in order to produce its antifolate effect. However, in contrast to trimetrexate, PT523 retains growth inhibitory activity in cells with the multidrug resistance phenotype. As part of the preclinical development of this drug, we have performed systematic modification of several regions of the PT523 molecule, with the aim of defining the optimal structural features for DHFR binding, influx into cells via the RFC, and the ability to inhibit cell growth. The following structure-activity correlations have emerged from this ongoing investigation, and are discussed: (1) the hemiphthaloylornithine side chain has the optimal length; (2) the preferred location of the aromatic carboxyl group is the ortho position; and (3) replacement of the phenyl ring of the para-aminobenzoic acid moiety by naphthalene, of nitrogen at the 10-position of the bridge by carbon, and of nitrogen at the 5- and/or 8-position of the B-ring by carbon are all well tolerated. Several of the second generation analogs of PT523 are more potent DHFR inhibitors and better RFC substrates than PT523 itself, and are more potent inhibitors of tumor cell growth in culture.


Asunto(s)
Antineoplásicos/farmacocinética , Antagonistas del Ácido Fólico/farmacocinética , Ornitina/análogos & derivados , Pterinas/farmacocinética , Ácido 4-Aminobenzoico/química , Ácido 4-Aminobenzoico/metabolismo , Aminopterina/análogos & derivados , Aminopterina/química , Antineoplásicos/farmacología , Sitios de Unión , División Celular , Resistencia a Múltiples Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/farmacología , Humanos , Ornitina/farmacocinética , Ornitina/farmacología , Pterinas/farmacología , Sustancias Reductoras , Relación Estructura-Actividad , Tetrahidrofolato Deshidrogenasa/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
17.
J Biol Chem ; 274(6): 3323-30, 1999 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-9920873

RESUMEN

The kinetics of xanthine oxidase has been investigated with the aim of addressing several outstanding questions concerning the reaction mechanism of the enzyme. Steady-state and rapid kinetic studies with the substrate 2,5-dihydroxybenzaldehyde demonstrated that (kcat/Km)app and kred/Kd exhibit comparable bell-shaped pH dependence with pKa values of 6.4 +/- 0.2 and 8.4 +/- 0.2, with the lower pKa assigned to an active-site residue of xanthine oxidase (possibly Glu-1261, by analogy to Glu-869 in the crystallographically known aldehyde oxidase from Desulfovibrio gigas) and the higher pKa to substrate. Early steps in the catalytic sequence have been investigated by following the reaction of the oxidized enzyme with a second aldehyde substrate, 2-aminopteridine-6-aldehyde. The absence of a well defined acid limb in this pH profile and other data indicate that this complex represents an Eox.S rather than Ered.P complex (i.e. no chemistry requiring the active-site base has taken place in forming the long wavelength-absorbing complex seen with this substrate). It appears that xanthine oxidase (and by inference, the closely related aldehyde oxidases) hydroxylates both aromatic heterocycles and aldehydes by a mechanism involving base-assisted catalysis. Single-turnover experiments following incorporation of 17O into the molybdenum center of the enzyme demonstrated that a single oxygen atom is incorporated at a site that gives rise to strong hyperfine coupling to the unpaired electron spin of the metal in the MoV oxidation state. By analogy to the hyperfine interactions seen in a homologous series of molybdenum model compounds, we conclude that this strongly coupled, catalytically labile site represents a metal-coordinated hydroxide rather than the Mo=O group and that this Mo-OH represents the oxygen that is incorporated into product in the course of catalysis.


Asunto(s)
Benzaldehídos/metabolismo , Gentisatos , Oxígeno/metabolismo , Xantina Oxidasa/metabolismo , Aminopterina/análogos & derivados , Aminopterina/química , Animales , Sitios de Unión , Catálisis , Bovinos , Hidroxibenzoatos/metabolismo , Cinética , Leche/enzimología , Oxidación-Reducción , Especificidad por Sustrato
18.
Cancer Chemother Pharmacol ; 42(4): 313-8, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9744777

RESUMEN

PURPOSE: These studies sought to evaluate the biochemical and cellular pharmacokinetic properties, cytotoxicity and antitumor efficacy of a new analogue of 10-deaza-aminopterin (PDX) against human tumors. METHODS: Studies were conducted with a group of human tumor cell lines in culture examining PDX and other folate analogues as permeants for mediated membrane transport, as inhibitors of dihdrofolate reductase and as substrates for folylpolyglutamate synthetase. These same analogues were examined for their cytotoxicity following a 3-h pulse exposure, in experiments providing a value for IC50. Other studies with these analogues were conducted in nude mice bearing subcutaneously implanted human tumors. Treatment of the mice was initiated 4 days after implantation of the tumor using a schedule of administration of one dose per day for 5 days. The tumors were measured 6 days after cessation of therapy and compared to controls for assessment of response. RESULTS: In the CCRF-CEM cell system, PDX was 2- to 3-fold less effective as an inhibitor of dihydrofolate reductase than aminopterin (AMT), methotrexate (MTX) or edatrexate (EDX) but much more effective as a permeant for one-carbon, reduced folate transport inward (PDX > AMT approximately equal to EDX > MTX) and substrate for folylpolyglutamate synthetase (PDX > AMT > EDX > MTX). As predicted by these results, PDX was 15- to 40-fold more cytotoxic than MTX and 3- to 4-fold more cytotoxic than the highly potent EDX following a 3-h pulse exposure in culture of CCRF-CEM cells and cells from a panel of three human breast and two human nonsmall-cell (NSC) lung cancers. The same relative differences were shown for the therapeutic efficacy of these three analogues at equitoxic doses in studies with the human MX-1 and LX-1 tumors and the human A549 NSC lung tumor xenografted in nude mice. On a schedule of qd x 5 given 3-4 days posttransplant, MTX was minimally active (modest tumor growth delay) against all three tumors. EDX was highly active (25-35% complete regressions and 5-10% cures) against the MX-1 and LX-1 tumors but very modestly active (no regressions) against the A549 tumor. In contrast, PDX was even more active (75-85% complete regressions and 25-30% cures) than EDX against the MX-1 and LX-1 tumors and highly active (30% complete regressions and 20% cures) against the A549 tumor. CONCLUSIONS: These studies showed significantly enhanced antitumor properties of PDX compared with MTX and EDX. Based upon these results, clinical trials of PDX in patients with metastatic breast and NSC lung cancer appear to be warranted.


Asunto(s)
Aminopterina/análogos & derivados , Antineoplásicos/farmacología , Antagonistas del Ácido Fólico/farmacología , Aminopterina/química , Aminopterina/farmacología , Animales , Antineoplásicos/farmacocinética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Antagonistas del Ácido Fólico/farmacocinética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Metotrexato/farmacología , Ratones , Ratones Desnudos , Relación Estructura-Actividad , Especificidad por Sustrato , Tetrahidrofolato Deshidrogenasa/metabolismo , Células Tumorales Cultivadas
19.
Pharm Res ; 14(8): 1058-65, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9279889

RESUMEN

PURPOSE: Low-dose methotrexate (MTX) is approved for the treatment of recalcitrant rheumatoid arthritis (RA). The objective of this study was to determine the effect of vehicles and penetration enhancers on the percutaneous absorption of MTX and its analog edatrexate (EDAM), and develop transdermal (TD) delivery systems of the drugs for the treatment of RA. METHODS: From previously published pharmacokinetic parameters with low-dose MTX therapy, and considering a 50 cm2 diffusional area, the target steady state in vitro TD flux for MTX was calculated to be 35 micrograms/cm2/hr. Modified Franz diffusion chambers and hairless mouse skin were used for in vitro skin permeation studies. Hairless mice were used for in vivo studies. Delivered amounts of MTX and EDAM were determined by assaying the receiver phase fluid (or blood) with validated reversed phase HPLC methods. RESULTS: Intrinsic partition coefficient of MTX was low (log P = -1.2). Target MTX fluxes of > or = 35 micrograms/cm2/hr were achievable only with 1-15% (v/v) Azone in propylene glycol (PG). Flux of EDAM (85 micrograms/cm2/hr) was higher than MTX from an isopropyl alcohol (IPA)-5% (v/v) Azone system. Clinically significant steady state in vivo blood concentration of MTX and EDAM was achieved using delivery systems containing > or = 2.5% Azone in PG. Area under the drug concentration-time curves (AUC0-24 hr) for MTX were 2379 and 3534 ng*hr/ml from PG-2.5% Azone and PG-7.5% Azone systems respectively. AUC0-24 hr of EDAM was 6893 ng*hr/ml using a PG-2.5% Azone system. CONCLUSIONS: Results of this study show the feasibility of using a transdermal delivery system of MTX and EDAM for the treatment of rheumatoid arthritis.


Asunto(s)
Aminopterina/análogos & derivados , Antirreumáticos/metabolismo , Metotrexato/metabolismo , Vehículos Farmacéuticos/farmacología , Absorción Cutánea/efectos de los fármacos , Administración Cutánea , Aminopterina/administración & dosificación , Aminopterina/química , Aminopterina/metabolismo , Animales , Antirreumáticos/administración & dosificación , Antirreumáticos/química , Azepinas/farmacología , Caprilatos/farmacología , Técnicas In Vitro , Metotrexato/administración & dosificación , Metotrexato/química , Ratones , Ratones Pelados , Permeabilidad/efectos de los fármacos , Solubilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...