Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharm ; 16(10): 4292-4301, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31503493

RESUMEN

2-(Phosphonomethyl)-pentanedioic acid (2-PMPA) is a potent (IC50 = 300 pM) and selective inhibitor of glutamate carboxypeptidase II (GCPII) with efficacy in multiple neurological and psychiatric disease preclinical models and more recently in models of inflammatory bowel disease (IBD) and cancer. 2-PMPA (1), however, has not been clinically developed due to its poor oral bioavailability (<1%) imparted by its four acidic functionalities (c Log P = -1.14). In an attempt to improve the oral bioavailability of 2-PMPA, we explored a prodrug approach using (5-methyl-2-oxo-1,3-dioxol-4-yl)methyl (ODOL), an FDA-approved promoiety, and systematically masked two (2), three (3), or all four (4) of its acidic groups. The prodrugs were evaluated for in vitro stability and in vivo pharmacokinetics in mice and dog. Prodrugs 2, 3, and 4 were found to be moderately stable at pH 7.4 in phosphate-buffered saline (57, 63, and 54% remaining at 1 h, respectively), but rapidly hydrolyzed in plasma and liver microsomes, across species. In vivo, in a single time-point screening study in mice, 10 mg/kg 2-PMPA equivalent doses of 2, 3, and 4 delivered significantly higher 2-PMPA plasma concentrations (3.65 ± 0.37, 3.56 ± 0.46, and 17.3 ± 5.03 nmol/mL, respectively) versus 2-PMPA (0.25 ± 0.02 nmol/mL). Given that prodrug 4 delivered the highest 2-PMPA levels, we next evaluated it in an extended time-course pharmacokinetic study in mice. 4 demonstrated an 80-fold enhancement in exposure versus oral 2-PMPA (AUC0-t: 52.1 ± 5.9 versus 0.65 ± 0.13 h*nmol/mL) with a calculated absolute oral bioavailability of 50%. In mouse brain, 4 showed similar exposures to that achieved with the IV route (1.2 ± 0.2 versus 1.6 ± 0.2 h*nmol/g). Further, in dogs, relative to orally administered 2-PMPA, 4 delivered a 44-fold enhanced 2-PMPA plasma exposure (AUC0-t for 4: 62.6 h*nmol/mL versus AUC0-t for 2-PMPA: 1.44 h*nmol/mL). These results suggest that ODOL promoieties can serve as a promising strategy for enhancing the oral bioavailability of multiply charged compounds, such as 2-PMPA, and enable its clinical translation.


Asunto(s)
Microsomas Hepáticos/metabolismo , Compuestos Organofosforados/metabolismo , Profármacos/metabolismo , Administración Oral , Animales , Disponibilidad Biológica , Perros , Masculino , Ratones , Compuestos Organofosforados/administración & dosificación , Compuestos Organofosforados/química , Compuestos Organofosforados/farmacocinética , Profármacos/administración & dosificación , Profármacos/química , Profármacos/farmacocinética , Distribución Tisular
2.
Mol Pharm ; 14(10): 3248-3257, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28763226

