Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Clin Transl Sci ; 16(10): 2021-2032, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37596712

RESUMEN

Mitapivat, a first-in-class, oral, small-molecule, allosteric activator of the red blood cell-specific form of pyruvate kinase (PKR), was approved for the treatment of hemolytic anemia in adults with pyruvate kinase (PK) deficiency. In this phase I mass balance study in healthy males, we administered a single ~120 mg oral dose of [14 C]mitapivat and a concomitant intravenous ~0.1 mg microdose of [13 C6 ]mitapivat. We determined (1) the routes of total radioactivity excretion, including the mass balance of total radioactivity in urine and feces; (2) the pharmacokinetics of mitapivat and [13 C6 ]mitapivat in plasma and total radioactivity in whole blood and plasma; (3) the absolute oral bioavailability of mitapivat; and (4) the metabolite profiles in plasma and excreta. Mean recovery of the radioactive dose was 89.1% (49.6% in urine and 39.6% in feces). [14 C]Mitapivat was rapidly absorbed and extensively metabolized as <4% of the total radioactive dose was excreted unaltered in urine and feces. Mean absolute oral bioavailability was 72.7%. A total of 17 metabolites were identified. Mitapivat accounted for 57% and 34% of plasma radioactivity in AUC0-24 and AUC0-72 pooled samples, respectively. The remaining radioactivity was attributable to several metabolites, each representing <10% of the total radioactivity in pooled samples; none were disproportionate metabolites as defined by the US Food and Drug Administration and International Conference on Harmonisation M3 guidelines. Metabolite structures suggest that the primary metabolic pathways for [14 C]mitapivat in humans include N-dealkylation of the cyclopropylmethyl moiety, oxygenation of the quinoline-8-sulfonamide, oxidation/unsaturation, scission of the piperazine moiety, and amide hydrolysis.


Asunto(s)
Piruvato Quinasa , Quinolinas , Adulto , Masculino , Humanos , Disponibilidad Biológica , Voluntarios Sanos , Piperazinas/farmacocinética , Administración Oral
2.
Blood ; 140(19): 2053-2062, 2022 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-35576529

RESUMEN

Polymerization of deoxygenated hemoglobin S underlies the pathophysiology of sickle cell disease (SCD). In activating red blood cell pyruvate kinase and glycolysis, mitapivat (AG-348) increases adenosine triphosphate (ATP) levels and decreases the 2,3-diphosphoglycerate (2,3-DPG) concentration, an upstream precursor in glycolysis. Both changes have therapeutic potential for patients with SCD. Here, we evaluated the safety and tolerability of multiple ascending doses of mitapivat in adults with SCD with no recent blood transfusions or changes in hydroxyurea or l-glutamine therapy. Seventeen subjects were enrolled; 1 subject was withdrawn shortly after starting the study. Sixteen subjects completed 3 ascending dose levels of mitapivat (5, 20, and 50 mg, twice daily [BID]) for 2 weeks each; following a protocol amendment, the dose was escalated to 100 mg BID in 9 subjects. Mitapivat was well tolerated at all dose levels, with the most common treatment-emergent adverse events (AEs) being insomnia, headache, and hypertension. Six serious AEs (SAEs) included 4 vaso-occlusive crises (VOCs), non-VOC-related shoulder pain, and a preexisting pulmonary embolism. Two VOCs occurred during drug taper and were possibly drug related; no other SAEs were drug related. Mean hemoglobin increase at the 50 mg BID dose level was 1.2 g/dL, with 9 of 16 (56.3%) patients achieving a hemoglobin response of a ≥1 g/dL increase compared with baseline. Mean reductions in hemolytic markers and dose-dependent decreases in 2,3-DPG and increases in ATP were also observed. This study provides proof of concept that mitapivat has disease-modifying potential in patients with SCD. This trial was registered at www.clinicaltrials.gov as #NCT04000165.


Asunto(s)
Anemia de Células Falciformes , Piruvato Quinasa , Adulto , Humanos , Ácido Pirúvico , 2,3-Difosfoglicerato , Anemia de Células Falciformes/tratamiento farmacológico , Hemoglobinas , Adenosina Trifosfato
3.
Invest New Drugs ; 37(2): 271-281, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30073466

RESUMEN

Background CLR457 is an orally bioavailable pan-phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) inhibitor. Methods CLR457 anti-tumor activity and pharmacokinetics (PK) were characterized by in vitro biochemical assays and in vivo tumor xenografts. A first-in-human study was conducted to determine the maximum tolerated dose (MTD), safety, PK, and efficacy of CLR457. Successive cohorts of patients with advanced solid tumors with PI3K pathway activation received increasing CLR457 doses according to a Bayesian escalation model based on the rate of dose limiting toxicity (DLT) in the first 28-day cycle. Results CLR457 inhibited p110α, p110ß, p110δ and p110γ isoforms with an IC50 of 89 ± 29 nM, 56 ± 35 nM, 39 ± 10 nM and 230 ± 31 nM, respectively. CLR457 exhibited dose-dependent antitumor activity and interfered with glucose homeostasis in PI3K-mutant tumor xenografts. 31 patients received doses ranging from 5 to 100 mg. DLTs included grade 3 hyperglycemia and rash (3). In the 100 mg cohort (n = 11), 3 (27.3%) patients had DLTs and all patients (100%) experienced ≥ grade 3 toxicity with rash (45.5%) as the most common event. The MTD was not determined. For the entire study population, stomatitis (45.2%), diarrhea (38.7%), rash (35.5%) were the most common any grade toxicities-51.6% patients experienced ≥ Grade 3 toxicity. CLR457 was rapidly absorbed with limited accumulation and linear PK. PK modeling indicated that pharmacologically active concentrations were achieved at the highest dose tested (100 mg), though no objective responses were observed. Conclusion CLR457 clinical development was terminated due to poor tolerability and limited antitumor activity. These results emphasize the difficulty of achieving a wide therapeutic index when targeting all class I PI3K-isoforms.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Compuestos Orgánicos/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Proliferación Celular , Relación Dosis-Respuesta a Droga , Femenino , Estudios de Seguimiento , Humanos , Masculino , Dosis Máxima Tolerada , Ratones , Ratones Desnudos , Persona de Mediana Edad , Neoplasias/metabolismo , Neoplasias/patología , Pronóstico , Ratas Desnudas , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Med Chem ; 61(18): 8120-8135, 2018 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30137981

RESUMEN

Chronic myelogenous leukemia (CML) arises from the constitutive activity of the BCR-ABL1 oncoprotein. Tyrosine kinase inhibitors (TKIs) that target the ATP-binding site have transformed CML into a chronic manageable disease. However, some patients develop drug resistance due to ATP-site mutations impeding drug binding. We describe the discovery of asciminib (ABL001), the first allosteric BCR-ABL1 inhibitor to reach the clinic. Asciminib binds to the myristate pocket of BCR-ABL1 and maintains activity against TKI-resistant ATP-site mutations. Although resistance can emerge due to myristate-site mutations, these are sensitive to ATP-competitive inhibitors so that combinations of asciminib with ATP-competitive TKIs suppress the emergence of resistance. Fragment-based screening using NMR and X-ray yielded ligands for the myristate pocket. An NMR-based conformational assay guided the transformation of these inactive ligands into ABL1 inhibitors. Further structure-based optimization for potency, physicochemical, pharmacokinetic, and drug-like properties, culminated in asciminib, which is currently undergoing clinical studies in CML patients.


Asunto(s)
Descubrimiento de Drogas , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Niacinamida/análogos & derivados , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Regulación Alostérica , Animales , Perros , Proteínas de Fusión bcr-abl/genética , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , Ratones , Modelos Moleculares , Estructura Molecular , Mutación , Niacinamida/química , Niacinamida/farmacología , Fosforilación , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Pirazoles/química , Ratas , Ratas Sprague-Dawley , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nature ; 543(7647): 733-737, 2017 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-28329763

RESUMEN

Chronic myeloid leukaemia (CML) is driven by the activity of the BCR-ABL1 fusion oncoprotein. ABL1 kinase inhibitors have improved the clinical outcomes for patients with CML, with over 80% of patients treated with imatinib surviving for more than 10 years. Second-generation ABL1 kinase inhibitors induce more potent molecular responses in both previously untreated and imatinib-resistant patients with CML. Studies in patients with chronic-phase CML have shown that around 50% of patients who achieve and maintain undetectable BCR-ABL1 transcript levels for at least 2 years remain disease-free after the withdrawal of treatment. Here we characterize ABL001 (asciminib), a potent and selective allosteric ABL1 inhibitor that is undergoing clinical development testing in patients with CML and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukaemia. In contrast to catalytic-site ABL1 kinase inhibitors, ABL001 binds to the myristoyl pocket of ABL1 and induces the formation of an inactive kinase conformation. ABL001 and second-generation catalytic inhibitors have similar cellular potencies but distinct patterns of resistance mutations, with genetic barcoding studies revealing pre-existing clonal populations with no shared resistance between ABL001 and the catalytic inhibitor nilotinib. Consistent with this profile, acquired resistance was observed with single-agent therapy in mice; however, the combination of ABL001 and nilotinib led to complete disease control and eradicated CML xenograft tumours without recurrence after the cessation of treatment.


Asunto(s)
Sitio Alostérico/efectos de los fármacos , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Niacinamida/análogos & derivados , Pirazoles/farmacología , Regulación Alostérica/efectos de los fármacos , Animales , Dominio Catalítico/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dasatinib/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Quimioterapia Combinada , Proteínas de Fusión bcr-abl/química , Proteínas de Fusión bcr-abl/genética , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Mutación , Niacinamida/farmacología , Niacinamida/uso terapéutico , Pirazoles/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Antimicrob Agents Chemother ; 55(5): 2113-21, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21300835

RESUMEN

Echinocandins have become a first-line therapy for invasive candidiasis (IC). Using phase 3 trial data for patients with IC, pharmacokinetic-pharmacodynamic (PK-PD) relationships for efficacy for micafungin were examined. Micafungin exposures were estimated using a population pharmacokinetic model, and univariable and multivariable logistic regressions were used to identify factors associated with outcome, including the micafungin area under the concentration-time curve (AUC)/MIC ratio. Monte Carlo simulation was used to evaluate the probability of achieving AUC/MIC ratios associated with efficacy. Mycological and clinical success rates for evaluable cases were 89.4 and 90.9, respectively. MIC50s and MIC90s for Candida species inhibition were 0.008 and 0.5 mg/liter, respectively. The median AUC/MIC ratio was 15,511 (range, 41.28 to 98,716). Univariable analyses revealed a significant relationship between the AUC/MIC ratio and mycological response, with the worst response being among patients with lower (≤3,000) AUC/MIC ratios (P=0.005). For patients with Candida parapsilosis, AUC/MIC ratios of ≥285 were predictive of a higher mycological response (P=0.11). Multivariable logistic regression demonstrated the AUC/MIC ratio, APACHE II score, and history of corticosteroid use to be significant independent predictors of a favorable response. PK-PD target attainment analyses suggested that 76.7% and 100% of patients would achieve an AUC/MIC ratio of ≥3,000 for an MIC of 0.03 mg/liter and an AUC/MIC ratio of ≥285 for an MIC of <0.5 mg/liter, respectively. The identification of a lower AUC/MIC ratio target for C. parapsilosis than other Candida species suggests consideration of species-specific echinocandin susceptibility breakpoints and values that are lower than those currently approved by regulatory agencies.


Asunto(s)
Antifúngicos/farmacocinética , Candidemia/tratamiento farmacológico , Candidiasis/tratamiento farmacológico , Equinocandinas/farmacocinética , Lipopéptidos/farmacocinética , Antifúngicos/uso terapéutico , Candidemia/sangre , Candidiasis/sangre , Ensayos Clínicos Fase III como Asunto , Equinocandinas/uso terapéutico , Femenino , Humanos , Lipopéptidos/uso terapéutico , Masculino , Micafungina , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Método de Montecarlo , Ensayos Clínicos Controlados Aleatorios como Asunto
7.
Exp Cell Res ; 313(12): 2586-96, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17553490

RESUMEN

Phosphatidylinositol 3-kinase inhibitors have been shown to affect endocytosis or subsequent intracellular sorting in various receptor systems. Agonist-activated beta(2)-adrenergic receptors undergo desensitization by mechanisms that include the phosphorylation, endocytosis and degradation of receptors. Following endocytosis, most internalized receptors are sorted to the cell surface, but some proportion is sorted to lysosomes for degradation. It is not known what governs the ratio of receptors that recycle versus receptors that undergo degradation. To determine if phosphatidylinositol 3-kinases regulate beta(2)-adrenergic receptor trafficking, HEK293 cells stably expressing these receptors were treated with the phosphatidylinositol 3-kinase inhibitors LY294002 or wortmannin. We then studied agonist-induced receptor endocytosis and postendocytic sorting, including recycling and degradation of the internalized receptors. Both inhibitors amplified the internalization of receptors after exposure to the beta-agonist isoproterenol, which was attributable to the sorting of a significant fraction of receptors to an intracellular compartment from which receptor recycling did not occur. The initial rate of beta(2)-adrenergic receptor endocytosis and the default rate of receptor recycling were not significantly altered. During prolonged exposure to agonist, LY294002 slowed the degradation rate of beta(2)-adrenergic receptors and caused the accumulation of receptors within rab7-positive vesicles. These results suggest that phosphatidylinositol 3-kinase inhibitors (1) cause a misrouting of beta(2)-adrenergic receptors into vesicles that are neither able to efficiently recycle to the surface nor sort to lysosomes, and (2) delays the movement of receptors from late endosomes to lysosomes.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Cromonas/farmacología , Endocitosis/efectos de los fármacos , Morfolinas/farmacología , Receptores Adrenérgicos beta 2/metabolismo , Agonistas Adrenérgicos beta/farmacología , Línea Celular , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Transporte de Proteínas/efectos de los fármacos , Transferrina/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
8.
J Neurochem ; 98(2): 495-505, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16749908

RESUMEN

Nociceptin (N/OFQ) is an endogenous neuropeptide that plays a role in the behavioral deficits associated with Parkinson's disease (PD). The purpose of the present study was to characterize the protective effects of prepro (pp)N/OFQ gene deletion against two dopamine toxins, MPTP and methamphetamine (METH). Results demonstrate that ppN/OFQ gene deletion attenuates the loss of both the number of tyrosine hydroxylase (TH)-positive neurons in the substantia nigra pars compacta (SNpc) and loss of TH and vesicular monoamine transporter-2 (VMAT) immunoreactivity in the caudate putamen (CPu) of MPTP-treated mice. This protection was unaffected by age or gender, although, when loss of TH exceeded 90% in 5-6 month-old mice, the protective effect was greatly diminished. In contrast, METH administration preferentially damaged dopaminergic terminals in the CPu with little effect on dopamine neurons in the SNpc, an effect not reversed by ppN/OFQ gene deletion. To determine if N/OFQ and MPP+ act directly and synergistically on dopamine neurons, differentiated SH-SY5Y cells were incubated with N/OFQ and/or MPP+. N/OFQ did not increase MPP+-mediated cell loss, suggesting an indirect action of N/OFQ. These studies demonstrate that inhibition of the endogenous N/OFQ system may represent a new therapeutic target for prevention of neuronal loss associated with PD.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/antagonistas & inhibidores , Dopaminérgicos/toxicidad , Inhibidores de Captación de Dopamina/antagonistas & inhibidores , Inhibidores de Captación de Dopamina/toxicidad , Dopamina/fisiología , Metanfetamina/antagonistas & inhibidores , Metanfetamina/toxicidad , Neuronas/efectos de los fármacos , Precursores de Proteínas/fisiología , Receptores Opioides/fisiología , Envejecimiento/fisiología , Animales , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Femenino , Eliminación de Gen , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Precursores de Proteínas/genética , Receptores Opioides/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Caracteres Sexuales
9.
Br J Pharmacol ; 147(3): 249-59, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16331289

RESUMEN

Activated beta2-adrenoceptors are rapidly desensitized by phosphorylation of Ser262 by protein kinase A (PKA) and of Ser355,356 by G-protein-coupled receptor kinase (GRK). We sought to determine whether the phosphorylation and subsequent dephosphorylation of these sites had similar kinetics and requirements for receptor endocytosis. The phosphorylation of the PKA and GRK sites were measured using antibodies that recognize phosphoserine 262 and phosphoserine 355,356. Endocytosis in stably transfected HEK293 cells was blocked by inducible expression of dominant-negative dynamin-1 K44A or by treatment with hypertonic sucrose. The phosphorylation of the GRK site Ser355,356 during a 10 microM isoprenaline treatment rapidly reached a steady state, and the extent of kinetics of phosphorylation were unaffected by dynamin-1 K44A expression, and minimally by hypertonic sucrose. In contrast, phosphorylation of the PKA site Ser262 during a 10 microM isoprenaline treatment peaked after 2 min and then rapidly declined, while inhibition of endocytosis enhanced and prolonged phosphorylation. Treatment with 300 pM isoprenaline, a concentration too low to provoke endocytosis, also resulted in prolonged PKA site phosphorylation. The dephosphorylation of these sites was measured after removal of agonist. Significant dephosphorylation of phosphoserines 262 and 355,356 was observed under conditions of very low endocytosis, however dephosphorylation of the GRK site was greater if antagonist was present after removal of agonist. The results indicate that the kinetics of beta2-adrenoceptor GRK and PKA site phosphorylation are distinct and differently affected by endocytosis, and that receptor dephosphorylation can occur either at the plasma membrane or in internal compartments.


Asunto(s)
Receptores Adrenérgicos beta 2/metabolismo , Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Dinamina I/análisis , Endocitosis/efectos de los fármacos , Quinasa 2 del Receptor Acoplado a Proteína-G , Quinasa 3 del Receptor Acoplado a Proteína-G , Humanos , Fosforilación , Receptores Adrenérgicos beta 2/química , Serina/metabolismo , Quinasas de Receptores Adrenérgicos beta/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...