Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Pharmacol Drug Dev ; 11(12): 1367-1381, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36331497

RESUMEN

Migalastat is approved for the treatment of Fabry disease (FD) with amenable variants. Objectives were to characterize effects of estimated glomerular filtration rate (eGFR) on oral clearance (CL), predict doses in mild to moderate renal impairment and in pediatric patients with FD, and to improve designs of FD studies. A 2-compartment model was fit to data from 260 subjects with/without FD and iteratively refined with evolving data. FD, eGFR, and weight affected CL, while weight and FD affected volume. Optimal sampling theory was used to choose pharmacokinetic sampling times for pediatric studies. Doses in patients with renal impairment and in pediatrics were determined by targeting exposure in adults receiving migalastat 123 mg every other day. A clinical study was conducted in 20 adolescent patients with FD ≥45 kg. eGFR had the largest effect on CL. Simulations showed that exposures in moderate renal impairment were within phase 2-3 exposures; patients aged 2-17 years require weight-based dosing; and predicted exposures in adolescent patients ≥45 kg receiving migalastat 123 mg every other day were similar to adults (data confirmed in a clinical study). Model-informed drug development optimized dosing and design of clinical studies and supported that no dose adjustments were needed in patients with mild to moderate renal impairment or in adolescent patients ≥45 kg.


Asunto(s)
Enfermedad de Fabry , Insuficiencia Renal , Adulto , Humanos , Adolescente , Niño , 1-Desoxinojirimicina/efectos adversos , Enfermedad de Fabry/tratamiento farmacológico , Tasa de Filtración Glomerular , Insuficiencia Renal/tratamiento farmacológico
2.
Clin Pharmacol Drug Dev ; 10(9): 1075-1088, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33876577

RESUMEN

Approved therapies for Fabry disease (FD) include migalastat, an oral pharmacological chaperone, and agalsidase beta and agalsidase alfa, 2 forms of enzyme replacement therapy. Broad tissue distribution may be beneficial for clinical efficacy in FD, which has severe manifestations in multiple organs. Here, migalastat and agalsidase beta biodistribution were assessed in mice and modeled using physiologically based pharmacokinetic (PBPK) analysis, and migalastat biodistribution was subsequently extrapolated to humans. In mice, migalastat concentration was highest in kidneys and the small intestine, 2 FD-relevant organs. Agalsidase beta was predominantly sequestered in the liver and spleen (organs unaffected in FD). PBPK modeling predicted that migalastat 123 mg every other day resulted in concentrations exceeding the in vitro half-maximal effective concentration in kidneys, small intestine, skin, heart, and liver in human subjects. However, extrapolation of mouse agalsidase beta concentrations to humans was unsuccessful. In conclusion, migalastat may distribute to tissues that are inaccessible to intravenous agalsidase beta in mice, and extrapolation of mouse migalastat concentrations to humans showed adequate tissue penetration, particularly in FD-relevant organs.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Isoenzimas/farmacocinética , Modelos Biológicos , alfa-Galactosidasa/farmacocinética , 1-Desoxinojirimicina/farmacocinética , Adulto , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Especificidad de la Especie , Distribución Tisular , Adulto Joven , alfa-Galactosidasa/genética
3.
Orphanet J Rare Dis ; 14(1): 36, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30736861

RESUMEN

Rare or orphan diseases often are inherited and overwhelmingly affect children. Many of these diseases have no treatments, are incurable, and have a devastating impact on patients and their families. Regulatory standards for drug approval for rare diseases must ensure that patients receive safe and efficacious treatments. However, regulatory bodies have shown flexibility in applying these standards to drug development in rare diseases, given the unique challenges that hinder efficient and effective traditional clinical trials, including low patient numbers, limited understanding of disease pathology and progression, variability in disease presentation, and a lack of established endpoints.To take steps toward improving rare disease clinical development strategies under current global regulatory statutes, Amicus Therapeutics, Inc. and BioNJ convened a 1-day meeting that included representatives from the Food and Drug Administration (FDA), biopharmaceutical industry, and not-for-profit agencies. The meeting focused on orphan diseases in pediatric and adult patients and was intended to identify potential strategies to overcome regulatory hurdles through open collaboration.During this meeting, several strategies were identified to minimize the limitations associated with low patient numbers in rare diseases, including the use of natural history to generate historical control data in comparisons, simulations, and identifying inclusion/exclusion criteria and appropriate endpoints. Novel approaches to clinical trial design were discussed to minimize patient exposure to placebo and to reduce the numbers of patients and clinical trials needed for providing substantial evidence. Novel statistical analysis approaches were also discussed to address the inherent challenges of small patient numbers. Areas of urgent unmet need were identified, including the need to develop registries that protect patient identities, to establish close collaboration and communication between the sponsor and regulatory bodies to address methodological and statistical challenges, to collaborate in pre-competitive opportunities within multiple sponsors and in conjunction with academia and disease-specific patient advocacy groups for optimal data sharing, and to develop harmonized guidelines for data extrapolation from source to target pediatric populations. Ultimately, these innovations will help in solving many regulatory challenges in rare disease drug development and encourage the availability of new treatments for patients with rare diseases.