RESUMEN

2-(Phosphonomethyl)pentanedioic acid (2-PMPA) is a potent and selective inhibitor of glutamate carboxypeptidase-II (GCPII) with efficacy in multiple neurological and psychiatric disease models, but its clinical utility is hampered by low brain penetration due to the inclusion of multiple acidic functionalities. We recently reported an improvement in the brain-to-plasma ratio of 2-PMPA after intranasal (IN) dosing in both rodents and primates. Herein, we describe the synthesis of several 2-PMPA prodrugs with further improved brain delivery of 2-PMPA after IN administration by masking of the γ-carboxylate. When compared to IN 2-PMPA in rats at 1 h post dose, γ-(4-acetoxybenzyl)-2-PMPA (compound 1) resulted in significantly higher 2-PMPA delivery to both plasma (4.1-fold) and brain (11-fold). Subsequent time-dependent evaluation of 1 also showed high brain as well as plasma 2-PMPA exposures with brain-to-plasma ratios of 2.2, 0.48, and 0.26 for olfactory bulb, cortex, and cerebellum, respectively, as well as an improved sciatic nerve to plasma ratio of 0.84. In contrast, IV administration of compound 1 resulted in similar plasma exposure of 2-PMPA versus the IN route (AUCIV: 76 ± 9 h·nmol/mL versus AUCIN: 99 ± 24 h·nmol/mL); but significantly lower nerve and brain tissue exposures with tissue-to-plasma ratios of 0.21, 0.03, 0.04, and 0.04 in nerve, olfactory bulb, cortex, and cerebellum, respectively. In primates, IN administration of 1 more than doubled 2-PMPA concentrations in the cerebrospinal fluid relative to previously reported levels following IN 2-PMPA. The results of these experiments provide a promising strategy for testing GCPII inhibition in neurological and psychiatric disorders.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Fármacos Neuroprotectores/farmacología , Compuestos Organofosforados/farmacología , Administración Intranasal , Administración Intravenosa , Animales , Líquido Cefalorraquídeo/efectos de los fármacos , Ésteres/análisis , Ésteres/química , Ésteres/farmacología , Macaca mulatta , Masculino , Fármacos Neuroprotectores/análisis , Fármacos Neuroprotectores/química , Compuestos Organofosforados/análisis , Compuestos Organofosforados/química , Profármacos/análisis , Profármacos/química , Profármacos/farmacología , Ratas , Ratas Wistar , Distribución Tisular
4.
Sci Adv ; 8(46): eabq5925, 2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36383674

RESUMEN

6-Diazo-5-oxo-l-norleucine (DON) is a glutamine antagonist that suppresses cancer cell metabolism but concurrently enhances the metabolic fitness of tumor CD8+ T cells. DON showed promising efficacy in clinical trials; however, its development was halted by dose-limiting gastrointestinal (GI) toxicities. Given its clinical potential, we designed DON peptide prodrugs and found DRP-104 [isopropyl(S)-2-((S)-2-acetamido-3-(1H-indol-3-yl)-propanamido)-6-diazo-5-oxo-hexanoate] that was preferentially bioactivated to DON in tumor while bioinactivated to an inert metabolite in GI tissues. In drug distribution studies, DRP-104 delivered a prodigious 11-fold greater exposure of DON to tumor versus GI tissues. DRP-104 affected multiple metabolic pathways in tumor, including decreased glutamine flux into the TCA cycle. In efficacy studies, both DRP-104 and DON caused complete tumor regression; however, DRP-104 had a markedly improved tolerability profile. DRP-104's effect was CD8+ T cell dependent and resulted in robust immunologic memory. DRP-104 represents a first-in-class prodrug with differential metabolism in target versus toxicity tissue. DRP-104 is now in clinical trials under the FDA Fast Track designation.


Asunto(s)
Neoplasias , Profármacos , Humanos , Profármacos/farmacología , Profármacos/uso terapéutico , Diazooxonorleucina/farmacología , Diazooxonorleucina/uso terapéutico , Glutamina/metabolismo , Linfocitos T CD8-positivos/metabolismo , Neoplasias/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico
5.
Future Med Chem ; 13(2): 157-171, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33275044

RESUMEN

In the present era of drug development, quantification of drug concentrations following pharmacokinetic studies has preferentially been performed using plasma as a matrix rather than whole blood. However, it is critical to realize the difference between measuring drug concentrations in blood versus plasma and the consequences thereof. Pharmacokinetics using plasma data may be misleading if concentrations differ between plasma and red blood cells (RBCs) because of differential binding in blood. In this review, factors modulating the partitioning of drugs into RBCs are discussed and the importance of determining RBC uptake of drugs for drug candidate selection is explored. In summary, the choice of matrix (plasma vs whole blood) is an important consideration to be factored in during drug discovery.


