Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Eur J Neurol ; 30(9): 2919-2945, 2023 09.
Article En | MEDLINE | ID: mdl-37209042

BACKGROUND: Since the results of previous studies regarding the safety and efficacy of miglustat in GM2 gangliosidosis (GM2g) were inconsistent, we aimed to assess miglustat therapy in GM2g patients. METHODS: This study followed the latest version of PRISMA. We included the observational or interventional studies reporting GM2g patients under miglustat therapy by searching PubMed, Web of Science, and Scopus. Data extracted included the natural history of individual patient data, as well as the safety and efficacy of miglustat in GM2g patients. The quality assessment was performed using the Joanna Briggs Institute Critical Appraisal checklist. RESULTS: A total of 1023 records were identified and reduced to 621 after removing duplicates. After screening and applying the eligibility criteria, 10 articles and 2 abstracts met the inclusion criteria. Overall, the studies represented 54 patients with GM2g under treatment with miglustat and 22 patients with GM2g in the control group. Among patients with available data, 14 and 54 have been diagnosed with Sandhoff disease and Tay-Sachs disease, respectively. Patients included in this review consisted of 23 infantile, 4 late-infantile, 18 juvenile, and 31 adult-onset GM2g. CONCLUSIONS: Although miglustat should not be considered a definite treatment for GM2g, it appears that patients, particularly those with infantile or late-infantile GM2g, could benefit from miglustat therapy to some extent. We also make some suggestions regarding future studies presenting their findings in a standard format to facilitate pooling the available data in such rare diseases for a more comprehensive conclusion.


Gangliosidoses, GM2 , Adult , Humans , Gangliosidoses, GM2/drug therapy , 1-Deoxynojirimycin/adverse effects
2.
Metab Brain Dis ; 34(2): 495-503, 2019 04.
Article En | MEDLINE | ID: mdl-30712135

Gangliosidoses (GM1 and GM2 gangliosidosis) are rare, autosomal recessive progressive neurodegenerative lysosomal storage disorders caused by defects in the degradation of glycosphingolipids. We aimed to investigate clinical, biochemical and molecular genetic spectrum of Turkish patients with infantile gangliosidoses and examined the potential role of serum aspartate transaminase levels as a biomarker. We confirmed the diagnosis of GM1 and GM2 gangliosidosis based on clinical findings with specific enzyme and/or molecular analyses. We retrospectively reviewed serum aspartate transaminase levels of patients with other biochemical parameters. Serum aspartate transaminase level was elevated in all GM1 and GM2 gangliosidosis patients in whom the test was performed, along with normal alanine transaminase. Aspartate transaminase can be a biochemical diagnostic clue for infantile gangliosidoses. It might be a simple but important biomarker for diagnosis, follow up, prognosis and monitoring of the response for the future therapies in these patients.


Aspartate Aminotransferases/metabolism , Biomarkers/analysis , Gangliosidoses/drug therapy , Sandhoff Disease/drug therapy , Aspartate Aminotransferases/drug effects , Female , Gangliosidoses, GM2/drug therapy , Gangliosidosis, GM1/drug therapy , Humans , Male , Retrospective Studies
3.
J Clin Invest ; 126(5): 1691-703, 2016 05 02.
Article En | MEDLINE | ID: mdl-27018595

GM2 gangliosidoses, including Tay-Sachs and Sandhoff diseases, are neurodegenerative lysosomal storage diseases that are caused by deficiency of ß-hexosaminidase A, which comprises an αß heterodimer. There are no effective treatments for these diseases; however, various strategies aimed at restoring ß-hexosaminidase A have been explored. Here, we produced a modified human hexosaminidase subunit ß (HexB), which we have termed mod2B, composed of homodimeric ß subunits that contain amino acid sequences from the α subunit that confer GM2 ganglioside-degrading activity and protease resistance. We also developed fluorescent probes that allow visualization of endocytosis of mod2B via mannose 6-phosphate receptors and delivery of mod2B to lysosomes in GM2 gangliosidosis models. In addition, we applied imaging mass spectrometry to monitor efficacy of this approach in Sandhoff disease model mice. Following i.c.v. administration, mod2B was widely distributed and reduced accumulation of GM2, asialo-GM2, and bis(monoacylglycero)phosphate in brain regions including the hypothalamus, hippocampus, and cerebellum. Moreover, mod2B administration markedly improved motor dysfunction and a prolonged lifespan in Sandhoff disease mice. Together, the results of our study indicate that mod2B has potential for intracerebrospinal fluid enzyme replacement therapy and should be further explored as a gene therapy for GM2 gangliosidoses.


Amino Acid Substitution , Brain/metabolism , Gangliosidoses, GM2/drug therapy , beta-Hexosaminidase beta Chain , Animals , Brain/diagnostic imaging , Disease Models, Animal , Gangliosidoses, GM2/diagnostic imaging , Gangliosidoses, GM2/metabolism , Humans , Mice , Mice, Knockout , Mutation, Missense , Peptide Hydrolases/chemistry , Proteolysis , beta-Hexosaminidase beta Chain/genetics , beta-Hexosaminidase beta Chain/pharmacology
4.
Ann Neurol ; 69(4): 691-701, 2011 Apr.
Article En | MEDLINE | ID: mdl-21520232

OBJECTIVE: Novel recombinant human lysosomal ß-hexosaminidase A (HexA) was developed for enzyme replacement therapy (ERT) for Tay-Sachs and Sandhoff diseases, ie, autosomal recessive GM2 gangliosidoses, caused by HexA deficiency. METHODS: A recombinant human HexA (Om4HexA) with a high mannose 6-phosphate (M6P)-type-N-glycan content, which was produced by a methylotrophic yeast strain, Ogataea minuta, overexpressing the OmMNN4 gene, was intracerebroventricularly (ICV) administered to Sandhoff disease model mice (Hexb⁻/⁻ mice) at different doses (0.5-2.5 mg/kg), and then the replacement and therapeutic effects were examined. RESULTS: The Om4HexA was widely distributed across the ependymal cell layer, dose-dependently restored the enzyme activity due to uptake via cell surface cation-independent M6P receptor (CI-M6PR) on neural cells, and reduced substrates, including GM2 ganglioside (GM2), asialo GM2 (GA2), and oligosaccharides with terminal N-acetylglucosamine residues (GlcNAc-oligosaccharides), accumulated in brain parenchyma. A significant inhibition of chemokine macrophage inflammatory protein-1 α (MIP-1α) induction was also revealed, especially in the hindbrain (< 63%). The decrease in central neural storage correlated with an improvement of motor dysfunction as well as prolongation of the lifespan. INTERPRETATION: This lysosome-directed recombinant human enzyme drug derived from methylotrophic yeast has the high therapeutic potential to improve the motor dysfunction and quality of life of the lysosomal storage diseases (LSDs) patients with neurological manifestations. We emphasize the importance of neural cell surface M6P receptor as a delivery target of neural cell-directed enzyme replacement therapy (NCDERT) for neurodegenerative metabolic diseases.


Enzyme Replacement Therapy , Gangliosidoses, GM2/drug therapy , Gangliosidoses, GM2/enzymology , Hexosaminidase A/administration & dosage , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Replacement Therapy/methods , Gangliosidoses, GM2/genetics , Gangliosidoses, GM2/pathology , Hexosaminidase A/genetics , Hexosaminidase B/genetics , Humans , Injections, Intraventricular , Lysosomes/enzymology , Mannose-6-Phosphate Isomerase/administration & dosage , Mice , Mice, Knockout , Receptors, CCR1/antagonists & inhibitors , Recombinant Proteins , Sandhoff Disease/drug therapy , Sandhoff Disease/enzymology , Tay-Sachs Disease/drug therapy , Tay-Sachs Disease/genetics , Treatment Outcome , Yeasts
5.
Mol Genet Metab ; 102(1): 6-12, 2011 Jan.
Article En | MEDLINE | ID: mdl-20926324

Late-onset GM2 gangliosidosis is an autosomal recessive, neurodegenerative, lysosomal storage disease, caused by deficiency of ß-hexosaminidase A (Hex A), resulting from mutations in the HEXA (Tay-Sachs variant) or the HEXB (Sandhoff variant) genes. The enzyme deficiency in many patients with juvenile or adult onset forms of the disease results from the production of an unstable protein, which becomes targeted for premature degradation by the quality control system of the smooth endoplasmic reticulum and is not transported to lysosomes. In vitro studies have shown that many mutations in either the α or ß subunit of Hex A can be partially rescued, i.e. enhanced levels of both enzyme protein and activity in lysosomes, following the growth of patient cells in the presence of the drug, pyrimethamine. The objectives of the present clinical trial were to establish the tolerability and efficacy of the treatment of late-onset GM2 gangliosidosis patients with escalating doses of pyrimethamine, to a maximum of 100 mg per day, administered orally in a single daily dose, over a 16-week period . The primary objective, tolerability, was assessed by regular clinical examinations, along with a panel of hematologic and biochemical studies. Although clinical efficacy could not be assessed in this short trial, treatment efficacy was evaluated by repeated measurements of leukocyte Hex A activity, expressed relative to the activity of lysosomal ß-glucuronidase. A total of 11 patients were enrolled, 8 males and 3 females, aged 23 to 50 years. One subject failed the initial screen, another was omitted from analysis because of the large number of protocol violations, and a third was withdrawn very early as a result of adverse events which were not drug-related. For the remaining 8 subjects, up to a 4-fold enhancement of Hex A activity at doses of 50 mg per day or less was observed. Additionally marked individual variations in the pharmacokinetics of the drug among the patients were noted. However, the study also found that significant side effects were experienced by most patients at or above 75 mg pyrimethamine per day. We concluded that pyrimethamine treatment enhances leukocyte Hex A activity in patients with late-onset GM2 gangliosidosis at doses lower than those associated with unacceptable side effects. Further plans are underway to extend these trials and to develop methods to assess clinical efficacy.


Gangliosidoses, GM2/drug therapy , Pyrimethamine/therapeutic use , Adult , Enzyme Assays , Female , Glucosylceramidase/blood , Hexosaminidase A/blood , Hexosaminidase B/blood , Humans , Male , Middle Aged , Pyrimethamine/adverse effects , Pyrimethamine/blood , Young Adult , beta-Galactosidase/blood
6.
Mol Genet Metab ; 98(1-2): 215-24, 2009.
Article En | MEDLINE | ID: mdl-19595619

Substrate reduction therapy (SRT) is considered to be a potential therapeutic option for juvenile GM2 gangliosidosis (jGM2g). We evaluated the efficacy of SRT in jGM2g, assessing neurological, neuropsychological and brain magnetic resonance imaging (MRI) outcomes over a 24-month period of treatment. In an open-label and single-center study, five jGM2g patients (mean age 14.6+/-4.5 years) received oral miglustat at doses of 100-200mg t.i.d. adjusted to body surface area. Patients underwent general and neurological examinations, neuropsychological, electrophysiological, and brain MRI studies. All patients showed neurological deterioration over the period of the study, with particularly notable worsening of gait, speech and coordination. One patient experienced acute psychosis, and another showed worsening of pre-existing epilepsy. Some neuropsychological tests showed no evidence of deterioration in the three patients with high enough cognitive functioning for reliable assessment. Profound cognitive impairment in two children precluded neuropsychological evaluation. In four patients, evaluation of brain MRI showed no changes in white matter signal abnormalities and cerebellar atrophy noted at baseline, while one patient showed progression of cerebellar and supratentorial brain atrophy. Transmission electron microscopy analysis of peripheral mononuclear cells showed reduction of intracytoplasmatic inclusions with treatment. SRT with miglustat of patients with jGM2g failed to ameliorate progressive neurological deterioration, but apparently no worsening of some areas of cognitive function tested and brain MRI lesions was noted over 24 months of treatment. The results must be interpreted with care owing to the small sample of patients and the lack of a control-arm.


1-Deoxynojirimycin/analogs & derivatives , Enzyme Inhibitors/therapeutic use , Gangliosidoses, GM2/drug therapy , 1-Deoxynojirimycin/therapeutic use , Adolescent , Adult , Brain Mapping , Child , Electrophysiological Phenomena , Enzyme Inhibitors/pharmacology , Female , Gangliosidoses, GM2/physiopathology , Humans , Inclusion Bodies/drug effects , Inclusion Bodies/ultrastructure , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/ultrastructure , Magnetic Resonance Imaging , Male , Neurologic Examination , Neuropsychological Tests , Substrate Specificity/drug effects , Treatment Outcome
7.
Mol Genet Metab ; 97(4): 284-91, 2009 Aug.
Article En | MEDLINE | ID: mdl-19447653

GM2 gangliosidosis (GM2g) is an inherited neurodegenerative disorder caused by deficiency of lysosomal beta-hexosaminidase A, resulting in accumulation of GM2 ganglioside, principally in the brain. Substrate reduction therapy is currently under investigation as a treatment. The study investigated the pharmacokinetics and safety of miglustat given as single and multiple doses in infantile and juvenile GM2g patients for 6- and 24-months, respectively. Eleven patients with infantile (n = 6) and juvenile (n = 5) GM2g received oral miglustat at 30-200 mg t.i.d. adjusted to the body surface area. Patients underwent pharmacokinetic assessments on day 1 and at month 3. The pharmacokinetics of miglustat were described by a 2-compartmental model with a lag time, median time to maximum concentration of 2.5 h, and terminal half-life of about 10 h. The pharmacokinetics were time-independent, and did not differ between infantile and juvenile cohorts. The accumulation index was 1.7. Among infantile GM2g patients, the major drug-related adverse events (DRAEs) were abdominal discomfort and flatulence. In the juvenile group, however, the major DRAEs observed were diarrhea and weight loss. One juvenile patient developed peripheral neuropathy, and others showed progression of already established neuropathy, which was judged to be part of the natural progression of the disease. Some mild laboratory abnormalities observed were either transient or attributable to concomitant medications. Miglustat showed similar pharmacokinetic parameters in all patients, with no specific difference between infantile and juvenile forms. Miglustat was shown to be a safe drug, with mild to moderate diarrhea, as an age-dependent DRAE, which was controlled by dietary modification.


1-Deoxynojirimycin/analogs & derivatives , Gangliosidoses, GM2/drug therapy , 1-Deoxynojirimycin/adverse effects , 1-Deoxynojirimycin/pharmacokinetics , Administration, Oral , Adolescent , Adult , Child , Child, Preschool , Diarrhea/chemically induced , Diarrhea/drug therapy , Drug-Related Side Effects and Adverse Reactions , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Glucosyltransferases/antagonists & inhibitors , Humans , Infant
8.
J Biol Chem ; 282(12): 9150-61, 2007 Mar 23.
Article En | MEDLINE | ID: mdl-17237499

Late-onset GM2 gangliosidosis is composed of two related, autosomal recessive, neurodegenerative diseases, both resulting from deficiency of lysosomal, heterodimeric beta-hexosaminidase A (Hex A, alphabeta). Pharmacological chaperones (PC) are small molecules that can stabilize the conformation of a mutant protein, allowing it to pass the quality control system of the endoplasmic reticulum. To date all successful PCs have also been competitive inhibitors. Screening for Hex A inhibitors in a library of 1040 Food Drug Administration-approved compounds identified pyrimethamine (PYR (2,4-diamino 5-(4-chlorophenyl)-6-ethylpyrimidine)) as the most potent inhibitor. Cell lines from 10 late-onset Tay-Sachs (11 alpha-mutations, 2 novel) and 7 Sandhoff (9 beta-mutations, 4 novel) disease patients, were cultured with PYR at concentrations corresponding to therapeutic doses. Cells carrying the most common late-onset mutation, alphaG269S, showed significant increases in residual Hex A activity, as did all 7 of the beta-mutants tested. Cells responding to PC treatment included those carrying mutants resulting in reduced Hex heat stability and partial splice junction mutations of the inherently less stable alpha-subunit. PYR, which binds to the active site in domain II, was able to function as PC even to domain I beta-mutants. We concluded that PYR functions as a mutation-specific PC, variably enhancing residual lysosomal Hex A levels in late-onset GM2 gangliosidosis patient cells.


Gangliosidoses, GM2/drug therapy , Gangliosidoses, GM2/metabolism , Pyrimethamine/pharmacology , Dimerization , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Folic Acid Antagonists/pharmacology , Hexosaminidase A , Humans , Lysosomes/metabolism , Models, Molecular , Molecular Chaperones , Mutation , Mutation, Missense , Protein Folding , beta-N-Acetylhexosaminidases/antagonists & inhibitors
9.
Brain ; 126(Pt 4): 974-87, 2003 Apr.
Article En | MEDLINE | ID: mdl-12615653

Mouse models of the GM2 gangliosidoses [Tay-Sachs, late onset Tay-Sachs (LOTS), Sandhoff] and GM1 gangliosidosis have been studied to determine whether there is a common neuro-inflammatory component to these disorders. During the disease course, we have: (i) examined the expression of a number of inflammatory markers in the CNS, including MHC class II, CD68, CD11b (CR3), 7/4, F4/80, nitrotyrosine, CD4 and CD8; (ii) profiled cytokine production [tumour necrosis factor alpha (TNF alpha), transforming growth factor (TGF beta 1) and interleukin 1 beta (IL1 beta)]; and (iii) studied blood-brain barrier (BBB) integrity. The kinetics of apoptosis and the expression of Fas and TNF-R1 were also assessed. In all symptomatic mouse models, a progressive increase in local microglial activation/expansion and infiltration of inflammatory cells was noted. Altered BBB permeability was evident in Sandhoff and GM1 mice, but absent in LOTS mice. Progressive CNS inflammation coincided with the onset of clinical signs in these mouse models. Substrate reduction therapy in the Sandhoff mouse model slowed the rate of accumulation of glycosphingolipids in the CNS, thus delaying the onset of the inflammatory process and disease pathogenesis. These data suggest that inflammation may play an important role in the pathogenesis of the gangliosidoses.


Antigens, CD/metabolism , Cytokines/metabolism , Gangliosidoses/etiology , Genes, MHC Class II/physiology , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/therapeutic use , Animals , Apoptosis , Biomarkers/analysis , Blood-Brain Barrier , Enzyme Inhibitors/therapeutic use , Gangliosidoses/drug therapy , Gangliosidoses/pathology , Gangliosidoses, GM2/drug therapy , Gangliosidoses, GM2/etiology , Gangliosidoses, GM2/pathology , Gangliosidosis, GM1/drug therapy , Gangliosidosis, GM1/etiology , Gangliosidosis, GM1/pathology , Immunohistochemistry , Inflammation/pathology , Mice , Sandhoff Disease/drug therapy , Sandhoff Disease/etiology , Sandhoff Disease/pathology , Tay-Sachs Disease/drug therapy , Tay-Sachs Disease/etiology , Tay-Sachs Disease/pathology
...