Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Genet Metab ; 142(1): 108444, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38555683

RESUMEN

Alpha-mannosidosis is an ultra-rare lysosomal disease that is caused by variants of the MAN2B1 gene on chromosome 19p13. These variants result in faulty or absent alpha-mannosidase in lysosomes, which leads to intracellular accumulation of mannose-containing oligosaccharides. Diagnosis of alpha-mannosidosis is often delayed, in part because of the rarity of the disease, its gradual onset and heterogeneity of presentation, but also because of the similarity of many signs and symptoms of the disease to those of other lysosomal diseases. Treatment of alpha-mannosidosis was previously limited to hematopoietic stem cell transplantation, but outcomes are variable and not all patients are eligible or have a suitable donor. Recently, an enzyme replacement therapy, recombinant human alpha-mannosidase (velmanase alfa), was approved for the treatment of non-neurological manifestations in adult and pediatric patients with alpha-mannosidosis. Treatment with velmanase alfa reduces serum levels of oligosaccharides, increases levels of immunoglobulin G, and improves patients' functional capacity and quality of life, although it is not effective for the neurologic phenotype because it does not cross the blood-brain barrier. Since the effects of velmanase alfa are more marked in children than adults, early diagnosis to allow early initiation of treatment has become more important. To support this, patient, parent/caregiver, and clinician awareness and education is imperative. A number of approaches can be taken to meet this goal, such as the development of disease registries, validated diagnostic algorithms, and screening tools, improved under-/post-graduate clinician education, easily accessible and reliable information for patients/families (such as that made available on the internet), and the formation of patient advocacy groups. Such approaches may raise awareness of alpha-mannosidosis, reduce the diagnostic delay and thus improve the lives of those affected.


Asunto(s)
Diagnóstico Tardío , Terapia de Reemplazo Enzimático , alfa-Manosidasa , alfa-Manosidosis , Humanos , alfa-Manosidosis/diagnóstico , alfa-Manosidosis/genética , alfa-Manosidasa/genética , Enfermedades Raras/diagnóstico , Enfermedades Raras/genética
2.
Int J Mol Sci ; 25(12)2024 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-38928321

RESUMEN

Glucocerebrosidase (GCase) is a lysosomal enzyme that catalyzes the breakdown of glucosylceramide in the presence of its activator saposin C (SapC). SapC arises from the proteolytical cleavage of prosaposin (encoded by PSAP gene), which gives rise to four saposins. GCase is targeted to the lysosomes by LIMP-2, encoded by SCARB2 gene. GCase deficiency causes Gaucher Disease (GD), which is mainly due to biallelic pathogenetic variants in the GCase-encoding gene, GBA1. However, impairment of GCase activity can be rarely caused by SapC or LIMP-2 deficiencies. We report a new case of LIMP-2 deficiency and a new case of SapC deficiency (missing all four saposins, PSAP deficiency), and measured common biomarkers of GD and GCase activity. Glucosylsphingosine and chitotriosidase activity in plasma were increased in GCase deficiencies caused by PSAP and GBA1 mutations, whereas SCARB2-linked deficiency showed only Glucosylsphingosine elevation. GCase activity was reduced in fibroblasts and leukocytes: the decrease was sharper in GBA1- and SCARB2-mutant fibroblasts than PSAP-mutant ones; LIMP-2-deficient leukocytes displayed higher residual GCase activity than GBA1-mutant ones. Finally, we demonstrated that GCase mainly undergoes proteasomal degradation in LIMP-2-deficient fibroblasts and lysosomal degradation in PSAP-deficient fibroblasts. Thus, we analyzed the differential biochemical profile of GCase deficiencies due to the ultra-rare PSAP and SCARB2 biallelic pathogenic variants in comparison with the profile observed in GBA1-linked GCase deficiency.


Asunto(s)
Enfermedad de Gaucher , Glucosilceramidasa , Proteínas de Membrana de los Lisosomas , Receptores Depuradores , Saposinas , Glucosilceramidasa/genética , Glucosilceramidasa/deficiencia , Glucosilceramidasa/metabolismo , Humanos , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/metabolismo , Saposinas/deficiencia , Saposinas/genética , Saposinas/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Proteínas de Membrana de los Lisosomas/genética , Receptores Depuradores/genética , Receptores Depuradores/metabolismo , Fibroblastos/metabolismo , Mutación , Lisosomas/metabolismo , Lisosomas/enzimología , Hexosaminidasas/metabolismo , Hexosaminidasas/genética , Hexosaminidasas/deficiencia , Masculino , Femenino
3.
Int J Mol Sci ; 24(13)2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37446383