Asunto(s)
Sangre/metabolismo , Didesoxinucleósidos/farmacocinética , Metazolamida/farmacocinética , Nifedipino/farmacocinética , Transporte Biológico , Evaluación Preclínica de Medicamentos , Eritrocitos/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Plasma/metabolismo , Unión Proteica , Estereoisomerismo , Temperatura
6.
Curr Drug Metab ; 22(9): 735-745, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34488583

RESUMEN

BACKGROUND: Metabolomic analyses from our group and others have shown that tumors treated with glutamine antagonists (GA) exhibit robust accumulation of formylglycinamide ribonucleotide (FGAR), an intermediate in the de novo purine synthesis pathway. The increase in FGAR is attributed to the inhibition of the enzyme FGAR amidotransferase (FGAR-AT) that catalyzes the ATP-dependent amidation of FGAR to formylglycinamidine ribonucleotide (FGAM). While perturbation of this pathway resulting from GA therapy has long been recognized, no study has reported systematic quantitation and analyses of FGAR in plasma and tumors. OBJECTIVE: Herein, we aimed to evaluate the efficacy of our recently discovered tumor-targeted GA prodrug, GA-607 (isopropyl 2-(6-acetamido-2-(adamantane-1-carboxamido)hexanamido)-6-diazo-5-oxohexanoate), and demonstrate its target engagement by quantification of FGAR in plasma and tumors. METHODS: Efficacy and pharmacokinetics of GA-607 were evaluated in a murine EL4 lymphoma model followed by global tumor metabolomic analysis. Liquid chromatography-mass spectrometry (LC-MS) based methods employing the ion-pair chromatography approach were developed and utilized for quantitative FGAR analyses in plasma and tumors. RESULTS: GA-607 showed preferential tumor distribution and robust single-agent efficacy in a murine EL4 lymphoma model. While several metabolic pathways were perturbed by GA-607 treatment, FGAR showed the highest increase qualitatively. Using our newly developed sensitive and selective LC-MS method, we showed a robust >80- and >10- fold increase in tumor and plasma FGAR levels, respectively, with GA-607 treatment. CONCLUSION: These studies describe the importance of FGAR quantification following GA therapy in cancer and underscore its importance as a valuable pharmacodynamic marker in the preclinical and clinical development of GA therapies.


Asunto(s)
Desarrollo de Medicamentos/métodos , Glutamina/antagonistas & inhibidores , Glicina/análogos & derivados , Neoplasias , Ribonucleótidos , Animales , Biomarcadores Farmacológicos/análisis , Biomarcadores Farmacológicos/metabolismo , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Cromatografía Liquida/métodos , Glicina/análisis , Glicina/metabolismo , Espectrometría de Masas/métodos , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Ribonucleótidos/análisis , Ribonucleótidos/metabolismo
7.
Eur J Drug Metab Pharmacokinet ; 45(4): 427-432, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32270425

RESUMEN

In the present scenario of drug discovery, several screening filters ensure a rigorous nomination of clinical candidates. One of these screens is the determination of IC50, the concentration of drug at half-maximal inhibitory concentration, also known as a potency assay. However, various nuances pertaining to the design, execution, and interpretation of in vitro potency results suggest a sizeable opportunity for the generation of erroneous data. The focus areas of this article include: (1) examining the requirement for the addition of serum albumin in in vitro potency assays, (2) problems encountered with cell lysates, and (3) drug candidate stability concerns during in vitro potency assays/high-throughput screening. Based on this assessment, the interpretation of the data generated using cell-based systems (i.e., lysates with or without the addition of fetal bovine serum) should be carried out with caution for in vitro potency testing, and the inclusion of a correction factor for non-specific protein binding should be considered. The addition of serum albumin to a cell-free system should be restricted to drugs having high protein binding (≥ 90%). Additionally, stability assessment of analytes should be considered to avoid dubious in vitro potency outcomes due to degraded material or active metabolite(s).


