Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Neurol ; 271(4): 1787-1801, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38057636

ABSTRACT

Cipaglucosidase alfa plus miglustat (cipa + mig) is a novel, two-component therapy for Pompe disease. We report data from the Phase I/II ATB200-02 study for up to 48 months of treatment. Four adult cohorts, including one non-ambulatory ERT-experienced (n = 6) and three ambulatory cohorts, (two enzyme replacement therapy [ERT]-experienced cohorts [2-6 years (n = 11) and ≥ 7 years (n = 6)]), one ERT-naïve cohort (n = 6), received 20 mg/kg intravenous-infused cipa plus 260 mg oral mig biweekly. Change from baseline (CFBL) for multiple efficacy endpoints at 12, 24, 36, and 48 months, pharmacodynamics, pharmacokinetics, safety, and immunogenicity data were assessed. Six-minute walking distance (% predicted) improved at 12, 24, 36, and 48 months: pooled ambulatory ERT-experienced cohorts, mean(± standard deviation [SD]) CFBL: 6.1(± 7.84), n = 16; 5.4(± 10.56), n = 13; 3.4(± 14.66), n = 12; 5.9(± 17.36), n = 9, respectively; ERT-naïve cohort: 10.7(± 3.93), n = 6; 11.0(± 5.06), n = 6; 9.0(± 7.98), n = 5; 11.7(± 7.69), n = 4, respectively. Percent predicted forced vital capacity was generally stable in ERT-experienced cohorts, mean(± SD) CFBL - 1.2(± 5.95), n = 16; 1.0(± 7.96), n = 13; - 0.3(± 6.68), n = 10; 1.0(± 6.42), n = 6, respectively, and improved in the ERT-naïve cohort: 3.2(± 8.42), n = 6; 4.7(± 5.09), n = 6; 6.2(± 3.35), n = 5; 8.3(± 4.50), n = 4, respectively. Over 48 months, CK and Hex4 biomarkers improved in ambulatory cohorts. Overall, cipa + mig was well tolerated with a safety profile like alglucosidase alfa. ATB200-02 results show the potential benefits of cipa + mig as a long-term treatment option for Pompe disease. Trial registration number: NCT02675465 January 26, 2016.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Glycogen Storage Disease Type II , Propionates , Adult , Humans , Glycogen Storage Disease Type II/therapy , Treatment Outcome , alpha-Glucosidases/therapeutic use , Indoles , Enzyme Replacement Therapy/methods
2.
Clin Pharmacol Drug Dev ; 11(12): 1367-1381, 2022 12.
Article in English | MEDLINE | ID: mdl-36331497

ABSTRACT

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.


Subject(s)
Fabry Disease , Renal Insufficiency , Adult , Humans , Adolescent , Child , 1-Deoxynojirimycin/adverse effects , Fabry Disease/drug therapy , Glomerular Filtration Rate , Renal Insufficiency/drug therapy
3.
Clin Pharmacol Drug Dev ; 10(9): 1075-1088, 2021 09.
Article in English | MEDLINE | ID: mdl-33876577

ABSTRACT

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.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Isoenzymes/pharmacokinetics , Models, Biological , alpha-Galactosidase/pharmacokinetics , 1-Deoxynojirimycin/pharmacokinetics , Adult , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Species Specificity , Tissue Distribution , Young Adult , alpha-Galactosidase/genetics
4.
Planta Med ; 87(4): 294-304, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33296937

ABSTRACT

Recent research demonstrates that Echinacea possesses cannabimimetic activity with potential applications beyond common contemporary uses for relief of cold and flu symptoms. In this study, we investigated the in vitro inhibitory effect of root extracts of Echinacea purpurea and Echinacea angustifolia on fatty acid amide hydrolase, the main enzyme that degrades the endocannabinoid anandamide. The objective was to relate variation in bioactivity between commercial Echinacea genotypes to their phytochemical profiles and to identify determinants of activity using biochemometric analysis. Forty root extracts of each of species were tested for inhibition of fatty acid amide hydrolase and analyzed by HPLC-DAD/MS to identify and quantitate alkylamides and caffeic acid derivatives. Fatty acid amide hydrolase inhibition ranged from 34 - 80% among E. angustifolia genotypes and from 33 - 87% among E. purpurea genotypes. Simple linear regression revealed the caffeic acid derivatives caftaric acid and cichoric acid, and the alkylamide dodeca-2E,4Z-diene-8,10-diynioc acid 2-methylbutylamide, as the strongest determinants of inhibition in E. purpurea (r* = 0.53, 0.45, and 0.20, respectively) while in E. angustifolia, only CADs were significantly associated with activity, most notably echinacoside (r* = 0.26). Regression analysis using compound groups generated by hierarchical clustering similarly indicated that caffeic acid derivatives contributed more than alkylamides to in vitro activity. Testing pure compounds identified as determinants of activity revealed cichoric acid (IC50 = 45 ± 4 µM) and dodeca-2E,4E,8Z,10E-tetraenoic acid isobutylamide (IC50 = 54 ± 2 µM) as the most active. The results suggest that several phytochemicals may contribute to Echinacea's cannabimimetic activity and that ample variation in genotypes exists for selection of high-activity germplasm in breeding programs.


Subject(s)
Echinacea , Amidohydrolases/genetics , Chromatography, High Pressure Liquid , Plant Extracts/pharmacology
5.
BMJ Case Rep ; 13(8)2020 Aug 11.
Article in English | MEDLINE | ID: mdl-32784238

ABSTRACT

The COVID-19 pandemic has had a significant impact on the structure and operation of healthcare services worldwide. We highlight a case of a 64-year-old man who presented to the emergency department with acute dyspnoea on a background of a 2-week history of fever, dry cough and shortness of breath. On initial assessment the patient was hypoxic (arterial oxygen saturation (SaO2) of 86% on room air), requiring 10 L/min of oxygen to maintain 98% SaO2 Examination demonstrated left-sided tracheal deviation and absent breath sounds in the right lung field on auscultation. A chest radiograph revealed a large right-sided tension pneumothorax which was treated with needle thoracocentesis and a definitive chest drain. A CT pulmonary angiogram demonstrated segmental left lower lobe acute pulmonary emboli, significant generalised COVID-19 parenchymal features, surgical emphysema and an iatrogenic pneumatocoele. This case emphasises the importance of considering coexisting alternative diagnoses in patients who present with suspected COVID-19.


Subject(s)
Betacoronavirus/isolation & purification , Coronavirus Infections/complications , Pneumonia, Viral/complications , Pneumothorax/complications , Pulmonary Embolism/complications , Anti-Bacterial Agents/therapeutic use , Anticoagulants/therapeutic use , COVID-19 , Coronavirus Infections/diagnosis , Coronavirus Infections/therapy , Drainage , Humans , Lung/diagnostic imaging , Male , Middle Aged , Oxygen Inhalation Therapy , Pandemics , Pneumonia, Viral/diagnosis , Pneumonia, Viral/therapy , Pneumothorax/diagnostic imaging , Pneumothorax/therapy , Pulmonary Embolism/diagnostic imaging , Pulmonary Embolism/drug therapy , Radiography, Thoracic , SARS-CoV-2 , Tinzaparin/therapeutic use , Tomography, X-Ray Computed
6.
Orphanet J Rare Dis ; 14(1): 36, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30736861

ABSTRACT

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.


Subject(s)
Rare Diseases , Drug Approval , Humans , United States , United States Food and Drug Administration
7.
Mol Ther ; 25(5): 1199-1208, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28341561

ABSTRACT

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.


Subject(s)
1-Deoxynojirimycin/therapeutic use , Glycogen Storage Disease Type II/drug therapy , Lysosomes/enzymology , Muscle, Skeletal/drug effects , alpha-Glucosidases/pharmacokinetics , Administration, Oral , Adult , Drug Administration Schedule , Drug Synergism , Drug Therapy, Combination , Enzyme Replacement Therapy/methods , Female , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/pathology , Humans , Infusions, Intravenous , Lysosomes/pathology , Male , Middle Aged , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Patient Safety , Treatment Outcome , alpha-Glucosidases/blood
8.
J Med Genet ; 54(4): 288-296, 2017 04.
Article in English | MEDLINE | ID: mdl-27834756

ABSTRACT

BACKGROUND: Fabry disease is an X-linked lysosomal storage disorder caused by GLA mutations, resulting in α-galactosidase (α-Gal) deficiency and accumulation of lysosomal substrates. Migalastat, an oral pharmacological chaperone being developed as an alternative to intravenous enzyme replacement therapy (ERT), stabilises specific mutant (amenable) forms of α-Gal to facilitate normal lysosomal trafficking. METHODS: The main objective of the 18-month, randomised, active-controlled ATTRACT study was to assess the effects of migalastat on renal function in patients with Fabry disease previously treated with ERT. Effects on heart, disease substrate, patient-reported outcomes (PROs) and safety were also assessed. RESULTS: Fifty-seven adults (56% female) receiving ERT (88% had multiorgan disease) were randomised (1.5:1), based on a preliminary cell-based assay of responsiveness to migalastat, to receive 18 months open-label migalastat or remain on ERT. Four patients had non-amenable mutant forms of α-Gal based on the validated cell-based assay conducted after treatment initiation and were excluded from primary efficacy analyses only. Migalastat and ERT had similar effects on renal function. Left ventricular mass index decreased significantly with migalastat treatment (-6.6 g/m2 (-11.0 to -2.2)); there was no significant change with ERT. Predefined renal, cardiac or cerebrovascular events occurred in 29% and 44% of patients in the migalastat and ERT groups, respectively. Plasma globotriaosylsphingosine remained low and stable following the switch from ERT to migalastat. PROs were comparable between groups. Migalastat was generally safe and well tolerated. CONCLUSIONS: Migalastat offers promise as a first-in-class oral monotherapy alternative treatment to intravenous ERT for patients with Fabry disease and amenable mutations. TRIAL REGISTRATION NUMBER: NCT00925301; Pre-results.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Fabry Disease/drug therapy , Molecular Chaperones/administration & dosage , alpha-Galactosidase/genetics , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/adverse effects , Administration, Oral , Adolescent , Adult , Aged , Enzyme Replacement Therapy/adverse effects , Fabry Disease/metabolism , Fabry Disease/physiopathology , Female , Humans , Lysosomes/genetics , Lysosomes/pathology , Male , Middle Aged , Molecular Chaperones/adverse effects , Treatment Outcome
9.
N Engl J Med ; 375(6): 545-55, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27509102

ABSTRACT

BACKGROUND: Fabry's disease, an X-linked disorder of lysosomal α-galactosidase deficiency, leads to substrate accumulation in multiple organs. Migalastat, an oral pharmacologic chaperone, stabilizes specific mutant forms of α-galactosidase, increasing enzyme trafficking to lysosomes. METHODS: The initial assay of mutant α-galactosidase forms that we used to categorize 67 patients with Fabry's disease for randomization to 6 months of double-blind migalastat or placebo (stage 1), followed by open-label migalastat from 6 to 12 months (stage 2) plus an additional year, had certain limitations. Before unblinding, a new, validated assay showed that 50 of the 67 participants had mutant α-galactosidase forms suitable for targeting by migalastat. The primary end point was the percentage of patients who had a response (≥50% reduction in the number of globotriaosylceramide inclusions per kidney interstitial capillary) at 6 months. We assessed safety along with disease substrates and renal, cardiovascular, and patient-reported outcomes. RESULTS: The primary end-point analysis, involving patients with mutant α-galactosidase forms that were suitable or not suitable for migalastat therapy, did not show a significant treatment effect: 13 of 32 patients (41%) who received migalastat and 9 of 32 patients (28%) who received placebo had a response at 6 months (P=0.30). Among patients with suitable mutant α-galactosidase who received migalastat for up to 24 months, the annualized changes from baseline in the estimated glomerular filtration rate (GFR) and measured GFR were -0.30±0.66 and -1.51±1.33 ml per minute per 1.73 m(2) of body-surface area, respectively. The left-ventricular-mass index decreased significantly from baseline (-7.7 g per square meter; 95% confidence interval [CI], -15.4 to -0.01), particularly when left ventricular hypertrophy was present (-18.6 g per square meter; 95% CI, -38.2 to 1.0). The severity of diarrhea, reflux, and indigestion decreased. CONCLUSIONS: Among all randomly assigned patients (with mutant α-galactosidase forms that were suitable or not suitable for migalastat therapy), the percentage of patients who had a response at 6 months did not differ significantly between the migalastat group and the placebo group. (Funded by Amicus Therapeutics; ClinicalTrials.gov numbers, NCT00925301 [study AT1001-011] and NCT01458119 [study AT1001-041].).


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Fabry Disease/drug therapy , Kidney/chemistry , Trihexosylceramides/analysis , alpha-Galactosidase/antagonists & inhibitors , 1-Deoxynojirimycin/adverse effects , 1-Deoxynojirimycin/therapeutic use , Adolescent , Adult , Aged , Diarrhea/drug therapy , Diarrhea/etiology , Double-Blind Method , Fabry Disease/complications , Female , Glomerular Filtration Rate , Heart Ventricles/diagnostic imaging , Humans , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/drug therapy , Kidney/physiopathology , Male , Middle Aged , Mutation , Trihexosylceramides/urine , Ultrasonography , Young Adult , alpha-Galactosidase/genetics
10.
PLoS One ; 10(8): e0134341, 2015.
Article in English | MEDLINE | ID: mdl-26252393

ABSTRACT

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.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Fabry Disease/drug therapy , Fabry Disease/enzymology , Isoenzymes/therapeutic use , alpha-Galactosidase/metabolism , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/blood , 1-Deoxynojirimycin/pharmacokinetics , 1-Deoxynojirimycin/therapeutic use , Administration, Oral , Adult , Area Under Curve , Demography , Fabry Disease/blood , Humans , Infusion Pumps , Isoenzymes/administration & dosage , Male , Middle Aged , Recombinant Proteins , Skin/enzymology , alpha-Galactosidase/administration & dosage , alpha-Galactosidase/blood , alpha-Galactosidase/therapeutic use
11.
Hortic Res ; 2: 15061, 2015.
Article in English | MEDLINE | ID: mdl-27087982

ABSTRACT

Apple (Malus × domestica Borkh.) is one of the most widely cultivated tree crops, and fruit storability is vital to the profitability of the apple fruit industry. Fruit of many apple cultivars can be stored for an extended period due to the introduction of advanced storage technologies, such as controlled atmosphere (CA) and 1-methylcyclopropane (1-MCP). However, CA storage can cause external CO2 injury for some apple cultivars. The molecular changes associated with the development of CO2 injury are not well elucidated. In this study, the global transcriptional regulations were investigated under different storage conditions and during development of CO2 injury symptoms on 'Golden Delicious' fruit. Fruit peel tissues under three different storage regimens, regular cold atmosphere, CA and CA storage and 1-MCP application were sampled at four storage durations over a 12-week period. Fruit physiological changes were affected differently under these storage regimens, and CO2 injury symptoms were detectable 2 weeks after CA storage. Identification of the differentially expressed genes and a gene ontology enrichment analysis revealed the specific transcriptome changes associated with each storage regimen. Overall, a profound transcriptome change was associated with CA storage regimen as indicated by the large number of differentially expressed genes. The lighter symptom was accompanied by reduced transcriptome changes under the CA storage and 1-MCP application regimen. Furthermore, the higher enrichment levels in the functional categories of oxidative stress response, glycolysis and protein post-translational modification were only associated with CA storage regime; therefore, these processes potentially contribute to the development of external CO2 injury or its symptom in apple.

12.
Clin Pharmacol Drug Dev ; 4(4): 256-61, 2015 07.
Article in English | MEDLINE | ID: mdl-27136905

ABSTRACT

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.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Glycoside Hydrolase Inhibitors/pharmacokinetics , Kidney Diseases/physiopathology , Kidney/physiopathology , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/adverse effects , 1-Deoxynojirimycin/blood , 1-Deoxynojirimycin/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Area Under Curve , Female , Glycoside Hydrolase Inhibitors/administration & dosage , Glycoside Hydrolase Inhibitors/adverse effects , Glycoside Hydrolase Inhibitors/blood , Half-Life , Humans , Kidney/metabolism , Kidney Diseases/blood , Kidney Diseases/diagnosis , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Severity of Illness Index , United States , Young Adult
13.
Clin Pharmacol Drug Dev ; 4(3): 193-202, 2015.
Article in English | MEDLINE | ID: mdl-27140799

ABSTRACT

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.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Food-Drug Interactions , Meals , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/adverse effects , 1-Deoxynojirimycin/blood , 1-Deoxynojirimycin/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Area Under Curve , Biological Availability , Capsules , Cross-Over Studies , Dietary Fats/administration & dosage , Drug Administration Schedule , Drug Compounding , Fasting/blood , Female , Half-Life , Healthy Volunteers , Humans , Male , Metabolic Clearance Rate , Middle Aged , Pharmaceutical Solutions , Postprandial Period , Texas , Therapeutic Equivalency , Young Adult
14.
Hum Psychopharmacol ; 29(3): 251-65, 2014 May.
Article in English | MEDLINE | ID: mdl-24911576

ABSTRACT

OBJECTIVE: This paper aimed to evaluate the effects of coadministered immediate-release morphine and ethanol on safety, pharmacokinetic, and pharmacodynamic measures. METHODS: In the first stage of a randomized, double-blind, placebo-controlled, crossover study, 16 healthy men with a history of moderate drinking received morphine 50 mg+ethanol 0.7 g/kg, morphine 50 mg+ethanol placebo, and morphine placebo+ethanol 0.7 g/kg. In the second stage, participants received either a lower (30 mg) or higher (80 mg) morphine dose (alone and in combination with ethanol) depending on their tolerability to treatments in stage 1. Safety, pharmacodynamic (including visual analog scales, pupillometry, capnography, and psychomotor and cognitive measures), and pharmacokinetic assessments were conducted. RESULTS: With the exception of one severe adverse event (AE), all others were mild or moderate in intensity. Morphine resulted in dose-related increases in AEs. When morphine was administered with ethanol, similar AEs were observed (dizziness, headache, somnolence, nausea, and vomiting), but these were sometimes more frequent compared with those observed with either drug alone. No consistent additive or interaction effects were observed on pharmacodynamic measures. Ethanol had no apparent effects on the pharmacokinetics of morphine or its metabolites. CONCLUSIONS: Coadministration of single doses of morphine and ethanol tested in this study did not affect the safety, pharmacodynamics, or pharmacokinetics of morphine or ethanol administered alone. Copyright © 2014 John Wiley & Sons, Ltd.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Central Nervous System Depressants/pharmacokinetics , Ethanol/pharmacokinetics , Morphine/pharmacokinetics , Administration, Oral , Adult , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Capnography , Central Nervous System Depressants/administration & dosage , Central Nervous System Depressants/adverse effects , Cognition/drug effects , Cross-Over Studies , Double-Blind Method , Drug Interactions , Ethanol/administration & dosage , Ethanol/adverse effects , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Hypnotics and Sedatives/pharmacokinetics , Male , Middle Aged , Morphine/administration & dosage , Morphine/adverse effects , Psychomotor Performance/drug effects , Reflex, Pupillary/drug effects , Substance-Related Disorders/physiopathology , Substance-Related Disorders/psychology , Visual Analog Scale , Young Adult
15.
Clin Pharmacol Drug Dev ; 2(2): 120-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-27121667

ABSTRACT

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.

16.
J Clin Pharmacol ; 52(5): 747-56, 2012 May.
Article in English | MEDLINE | ID: mdl-21593282

ABSTRACT

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.


Subject(s)
Alcohol Drinking/metabolism , Analgesics, Opioid/pharmacokinetics , Ethanol/administration & dosage , Morphine/pharmacokinetics , Naltrexone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Adult , Alcohol Drinking/blood , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/blood , Analgesics, Opioid/chemistry , Analysis of Variance , Area Under Curve , Capsules , Cross-Over Studies , Delayed-Action Preparations , Drug Combinations , Drug Interactions , Ethanol/blood , Female , Humans , Male , Metabolic Clearance Rate , Models, Biological , Morphine/administration & dosage , Morphine/blood , Morphine/chemistry , Naltrexone/administration & dosage , Naltrexone/blood , Naltrexone/chemistry , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/blood , Narcotic Antagonists/chemistry , Young Adult
17.
Drugs R D ; 11(3): 259-75, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21902287

ABSTRACT

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.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Naltrexone/pharmacology , Opioid-Related Disorders/drug therapy , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacokinetics , Cross-Over Studies , Double-Blind Method , Drug Combinations , Drug Interactions , Euphoria/drug effects , Humans , Injections, Intravenous , Male , Morphine/administration & dosage , Morphine/adverse effects , Morphine/pharmacokinetics , Naltrexone/administration & dosage , Naltrexone/adverse effects , Naltrexone/pharmacokinetics , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/adverse effects , Narcotic Antagonists/pharmacokinetics , Opioid-Related Disorders/metabolism , Pain/drug therapy , Pain Measurement/drug effects , Substance-Related Disorders/drug therapy , Substance-Related Disorders/metabolism , Young Adult
18.
Pain Physician ; 14(4): 391-406, 2011.
Article in English | MEDLINE | ID: mdl-21785483

ABSTRACT

BACKGROUND: Morphine sulfate and naltrexone hydrochloride extended-release capsules (EMBEDA, King Pharmaceuticals, Inc., Bristol, TN), indicated for management of chronic, moderate-to-severe pain, contain pellets of extended-release morphine sulfate with a sequestered naltrexone core (MS-sNT). Taken as directed, morphine provides analgesia while naltrexone remains sequestered; if tampered with by crushing, naltrexone is released to mitigate morphine-induced euphoric effects. While it is necessary to establish that formulations intended to reduce attractiveness for abuse are successful in doing so, it is also necessary to demonstrate that product therapeutic integrity is maintained for patients. OBJECTIVES: Data were reviewed from 3 studies to determine: 1) the quantity of naltrexone released when MS-sNT pellets are crushed (MS-sNTC) for at least 2 minutes with mortar and pestle); 2) the extent to which the naltrexone released upon crushing mitigated morphine-induced subjective effects; and 3) whether sequestered naltrexone precipitates opioid withdrawal when MS-sNT is taken as directed. METHODS: The naltrexone bioavailability study compared naltrexone release from MS-sNTC with that from whole intact MS-sNT capsules (MS-sNTW) and an equal naltrexone solution (NS) dose. Equivalent bioavailability was established if 90% confidence intervals (CIs) for geometric mean ratios (maximum plasma naltrexone concentration [Cmax] and area under the concentration-time curve extrapolated to infinity [AUC∞]) fell between 80% and 125%. The oral pharmacodynamic study assessed drug liking and euphoria and pharmacokinetic properties of MS-sNTC and MS-sNTW compared with morphine sulfate solution (MSS) and placebo. The 12-month, open-label (OL) safety study evaluated safety of MS-sNT administered orally as directed in patients with chronic, moderate-to-severe pain. Safety assessments included withdrawal symptoms based on the Clinical Opiate Withdrawal Scale (COWS). RESULTS: Naltrexone from MS-sNTC met criteria for equivalent bioavailability to NS. Although morphine relative bioavailability was similar for MS-sNTC and MSS, mean peak (Emax) visual analog scale (VAS) scores for drug liking and Cole/Addiction Research Center Inventory Stimulation-Euphoria were significantly reduced for MS-sNTC vs MSS (p < 0.001). In these 2 studies, a total of 6 participants had one measurement of plasma naltrexone after MS-sNTW that was above the lower limit of quantification. In the OL safety study, 72/93 participants (77%) had no quantifiable naltrexone concentrations. There was neither evidence of naltrexone accumulation for any participant nor any significant correlation with MS-sNT dose, age, or sex. Of 4 participants with the highest naltrexone concentrations, none had COWS scores consistent with moderate opioid withdrawal symptoms. Only 5 participants had COWS scores consistent with moderate opioid withdrawal; all 5 had not taken MS-sNT as directed. LIMITATIONS: Study populations may not be fully representative of patients receiving opioid therapy for the management of chronic, moderate-to-severe pain and of opioid abusers. CONCLUSIONS: When MS-sNT capsules are crushed, all of the sequestered naltrexone (relative to oral NS) is released and immediately available to mitigate morphine-induced effects. When MS-sNT was crushed, the naltrexone released abated drug liking and euphoria relative to that from an equal dose of immediate-release morphine from MSS administration in a majority of participants. Naltrexone concentrations were low over a period of 12 months without evidence of accumulation, and there were no observable opioid withdrawal symptoms when MS-sNT was taken as directed.


Subject(s)
Morphine/pharmacokinetics , Naltrexone/pharmacokinetics , Area Under Curve , Biological Availability , Capsules , Drug Combinations , Humans , ROC Curve
19.
Am J Ther ; 18(1): 2-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20864883

ABSTRACT

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%.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Morphine/pharmacokinetics , Naltrexone/pharmacokinetics , Adult , Analgesics, Opioid/administration & dosage , Area Under Curve , Biological Availability , Biotransformation , Capsules , Chemistry, Pharmaceutical , Cross-Over Studies , Delayed-Action Preparations , Double-Blind Method , Drug Combinations , Fasting/metabolism , Female , Half-Life , Humans , Male , Middle Aged , Morphine/administration & dosage , Naltrexone/administration & dosage , Therapeutic Equivalency , Young Adult
20.
J Pain Symptom Manage ; 40(5): 734-46, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21075272

ABSTRACT

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.


Subject(s)
Morphine/administration & dosage , Naltrexone/administration & dosage , Pain/drug therapy , Adolescent , Adult , Aged , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Chronic Disease/drug therapy , Constipation/chemically induced , Delayed-Action Preparations , Drug Combinations , Female , Humans , Male , Middle Aged , Morphine/adverse effects , Naltrexone/adverse effects , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/adverse effects , Nausea/chemically induced , Pain Measurement/drug effects , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...