RESUMEN

Gaucher disease (GD) is caused by biallelic pathogenic variants in the acid ß-glucosidase gene (GBA1), leading to a deficiency in the ß-glucocerebrosidase (GCase) enzyme activity resulting in the intracellular accumulation of sphingolipids. Skeletal alterations are one of the most disabling features in GD patients. Although both defective bone formation and increased bone resorption due to osteoblast and osteoclast dysfunction contribute to GD bone pathology, the molecular bases are not fully understood, and bone disease is not completely resolved with currently available specific therapies. For this reason, using editing technology, our group has developed a reliable, isogenic, and easy-to-handle cellular model of GD monocytes (GBAKO-THP1) to facilitate GD pathophysiology studies and high-throughput drug screenings. In this work, we further characterized the model showing an increase in proinflammatory cytokines (Interleukin-1ß and Tumor Necrosis Factor-α) release and activation of osteoclastogenesis. Furthermore, our data suggest that GD monocytes would display an increased osteoclastogenic potential, independent of their interaction with the GD microenvironment or other GD cells. Both proinflammatory cytokine production and osteoclastogenesis were restored at least, in part, by treating cells with the recombinant human GCase, a substrate synthase inhibitor, a pharmacological chaperone, and an anti-inflammatory compound. Besides confirming that this model would be suitable to perform high-throughput screening of therapeutic molecules that act via different mechanisms and on different phenotypic features, our data provided insights into the pathogenic cascade, leading to osteoclastogenesis exacerbation and its contribution to bone pathology in GD.


Asunto(s)
Enfermedad de Gaucher , Humanos , Enfermedad de Gaucher/patología , Osteogénesis , Monocitos/patología , Sistemas CRISPR-Cas , Diferenciación Celular
4.
Genet Med ; 24(7): 1425-1436, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35471153

RESUMEN

PURPOSE: This trial aimed to assess the efficacy and safety of olipudase alfa enzyme replacement therapy for non-central nervous system manifestations of acid sphingomyelinase deficiency (ASMD) in adults. METHODS: A phase 2/3, 52 week, international, double-blind, placebo-controlled trial (ASCEND; NCT02004691/EudraCT 2015-000371-26) enrolled 36 adults with ASMD randomized 1:1 to receive olipudase alfa or placebo intravenously every 2 weeks with intrapatient dose escalation to 3 mg/kg. Primary efficacy endpoints were percent change from baseline to week 52 in percent predicted diffusing capacity of the lung for carbon monoxide and spleen volume (combined with splenomegaly-related score in the United States). Other outcomes included liver volume/function/sphingomyelin content, pulmonary imaging/function, platelet levels, lipid profiles, and pharmacodynamics. RESULTS: Least square mean percent change from baseline to week 52 favored olipudase alfa over placebo for percent predicted diffusing capacity of the lung for carbon monoxide (22% vs 3.0% increases, P = .0004), spleen volume (39% decrease vs 0.5% increase, P < .0001), and liver volume (28% vs 1.5% decreases, P < .0001). Splenomegaly-related score decreased in both groups (P = .64). Other clinical outcomes improved in the olipudase alfa group compared with the placebo group. There were no treatment-related serious adverse events or adverse event-related discontinuations. Most adverse events were mild. CONCLUSION: Olipudase alfa was well tolerated and associated with significant and comprehensive improvements in disease pathology and clinically relevant endpoints compared with placebo in adults with ASMD.


Asunto(s)
Enfermedad de Niemann-Pick Tipo A , Adulto , Monóxido de Carbono/uso terapéutico , Método Doble Ciego , Terapia de Reemplazo Enzimático/métodos , Humanos , Proteínas Recombinantes , Esfingomielina Fosfodiesterasa , Esplenomegalia
5.
Genet Med ; 23(8): 1543-1550, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33875845

RESUMEN

PURPOSE: To assess olipudase alfa enzyme replacement therapy for non-central nervous system manifestations of acid sphingomyelinase deficiency (ASMD) in children. METHODS: This phase 1/2, international, multicenter, open-label trial (ASCEND-Peds/NCT02292654) administered intravenous olipudase alfa every 2 weeks with intrapatient dose escalation to 3 mg/kg. Primary outcome was safety through week 64. Secondary outcomes included pharmacokinetics, spleen and liver volumes, lung diffusing capacity (DLCO), lipid profiles, and height through week 52. RESULTS: Twenty patients were enrolled: four adolescents (12-17 years), nine children (6-11 years), and seven infants/early child (1-5 years). Most adverse events were mild or moderate, including infusion-associated reactions (primarily urticaria, pyrexia, and/or vomiting) in 11 patients. Three patients had serious treatment-related events: one with transient asymptomatic alanine aminotransferase increases, another with urticaria and rash (antidrug antibody positive [ADA+]), and a third with an anaphylactic reaction (ADA+) who underwent desensitization and reached the 3 mg/kg maintenance dose. Mean splenomegaly and hepatomegaly improved by >40% (p < 0.0001). Mean % predicted DLCO improved by 32.9% (p = 0.0053) in patients able to perform the test. Lipid profiles and elevated liver transaminase levels normalized. Mean height Z-scores improved by 0.56 (p < 0.0001). CONCLUSION: In this study in children with chronic ASMD, olipudase alfa was generally well-tolerated with significant, comprehensive improvements in disease pathology across a range of clinically relevant endpoints.


Asunto(s)
Enfermedad de Niemann-Pick Tipo A , Adolescente , Niño , Preescolar , Terapia de Reemplazo Enzimático , Humanos , Lactante , Hígado , Enfermedad de Niemann-Pick Tipo A/tratamiento farmacológico , Enfermedad de Niemann-Pick Tipo A/genética , Proteínas Recombinantes/uso terapéutico , Esfingomielina Fosfodiesterasa/genética
6.
J Inherit Metab Dis ; 44(5): 1124-1135, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33844307

RESUMEN

Patients with inborn errors of metabolism causing fasting intolerance can experience acute metabolic decompensations. Long-term data on outcomes using emergency letters are lacking. This is a retrospective, observational, single-center study of the use of emergency letters based on a generic emergency protocol in patients with hepatic glycogen storage diseases (GSD) or fatty acid oxidation disorders (FAOD). Data on hospital admissions, initial laboratory results, and serious adverse events were collected. Subsequently, the website www.emergencyprotocol.net was generated in the context of the CONNECT MetabERN eHealth project following multiple meetings, protocol revisions, and translations. Representing 470 emergency protocol years, 127 hospital admissions were documented in 54/128 (42%) patients who made use of emergency letters generated based on the generic emergency protocol. Hypoglycemia (here defined as glucose concentration < 3.9 mmol/L) was reported in only 15% of hospital admissions and was uncommon in patients with ketotic GSD and patients with FAOD aged >5 years. Convulsions, coma, or death was not documented. By providing basic information, emergency letters for individual patients with hepatic GSD or the main FAOD can be generated at www.emergencyprotocol.net, in nine different languages. Generic emergency protocols are safe and easy for home management by the caregivers and the first hour in-hospital management to prevent metabolic emergencies in patients with hepatic GSD and medium-chain Acyl CoA dehydrogenase deficiency. The website www.emergencyprotocol.net is designed to support families and healthcare providers to generate personalized emergency letters for patients with hepatic GSD and the main FAOD.


Asunto(s)
Tratamiento de Urgencia/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Hipoglucemia/terapia , Errores Innatos del Metabolismo Lipídico/metabolismo , Telemedicina , Adolescente , Adulto , Niño , Preescolar , Ayuno , Ácidos Grasos/metabolismo , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo I/fisiopatología , Humanos , Hipoglucemia/etiología , Lactante , Recién Nacido , Errores Innatos del Metabolismo Lipídico/fisiopatología , Masculino , Persona de Mediana Edad , Oxidación-Reducción , Estudios Retrospectivos , Adulto Joven
7.
Int J Mol Sci ; 22(23)2021 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-34884674

RESUMEN

Acid sphingomyelinase deficiency (ASMD) is a lysosomal storage disease caused by deficient activity of acid sphingomyelinase (ASM) enzyme, leading to the accumulation of varying degrees of sphingomyelin. Lipid storage leads to foam cell infiltration in tissues, and clinical features including hepatosplenomegaly, pulmonary insufficiency and in some cases central nervous system involvement. ASM enzyme replacement therapy is currently in clinical trial being the first treatment addressing the underlying pathology of the disease. Therefore, presently, it is critical to better comprehend ASMD to improve its diagnose and monitoring. Lung disease, including recurrent pulmonary infections, are common in ASMD patients. Along with lung disease, several immune system alterations have been described both in patients and in ASMD animal models, thus highlighting the role of ASM enzyme in the immune system. In this review, we summarized the pivotal roles of ASM in several immune system cells namely on macrophages, Natural Killer (NK) cells, NKT cells, B cells and T cells. In addition, an overview of diagnose, monitoring and treatment of ASMD is provided highlighting the new enzyme replacement therapy available.


Asunto(s)
Enfermedades por Almacenamiento Lisosomal/inmunología , Esfingomielina Fosfodiesterasa/deficiencia , Animales , Terapia de Reemplazo Enzimático , Humanos , Enfermedades Pulmonares/enzimología , Enfermedades por Almacenamiento Lisosomal/diagnóstico , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/terapia , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/inmunología
8.
Int J Mol Sci ; 22(11)2021 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-34073924

RESUMEN

Gaucher disease (GD) is an autosomal recessive lysosomal disorder due to beta-glucosidase gene (GBA) mutations. The molecular diagnosis of GD is complicated by the presence of recombinant alleles originating from a highly homologous pseudogene. Clinical exome sequencing (CES) is a rapid genetic approach for identifying disease-causing mutations. However, copy number variation and recombination events are poorly detected, and further investigations are required to avoid mis-genotyping. The aim of this work was to set-up an integrated strategy for GD patients genotyping using CES as a first-line test. Eight patients diagnosed with GD were analyzed by CES. Five patients were fully genotyped, while three were revealed to be homozygous for mutations that were not confirmed in the parents. Therefore, MLPA (multiplex ligation-dependent probe amplification) and specific long-range PCR were performed, and two recombinant alleles, one of them novel, and one large deletion were identified. Furthermore, an MLPA assay performed in one family resulted in the identification of an additional novel mutation (p.M124V) in a relative, in trans with the known p.N409S mutation. In conclusion, even though CES has become extensively used in clinical practice, our study emphasizes the importance of a comprehensive molecular strategy to provide proper GBA genotyping and genetic counseling.


Asunto(s)
Exoma/genética , Enfermedad de Gaucher/diagnóstico , Reacción en Cadena de la Polimerasa Multiplex/métodos , beta-Glucosidasa/genética , Alelos , Variaciones en el Número de Copia de ADN , Familia , Femenino , Enfermedad de Gaucher/genética , Genotipo , Células HEK293 , Homocigoto , Humanos , Masculino , Mutación , Linaje
9.
Mol Genet Metab ; 130(3): 170-171, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32386848

RESUMEN

The direct and indirect effects of Coronavirus Disease-19 (COVID-19) pandemic, on Italian patients with lysosomal storage disorders receiving therapy, were analyzed by a phone questionnaire. No proved COVID-19 emerged among 102 interviewed. No problems were reported by patients receiving oral treatments. Forty-nine% of patients receiving enzyme replacement therapy in hospitals experienced disruptions, versus 6% of those home-treated. The main reasons of missed infusions were fear of infection (62.9%) and re-organization of the infusion centers (37%).


Asunto(s)
Infecciones por Coronavirus/epidemiología , Enfermedades por Almacenamiento Lisosomal/terapia , Neumonía Viral/epidemiología , Adulto , COVID-19 , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/psicología , Infecciones por Coronavirus/terapia , Terapia de Reemplazo Enzimático , Miedo , Femenino , Humanos , Italia/epidemiología , Enfermedades por Almacenamiento Lisosomal/complicaciones , Masculino , Persona de Mediana Edad , Pandemias , Aceptación de la Atención de Salud , Manejo de Atención al Paciente , Neumonía Viral/complicaciones , Neumonía Viral/psicología , Neumonía Viral/terapia , Encuestas y Cuestionarios , Adulto Joven
10.
Eur J Pediatr ; 178(5): 739-753, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30809705

RESUMEN

Mucopolysaccharidoses (MPS) are a subgroup of 11 monogenic lysosomal storage disorders due to the deficit of activity of the lysosomal hydrolases deputed to the degradation of mucopolysaccharides. Although individually rare, all together they account for at least 1:25,000 live births. In this study, we present the genetic analysis of a population of 71 MPS patients enrolled in a multicenter Italian study. We re-annotated all variants, according to the latest recommendations, and re-classified them as suggested by the American College of Medical Genetics and Genomics. Variant distribution per type was mainly represented by missense mutations. Overall, 10 patients had received no molecular diagnosis, although 6 of them had undergone either HSCT or ERT, based on clinical and enzymatic evaluations. Moreover, nine novel variants are reported.Conclusions: Our analysis underlines the need to complete the molecular diagnosis in patients previously diagnosed only on a biochemical basis, suggests a periodical re-annotation of the variants and solicits their deposition in public databases freely available to clinicians and researchers. We strongly recommend a molecular diagnosis based on the analysis of the "trio" instead of the sole proband. These recommendations will help to obtain a complete and correct diagnosis of mucopolysaccharidosis, rendering also possible genetic counseling. What is known • MPS are a group of 11 metabolic genetic disorders due to deficits of enzymes involved in the mucopolysaccharides degradation. • Molecular analysis is commonly performed to confirm enzymatic assays. What is new • Eighty-six percent of the 71 patients we collected received a molecular diagnosis; among them, 9 novel variants were reported. • We stress the importance of molecular diagnosis in biochemically diagnosed patients, encourage a periodical re-annotation of variants according to the recent nomenclature and their publication in open databases.