Asunto(s)
Enfermedades Raras , Aprobación de Drogas , Humanos , Estados Unidos , United States Food and Drug Administration
4.
Mol Ther ; 25(5): 1199-1208, 2017 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-28341561

RESUMEN

Duvoglustat HCl (AT2220, 1-deoxynojirimycin) is an investigational pharmacological chaperone for the treatment of acid α-glucosidase (GAA) deficiency, which leads to the lysosomal storage disorder Pompe disease, which is characterized by progressive accumulation of lysosomal glycogen primarily in heart and skeletal muscles. The current standard of care is enzyme replacement therapy with recombinant human GAA (alglucosidase alfa [AA], Genzyme). Based on preclinical data, oral co-administration of duvoglustat HCl with AA increases exposure of active levels in plasma and skeletal muscles, leading to greater substrate reduction in muscle. This phase 2a study consisted of an open-label, fixed-treatment sequence that evaluated the effect of single oral doses of 50 mg, 100 mg, 250 mg, or 600 mg duvoglustat HCl on the pharmacokinetics and tissue levels of intravenously infused AA (20 mg/kg) in Pompe patients. AA alone resulted in increases in total GAA activity and protein in plasma compared to baseline. Following co-administration with duvoglustat HCl, total GAA activity and protein in plasma were further increased 1.2- to 2.8-fold compared to AA alone in all 25 Pompe patients; importantly, muscle GAA activity was increased for all co-administration treatments from day 3 biopsy specimens. No duvoglustat-related adverse events or drug-related tolerability issues were identified.


Asunto(s)
1-Desoxinojirimicina/uso terapéutico , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Lisosomas/enzimología , Músculo Esquelético/efectos de los fármacos , alfa-Glucosidasas/farmacocinética , Administración Oral , Adulto , Esquema de Medicación , Sinergismo Farmacológico , Quimioterapia Combinada , Terapia de Reemplazo Enzimático/métodos , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Humanos , Infusiones Intravenosas , Lisosomas/patología , Masculino , Persona de Mediana Edad , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Seguridad del Paciente , Resultado del Tratamiento , alfa-Glucosidasas/sangre
5.
PLoS One ; 10(8): e0134341, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26252393

RESUMEN

UNLABELLED: Migalastat HCl (AT1001, 1-Deoxygalactonojirimycin) is an investigational pharmacological chaperone for the treatment of α-galactosidase A (α-Gal A) deficiency, which leads to Fabry disease, an X-linked, lysosomal storage disorder. The currently approved, biologics-based therapy for Fabry disease is enzyme replacement therapy (ERT) with either agalsidase alfa (Replagal) or agalsidase beta (Fabrazyme). Based on preclinical data, migalastat HCl in combination with agalsidase is expected to result in the pharmacokinetic (PK) enhancement of agalsidase in plasma by increasing the systemic exposure of active agalsidase, thereby leading to increased cellular levels in disease-relevant tissues. This Phase 2a study design consisted of an open-label, fixed-treatment sequence that evaluated the effects of single oral doses of 150 mg or 450 mg migalastat HCl on the PK and tissue levels of intravenously infused agalsidase (0.2, 0.5, or 1.0 mg/kg) in male Fabry patients. As expected, intravenous administration of agalsidase alone resulted in increased α-Gal A activity in plasma, skin, and peripheral blood mononuclear cells (PBMCs) compared to baseline. Following co-administration of migalastat HCl and agalsidase, α-Gal A activity in plasma was further significantly increased 1.2- to 5.1-fold compared to agalsidase administration alone, in 22 of 23 patients (95.6%). Importantly, similar increases in skin and PBMC α-Gal A activity were seen following co-administration of migalastat HCl and agalsidase. The effects were not related to the administered migalastat HCl dose, as the 150 mg dose of migalastat HCl increased α-Gal A activity to the same extent as the 450 mg dose. Conversely, agalsidase had no effect on the plasma PK of migalastat. No migalastat HCl-related adverse events or drug-related tolerability issues were identified. TRIAL REGISTRATION: ClinicalTrials.gov NCT01196871.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Enfermedad de Fabry/tratamiento farmacológico , Enfermedad de Fabry/enzimología , Isoenzimas/uso terapéutico , alfa-Galactosidasa/metabolismo , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/sangre , 1-Desoxinojirimicina/farmacocinética , 1-Desoxinojirimicina/uso terapéutico , Administración Oral , Adulto , Área Bajo la Curva , Demografía , Enfermedad de Fabry/sangre , Humanos , Bombas de Infusión , Isoenzimas/administración & dosificación , Masculino , Persona de Mediana Edad , Proteínas Recombinantes , Piel/enzimología , alfa-Galactosidasa/administración & dosificación , alfa-Galactosidasa/sangre , alfa-Galactosidasa/uso terapéutico
6.
Clin Pharmacol Drug Dev ; 4(4): 256-61, 2015 07.
Artículo en Inglés | MEDLINE | ID: mdl-27136905

RESUMEN

OBJECTIVES: Renal function may progressively decline in patients with Fabry disease. This study assessed pharmacokinetics, safety, and tolerability of a single oral dose of migalastat HCl 150 mg in subjects with normal or mildly, moderately, or severely impaired renal function. METHODS: Volunteers were enrolled into two cohorts stratified for renal function calculated using the Cockcroft-Gault equation for creatinine clearance. Pharmacokinetic parameters determined were: area under the concentration-time curve (AUC) from time zero to the last measurable concentration postdose (AUC0-t ) and extrapolated to infinity (AUC0-∞ ), maximum observed concentration (Cmax ), time to Cmax (tmax ), concentration at 48 hours postdose (C48h ), terminal elimination half-life (t1/2 ), oral clearance (CL/F), and apparent terminal elimination rate constant (λz) (ClinicalTrials.gov registration: NCT01730469). RESULTS: Thirty-two subjects enrolled and completed the study (Cohort 1: n = 24; Cohort 2: n = 8). Migalastat clearance decreased with increasing renal impairment, resulting in increases in migalastat HCl plasma t1/2 , AUC0-∞ , and C48h compared with subjects with normal renal function. Incidence of adverse events was comparable across all renal function groups. CONCLUSIONS: Plasma migalastat clearance decreased as degree of renal impairment increased. Data from the migalastat HCl clinical program will guide dosing and intervals for patients with Fabry disease with renal impairment.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Inhibidores de Glicósido Hidrolasas/farmacocinética , Enfermedades Renales/fisiopatología , Riñón/fisiopatología , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/efectos adversos , 1-Desoxinojirimicina/sangre , 1-Desoxinojirimicina/farmacocinética , Administración Oral , Adolescente , Adulto , Anciano , Área Bajo la Curva , Femenino , Inhibidores de Glicósido Hidrolasas/administración & dosificación , Inhibidores de Glicósido Hidrolasas/efectos adversos , Inhibidores de Glicósido Hidrolasas/sangre , Semivida , Humanos , Riñón/metabolismo , Enfermedades Renales/sangre , Enfermedades Renales/diagnóstico , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Modelos Biológicos , Índice de Severidad de la Enfermedad , Estados Unidos , Adulto Joven
7.
Clin Pharmacol Drug Dev ; 4(3): 193-202, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27140799

RESUMEN

Migalastat HCl is an investigational, pharmacological chaperone for mutant α-galactosidase A, which is responsible for Fabry disease, an X-linked, lysosomal storage disorder. Two Phase I studies evaluated relative bioavailability, effect of meal type and timing on pharmacokinetics, safety, and tolerability of migalastat HCl in healthy volunteers. Study 1 (N = 15, 19-55 years): single 100-mg doses of migalastat HCl capsule and solution formulations were bioequivalent. The ratios of LSM (90% CIs) for Cmax were 97.1% (86.8-109) and AUC0-inf 97.9% (88.8-108) under fasted conditions. Single 100-mg doses of migalastat HCl capsules administered with a high-fat meal decreased Cmax by 40% and AUC0-inf by 37%. A high-fat meal delayed tmax by approximately 1 hour. Study 2 (N = 20, 18-65 years): A high-fat or light meal up to 1 hour before or after administration of single 150 mg doses of migalastat HCl capsules decreased Cmax and AUC0-inf up to 40%, but had no apparent effect on tmax (range of medians with food: 1.5-3 hours, median fasted: 3 hours). A 50-g glucose drink co-administered with migalastat HCL did not result in clinically significant changes in migalastat absorption. No serious safety or tolerability issues were identified.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Interacciones Alimento-Droga , Comidas , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/efectos adversos , 1-Desoxinojirimicina/sangre , 1-Desoxinojirimicina/farmacocinética , Administración Oral , Adolescente , Adulto , Anciano , Área Bajo la Curva , Disponibilidad Biológica , Cápsulas , Estudios Cruzados , Grasas de la Dieta/administración & dosificación , Esquema de Medicación , Composición de Medicamentos , Ayuno/sangre , Femenino , Semivida , Voluntarios Sanos , Humanos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Soluciones Farmacéuticas , Periodo Posprandial , Texas , Equivalencia Terapéutica , Adulto Joven
8.
Clin Pharmacol Drug Dev ; 2(2): 120-32, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27121667

RESUMEN

Migalastat HCl is an investigational, oral treatment for Fabry disease, an X-linked lysosomal storage disorder. Four Phase 1 studies were conducted to determine the pharmacokinetics, pharmacodynamics, safety, and tolerability of migalastat. Healthy volunteers (N = 124), 18-55 years old, received migalastat HCl single (25 mg-2000 mg) or twice-daily doses (50 mg, 150 mg) for 7 days in a double-blind, placebo-controlled fashion. Migalastat pharmacokinetics were dose-proportional (AUC∞ range: 1129-72 838 ng h/mL, Cmax range: 200.5-13 844 ng/mL, t1/2 3-4 hours). Steady state was achieved by Day 7. Up to 67% of the dose was excreted as unchanged drug in urine. Increased α-Gal A activity was dose related. No abnormal cardiac effects, including prolonged QTc intervals, were observed. The pharmacokinetics of migalastat were well characterized in these Phase 1 studies conducted healthy volunteers. The 150 mg dose of migalastat HCl administered BID for 7 days was generally safe and well tolerated. A TQT study demonstrated lack of a positive signal at therapeutic and supra-therapeutic doses. Increases in α-Gal A enzyme activity for the 150 mg dose observed in healthy subjects suggested a successful proof of mechanism for further investigations.

9.
J Clin Pharmacol ; 52(5): 747-56, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21593282

RESUMEN

Although contraindicated, coingestion of alcohol and opioids by patients or drug abusers is a major health concern because of dangerous additive and potentially life-threatening sedative and respiratory effects. In addition, alcohol has been shown to disrupt the extended-release characteristics of certain extended-release opioid formulations, releasing a hazardous amount of opioid over a short time period. Morphine sulfate and naltrexone hydrochloride extended release capsules (MS-sNT), which contain naltrexone sequestered in each pellet core, are indicated for management of chronic, moderate to severe pain. Sequestered naltrexone is designed for release upon product tampering (crushing) to potentially mitigate morphine-induced subjective effects. This open-label, single-dose, 4-way crossover, pharmacokinetic drug interaction study compared the relative bioavailability of morphine and naltrexone when MS-sNT is administered (under fasting conditions) with increasing doses of alcohol. Thirty-two healthy, opioid-naive adults were randomized to MS-sNT administered with 240 mL of 4%, 20%, or 40% alcohol or water. No drug interaction was found between morphine in MS-sNT and 4% or 20% alcohol. Administration with 40% alcohol did not affect overall morphine exposure but was associated with a 2-fold increase in C(max) and reduction of t(max) from 9 to 4 hours versus MS-sNT taken with water. Naltrexone sequestering was successful in all treatment arms and not affected by coadministration with alcohol over the dose range tested.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Analgésicos Opioides/farmacocinética , Etanol/administración & dosificación , Morfina/farmacocinética , Naltrexona/farmacocinética , Antagonistas de Narcóticos/farmacocinética , Adulto , Consumo de Bebidas Alcohólicas/sangre , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/sangre , Analgésicos Opioides/química , Análisis de Varianza , Área Bajo la Curva , Cápsulas , Estudios Cruzados , Preparaciones de Acción Retardada , Combinación de Medicamentos , Interacciones Farmacológicas , Etanol/sangre , Femenino , Humanos , Masculino , Tasa de Depuración Metabólica , Modelos Biológicos , Morfina/administración & dosificación , Morfina/sangre , Morfina/química , Naltrexona/administración & dosificación , Naltrexona/sangre , Naltrexona/química , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/sangre , Antagonistas de Narcóticos/química , Adulto Joven
10.
Drugs R D ; 11(3): 259-75, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21902287

RESUMEN

BACKGROUND: Opioid analgesics can be abused by crushing followed by solubilization and intravenous injection to attain rapid absorption. Morphine sulfate and naltrexone hydrochloride extended release capsules (EMBEDA, MS-sNT), indicated for management of chronic, moderate to severe pain, contain pellets of morphine sulfate with a sequestered naltrexone core. Should product tampering by crushing occur, the sequestered naltrexone is intended for release to reduce morphine-induced subjective effects. OBJECTIVE: This study compared self-reports of high, euphoria, and drug-liking effects of intravenous morphine alone versus intravenous morphine combined with naltrexone in a clinical simulation of intravenous abuse of crushed MS-sNT. METHODS: This single-center, randomized, double-blind, crossover study characterized subjective effects of naltrexone administered intravenously at the same ratio to morphine present in MS-sNT. Subjects were male and had used prescription opioids five or more times within the previous 12 months to get 'high' but were not physically dependent on opioids. The primary outcome was the response to the Drug Effects Questionnaire (DEQ) question #5, "How high are you now?" (100 mm Visual Analog Scale [VAS]). The secondary outcome was the response to a Cole/Addiction Research Center Inventory (ARCI) Stimulation-Euphoria modified scale. Additional outcomes included response to VAS drug liking, the remaining DEQ questions, and pupillometry. RESULTS: Administration of intravenous naltrexone following intravenous morphine diminished mean high (29.8 vs 85.2 mm), Cole/ARCI Stimulation-Euphoria (13.7 vs 27.8 mm), and drug-liking (38.9 vs 81.4 mm) scores (all p < 0.0001) compared with intravenous morphine alone. No serious adverse events occurred as a result of the tested ratio of naltrexone to morphine. CONCLUSIONS: Results in this study population suggest that naltrexone added to morphine in the 4% ratio within MS-sNT mitigates the high, euphoria, and drug liking of morphine alone, potentially reducing the attractiveness for product tampering. Assessment of the true clinical significance of these findings will require further study.