Asunto(s)
Descubrimiento de Drogas , Preparaciones Farmacéuticas/metabolismo , Albúmina Sérica/metabolismo , Animales , Sistema Libre de Células , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Desarrollo de Medicamentos , Estabilidad de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Concentración 50 Inhibidora , Preparaciones Farmacéuticas/química , Unión Proteica
8.
Drug Saf ; 43(8): 711-725, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32367507

RESUMEN

The introduction of novel, small-molecule Janus kinase inhibitors namely tofacitinib, baricitinib and upadacitinib has provided an alternative treatment option for patients with rheumatoid arthritis outside of traditional drugs and expensive biologics. This review aimed to critically assess the drug-drug interaction potential of tofacitinib, baricitinib and upadacitinib and provide a balanced perspective for choosing the most appropriate Janus kinase inhibitor based on the needs of patients with rheumatoid arthritis including co-medications and renal/hepatic impairment status. Based on the critical assessment, all three approved Janus kinase inhibitors generally provide a favourable opportunity for co-prescription with a plethora of drugs. While cytochrome P450 3A4-related inhibition or induction altered the exposures (area under the curve) of tofacitinib and upadacitinib, it did not impact the exposure of baricitinib. Transporter drug-drug interaction studies revealed that the disposition of baricitinib was altered with certain transporter inhibitors as compared with either tofacitinib or upadacitinib. Adjustment of tofacitinib or baricitinib dosages but not that of upadacitinib is required with the progression of renal impairment from a mild to a severe condition. While the dosage of tofacitinib needs to be adjusted for patients with moderate hepatic impairment status, it is not the case for either baricitinib or upadacitinib. Assessment of the drug-drug interaction potential suggests that tofacitinib, baricitinib and upadacitinib generally show a favourable disposition with no perpetrator activity; however, as victim drugs, they show subtle pharmacokinetic differences that may be considered during polypharmacy. Moreover, careful choice of the three drugs could be made in patients with rheumatoid arthritis with varying degrees of renal/hepatic impairments.


Asunto(s)
Antirreumáticos/efectos adversos , Azetidinas/efectos adversos , Interacciones Farmacológicas , Compuestos Heterocíclicos con 3 Anillos/efectos adversos , Quinasas Janus/antagonistas & inhibidores , Farmacocinética , Piperidinas/efectos adversos , Inhibidores de Proteínas Quinasas/efectos adversos , Purinas/efectos adversos , Pirazoles/efectos adversos , Pirimidinas/efectos adversos , Sulfonamidas/efectos adversos , Animales , Antirreumáticos/farmacocinética , Antirreumáticos/uso terapéutico , Azetidinas/farmacocinética , Azetidinas/toxicidad , Compuestos Heterocíclicos con 3 Anillos/farmacocinética , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Humanos , Piperidinas/farmacocinética , Piperidinas/toxicidad , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/uso terapéutico , Purinas/farmacocinética , Purinas/toxicidad , Pirazoles/farmacocinética , Pirazoles/toxicidad , Pirimidinas/farmacocinética , Pirimidinas/toxicidad , Sulfonamidas/farmacocinética , Sulfonamidas/toxicidad
9.
Sci Adv ; 6(44)2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33115736

RESUMEN

Extended-release gastrointestinal (GI) luminal delivery substantially increases the ease of administration of drugs and consequently the adherence to therapeutic regimens. However, because of clearance by intrinsic GI motility, device gastroretention and extended drug release over a prolonged duration are very challenging. Here, we report that GI parasite-inspired active mechanochemical therapeutic grippers, or theragrippers, can reside within the GI tract of live animals for 24 hours by autonomously latching onto the mucosal tissue. We also observe a notable sixfold increase in the elimination half-life using theragripper-mediated delivery of a model analgesic ketorolac tromethamine. These results provide first-in-class evidence that shape-changing and self-latching microdevices enhance the efficacy of extended drug delivery.


Asunto(s)
Sistemas de Liberación de Medicamentos , Ketorolaco Trometamina , Animales , Liberación de Fármacos , Tracto Gastrointestinal , Preparaciones Farmacéuticas
10.
Mol Cancer Ther ; 19(2): 397-408, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31594823

RESUMEN

The carbon and nitrogen components of glutamine are used for multiple biosynthetic processes by tumors. Glutamine metabolism and the therapeutic potential of glutamine antagonists (GA), however, are incompletely understood in malignant peripheral nerve sheath tumor (MPNST), an aggressive soft tissue sarcoma observed in patients with neurofibromatosis type I. We investigated glutamine dependence of MPNST using JHU395, a novel orally bioavailable GA prodrug designed to circulate inert in plasma, but permeate and release active GA within target tissues. Human MPNST cells, compared with Schwann cells derived from healthy peripheral nerve, were selectively susceptible to both glutamine deprivation and GA dose-dependent growth inhibition. In vivo, orally administered JHU395 delivered active GA to tumors with over 2-fold higher tumor-to-plasma exposure, and significantly inhibited tumor growth in a murine flank MPNST model without observed toxicity. Global metabolomics studies and stable isotope-labeled flux analyses in tumors identified multiple glutamine-dependent metabolites affected, including prominent effects on purine synthesis. These data demonstrate that glutamine antagonism is a potential antitumor strategy for MPNST.


Asunto(s)
Glutamina/antagonistas & inhibidores , Neoplasias de la Vaina del Nervio/tratamiento farmacológico , Profármacos/farmacología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Med Chem ; 63(11): 6028-6056, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32298582

RESUMEN

Neutral sphingomyelinase 2 (nSMase2) catalyzes the cleavage of sphingomyelin to phosphorylcholine and ceramide, an essential step in the formation and release of exosomes from cells that is critical for intracellular communication. Chronic increase of brain nSMase2 activity and related exosome release have been implicated in various pathological processes, including the progression of Alzheimer's disease (AD), making nSMase2 a viable therapeutic target. Recently, we identified phenyl (R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)carbamate 1 (PDDC), the first nSMase2 inhibitor that possesses both favorable pharmacodynamics and pharmacokinetic (PK) parameters, including substantial oral bioavailability, brain penetration, and significant inhibition of exosome release from the brain in vivo. Herein we demonstrate the efficacy of 1 (PDDC) in a mouse model of AD and detail extensive structure-activity relationship (SAR) studies with 70 analogues, unveiling several that exert similar or higher activity against nSMase2 with favorable pharmacokinetic properties.


Asunto(s)
Inhibidores Enzimáticos/química , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Animales , Peso Corporal/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Exosomas/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Piridazinas/química , Piridazinas/metabolismo , Piridazinas/uso terapéutico , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielina Fosfodiesterasa/farmacología , Relación Estructura-Actividad
12.
J Med Chem ; 62(7): 3524-3538, 2019 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-30892035

RESUMEN

6-Diazo-5-oxo-l-norleucine (DON) is a glutamine antagonist with robust anticancer efficacy; however, its therapeutic potential was hampered by its biodistribution and toxicity to normal tissues, specifically gastrointestinal (GI) tissues. To circumvent DON's toxicity, we synthesized a series of tumor-targeted DON prodrugs designed to circulate inert in plasma and preferentially activate over DON in tumor. Our best prodrug 6 (isopropyl 2-(6-acetamido-2-(adamantane-1-carboxamido)hexanamido)-6-diazo-5-oxohexanoate) showed stability in plasma, liver, and intestinal homogenates yet was readily cleaved to DON in P493B lymphoma cells, exhibiting a 55-fold enhanced tumor cell-to-plasma ratio versus that of DON and resulting in a dose-dependent inhibition of cell proliferation. Using carboxylesterase 1 knockout mice that were shown to mimic human prodrug metabolism, systemic administration of 6 delivered 11-fold higher DON exposure to tumor (target tissue; AUC0- t = 5.1 nmol h/g) versus GI tissues (toxicity tissue; AUC0- t = 0.45 nmol h/g). In summary, these studies describe the discovery of a glutamine antagonist prodrug that provides selective tumor exposure.