Asunto(s)
Pruebas Genéticas , Técnicas de Diagnóstico Molecular , Mucopolisacaridosis/diagnóstico , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Asesoramiento Genético , Marcadores Genéticos , Humanos , Lactante , Italia , Masculino , Mucopolisacaridosis/genética , Mutación Missense , Adulto Joven
11.
Int J Mol Sci ; 20(8)2019 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-31022913

RESUMEN

Mucopolysaccharidosis type II (MPSII) is a lysosomal storage disorder due to the deficit of the enzyme iduronate 2-sulfatase (IDS), which leads to the accumulation of glycosaminoglycans in most organ-systems, including the brain, and resulting in neurological involvement in about two-thirds of the patients. The main treatment is represented by a weekly infusion of the functional enzyme, which cannot cross the blood-brain barrier and reach the central nervous system. In this study, a tailored nanomedicine approach based on brain-targeted polymeric nanoparticles (g7-NPs), loaded with the therapeutic enzyme, was exploited. Fibroblasts from MPSII patients were treated for 7 days with NPs loaded with the IDS enzyme; an induced IDS activity like the one detected in healthy cells was measured, together with a reduction of GAG content to non-pathological levels. An in vivo short-term study in MPSII mice was performed by weekly administration of g7-NPs-IDS. Biochemical, histological, and immunohistochemical evaluations of liver and brain were performed. The 6-weeks treatment produced a significant reduction of GAG deposits in liver and brain tissues, as well as a reduction of some neurological and inflammatory markers (i.e., LAMP2, CD68, GFAP), highlighting a general improvement of the brain pathology. The g7-NPs-IDS approach allowed a brain-targeted enzyme replacement therapy. Based on these positive results, the future aim will be to optimize NP formulation further to gain a higher efficacy of the proposed approach.


Asunto(s)
Encéfalo/efectos de los fármacos , Portadores de Fármacos/metabolismo , Sistemas de Liberación de Medicamentos , Iduronato Sulfatasa/administración & dosificación , Mucopolisacaridosis II/tratamiento farmacológico , Nanopartículas/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Encéfalo/patología , Portadores de Fármacos/química , Terapia de Reemplazo Enzimático , Glicopéptidos/química , Glicopéptidos/metabolismo , Humanos , Iduronato Sulfatasa/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis II/enzimología , Mucopolisacaridosis II/metabolismo , Mucopolisacaridosis II/patología , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química
12.
N Engl J Med ; 373(11): 1010-20, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26352813

RESUMEN

BACKGROUND: Lysosomal acid lipase is an essential lipid-metabolizing enzyme that breaks down endocytosed lipid particles and regulates lipid metabolism. We conducted a phase 3 trial of enzyme-replacement therapy in children and adults with lysosomal acid lipase deficiency, an underappreciated cause of cirrhosis and severe dyslipidemia. METHODS: In this multicenter, randomized, double-blind, placebo-controlled study involving 66 patients, we evaluated the safety and effectiveness of enzyme-replacement therapy with sebelipase alfa (administered intravenously at a dose of 1 mg per kilogram of body weight every other week); the placebo-controlled phase of the study was 20 weeks long and was followed by open-label treatment for all patients. The primary end point was normalization of the alanine aminotransferase level. Secondary end points included additional disease-related efficacy assessments, safety, and side-effect profile. RESULTS: Substantial disease burden at baseline included a very high level of low-density lipoprotein cholesterol (≥190 mg per deciliter) in 38 of 66 patients (58%) and cirrhosis in 10 of 32 patients (31%) who underwent biopsy. A total of 65 of the 66 patients who underwent randomization completed the double-blind portion of the trial and continued with open-label treatment. At 20 weeks, the alanine aminotransferase level was normal in 11 of 36 patients (31%) in the sebelipase alfa group and in 2 of 30 (7%) in the placebo group (P=0.03), with mean changes from baseline of -58 U per liter versus -7 U per liter (P<0.001). With respect to prespecified key secondary efficacy end points, we observed improvements in lipid levels and reduction in hepatic fat content (P<0.001 for all comparisons, except P=0.04 for triglycerides). The number of patients with adverse events was similar in the two groups; most events were mild and were considered by the investigator to be unrelated to treatment. CONCLUSIONS: Sebelipase alfa therapy resulted in a reduction in multiple disease-related hepatic and lipid abnormalities in children and adults with lysosomal acid lipase deficiency. (Funded by Synageva BioPharma and others; ARISE ClinicalTrials.gov number, NCT01757184.).


Asunto(s)
Esterol Esterasa/uso terapéutico , Enfermedad de Wolman/tratamiento farmacológico , Adolescente , Adulto , Anciano , Alanina Transaminasa/sangre , Biopsia , Niño , Preescolar , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Método Doble Ciego , Dislipidemias/tratamiento farmacológico , Dislipidemias/genética , Femenino , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Persona de Mediana Edad , Esterol Esterasa/efectos adversos , Esterol Esterasa/farmacología , Enfermedad de Wolman/sangre , Adulto Joven , Enfermedad de Wolman
13.
Acta Paediatr ; 107(8): 1402-1408, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29797470

RESUMEN

AIM: The aim of this study was to develop an algorithm to prompt early clinical suspicion of mucopolysaccharidosis type I (MPS I). METHODS: An international working group was established in 2016 that comprised 11 experts in paediatrics, rare diseases and inherited metabolic diseases. They reviewed real-world clinical cases, selected key signs or symptoms based on their prevalence and specificity and reached consensus about the algorithm. The algorithm was retrospectively tested. RESULTS: An algorithm was developed. In patients under two years of age, kyphosis or gibbus deformity were the key symptoms that raised clinical suspicion of MPS I and in those over two years they were kyphosis or gibbus deformity, or joint stiffness or contractures without inflammation. The algorithm was tested on 35 cases, comprising 16 Hurler, 10 Hurler-Scheie, and nine Scheie patients. Of these 35 cases, 32 (91%) - 16 Hurler, nine Hurler-Scheie and seven Scheie patients - would have been referred earlier if the algorithm had been used. CONCLUSION: The expert panel developed and tested an algorithm that helps raise clinical suspicion of MPS I and would lead to a more prompt final diagnosis and allow earlier treatment.


Asunto(s)
Algoritmos , Diagnóstico Precoz , Mucopolisacaridosis I/diagnóstico , Mucopolisacaridosis I/terapia , Tamizaje Neonatal/métodos , Factores de Edad , Niño , Preescolar , Consenso , Progresión de la Enfermedad , Femenino , Humanos , Recién Nacido , Internacionalidad , Masculino , Multimorbilidad , Pronóstico , Estudios Retrospectivos , Medición de Riesgo , Índice de Severidad de la Enfermedad , Factores Sexuales
14.
Acta Paediatr ; 107(12): 2059-2065, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30242902

RESUMEN

AIM: Mucopolysaccharidosis type I is a lysosomal storage disorder that can result in significant disease burden, disability and premature death, if left untreated. The aim of this review was to elaborate on the diagnosis of mucopolysaccharidosis type I and the pros and cons of newborn screening. METHODS: An international working group was established to discuss ways to improve the early diagnosis of mucopolysaccharidosis type I. It consisted of 13 experts in paediatrics, rare diseases and inherited metabolic diseases from Europe and the Middle East. RESULTS: It is becoming increasingly clearer that the delay between symptom onset and clinical diagnosis is considerable for mucopolysaccharidosis type I and other rare lysosomal storage disorders, despite numerous awareness campaigns since therapies became available. Diagnosis currently depends on recognising the signs and symptoms of the disease. The practice of newborn screening, which is being explored by pilot programmes around the world, enables early diagnosis and consequently early treatment. However, these studies have highlighted numerous new problems and pitfalls that must be faced before newborn screening becomes generally available. CONCLUSION: Newborn screening for mucopolysaccharidosis type I offers the potential for early diagnosis and early pre-symptomatic treatment, but existing hurdles need to be overcome.


Asunto(s)
Mucopolisacaridosis I/diagnóstico , Tamizaje Neonatal , Humanos , Recién Nacido
16.
Mol Genet Metab ; 122S: 55-61, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29170080

RESUMEN

The mucopolysaccharidosis (MPS) disorders are caused by deficiencies of specific lysosomal enzymes involved in the catabolism of glycosaminoglycans (GAGs). The resulting GAG accumulation in cells and tissues throughout the body leads to progressive multi-organ dysfunction. MPS patients present with several somatic manifestations, including short stature, musculoskeletal abnormalities, and cardiorespiratory dysfunction, and several primary and secondary neurological signs and symptoms. Epileptic seizures are neurological signs of MPS thought to develop due to accumulation of GAGs in the brain, triggering alterations in neuronal connectivity and signaling, and release of inflammatory mediators. The amount of literature on the prevalence, pathophysiology, clinical features, and management of epileptic seizures in patients with MPS is limited. This review discusses current knowledge on this topic, as well as two case examples, presented and discussed during a closed meeting on MPS and the brain among an international group of experts with extensive experience in managing and treating MPS.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Encéfalo/diagnóstico por imagen , Epilepsia/diagnóstico , Glicosaminoglicanos/toxicidad , Mucopolisacaridosis/complicaciones , Adolescente , Anticonvulsivantes/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/metabolismo , Niño , Electroencefalografía/métodos , Epilepsia/tratamiento farmacológico , Epilepsia/epidemiología , Epilepsia/etiología , Femenino , Glicosaminoglicanos/metabolismo , Humanos , Imagen por Resonancia Magnética , Masculino , Mucopolisacaridosis/genética , Mucopolisacaridosis/patología , Mucopolisacaridosis/terapia , Prevalencia , Resultado del Tratamiento
17.
Mol Genet Metab ; 122S: 25-34, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29153844

RESUMEN

The mucopolysaccharidosis (MPS) disorders are a group of lysosomal storage diseases caused by lysosomal enzyme deficits that lead to glycosaminoglycan accumulation, affecting various tissues throughout the body based on the specific enzyme deficiency. These disorders are characterized by their progressive nature and a variety of somatic manifestations and neurological symptoms. There are established treatments for some MPS disorders, but these mostly alleviate somatic and non-neurological symptoms and do not cure the disease. Patients with MPS I, II, III, and VII can present with neurological manifestations such as neurocognitive decline and behavioral problems. Treatment of these neurological manifestations remains challenging due to the blood-brain barrier (BBB) that limits delivery of therapeutic agents to the central nervous system (CNS). New therapies that circumvent this barrier and target brain disease in MPS are currently under development. They primarily focus on facilitating penetration of drugs through the BBB, delivery of recombinant enzyme to the brain by gene therapy, or direct CNS administration. This review summarizes existing and potential future treatment approaches that target brain disease in MPS. The information in this review is based on current literature and presentations and discussions during a closed meeting by an international group of experts with extensive experience in managing and treating MPS.


Asunto(s)
Encéfalo/efectos de los fármacos , Disfunción Cognitiva/terapia , Terapia de Reemplazo Enzimático/métodos , Mucopolisacaridosis/terapia , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/citología , Encéfalo/metabolismo , Niño , Conducta Infantil/efectos de los fármacos , Desarrollo Infantil/efectos de los fármacos , Preescolar , Ensayos Clínicos como Asunto , Disfunción Cognitiva/diagnóstico , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Congresos como Asunto , Portadores de Fármacos/química , Terapia Genética/métodos , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/toxicidad , Trasplante de Células Madre Hematopoyéticas , Humanos , Inyecciones Intraventriculares , Inyecciones Espinales , Chaperonas Moleculares/uso terapéutico , Mucopolisacaridosis/diagnóstico , Mucopolisacaridosis/genética , Mucopolisacaridosis/patología , Nanopartículas/química , Proteínas Recombinantes/uso terapéutico
18.
Eur J Pediatr ; 176(3): 395-405, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28093642

RESUMEN

The need for performing clinical trials to develop well-studied and appropriate medicines for inherited neurometabolic disease patients faces ethical concerns mainly raising from four aspects: the diseases are rare; include young and very young patients; the neurological impairment may compromise the capability to provide 'consent'; and the genetic nature of the disease leads to further ethical implications. This work is intended to identify the ethical provisions applicable to clinical research involving these patients and to evaluate if these cover the ethical issues. Three searches have been performed on the European regulatory/legal framework, the literature and European Union-funded projects. The European legal framework offers a number of ethical provisions ruling the clinical research on paediatric, rare, inherited diseases with neurological symptoms. In the literature, relevant publications deal with informed consent, newborn genetic screenings, gene therapy and rights/interests of research participants. Additional information raised from European projects on sharing patients' data from different countries, the need to fill the gap of the regulatory framework and to improve information to stakeholders and patients/families. CONCLUSION: Several recommendations and guidelines on ethical aspects are applicable to the inherited neurometabolic disease research in Europe, even though they suffer from the lack of a common ethical approach. What is Known: • When planning and conducting clinical trials, sponsors and researchers know that clinical trials are to be performed according to well-established ethical rules, and patients should be aware about their rights. • In the cases of paediatric patients, vulnerable patients unable to provide consent, genetic diseases' further rules apply. What is New: • This work discusses which ethical rules apply to ensure protection of patient's rights if all the above-mentioned features coexist. • This work shows available data and information on how these rules have been applied.


Asunto(s)
Investigación Biomédica/ética , Ensayos Clínicos como Asunto/ética , Consentimiento Informado/legislación & jurisprudencia , Enfermedades Metabólicas , Enfermedades del Sistema Nervioso , Enfermedades Raras , Niño , Europa (Continente) , Unión Europea , Humanos
19.
Metab Brain Dis ; 32(5): 1529-1536, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28577204

RESUMEN

Juvenile and adult GM1-gangliosidosis are invariably characterized by progressive neurological deterioration. To date only symptomatic therapies are available. We report for the first time the positive results of Miglustat (OGT 918, N-butyl-deoxynojirimycin) treatment on three Italian GM1-gangliosidosis patients. The first two patients had a juvenile form (enzyme activity ≤5%, GLB1 genotype p.R201H/c.1068 + 1G > T; p.R201H/p.I51N), while the third patient had an adult form (enzyme activity about 7%, p.T329A/p.R442Q). Treatment with Miglustat at the dose of 600 mg/day was started at the age of 10, 17 and 28 years; age at last evaluation was 21, 20 and 38 respectively. Response to treatment was evaluated using neurological examinations in all three patients every 4-6 months, the assessment of Movement Disorder-Childhood Rating Scale (MD-CRS) in the second patient, and the 6-Minute Walking Test (6-MWT) in the third patient. The baseline neurological status was severely impaired, with loss of autonomous ambulation and speech in the first two patients, and gait and language difficulties in the third patient. All three patients showed gradual improvement while being treated; both juvenile patients regained the ability to walk without assistance for few meters, and increased alertness and vocalization. The MD-CRS class score in the second patient decreased from 4 to 2. The third patient improved in movement and speech control, the distance covered during the 6-MWT increased from 338 to 475 m. These results suggest that Miglustat may help slow down or reverse the disease progression in juvenile/adult GM1-gangliosidosis.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Gangliosidosis GM1/tratamiento farmacológico , Inhibidores de Glicósido Hidrolasas/uso terapéutico , 1-Desoxinojirimicina/uso terapéutico , Adulto , Factores de Edad , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/psicología , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Marcha , Gangliosidosis GM1/genética , Gangliosidosis GM1/psicología , Genotipo , Humanos , Trastornos del Lenguaje/tratamiento farmacológico , Trastornos del Lenguaje/psicología , Destreza Motora , Trastornos del Movimiento/tratamiento farmacológico , Trastornos del Movimiento/psicología , Pruebas Neuropsicológicas , Resultado del Tratamiento , Caminata , Adulto Joven , beta-Galactosidasa/genética
20.
Klin Padiatr ; 229(3): 168-174, 2017 May.
Artículo en Alemán | MEDLINE | ID: mdl-28464188

RESUMEN

Background Due to their rarity studies in (ultra-) rare diseases are difficult. Only for a minority of these diseases causal therapies are available. Development and production of enzyme replacement therapies (ERT) for example are challenging and expensive. The number of patients is low, costs per patient are high. We will focus on the challenges of providing long-term ERT to patients with lysosomal storage diseases (LSD) in an out- and inpatient setting based on a literature search in Pubmed and own experience. Many ERTs for LSDs have a positive cost-benefit ratio. Possible side-effects are severe allergic reactions. ERT is covered by the insurance companies when prescribed by a physician, however they are liable to recourse by the insurance company as the expenses for drugs of the prescribing physician will be above average. In most cases the recourse can be averted if diagnoses of individual patients are disclosed. Intravenous infusion of ERT is not well-regulated in Germany/Austria. Infusion on a ward is safe however often not covered by the insurance companies as patients do not stay overnight. Another option is infusion in a day-care setting, however the lump sum paid for infusion does not cover costs for ERT. On an individual basis, reimbursement for medication (ERT) has to be negotiated with the insurance companies before infusion takes place. Home infusions are feasible, however careful evaluations of the infusion-team and the risk for side-effects have to be performed on an individual basis, legal issues have to be considered. In- and outpatient ERT of patients with LSDs is challenging but feasible after individual evaluation of patient and infusion team.


Asunto(s)
Terapia de Reemplazo Enzimático , Comunicación Interdisciplinaria , Colaboración Intersectorial , Enfermedades por Almacenamiento Lisosomal/terapia , Niño , Terapia de Reemplazo Enzimático/efectos adversos , Terapia de Reemplazo Enzimático/economía , Alemania , Terapia de Infusión a Domicilio/economía , Hospitalización/economía , Humanos , Cobertura del Seguro/economía , Cuidados a Largo Plazo/economía , Enfermedades por Almacenamiento Lisosomal/diagnóstico , Enfermedades por Almacenamiento Lisosomal/economía , Enfermedades por Almacenamiento Lisosomal/enzimología , Programas Nacionales de Salud/economía , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA