Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
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
2.
J Inherit Metab Dis ; 42(3): 534-544, 2019 05.
Article in English | MEDLINE | ID: mdl-30834538

ABSTRACT

Pegunigalsidase alfa, a novel PEGylated, covalently crosslinked form of α-galactosidase A developed as enzyme replacement therapy (ERT) for Fabry disease (FD), was designed to increase plasma half-life and reduce immunogenicity, thereby enhancing efficacy compared with available products. Symptomatic adults with FD participated in this open-label, 3-month dose-ranging study, followed by a 9-month extension. Three cohorts were enrolled in a stepwise manner, each receiving increased doses of pegunigalsidase alfa: 0.2, 1.0, 2.0 mg/kg, via intravenous infusion every other week. Pharmacokinetic analysis occurred on Day 1 and Months 3, 6, and 12. Kidney biopsies at baseline and Month 6 assessed peritubular capillary globotriaosylceramide (Gb3) content. Renal function, cardiac parameters, and other clinical endpoints were assessed throughout. Treatment-emergent adverse events (AEs) and presence of immunoglobulin G (IgG) antidrug antibodies (ADAs) were assessed. Sixteen patients completed 1 year's treatment. Mean terminal plasma half-life (each cohort) ranged from 53 to 121 hours. All 11 male and 1 of 7 female patients presented with classic FD phenotype, in whom renal peritubular capillary Gb3 inclusions were reduced by 84%. Mean estimated glomerular filtration rate was 111 mL/min/1.73 m2 at baseline, remaining stable throughout treatment. Three patients developed treatment-induced IgG ADAs; following 1 year's treatment, all became ADA-negative. Nearly all treatment-emergent AEs were mild or moderate. One patient withdrew from the study following a serious related AE. Pegunigalsidase alfa may represent an advance in ERT for FD, based on its unique pharmacokinetics and apparent low immunogenicity.


Subject(s)
Enzyme Replacement Therapy , Fabry Disease/drug therapy , Trihexosylceramides/metabolism , alpha-Galactosidase/administration & dosage , alpha-Galactosidase/pharmacokinetics , Adolescent , Adult , Female , Glomerular Filtration Rate , Heart/physiopathology , Humans , Internationality , Kidney/physiopathology , Male , Middle Aged , Treatment Outcome , Young Adult
3.
J Inherit Metab Dis ; 42(3): 527-533, 2019 05.
Article in English | MEDLINE | ID: mdl-30746723

ABSTRACT

Moss-aGalactosidase A (moss-aGal) is a moss-derived version of human α-galactosidase developed for enzyme replacement therapy in patients with Fabry disease. It exhibits a homogenous N-glycosylation profile with >90% mannose-terminated glycans. In contrast to mammalian cell produced α-galactosidase, moss-aGal does not rely on mannose-6-phosphate receptor mediated endocytosis but targets the mannose receptor for tissue uptake. We conducted a phase 1 clinical trial with moss-aGal in six patients with confirmed diagnosis of Fabry disease during a 28-day schedule. All patients received a single dose of 0.2 mg/kg moss-aGal by i.v.-infusion. Primary endpoints of the trial were safety and pharmacokinetics; secondary endpoints were pharmacodynamics by analyzing urine and plasma Gb3 and lyso-Gb3 concentrations. In all patients, the administered single dose was well tolerated. No safety issues were observed. Pharmacokinetic data revealed a stable nonlinear profile with a short plasma half-life of moss-aGal of 14 minutes. After one single dose of moss-aGal, urinary Gb3 concentrations decreased up to 23% 7 days and up to 60% 28 days post-dose. Plasma concentrations of lyso-Gb3 decreased by 3.8% and of Gb3 by 11% 28 days post-dose. These data reveal that a single dose of moss-aGal was safe, well tolerated, and led to a prolonged reduction of Gb3 excretion. As previously shown, moss-aGal is taken up via the mannose receptor, which is expressed on macrophages but also on endothelial and kidney cells. Thus, these data indicate that moss-aGal may target kidney cells. After these promising results, phase 2/3 clinical trials are in preparation.


Subject(s)
Enzyme Replacement Therapy , Fabry Disease/drug therapy , Glycolipids/blood , Glycolipids/urine , Sphingolipids/blood , Sphingolipids/urine , alpha-Galactosidase/pharmacology , alpha-Galactosidase/pharmacokinetics , Adult , Fabry Disease/blood , Fabry Disease/urine , Female , Germany , Humans , Infusions, Intravenous , Male , Middle Aged , Treatment Outcome
4.
Biochem Biophys Res Commun ; 501(3): 648-653, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29746864

ABSTRACT

Cell-penetrating peptides (CPPs) can deliver payloads into cells by forming complexes with bioactive molecules via covalent or non-covalent bonds. Various CPPs have been applied in CPP-modified liposomes, and their effectiveness is highly regarded in liposomal drug delivery systems (DDSs). Previously, we have reported on the polyhistidine peptide (H16 peptide: HHHHHHHHHHHHHHHH-NH2) as a new CPP. The H16 peptide has a higher cell-penetrating capacity than well-known CPPs and delivers small molecules such as fluorescent dyes, bioactive peptides, and proteins into mammalian cells. However, it is not known whether the H16 peptide can deliver large cargos such as liposomes into cells. To assess the potential of the H16 peptide, in this study, we developed H16 peptide-modified liposomes (H16-Lipo) and evaluated their effectiveness in a liposomal DDS. The H16-Lipo was prepared by inserting a stearyl-H16 peptide into the hydrophobic region of a liposome. The H16-Lipo was internalized into human fibrosarcoma cells via multiple endocytosis pathways and localized to intracellular lysosomes. Based on this result, we used the H16-Lipo as a lysosome-targeting DDS. The H16-Lipo delivered alpha-galactosidase A (GLA), one of the lysosomal enzymes, to intracellular lysosomes and improved the proliferation of GLA-knockdown cells. These results suggest that the H16-Lipo is an effective drug carrier for lysosomal enzymes in a lysosome-targeting DDS. The loss of lysosomal enzymes has been known to induce metabolic disorders, called lysosomal storage diseases (LSDs). Our findings indicate that this combination of the H16 peptide and a liposome is a promising candidate as a DDS for the treatment of LSDs.


Subject(s)
Histidine/metabolism , Liposomes/metabolism , Lysosomes/metabolism , alpha-Galactosidase/administration & dosage , Cell Line, Tumor , Drug Delivery Systems , Histidine/chemistry , Humans , Liposomes/chemistry , alpha-Galactosidase/pharmacokinetics
5.
Bioconjug Chem ; 29(5): 1630-1639, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29698600

ABSTRACT

The current treatment of Fabry disease by enzyme replacement therapy with commercially available recombinant human α-Galactosidase A shows a continuous deterioration of the disease patients. Human recombinant α-Galactosidase A is a homodimer with noncovalently bound subunits and is expressed in the ProCellEx plant cell-based protein expression platform to produce pegunigalsidase alfa. The effect of covalent bonding between two α-Galactosidase A subunits by PEG-based cross-linkers of various lengths was evaluated in this study. The results show that cross-linking by a bifunctional PEG polymer of 2000 Da produces a more stable protein with improved pharmacokinetic and biodistribution properties. The chemical modification did not influence the tertiary protein structure but led to an increased thermal stability and showed partial masking of immune epitopes. The developed pegunigalsidase alfa is currently tested in phase III clinical trials and has a potential to show superior efficacy versus the currently used enzyme replacement therapies in the treatment of Fabry disease patients.


Subject(s)
Cross-Linking Reagents/chemistry , Polyethylene Glycols/chemistry , alpha-Galactosidase/chemistry , Animals , Cell Line , Enzyme Stability , Fabry Disease/drug therapy , Humans , Mice , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Tissue Distribution , Nicotiana/genetics , alpha-Galactosidase/genetics , alpha-Galactosidase/pharmacokinetics , alpha-Galactosidase/therapeutic use
6.
Mol Genet Metab ; 114(2): 259-67, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25155442

ABSTRACT

Fabry disease is an X-linked recessive disorder caused by the loss of function of the lysosomal enzyme α-Galactosidase-A. Although two enzyme replacement therapies (ERTs) are commercially available, they may not effectively reverse some of the Fabry pathology. PRX-102 is a novel enzyme for the therapy of Fabry disease expressed in a BY2 Tobacco cell culture. PRX-102 is chemically modified, resulting in a cross-linked homo-dimer. We have characterized the in-vitro and in-vivo properties of PRX-102 and compared the results with the two commercially produced α-Galactosidase-A enzymes. Results show that PRX-102 has prolonged in-vitro stability in plasma, after 1h incubation it retains 30% activity compared with complete inactivation of the commercial enzymes. Under lysosomal-like conditions PRX-102 maintains over 80% activity following 10 days of incubation, while commercial enzymes become inactive after 2days. Pharmacokinetic profile of PRX-102 measured in male Fabry mice shows a 10 fold increase in t1/2 in mice (581min) compared to approved drugs. The enzyme has significantly different kinetic parameters to the alternative ERTs available (p-value<0.05, one way ANOVA), although these differences do not indicate any significant biochemical variations. PRX-102 is uptaken to primary human Fabry fibroblasts. The repeat administration of the enzyme to Fabry mice caused significant reduction (p-value<0.05) of Gb3 in various tissues (the measured residual content was 64% in kidney, liver was cleaned, 23% in heart, 5.7% in skin and 16.2% in spleen). PRX-102 has a relatively simple glycosylation pattern, characteristic to plants, having mainly tri-mannose structures with the addition of either α(1-3)-linked fucose or ß(1-2)-linked xylose, or both, in addition to various high mannose structures, while agalsidase beta has a mixture of sialylated glycans in addition to high mannose structures. This study concludes that PRX-102 is equivalent in functionality to the current ERTs available, with superior stability and prolonged circulatory half-life. Therefore we propose that PRX-102 is a promising alternative for treatment of Fabry disease.


Subject(s)
Enzyme Replacement Therapy , Fabry Disease/drug therapy , alpha-Galactosidase/genetics , alpha-Galactosidase/therapeutic use , Animals , Cells, Cultured , Enzyme Stability , Heart , Isoenzymes/therapeutic use , Kidney/enzymology , Liver/enzymology , Male , Mice , Recombinant Proteins/therapeutic use , Skin/enzymology , Spleen/enzymology , Nicotiana/genetics , alpha-Galactosidase/pharmacokinetics
7.
J Clin Pharmacol ; 54(6): 675-81, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24408305

ABSTRACT

ISU303 is a new recombinant agalsidase beta (Agal) enzyme replacement therapy under investigation for Fabry disease, caused by a deficiency in α-galactosidase A activity that leads to fatty deposits in tissues. We evaluated the pharmacokinetic (PK) parameters, safety and tolerability of ISU303 in healthy adult volunteers. The study was a dose block-randomized, double-blinded, placebo-controlled, single-dosing, and dose escalation phase 1 clinical trial. A total of 18 healthy subjects were enrolled (0.3 mg/kg, n = 6; 1.0 mg/kg, n = 6; placebo, n = 6). Blood samples for PK analysis were collected according to planned time. The PK parameters in each 0.3 and 1.0 mg/kg Agal group were as follows: Cmax (mU/mL) 43.19 ± 5.9 and 195.86 ± 32.3; AUClast (h·mU/mL) 207.91 ± 25.1 and 939.96 ± 158.3; t1/2 (hours) 1.13 ± 0.3 and 1.46 ± 0.2; Cl (mL/min/kg) 1.79 ± 0.2 and 1.34 ± 0.2, respectively. There were seven adverse events (AE) overall. All AEs were resolved without any complications. None were related to the study drug. There were no immunogenicity or any significant infusion-related reactions. The new Agal product exhibited a dose-dependent PK and was well tolerated with no significant AEs in healthy adult volunteers.


Subject(s)
Isoenzymes/pharmacokinetics , alpha-Galactosidase/pharmacokinetics , Adult , Double-Blind Method , Healthy Volunteers , Humans , Immunoglobulin G/blood , Isoenzymes/adverse effects , Isoenzymes/blood , Isoenzymes/immunology , Male , Recombinant Proteins/adverse effects , Recombinant Proteins/blood , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Young Adult , alpha-Galactosidase/adverse effects , alpha-Galactosidase/blood , alpha-Galactosidase/immunology
8.
J Vis Exp ; (80): e50638, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24192611

ABSTRACT

Sub-micrometer carriers (nanocarriers; NCs) enhance efficacy of drugs by improving solubility, stability, circulation time, targeting, and release. Additionally, traversing cellular barriers in the body is crucial for both oral delivery of therapeutic NCs into the circulation and transport from the blood into tissues, where intervention is needed. NC transport across cellular barriers is achieved by: (i) the paracellular route, via transient disruption of the junctions that interlock adjacent cells, or (ii) the transcellular route, where materials are internalized by endocytosis, transported across the cell body, and secreted at the opposite cell surface (transyctosis). Delivery across cellular barriers can be facilitated by coupling therapeutics or their carriers with targeting agents that bind specifically to cell-surface markers involved in transport. Here, we provide methods to measure the extent and mechanism of NC transport across a model cell barrier, which consists of a monolayer of gastrointestinal (GI) epithelial cells grown on a porous membrane located in a transwell insert. Formation of a permeability barrier is confirmed by measuring transepithelial electrical resistance (TEER), transepithelial transport of a control substance, and immunostaining of tight junctions. As an example, ~200 nm polymer NCs are used, which carry a therapeutic cargo and are coated with an antibody that targets a cell-surface determinant. The antibody or therapeutic cargo is labeled with (125)I for radioisotope tracing and labeled NCs are added to the upper chamber over the cell monolayer for varying periods of time. NCs associated to the cells and/or transported to the underlying chamber can be detected. Measurement of free (125)I allows subtraction of the degraded fraction. The paracellular route is assessed by determining potential changes caused by NC transport to the barrier parameters described above. Transcellular transport is determined by addressing the effect of modulating endocytosis and transcytosis pathways.


Subject(s)
Drug Carriers/administration & dosage , Drug Carriers/pharmacokinetics , Drug Delivery Systems/methods , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Intestinal Mucosa/metabolism , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Caco-2 Cells , Cell Membrane Permeability , Electric Impedance , Gastric Mucosa/cytology , Humans , Intestinal Absorption , Intestinal Mucosa/cytology , Iodine Radioisotopes/chemistry , Isotope Labeling , Staining and Labeling/methods , Tight Junctions/metabolism , alpha-Galactosidase/administration & dosage , alpha-Galactosidase/pharmacokinetics
9.
J Inherit Metab Dis ; 35(6): 1107-17, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22450713

ABSTRACT

The success of enzymatic replacement in Gaucher disease has stimulated development of targeted protein replacement for other lysosomal disorders, including Anderson-Fabry disease, which causes fatal cardiac, cerebrovascular and renal injury: deficiency of lysosomal α-Galactosidase A induces accumulation of glycosphingolipids. Endothelial cell storage was the primary endpoint in a clinical trial that led to market authorization. Two α-Galactosidase A preparations are licensed worldwide, but fatal outcomes persist, with storage remaining in many tissues. We compare mechanisms of uptake of α -Galactosidase A into cells relevant to Fabry disease, in order to investigate if the enzyme is targeted to the lysosomes in a mannose-6-phosphate receptor dependent fashion, as generally believed. α -Galactosidase A uptake was examined in fibroblasts, four different endothelial cell models, and hepatic cells in vitro. Uptake of europium-labeled human α -Galactosidase A was measured by time-resolved fluorescence. Ligand-specific uptake was quantified in inhibitor studies. Targeting to the lysosome was determined by precipitation and by confocal microscopy. The quantity and location of cation-independent mannose-6-phosphate receptors in the different cell models were investigated using confocal microscopy. Uptake and delivery of α -Galactosidase A to lysosomes in fibroblasts is mediated by the canonical mannose-6-phosphate receptor pathway, but in endothelial cells in vitro this mechanism does not operate. Moreover, this observation is supported by a striking paucity of expression of cation independent mannose-6-phosphate receptors on the plasma membrane of the four endothelial cell models and by little delivery of enzyme to lysosomes, when compared with fibroblasts. If these observations are confirmed in vivo, alternative mechanisms will be needed to explain the ready clearance of storage from endothelial cells in patients undergoing enzyme replacement therapy.


Subject(s)
Enzyme Replacement Therapy/methods , Fabry Disease/drug therapy , Fabry Disease/enzymology , Lysosomes/enzymology , Cell Line , Cells, Cultured , Drug Delivery Systems/methods , Endothelial Cells/enzymology , Fibroblasts/enzymology , Humans , Isoenzymes/administration & dosage , Isoenzymes/pharmacokinetics , Microscopy, Confocal , Models, Biological , Receptor, IGF Type 2/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , alpha-Galactosidase/administration & dosage , alpha-Galactosidase/pharmacokinetics
10.
Mol Med ; 18: 76-82, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22033676

ABSTRACT

To economically produce recombinant human α-galactosidase A (GLA) with a cell culture system that does not require bovine serum, we chose methylotrophic yeast cells with the OCH1 gene, which encodes α-1,6-mannosyltransferase, deleted and over-expressing the Mnn4p (MNN4) gene, which encodes a positive regulator of mannosylphosphate transferase, as a host cell line. The enzyme (yr-hGLA) produced with the gene-manipulated yeast cells has almost the same enzymological parameters as those of the recombinant human GLA produced with cultured human fibroblasts (agalsidase alfa), which is currently used for enzyme replacement therapy for Fabry disease. However, the basic structures of their sugar chains are quite different. yr-hGLA has a high content of phosphorylated N-glycans and is well incorporated into the kidneys, the main target organ in Fabry disease, where it cleaves the accumulated glycosphingolipids. A glycoprotein production system involving this gene-manipulated yeast cell line will be useful for the development of a new enzyme replacement therapy for Fabry disease.


Subject(s)
Fabry Disease/drug therapy , Kidney/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Yeasts/metabolism , alpha-Galactosidase/metabolism , alpha-Galactosidase/therapeutic use , Animals , Fabry Disease/metabolism , Female , Humans , Male , Mice , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics , Yeasts/genetics , alpha-Galactosidase/genetics , alpha-Galactosidase/pharmacokinetics
11.
J Control Release ; 149(3): 323-31, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21047542

ABSTRACT

Fabry disease, due to the deficiency of α-galactosidase A (α-Gal), causes lysosomal accumulation of globotriaosylceramide (Gb3) in multiple tissues and prominently in the vascular endothelium. Although enzyme replacement therapy (ERT) by injection of recombinant α-Gal improves the disease outcome, the effects on the vasculopathy associated with life-threatening cerebrovascular, cardiac and renal complications are still limited. We designed a strategy to enhance the delivery of α-Gal to organs and endothelial cells (ECs). We targeted α-Gal to intercellular adhesion molecule 1 (ICAM-1), a protein expressed on ECs throughout the vasculature, by loading this enzyme on nanocarriers coated with anti-ICAM (anti-ICAM/α-Gal NCs). In vitro radioisotope tracing showed efficient loading of α-Gal on anti-ICAM NCs, stability of this formulation under storage and in model physiological fluids, and enzyme release in response to lysosome environmental conditions. In mice, the delivery of (125)I-α-Gal was markedly enhanced by anti-ICAM/(125)I-α-Gal NCs in brain, kidney, heart, liver, lung, and spleen, and transmission electron microscopy showed anti-ICAM/α-Gal NCs attached to and internalized into the vascular endothelium. Fluorescence microscopy proved targeting, endocytosis and lysosomal transport of anti-ICAM/α-Gal NCs in macro- and micro-vascular ECs and a marked enhancement of Gb3 degradation. Therefore, this ICAM-1-targeting strategy may help improve the efficacy of therapeutic enzymes for Fabry disease.


Subject(s)
Drug Carriers/chemistry , Endothelium, Vascular/cytology , Fabry Disease/drug therapy , Intercellular Adhesion Molecule-1/immunology , alpha-Galactosidase/administration & dosage , alpha-Galactosidase/therapeutic use , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antigens, Tumor-Associated, Carbohydrate/metabolism , Cell Line , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enzyme Replacement Therapy , Fabry Disease/enzymology , Fabry Disease/pathology , Humans , Lysosomes/metabolism , Lysosomes/pathology , Lysosomes/ultrastructure , Mice , Mice, Inbred C57BL , alpha-Galactosidase/pharmacokinetics
12.
J Pharmacokinet Pharmacodyn ; 36(5): 443-59, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19789963

ABSTRACT

Patients that are exposed to biotechnology-derived therapeutics often develop antibodies to the therapeutic, the magnitude of which is assessed by measuring antibody titers. A statistical approach for analyzing antibody titer data conditional on seroconversion is presented. The proposed method is to first transform the antibody titer data based on a geometric series using a common ratio of 2 and a scale factor of 50 and then analyze the exponent using a zero-inflated or hurdle model assuming a Poisson or negative binomial distribution with random effects to account for patient heterogeneity. Patient specific covariates can be used to model the probability of developing an antibody response, i.e., seroconversion, as well as the magnitude of the antibody titer itself. The method was illustrated using antibody titer data from 87 male seroconverted Fabry patients receiving Fabrazyme. Titers from five clinical trials were collected over 276 weeks of therapy with anti-Fabrazyme IgG titers ranging from 100 to 409,600 after exclusion of seronegative patients. The best model to explain seroconversion was a zero-inflated Poisson (ZIP) model where cumulative dose (under a constant dose regimen of dosing every 2 weeks) influenced the probability of seroconversion. There was an 80% chance of seroconversion when the cumulative dose reached 210 mg (90% confidence interval: 194-226 mg). No difference in antibody titers was noted between Japanese or Western patients. Once seroconverted, antibody titers did not remain constant but decreased in an exponential manner from an initial magnitude to a new lower steady-state value. The expected titer after the new steady-state titer had been achieved was 870 (90% CI: 630-1109). The half-life to the new steady-state value after seroconversion was 44 weeks (90% CI: 17-70 weeks). Time to seroconversion did not appear to be correlated with titer at the time of seroconversion. The method can be adequately used to model antibody titer data.


Subject(s)
Antibodies/blood , Isoenzymes/immunology , Isoenzymes/pharmacokinetics , alpha-Galactosidase/immunology , alpha-Galactosidase/pharmacokinetics , Adolescent , Algorithms , Antibody Formation/physiology , Asian People , Child , Data Interpretation, Statistical , Enzyme-Linked Immunosorbent Assay , Fabry Disease/drug therapy , Fabry Disease/metabolism , Half-Life , Humans , Isoenzymes/therapeutic use , Male , Models, Statistical , Multicenter Studies as Topic , Poisson Distribution , Proteins/immunology , Randomized Controlled Trials as Topic , White People , Young Adult , alpha-Galactosidase/therapeutic use
13.
Arch. Soc. Esp. Oftalmol ; 83(12): 713-718, dic. 2008. ilus
Article in Es | IBECS | ID: ibc-70394

ABSTRACT

Caso clínico: La enfermedad de Fabry es una enfermedad producida por una alteración en el catabolismo de los glucoesfingolípidos. Se muestran las alteraciones oftalmológicas de cuatro pacientes detectados tras evaluar desde el punto de vista analítico, cardiológico y genético a 113 enfermos. Discusión: La enfermedad de Fabry es una enfermedad infrecuente con afectación oftalmológica inconstante existiendo enfermos con Fabry sin afectación ocular y portadores sanos, con importantes alteraciones oculares. El depósito de glucoesfingolípidos produce afectación a nivel corneal, cristaliniano, vascular y retiniano. Las alteraciones vasculares afectan no sólo a las venas sino también a las arterias como mostramos en nuestros pacientes


Case report: Fabrys disease is an illness produced by an alteration in the catabolism of the glycosphingolipids. We report ophthalmologic findings in 4 people, detected after 113 patient evaluations from an analytical, cardiological and genetic point of view. Discussion: Fabry’s disease is uncommon and shows variable ophthalmologic affectation. Some patients with Fabry’s disease do not present ocular affectation, while, on the other hand, healthy carriers with important ocular alterations have been described. The deposit of glycosphingolipids produces affectation at the corneal, crystalline, vascular and retinal levels. The vascular alterations affect not only the veins but also the arteries, as we report in our patients (Arch Soc Esp Oftalmol 2008; 83: 713-718)


Subject(s)
Humans , Male , Middle Aged , Fabry Disease/complications , Fabry Disease/diagnosis , Graves Disease/complications , Graves Disease/diagnosis , Angiography/methods , alpha-Galactosidase , alpha-Galactosidase , alpha-Galactosidase/pharmacokinetics , alpha-Galactosidase/metabolism , Sclerosis/complications , Sclerosis/epidemiology , Cornea/pathology , Cornea
14.
J Hum Genet ; 52(12): 1018-1025, 2007.
Article in English | MEDLINE | ID: mdl-17965825

ABSTRACT

Peripheral neuropathy is one of the important manifestations of Fabry disease. Enzyme replacement therapy with presently available recombinant alpha-galactosidases does not always improve the Fabry neuropathy. But the reason has not been determined yet. We established a Schwann cell line from Fabry mice, characterized it, and then examined the uptake of alpha-galactosidase by cells and its effect on the degradation of accumulated substrate. The cells exhibited a distinct Schwann cell morphology and biochemical phenotype (alpha-Galactosidase activity was deficient, and numerous cytoplasmic inclusion bodies were present in the cells). A recombinant alpha-galactosidase added to the culture medium was incorporated into the cultured Fabry Schwann cells dose dependently. But the increase in cell-associated enzyme activity was less than that in the cases of human and mouse Fabry fibroblasts. The administration of a high dose of the enzyme improved the pathological changes in cells, although a low dose of it did not. Cellular uptake of the enzyme was strongly inhibited in the presence of mannose 6-phosphate. This suggests that the enzyme is incorporated via cation-independent mannose 6-phosphate receptors in Schwann cells. The low expression of cation-independent mannose 6-phosphate receptors in Schwann cells must be one of the reasons their uptake of the present enzymes was low. The administration of a high dose of the enzyme or the development of an enzyme containing many mannose 6-phosphate residues is required to improve Fabry neuropathy.


Subject(s)
Fabry Disease/drug therapy , Schwann Cells/metabolism , alpha-Galactosidase/pharmacokinetics , Animals , Biological Transport , Dose-Response Relationship, Drug , Fabry Disease/enzymology , Fabry Disease/metabolism , Humans , Mice , Mice, Inbred C57BL , Receptor, IGF Type 2 , Recombinant Proteins , Schwann Cells/drug effects , alpha-Galactosidase/antagonists & inhibitors
15.
J Clin Pharmacol ; 47(10): 1222-30, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17698592

ABSTRACT

This multicenter, open-label study evaluated pharmacokinetics, pharmacodynamics, and safety of agalsidase alpha in pediatric compared with adult patients with Fabry disease. The pharmacokinetic parameters of pediatric patients (19 boys, 5 girls, 6-18 years old; mean age, 11.8 years) were compared to those of adult male and female patients who participated in other clinical studies. All patients received agalsidase alpha at a dose of 0.2 mg/kg infused over 40 minutes every other week. Agalsidase alpha exhibited a biphasic serum elimination profile with a maximum serum concentration at the end of the 40-minute infusion; <1% of the maximum concentration was detected 8 hours after dosing. In children, serum clearance was 2.0 to 9.4 mL/min/kg and tended to decrease with increasing age. The average clearance in children, 3.7 +/- 1.5 mL/min/kg (mean +/- SD), was significantly greater than that measured in 33 adults (2.3 +/- 0.7 mL/min/kg, P < .0001). Mean terminal elimination half-life of agalsidase alpha was prolonged in week 25 compared with baseline (150 vs 66 minutes) in 8 of 19 male children. The magnitude of the reduction of plasma globotriaosylceremide was similar in all age groups and was independent of area under the curve and other pharmacokinetic parameters. Except for clearance in younger patients, agalsidase alpha appears to have comparable pharmacokinetic and pharmacodynamic profiles in pediatric and adult Fabry patients of both genders.


Subject(s)
Fabry Disease/drug therapy , alpha-Galactosidase/pharmacokinetics , alpha-Galactosidase/therapeutic use , Adolescent , Adult , Age Factors , Aged , Area Under Curve , Child , Drug Administration Schedule , Female , Half-Life , Humans , Immunoglobulin G/blood , Infusions, Intravenous , Isoenzymes/administration & dosage , Isoenzymes/immunology , Isoenzymes/pharmacokinetics , Isoenzymes/therapeutic use , Male , Middle Aged , Sex Factors , Treatment Outcome , Trihexosylceramides/blood , alpha-Galactosidase/administration & dosage , alpha-Galactosidase/immunology
16.
Genet Med ; 9(8): 504-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17700388

ABSTRACT

PURPOSE: This 10-week study was conducted to determine the pharmacokinetics of varying doses of agalsidase alfa and evaluate the effect of dose and dosing frequency on plasma Gb3 levels. METHODS: Eighteen adult male Fabry patients, naive to enzyme replacement therapy, were randomized to one of five regimens: 0.1, 0.2, or 0.4 mg/kg weekly; 0.2 mg/kg every other week (the approved dose); or 0.4 mg/kg every other week. Intravenous infusion rate was 0.1 mg/kg per 20 minutes. Plasma Gb3 levels were assessed at baseline and periodically during the study. RESULTS: The mean half-life was 56-76 minutes, and the mean volume of distribution at steady state was 17%-18% of body weight, with no significant association between dose and half-life, clearance, or volume of distribution at steady state. The area under the curve was linearly proportional to the dose from 0.1 to 0.4 mg/kg. Baseline average plasma Gb3 was 9.12 +/- 2.61 nmol/mL and after 10 weeks of treatment was significantly reduced by about 50% in each group with no statistically significant differences between groups. CONCLUSIONS: Reduction of plasma Gb3 levels was independent of dose or dose frequency in the range tested. These observations, coupled with the clinical trial experience of both agalsidase alfa and agalsidase beta, indicate that the standard dose of agalsidase alfa is sufficient to maximally reduce plasma Gb3. However, because plasma Gb3 is not a validated surrogate of disease severity in Fabry disease, further clinical study will be required to determine the optimal dosing regimen for providing maximal clinical benefit.


Subject(s)
Fabry Disease/therapy , alpha-Galactosidase/pharmacology , Adolescent , Adult , Clinical Protocols , Dose-Response Relationship, Drug , Drug Administration Schedule , Fabry Disease/enzymology , Humans , Infusions, Intravenous , Isoenzymes/administration & dosage , Isoenzymes/adverse effects , Isoenzymes/pharmacokinetics , Isoenzymes/pharmacology , Male , Recombinant Proteins , Trihexosylceramides/blood , alpha-Galactosidase/administration & dosage , alpha-Galactosidase/adverse effects , alpha-Galactosidase/pharmacokinetics
17.
Drugs ; 67(3): 435-55, 2007.
Article in English | MEDLINE | ID: mdl-17335299

ABSTRACT

Agalsidase beta (Fabrazyme) is a recombinant human alpha-galactosidase A enzyme approved for intravenous use in the treatment of Fabry disease. Fabry disease is a progressive, multisystemic, potentially life threatening disorder caused by a deficiency of alpha-galactosidase A. This deficiency results in accumulation of glycosphingolipids, particularly globotriaosylceramide (GL-3), in the lysosomes of various tissues. This accumulation is the underlying driver of disease progression. Agalsidase beta provides an exogenous source of alpha-galactosidase A.Intravenous agalsidase beta is effective and well tolerated in patients with Fabry disease. In a phase III trial, agalsidase beta was shown to clear GL-3 from various target cells and, in a subsequent extension of this trial, prevent GL-3 reaccumulation. In a post-approval trial, agalsidase beta was shown to provide significant clinical benefit by reducing the risk of a major clinical event. Thus, agalsidase beta represents an important advance in the treatment of Fabry disease, and agalsidase beta therapy should be strongly considered in patients with Fabry disease who are suitable candidates.


Subject(s)
Fabry Disease/drug therapy , Isoenzymes/therapeutic use , Recombinant Proteins/therapeutic use , alpha-Galactosidase/therapeutic use , Adult , Asian People , Child , Female , Humans , Isoenzymes/adverse effects , Isoenzymes/pharmacokinetics , Isoenzymes/pharmacology , Japan , Male , Quality of Life , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Trihexosylceramides , alpha-Galactosidase/adverse effects , alpha-Galactosidase/pharmacokinetics , alpha-Galactosidase/pharmacology
18.
Nephrol Dial Transplant ; 22(7): 1920-5, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17395657

ABSTRACT

BACKGROUND: Fabry disease (FD) is caused by an X-linked deficiency in the activity of alpha-galactosidase A and the resultant accumulation of globotriaosylceramide (Gb3) in multiple tissues. Nearly all classically affected males with FD experience kidney dysfunction, with progression to end-stage renal disease (ESRD) in the third decade of life or shortly thereafter. METHODS: Twenty-two FD patients (20 men and 2 women) receiving dialysis or who had a history of kidney transplantation were treated with agalsidase alfa in an open label setting using the same dosing regimen given to patients without ESRD (0.2 mg/kg every other week). Pharmacokinetics (PK) were determined during and following the initial dose, and safety was evaluated during therapy. Change in plasma Gb3 level was used as a surrogate marker of enzyme activity in vivo. RESULTS: A typical biphasic plasma elimination profile was seen in both dialysis and transplant patients, similar to that observed in 18 non-ESRD FD patients. Calculated PK parameters were similar to the three patient groups. In the male patients, plasma Gb3 level declined by 43% after 6 months (P<0.001). Infusion reactions were experienced by 8 of 21 (38%) patients, but did not result in any infusions being stopped prematurely. Anti-agalsidase alfa IgG antibodies were detected in 15.8% of males and 0% female patients. No anti-agalsidase alfa IgE antibodies were detected. CONCLUSIONS: The same dosing regimen of agalsidase alfa may be safely administered to FD patients with ESRD as given to those without ESRD.


Subject(s)
Fabry Disease/complications , Fabry Disease/drug therapy , Kidney Failure, Chronic/etiology , alpha-Galactosidase/therapeutic use , Adult , Aged , Fabry Disease/blood , Fabry Disease/metabolism , Female , Humans , Immunoglobulin G/blood , Isoenzymes/adverse effects , Isoenzymes/immunology , Isoenzymes/pharmacokinetics , Isoenzymes/therapeutic use , Kidney Failure, Chronic/therapy , Kidney Transplantation , Male , Medical Records , Middle Aged , Recombinant Proteins , Renal Dialysis , Time Factors , Trihexosylceramides/blood , alpha-Galactosidase/adverse effects , alpha-Galactosidase/immunology , alpha-Galactosidase/pharmacokinetics
19.
Mol Genet Metab ; 90(3): 307-12, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17188539

ABSTRACT

alpha-Galactosidase A is the lysosomal hydrolase that is deficient in patients with Fabry disease. Intravenous infusion of agalsidase alfa, a preparation of alpha-Galactosidase A, is used for enzyme replacement therapy (ERT) in patients with Fabry disease. Although ERT appears to show some beneficial effects, most patients show only a modest response. We investigated using immunohistochemistry the relative tissue and cellular distribution of agalsidase alfa after a single intravenous injection in a mouse knockout model of Fabry disease. Specific immunostaining for agalsidase alfa was found only in liver, kidney, heart, testes, adrenal gland, spleen and bone marrow. There was no difference in distribution of the infused enzyme distribution among tissues sampled 4, 24, and 48h post-injection. The intracellular localization of immunopositivity varied considerably between organs with vascular endothelium being the most commonly positive site. alpha-Galactosidase A specific activity in tissue homogenates matched the relative extent of agalsidase alfa immunostaining distribution in the same organs. We conclude that intravenously injected agalsidase alfa has a very heterogeneous systemic distribution using an immunostaining technique.


Subject(s)
alpha-Galactosidase/administration & dosage , alpha-Galactosidase/pharmacokinetics , Animals , Disease Models, Animal , Fabry Disease/drug therapy , Fabry Disease/enzymology , Fabry Disease/genetics , Humans , Immunohistochemistry , Injections, Intravenous , Isoenzymes/administration & dosage , Isoenzymes/pharmacokinetics , Male , Mice , Mice, Knockout , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Tissue Distribution , alpha-Galactosidase/genetics
20.
J Hum Genet ; 51(4): 341-352, 2006.
Article in English | MEDLINE | ID: mdl-16532254

ABSTRACT

We have previously reported the production of a recombinant alpha-galactosidase with engineered N-linked sugar chains facilitating uptake and transport to lysosomes in a Saccharomyces cerevisiae mutant. In this study, we improved the purification procedure, allowing us to obtain a large amount of highly purified enzyme protein with mannose-6-phosphate residues at the non-reducing ends of sugar chains. The products were incorporated into cultured fibroblasts derived from a patient with Fabry disease via mannose-6-phosphate receptors. The ceramide trihexoside (CTH) accumulated in lysosomes was cleaved dose-dependently, and the disappearance of deposited CTH was maintained for at least 7 days after administration. We next examined the effect of the recombinant alpha-galactosidase on Fabry mice. Repeated intravascular administration of the enzyme led to successful degradation of CTH accumulated in the liver, kidneys, heart, and spleen. However, cleavage of the accumulated CTH in the dorsal root ganglia was insufficient. As the culture of yeast cells is easy and economical, and does not require fetal calf serum, the recombinant alpha-galactosidase produced in yeast cells is highly promising as an enzyme source for enzyme replacement therapy in Fabry disease.


Subject(s)
Drug Delivery Systems , Fabry Disease/therapy , Lysosomes/enzymology , Saccharomyces cerevisiae/enzymology , alpha-Galactosidase/administration & dosage , alpha-Galactosidase/pharmacology , Animals , Carbohydrate Sequence , Cells, Cultured , Chromatography, High Pressure Liquid , Culture Media/chemistry , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Fibroblasts/drug effects , Glycosphingolipids/metabolism , Humans , Mice , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Distribution , alpha-Galactosidase/biosynthesis , alpha-Galactosidase/chemistry , alpha-Galactosidase/genetics , alpha-Galactosidase/isolation & purification , alpha-Galactosidase/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...