Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Clin Pharmacol ; 64(1): 94-102, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37566903

RESUMEN

Lasmiditan is an in vitro inhibitor of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) efflux transporters. We aimed to confirm predictions from physiologically based pharmacokinetic models of lasmiditan, and assess the safety and tolerability of rosuvastatin and dabigatran co-administered with lasmiditan. In this open-label, post-marketing drug-drug interaction, phase 1 clinical trial, eligible participants were adults aged 21-70 years with a body mass index of 18.5-35.0 kg/m2 . Part 1 (P-gp, 150 mg dabigatran etexilate with 200 mg lasmiditan) and part 2 (BCRP, 10 mg rosuvastatin with 200 mg lasmiditan) employed similar designs: a single dose of probe substrate administered on day -2 with pharmacokinetic evaluation; 1-week washout; lasmiditan administered on days 8 and 9 alone; lasmiditan co-administered with a single dose of probe substrate on day 10, with pharmacokinetic evaluation of probe substrate and lasmiditan. Sixty-six participants were included in part 1 and 30 participants were included in part 2. Following dabigatran co-administration with lasmiditan, versus dabigatran alone, 90% confidence intervals for geometric least-squares (LS) mean ratios of area under the plasma concentration-time curve from time 0 extrapolated to infinity (AUC0-∞ ) and maximum observed drug concentration (Cmax ) were not contained within the non-effect boundaries (0.80 to 1.25). Dabigatran AUC0-∞ increased by 25% and Cmax increased by 22%. The median time of maximum observed drug concentration (tmax ) for dabigatran was 2.0 to 3.0 hours. Following rosuvastatin co-administration with lasmiditan, versus rosuvastatin alone, 90%CIs for geometric LS mean ratios of AUC0-∞ and Cmax were contained within non-effect boundaries (0.80-1.25). Rosuvastatin AUC0-∞ increased by 15% and Cmax increased by 7%. The median tmax for rosuvastatin was 4.0 hours. Results suggest that lasmiditan has a weak effect on P-gp substrates and no clinically relevant effect on BCRP substrates.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Neoplasias de la Mama , Adulto , Femenino , Humanos , Área Bajo la Curva , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Estudios Cruzados , Dabigatrán , Interacciones Farmacológicas , Proteínas de Neoplasias , Rosuvastatina Cálcica/farmacocinética , Adulto Joven , Persona de Mediana Edad , Anciano
2.
Clin Transl Sci ; 16(7): 1149-1162, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37212649

RESUMEN

Soticlestat is a first-in-class, selective inhibitor of cholesterol 24-hydroxylase (CH24H), which catabolizes cholesterol to 24S-hydroxycholesterol (24HC) in the brain, in phase III development for Dravet syndrome and Lennox-Gastaut syndrome treatment. This study aimed to develop a model of soticlestat pharmacokinetics (PKs) and pharmacodynamics (PDs) using 24HC plasma concentrations and CH24H enzyme occupancy (EO) time profiles. Subsequently, model-based simulations were conducted to identify dosing strategies for phase II trials in children and adults with developmental and epileptic encephalopathies (DEEs). Four phase I trials of healthy adults involving oral administration of soticlestat 15-1350 mg were used to develop the mixed-effect population PK/EO/PD model. The population PK analysis utilized 1727 observations (104 individuals), PK/EO analysis utilized 20 observations (11 individuals), and PK/PD analysis utilized 2270 observations (99 individuals). Optimal dosing strategies were identified from model-based PK, EO, and PD simulations. The PK/EO/PD model described the observed data well and comprised a two-compartment model with dose as a covariate on peripheral volume, linear elimination, and intercompartmental clearance. Transit and effect-site compartments were included to accommodate different dosage forms and the delay between plasma drug concentrations and EO. Model-based simulations indicated that soticlestat 100-300 mg twice daily may be an optimal adult dosing regimen with weight-adjusted pediatric dosing strategies identified for evaluation in phase II trials. The population PK/EO/PD model provided understanding of the soticlestat PK/PD relationship with partial delineation of sources of variability, and identified dosing strategies for phase II trials of children and adults with DEEs.


Asunto(s)
Modelos Biológicos , Humanos , Adulto , Niño , Colesterol 24-Hidroxilasa , Administración Oral
3.
Clin Pharmacol Drug Dev ; 11(3): 324-332, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34664427

RESUMEN

Mevidalen (LY3154207) is a positive allosteric modulator of the dopamine D1 receptor that enhances the affinity of dopamine for the D1 receptor. The safety, tolerability, motor effects, and pharmacokinetics of mevidalen were studied in patients with Parkinson disease. Mevidalen or placebo was given once daily for 14 days to 2 cohorts of patients (cohort 1, 75 mg; cohort 2, titration from 15 to 75 mg). For both cohorts, the median time to maximum concentration for mevidalen plasma concentration was about 2 hours, the apparent steady-state clearance was 20-25 L/h, and mevidalen plasma concentrations were similar between the 1st and 14th administration in cohort 1, indicating minimal accumulation upon repeated dosing. Mevidalen was well tolerated, and most treatment-emergent adverse events were mild. Blood pressure and pulse rate increased when taking mevidalen, but there was considerable overlap with patients taking placebo, and vital signs normalized with repeated dosing. In the Movement Disorder Society-United Parkinson's Disease Rating Scale, all patients taking mevidalen showed a better motor examination sub-score on day 6 compared to only some patients in the placebo group. These data support examining mevidalen for symptomatic treatment of patients with Parkinson disease and Lewy body dementia.


