Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
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
2.
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
3.
Genet Med ; 19(4): 430-438, 2017 04.
Article in English | MEDLINE | ID: mdl-27657681

ABSTRACT

PURPOSE: Fabry disease is an X-linked lysosomal storage disorder caused by mutations in the α-galactosidase A gene. Migalastat, a pharmacological chaperone, binds to specific mutant forms of α-galactosidase A to restore lysosomal activity. METHODS: A pharmacogenetic assay was used to identify the α-galactosidase A mutant forms amenable to migalastat. Six hundred Fabry disease-causing mutations were expressed in HEK-293 (HEK) cells; increases in α-galactosidase A activity were measured by a good laboratory practice (GLP)-validated assay (GLP HEK/Migalastat Amenability Assay). The predictive value of the assay was assessed based on pharmacodynamic responses to migalastat in phase II and III clinical studies. RESULTS: Comparison of the GLP HEK assay results in in vivo white blood cell α-galactosidase A responses to migalastat in male patients showed high sensitivity, specificity, and positive and negative predictive values (≥0.875). GLP HEK assay results were also predictive of decreases in kidney globotriaosylceramide in males and plasma globotriaosylsphingosine in males and females. The clinical study subset of amenable mutations (n = 51) was representative of all 268 amenable mutations identified by the GLP HEK assay. CONCLUSION: The GLP HEK assay is a clinically validated method of identifying male and female Fabry patients for treatment with migalastat.Genet Med 19 4, 430-438.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Fabry Disease/genetics , Mutation , alpha-Galactosidase/genetics , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/pharmacology , Biological Assay , Cell Line , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Fabry Disease/drug therapy , Female , HEK293 Cells , Humans , Leukocytes/drug effects , Leukocytes/enzymology , Male , Predictive Value of Tests , Validation Studies as Topic
4.
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
5.
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
6.
Mol Ther ; 23(7): 1169-1181, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25915924

ABSTRACT

Fabry disease is an X-linked lysosomal storage disorder caused by mutations in the gene that encodes α-galactosidase A and is characterized by pathological accumulation of globotriaosylceramide and globotriaosylsphingosine. Earlier, the authors demonstrated that oral coadministration of the pharmacological chaperone AT1001 (migalastat HCl; 1-deoxygalactonojirimycin HCl) prior to intravenous administration of enzyme replacement therapy improved the pharmacological properties of the enzyme. In this study, the authors investigated the effects of coformulating AT1001 with a proprietary recombinant human α-galactosidase A (ATB100) into a single intravenous formulation. AT1001 increased the physical stability and reduced aggregation of ATB100 at neutral pH in vitro, and increased the potency for ATB100-mediated globotriaosylceramide reduction in cultured Fabry fibroblasts. In Fabry mice, AT1001 coformulation increased the total exposure of active enzyme, and increased ATB100 levels in cardiomyocytes, cardiac vascular endothelial cells, renal distal tubular epithelial cells, and glomerular cells, cell types that do not show substantial uptake with enzyme replacement therapy alone. Notably, AT1001 coformulation also leads to greater tissue globotriaosylceramide reduction when compared with ATB100 alone, which was positively correlated with reductions in plasma globotriaosylsphingosine. Collectively, these data indicate that intravenous administration of ATB100 coformulated with AT1001 may provide an improved therapy for Fabry disease and thus warrants further investigation.


Subject(s)
Fabry Disease/drug therapy , Molecular Chaperones/administration & dosage , Oligopeptides/administration & dosage , alpha-Galactosidase/administration & dosage , Animals , Disease Models, Animal , Drug Combinations , Enzyme Replacement Therapy , Fabry Disease/pathology , Fibroblasts/drug effects , Humans , Mice , Mutation , Substrate Specificity
7.
Neurotherapeutics ; 11(4): 840-56, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25037721

ABSTRACT

Mutation of the lysosomal hydrolase acid-ß-glucosidase (GCase), which leads to reduced GCase activity, is one of the most frequent genetic risk factors for Parkinson's disease (PD) and promotes α-synuclein accumulation in the brain, a hallmark of PD and other synucleinopathies. Whether targeting GCase pharmacologically is a valid therapeutic strategy for sporadic PD in the absence of GCase mutation is unknown. We have investigated whether increasing the stability, trafficking, and activity of wild-type GCase could be beneficial in synucleinopathies by administering the pharmacological chaperone AT2101 (afegostat-tartrate, isofagomine) to mice that overexpress human wild-type α-synuclein (Thy1-aSyn mice). AT2101 administered orally for 4 months to Thy1-aSyn mice improved motor and nonmotor function, abolished microglial inflammatory response in the substantia nigra, reduced α-synuclein immunoreactivity in nigral dopaminergic neurons, and reduced the number of small α-synuclein aggregates, while increasing the number of large α-synuclein aggregates. These data support the further investigation of pharmacological chaperones that target GCase as a therapeutic approach for sporadic PD and other synucleinopathies, even in the absence of glucocerebrosidase mutations.


Subject(s)
Brain/drug effects , Brain/enzymology , Parkinson Disease/enzymology , Parkinson Disease/prevention & control , alpha-Synuclein/metabolism , beta-Glucosidase/antagonists & inhibitors , Animals , Brain/metabolism , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Humans , Imino Pyranoses/pharmacology , Male , Mice , Motor Activity/drug effects , Protein Aggregates/drug effects , Protein Transport/drug effects , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tartrates
8.
PLoS One ; 9(7): e102092, 2014.
Article in English | MEDLINE | ID: mdl-25036864

ABSTRACT

Pompe disease is an inherited lysosomal storage disorder that results from a deficiency in acid α-glucosidase (GAA) activity due to mutations in the GAA gene. Pompe disease is characterized by accumulation of lysosomal glycogen primarily in heart and skeletal muscles, which leads to progressive muscle weakness. We have shown previously that the small molecule pharmacological chaperone AT2220 (1-deoxynojirimycin hydrochloride, duvoglustat hydrochloride) binds and stabilizes wild-type as well as multiple mutant forms of GAA, and can lead to higher cellular levels of GAA. In this study, we examined the effect of AT2220 on mutant GAA, in vitro and in vivo, with a primary focus on the endoplasmic reticulum (ER)-retained P545L mutant form of human GAA (P545L GAA). AT2220 increased the specific activity of P545L GAA toward both natural (glycogen) and artificial substrates in vitro. Incubation with AT2220 also increased the ER export, lysosomal delivery, proteolytic processing, and stability of P545L GAA. In a new transgenic mouse model of Pompe disease that expresses human P545L on a Gaa knockout background (Tg/KO) and is characterized by reduced GAA activity and elevated glycogen levels in disease-relevant tissues, daily oral administration of AT2220 for 4 weeks resulted in significant and dose-dependent increases in mature lysosomal GAA isoforms and GAA activity in heart and skeletal muscles. Importantly, oral administration of AT2220 also resulted in significant glycogen reduction in disease-relevant tissues. Compared to daily administration, less-frequent AT2220 administration, including repeated cycles of 4 or 5 days with AT2220 followed by 3 or 2 days without drug, respectively, resulted in even greater glycogen reductions. Collectively, these data indicate that AT2220 increases the specific activity, trafficking, and lysosomal stability of P545L GAA, leads to increased levels of mature GAA in lysosomes, and promotes glycogen reduction in situ. As such, AT2220 may warrant further evaluation as a treatment for Pompe disease.


Subject(s)
1-Deoxynojirimycin/pharmacology , Glucan 1,4-alpha-Glucosidase/genetics , Glucan 1,4-alpha-Glucosidase/metabolism , Glycogen Storage Disease Type II/metabolism , Glycogen/metabolism , Lysosomes/drug effects , Mutation , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/pharmacokinetics , Administration, Oral , Animals , Biocatalysis/drug effects , Biological Availability , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Enzyme Stability/drug effects , Gene Knockout Techniques , Glucan 1,4-alpha-Glucosidase/biosynthesis , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/pathology , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Lysosomes/metabolism , Mice , Mice, Transgenic , Mutant Proteins/biosynthesis , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Transport/drug effects , Proteolysis/drug effects
9.
J Am Heart Assoc ; 3(1): e000394, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24496231

ABSTRACT

BACKGROUND: Elevated urinary globotriaosylceramide (Gb3) has been considered a hallmark of Fabry disease, an X-linked lysosomal disorder that is a risk factor for most types of heart disease. METHODS AND RESULTS: We screened 1421 consecutive patients with common forms of heart disease for Fabry disease by measuring urinary Gb3 in whole urine using tandem mass spectrometry, α-galactosidase A activity in dried blood spots, and we looked for GLA mutations by parallel sequencing of the whole gene (exons and introns) in pooled genomic DNA samples followed by Sanger sequencing verification. GLA variants were found in 13 patients. In the 1408 patients without GLA mutations, urinary Gb3 levels were significantly higher in heart disease patients compared to 116 apparently healthy controls (median difference=10.0 ng/mL and P<0.001). Urinary lipid profiling showed that levels of 5 other lipids significantly distinguished between urine of patients with Fabry disease (n=7) and heart disease patients with elevated urinary Gb3 (n=6). Sphingomyelin and Gb3 levels were abnormal in the left ventricular wall of patients with ischemic heart failure. Elevated levels of urinary Gb3 were independently associated with increased risk of death in the average follow-up of 17 months (hazard ratio=1.59 for increase in Gb3 of 200, 95% CI=1.36 and 1.87, and P<0.0001). CONCLUSIONS: In heart disease patients who do not have Fabry disease or GLA gene mutations, a higher level of urinary Gb3 is positively associated with near-term mortality. The elevation of urinary Gb3 and that of other lipids suggests that heart disease is associated with multiorgan lipid abnormalities. CLINICAL TRIAL REGISTRATION URL: clinicaltrials.gov. Unique Identifier: NCT01019629.


Subject(s)
Fabry Disease/mortality , Fabry Disease/urine , Heart Diseases/mortality , Heart Diseases/urine , Trihexosylceramides/urine , Adult , Aged , Biomarkers/urine , Case-Control Studies , Clinical Enzyme Tests , DNA Mutational Analysis , Fabry Disease/complications , Fabry Disease/diagnosis , Fabry Disease/genetics , Female , Heart Diseases/diagnosis , Heart Diseases/etiology , Humans , Male , Middle Aged , Mutation , Prognosis , Prospective Studies , Risk Factors , Tandem Mass Spectrometry , Up-Regulation , alpha-Galactosidase/blood , alpha-Galactosidase/genetics
10.
PLoS One ; 8(3): e57631, 2013.
Article in English | MEDLINE | ID: mdl-23472096

ABSTRACT

Fabry disease (FD) results from mutations in the gene (GLA) that encodes the lysosomal enzyme α-galactosidase A (α-Gal A), and involves pathological accumulation of globotriaosylceramide (GL-3) and globotriaosylsphingosine (lyso-Gb3). Migalastat hydrochloride (GR181413A) is a pharmacological chaperone that selectively binds, stabilizes, and increases cellular levels of α-Gal A. Oral administration of migalastat HCl reduces tissue GL-3 in Fabry transgenic mice, and in urine and kidneys of some FD patients. A liquid chromatography-tandem mass spectrometry method was developed to measure lyso-Gb3 in mouse tissues and human plasma. Oral administration of migalastat HCl to transgenic mice reduced elevated lyso-Gb3 levels up to 64%, 59%, and 81% in kidney, heart, and skin, respectively, generally equal to or greater than observed for GL-3. Furthermore, baseline plasma lyso-Gb3 levels were markedly elevated in six male FD patients enrolled in Phase 2 studies. Oral administration of migalastat HCl (150 mg QOD) reduced urine GL-3 and plasma lyso-Gb3 in three subjects (range: 15% to 46% within 48 weeks of treatment). In contrast, three showed no reductions in either substrate. These results suggest that measurement of tissue and/or plasma lyso-Gb3 is feasible and may be warranted in future studies of migalastat HCl or other new potential therapies for FD.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Fabry Disease/genetics , Glycolipids/metabolism , Sphingolipids/metabolism , Sphingosine/metabolism , Trihexosylceramides/metabolism , 1-Deoxynojirimycin/pharmacology , Administration, Oral , Animals , Fabry Disease/blood , Fabry Disease/drug therapy , Glycolipids/blood , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Reproducibility of Results , Sphingolipids/blood , Trihexosylceramides/blood , alpha-Galactosidase/genetics
11.
Orphanet J Rare Dis ; 7: 91, 2012 Nov 24.
Article in English | MEDLINE | ID: mdl-23176611

ABSTRACT

BACKGROUND: Fabry disease (FD) is a genetic disorder resulting from deficiency of the lysosomal enzyme α-galactosidase A (α-Gal A), which leads to globotriaosylceramide (GL-3) accumulation in multiple tissues. We report on the safety and pharmacodynamics of migalastat hydrochloride, an investigational pharmacological chaperone given orally at 150 mg every-other-day. METHODS: Two open-label uncontrolled phase 2 studies of 12 and 24 weeks (NCT00283959 and NCT00283933) in 9 males with FD were combined. At multiple time points, α-Gal A activity and GL-3 levels were quantified in blood cells, kidney and skin. GL-3 levels were also evaluated through skin and renal histology. RESULTS: Compared to baseline, increased α-Gal A activity of at least 50% was demonstrated in blood, skin and kidney in 6 of 9 patients. Patients' increased α-Gal A activities paralleled the α-Gal A increases observed in vitro in HEK-293 cells transfected with the corresponding mutant form of the enzyme. The same 6 patients who demonstrated increases of α-Gal A activity also had GL-3 reduction in skin, urine and/or kidney, and had α-Gal A mutations that responded in transfected cells incubated with the drug. The 3 patients who did not show a consistent response in vivo had α-Gal A mutations that did not respond to migalastat HCl in transfected cells. Migalastat HCl was well tolerated. CONCLUSIONS: Migalastat HCl is a candidate pharmacological chaperone that provides a novel genotype-specific treatment for FD. It enhanced α-Gal A activity and resulted in GL-3 substrate decrease in patients with responsive GLA mutations. Phase 3 studies are ongoing.


Subject(s)
Fabry Disease/drug therapy , Molecular Chaperones/therapeutic use , Trihexosylceramides/metabolism , alpha-Galactosidase/metabolism , Fabry Disease/enzymology , Fabry Disease/metabolism , Fabry Disease/pathology , Humans , Male , Molecular Chaperones/adverse effects
12.
PLoS One ; 7(7): e40776, 2012.
Article in English | MEDLINE | ID: mdl-22815812

ABSTRACT

Pompe disease is an inherited lysosomal storage disease that results from a deficiency in the enzyme acid α-glucosidase (GAA), and is characterized by progressive accumulation of lysosomal glycogen primarily in heart and skeletal muscles. Recombinant human GAA (rhGAA) is the only approved enzyme replacement therapy (ERT) available for the treatment of Pompe disease. Although rhGAA has been shown to slow disease progression and improve some of the pathophysiogical manifestations, the infused enzyme tends to be unstable at neutral pH and body temperature, shows low uptake into some key target tissues, and may elicit immune responses that adversely affect tolerability and efficacy. We hypothesized that co-administration of the orally-available, small molecule pharmacological chaperone AT2220 (1-deoxynojirimycin hydrochloride, duvoglustat hydrochloride) may improve the pharmacological properties of rhGAA via binding and stabilization. AT2220 co-incubation prevented rhGAA denaturation and loss of activity in vitro at neutral pH and 37°C in both buffer and blood. In addition, oral pre-administration of AT2220 to rats led to a greater than two-fold increase in the circulating half-life of intravenous rhGAA. Importantly, co-administration of AT2220 and rhGAA to GAA knock-out (KO) mice resulted in significantly greater rhGAA levels in plasma, and greater uptake and glycogen reduction in heart and skeletal muscles, compared to administration of rhGAA alone. Collectively, these preclinical data highlight the potentially beneficial effects of AT2220 on rhGAA in vitro and in vivo. As such, a Phase 2 clinical study has been initiated to investigate the effects of co-administered AT2220 on rhGAA in Pompe patients.


Subject(s)
1-Deoxynojirimycin/therapeutic use , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/enzymology , Glycogen/metabolism , Recombinant Proteins/metabolism , alpha-Glucosidases/metabolism , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/pharmacology , Animals , Buffers , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Stability/drug effects , Half-Life , Humans , Mice , Mice, Knockout , Protein Denaturation/drug effects , Rats , Recombinant Proteins/blood , alpha-Glucosidases/administration & dosage , alpha-Glucosidases/blood
13.
J Neurosci ; 32(15): 5223-36, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22496568

ABSTRACT

Alterations in the lipid composition of endosomal-lysosomal membranes may constitute an early event in Alzheimer's disease (AD) pathogenesis. In this study, we investigated the possibility that GM2 ganglioside accumulation in a mouse model of Sandhoff disease might be associated with the accumulation of intraneuronal and extracellular proteins commonly observed in AD. Our results show intraneuronal accumulation of amyloid-ß peptide (Aß)-like, α-synuclein-like, and phospho-tau-like immunoreactivity in the brains of ß-hexosaminidase knock-out (HEXB KO) mice. Biochemical and immunohistochemical analyses confirmed that at least some of the intraneuronal Aß-like immunoreactivity (iAß-LIR) represents amyloid precursor protein C-terminal fragments (APP-CTFs) and/or Aß. In addition, we observed increased levels of Aß40 and Aß42 peptides in the lipid-associated fraction of HEXB KO mouse brains, and intraneuronal accumulation of ganglioside-bound Aß (GAß) immunoreactivity in a brain region-specific manner. Furthermore, α-synuclein and APP-CTFs and/or Aß were found to accumulate in different regions of the substantia nigra, indicating different mechanisms of accumulation or turnover pathways. Based on the localization of the accumulated iAß-LIR to endosomes, lysosomes, and autophagosomes, we conclude that a significant accumulation of iAß-LIR may be associated with the lysosomal-autophagic turnover of Aß and fragments of APP-containing Aß epitopes. Importantly, intraneuronal GAß immunoreactivity, a proposed prefibrillar aggregate found in AD, was found to accumulate throughout the frontal cortices of postmortem human GM1 gangliosidosis, Sandhoff disease, and Tay-Sachs disease brains. Together, these results establish an association between the accumulation of gangliosides, autophagic vacuoles, and the intraneuronal accumulation of proteins associated with AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Gangliosides/metabolism , Hexosaminidase B/genetics , Lysosomes/physiology , Sandhoff Disease/pathology , Adult , Animals , Blotting, Western , Brain Chemistry/genetics , Brain Chemistry/physiology , Child, Preschool , G(M2) Ganglioside/metabolism , Humans , Immunohistochemistry , Infant , Lipid Metabolism , Medulla Oblongata/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Spinal Cord/metabolism , Substantia Nigra/metabolism , Young Adult , alpha-Synuclein/metabolism , tau Proteins/metabolism
14.
Mol Ther ; 20(4): 717-26, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22215019

ABSTRACT

Fabry disease is an X-linked lysosomal storage disorder (LSD) caused by mutations in the gene (GLA) that encodes the lysosomal hydrolase α-galactosidase A (α-Gal A), and is characterized by pathological accumulation of the substrate, globotriaosylceramide (GL-3). Regular infusion of recombinant human α-Gal A (rhα-Gal A), termed enzyme replacement therapy (ERT), is the primary treatment for Fabry disease. However, rhα-Gal A has low physical stability, a short circulating half-life, and variable uptake into different disease-relevant tissues. We hypothesized that coadministration of the orally available, small molecule pharmacological chaperone AT1001 (GR181413A, 1-deoxygalactonojirimycin, migalastat hydrochloride) may improve the pharmacological properties of rhα-Gal A via binding and stabilization. AT1001 prevented rhα-Gal A denaturation and activity loss in vitro at neutral pH and 37 °C. Coincubation of Fabry fibroblasts with rhα-Gal A and AT1001 resulted in up to fourfold higher cellular α-Gal A and ~30% greater GL-3 reduction compared to rhα-Gal A alone. Furthermore, coadministration of AT1001 to rats increased the circulating half-life of rhα-Gal A by >2.5-fold, and in GLA knockout mice resulted in up to fivefold higher α-Gal A levels and fourfold greater GL-3 reduction than rhα-Gal A alone. Collectively, these data highlight the potentially beneficial effects of AT1001 on rhα-Gal A, thus warranting clinical investigation.


Subject(s)
Enzyme Replacement Therapy/methods , Fabry Disease/drug therapy , Oligopeptides/therapeutic use , Recombinant Proteins/therapeutic use , alpha-Galactosidase/therapeutic use , Animals , Blotting, Western , Fabry Disease/metabolism , Fluorescent Antibody Technique , Humans , Mice , Rats , Trihexosylceramides/metabolism
15.
Hum Mutat ; 32(8): 965-77, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21598360

ABSTRACT

Fabry disease is caused by mutations in the gene (GLA) that encodes α-galactosidase A (α-Gal A). The iminosugar AT1001 (GR181413A, migalastat hydrochloride, 1-deoxygalactonojirimycin) is a pharmacological chaperone that selectively binds and stabilizes α-Gal A, increasing total cellular levels and activity for some mutant forms (defined as "responsive"). In this study, we developed a cell-based assay in cultured HEK-293 cells to identify mutant forms of α-Gal A that are responsive to AT1001. Concentration-dependent increases in α-Gal A activity in response to AT1001 were shown for 49 (60%) of 81 mutant forms. The responses of α-Gal A mutant forms were generally consistent with the responses observed in male Fabry patient-derived lymphoblasts. Importantly, the HEK-293 cell responses of 19 α-Gal A mutant forms to a clinically achievable concentration of AT1001 (10 µM) were generally consistent with observed increases in α-Gal A activity in peripheral blood mononuclear cells from male Fabry patients orally administered AT1001 during Phase 2 clinical studies. This indicates that the cell-based responses can identify mutant forms of α-Gal A that are likely to respond to AT1001 in vivo. Thus, the HEK-293 cell-based assay may be a useful aid in the identification of Fabry patients with AT1001-responsive mutant forms.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Fabry Disease/genetics , Mutant Proteins/analysis , alpha-Galactosidase/genetics , 1-Deoxynojirimycin/metabolism , 1-Deoxynojirimycin/pharmacology , Biological Assay , Enzyme Activation/drug effects , Fabry Disease/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Mutant Proteins/metabolism , Point Mutation/genetics , Protein Conformation , alpha-Galactosidase/chemistry , alpha-Galactosidase/metabolism
16.
FEBS J ; 277(7): 1618-38, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20148966

ABSTRACT

Gaucher disease is caused by mutations in the gene that encodes the lysosomal enzyme acid beta-glucosidase (GCase). We have shown previously that the small molecule pharmacological chaperone isofagomine (IFG) binds and stabilizes N370S GCase, resulting in increased lysosomal trafficking and cellular activity. In this study, we investigated the effect of IFG on L444P GCase. Incubation of Gaucher patient-derived lymphoblastoid cell lines (LCLs) or fibroblasts with IFG led to approximately 3.5- and 1.3-fold increases in L444P GCase activity, respectively, as measured in cell lysates. The effect in fibroblasts was increased approximately 2-fold using glycoprotein-enrichment, GCase-immunocapture, or by incubating cells overnight in IFG-free media prior to assay, methods designed to maximize GCase activity by reducing IFG carryover and inhibition in the enzymatic assay. IFG incubation also increased the lysosomal trafficking and in situ activity of L444P GCase in intact cells, as measured by reduction in endogenous glucosylceramide levels. Importantly, this reduction was seen only following three-day incubation in IFG-free media, underscoring the importance of IFG removal to restore lysosomal GCase activity. In mice expressing murine L444P GCase, oral administration of IFG resulted in significant increases (2- to 5-fold) in GCase activity in disease-relevant tissues, including brain. Additionally, eight-week IFG administration significantly lowered plasma chitin III and IgG levels, and 24-week administration significantly reduced spleen and liver weights. Taken together, these data suggest that IFG can increase the lysosomal activity of L444P GCase in cells and tissues. Moreover, IFG is orally available and distributes into multiple tissues, including brain, and may thus merit therapeutic evaluation for patients with neuronopathic and non-neuronopathic Gaucher disease.


Subject(s)
Gaucher Disease/genetics , Imino Pyranoses/chemistry , Lysosomal Storage Diseases/genetics , Mutation , beta-Glucosidase/genetics , Animals , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Glucosylceramidase/metabolism , Humans , Male , Mice , Microscopy, Confocal/methods , Molecular Chaperones/metabolism , Rats , Rats, Sprague-Dawley
17.
Mol Ther ; 18(1): 23-33, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19773742

ABSTRACT

Fabry disease is an X-linked lysosomal storage disorder caused by a deficiency in alpha-galactosidase A (alpha-Gal A) activity and subsequent accumulation of the substrate globotriaosylceramide (GL-3), which contributes to disease pathology. The pharmacological chaperone (PC) DGJ (1-deoxygalactonojirimycin) binds and stabilizes alpha-Gal A, increasing enzyme levels in cultured cells and in vivo. The ability of DGJ to reduce GL-3 in vivo was investigated using transgenic (Tg) mice that express a mutant form of human alpha-Gal A (R301Q) on a knockout background (Tg/KO), which leads to GL-3 accumulation in disease-relevant tissues. Four-week daily oral administration of DGJ to Tg/KO mice resulted in significant and dose-dependent increases in alpha-Gal A activity, with concomitant GL-3 reduction in skin, heart, kidney, brain, and plasma; 24-week administration resulted in even greater reductions. Compared to daily administration, less frequent DGJ administration, including repeated cycles of 4 days with DGJ followed by 3 days without or every other day with DGJ, resulted in even greater GL-3 reductions that were comparable to those obtained with Fabrazyme. Collectively, these data indicate that oral administration of DGJ increases mutant alpha-Gal A activity and reduces GL-3 in disease-relevant tissues in Tg/KO mice, and thus merits further evaluation as a treatment for Fabry disease.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Fabry Disease/drug therapy , Trihexosylceramides/metabolism , 1-Deoxynojirimycin/therapeutic use , Animals , Blotting, Western , Disease Models, Animal , Fabry Disease/genetics , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , alpha-Galactosidase/antagonists & inhibitors , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism
18.
Hum Mutat ; 30(12): 1683-92, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19862843

ABSTRACT

Pompe disease is a lysosomal storage disorder (LSD) caused by mutations in the gene that encodes acid alpha-glucosidase (GAA). Recently, small molecule pharmacological chaperones have been shown to increase protein stability and cellular levels for mutant lysosomal enzymes and have emerged as a new therapeutic strategy for the treatment of LSDs. In this study, we characterized the pharmacological chaperone 1-deoxynojirimycin (DNJ) on 76 different mutant forms of GAA identified in Pompe disease. DNJ significantly increased enzyme activity and protein levels for 16 different GAA mutants in patient-derived fibroblasts and in transiently transfected COS-7 cells. Additionally, DNJ increased the processing of these GAA mutants to their mature lysosomal forms, suggesting facilitated trafficking through the secretory pathway. Immunofluorescence microscopy studies showed increased colocalization of GAA with the lysosomal marker LAMP2 after incubation with DNJ, confirming increased lysosomal trafficking. Lastly, a GAA structural model was constructed based on the related eukaryotic glucosidase maltase-glucoamylase. The mutated residues identified in responsive forms of GAA are located throughout most of the structural domains, with half of these residues located in two short regions within the catalytic domain. Taken together, these data support further evaluation of DNJ as a potential treatment for Pompe disease in patients that express responsive forms of GAA.


Subject(s)
1-Deoxynojirimycin/pharmacology , Lysosomes/drug effects , Lysosomes/enzymology , Mutant Proteins/metabolism , alpha-Glucosidases/metabolism , Adolescent , Adult , Animals , COS Cells , Chlorocebus aethiops , Enzyme Stability/drug effects , Fibroblasts/drug effects , Fibroblasts/enzymology , Glycogen Storage Disease Type II/enzymology , Humans , Infant , Models, Molecular , Protein Structure, Secondary , Protein Transport/drug effects , Recombinant Proteins/metabolism , alpha-Glucosidases/chemistry
20.
J Am Chem Soc ; 130(51): 17568-74, 2008 Dec 24.
Article in English | MEDLINE | ID: mdl-19053485

ABSTRACT

The upstream protein kinases responsible for thousands of phosphorylation events in the phosphoproteome remain to be discovered. We developed a three-component chemical reaction which converts the transient noncovalent substrate-kinase complex into a covalently cross-linked product by utilizing a dialdehyde-based cross-linker, 1. Unfortunately, the reaction of 1 with a lysine in the kinase active site and an engineered cysteine on the substrate to form an isoindole cross-linked product could not be performed in the presence of competing cellular proteins due to nonspecific side reactions. In order to more selectively target the cross-linker to protein kinases in cell lysates, we replaced the weak, kinase-binding adenosine moiety of 1 with a potent protein kinase inhibitor scaffold. In addition, we replaced the o-phthaldialdehyde moiety in 1 with a less-reactive thiophene-2,3-dicarboxaldehyde moiety. The combination of these two structural modifications provides for cross-linking of a cysteine-containing substrate to its corresponding kinase in the presence of competing cellular proteins.


Subject(s)
Phosphotransferases/chemistry , Adenosine/chemistry , Cross-Linking Reagents/chemistry , Cysteine/chemistry , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Lysine/chemistry , Models, Chemical , Molecular Conformation , Peptides/chemistry , Phosphorylation , Phosphotransferases/metabolism , Protein Structure, Tertiary , Proteins/chemistry , Thiophenes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...