Asunto(s)
Antineoplásicos/administración & dosificación , Diazooxonorleucina/administración & dosificación , Sistemas de Liberación de Medicamentos , Lisina/química , Profármacos/química , Acetilación , Animales , Área Bajo la Curva , Hidrolasas de Éster Carboxílico/genética , Línea Celular Tumoral , Diazooxonorleucina/farmacocinética , Humanos , Ratones , Ratones Noqueados , Porcinos
13.
Eur J Med Chem ; 170: 276-289, 2019 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-30921693

RESUMEN

Neutral sphingomyelinase 2 (nSMase2), a key enzyme in ceramide biosynthesis, is a new therapeutic target for the treatment of neurological disorders and cancer. Using 2,6-dimethoxy-4-[4-phenyl-5-(2-thienyl)-1H-imidazol-2-yl]phenol (DPTIP), our initial hit compound (IC50 = 30 nM) from nSMase2 screening efforts, as a molecular template, a series of 4-(1H-imidazol-2-yl)-2,6-dialkoxyphenol derivatives were designed, synthesized, and evaluated. Systematic examination of various regions of DPTIP identified the key pharmacophore required for potent nSMase2 inhibition as well as a number of compounds with the 4-(1H-imidazol-2-yl)-2,6-dialkoxyphenol scaffold with similar or higher inhibitory potency against nSMase2 as compared to DPTIP. Among them, 4-(4,5-diisopropyl-1H-imidazol-2-yl)-2,6-dimethoxyphenol (25b) was found to be metabolically stable against P450 metabolism in liver microsomes and displayed higher plasma exposure following oral administration as compared to DPTIP. Analysis of plasma samples identified an O-glucuronide as the major metabolite. Blockade of the phase II metabolism should further facilitate our efforts to identify potent nSMase2 inhibitors with desirable ADME properties.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Imidazoles/química , Imidazoles/farmacología , Fenoles/química , Fenoles/farmacología , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Animales , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Humanos , Imidazoles/metabolismo , Imidazoles/farmacocinética , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Fenoles/metabolismo , Fenoles/farmacocinética , Esfingomielina Fosfodiesterasa/metabolismo
14.
Br J Pharmacol ; 176(19): 3857-3870, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31273753

RESUMEN

BACKGROUND AND PURPOSE: Extracellular vesicles (EVs) are constitutively shed from cells and released by various stimuli. Their protein and RNA cargo are modified by the stimulus, and in disease conditions can carry pathological cargo involved in disease progression. Neutral sphingomyelinase 2 (nSMase2) is a major regulator in at least one of several independent routes of EV biogenesis, and its inhibition is a promising new therapeutic approach for neurological disorders. Unfortunately, known inhibitors exhibit µM potency, poor physicochemical properties, and/or limited brain penetration. Here, we sought to identify a drug-like inhibitor of nSMase2. EXPERIMENTAL APPROACH: We conducted a human nSMase2 high throughput screen (>365,000 compounds). Selected hits were optimized focusing on potency, selectivity, metabolic stability, pharmacokinetics, and ability to inhibit EV release in vitro and in vivo. KEY RESULTS: We identified phenyl(R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)-carbamate (PDDC), a potent (pIC50  = 6.57) and selective non-competitive inhibitor of nSMase2. PDDC was metabolically stable, with excellent oral bioavailability (%F = 88) and brain penetration (AUCbrain /AUCplasma  = 0.60). PDDC dose-dependently (pEC50  = 5.5) inhibited release of astrocyte-derived extracellular vesicles (ADEV). In an in vivo inflammatory brain injury model, PDDC robustly inhibited ADEV release and the associated peripheral immunological response. A closely related inactive PDDC analogue was ineffective. CONCLUSION AND IMPLICATIONS: PDDC is a structurally novel, potent, orally available, and brain penetrant inhibitor of nSMase2. PDDC inhibited release of ADEVs in tissue culture and in vivo. PDDC is actively being tested in animal models of neurological disease and, along with closely related analogues, is being considered for clinical translation.


Asunto(s)
Encéfalo/efectos de los fármacos , Vesículas Extracelulares/efectos de los fármacos , Animales , Astrocitos/química , Astrocitos/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Vesículas Extracelulares/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Ratones , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
15.
J Neuroimmune Pharmacol ; 14(3): 391-400, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31209775

RESUMEN

HIV-associated neurocognitive disorders (HAND) have been linked to dysregulation of glutamate metabolism in the central nervous system (CNS) culminating in elevated extracellular glutamate and disrupted glutamatergic neurotransmission. Increased glutamate synthesis via upregulation of glutaminase (GLS) activity in brain immune cells has been identified as one potential source of excess glutamate in HAND. However, direct evidence for this hypothesis in an animal model is lacking, and the viability of GLS as a drug target has not been explored. In this brief report, we demonstrate that GLS inhibition with the glutamine analogue 6-diazo-5-oxo-L-norleucine (DON) can reverse cognitive impairment in the EcoHIV-infected mouse model of HAND. However, due to peripheral toxicity DON is not amenable to clinical use in a chronic disease such as HAND. We thus tested JHU083, a novel, brain penetrant DON prodrug predicted to exhibit improved tolerability. Systemic administration of JHU083 reversed cognitive impairment in EcoHIV-infected mice similarly to DON, and simultaneously normalized EcoHIV-induced increases in cerebrospinal fluid (CSF) glutamate and GLS activity in microglia-enriched brain CD11b + cells without observed toxicity. These studies support the mechanistic involvement of elevated microglial GLS activity in HAND pathogenesis, and identify JHU083 as a potential treatment option. Graphical Abstract Please provide Graphical Abstract caption.Glutamine Antagonist JHU083 Normalizes Aberrant Glutamate Production and Cognitive Deficits in the EcoHIV Murine Model of HIV-Associated Neurocognitive Disorders .


Asunto(s)
Complejo SIDA Demencia , Compuestos Azo/uso terapéutico , Caproatos/uso terapéutico , Trastornos del Conocimiento/tratamiento farmacológico , Glutamatos/biosíntesis , Glutamina/antagonistas & inhibidores , Profármacos/uso terapéutico , Animales , Compuestos Azo/farmacocinética , Antígeno CD11b/análisis , Caproatos/farmacocinética , Trastornos del Conocimiento/líquido cefalorraquídeo , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/virología , Condicionamiento Clásico/efectos de los fármacos , Miedo , Glutamatos/líquido cefalorraquídeo , VIH-1/genética , VIH-1/patogenicidad , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/patogenicidad , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Norleucina/análogos & derivados , Norleucina/uso terapéutico , Profármacos/farmacocinética , Virus Reordenados/genética , Virus Reordenados/patogenicidad , Aprendizaje Espacial/efectos de los fármacos
16.
J Med Chem ; 61(9): 3918-3929, 2018 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-29648826

RESUMEN

Mebendazole (MBZ) was developed as a broad-spectrum anthelmintic but has recently shown efficacy as an anticancer agent. The use of MBZ for cancer, however, is challenging due to its poor solubility leading to poor bioavailability. Herein, we developed a prodrug approach with various N-linked promoieties including acyloxymethyl, aminoacyloxymethyl, and substituted phosphonooxymethyl in attempt to improve these characteristics. Compound 12, containing an (((((isopropoxycarbonyl)oxy)methoxy)phosphoryl)oxy)methyl promoiety, showed a >10 000-fold improvement in aqueous solubility. When evaluated in mice, 12 displayed a 2.2-fold higher plasma AUC0- t and a 1.7-fold improvement in brain AUC0- t with a calculated oral bioavailability of 52%, as compared to 24% for MBZ-polymorph C (MBZ-C), the most bioavailable polymorph. In dogs, 12 showed a 3.8-fold higher plasma AUC0- t with oral bioavailability of 41% compared to 11% for MBZ-C. In summary, we have identified a prodrug of MBZ with better physicochemical properties and enhanced bioavailability in both mice and dog.


Asunto(s)
Antihelmínticos/metabolismo , Mebendazol/metabolismo , Nitrógeno/química , Profármacos/química , Profármacos/farmacocinética , Agua/química , Administración Oral , Animales , Disponibilidad Biológica , Perros , Estabilidad de Medicamentos , Masculino , Ratones , Profármacos/administración & dosificación , Profármacos/metabolismo , Solubilidad , Relación Estructura-Actividad , Distribución Tisular
17.
J Med Chem ; 60(18): 7799-7809, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28759215

RESUMEN

4-Carboxy-α-[3-(hydroxyamino)-3-oxopropyl]-benzenepropanoic acid 1 is a potent hydroxamate-based inhibitor of glutamate carboxypeptidase II. In an attempt to improve its poor oral pharmacokinetics, we synthesized a series of prodrugs by masking its hydrophilic hydroxamate group. Prodrugs were evaluated for oral availability in mice and showed varying degree of plasma exposure to 1. Of these, para-acetoxybenzyl-based, 4-(5-(((4-acetoxybenzyl)oxy)amino)-2-carboxy-5-oxopentyl)benzoic acid, 12, provided 5-fold higher plasma levels of 1 compared to oral administration of 1 itself. Subsequently, para-acetoxybenzyl-based prodrugs with additional ester promoiety(ies) on carboxylate(s) were examined for their ability to deliver 1 to plasma. Isopropyloxycarbonyloxymethyl (POC) ester 30 was the only prodrug that achieved substantial plasma levels of 1. In vitro metabolite identification studies confirmed stability of the ethyl ester of benzoate while the POC group was rapidly hydrolyzed. At oral daily dose-equivalent of 3 mg/kg, 12 exhibited analgesic efficacy comparable to dose of 10 mg/kg of 1 in the rat chronic constrictive injury model of neuropathic pain.


Asunto(s)
Analgésicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Ácidos Hidroxámicos/uso terapéutico , Neuralgia/tratamiento farmacológico , Profármacos/uso terapéutico , Administración Oral , Analgésicos/química , Analgésicos/farmacocinética , Analgésicos/farmacología , Animales , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Esterificación , Glutamato Carboxipeptidasa II/metabolismo , Humanos , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacocinética , Ácidos Hidroxámicos/farmacología , Masculino , Ratones , Neuralgia/enzimología , Profármacos/química , Profármacos/farmacocinética , Profármacos/farmacología , Ratas , Ratas Sprague-Dawley
18.
J Med Chem ; 60(16): 7186-7198, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28759224

RESUMEN

Aberrant excitatory neurotransmission associated with overproduction of glutamate has been implicated in the development of HIV-associated neurocognitive disorders (HAND). The glutamine antagonist 6-diazo-5-oxo-l-norleucine (DON, 14) attenuates glutamate synthesis in HIV-infected microglia/macrophages, offering therapeutic potential for HAND. We show that 14 prevents manifestation of spatial memory deficits in chimeric EcoHIV-infected mice, a model of HAND. 14 is not clinically available, however, because its development was hampered by peripheral toxicities. We describe the synthesis of several substituted N-(pivaloyloxy)alkoxy-carbonyl prodrugs of 14 designed to circulate inert in plasma and be taken up and biotransformed to 14 in the brain. The lead prodrug, isopropyl 6-diazo-5-oxo-2-(((phenyl(pivaloyloxy)methoxy)carbonyl)amino)hexanoate (13d), was stable in swine and human plasma but liberated 14 in swine brain homogenate. When dosed systemically in swine, 13d provided a 15-fold enhanced CSF-to-plasma ratio and a 9-fold enhanced brain-to-plasma ratio relative to 14, opening a possible clinical path for the treatment of HAND.


Asunto(s)
Aminocaproatos/farmacología , Compuestos Azo/farmacología , Diazooxonorleucina/farmacología , Trastornos Neurocognitivos/tratamiento farmacológico , Nootrópicos/farmacología , Profármacos/farmacología , Aminocaproatos/administración & dosificación , Aminocaproatos/síntesis química , Animales , Compuestos Azo/administración & dosificación , Compuestos Azo/síntesis química , Sangre/metabolismo , Encéfalo/metabolismo , Diazooxonorleucina/administración & dosificación , Estabilidad de Medicamentos , Femenino , Ácido Glutámico/metabolismo , Glutaminasa/antagonistas & inhibidores , Infecciones por VIH/complicaciones , Humanos , Masculino , Ratones Endogámicos C57BL , Trastornos Neurocognitivos/etiología , Nootrópicos/administración & dosificación , Nootrópicos/síntesis química , Profármacos/administración & dosificación , Profármacos/síntesis química , Porcinos , Carga Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA