Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Plant Biotechnol J ; 13(8): 1033-40, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25828481

ABSTRACT

Gaucher's disease (GD), a lysosomal storage disorder caused by mutations in the gene encoding glucocerebrosidase (GCD), is currently treated by enzyme replacement therapy (ERT) using recombinant GCD that is administered intravenously every 2 weeks. However, intravenous administration includes discomfort or pain and might cause local and systemic infections that may lead to low patient compliance. An orally administered drug has the potential to alleviate these problems. In this study, we describe the potential use of plant cells as a vehicle for the oral delivery of recombinant human GCD (prGCD) expressed in carrot cells. The in vitro results demonstrate that the plant cells protect the recombinant protein in the gastric fluids and may enable absorption into the blood. Feeding experiments, with rat and pig as model animals, using carrot cells containing prGCD, show that active recombinant prGCD was found in the digestive tract and blood system and reached both, liver and spleen, the target organs in GD. These results demonstrate that the oral administration of proteins encapsulated in plant cells is feasible. Specifically, carrot cells containing recombinant human prGCD can be used as an oral delivery system and are a feasible alternative to intravenous administration of ERT for GD.


Subject(s)
Enzyme Replacement Therapy , Gaucher Disease/drug therapy , Glucosylceramidase/administration & dosage , Glucosylceramidase/therapeutic use , Nicotiana/metabolism , Administration, Oral , Animals , Body Fluids/metabolism , Caco-2 Cells , Enzyme Stability , Glucosylceramidase/pharmacokinetics , Humans , Intestinal Mucosa/metabolism , Organ Specificity , Plant Cells/metabolism , Rats , Recombinant Proteins/administration & dosage , Sus scrofa , Tissue Distribution , Transcytosis
2.
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
3.
Biosci Rep ; 33(5)2013 Sep 25.
Article in English | MEDLINE | ID: mdl-23980545

ABSTRACT

The glycosylation of recombinant ß-glucocerebrosidase, and in particular the exposure of mannose residues, has been shown to be a key factor in the success of ERT (enzyme replacement therapy) for the treatment of GD (Gaucher disease). Macrophages, the target cells in GD, internalize ß-glucocerebrosidase through MRs (mannose receptors). Three enzymes are commercially available for the treatment of GD by ERT. Taliglucerase alfa, imiglucerase and velaglucerase alfa are each produced in different cell systems and undergo various post-translational or post-production glycosylation modifications to expose their mannose residues. This is the first study in which the glycosylation profiles of the three enzymes are compared, using the same methodology and the effect on functionality and cellular uptake is evaluated. While the major differences in glycosylation profiles reside in the variation of terminal residues and mannose chain length, the enzymatic activity and stability are not affected by these differences. Furthermore, the cellular uptake and in-cell stability in rat and human macrophages are similar. Finally, in vivo studies to evaluate the uptake into target organs also show similar results for all three enzymes. These results indicate that the variations of glycosylation between the three regulatory-approved ß-glucocerebrosidase enzymes have no effect on their function or distribution.


Subject(s)
Glucosylceramidase/metabolism , Protein Processing, Post-Translational , Animals , Biological Transport , Carbohydrate Conformation , Carbohydrate Sequence , Cell Line , Enzyme Stability , Glucosylceramidase/chemistry , Glucosylceramidase/pharmacokinetics , Glycosylation , Humans , Kinetics , Macrophages, Alveolar/enzymology , Male , Mice , Mice, Inbred ICR , Molecular Sequence Data , Rats , Recombinant Proteins/metabolism , Tissue Distribution
4.
Blood ; 118(22): 5767-73, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-21900191

ABSTRACT

Taliglucerase alfa (Protalix Biotherapeutics, Carmiel, Israel) is a novel plant cell-derived recombinant human ß-glucocerebrosidase for Gaucher disease. A phase 3, double-blind, randomized, parallel-group, comparison-dose (30 vs 60 U/kg body weight/infusion) multinational clinical trial was undertaken. Institutional review board approvals were received. A 9-month, 20-infusion trial used inclusion/exclusion criteria in treatment-naive adult patients with splenomegaly and thrombocytopenia. Safety end points were drug-related adverse events: Ab formation and hypersensitivity reactions. Primary efficacy end point was reduction in splenic volume measured by magnetic resonance imaging. Secondary end points were: changes in hemoglobin, hepatic volume, and platelet counts. Exploratory parameters included biomarkers and bone imaging. Twenty-nine patients (11 centers) completed the protocol. There were no serious adverse events; drug-related adverse events were mild/moderate and transient. Two patients (6%) developed non-neutralizing IgG Abs; 2 other patients (6%) developed hypersensitivity reactions. Statistically significant spleen reduction was achieved at 9 months: 26.9% (95% confidence interval [CI]: -31.9, -21.8) in the 30-unit dose group and 38.0% (95% CI: -43.4, -32.8) in the 60-unit dose group (both P < .0001); and in all secondary efficacy end point measures, except platelet counts at the lower dose. These results support safety and efficacy of taliglucerase alfa for Gaucher disease.


Subject(s)
Enzyme Replacement Therapy , Gaucher Disease/drug therapy , Glucosylceramidase/therapeutic use , Plant Cells/metabolism , Adult , Aged , Algorithms , Double-Blind Method , Enzyme Replacement Therapy/methods , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Plant , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Humans , Male , Middle Aged , Placebos , Plant Cells/enzymology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Treatment Outcome , Young Adult
5.
Org Biomol Chem ; 9(11): 4160-7, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21483943

ABSTRACT

Cyclodextrin-based host-guest chemistry has been exploited to facilitate co-crystallization of recombinant human acid ß-glucosidase (ß-glucocerebrosidase, GlcCerase) with amphiphilic bicyclic nojirimycin analogues of the sp(2)-iminosugar type. Attempts to co-crystallize GlcCerase with 5-N,6-O-[N'-(n-octyl)iminomethylidene]nojirimycin (NOI-NJ) or with 5-N,6-S-[N'-(n-octyl)iminomethylidene]-6-thionojirimycin (6S-NOI-NJ), two potent inhibitors of the enzyme with promising pharmacological chaperone activity for several Gaucher disease-associated mutations, were unsuccessful probably due to the formation of aggregates that increase the heterogeneity of the sample and affect nucleation and growth of crystals. Cyclomaltoheptaose (ß-cyclodextrin, ßCD) efficiently captures NOI-NJ and 6S-NOI-NJ in aqueous media to form inclusion complexes in which the lipophilic tail is accommodated in the hydrophobic cavity of the cyclooligosaccharide. The dissociation constant of the complex of the amphiphilic sp(2)-iminosugars with ßCD is two orders of magnitude higher than that of the corresponding complex with GlcCerase, allowing the efficient transfer of the inhibitor from the ßCD cavity to the GlcCerase active site. Enzyme-inhibitor complexes suitable for X-ray analysis were thus grown in the presence of ßCD. In contrast to what was previously observed for the complex of GlcCerase with the more basic derivative, 6-amino-6-deoxy-5-N,6-N-[N'-(n-octyl)iminomethylidene]nojirimycin (6N-NOI-NJ), the ß-anomers of both NOI-NJ and 6S-NOI-NJ were seen in the active site, even though the α-anomer was exclusively detected both in aqueous solution and in the corresponding ßCD:sp(2)-iminosugar complexes. Our results further suggest that cyclodextrin derivatives might serve as suitable delivery systems of amphiphilic glycosidase inhibitors in a biomedical context.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cyclodextrins/chemistry , Glucosylceramidase/chemistry , 1-Deoxynojirimycin/chemistry , Crystallography, X-Ray , Glucosylceramidase/metabolism , Hydrogen Bonding , Models, Molecular , Molecular Structure , Stereoisomerism
7.
PLoS One ; 4(3): e4792, 2009.
Article in English | MEDLINE | ID: mdl-19277123

ABSTRACT

UNLABELLED: Gaucher disease is a progressive lysosomal storage disorder caused by the deficiency of glucocerebrosidase leading to the dysfunction in multiple organ systems. Intravenous enzyme replacement is the accepted standard of treatment. In the current report, we evaluate the safety and pharmacokinetics of a novel human recombinant glucocerebrosidase enzyme expressed in transformed plant cells (prGCD), administered to primates and human subjects. Short term (28 days) and long term (9 months) repeated injections with a standard dose of 60 Units/kg and a high dose of 300 Units/kg were administered to monkeys (n = 4/sex/dose). Neither clinical drug-related adverse effects nor neutralizing antibodies were detected in the animals. In a phase I clinical trial, six healthy volunteers were treated by intravenous infusions with escalating single doses of prGCD. Doses of up to 60 Units/kg were administered at weekly intervals. prGCD infusions were very well tolerated. Anti-prGCD antibodies were not detected. The pharmacokinetic profile of the prGCD revealed a prolonged half-life compared to imiglucerase, the commercial enzyme that is manufactured in a costly mammalian cell system. These studies demonstrate the safety and lack of immunogenicity of prGCD. Following these encouraging results, a pivotal phase III clinical trial for prGCD was FDA approved and is currently ongoing. TRIAL REGISTRATION: ClinicalTrials.gov NCT00258778.


Subject(s)
Gaucher Disease/drug therapy , Glucosylceramidase/therapeutic use , Adult , Animals , Antibody Formation , Cells, Cultured/enzymology , Clinical Trials, Phase III as Topic , Daucus carota/cytology , Drug Evaluation, Preclinical , Female , Gaucher Disease/enzymology , Gaucher Disease/genetics , Glucosylceramidase/adverse effects , Glucosylceramidase/economics , Glucosylceramidase/genetics , Glucosylceramidase/immunology , Glucosylceramidase/isolation & purification , Glucosylceramidase/pharmacokinetics , Half-Life , Humans , Infusions, Intravenous , Macaca fascicularis , Male , Neutralization Tests , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/economics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Transfection , Young Adult
8.
J Biol Chem ; 282(39): 29052-29058, 2007 Sep 28.
Article in English | MEDLINE | ID: mdl-17666401

ABSTRACT

Gaucher disease is caused by mutations in the gene encoding acid beta-glucosidase (GlcCerase), resulting in glucosylceramide (GlcCer) accumulation. The only currently available orally administered treatment for Gaucher disease is N-butyl-deoxynojirimycin (Zavesca, NB-DNJ), which partially inhibits GlcCer synthesis, thus reducing levels of GlcCer accumulation. NB-DNJ also acts as a chemical chaperone for GlcCerase, although at a different concentration than that required to completely inhibit GlcCer synthesis. We now report the crystal structures, at 2A resolution, of complexes of NB-DNJ and N-nonyl-deoxynojirimycin (NN-DNJ) with recombinant human GlcCerase, expressed in cultured plant cells. Both inhibitors bind at the active site of GlcCerase, with the imino sugar moiety making hydrogen bonds to side chains of active site residues. The alkyl chains of NB-DNJ and NN-DNJ are oriented toward the entrance of the active site where they undergo hydrophobic interactions. Based on these structures, we make a number of predictions concerning (i) involvement of loops adjacent to the active site in the catalytic process, (ii) the nature of nucleophilic attack by Glu-340, and (iii) the role of a conserved water molecule located in a solvent cavity adjacent to the active site. Together, these results have significance for understanding the mechanism of action of GlcCerase and the mode of GlcCerase chaperoning by imino sugars.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Enzyme Inhibitors/chemistry , Gaucher Disease/enzymology , Glucosylceramidase/chemistry , Molecular Chaperones/chemistry , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/chemistry , Administration, Oral , Binding Sites/physiology , Crystallography, X-Ray , Enzyme Inhibitors/administration & dosage , Gaucher Disease/drug therapy , Glucosylceramides/biosynthesis , Glucosylceramides/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Recombinant Proteins/chemistry , Structure-Activity Relationship
9.
Plant Biotechnol J ; 5(5): 579-90, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17524049

ABSTRACT

Gaucher's disease, a lysosomal storage disorder caused by mutations in the gene encoding glucocerebrosidase (GCD), is currently treated by enzyme replacement therapy using recombinant GCD (Cerezyme) expressed in Chinese hamster ovary (CHO) cells. As complex glycans in mammalian cells do not terminate in mannose residues, which are essential for the biological uptake of GCD via macrophage mannose receptors in human patients with Gaucher's disease, an in vitro glycan modification is required in order to expose the mannose residues on the glycans of Cerezyme. In this report, the production of a recombinant human GCD in a carrot cell suspension culture is described. The recombinant plant-derived GCD (prGCD) is targeted to the storage vacuoles, using a plant-specific C-terminal sorting signal. Notably, the recombinant human GCD expressed in the carrot cells naturally contains terminal mannose residues on its complex glycans, apparently as a result of the activity of a special vacuolar enzyme that modifies complex glycans. Hence, the plant-produced recombinant human GCD does not require exposure of mannose residues in vitro, which is a requirement for the production of Cerezyme. prGCD also displays a level of biological activity similar to that of Cerezyme produced in CHO cells, as well as a highly homologous high-resolution three-dimensional structure, determined by X-ray crystallography. A single-dose toxicity study with prGCD in mice demonstrated the absence of treatment-related adverse reactions or clinical findings, indicating the potential safety of prGCD. prGCD is currently undergoing clinical studies, and may offer a new and alternative therapeutic option for Gaucher's disease.


Subject(s)
Gaucher Disease/drug therapy , Glucosylceramidase/metabolism , Glucosylceramidase/therapeutic use , Polysaccharides/metabolism , Animals , Blotting, Western , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Crystallography, X-Ray , Daucus carota/cytology , Daucus carota/enzymology , Daucus carota/metabolism , Drug Evaluation, Preclinical/methods , Female , Gaucher Disease/enzymology , Gaucher Disease/genetics , Glucosylceramidase/genetics , Humans , Macrophages/metabolism , Male , Mannose/chemistry , Mannose/metabolism , Mice , Mice, Inbred ICR , Polysaccharides/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use
10.
Curr Drug Targets ; 4(5): 353-65, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12816345

ABSTRACT

Achondroplasia, the most common form of human dwarfism is a sporadic autosomal dominant condition that occurs in approximately 1:20,000 births. The major clinical outcome of Achondroplasia is attenuated growth, rhizomelic shortening of the long bones and craniofacial abnormalities. As of today there is no pharmacological treatment for Achondroplasia. Some improvement in the patients well being and daily function can be achieved by a surgical limb lengthening procedure. Growth hormone treatment seems to have only modest short term success and to lack long term benefits. Achondroplasia results from a single point mutation in Fibroblast Growth Factor Receptor 3 (FGFR3). In 97% of the patients, there is a Glycine to Arginine substitution at position 380 within the FGFR-3 transmembrane domain leading to receptor overactivation. This FGF receptor tyrosine kinase is expressed by chondrocytes in the growth plate of developing long bones and plays a crucial role in bone growth. Genetic disruption of the FGFR-3 gene in mice leads to a remarkable increase in the length of the vertebral column and long bones. This suggests that overaction of FGFR3 signaling may specifically impair chondrocyte function within the epiphyseal growth plates and cause Achondroplasia. Reconstituted normal bone growth may therefore be achieved by attenuation of FGFR3 signaling in the appropriate cells within the growth plate. It is highly conceivable that drug development strategies aimed either towards blocking extracellular ligand binding or towards intracellular checkpoints along the FGF signal transduction cascade, may prove successful in the treatment of Achondroplasia. This review focuses on the possible approaches for developing a drug for Achondroplasia and related skeletal disorders, using chemical, biochemical and molecular strategies.


Subject(s)
Achondroplasia/genetics , Receptors, Fibroblast Growth Factor/genetics , Achondroplasia/epidemiology , Achondroplasia/therapy , Animals , Antibodies/therapeutic use , Disease Models, Animal , Genetic Therapy , Humans , Mice , Mice, Knockout , Point Mutation , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/drug effects , Receptors, Fibroblast Growth Factor/immunology , Signal Transduction/drug effects
11.
Cardiovasc Res ; 53(1): 232-41, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11744033

ABSTRACT

OBJECTIVE: One of the key events in post-angioplasty restenosis is the migration and proliferation of medial smooth muscle cells leading to neo-intima formation. This phase is mediated by several growth factors, mainly platelet-derived growth factor (PDGF), basic fibroblast growth factor (FGF2/bFGF) and heparin-binding epidermal growth factor (HB-EGF). In this study, we have focused on the role of FGF2, which requires heparan sulfate proteoglycans (HSPG) as cofactors for binding and activation of its cell surface tyrosine kinase receptor. The aim of this study was to identify and explore the effect of novel FGF antagonists on vascular smooth muscle cell (VSMC) proliferation. METHODS: We have recently identified a novel class of small, positively charged molecules sharing a porphyrin core as inhibitors of FGF2 and vascular endothelial growth factor (VEGF) activity. Here we investigated the inhibitory effect of these compounds on VSMC proliferation and their effect on heparin-induced FGF receptor activity. RESULTS: We found that these molecules exert a marked inhibitory effect on FGF2-mediated smooth muscle cell (SMC) proliferation, manifested by reduced cell growth and DNA synthesis, which occurred in a dose-dependent manner with an IC(50) of approximately 1 microM of inhibitor. We demonstrate that the molecule, 5, 10, 15, 20-tetrakis (methyl-4-pyridyl)-21H, 23H-porphine tetra-p-tosylate salt (TMPP), inhibits binding of radiolabeled FGF2 to SMCs and to soluble FGF receptor 1 (FGFR1) in a manner that interferes with both ligand and receptor interactions with heparin, thereby blocking growth factor mediated SMC proliferation. CONCLUSION: We have identified an FGF antagonist, which may serve in clinical practice as a preventive measure of restenosis.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Muscle, Smooth, Vascular/cytology , Porphyrins/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Aorta , Binding, Competitive , Cattle , Cell Division/drug effects , Cells, Cultured , DNA/biosynthesis , Depression, Chemical , Fibroblast Growth Factor 2/metabolism , Heparin/metabolism , Heparin/pharmacology , Muscle, Smooth, Vascular/drug effects , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...