Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Orphanet J Rare Dis ; 18(1): 381, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38057861

ABSTRACT

BACKGROUND: Studies indicate that doses of alglucosidase alfa (ALGLU) higher than label dose (20 mg/kg every other week) improve clinical outcomes in infantile-onset Pompe disease (IOPD). We investigated data from the Pompe Registry to determine the association between ALGLU dose and survival in IOPD. RESULTS: We included 332 IOPD patients from the Registry as of January 2022 who had cardiomyopathy and were first treated at age < 1 year. We used Cox proportional hazards models to estimate hazard ratios (HR) and 95% confidence intervals (CI) for the association between ALGLU as a time-varying exposure and survival, adjusting for age at first treatment, sex, and cross-reactive immunologic material (CRIM)/immune tolerance induction (ITI) status. Dose was measured as average relative dose received over time (in multiples of label dose, range > 0 to 4 times label dose), current dose, and lagged dose. 81% patients received label dose at treatment initiation. Over time, 52% received a higher dose. Higher ALGLU dose over time was associated with improved survival: adjusted HR 0.40 (95% CI 0.22-0.73, p = 0.003) per 1-unit increase in average relative dose, with similar results for invasive ventilation-free survival (adjusted HR 0.48, 95% CI 0.28-0.84; p = 0.010). The association was consistent in patients first treated before or after 3 months of age and did not vary significantly by CRIM status. Results for current and lagged dose were similar to average dose. CONCLUSIONS: Higher ALGLU doses were associated with significantly improved overall and invasive ventilator-free survival in IOPD. Results were consistent across sensitivity analyses.


Subject(s)
Glycogen Storage Disease Type II , Humans , Glycogen Storage Disease Type II/drug therapy , alpha-Glucosidases/therapeutic use , Registries , Enzyme Replacement Therapy/methods
2.
Genet Med ; 25(2): 100328, 2023 02.
Article in English | MEDLINE | ID: mdl-36542086

ABSTRACT

PURPOSE: Mini-COMET (NCT03019406; Sanofi) is a phase 2, open-label, ascending-dose, 3-cohort study, evaluating avalglucosidase alfa safety, pharmacokinetics, and efficacy in individuals with infantile-onset Pompe disease aged <18 years who previously received alglucosidase alfa and showed clinical decline (cohorts 1 and 2) or suboptimal response (cohort 3). METHODS: During a 25-week primary analysis period, cohorts 1 and 2 received avalglucosidase alfa 20 and 40 mg/kg every other week, respectively, for 6 months, whereas cohort 3 individuals were randomized (1:1) to receive avalglucosidase alfa 40 mg/kg every other week or alglucosidase alfa (current stable dose) for 6 months. RESULTS: In total, 22 individuals were enrolled (cohort 1 [n = 6], cohort 2 [n = 5], cohort 3-avalglucosidase alfa [n = 5], and cohort 3-alglucosidase alfa [n = 6]). Median treatment compliance was 100%. None of the individuals discontinued treatment or died. Percentages of individuals with treatment-emergent adverse events were similar across dose and treatment groups. No serious or severe treatment-related treatment-emergent adverse events occurred. Trends for better motor function from baseline to week 25 were observed for 40 mg/kg every other week avalglucosidase alfa compared with either 20 mg/kg every other week avalglucosidase alfa or alglucosidase alfa up to 40 mg/kg weekly. CONCLUSION: These data support the positive clinical effect of avalglucosidase alfa in patients with infantile-onset Pompe disease previously declining on alglucosidase alfa.


Subject(s)
Glycogen Storage Disease Type II , Humans , Glycogen Storage Disease Type II/drug therapy , Cohort Studies , Treatment Outcome , alpha-Glucosidases/adverse effects , Research , Enzyme Replacement Therapy/adverse effects
3.
J Neuromuscul Dis ; 9(6): 713-730, 2022.
Article in English | MEDLINE | ID: mdl-36214004

ABSTRACT

BACKGROUND: ADVANCE (NCT01526785) presented an opportunity to obtain a more nuanced understanding of motor function changes in treatment-experienced children with Pompe disease receiving 4000L-production-scale alglucosidase alfa for 52 weeks. OBJECTIVE: To estimate minimal detectable change (MDC) and effect size on Gross Motor Function Measure-88 (GMFM-88) after 52 weeks of 4000L alglucosidase alfa (complete data N =  90). METHODS: The GMFM-88 mean total % score changes, MDC, and effect size were analyzed post hoc by Pompe Motor Function Level at enrollment, age groups at enrollment, and fraction of life on pre-study 160L-production-scale alglucosidase alfa. RESULTS: Overall, participants aged < 2 years surpassed MDC at Week 52 (change [mean±standard deviation] 21.1±14.1, MDC range 5.7-13.3, effect size 1.1), whereas participants aged≥2 years did not attain this (change -0.9±15.3, MDC range 10.8-25.2, effect size -0.03). In participants aged < 2 years, improvements surpassed the MDC for walkers (change 17.1±13.3, MDC range 3.0-6.9, effect size 1.7), supported standers (change 35.2±18.0, MDC range 5.9-13.7, effect size 1.8) and sitters (change 24.1±12.1, MDC range 2.6-6.2, effect size 2.7). Age-independent MDC ranges were only attained by walkers (change 7.7±12.3, MDC range 6.4-15.0, effect size 0.4) and sitters (change 9.9±17.2, MDC range 3.3-7.7, effect size 0.9). CONCLUSIONS: These first GMFM-88 minimal-detectable-change estimates for alglucosidase alfa-treated Pompe disease offer utility for monitoring motor skills. TRIAL REGISTRATION: ClinicalTrials.gov; NCT01526785; Registered 6 February 2012; https://clinicaltrials.gov/ct2/show/NCT01526785.


Subject(s)
Glycogen Storage Disease Type II , Child , Humans , Glycogen Storage Disease Type II/drug therapy , Enzyme Replacement Therapy , Cohort Studies , Motor Skills
4.
Cardiol Young ; 32(3): 364-373, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34420548

ABSTRACT

Pompe disease results from lysosomal acid α-glucosidase deficiency, which leads to cardiomyopathy in all infantile-onset and occasional late-onset patients. Cardiac assessment is important for its diagnosis and management. This article presents unpublished cardiac findings, concomitant medications, and cardiac efficacy and safety outcomes from the ADVANCE study; trajectories of patients with abnormal left ventricular mass z score at enrolment; and post hoc analyses of on-treatment left ventricular mass and systolic blood pressure z scores by disease phenotype, GAA genotype, and "fraction of life" (defined as the fraction of life on pre-study 160 L production-scale alglucosidase alfa). ADVANCE evaluated 52 weeks' treatment with 4000 L production-scale alglucosidase alfa in ≥1-year-old United States of America patients with Pompe disease previously receiving 160 L production-scale alglucosidase alfa. M-mode echocardiography and 12-lead electrocardiography were performed at enrolment and Week 52. Sixty-seven patients had complete left ventricular mass z scores, decreasing at Week 52 (infantile-onset patients, change -0.8 ± 1.83; 95% confidence interval -1.3 to -0.2; all patients, change -0.5 ± 1.71; 95% confidence interval -1.0 to -0.1). Patients with "fraction of life" <0.79 had left ventricular mass z score decreasing (enrolment: +0.1 ± 3.0; Week 52: -1.1 ± 2.0); those with "fraction of life" ≥0.79 remained stable (enrolment: -0.9 ± 1.5; Week 52: -0.9 ± 1.4). Systolic blood pressure z scores were stable from enrolment to Week 52, and no cohort developed systemic hypertension. Eight patients had Wolff-Parkinson-White syndrome. Cardiac hypertrophy and dysrhythmia in ADVANCE patients at or before enrolment were typical of Pompe disease. Four-thousand L alglucosidase alfa therapy maintained fractional shortening, left ventricular posterior and septal end-diastolic thicknesses, and improved left ventricular mass z score.Trial registry: ClinicalTrials.gov Identifier: NCT01526785 https://clinicaltrials.gov/ct2/show/NCT01526785.Social Media Statement: Post hoc analyses of the ADVANCE study cohort of 113 children support ongoing cardiac monitoring and concomitant management of children with Pompe disease on long-term alglucosidase alfa to functionally improve cardiomyopathy and/or dysrhythmia.


Subject(s)
Glycogen Storage Disease Type II , Cardiomegaly/drug therapy , Cardiomegaly/etiology , Cohort Studies , Enzyme Replacement Therapy/adverse effects , Enzyme Replacement Therapy/methods , Genotype , Glycogen Storage Disease Type II/complications , Glycogen Storage Disease Type II/diagnosis , Glycogen Storage Disease Type II/drug therapy , Humans , Phenotype
5.
Am J Hum Genet ; 108(5): 857-873, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33961779

ABSTRACT

The ALF transcription factor paralogs, AFF1, AFF2, AFF3, and AFF4, are components of the transcriptional super elongation complex that regulates expression of genes involved in neurogenesis and development. We describe an autosomal dominant disorder associated with de novo missense variants in the degron of AFF3, a nine amino acid sequence important for its binding to ubiquitin ligase, or with de novo deletions of this region. The sixteen affected individuals we identified, along with two previously reported individuals, present with a recognizable pattern of anomalies, which we named KINSSHIP syndrome (KI for horseshoe kidney, NS for Nievergelt/Savarirayan type of mesomelic dysplasia, S for seizures, H for hypertrichosis, I for intellectual disability, and P for pulmonary involvement), partially overlapping the AFF4-associated CHOPS syndrome. Whereas homozygous Aff3 knockout mice display skeletal anomalies, kidney defects, brain malformations, and neurological anomalies, knockin animals modeling one of the microdeletions and the most common of the missense variants identified in affected individuals presented with lower mesomelic limb deformities like KINSSHIP-affected individuals and early lethality, respectively. Overexpression of AFF3 in zebrafish resulted in body axis anomalies, providing some support for the pathological effect of increased amount of AFF3. The only partial phenotypic overlap of AFF3- and AFF4-associated syndromes and the previously published transcriptome analyses of ALF transcription factors suggest that these factors are not redundant and each contributes uniquely to proper development.


Subject(s)
Brain Diseases/genetics , Epilepsy/genetics , Fused Kidney/genetics , Intellectual Disability/genetics , Mutation, Missense , Nuclear Proteins/genetics , Osteochondrodysplasias/genetics , Adolescent , Amino Acid Sequence , Animals , Brain Diseases/etiology , Child , Child, Preschool , Epilepsy/complications , Evolution, Molecular , Female , Gene Frequency , Humans , Infant , Male , Mice , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/deficiency , Phenotype , Protein Stability , Syndrome , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/genetics , Young Adult , Zebrafish/genetics
6.
Neurosci Biobehav Rev ; 124: 267-290, 2021 05.
Article in English | MEDLINE | ID: mdl-33581222

ABSTRACT

For patients diagnosed with a rare musculoskeletal or neuromuscular disease, pain may transition from acute to chronic; the latter yielding additional challenges for both patients and care providers. We assessed the present understanding of pain across a set of ten rare, noninfectious, noncancerous disorders; Osteogenesis Imperfecta, Ehlers-Danlos Syndrome, Achondroplasia, Fibrodysplasia Ossificans Progressiva, Fibrous Dysplasia/McCune-Albright Syndrome, Complex Regional Pain Syndrome, Duchenne Muscular Dystrophy, Infantile- and Late-Onset Pompe disease, Charcot-Marie-Tooth Disease, and Amyotrophic Lateral Sclerosis. Through the integration of natural history, cross-sectional, retrospective, clinical trials, & case studies we described pathologic and genetic factors, pain sources, phenotypes, and lastly, existing therapeutic approaches. We highlight that while rare diseases possess distinct core pathologic features, there are a number of shared pain phenotypes and mechanisms that may be prospectively examined and therapeutically targeted in a parallel manner. Finally, we describe clinical and research approaches that may facilitate more accurate diagnosis, monitoring, and treatment of pain as well as elucidation of the evolving nature of pain phenotypes in rare musculoskeletal or neuromuscular illnesses.


Subject(s)
Neuromuscular Diseases , Cross-Sectional Studies , Humans , Neuromuscular Diseases/complications , Neuromuscular Diseases/genetics , Pain , Phenotype , Retrospective Studies
7.
Mol Genet Metab ; 131(1-2): 135-146, 2020.
Article in English | MEDLINE | ID: mdl-33342467

ABSTRACT

Phosphoglucomutase 1 deficiency is a congenital disorder of glycosylation (CDG) with multiorgan involvement affecting carbohydrate metabolism, N-glycosylation and energy production. The metabolic management consists of dietary D-galactose supplementation that ameliorates hypoglycemia, hepatic dysfunction, endocrine anomalies and growth delay. Previous studies suggest that D-galactose administration in juvenile patients leads to more significant and long-lasting effects, stressing the urge of neonatal diagnosis (0-6 months of age). Here, we detail the early clinical presentation of PGM1-CDG in eleven infantile patients, and applied the modified Beutler test for screening of PGM1-CDG in neonatal dried blood spots (DBSs). All eleven infants presented episodic hypoglycemia and elevated transaminases, along with cleft palate and growth delay (10/11), muscle involvement (8/11), neurologic involvement (5/11), cardiac defects (2/11). Standard dietary measures for suspected lactose intolerance in four patients prior to diagnosis led to worsening of hypoglycemia, hepatic failure and recurrent diarrhea, which resolved upon D-galactose supplementation. To investigate possible differences in early vs. late clinical presentation, we performed the first systematic literature review for PGM1-CDG, which highlighted respiratory and gastrointestinal symptoms as significantly more diagnosed in neonatal age. The modified Butler-test successfully identified PGM1-CDG in DBSs from seven patients, including for the first time Guthrie cards from newborn screening, confirming the possibility of future inclusion of PGM1-CDG in neonatal screening programs. In conclusion, severe infantile morbidity of PGM1-CDG due to delayed diagnosis could be prevented by raising awareness on its early presentation and by inclusion in newborn screening programs, enabling early treatments and galactose-based metabolic management.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Glycogen Storage Disease/blood , Hypoglycemia/genetics , Phosphoglucomutase/blood , Cleft Palate/blood , Cleft Palate/complications , Cleft Palate/genetics , Congenital Disorders of Glycosylation/blood , Congenital Disorders of Glycosylation/complications , Congenital Disorders of Glycosylation/enzymology , Dried Blood Spot Testing , Female , Glycogen Storage Disease/enzymology , Glycogen Storage Disease/genetics , Humans , Hypoglycemia/blood , Hypoglycemia/complications , Infant , Infant, Newborn , Male , Neonatal Screening , Phenotype , Phosphoglucomutase/genetics
8.
Neoreviews ; 20(5): e280-e291, 2019 05.
Article in English | MEDLINE | ID: mdl-31261080

ABSTRACT

Newborn screening (NBS) is the largest public health program in the United States, affecting every newborn. The purpose of newborn screening is to identify newborns at risk for selected disorders during the presymptomatic phase, with the hope that early intervention can prevent disease progression. NBS began in the early 1960s following the pioneering work of Robert Guthrie with phenylketonuria. Since then, NBS has expanded, with testing available for more than 50 disorders in most states. Screening tests need to be highly automated, with high sensitivity and specificity to avoid missing patients with disease, and ensuring manageable false-positive rates. Current initiatives in NBS include timeliness to ensure that results of the screen are available by 5 days after birth for a core set of critical conditions. This has resulted in the current recommendation for NBS specimens to be collected at 24 to 48 hours after birth. False-positive rates are higher in the NICU, because of the metabolic instability of sick neonates and the immaturity of premature enzyme systems. The recommended uniform screen panel (RUSP) contains the current list of disorders screened for by most states. Additional disorders continue to be added to the RUSP as medical progress allows previously untreatable disorders to be managed successfully, and thus the need to screen emerges. The costs associated with NBS continue to climb, because despite state-mandated screening, the diagnostic evaluation and treatment of these conditions has no such mandate. This is a particular concern for disorders with annual treatment costs of several hundred thousand dollars.


Subject(s)
Infant, Newborn, Diseases/diagnosis , Intensive Care Units, Neonatal/standards , Neonatal Screening/methods , Neonatal Screening/standards , Practice Guidelines as Topic/standards , Humans , Infant, Newborn , Infant, Newborn, Diseases/genetics , Infant, Newborn, Diseases/metabolism
9.
Genet Med ; 21(11): 2543-2551, 2019 11.
Article in English | MEDLINE | ID: mdl-31086307

ABSTRACT

PURPOSE: To characterize clinical characteristics and genotypes of patients in the ADVANCE study of 4000 L-scale alglucosidase alfa (NCT01526785), the largest prospective United States Pompe disease cohort to date. METHODS: Patients aged ≥1 year with confirmed Pompe disease previously receiving 160 L alglucosidase alfa were eligible. GAA genotypes were determined before/at enrollment. Baseline assessments included histories/physical exams, Gross Motor Function Measure-88 (GMFM-88), pulmonary function tests, and cardiac assessments. RESULTS: Of 113 enrollees (60 male/53 female) aged 1-18 years, 87 had infantile-onset Pompe disease (IOPD) and 26 late-onset (LOPD). One hundred eight enrollees with GAA genotypes had 215 pathogenic variants (220 including combinations): 118 missense (4 combinations), 23 splice, 35 nonsense, 34 insertions/deletions, 9 duplications (1 combination), 6 other; c.2560C>T (n = 23), c.-32-13T>G (n = 13), and c.525delT (n = 12) were most common. Four patients had previously unpublished variants, and 14/83 (17%) genotyped IOPD patients were cross-reactive immunological material-negative. All IOPD and 6/26 LOPD patients had cardiac involvement, all without c.-32-13T>G. Thirty-two (26 IOPD, 6 LOPD) were invasively ventilated. GMFM-88 total %scores (mean ± SD, median, range): overall 46.3 ± 33.0% (47.9%, 0.0-100.0%), IOPD 41.6 ± 31.64% (38.9%, 0.0-99.7%), LOPD: 61.8 ± 33.2 (70.9%, 0.0-100.0%). CONCLUSION: ADVANCE, a uniformly assessed cohort comprising most US children and adolescents with treated Pompe disease, expands understanding of the phenotype and observed variants in the United States.


Subject(s)
Glycogen Storage Disease Type II/epidemiology , Glycogen Storage Disease Type II/genetics , alpha-Glucosidases/genetics , Adolescent , Child , Child, Preschool , Cohort Studies , Enzyme Replacement Therapy/methods , Female , Genotype , Humans , Infant , Male , Phenotype , Prospective Studies , United States/epidemiology , alpha-Glucosidases/metabolism
10.
J Pediatr Genet ; 8(2): 63-68, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31061748

ABSTRACT

Unexplained childhood fracture(s) warrant consideration of physical abuse and osteogenesis imperfecta (OI). Genetic OI testing may identify "variants of unknown significance (VUS)." Interpretation of VUS in context of potential abuse may have protective, criminal, and medical impacts. This case series explores practices regarding clinicians' interpretation of VUS during child abuse evaluations. Variability was noted regarding factors considered for interpreting clinical significance. Based on these cases, recommendations for careful and thorough evaluation are detailed, including proposed use of a limited follow-up skeletal survey in 3 months, as a consideration to assess healing of prior fractures and to look for any additional injuries.

11.
Genet Med ; 21(4): 887-895, 2019 04.
Article in English | MEDLINE | ID: mdl-30214072

ABSTRACT

PURPOSE: To investigate immune tolerance induction with transient low-dose methotrexate (TLD-MTX) initiated with recombinant human acid α-glucosidase (rhGAA), in treatment-naïve cross-reactive immunologic material (CRIM)-positive infantile-onset Pompe disease (IOPD) patients. METHODS: Newly diagnosed IOPD patients received subcutaneous or oral 0.4 mg/kg TLD-MTX for 3 cycles (3 doses/cycle) with the first 3 rhGAA infusions. Anti-rhGAA IgG titers, classified as high-sustained (HSAT; ≥51,200, ≥2 times after 6 months), sustained intermediate (SIT; ≥12,800 and <51,200 within 12 months), or low (LT; ≤6400 within 12 months), were compared with those of 37 CRIM-positive IOPD historic comparators receiving rhGAA alone. RESULTS: Fourteen IOPD TLD-MTX recipients at the median age of 3.8 months (range, 0.7-13.5 months) had a median last titer of 150 (range, 0-51,200) at median rhGAA duration ~83 weeks (range, 36-122 weeks). One IOPD patient (7.1%) developed titers in the SIT range and one patient (7.1%) developed titers in the HSAT range. Twelve of the 14 patients (85.7%) that received TLD-MTX remained LT, versus 5/37 HSAT (peak 51,200-409,600), 7/37 SIT (12,800-51,000), and 23/37 LT (200-12,800) among comparators. CONCLUSION: Results of TLD-MTX coinitiated with rhGAA are encouraging and merit a larger longitudinal study.


Subject(s)
Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/immunology , Immune Tolerance/genetics , Methotrexate/administration & dosage , Age of Onset , Cross Reactions/immunology , Enzyme Replacement Therapy , Female , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , Humans , Infant , Infant, Newborn , Male , alpha-Glucosidases/administration & dosage , alpha-Glucosidases/genetics
12.
Genome Res ; 28(7): 1039-1052, 2018 07.
Article in English | MEDLINE | ID: mdl-29773658

ABSTRACT

Current approaches to detect and characterize mosaic chromosomal aneuploidy are limited by sensitivity, efficiency, cost, or the need to culture cells. We describe the mosaic aneuploidy detection by massively parallel sequencing (MAD-seq) capture assay and the MADSEQ analytical approach that allow low (<10%) levels of mosaicism for chromosomal aneuploidy or regional loss of heterozygosity to be detected, assigned to a meiotic or mitotic origin, and quantified as a proportion of the cells in the sample. We show results from a multi-ethnic MAD-seq (meMAD-seq) capture design that works equally well in populations of diverse racial and ethnic origins and how the MADSEQ analytical approach can be applied to exome or whole-genome sequencing data, revealing previously unrecognized aneuploidy or copy number neutral loss of heterozygosity in samples studied by the 1000 Genomes Project, cell lines from public repositories, and one of the Illumina Platinum Genomes samples. We have made the meMAD-seq capture design and MADSEQ analytical software open for unrestricted use, with the goal that they can be applied in clinical samples to allow new insights into the unrecognized prevalence of mosaic chromosomal aneuploidy in humans and its phenotypic associations.


Subject(s)
Chromosomes/genetics , High-Throughput Nucleotide Sequencing/methods , Aneuploidy , Exome/genetics , Female , Genome/genetics , Humans , Male , Mosaicism , Software
13.
Genet Med ; 20(10): 1284-1294, 2018 10.
Article in English | MEDLINE | ID: mdl-29565424

ABSTRACT

PURPOSE: Pompe disease results from lysosomal acid α-glucosidase (GAA) deficiency and its associated glycogen accumulation and muscle damage. Alglucosidase alfa (recombinant human GAA (rhGAA)) received approval in 2006 as a treatment for Pompe disease at the 160 L production scale. In 2010, larger-scale rhGAA was approved for patients up to 8 years old without cardiomyopathy. NCT01526785 evaluated 4,000 L rhGAA efficacy/safety in US infantile- or late-onset Pompe disease (IOPD, LOPD) patients up to 1 year old transitioned from 160 L rhGAA. METHODS: A total of 113 patients (87 with IOPD; 26 with LOPD) received 4,000 L rhGAA for 52 weeks dosed the same as previous 160 L rhGAA. Efficacy was calculated as the percentage of patients stable/improved at week 52 (without death, new requirement for invasive ventilation, left ventricular mass z-score increase >1 if baseline was >2, upright forced vital capacity decrease ≥15% predicted, or Gross Motor Function Measure-88 decrease ≥8 percentage points). Safety evaluation included an extension ≤20 months. RESULTS: Week 52 data was available for 104 patients, 100 of whom entered the extension. At week 52, 87/104 (83.7%) were stable/improved. Overall survival was 98.1% overall, 97.6% IOPD, 100% LOPD; 92.4% remained invasive ventilator-free (93.4% IOPD, 88.7% LOPD). Thirty-five patients had infusion-associated reactions. Eight IOPD patients died of drug-unrelated causes. CONCLUSIONS: Most Pompe disease patients were clinically stable/improved after transitioning to 4,000 L rhGAA. Safety profiles of both rhGAA forms were consistent.


Subject(s)
Glycogen Storage Disease Type II/drug therapy , Recombinant Proteins/administration & dosage , alpha-Glucosidases/administration & dosage , Age of Onset , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , Humans , Infant , Infant, Newborn , Kaplan-Meier Estimate , Male , Recombinant Proteins/adverse effects , alpha-Glucosidases/adverse effects
14.
Am J Hum Genet ; 102(1): 188-195, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29304374

ABSTRACT

Fucosyltransferase 8 (FUT8) encodes a Golgi-localized α1,6 fucosyltransferase that is essential for transferring the monosaccharide fucose into N-linked glycoproteins, a process known as "core fucosylation." Here we describe three unrelated individuals, who presented with intrauterine growth retardation, severe developmental and growth delays with shortened limbs, neurological impairments, and respiratory complications. Each underwent whole-exome sequencing and was found to carry pathogenic variants in FUT8. The first individual (consanguineous family) was homozygous for c.715C>T (p.Arg239∗), while the second (non-consanguineous family) was compound heterozygous for c.1009C>G (p.Arg337Gly) and a splice site variant c.1259+5G>T. The third individual (consanguineous family) was homozygous for a c.943C>T (p.Arg315∗). Splicing analysis confirmed the c.1259+5G>T resulted in expression of an abnormal FUT8 transcript lacking exon 9. Functional studies using primary fibroblasts from two affected individuals revealed a complete lack of FUT8 protein expression that ultimately resulted in substantial deficiencies in total core fucosylated N-glycans. Furthermore, serum samples from all three individuals showed a complete loss of core fucosylation. Here, we show that loss of function mutations in FUT8 cause a congenital disorder of glycosylation (FUT8-CDG) characterized by defective core fucosylation that phenotypically parallels some aspects of the Fut8-/- knockout mouse. Importantly, identification of additional affected individuals can be easily achieved through analysis of core fucosylation of N-glycans.


Subject(s)
Alleles , Fucose/genetics , Fucosyltransferases/genetics , Mutation/genetics , Alternative Splicing/genetics , Cells, Cultured , Child , Child, Preschool , Fatal Outcome , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Glycosylation , Humans , Lectins/metabolism , Male , Polysaccharides/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Pediatrics ; 140(Suppl 1): S14-S23, 2017 Jul.
Article in English | MEDLINE | ID: mdl-29162674

ABSTRACT

Newborn screening (NBS) for Pompe disease is done through analysis of acid α-glucosidase (GAA) activity in dried blood spots. When GAA levels are below established cutoff values, then second-tier testing is required to confirm or refute a diagnosis of Pompe disease. This article in the "Newborn Screening, Diagnosis, and Treatment for Pompe Disease" guidance supplement provides recommendations for confirmatory testing after a positive NBS result indicative of Pompe disease is obtained. Two algorithms were developed by the Pompe Disease Newborn Screening Working Group, a group of international experts on both NBS and Pompe disease, based on whether DNA sequencing is performed as part of the screening method. Using the recommendations in either algorithm will lead to 1 of 3 diagnoses: classic infantile-onset Pompe disease, late-onset Pompe disease, or no disease/not affected/carrier. Mutation analysis of the GAA gene is essential for confirming the biochemical diagnosis of Pompe disease. For NBS laboratories that do not have DNA sequencing capabilities, the responsibility of obtaining sequencing of the GAA gene will fall on the referral center. The recommendations for confirmatory testing and the initial evaluation are intended for a broad global audience. However, the Working Group recognizes that clinical practices, standards of care, and resource capabilities vary not only regionally, but also by testing centers. Individual patient needs and health status as well as local/regional insurance reimbursement programs and regulations also must be considered.


Subject(s)
Glycogen Storage Disease Type II/diagnosis , Neonatal Screening/methods , Algorithms , Humans , Infant, Newborn
16.
Pediatrics ; 140(Suppl 1): S24-S45, 2017 Jul.
Article in English | MEDLINE | ID: mdl-29162675

ABSTRACT

After a Pompe disease diagnosis is confirmed in infants identified through newborn screening (NBS), when and if to start treatment with enzyme replacement therapy (ERT) with alglucosidase alfa must be determined. In classic infantile-onset Pompe disease, ERT should start as soon as possible. Once started, regular, routine follow-up is necessary to monitor for treatment effects, disease progression, and adverse effects. Decision-making for when or if to start ERT in late-onset Pompe disease (LOPD) is more challenging because patients typically have no measurable signs or symptoms or predictable time of symptom onset at NBS. With LOPD, adequate, ongoing follow-up and assessments for onset or progression of signs and symptoms are important to track disease state and monitor and adjust care before and after treatment is started. Because numerous tests are used to monitor patients at variable frequencies, a standardized approach across centers is lacking. Significant variability in patient assessments may result in missed opportunities for early intervention. Management of Pompe disease requires a comprehensive, multidisciplinary approach with timely disease-specific interventions that target the underlying disease process and symptom-specific manifestations. Regardless of how identified, all patients who have signs or symptoms of the disease require coordinated medical care and follow-up tailored to individual needs throughout their lives. The Pompe Disease Newborn Screening Working Group identifies key considerations before starting and during ERT; summarizes what comprises an indication to start ERT; and provides guidance on how to determine appropriate patient management and monitoring and guide the frequency and type of follow-up assessments for all patients identified through NBS.


Subject(s)
Enzyme Replacement Therapy/methods , Glycogen Storage Disease Type II/diagnosis , Neonatal Screening/methods , Disease Progression , Glycogen Storage Disease Type II/therapy , Humans , Infant, Newborn
17.
JCI Insight ; 2(16)2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28814660

ABSTRACT

BACKGROUND: Cross-reactive immunological material-negative (CRIM-negative) infantile Pompe disease (IPD) patients develop an immune response against enzyme replacement therapy (ERT) with alglucosidase alfa that nullifies ERT efficacy. Prophylactic immune tolerance induction (ITI) with rituximab, methotrexate, and IVIG successfully prevents development of deleterious rhGAA IgG antibodies; however, safety, likelihood of success, and long-term efficacy of ITI in a larger cohort remain unknown. METHODS: Clinical data were analyzed for 19 CRIM-negative IPD patients who received ITI with rituximab, methotrexate, and IVIG in the ERT-naive setting (ERT+ITI) and compared to a historical cohort of 10 CRIM-negative IPD patients on ERT monotherapy. RESULTS: ITI was safely tolerated, although infections were reported in 4 patients. Fourteen (74%) ERT+ITI patients were alive, with a median age of 44.2 months at their final assessment. The eldest survivor was 103.9 months old, with 100.2 months of follow-up after initiation of ERT+ITI. Death (n = 5) occurred at a median age of 29.2 months and was unrelated to the administration of ITI. Fifteen patients either did not seroconvert (n = 8) or maintained low titers (n = 7; defined as titers of ≤6,400 throughout the course of ERT) following ERT+ITI. Only one patient developed high and sustained antibody titers (defined as titers of ≥51,200 at or beyond 6 months on ERT). Left ventricular mass index (LVMI) decreased from a median of 248.5 g/m2 at baseline to 76.8 g/m2 at a median time from ERT+ITI initiation to 59 weeks. ERT+ITI significantly improved overall survival (P = 0.001), eliminated/reduced antibodies at values of ≤6,400 at week 52 on ERT (P = 0.0004), and improved LVMI at week 52 on ERT (P = 0.02) when compared with ERT monotherapy. CONCLUSION: Evidence from this international cohort of CRIM-negative IPD patients further supports the safety, feasibility, and efficacy of ITI in the prevention of immune responses to ERT. TRIAL REGISTRATION: Clinicaltrials.gov NCT01665326. FUNDING: This research was supported in part by the Lysosomal Disease Network, a part of NIH Rare Diseases Clinical Research Network, and by a grant from Genzyme, a Sanofi company.

18.
Genet Med ; 18(12): 1235-1243, 2016 12.
Article in English | MEDLINE | ID: mdl-27171547

ABSTRACT

BACKGROUND: Early infantile Krabbe disease is rapidly fatal, but hematopoietic stem cell transplantation (HSCT) may improve outcomes if performed soon after birth. New York State began screening all newborns for Krabbe disease in 2006. METHODS: Infants with abnormal newborn screen results for Krabbe disease were referred to specialty-care centers. Newborns found to be at high risk for Krabbe disease underwent a neurodiagnostic battery to determine the need for emergent HSCT. RESULTS: Almost 2 million infants were screened. Five infants were diagnosed with early infantile Krabbe disease. Three died, two from HSCT-related complications and one from untreated disease. Two children who received HSCT have moderate to severe developmental delays. Forty-six currently asymptomatic children are considered to be at moderate or high risk for development of later-onset Krabbe disease. CONCLUSIONS: These results show significant HSCT-associated morbidity and mortality in early infantile Krabbe disease and raise questions about its efficacy when performed in newborns diagnosed through newborn screening. The unanticipated identification of "at risk" children introduces unique ethical and medicolegal issues. New York's experience raises questions about the risks, benefits, and practicality of screening newborns for Krabbe disease. It is imperative that objective assessments be made on an ongoing basis as additional states begin screening for this disorder.Genet Med 18 12, 1235-1243.


Subject(s)
Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/therapy , Mass Screening , Neonatal Screening , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infant , Infant, Newborn , Leukodystrophy, Globoid Cell/diagnosis , Leukodystrophy, Globoid Cell/mortality , New York , Risk Factors
19.
Genet Med ; 18(3): 239-48, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26795590

ABSTRACT

PURPOSE: Krabbe disease (KD) results from galactocerebrosidase (GALC) deficiency. Infantile KD symptoms include irritability, progressive stiffness, developmental delay, and death. The only potential treatment is hematopoietic stem cell transplantation. New York State (NYS) implemented newborn screening for KD in 2006. METHODS: Dried blood spots from newborns were assayed for GALC enzyme activity using mass spectrometry, followed by molecular analysis for those with low activity (≤12% of the daily mean). Infants with low enzyme activity and one or more mutations were referred for follow-up diagnostic testing and neurological examination. RESULTS: Of >1.9 million screened, 620 infants were subjected to molecular analysis and 348 were referred for diagnostic testing. Five had enzyme activities and mutations consistent with infantile KD and manifested clinical/neurodiagnostic abnormalities. Four underwent transplantation, two are surviving with moderate to severe handicaps, and two died from transplant-related complications. The significance of many sequence variants identified is unknown. Forty-six asymptomatic infants were found to be at moderate to high risk for disease. CONCLUSIONS: The positive predictive value of KD screening in NYS is 1.4% (5/346) considering confirmed infantile cases. The incidence of infantile KD in NYS is approximately 1 in 394,000, but it may be higher for later-onset forms.


Subject(s)
Galactosylceramidase/genetics , Galactosylceramidase/metabolism , Leukodystrophy, Globoid Cell/diagnosis , Neonatal Screening/methods , Polymorphism, Single Nucleotide , Algorithms , Dried Blood Spot Testing , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infant, Newborn , Leukodystrophy, Globoid Cell/enzymology , Leukodystrophy, Globoid Cell/therapy , Mass Spectrometry , New York , Predictive Value of Tests , Treatment Outcome
20.
Mol Genet Metab ; 116(1-2): 29-34, 2015.
Article in English | MEDLINE | ID: mdl-26296711

ABSTRACT

BACKGROUND: Health care outcomes have been increasingly assessed through health-related quality of life (HRQoL) measures. While the introduction of nitrogen-scavenging medications has improved survival in patients with urea cycle disorders (UCDs), they are often associated with side effects that may affect patient compliance and outcomes. METHODS: Symptoms commonly associated with nitrogen-scavenging medications were evaluated in 100 adult and pediatric participants using a non-validated UCD-specific questionnaire. Patients or their caregivers responded to a pre-defined list of symptoms known to be associated with the use of these medications. Responses were collected at baseline (while patients were receiving sodium phenylbutyrate [NaPBA]) and during treatment with glycerol phenylbutyrate (GPB). RESULTS: After 3 months of GPB dosing, there were significant reductions in the proportion of patients with treatment-associated symptoms (69% vs. 46%; p<0.0001), the number of symptoms per patient (2.5 vs. 1.1; p<0.0001), and frequency of the more commonly reported individual symptoms such as body odor, abdominal pain, nausea, burning sensation in mouth, vomiting, and heartburn (p<0.05). The reduction in symptoms was observed in both pediatric and adult patients. The presence or absence of symptoms or change in severity did not correlate with plasma ammonia levels or NaPBA dose. CONCLUSIONS: The reduction in symptoms following 3 months of open-label GPB dosing was similar in pediatric and adult patients and may be related to chemical structure and intrinsic characteristics of the product rather than its effect on ammonia control.


Subject(s)
Glycerol/analogs & derivatives , Phenylbutyrates/adverse effects , Quality of Life , Self Report , Urea Cycle Disorders, Inborn/drug therapy , Adolescent , Adult , Aged , Ammonia/blood , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , Female , Glycerol/adverse effects , Glycerol/chemistry , Glycerol/therapeutic use , Humans , Infant , Male , Middle Aged , Phenylbutyrates/chemistry , Phenylbutyrates/therapeutic use , Surveys and Questionnaires , Urea Cycle Disorders, Inborn/blood , Urea Cycle Disorders, Inborn/psychology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...