Asunto(s)
Analgésicos Opioides/farmacología , Morfina/farmacología , Naltrexona/farmacología , Trastornos Relacionados con Opioides/tratamiento farmacológico , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacocinética , Estudios Cruzados , Método Doble Ciego , Combinación de Medicamentos , Interacciones Farmacológicas , Euforia/efectos de los fármacos , Humanos , Inyecciones Intravenosas , Masculino , Morfina/administración & dosificación , Morfina/efectos adversos , Morfina/farmacocinética , Naltrexona/administración & dosificación , Naltrexona/efectos adversos , Naltrexona/farmacocinética , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/efectos adversos , Antagonistas de Narcóticos/farmacocinética , Trastornos Relacionados con Opioides/metabolismo , Dolor/tratamiento farmacológico , Dimensión del Dolor/efectos de los fármacos , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Trastornos Relacionados con Sustancias/metabolismo , Adulto Joven
11.
Am J Ther ; 18(1): 2-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20864883

RESUMEN

Morphine sulfate and naltrexone hydrochloride extended release capsules (EMBEDA®, King Pharmaceuticals®, Inc., Bristol, TN), indicated for the management of chronic, moderate to severe pain, contain extended release morphine pellets with a sequestered naltrexone core (MS-sNT). If the product is tampered with by crushing, naltrexone, a µ-opioid antagonist, is intended for release to mitigate morphine-induced subjective effects. The primary end point of this randomized 2-way crossover study in healthy fasted volunteers was evaluation of morphine bioequivalence between MS-sNT (treatment A) and morphine sulfate extended release capsules (KADIAN®, treatment B). Morphine pharmacokinetics were assessed predose to 72 hours postdose of single 100-mg doses of treatment A or B. Analysis of variance of ln-transformed ratios of least squares mean of the area under the concentration time curve (AUC) from time 0 to last measurable concentration (AUC0-t) and AUC from time 0 to infinity (AUCinf) and maximum serum concentration (Cmax) for treatments A versus B were performed. Ratios and 90% confidence intervals for least squares mean for AUC0-t (102.2%; 98.6-105.9%), AUCinf (97.4%; 91.2-104.1%), and Cmax (93.8%; 82.4-106.7%) indicated bioequivalence between the 2 formulations. When subjects who vomited during the 12-hour dosing interval were excluded, the confidence interval for AUC0-t and AUCinf fell within the 80%-125% range, but the lower limit for Cmax was 76.9%.


Asunto(s)
Analgésicos Opioides/farmacocinética , Morfina/farmacocinética , Naltrexona/farmacocinética , Adulto , Analgésicos Opioides/administración & dosificación , Área Bajo la Curva , Disponibilidad Biológica , Biotransformación , Cápsulas , Química Farmacéutica , Estudios Cruzados , Preparaciones de Acción Retardada , Método Doble Ciego , Combinación de Medicamentos , Ayuno/metabolismo , Femenino , Semivida , Humanos , Masculino , Persona de Mediana Edad , Morfina/administración & dosificación , Naltrexona/administración & dosificación , Equivalencia Terapéutica , Adulto Joven
12.
J Pain Symptom Manage ; 40(5): 734-46, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21075272

RESUMEN

CONTEXT: Morphine sulfate and naltrexone hydrochloride extended release capsules contain extended-release pellets of morphine with a sequestered naltrexone core (MS-sNT). Taken whole, as intended, morphine is released to provide pain relief; if tampered with by crushing, naltrexone is released to mitigate subjective effects of morphine. OBJECTIVES: This open-label study assessed long-term (12-month) safety of MS-sNT in patients with chronic, moderate to severe pain. METHODS: Safety assessments included determining adverse events (AEs), laboratory assessments, and the Clinical Opiate Withdrawal Scale (COWS). Analgesic efficacy was assessed (diary) as worst, least, average, and current pain using an 11-point numeric scale (0=none; 10=worst). RESULTS: Of 465 patients receiving one or more doses, 160 completed the study. Most patients (81.3%) experienced one or more AEs, most commonly constipation (31.8%) or nausea (25.2%). Thirty-three patients (7.1%) reported serious AEs; one patient's severe gastrointestinal inflammation and colitis were considered possibly study drug-related. Most discontinuations (30%) occurred in the first month, most often because of AEs (23.7%). There were no clinically relevant changes in laboratory results or vital signs, and no clinically significant electrocardiogram changes deemed study drug-related. During each visit after Week 1, 5% or fewer patients had COWS scores indicating mild withdrawal symptoms (range, 0%-4.8%). Five patients, who did not take the study drug as instructed, had scores consistent with moderate withdrawal. MS-sNT yielded statistically significant improvements from baseline in mean scores for all pain diary items for all visits, except Week 1 for least pain. CONCLUSION: In this study population, when MS-sNT was taken as directed for chronic, moderate to severe pain for up to 12 months, most AEs were typical opioid-related side effects. Mean COWS scores remained low, indicating lack of withdrawal symptoms and appropriate transition off the study drug at completion.


Asunto(s)
Morfina/administración & dosificación , Naltrexona/administración & dosificación , Dolor/tratamiento farmacológico , Adolescente , Adulto , Anciano , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Enfermedad Crónica/tratamiento farmacológico , Estreñimiento/inducido químicamente , Preparaciones de Acción Retardada , Combinación de Medicamentos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Morfina/efectos adversos , Naltrexona/efectos adversos , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/efectos adversos , Náusea/inducido químicamente , Dimensión del Dolor/efectos de los fármacos , Resultado del Tratamiento
13.
Clin Ther ; 32(6): 1149-64, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20637968

RESUMEN

BACKGROUND: Morphine sulfate/sequestered naltrexone hydrochloride (HCl) (MS-sNT) extended-release fixed-dose combination capsules, approved by the US Food and Drug Administration (FDA) in August 2009 for chronic moderate to severe pain, contain extended-release morphine pellets with a sequestered core of the opioid antagonist naltrexone. MS-sNT was designed so that if the product is tampered with by crushing, the naltrexone becomes bioavailable to mitigate morphine-induced subjective effects, rendering the product less attractive for tampering. OBJECTIVES: The primary aim of this study was to compare the oral bioavailability of naltrexone and its metabolite 6-beta-naltrexol, derived from crushed pellets from MS-sNT capsules, to naltrexone solution. This study also assessed the relative bioavailability of morphine from crushed pellets from MS-sNT capsules and that from the whole, intact product. METHODS: This single-dose, randomized-sequence, open-label, 3-period, 3-treatment crossover trial was conducted in healthy volunteers. Adults admitted to the study center underwent a 10-hour overnight fast before study drug administration. Each subject received all 3 of the following treatments, 1 per session, separated by a 14-day washout: tampered pellets (crushed for >or=2 minutes with a mortar and pestle) from a 60-mg MS-sNT capsule (60 mg morphine/2.4 mg naltrexone); 60-mg whole, intact MS-sNT capsule; and oral naltrexone HCl (2.4 mg) solution. Plasma concentrations of naltrexone and 6-beta-naltrexol were measured 0 to 168 hours after administration. Morphine pharmaco-kinetics of crushed and whole pellets were determined 0 to 72 hours after administration. The analysis of relative bioavailability was based on conventional FDA criteria for assuming bioequivalence; that is, 90% CIs for ratios of geometric means (natural logarithm [In]-transformed C(max) and AUC) fell within the range of 80% to 125%. Subjects underwent physical examinations, clinical laboratory tests, and ECG at screening and study discharge and were monitored for adverse events (AEs) throughout the study. RESULTS: Of the 24 subjects enrolled in the study, 23 completed it. Most subjects were white (79%) and male (63%); the mean (SD) age was 39.3 (11.2) years and the mean weight was 77.6 (13.5) kg (range, 55.0102.5 kg). Plasma C(max) and AUC(0-t) of naltrexone after the administration of crushed pellets of MS-sNT (579 pg/mL and 1811 h . pg/mL, respectively) and naltrexone solution (584 pg/mL and 1954 h . pg/mL) were not significantly different; 90% CIs were 83.8% to 116% and 83.3% to 102%, meeting the regulatory requirements for assuming bioequivalence in this study population. Plasma naltrexone concentration was below the lower limit of quantitation (4.0 pg/mL) in 23 of 24 subjects (96%) after whole MS-sNT administration. Morphine AUC(0-t) was not significantly different whether MS-sNT was crushed (163 h . ng/mL) or administered whole (174 h . ng/mL), but C(max) was numerically higher (24.5 vs 7.7 ng/mL) and T(max) was numerically shorter (2.00 vs 7.03 hours) with MS-sNT crushed versus whole. The most commonly reported AEs were nausea (8/23 [35%], 10/24 [42%], and 3/23 [13%] subjects in the crushed, whole, and naltrexone groups, respectively) and emesis (6 [26%], 7 [29%], and 2 [9%]). CONCLUSIONS: In this single-dose study, when pellets from MS-sNT were crushed, naltrexone appeared to be completely released and available to mitigate morphine-induced effects. When MS-sNT was administered whole, morphine was released in an extended-release fashion while naltrexone remained sequestered.


Asunto(s)
Morfina/farmacocinética , Naltrexona/farmacocinética , Antagonistas de Narcóticos/farmacocinética , Narcóticos/farmacocinética , Administración Oral , Adulto , Área Bajo la Curva , Disponibilidad Biológica , Estudios Cruzados , Preparaciones de Acción Retardada , Combinación de Medicamentos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Morfina/efectos adversos , Morfina/sangre , Naltrexona/efectos adversos , Naltrexona/análogos & derivados , Naltrexona/sangre , Antagonistas de Narcóticos/efectos adversos , Antagonistas de Narcóticos/sangre , Narcóticos/efectos adversos , Narcóticos/sangre
14.
J Pediatr Gastroenterol Nutr ; 38(4): 442-51, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15085026

RESUMEN

OBJECTIVES: To characterize the impact of development on the pharmacokinetics and pharmacodynamics of nizatidine. METHODS: Children (age range, 5 days-18 years) and adults (age range, 18-50 years) were enrolled in four open-label trials. Nizatidine formulation and dose were determined by age: infants received 2 or 4 mg/kg i.v., children 2.5 or 5 mg/kg in one of three oral liquid formulations, and adolescents and adults received a fixed 150-mg capsule. Nizatidine and N-desmethylnizatidine concentrations were measured in serial post-dose plasma samples by a high-performance liquid chromatographic assay with mass spectrometric detection. Intragastric pH was recorded during a 24-hour post-dose interval. RESULTS: Data on 93 subjects were combined with previous values from 36 individuals to cover an age group not adequately captured and to control for formulation effects. Dose-normalized exposure estimates revealed no apparent age dependence; however, maximum plasma concentration (298.5 +/- 100.7 v 552.8 +/- 152.4 ng/mL per mg/kg dose) and AUC0-infinity (954.4 +/- 379.8 v 1,573.0 +/- 347.4 ng*hour/mL per mg/kg dose) were reduced in extemporaneous formulations in apple juice. The apparent modest age dependence observed for total body clearance (Cl/F) (r = 0.365) and Vss/F (r = 0.221) reflected a formulation-dependent decrease in bioavailability rather than a true age effect. The age-associated changes in lambda z observed for nizatidine and its metabolite were predictable and consistent with developmental acquisition of renal function. Mean and median pH, as well as fraction of time that the dosing interval remained above target pH values, were significantly greater with administration of the drug than without. CONCLUSIONS: The biodisposition of nizatidine in children and adults is similar; however, response after a comparable weight-based dose is equal and potentially greater in children.


Asunto(s)
Reflujo Gastroesofágico/tratamiento farmacológico , Antagonistas de los Receptores H2 de la Histamina/farmacología , Antagonistas de los Receptores H2 de la Histamina/farmacocinética , Nizatidina/farmacología , Nizatidina/farmacocinética , Administración Oral , Adolescente , Adulto , Área Bajo la Curva , Disponibilidad Biológica , Cápsulas , Niño , Preescolar , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Femenino , Determinación de la Acidez Gástrica , Reflujo Gastroesofágico/metabolismo , Antagonistas de los Receptores H2 de la Histamina/uso terapéutico , Humanos , Lactante , Recién Nacido , Inyecciones Intravenosas , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Nizatidina/uso terapéutico , Resultado del Tratamiento
15.
J Clin Pharmacol ; 43(2): 148-53, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12616667

RESUMEN

Nizatidine (Axid) is an H2-receptor antagonist used for the treatment of acid-related gastrointestinal disorders. Given the frequency of these conditions in children and the potential for pediatric use of nizatidine, an oral liquid dosage formulation would provide an alternative treatment option for patients unable to swallow solid oral dosage forms. This study was designed as an open-label, single-dose, four-way crossover trial to investigate the bioequivalence of 150 mg nizatidine administered in three oral liquid formulations (a commercially prepared oral syrup, an extemporaneous solution in apple juice, and an extemporaneous suspension in infant formula) relative to the marketed capsule formulation. Twenty-four adult subjects (ages 31.2 +/- 7.5 years; weight 71.1 +/- 11.8 kg) were enrolled, and blood samples for determination of plasma nizatidine concentrations were collected prior to drug administration and at 19 discrete intervals over a 24-hour postdose interval. Nizatidine was quantitated from plasma using a validated HPLC-MS assay, and a noncompartmental approach was used to describe nizatidine biodisposition in all subjects. Significant treatment effects were observed for log-normalized Cmax, AUC0-n, and AUC0-infinity (p < 0.001). Further evaluation revealed that nizatidine prepared in apple juice was markedly less bioavailable than the reference capsule, with 90% confidence intervals (CIs) of 0.518-0.626, 0.682-0.751, and 0.696-0.763 for Cmax, AUC0-n, and AUC0-infinity, respectively. The remaining two oral formulations demonstrated 90% CI within the guidelines established by the Food and Drug Administration (e.g., 0.80-1.25). Thus, nizatidine in infant formula and the commercially prepared oral syrup can be considered bioequivalent to the reference capsule.


Asunto(s)
Nizatidina/farmacocinética , Adulto , Área Bajo la Curva , Disponibilidad Biológica , Cápsulas , Química Farmacéutica , Femenino , Semivida , Humanos , Masculino , Nizatidina/administración & dosificación , Nizatidina/sangre , Equivalencia Terapéutica , Distribución Tisular
16.
J Clin Pharmacol ; 43(1): 74-83, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12520631

RESUMEN

The pharmacokinetics and intragastric pH effects of a novel nizatidine controlled-release (CR) formulation were compared to a currently marketed immediate-release (IR) nizatidine formulation (Axid). The bimodal pulsatile release characteristics of nizatidine CR decreased its Cmax by approximately 42% compared to nizatidine IR while maintaining 90% relative bioavailability; tmax was approximately 1.6 times longer with the CR formulation. These characteristics enabled controlled-release nizatidine to sustain effective plasma drug concentrations for a greater duration than immediate-release nizatidine over the dosing intervals. In multiple doses, the 24-hour AUC ratio for all comparisons of nizatidine CR 150 mg bid, nizatidine CR 300 mg daily, and nizatidine IR 150 mg bid was between 97% and 99%. Mean pH AUC values for nizatidine CR 150 mg bid and nizatidine IR 150 mg bid were similar overall during the 0- to 14-hour and 14- to 24-hour dosing intervals. For the 14- to 24-hour dosing interval, nizatidine CR 150 mg maintained gastric pH over 3.0 and 4.0 for 42% and 27% of the time compared to 39% and 23% for nizatidine IR, respectively. Nizatidine CR 300 mg, compared to the 150-mg CR and IR regimens, had a greater effect on increasing evening intragastric pH, thus providing support for the potential utility of nizatidine CR 300 mg dosed at night in alleviating nocturnal symptoms of gastroesophageal reflux disease.


Asunto(s)
Antagonistas de los Receptores H2 de la Histamina/farmacología , Antagonistas de los Receptores H2 de la Histamina/farmacocinética , Nizatidina/farmacología , Nizatidina/farmacocinética , Adolescente , Adulto , Estudios Cruzados , Preparaciones de Acción Retardada , Femenino , Humanos , Masculino , Persona de Mediana Edad
17.
J Clin Pharmacol ; 42(10): 1089-96, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12362922

RESUMEN

The pharmacokinetics of nizatidine following a single 5.0 mg/kg oral dose given as an extemporaneous liquid formulation in apple juice was examined in 12 healthy children (8.0 +/- 2.4 years, 30.7 +/- 8.4 kg). Nizatidine and N-desmethylnizatidine were quantitated by HPLC/MS from five post dose blood samples taken over a 12-hour period. The apparent terminal elimination rate constant for nizatidine in the pediatric subjects (0.58 +/- 0.8h(-1)) was virtually identical to that (0.54 +/- 0.13 h(-1)) previously reported from adult studies. When corrected for an estimated 30% reduction in nizatidine oral bioavailability observed in adults upon coingestion of the drug with other fruit/vegetable juices, nizatidine pharmacokinetic parameter estimates (e.g., Cmax, CL/F, Vss/F) in our pediatric subjects were similar to those previously reported in adults who were administered dimensionally similar (e.g., approximately 4 mg/kg) solid oral doses of the drug. Examination of the mean area under the curve (i.e., AUC0-infinity for nizatidine and N-desmethylnizatidine suggested an approximate 15% metabolic conversion of the parent drug. Finally, nizatidine plasma concentrations in pediatric patients following a single 5.0 mg/kg oral dose exceeded the EC50 value of the drug for gastric acid suppression determined from adult studies for approximately 6 hours.


Asunto(s)
Antagonistas de los Receptores H2 de la Histamina/farmacocinética , Nizatidina/análogos & derivados , Nizatidina/farmacocinética , Administración Oral , Área Bajo la Curva , Niño , Preescolar , Cromatografía Líquida de Alta Presión , Femenino , Semivida , Antagonistas de los Receptores H2 de la Histamina/administración & dosificación , Antagonistas de los Receptores H2 de la Histamina/sangre , Humanos , Lactante , Masculino , Nizatidina/administración & dosificación , Nizatidina/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...