Asunto(s)
Fármacos Neuroprotectores , Enfermedad de Parkinson , Humanos , Isoquinolinas/farmacocinética , Fármacos Neuroprotectores/efectos adversos , Enfermedad de Parkinson/tratamiento farmacológico , Receptores de Dopamina D1
4.
Clin Pharmacokinet ; 60(6): 819-828, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33565026

RESUMEN

INTRODUCTION: Lasmiditan is a selective serotonin (5-HT1F) receptor agonist approved in the US for the acute treatment ofmigraine in adults. This phase I, open-label, two-cohort study assessed the pharmacokinetics (PK), safety, and tolerability of lasmiditan in patients with migraine aged 6 to < 18 years. METHODS: Cohort 1 (15 to ≤ 40 kg) and Cohort 2 (> 40 to ≤ 55 kg) received single oral doses of lasmiditan (100 mg and 200 mg, respectively).Blood samples for the assessment of PK and safety parameters were collected over a 24-h period. Follow-up was approximately 14 days after dosing. RESULTS: Eighteen patients received lasmiditan (11 in Cohort 1, 7 in Cohort 2) and 17 patients completed the study. One patient in Cohort 2 discontinued due to adverse events. Plasma concentrations peaked at 1.5-2 h post dose and then declined, with a terminal half-life of approximately 4 h in both cohorts. While the exposure to lasmiditan was generally similar between cohorts, PK parameters, such as apparent total body clearance and volume of distribution, were greater for the 200 mg cohort relative to the 100 mg cohort. No deaths or serious adverse events were reported. The frequency and severity of adverse events (including somnolence, dizziness, and fatigue) were generally mild and similar to those in adult studies. CONCLUSION: The PK results support weight-based dosing of lasmiditan in pediatric patients with migraine and no new safety or tolerability issues were identified. These findings support further investigation of lasmiditan as a potential treatment in pediatric patients with migraine. Clinical Trial Registration Numbers NCT03988088 and EMEA-002166-PIP01-17M02.


Asunto(s)
Trastornos Migrañosos , Agonistas de Receptores de Serotonina , Adolescente , Benzamidas , Niño , Estudios de Cohortes , Método Doble Ciego , Humanos , Trastornos Migrañosos/tratamiento farmacológico , Piperidinas , Piridinas , Resultado del Tratamiento
5.
Clin Pharmacol Drug Dev ; 10(4): 393-403, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33029934

RESUMEN

Activation of the brain dopamine D1 receptor has attracted attention because of its promising role in neuropsychiatric diseases. Although efforts to develop D1 agonists have been challenging, a positive allosteric modulator (PAM), represents an attractive approach with potential better drug-like properties. Phase 1 single-ascending-dose (SAD; NCT03616795) and multiple-ascending-dose (MAD; NCT02562768) studies with the D1PAM mevidalen (LY3154207) were conducted with healthy subjects. There were no treatment-related serious adverse events (AEs) in these studies. In the SAD study, 25-200 mg administered orally showed dose-proportional pharmacokinetics (PK) and acute dose-related increases in systolic blood pressure (SBP) and diastolic blood pressure DBP) and pulse rate at doses ≥ 75 mg. AE related to central activation were seen at doses ≥ 75 mg. At 25 and 75 mg, central penetration of mevidalen was confirmed by measurement of mevidalen in cerebrospinal fluid. In the MAD study, once-daily doses of mevidalen at 15-150 mg for 14 days showed dose-proportional PK. Acute dose-dependent increases in SBP, DBP, and PR were observed on initial administration, but with repeated dosing the effects diminished and returned toward baseline levels. Overall, these findings support further investigation of mevidalen as a potential treatment for a range of neuropsychiatric disorders.


Asunto(s)
Dopaminérgicos , Isoquinolinas , Receptores de Dopamina D1 , Adolescente , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Administración Oral , Regulación Alostérica/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Estudios de Cohortes , Dopaminérgicos/administración & dosificación , Dopaminérgicos/farmacocinética , Dopaminérgicos/farmacología , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Frecuencia Cardíaca/efectos de los fármacos , Isoquinolinas/administración & dosificación , Isoquinolinas/farmacocinética , Isoquinolinas/farmacología , Receptores de Dopamina D1/efectos de los fármacos , Receptores de Dopamina D1/metabolismo
6.
Psychopharmacology (Berl) ; 238(5): 1279-1289, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-30536081

RESUMEN

RATIONALE: Schizophrenia is associated with impairments in cognitive functioning yet there are no approved drugs to treat these deficits. OBJECTIVES: Based on animal models, we investigated the potential for roflumilast, a selective inhibitor of phosphodiesterase type 4 (PDE4), to improve cognition, which may act by increasing intracellular cyclic adenosine monophosphate in brain regions underlying cognitive deficits in schizophrenia. METHODS: This study consisted of a randomised, double-blind, placebo-controlled, crossover design involving 15 schizophrenia patients. In 3 treatment periods, patients were given 8 days of placebo or one of the two doses of roflumilast (100 and 250 µg daily) with 14 days of washout between treatments. The primary endpoints were dorsolateral prefrontal cortex (DLPFC) activation during a visuospatial working memory task measured with fMRI on dosing day 8 and verbal memory and working memory performance change from baseline to day 8. Least square mean change scores were calculated for behavioural outcomes; fMRI data were analysed in SPM12 with bilateral DLPFC as regions of interest. RESULTS: Verbal memory was significantly improved under 250 µg roflumilast (effect size (ES) = 0.77) compared to placebo. fMRI analyses revealed that increasing dose of roflumilast was associated with reduction of bilateral DLPFC activation during working memory compared to placebo, although this was not statistically significant (ES = 0.31 for the higher dose). Working memory was not improved (ES = 0.03). CONCLUSIONS: Results support the mechanistic validation of potential novel strategies for improving cognitive dysfunction in schizophrenia and suggest that PDE4 inhibition may be beneficial for cognitive dysfunction in schizophrenia. TRIAL REGISTRATION: NCT02079844 .


Asunto(s)
Aminopiridinas/farmacología , Benzamidas/farmacología , Inhibidores de Fosfodiesterasa 4/farmacología , Esquizofrenia/tratamiento farmacológico , Adulto , Animales , Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/fisiopatología , Estudios Cruzados , Ciclopropanos/farmacología , Método Doble Ciego , Femenino , Humanos , Masculino , Memoria Episódica , Memoria a Corto Plazo/efectos de los fármacos , Persona de Mediana Edad , Corteza Prefrontal/efectos de los fármacos , Esquizofrenia/fisiopatología
7.
J Psychopharmacol ; 35(1): 15-22, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32854568

RESUMEN

BACKGROUND: Patients with schizophrenia have significant cognitive deficits, which may profoundly impair quality of life. These deficits are also evident at the neurophysiological level with patients demonstrating altered event-related potential in several stages of cognitive processing compared to healthy controls; within the auditory domain, for example, there are replicated alterations in Mismatch Negativity, P300 and Auditory Steady State Response. However, there are no approved pharmacological treatments for cognitive deficits in schizophrenia. AIMS: Here we examine whether the phosphodiesterase-4 inhibitor, roflumilast, can improve neurophysiological deficits in schizophrenia. METHODS: Using a randomised, double-blind, placebo-controlled, crossover design study in 18 patients with schizophrenia, the effect of the phosphodiesterase-4 inhibitor, roflumilast (100 µg and 250 µg) on auditory steady state response (early stage), mismatch negativity and theta (intermediate stage) and P300 (late stage) was examined using electroencephalogram. A total of 18 subjects were randomised and included in the analysis. RESULTS: Roflumilast 250 µg significantly enhanced the amplitude of both the mismatch negativity (p=0.04) and working memory-related theta oscillations (p=0.02) compared to placebo but not in the other (early- or late-stage) cognitive markers. CONCLUSIONS: The results suggest that phosphodiesterase-4 inhibition, with roflumilast, can improve electroencephalogram cognitive markers, which are impaired in schizophrenia, and that phosphodiesterase-4 inhibition acts at an intermediate rather than early or late cognitive processing stage. This study also underlines the use of neurophysiological measures as cognitive biomarkers in experimental medicine.


Asunto(s)
Aminopiridinas , Benzamidas , Cognición , Disfunción Cognitiva , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Potenciales Evocados/efectos de los fármacos , Esquizofrenia , Adulto , Aminopiridinas/administración & dosificación , Aminopiridinas/efectos adversos , Benzamidas/administración & dosificación , Benzamidas/efectos adversos , Cognición/efectos de los fármacos , Cognición/fisiología , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/psicología , Estudios Cruzados , Ciclopropanos/administración & dosificación , Ciclopropanos/efectos adversos , Método Doble Ciego , Monitoreo de Drogas/métodos , Electroencefalografía/efectos de los fármacos , Electroencefalografía/métodos , Femenino , Humanos , Masculino , Procesos Mentales/efectos de los fármacos , Procesos Mentales/fisiología , Inhibidores de Fosfodiesterasa 4/administración & dosificación , Inhibidores de Fosfodiesterasa 4/efectos adversos , Escalas de Valoración Psiquiátrica , Esquizofrenia/diagnóstico , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/fisiopatología , Resultado del Tratamiento
8.
Hum Psychopharmacol ; 35(5): e2732, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32449213

RESUMEN

OBJECTIVE: To evaluate the impact of lasmiditan, an oral, centrally-penetrant, selective serotonin 1F (5-HT1F ) receptor agonist developed for the acute treatment of migraine, on simulated driving. METHODS: Healthy adult volunteers enrolled in two randomized, placebo and active comparator-controlled, crossover studies. Study 1 (N = 90) tested lasmiditan (50-, 100-, 200-mg), alprazolam (1-mg), and placebo at 1.5 hr post-dose. Study 2 (N = 68) tested lasmiditan (100-, 200-mg), diphenhydramine (50-mg, administered 2 hr pre-assessments), and placebo at 8, 12 and 24 hr post-dose. Driving performance was assessed using a validated driving simulator employing a 100 km driving scenario. Standard deviation of lateral position (SDLP), a measure of lane position control, was the primary endpoint. RESULTS: Assay sensitivity was confirmed by increased SDLP for active comparators at 1.5- and 8-hr time points. Lasmiditan doses showed significant driving impairment versus placebo at 1.5 hr post-dose. Lasmiditan doses were non-inferior to placebo at 8 hr. Driving impairment was concentration-dependent at 1.5 hr but not at 8 hr. Common adverse events were central nervous system-related and mild-to-moderate in severity. CONCLUSIONS: Lasmiditan was associated with impaired simulated driving performance at 1.5 hr post-dose, but showed no clinically meaningful impairment at 8 hr post-dose.


Asunto(s)
Conducción de Automóvil , Benzamidas/efectos adversos , Piperidinas/efectos adversos , Piridinas/efectos adversos , Agonistas de Receptores de Serotonina/efectos adversos , Adulto , Benzamidas/administración & dosificación , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Piperidinas/administración & dosificación , Piridinas/administración & dosificación , Receptores de Serotonina/efectos de los fármacos , Agonistas de Receptores de Serotonina/administración & dosificación , Factores de Tiempo , Adulto Joven , Receptor de Serotonina 5-HT1F
9.
Clin Pharmacol Drug Dev ; 9(5): 629-638, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31950732

RESUMEN

Lasmiditan (LY573144/COL-144) is a high-affinity, centrally penetrant, selective 5-HT1F receptor agonist currently under investigation for acute treatment of migraine. Although lasmiditan is not known to induce vasoconstriction, it remains important to understand its effect on cardiovascular parameters because it is likely to be coadministered with ß-adrenergic receptor antagonists used for migraine prophylaxis, such as propranolol. This phase 1, single-center, open-label, fixed-sequence study evaluated the cardiovascular and pharmacokinetic effects of 200 mg lasmiditan in 44 healthy subjects receiving repeated oral doses of twice-daily 80 mg propranolol under fasting conditions. Coadministration caused statistically significant decreases in mean hourly heart rate relative to propranolol alone, but the maximum magnitude of this effect was -6.5 bpm and recovered to predose levels by 3 to 4 hours before stabilizing. Additionally, short-lived (≤2.5 hours) statistically significant increases in systolic blood pressure (8.3 mm Hg) and diastolic blood pressure (6.4 mm Hg) were observed following coadministration. Consistent with the largely nonoverlapping metabolic pathways of lasmiditan and propranolol, exposure to either drug was not affected by coadministration. Overall, compared with administration of either drug alone, coadministration was generally well tolerated.


Asunto(s)
Benzamidas/farmacocinética , Sistema Cardiovascular/efectos de los fármacos , Trastornos Migrañosos/tratamiento farmacológico , Piperidinas/farmacocinética , Piridinas/farmacocinética , Agonistas de Receptores de Serotonina/farmacocinética , Administración Oral , Antagonistas Adrenérgicos beta/administración & dosificación , Antagonistas Adrenérgicos beta/efectos adversos , Antagonistas Adrenérgicos beta/farmacología , Adulto , Anciano , Benzamidas/administración & dosificación , Benzamidas/efectos adversos , Benzamidas/farmacología , Presión Sanguínea/efectos de los fármacos , Interacciones Farmacológicas , Quimioterapia Combinada/efectos adversos , Ayuno/sangre , Femenino , Voluntarios Sanos/estadística & datos numéricos , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Trastornos Migrañosos/prevención & control , Piperidinas/administración & dosificación , Piperidinas/efectos adversos , Piperidinas/farmacología , Propranolol/administración & dosificación , Propranolol/efectos adversos , Propranolol/farmacología , Piridinas/administración & dosificación , Piridinas/efectos adversos , Piridinas/farmacología , Agonistas de Receptores de Serotonina/administración & dosificación , Agonistas de Receptores de Serotonina/efectos adversos , Agonistas de Receptores de Serotonina/farmacología
10.
J Clin Pharmacol ; 60(4): 495-504, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31745991

RESUMEN

Lasmiditan is a centrally penetrant, highly selective 5-hydroxytryptamine (serotonin) receptor 1F (5HT1F ) agonist under development as a novel therapy for acute treatment of migraine. A phase 1 randomized, placebo- and positive-controlled crossover study assessed the abuse potential of lasmiditan in adult recreational polydrug users. Following a qualification phase, subjects were randomized into treatment sequences, each consisting of 5 study treatments: placebo, alprazolam 2 mg, lasmiditan 100, 200 (lasmiditan 100 and 200 mg are proposed therapeutic doses), and 400 mg (supratherapeutic). The abuse potential of lasmiditan was investigated and compared with alprazolam and with placebo using the maximal effect score (Emax ) of the Drug-Liking Visual Analog Scale as the primary end point. Lasmiditan was not similar to placebo in drug-liking scores at all doses tested, with a maximum difference observed with the lasmiditan 400-mg dose (upper 90% confidence limit on difference in least-squares [LS] means > 14 for all lasmiditan doses). Drug-liking scores for lasmiditan 400 mg were not significantly different from alprazolam (lower 90% confidence limit on difference in LS means < 5), but drug-liking scores at lower doses (100 and 200 mg) were significantly different from alprazolam. During the treatment phase, the incidence of treatment-emergent adverse events (TEAEs) increased with increasing dose of lasmiditan; all TEAEs reported with lasmiditan treatment were mild. Subjective drug-liking effects for lasmiditan versus placebo and versus alprazolam, and the safety and tolerability profile of lasmiditan suggest that lasmiditan has a low potential for abuse.


Asunto(s)
Benzamidas/efectos adversos , Piperidinas/efectos adversos , Piridinas/efectos adversos , Agonistas de Receptores de Serotonina/efectos adversos , Administración Oral , Adolescente , Adulto , Alprazolam/efectos adversos , Alprazolam/uso terapéutico , Benzamidas/administración & dosificación , Benzamidas/farmacocinética , Estudios Cruzados , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Piperidinas/administración & dosificación , Piperidinas/farmacocinética , Piridinas/administración & dosificación , Piridinas/farmacocinética , Uso Recreativo de Drogas , Medición de Riesgo , Agonistas de Receptores de Serotonina/administración & dosificación , Agonistas de Receptores de Serotonina/farmacocinética , Trastornos Relacionados con Sustancias , Adulto Joven
11.
Neurobiol Aging ; 77: 37-43, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30776650

RESUMEN

There is ample evidence that phosphodiesterase 4 (PDE4) inhibition can improve memory performance in animal studies. In the present study, we examined the acute effects of the PDE4 inhibitor roflumilast on memory performance in healthy individuals (60-80 years of age). We tested the effects of acute roflumilast administration (100, 250, 1000 µg) in a double-blind, placebo-controlled, 4-way crossover design. Participants were first screened for their verbal word memory performance to ensure normal memory performance (within 0.5 standard deviation from norm score; n = 20) Drug effects on memory performance were tested in a verbal memory test and a spatial memory test. Reported side effects of drug treatment were registered. Roflumilast (100 µg) improved the delayed recall performance of the participants (Cohen's d, 0.69). No effects were observed in the spatial memory task. Roflumilast was well tolerated at this low dose. Although no clear adverse side effects were reported at the low dose, mild adverse events (including headache, dizziness, insomnia, and diarrhea) were reported after the 1000 µg dose. The present study provides first evidence that the PDE4 inhibitor roflumilast improves verbal memory performance in old participants. The current data encourage further development of PDE4 inhibitors for improving memory.


Asunto(s)
Aminopiridinas/administración & dosificación , Aminopiridinas/farmacología , Benzamidas/administración & dosificación , Benzamidas/farmacología , Envejecimiento Saludable/psicología , Memoria/efectos de los fármacos , Inhibidores de Fosfodiesterasa 4/administración & dosificación , Inhibidores de Fosfodiesterasa 4/farmacología , Conducta Verbal/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Aminopiridinas/efectos adversos , Benzamidas/efectos adversos , Cognición/efectos de los fármacos , Estudios Cruzados , AMP Cíclico/fisiología , Ciclopropanos/administración & dosificación , Ciclopropanos/efectos adversos , Ciclopropanos/farmacología , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Humanos , Recuerdo Mental/efectos de los fármacos , Persona de Mediana Edad , Inhibidores de Fosfodiesterasa 4/efectos adversos , Estimulación Química
12.
Mol Imaging Biol ; 20(4): 615-622, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29441434

RESUMEN

PURPOSE: Phosphodiesterase 4 (PDE4) inhibition in the brain has been reported to improve cognitive function in animal models. Therefore, PDE4 inhibitors are one of key targets potential for drug development. Investigation of brain PDE4 occupancy would help to understand the effects of PDE4 inhibition to cognitive functions. Roflumilast is a selective phosphodiesterase type 4 (PDE4) inhibitor used clinically for severe chronic obstructive pulmonary disease, but the effects to the brain have not been well investigated. In this study, we aimed to investigate whether roflumilast entered the brain and occupied PDE4 in nonhuman primates. PROCEDURES: Positron emission tomography (PET) measurements with (R)-[11C]rolipram were performed at baseline and after intravenous (i.v.) administration of roflumilast (3.6 to 200 µg/kg) in three female rhesus monkeys. Arterial blood samples were taken to obtain the input function. Protein binding was measured to obtain the free fraction (fp) of the radioligand. Total distribution volume (VT) and VT/fp were calculated as outcome measures from two tissue compartment model. Lassen plot approach was taken to estimate the target occupancy. RESULTS: The brain uptake of (R)-[11C]rolipram decreased after roflumilast administration. PDE 4 occupancy by roflumilast showed dose- and plasma concentration-dependent increase, although PDE4 occupancy did not reach 50 % even after the administration of up to 200 µg/kg of roflumilast, regardless of outcome measures, VT or VT/fp. CONCLUSIONS: This PET study showed that the brain PDE4 binding was blocked to a certain extent after i.v. administration of clinical relevant doses of roflumilast in nonhuman primates. Further clinical PET evaluation is needed to understand the relationship between PDE4 inhibition and potential improvement of cognitive function in human subjects.


Asunto(s)
Aminopiridinas/farmacología , Benzamidas/farmacología , Encéfalo/enzimología , Radioisótopos de Carbono/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Tomografía de Emisión de Positrones , Rolipram/farmacología , Aminopiridinas/sangre , Aminopiridinas/química , Aminopiridinas/farmacocinética , Animales , Benzamidas/sangre , Benzamidas/química , Benzamidas/farmacocinética , Ciclopropanos/sangre , Ciclopropanos/química , Ciclopropanos/farmacocinética , Ciclopropanos/farmacología , Femenino , Humanos , Macaca mulatta , Rolipram/sangre , Rolipram/química , Rolipram/farmacocinética
13.
Drugs R D ; 17(4): 631-643, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29103081

RESUMEN

BACKGROUND: Phosphodiesterase 10A (PDE10A) is selectively expressed in medium spiny neurons of the striatum. TAK-063 is a selective inhibitor of PDE10A in clinical development for the treatment of schizophrenia. OBJECTIVES: Safety, tolerability, and pharmacokinetics (PK) of TAK-063 were evaluated following multiple rising oral doses, and PK/adverse event (AE) models were developed to characterize the relationship between TAK-063 exposure and incidence of specific AEs. METHODS: Healthy Japanese subjects (HJS) aged 20-55 years and subjects with stable schizophrenia (SSS) aged 18-55 years were enrolled and randomized to either TAK-063 or placebo. Study medication was administered as a tablet once daily (at night) with food over a 7-day period. RESULTS: TAK-063 and placebo groups consisted of 62 and 15 subjects, respectively. A majority of subjects (71 of 77) completed the study. AEs were mostly of mild or moderate severity, and no deaths were reported. The most common AE was somnolence. For equivalent doses, the rate of extrapyramidal syndromes (EPS) was higher in SSS than in HJS. PK parameters were comparable between HJS and SSS at equivalent doses. The incidence of somnolence and EPS symptoms increased with exposure, and this was described with the PK/AE model. A maximum tolerated dose was not determined. CONCLUSIONS: Multiple doses of TAK-063 were safe and well tolerated. PK/AE models characterized the incidence of somnolence and EPS with increasing TAK-063 exposure, and simulations suggested that a once-daily dose range of up to 30 mg would be suitable for future studies. CLINICALTRIALS. GOV IDENTIFIER: NCT01879722.


Asunto(s)
Modelos Biológicos , Hidrolasas Diéster Fosfóricas/metabolismo , Pirazoles , Piridazinas , Esquizofrenia/tratamiento farmacológico , Administración Oral , Adolescente , Adulto , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Humanos , Japón , Persona de Mediana Edad , Inhibidores de Fosfodiesterasa/administración & dosificación , Inhibidores de Fosfodiesterasa/efectos adversos , Inhibidores de Fosfodiesterasa/farmacocinética , Pirazoles/administración & dosificación , Pirazoles/efectos adversos , Pirazoles/farmacocinética , Piridazinas/administración & dosificación , Piridazinas/efectos adversos , Piridazinas/farmacocinética , Esquizofrenia/metabolismo , Adulto Joven
14.
Eur J Clin Pharmacol ; 73(3): 279-288, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27999883

RESUMEN

PURPOSE: The aim of this study is to determine the pharmacokinetics (PK) and pharmacodynamics (PD) of a single 12.5- or 25-mg dose of alogliptin, a dipeptidyl peptidase-4 (DPP-4) inhibitor, in pediatric (children and adolescents) and adult subjects with type 2 diabetes mellitus (T2DM). METHODS: A randomized, open-label, multicenter study was conducted in pediatric and adult subjects. Subjects in two pediatric groups (children and adolescents) were randomized 1:1 to receive a single oral dose of alogliptin 12.5 or 25 mg, respectively; all gender- and race-matched adult subjects received alogliptin 25 mg. Blood and urine samples were collected at prespecified time points for PK/PD analyses. A PK/PD model was developed using data from the study for steady-state simulations. Safety was also assessed. RESULTS: In pediatric subjects receiving the 25-mg dose, the mean alogliptin peak plasma concentrations (Cmax) and AUC0-inf values were 26 and 23% lower, respectively, than in adults receiving the 25-mg dose, but maximum observed DPP-4 inhibition effect (Emax) and AUEC0-24 values were similar to those in adults. In pediatric subjects receiving the 12.5-mg dose, the mean alogliptin Cmax and AUC0-inf values were 58 and 54% lower, respectively, than those in adults, hence Emax and AUEC0-24 values were also lower by 11 and 17%, respectively. The PK/PD model simulated data were consistent with study results. No safety concern was found. CONCLUSIONS: A 25-mg dose of alogliptin in pediatric subjects achieved alogliptin exposures and DPP-4 inhibition similar to those in adult T2DM patients without safety concerns; therefore, this dose is recommended for a pediatric phase 3 trial.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Hipoglucemiantes/uso terapéutico , Piperidinas/uso terapéutico , Uracilo/análogos & derivados , Adolescente , Adulto , Área Bajo la Curva , Niño , Diabetes Mellitus Tipo 2/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/efectos adversos , Inhibidores de la Dipeptidil-Peptidasa IV/farmacocinética , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Femenino , Semivida , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/farmacología , Masculino , Piperidinas/efectos adversos , Piperidinas/farmacocinética , Piperidinas/farmacología , Uracilo/efectos adversos , Uracilo/farmacocinética , Uracilo/farmacología , Uracilo/uso terapéutico , Adulto Joven
15.
Psychopharmacology (Berl) ; 233(21-22): 3787-3795, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27572830

RESUMEN

RATIONALE: Schizophrenia is a complex neuropsychiatric disorder characterized, in part, by impaired dopamine signaling. TAK-063 is a selective inhibitor of phosphodiesterase 10A, a key regulator of intracellular signaling pathways that is highly expressed in the striatum. OBJECTIVE: Safety, tolerability, and pharmacokinetics of TAK-063 were evaluated in a phase 1 study. METHODS: Healthy Japanese and non-Japanese volunteers were randomized into dose cohorts of 3, 10, 30, 100, 300, and 1000 mg. Each fasting volunteer randomly received a single dose of TAK-063 or placebo. Individuals from the 100-mg cohort also received a post-washout, 100-mg dose under fed conditions. A total of 84 volunteers enrolled (14 per cohort). RESULTS: The most common drug-related adverse events (AEs) were somnolence (33.3 %), orthostatic tachycardia (19.7 %), and orthostatic hypotension (9.1 %). The three severe AEs recorded occurred at the highest doses: orthostatic hypotension (n = 1; 300 mg) and somnolence (n = 2; 1000 mg). There were no deaths, serious AEs, or discontinuations due to AEs. TAK-063 exposure increased in a dose-dependent manner. Median T max was reached 3 to 4 h postdose. Fed conditions slowed absorption (T max = 6 h) and increased oral bioavailability. Renal elimination was negligible. Safety and pharmacokinetic parameters were similar between Japanese and non-Japanese subjects. Impairments in cognitive function consistent with the effects of other sedative or hypnotic agents were detected using a validated, computerized cognition battery, CNS Vital Signs. CONCLUSIONS: TAK-063 was safe and well tolerated at doses up to 1000 mg and demonstrated a pharmacokinetic profile supporting once-daily dosing. Further evaluation of the clinical safety and efficacy of TAK-063 is warranted.


Asunto(s)
Inhibidores de Fosfodiesterasa/farmacología , Pirazoles/farmacología , Piridazinas/farmacología , Adulto , Trastornos de Somnolencia Excesiva/inducido químicamente , Relación Dosis-Respuesta a Droga , Ayuno , Femenino , Voluntarios Sanos , Humanos , Hipotensión Ortostática/inducido químicamente , Masculino , Persona de Mediana Edad , Inhibidores de Fosfodiesterasa/administración & dosificación , Inhibidores de Fosfodiesterasa/efectos adversos , Hidrolasas Diéster Fosfóricas , Pirazoles/administración & dosificación , Pirazoles/efectos adversos , Piridazinas/administración & dosificación , Piridazinas/efectos adversos , Taquicardia/inducido químicamente , Adulto Joven
16.
Neuroimage ; 141: 10-17, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27423256

RESUMEN

Phosphodiesterase 10A (PDE10A) is selectively expressed in the striatal regions in the brain and may play a role in modulating dopaminergic and glutamatergic second messenger pathways. PDE10A inhibitors are expected to be useful in treating neuropsychiatric disorders such as schizophrenia and Huntington's disease. In this study, the brain kinetics of [(11)C]T-773 in the human brain and test-retest reproducibility of the outcome measures were evaluated. Subsequently, the occupancy of a novel PDE10A inhibitor, TAK-063, was measured using [(11)C]T-773. Dynamic PET measurements were conducted three times for 12 healthy male subjects after intravenous bolus injection of [(11)C]T-773: two baseline PETs and one postdose PET (3hours) after oral administration of TAK-063 for four subjects, and one baseline PET and two postdose PET (3hours and 23hours) for eight subjects. Kinetic model analysis was performed with arterial input functions. PDE10A occupancy was calculated as the percent change of the binding specific to PDE10A (Vs) total distribution volume (VT), which was calculated as the VT of the putamen minus the VT of the cerebellum. Regional brain uptake was highest in the putamen. Time-activity curves of the brain regions were described with two tissue-compartment (2TC) models. The mean VT was 5.5±0.7 in the putamen and 2.3±0.5 in the cerebellum in the baseline PET. Absolute VT variability between the two baseline scans was less than 7%. Reproducibility of VT was excellent. PDE10A occupancy in the putamen ranged from 2.8% to 72.1% at 3hours after a single administration of 3 to 1000mg of TAK-063, and increased in a dose- and plasma concentration-dependent manner. At 23hours postdose, PDE10A occupancy in the putamen was 0 to 42.8% following administration of 3 to 100mg of TAK-063. In conclusion, [(11)C]T-773 showed good characteristics as a PET radioligand for PDE10A in the human brain.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Imagen Molecular/métodos , Hidrolasas Diéster Fosfóricas/metabolismo , Pirazoles/administración & dosificación , Pirazoles/farmacocinética , Piridazinas/administración & dosificación , Piridazinas/farmacocinética , Administración Oral , Adulto , Relación Dosis-Respuesta a Droga , Monitoreo de Drogas , Humanos , Masculino , Tasa de Depuración Metabólica/efectos de los fármacos , Persona de Mediana Edad , Inhibidores de Fosfodiesterasa/administración & dosificación , Hidrolasas Diéster Fosfóricas/efectos de los fármacos , Tomografía de Emisión de Positrones/métodos , Unión Proteica , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Distribución Tisular/efectos de los fármacos
17.
Eur J Clin Pharmacol ; 72(4): 447-57, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26725367

RESUMEN

PURPOSE: This open-label, multicenter, single-dose study characterized the pharmacokinetics and short-term safety of azilsartan medoxomil (AZL-M) in hypertensive pediatric subjects (12-16 years [cohort 1a; n = 9]; 6-11 years [cohort 2; n = 8]; 4-5 years [cohort 3; n = 3]). METHODS: Model-based simulations were performed to guide dosing, especially in 1-5-year olds, who were difficult to enroll. AZL-M was dosed according to body weight (20-60-mg tablet, cohorts 1a and 2; 0.66 mg/kg granule suspension, cohort 3). In cohort 1, gender-matched healthy adults (cohort 1b; n = 9) received AZL-M 80 mg. RESULTS: Exposure to AZL (active moiety of AZL-M), measured by dose-/body weight-normalized C max and AUC0-∞, was ∼15-30 % lower in pediatric subjects versus adults. In simulations, exposure with 0.66 mg/kg AZL-M in pediatric subjects weighing 8-25 kg approximated to AZL-M 40 mg (typical starting dose) in adults. The simulations suggest that 25-50-kg subjects require half the adult dose (10-40 mg), whereas 50-100-kg subjects can use the same dosing as adults. Adverse events were mild in intensity, apart from one moderate event (migraine). CONCLUSIONS: This dosing strategy should be safe in pediatric patients, as AZL exposure would not exceed that seen in adults with the highest approved AZL-M dose (80 mg).


Asunto(s)
Antihipertensivos/administración & dosificación , Antihipertensivos/farmacocinética , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacocinética , Hipertensión/tratamiento farmacológico , Oxadiazoles/administración & dosificación , Oxadiazoles/farmacocinética , Adolescente , Adulto , Antihipertensivos/efectos adversos , Área Bajo la Curva , Bencimidazoles/efectos adversos , Niño , Preescolar , Femenino , Voluntarios Sanos , Humanos , Masculino , Oxadiazoles/efectos adversos
18.
J Clin Pharmacol ; 56(8): 988-98, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26632101

RESUMEN

Population pharmacokinetic and exposure-response models for azilsartan medoxomil (AZL-M) and chlorthalidone (CLD) were developed using data from an 8-week placebo-controlled phase 3, factorial study of 20, 40, and 80 mg AZL-M every day (QD) and 12.5 and 25 mg CLD QD in fixed-dose combination (FDC) in subjects with moderate to severe essential hypertension. A 2-compartment model with first-order absorption and elimination was developed to describe pharmacokinetics. An Emax model for exposure-response analysis evaluated AZL-M/CLD effects on ambulatory systolic blood pressure (SBP). Estimated oral clearance and apparent volume of distribution (central compartment) were 1.47 L/h and 3.98 L for AZL, and 4.13 L/h and 62.1 L for CLD. Age as a covariate had the largest effect on AZL and CLD exposure (±20% change). Predicted maximal SBP responses (Emax ) were -15.6 and -23.9 mm Hg for AZL and CLD. Subgroup analysis identified statistically significant Emax differences for black vs nonblack subjects, whereby the reduced AZL response in black subjects was offset by greater response to CLD. The estimated Emax for AZL and CLD was generally greater in subjects with higher baseline BP. In conclusion, no dose adjustments to AZL-M or CLD are warranted based on identified covariates, and antihypertensive efficacy of AZL-M/CLD combination therapy is comparable in black and nonblack subjects.


Asunto(s)
Bencimidazoles/administración & dosificación , Bencimidazoles/sangre , Clortalidona/administración & dosificación , Clortalidona/sangre , Hipertensión/sangre , Hipertensión/tratamiento farmacológico , Oxadiazoles/administración & dosificación , Oxadiazoles/sangre , Anciano , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/sangre , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacocinética , Antihipertensivos/administración & dosificación , Antihipertensivos/sangre , Antihipertensivos/farmacocinética , Bencimidazoles/farmacocinética , Clortalidona/farmacocinética , Método Doble Ciego , Quimioterapia Combinada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oxadiazoles/farmacocinética , Resultado del Tratamiento
19.
AAPS J ; 16(3): 424-39, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24570339

RESUMEN

This study established a multiscale computational model for intraperitoneal (IP) chemotherapy, to depict the time-dependent and spatial-dependent drug concentrations in peritoneal tumors as functions of drug properties (size, binding, diffusivity, permeability), transport mechanisms (diffusion, convection), spatial-dependent tumor heterogeneities (vessel density, cell density, pressure gradient), and physiological properties (peritoneal pressure, peritoneal fluid volume). Equations linked drug transport and clearance on three scales (tumor, IP cavity, whole organism). Paclitaxel was the test compound. The required model parameters (tumor diffusivity, tumor hydraulic conductivity, vessel permeability and surface area, microvascular hydrostatic pressure, drug association with cells) were obtained from literature reports, calculation, and/or experimental measurements. Drug concentration-time profiles in peritoneal fluid and plasma were the boundary conditions for tumor domain and blood vessels, respectively. The finite element method was used to numerically solve the nonlinear partial differential equations for fluid and solute transport. The resulting multiscale model accounted for intratumoral spatial heterogeneity, depicted diffusive and convective drug transport in tumor interstitium and across blood vessels, and provided drug flux and concentration as a function of time and spatial position in the tumor. Comparison of model-predicted tumor spatiokinetics with experimental results (autoradiographic data of 3H-paclitaxel in IP ovarian tumors in mice, 6 h posttreatment) showed good agreement (1% deviation for area under curve and 23% deviations for individual data points, which were several-fold lower compared to the experimental intertumor variations). The computational multiscale model provides a tool to quantify the effects of drug-, tumor-, and host-dependent variables on the concentrations and residence time of IP therapeutics in tumors.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Inyecciones Intraperitoneales/métodos , Algoritmos , Animales , Líquido Ascítico/metabolismo , Simulación por Computador , Difusión , Absorción Intestinal , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Distribución Tisular
20.
Curr Cancer Drug Targets ; 14(1): 70-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24200079

RESUMEN

Intraperitoneal (IP) chemotherapy confers significant survival benefits in cancer patients. However, several problems, including local toxicity and ineffectiveness against bulky tumors, have prohibited it from becoming a standard of care. We have developed drug-loaded, polymeric tumor-penetrating microparticles (TPM) to address these problems. Initial studies showed that TPM provides tumor-selective delivery and is effective against ovarian SKOV3 tumors of relatively small size (<50 mg). The present study evaluated whether the TPM activity extends to other tumor types that are more bulky and have different morphologies and disease presentation. We evaluated TPM in mice bearing two IP human pancreatic tumors with different growth characteristics and morphologies (rapidly growing, large and porous Hs766T vs. slowly growing, smaller and densely packed MiaPaCa2), and at different disease stage (early stage with smaller tumors vs. late stage with larger tumors plus peritoneal carcinomatosis). Comparison of treatments with TPM or paclitaxel in Cremophor micelles, at equi-toxic doses, shows, in all tumor types: (a) higher paclitaxel levels in tumors (up to 55-fold) for TPM, (b) greater efficacy for TPM, including significantly longer survival and higher cure rate, and (c) a single dose of TPM was equally efficacious as multiple doses of paclitaxel/Cremophor. The results indicate tumor targeting property and superior antitumor activity of paclitaxel-loaded TPM are generalizable to small and large peritoneal tumors, with or without accompanying carcinomatosis.


Asunto(s)
Antineoplásicos/administración & dosificación , Portadores de Fármacos/farmacología , Paclitaxel/administración & dosificación , Neoplasias Pancreáticas/tratamiento farmacológico , Polietilenglicoles/administración & dosificación , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Portadores de Fármacos/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Inyecciones Intraperitoneales , Ratones , Ratones Desnudos , Paclitaxel/farmacocinética , Paclitaxel/farmacología , Neoplasias Pancreáticas/patología , Tamaño de la Partícula , Polietilenglicoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA