1
|
Sunaga Y, Sakashita T, Koga T, Sawada T, Yamane S, Ikeda M. [Importance of early treatment and quantitative evaluation of enzyme replacement therapy for Pompe disease: alglucosidase alfa post-marketing surveillance additional analysis]. Rinsho Shinkeigaku 2024; 64:866-877. [PMID: 39566964 DOI: 10.5692/clinicalneurol.cn-001894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
We conducted an additional analysis using the data from the post-marketing surveillance of Alglucosidase alfa for Pompe disease. We aimed to investigate the changes in the percentage of predicted forced vital capacity (%FVC) and the changes in the distance of the 6-min walk test (6MWT) by overall improvement and to investigate the %FVC change by the duration from symptom onset to survey registration (shorter/longer groups) using a linear mixed model. Thirty-seven and eighteen survey participants had %FVC and 6MWT data available, respectively; of the patients whose overall improvement was rated as "relatively improved," %FVC and 6MWT worsened in 71.4% and 66.7%, respectively. The %FVC at the survey registration estimated using a linear mixed model was significantly higher in the shorter group than in the longer group (P = 0.0413). The estimated slope of %FVC was significantly lower in the shorter group than in the longer group (P = 0.0051). These results suggest the importance of early treatment initiation and quantitative evaluation of each symptom.
Collapse
Affiliation(s)
- Yoshinori Sunaga
- Medical affairs, Real world evidence generation partnering, Sanofi K.K
| | | | | | | | - Shiho Yamane
- Medical Affairs, Post-Authorization Regulatory Studies, Sanofi K.K
| | | |
Collapse
|
2
|
Parenti G, Fecarotta S, Alagia M, Attaianese F, Verde A, Tarallo A, Gragnaniello V, Ziagaki A, Guimaraes MJ, Aguiar P, Hahn A, Azevedo O, Donati MA, Kiec-Wilk B, Scarpa M, van der Beek NAME, Del Toro Riera M, Germain DP, Huidekoper H, van den Hout JMP, van der Ploeg AT. The European reference network for metabolic diseases (MetabERN) clinical pathway recommendations for Pompe disease (acid maltase deficiency, glycogen storage disease type II). Orphanet J Rare Dis 2024; 19:408. [PMID: 39482698 PMCID: PMC11529438 DOI: 10.1186/s13023-024-03373-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 09/18/2024] [Indexed: 11/03/2024] Open
Abstract
Clinical pathway recommendations (CPR) are based on existing guidelines and deliver a short overview on how to deal with a specific diagnosis, resulting therapy and follow-up. In this paper we propose a methodology for developing CPRs for Pompe disease, a metabolic myopathy caused by deficiency of lysosomal acid alpha-glucosidase. The CPR document was developed within the activities of the MetabERN, a non-profit European Reference Network for Metabolic Diseases established by the European Union. A working group was selected among members of the MetabERN lysosomal storage disease subnetwork, with specific expertise in the care of Pompe disease, and patient support group representatives. The working strategy was based on a systematic literature search to develop a database, followed by quality assessment of the studies selected from the literature, and by the development of the CPR document according to a matrix provided by MetabERN. Quality assessment of the literature and collection of citations was conducted according to the AGREE II criteria and Grading of Recommendations, Assessment, Development and Evaluation methodology. General aspects were addressed in the document, including pathophysiology, genetics, frequency, classification, manifestations and clinical approach, laboratory diagnosis and multidisciplinary evaluation, therapy and supportive measures, follow-up, monitoring, and pregnancy. The CPR document that was developed was intended to be a concise and easy-to-use tool for standardization of care for patients among the healthcare providers that are members of the network or are involved in the care for Pompe disease patients.
Collapse
Affiliation(s)
- Giancarlo Parenti
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands.
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, Naples, Italy.
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples, Italy.
- Azienda Ospedaliera Universitaria Federico II, Naples, Italy.
| | - Simona Fecarotta
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples, Italy
- Azienda Ospedaliera Universitaria Federico II, Naples, Italy
| | - Marianna Alagia
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples, Italy
- Azienda Ospedaliera Universitaria Federico II, Naples, Italy
| | - Federica Attaianese
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples, Italy
| | - Alessandra Verde
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples, Italy
- Azienda Ospedaliera Universitaria Federico II, Naples, Italy
| | - Antonietta Tarallo
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples, Italy
| | - Vincenza Gragnaniello
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples, Italy
| | - Athanasia Ziagaki
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Department of Endocrinology and Metabolism, Center of Excellence for Rare Metabolic Diseases in Adults, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Jose' Guimaraes
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Pneumology Department, Reference Center on Lysosomal Storage Disorders, Hospital Senhora da Oliveira, Guimarães, Portugal
| | - Patricio Aguiar
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Clinica Universitaria de Medicina I, Universidade de Lisboa, Lisbon, Portugal
| | - Andreas Hahn
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Department of Child Neurology, Justus-Liebig University, Giessen, Germany
| | - Olga Azevedo
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Cardiology Department, Reference Center on Lysosomal Storage Disorders, Hospital Senhora da Oliveira, Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3Bs PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Maria Alice Donati
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Metabolic and Neuromuscular Unit, Meyer Children Hospital-University of Florence, Florence, Italy
| | - Beata Kiec-Wilk
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Unit of Rare Metabolic Diseases, Jagiellonian University Medical College, Kraków, Poland
- The John Paul II Specjalist Hospital in Kraków, Kraków, Poland
| | - Maurizio Scarpa
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Centro Coordinamento Regionale Malattie Rare, Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | - Nadine A M E van der Beek
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Mireja Del Toro Riera
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Metabolic Unit, Department of Pediatric Neurology, Hospital Universitario Vall d'Hebron Barcelona, Barcelona, Spain
| | - Dominique P Germain
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Division of Medical Genetics, University of Versailles, Montigny, France
| | - Hidde Huidekoper
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Johanna M P van den Hout
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ans T van der Ploeg
- MetabERN Subnetwork for Lysosomal Disorders, Rotterdam, The Netherlands.
- Center for Lysosomal and Metabolic Diseases, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
3
|
Nunes Campos L, Davila Rivera I, Ibañez Alegre DM, Del Puerto González FN, Garrido San Juan M, Fernandez Zelcer F, Borgobello D, Gerk A, Sosa LF, Miretti MM, Stegmann J, Argüelles CF. Navigating Pompe Disease Assessment: A Comprehensive Scoping Review. Cureus 2024; 16:e73593. [PMID: 39677172 PMCID: PMC11645167 DOI: 10.7759/cureus.73593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2024] [Indexed: 12/17/2024] Open
Abstract
Pompe disease (PD) is a rare progressive autosomal recessive disorder resulting from the deficiency of acid alpha-glucosidase (GAA) enzyme activity. Due to its multisystemic involvement, PD leads to significant morbidity and impacts patients' quality of life. Despite the availability of approved disease-modifying treatments, the prompt diagnosis and management of PD, which are crucial for patient outcomes, still present several challenges. This scoping review aimed to synthesize the evidence regarding methods for screening, diagnosing, and following up PD. We searched articles in English and Spanish published from 2017 to February 8, 2022, across 11 databases (i.e., Cochrane Database of Systematic Reviews, Directory of Open Access Journals (DOAJ), Epistemonikos, Ingenta Connect, Medline/PubMed, SAGE, SciELO Citation Index, ScienceDirect, Springer Link, Virtual Health Library, and Wiley Online Library). We included primary studies (i.e., case reports, case series, cross-sectional studies, case controls, cohorts, clinical trials, and qualitative studies), reviews, and guidelines that described at least one assessment method for patients with confirmed clinical, genetic, or biochemical PD. Two independent reviewers screened and extracted data from articles, with a third reviewer solving conflicts. We synthesized data with narrative summaries and descriptive statistics. After screening 2,139 titles and abstracts, we included 96 eligible articles. Cross-sectional studies (n = 30) and guidelines (n = 1) were the most and least prevalent designs, respectively. Most studies targeted late-onset PD (LOPD, n = 48) and infantile-onset PD (IOPD, n = 21). Eleven articles described newborn screening programs, highlighting their potential to improve PD prevalence estimations and still limited availability among countries. Overall, 81 articles documented clinical manifestations of PD. Hypotonia (n = 7) and hypertrophic cardiomyopathy (n = 7) were the most documented for IOPD, while progressive muscle weakness (n = 21) and dyspnea (n = 11) were the most prevalent for LOPD. We found 26 articles reporting biochemical assays, with dried blood spots (DBS) for GAA enzyme deficiency detection being the most cited (n = 19). We also noted a lack of standardization in documenting DBS results. Additionally, 21 articles mentioned genetic studies, with next-generation sequencing emerging as the gold standard for identifying mutated alleles. Functional studies were the most utilized to follow up with patients. However, monitoring strategies for pediatric and adult PD lacked consensus, and only one article assessed patients' quality of life. This review comprehensively evaluated the literature on PD screening, diagnosis, and follow-up methods, identifying prevalent techniques within each assessment category. We emphasized the need for a more standardized approach to reporting biochemical assays, genetic testing, and clinical presentations. Our review also underscored the critical lack of standardization in PD follow-up. Addressing these gaps will enhance the comparability of future research findings and improve the quality of PD-related healthcare. Limitations of this review included restricting eligible languages and publication years to the latest five, the methodological heterogeneity of selected articles, and the lack of individual study bias assessment.
Collapse
Affiliation(s)
| | | | | | - Fabiana N Del Puerto González
- Rare Diseases, Rare Diseases Community (RDCom), Buenos Aires, ARG
- Genetics, GIGA, Instituto de Biología Subtropical, Nodo Posadas, Universidad Nacional de Misiones (UNaM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Posadas, ARG
| | | | | | | | - Ayla Gerk
- Rare Diseases, Rare Diseases Community (RDCom), Buenos Aires, ARG
| | - Laura F Sosa
- Rare Diseases, Rare Diseases Community (RDCom), Buenos Aires, ARG
| | - Marcos M Miretti
- Genetics, GIGA, Instituto de Biología Subtropical, Nodo Posadas, Universidad Nacional de Misiones (UNaM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Posadas, ARG
- Rare Diseases, Rare Diseases Community (RDCom), Buenos Aires, ARG
| | | | - Carina F Argüelles
- Rare Diseases, Rare Diseases Community (RDCom), Buenos Aires, ARG
- Faculty of Health Sciences, Universidad Católica de las Misiones, Posadas, ARG
- Genetics, GIGA, Instituto de Biología Subtropical, Nodo Posadas, Universidad Nacional de Misiones (UNaM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Posadas, ARG
| |
Collapse
|
4
|
Colella P. Advances in Pompe Disease Treatment: From Enzyme Replacement to Gene Therapy. Mol Diagn Ther 2024; 28:703-719. [PMID: 39134822 DOI: 10.1007/s40291-024-00733-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 10/27/2024]
Abstract
Pompe disease is a neuromuscular disorder caused by a deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA), hydrolyzing glycogen to glucose. Pathological glycogen storage, the hallmark of the disease, disrupts the metabolism and function of various cell types, especially muscle cells, leading to cardiac, motor, and respiratory dysfunctions. The spectrum of Pompe disease manifestations spans two main forms: classical infantile-onset (IOPD) and late-onset (LOPD). IOPD, caused by almost complete GAA deficiency, presents at birth and leads to premature death by the age of 2 years without treatment. LOPD, less severe due to partial GAA activity, appears in childhood, adolescence, or adulthood with muscle weakness and respiratory problems. Since 2006, enzyme replacement therapy (ERT) has been approved for Pompe disease, offering clinical benefits but not a cure. However, advances in early diagnosis through newborn screening, recognizing disease manifestations, and developing improved treatments are set to enhance Pompe disease care. This article reviews recent progress in ERT and ongoing translational research, including the approval of second-generation ERTs, a clinical trial of in utero ERT, and preclinical development of gene and substrate reduction therapies. Notably, gene therapy using intravenous delivery of adeno-associated virus (AAV) vectors is in phase I/II clinical trials for both LOPD and IOPD. Promising data from LOPD trials indicate that most participants met the criteria to discontinue ERT several months after gene therapy. The advantages and challenges of this approach are discussed. Overall, significant progress is being made towards curative therapies for Pompe disease. While several challenges remain, emerging data are promising and suggest the potential for a once-in-a-lifetime treatment.
Collapse
Affiliation(s)
- Pasqualina Colella
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
5
|
Tan L, Zschüntzsch J, Meyer S, Stobbe A, Bruex H, Regensburger AP, Claßen M, Alves F, Jüngert J, Rother U, Li Y, Danko V, Lang W, Türk M, Schmidt S, Vorgerd M, Schlaffke L, Woelfle J, Hahn A, Mensch A, Winterholler M, Trollmann R, Heiß R, Wagner AL, Raming R, Knieling F. Non-invasive optoacoustic imaging of glycogen-storage and muscle degeneration in late-onset Pompe disease. Nat Commun 2024; 15:7843. [PMID: 39245687 PMCID: PMC11381542 DOI: 10.1038/s41467-024-52143-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 08/26/2024] [Indexed: 09/10/2024] Open
Abstract
Pompe disease (PD) is a rare autosomal recessive glycogen storage disorder that causes proximal muscle weakness and loss of respiratory function. While enzyme replacement therapy (ERT) is the only effective treatment, biomarkers for disease monitoring are scarce. Following ex vivo biomarker validation in phantom studies, we apply multispectral optoacoustic tomography (MSOT), a laser- and ultrasound-based non-invasive imaging approach, in a clinical trial (NCT05083806) to image the biceps muscles of 10 late-onset PD (LOPD) patients and 10 matched healthy controls. MSOT is compared with muscle magnetic resonance imaging (MRI), ultrasound, spirometry, muscle testing and quality of life scores. Next, results are validated in an independent LOPD patient cohort from a second clinical site. Our study demonstrates that MSOT enables imaging of subcellular disease pathology with increases in glycogen/water, collagen and lipid signals, providing higher sensitivity in detecting muscle degeneration than current methods. This translational approach suggests implementation in the complex care of these rare disease patients.
Collapse
Affiliation(s)
- Lina Tan
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
- Translational Pediatrics, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Jana Zschüntzsch
- Neuromuscular Disease Research, Clinic for Neurology, University Medical Center Göttingen (UMG), Göttingen, 37075, Germany
| | - Stefanie Meyer
- Neuromuscular Disease Research, Clinic for Neurology, University Medical Center Göttingen (UMG), Göttingen, 37075, Germany
| | - Alica Stobbe
- Neuromuscular Disease Research, Clinic for Neurology, University Medical Center Göttingen (UMG), Göttingen, 37075, Germany
| | - Hannah Bruex
- Neuromuscular Disease Research, Clinic for Neurology, University Medical Center Göttingen (UMG), Göttingen, 37075, Germany
| | - Adrian P Regensburger
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
- Translational Pediatrics, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Merle Claßen
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
- Translational Pediatrics, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Frauke Alves
- Translational Molecular Imaging, Max-Planck Institute for Multidisciplinary Sciences (MPI-NAT), City Campus, Göttingen, 37075, Germany
- Clinic for Haematology and Medical Oncology, Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen (UMG), Göttingen, 37075, Germany
| | - Jörg Jüngert
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Ulrich Rother
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Yi Li
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Vera Danko
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
- Translational Pediatrics, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Werner Lang
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Matthias Türk
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Sandy Schmidt
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Matthias Vorgerd
- Department of Neurology, BG-University Hospital Bergmannsheil, Ruhr-University Bochum, 44789, Bochum, Germany
- Heimer Institute for Muscle Research, BG-University Hospital Bergmannsheil, 44789, Bochum, Germany
| | - Lara Schlaffke
- Department of Neurology, BG-University Hospital Bergmannsheil, Ruhr-University Bochum, 44789, Bochum, Germany
| | - Joachim Woelfle
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Andreas Hahn
- Department of Child Neurology, Justus-Liebig-Universität Giessen, 35385, Giessen, Germany
| | - Alexander Mensch
- Department of Neurology, Martin-Luther-Universität Halle-Wittenberg, 06120, Halle (Saale), Germany
| | | | - Regina Trollmann
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
- Center for Social Pediatrics, University Hospital Erlangen: Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Rafael Heiß
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Alexandra L Wagner
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
- Translational Pediatrics, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
- Department of Pediatric Neurology, Center for Chronically Sick Children, Charité Berlin, 13353, Berlin, Germany
| | - Roman Raming
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
- Translational Pediatrics, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Ferdinand Knieling
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany.
- Translational Pediatrics, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, 91054, Germany.
| |
Collapse
|
6
|
Kishnani PS, Chien YH, Berger KI, Thibault N, Sparks S. Clinical insight meets scientific innovation to develop a next generation ERT for Pompe disease. Mol Genet Metab 2024; 143:108559. [PMID: 39154400 DOI: 10.1016/j.ymgme.2024.108559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/20/2024]
Abstract
Years of research into the structure, processing, and function of acid alpha-glucosidase led to the development and 2006 approval of alglucosidase alfa (recombinant human acid alpha-glucosidase, Myozyme®/Lumizyme®), an enzyme replacement therapy and the first approved treatment for Pompe disease. Alglucosidase alfa has been a lifesaving treatment for patients with infantile-onset Pompe disease and radically improved daily life for patients with late-onset Pompe disease; however, long-term experience with alglucosidase alfa unraveled key unmet needs in these populations. Despite treatment, Pompe disease continues to progress, especially from a skeletal muscle perspective, resulting in a multitude of functional limitations. Strong collaboration between the scientific and patient communities led to increased awareness of Pompe disease, a better understanding of disease pathophysiology, knowledge of the clinical course of the disease as patients surpassed the first decade of life, and the strengths and limitations of enzyme replacement therapy. Taken together, these advancements spurred the need for development of a next generation of enzyme replacement therapy and provided a framework for progress toward other novel treatments. This review provides an overview of the development of avalglucosidase alfa as a model to highlight the interaction between clinical experience with existing treatments, the role of the clinician scientist, translational research at both system and cellular levels, and the iterative and collaborative process that optimizes the development of therapeutics.
Collapse
Affiliation(s)
- Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| | - Yin-Hsiu Chien
- Department of Medical Genetics and Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | |
Collapse
|
7
|
Hirachan R, Horman A, Burke D, Heales S. Evaluation, in a highly specialised enzyme laboratory, of a digital microfluidics platform for rapid assessment of lysosomal enzyme activity in dried blood spots. JIMD Rep 2024; 65:124-131. [PMID: 38444576 PMCID: PMC10910220 DOI: 10.1002/jmd2.12413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
Lysosomal storage disorders (LSDs) are predominantly enzyme deficiencies leading to substrate accumulation, causing progressive damage to multiple organs. To date, a crucial part of diagnosing LSDs is measuring enzymatic activity in leucocytes, plasma, or dried blood spots (DBS). Here, we present results from a proof-of-principle study, evaluating an innovative digital microfluidics (DMF) platform, referred to as SEEKER®, that can measure the activity of the following four lysosomal enzymes from DBS: α-L-iduronidase (IDUA) for mucopolysaccharidosis I (MPS I), acid α-glucosidase (GAA) for Pompe disease, β-glucosidase (GBA) for Gaucher disease, and α-galactosidase A (GLA) for Fabry disease. Over 900 DBS were analysed from newborns, children, and adults. DMF successfully detected known patients with MPS I, Pompe disease, and Gaucher disease, and known males with Fabry disease. This is the first demonstration of this multiplexed DMF platform for identification of patients with LSDs in a specialised diagnostic enzyme laboratory environment. We conclude that this DMF platform is relatively simple, high-throughput, and could be readily accommodated into a specialised laboratory as a first-tier test for MPS I, Pompe disease, and Gaucher disease for all patients, and Fabry disease for male patients only.
Collapse
Affiliation(s)
- Rohit Hirachan
- Chemical PathologyCamelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Alistair Horman
- Chemical PathologyCamelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Derek Burke
- Chemical PathologyCamelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Simon Heales
- Chemical PathologyCamelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Foundation TrustLondonUK
- Neurometabolic UnitNational Hospital for Neurology and NeurosurgeryLondonUK
| |
Collapse
|
8
|
Mackels L, Servais L. The Importance of Early Treatment of Inherited Neuromuscular Conditions. J Neuromuscul Dis 2024; 11:253-274. [PMID: 38306060 DOI: 10.3233/jnd-230189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
There has been tremendous progress in treatment of neuromuscular diseases over the last 20 years, which has transformed the natural history of these severely debilitating conditions. Although the factors that determine the response to therapy are many and in some instance remain to be fully elucidated, early treatment clearly has a major impact on patient outcomes across a number of inherited neuromuscular conditions. To improve patient care and outcomes, clinicians should be aware of neuromuscular conditions that require prompt treatment initiation. This review describes data that underscore the importance of early treatment of children with inherited neuromuscular conditions with an emphasis on data resulting from newborn screening efforts.
Collapse
Affiliation(s)
- Laurane Mackels
- MDUK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Adult Neurology Department, Citadelle Hospital, Liège, Belgium
| | - Laurent Servais
- Neuromuscular Centre, Division of Paediatrics, University and University Hospital of Liège, Liège, Belgium
- MDUK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford & NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
9
|
Gragnaniello V, Cazzorla C, Gueraldi D, Puma A, Loro C, Porcù E, Stornaiuolo M, Miglioranza P, Salviati L, Burlina AP, Burlina AB. Light and Shadows in Newborn Screening for Lysosomal Storage Disorders: Eight Years of Experience in Northeast Italy. Int J Neonatal Screen 2023; 10:3. [PMID: 38248631 PMCID: PMC10801488 DOI: 10.3390/ijns10010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
In the last two decades, the development of high-throughput diagnostic methods and the availability of effective treatments have increased the interest in newborn screening for lysosomal storage disorders. However, long-term follow-up experience is needed to clearly identify risks, benefits and challenges. We report our 8-year experience of screening and follow-up on about 250,000 neonates screened for four lysosomal storage diseases (Pompe disease, mucopolysaccharidosis type I, Fabry disease, Gaucher disease), using the enzyme activity assay by tandem mass spectrometry, and biomarker quantification as a second-tier test. Among the 126 positive newborns (0.051%), 51 infants were confirmed as affected (positive predictive value 40%), with an overall incidence of 1:4874. Of these, three patients with infantile-onset Pompe disease, two with neonatal-onset Gaucher disease and four with mucopolysaccharidosis type I were immediately treated. Furthermore, another four Gaucher disease patients needed treatment in the first years of life. Our study demonstrates the feasibility and effectiveness of newborn screening for lysosomal storage diseases. Early diagnosis and treatment allow the achievement of better patient outcomes. Challenges such as false-positive rates, the diagnosis of variants of uncertain significance or late-onset forms and the lack of treatment for neuronopathic forms, should be addressed.
Collapse
Affiliation(s)
- Vincenza Gragnaniello
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
- Division of Inherited Metabolic Diseases, Department of Women’s and Children’s Health, University of Padua, 35128 Padua, Italy
| | - Chiara Cazzorla
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Daniela Gueraldi
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Andrea Puma
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Christian Loro
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Elena Porcù
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Maria Stornaiuolo
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Paolo Miglioranza
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women’s and Children’s Health, University of Padua, 35128 Padua, Italy;
| | | | - Alberto B. Burlina
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
- Division of Inherited Metabolic Diseases, Department of Women’s and Children’s Health, University of Padua, 35128 Padua, Italy
| |
Collapse
|
10
|
Gómez-Cebrián N, Gras-Colomer E, Poveda Andrés JL, Pineda-Lucena A, Puchades-Carrasco L. Omics-Based Approaches for the Characterization of Pompe Disease Metabolic Phenotypes. BIOLOGY 2023; 12:1159. [PMID: 37759559 PMCID: PMC10525434 DOI: 10.3390/biology12091159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023]
Abstract
Lysosomal storage disorders (LSDs) constitute a large group of rare, multisystemic, inherited disorders of metabolism, characterized by defects in lysosomal enzymes, accessory proteins, membrane transporters or trafficking proteins. Pompe disease (PD) is produced by mutations in the acid alpha-glucosidase (GAA) lysosomal enzyme. This enzymatic deficiency leads to the aberrant accumulation of glycogen in the lysosome. The onset of symptoms, including a variety of neurological and multiple-organ pathologies, can range from birth to adulthood, and disease severity can vary between individuals. Although very significant advances related to the development of new treatments, and also to the improvement of newborn screening programs and tools for a more accurate diagnosis and follow-up of patients, have occurred over recent years, there exists an unmet need for further understanding the molecular mechanisms underlying the progression of the disease. Also, the reason why currently available treatments lose effectiveness over time in some patients is not completely understood. In this scenario, characterization of the metabolic phenotype is a valuable approach to gain insights into the global impact of lysosomal dysfunction, and its potential correlation with clinical progression and response to therapies. These approaches represent a discovery tool for investigating disease-induced modifications in the complete metabolic profile, including large numbers of metabolites that are simultaneously analyzed, enabling the identification of novel potential biomarkers associated with these conditions. This review aims to highlight the most relevant findings of recently published omics-based studies with a particular focus on describing the clinical potential of the specific metabolic phenotypes associated to different subgroups of PD patients.
Collapse
Affiliation(s)
- Nuria Gómez-Cebrián
- Drug Discovery Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Elena Gras-Colomer
- Pharmacy Department, Hospital Manises of Valencia, 46940 Valencia, Spain
| | | | - Antonio Pineda-Lucena
- Molecular Therapeutics Program, Centro de Investigación Médica Aplicada, 31008 Pamplona, Spain
| | | |
Collapse
|
11
|
Chan MY, Jalil JA, Yakob Y, Wahab SAA, Ali EZ, Khalid MKNM, Leong HY, Chew HB, Sivabalakrishnan JB, Ngu LH. Genotype, phenotype and treatment outcomes of 17 Malaysian patients with infantile-onset Pompe disease and the identification of 3 novel GAA variants. Orphanet J Rare Dis 2023; 18:231. [PMID: 37542277 PMCID: PMC10403872 DOI: 10.1186/s13023-023-02848-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/28/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Pompe disease is a rare glycogen storage disorder caused by deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA), leading to glycogen deposition in multiple tissues. Infantile-onset Pompe disease (IOPD) patients present within the first year of life with profound hypotonia and hypertrophic cardiomyopathy. Treatment with enzyme replacement therapy (ERT) has significantly improved survival for this otherwise lethal disorder. This study aims to describe the clinical and molecular spectrum of Malaysian IOPD patients, and to analyze their long term treatment outcomes. METHODS Seventeen patients diagnosed with IOPD between 2000 and 2020 were included in this retrospective cohort study. Clinical and biochemical data were collated and analyzed using descriptive statistics. GAA enzyme levels were performed on dried blood spots. Molecular analysis of the GAA gene was performed by polymerase chain reaction and Sanger sequencing. Structural modelling was used to predict the effect of the novel mutations on enzyme structure. RESULTS Our cohort had a median age of presentation of 3 months and median age of diagnosis of 6 months. Presenting features were hypertrophic cardiomyopathy (100%), respiratory insufficiency (94%), hypotonia (88%), failure to thrive (82%), feeding difficulties (76%), and hepatomegaly (76%). Fourteen different mutations in the GAA gene were identified, with three novel mutations, c.1552-14_1552-1del, exons 2-3 deletion and exons 6-10 deletion. The most common mutation identified was c.1935C > A p.(D645E), with an allele frequency of 33%. Sixteen patients received ERT at the median age of 7 months. Overall survival was 29%. Mean age of death was 17.5 months. Our longest surviving patient has atypical IOPD and is currently 20 years old. CONCLUSIONS This is the first study to analyze the genotype and phenotype of Malaysian IOPD patients, and has identified the c.1935C > A p.(D645E) as the most common mutation. The three novel mutations reported in this study expands the mutation spectrum for IOPD. Our low survival rate underscores the importance of early diagnosis and treatment in achieving better treatment outcomes.
Collapse
Affiliation(s)
- Mei-Yan Chan
- Department of Genetics, Hospital Kuala Lumpur, Ministry of Health Malaysia, Jalan Pahang, 50586, Kuala Lumpur, Malaysia.
| | - Julaina Abdul Jalil
- Unit of Biochemistry, Institute for Medical Research, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
| | - Yusnita Yakob
- Unit of Molecular Diagnostics, Specialised Diagnostics Centre, National Institutes of Health, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
| | - Siti Aishah Abdul Wahab
- Unit of Molecular Diagnostics, Specialised Diagnostics Centre, National Institutes of Health, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
| | - Ernie Zuraida Ali
- Unit of Inborn Errors of Metabolism and Genetic, Nutrition, Metabolism and Cardiovascular Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
| | - Mohd Khairul Nizam Mohd Khalid
- Unit of Molecular Diagnostics, Specialised Diagnostics Centre, National Institutes of Health, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
| | - Huey-Yin Leong
- Department of Genetics, Hospital Kuala Lumpur, Ministry of Health Malaysia, Jalan Pahang, 50586, Kuala Lumpur, Malaysia
| | - Hui-Bein Chew
- Department of Genetics, Hospital Kuala Lumpur, Ministry of Health Malaysia, Jalan Pahang, 50586, Kuala Lumpur, Malaysia
| | | | - Lock-Hock Ngu
- Department of Genetics, Hospital Kuala Lumpur, Ministry of Health Malaysia, Jalan Pahang, 50586, Kuala Lumpur, Malaysia
| |
Collapse
|
12
|
Brassier A, Pichard S, Schiff M, Bouchereau J, Bérat CM, Caillaud C, Pion A, Khraiche D, Fauroux B, Oualha M, Barnerias C, Desguerre I, Hully M, Maquet M, Deladrière E, de Lonlay P, Gitiaux C. Motor outcomes in patients with infantile and juvenile Pompe disease: Lessons from neurophysiological findings. Mol Genet Metab 2023; 139:107650. [PMID: 37454519 DOI: 10.1016/j.ymgme.2023.107650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
In Infantile Onset Pompe Disease (IOPD), enzyme replacement therapy (ERT) may improve survival, cardiac function, and motor development. However, even with early enzyme replacement therapy, some patients experienced poor response to ERT and abnormal motor milestones that could be due to motor neuron involvement. In this long-term retrospective study, we analyzed concomitant clinical motor outcomes and electroneuromyography (ENMG) findings in patients with IOPD and Juvenile Onset Pompe Disease (JOPD). Twenty-nine pediatric patients were included and 20 surviving were analyzed for neuromotor studies: 12 had IOPD (group 1), 4 had JOPD (group 2) and 4 (group 3) received ERT in the first month of age. Motor nerve conduction studies were mostly normal. Needle EMG performed at diagnosis always indicated the existence of myopathy that responded to ERT. Two IOPD patients (group 1) presenting with mixed motor neuropathy and myopathy displayed a poor outcome and never walked. Two patients became non-walkers (one IOPD patient and one patient of group 3) at respectively 9 and 3 years of age. One JOPD patient is about to lose walking ability. This motor deterioration was associated with the development of a motor neuropathy. Patients older than 10 years of age develop a motor neuropathy. Initial or secondary motor neuron involvement seems to be associated with a poor motor outcome showing that ERT may fail to prevent the accumulation of glycogen in motor neuron. Neurophysiological findings are important to assess severity of motor neuron damage in all Pompe pediatric patients and should be systematically performed.
Collapse
Affiliation(s)
- Anaïs Brassier
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, G2M, MetabERN, Paris, France.
| | - Samia Pichard
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, G2M, MetabERN, Paris, France
| | - Manuel Schiff
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, G2M, MetabERN, Paris, France; INSERM UMRS_1163, Imagine Institute, Paris, France
| | - Juliette Bouchereau
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, G2M, MetabERN, Paris, France
| | - Claire-Marine Bérat
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, G2M, MetabERN, Paris, France
| | - Catherine Caillaud
- Biochemistry Unit, Biology Department, Assistance Publique Hôpitaux de Paris (AP-HP), Necker-Enfants-Malades University Hospital, Paris, France; Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Aude Pion
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, G2M, MetabERN, Paris, France
| | - Diala Khraiche
- Department of Pediatric cardiology, Necker-Enfants malades Hospital, University of Paris, AP-HP, Paris, France
| | - Brigitte Fauroux
- Pediatric Noninvasive Ventilation and Sleep Unit, Necker University Hospital, Paris, Paris Descartes University, Paris, Research Unit INSERM U 955, Team 13, Creteil, France
| | - Mehdi Oualha
- Pediatric Intensive Care Unit, Necker-Enfants-Malades University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Christine Barnerias
- Reference Center for neuromuscular diseases, Necker-Enfants-Malades University Hospital, APHP, FILNEMUS, Paris, France
| | - Isabelle Desguerre
- Reference Center for neuromuscular diseases, Necker-Enfants-Malades University Hospital, APHP, FILNEMUS, Paris, France
| | - Marie Hully
- Department of Pediatric Rehabilitation, Necker-Enfants malades Hospital, University of Paris, AP-HP, Paris, France
| | - Marion Maquet
- Department of Pediatric Rehabilitation, Necker-Enfants malades Hospital, University of Paris, AP-HP, Paris, France
| | - Elodie Deladrière
- Department of Pediatric Rehabilitation, Necker-Enfants malades Hospital, University of Paris, AP-HP, Paris, France
| | - Pascale de Lonlay
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, G2M, MetabERN, Paris, France; INSERM U1151, Institut Necker Enfants-Malades (INEM), Paris, France
| | - Cyril Gitiaux
- Reference Center for neuromuscular diseases, Necker-Enfants-Malades University Hospital, APHP, FILNEMUS, Paris, France; Department of Pediatric Neurophysiology, Necker-Enfants malades Hospital, University of Paris, AP-HP, Paris, France
| |
Collapse
|
13
|
Zhang WC, Mao YY, Chen Q. [Research progress of nervous system damage in Pompe disease]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:420-424. [PMID: 37073849 PMCID: PMC10120337 DOI: 10.7499/j.issn.1008-8830.2211052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Pompe disease, also known as glycogen storage disease type Ⅱ, is a rare autosomal recessive disease. With the application of enzyme replacement therapy, more and more patients with Pompe disease can survive to adulthood, and nervous system-related clinical manifestations gradually emerge. Nervous system involvement seriously affects the quality of life of patients with Pompe disease, and a systematic understanding of the clinical manifestations, imaging features and pathological changes of nervous system injury in Pompe disease is of great significance for the early identification and intervention of Pompe disease. This article reviews the research progress of neurological damage in Pompe disease.
Collapse
Affiliation(s)
- Wen-Chao Zhang
- Department of Neurology, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Ying-Ying Mao
- Department of Neurology, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Qian Chen
- Department of Neurology, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| |
Collapse
|
14
|
Enzyme Replacement Therapy (ERT) on Heart Function Changes the Outcome in Patients with Infantile-Onset Pompe Disease: A Familial History. Case Rep Pediatr 2023; 2023:8470341. [PMID: 36845322 PMCID: PMC9957634 DOI: 10.1155/2023/8470341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/08/2022] [Accepted: 02/06/2023] [Indexed: 02/19/2023] Open
Abstract
Background Lysosomal acid alpha-glucosidase (GAA) deficiency, also known as Pompe disease, is an autosomal recessive disorder that leads to the accumulation of glycogen in lysosomes and cytoplasm, resulting in tissue destruction. Infantile-onset GAA deficiency is characterized by cardiomyopathy and severe generalized hypotonia. Without treatment, most patients die within the first two years of life. The demonstration of reduced GAA activity, followed by sequencing of the GAA gene, confirms the disease. GAA deficiency is currently treated with enzyme replacement therapy (ERT) with improved clinical outcomes and survival. Case Presentation. We describe the case of DGAA in two siblings, in which the diagnostic time point, treatment, and outcomes were completely different. The girl was diagnosed with DGAA at the age of 6 months during investigations for poor weight gain and excessive sleepiness. The finding of severe cardiomyopathy through EKG and echocardiography led to the suspicion of storage disease, and the GAA deficiency was later confirmed by genetic analysis. The girl died of complications due to the clinical picture before starting ERT. Conversely, her younger brother had the opportunity to receive an early diagnosis and the rapid onset of ERT. He is showing a regression of cardiac hypertrophy. Conclusion The advent of ERT improved clinical outcomes and survival in infantile-onset PD. Its impact on cardiac function is still under study, but different reports in the literature have shown encouraging data. Early recognition of DGAA and prompt initiation of ERT is therefore crucial to prevent the progression of the disease and improve the outcomes.
Collapse
|
15
|
Treatment Dilemma in Children with Late-Onset Pompe Disease. Genes (Basel) 2023; 14:genes14020362. [PMID: 36833288 PMCID: PMC9957524 DOI: 10.3390/genes14020362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/10/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
In recent years, there has been a significant increase in the diagnosis of asymptomatic Late-Onset Pompe Disease (LOPD) patients, who are detected via family screening or Newborn Screening (NBS). The dilemma is when to start Enzyme Replacement Therapy (ERT) in patients without any clinical sign of the disease, considering its important benefits in terms of loss of muscle but also its very high cost, risk of side effects, and long-term immunogenicity. Muscle Magnetic Resonance Imaging (MRI) is accessible, radiation-free, and reproducible; therefore, it is an important instrument for the diagnosis and follow-up of patients with LOPD, especially in asymptomatic cases. European guidelines suggest monitoring in asymptomatic LOPD cases with minimal MRI findings, although other guidelines consider starting ERT in apparently asymptomatic cases with initial muscle involvement (e.g., paraspinal muscles). We describe three siblings affected by LOPD who present compound heterozygosis and wide phenotypic variability. The three cases differ in age at presentation, symptoms, urinary tetrasaccharide levels, and MRI findings, confirming the significant phenotypic variability of LOPD and the difficulty in deciding when to start therapy.
Collapse
|
16
|
Xi H, Li X, Ma L, Yin X, Yang P, Zhang L. Infantile Pompe disease with intrauterine onset: a case report and literature review. Ital J Pediatr 2022; 48:187. [PMID: 36411466 PMCID: PMC9677902 DOI: 10.1186/s13052-022-01379-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/02/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Pompe disease is a rare autosomal recessive disease. Acid alpha-glucosidase (GAA) deficiency leads to glycogen storage in lysosomes, causing skeletal, cardiac, and smooth muscle lesions. Pompe disease is progressive, and its severity depends on the age of onset. Classic infantile Pompe disease, the most severe form, is characterized by an age of onset before 12 months. Pompe disease with intrauterine onset has rarely been reported. CASE PRESENTATION The proband was born at a gestational age of 40 weeks and 3 days and admitted to our hospital because of intrauterine cardiac hypertrophy, shortness of breath, and cyanosis until 13 min postnatally. Physical examination at admission revealed poor responsiveness, pale skin, shortness of breath, reduced limb muscle tone, and bilateral pedal edema. The heart sounds were weak, and no heart murmur was heard. Echocardiography showed left (9 mm) and right (5 mm) ventricular hypertrophies. The patient was subjected to non-invasive ventilator-assisted respiration, fluid restriction, diuresis, and metoprolol treatment. Infantile Pompe disease was diagnosed on day 16 with a GAA enzymatic activity of 0.31 µmol/L/h and with the full-penetrance genetic test showing the homozygous gene mutation c.1844G>T(p.Gly615Val). Enzyme replacement therapy was refused by the patient's parents, and the patient died at seven months of age from cardiopulmonary failure. CONCLUSION Infants with intrauterine-onset Pompe disease usually have early manifestations of heart disease. Prompt GAA enzymatic activity determination and molecular genetic testing are helpful in aiding the parents' decision and planning the treatment.
Collapse
Affiliation(s)
- Hongmin Xi
- grid.412521.10000 0004 1769 1119Neonatology Department, The Affiliated Hospital of Qingdao University, NO.16 Jiangsu Road, Shinan district, Qingdao, 266003 Shandong China
| | - Xianghong Li
- grid.412521.10000 0004 1769 1119Neonatology Department, The Affiliated Hospital of Qingdao University, NO.16 Jiangsu Road, Shinan district, Qingdao, 266003 Shandong China
| | - Lili Ma
- grid.412521.10000 0004 1769 1119Neonatology Department, The Affiliated Hospital of Qingdao University, NO.16 Jiangsu Road, Shinan district, Qingdao, 266003 Shandong China
| | - Xiangyun Yin
- grid.412521.10000 0004 1769 1119Neonatology Department, The Affiliated Hospital of Qingdao University, NO.16 Jiangsu Road, Shinan district, Qingdao, 266003 Shandong China
| | - Ping Yang
- grid.412521.10000 0004 1769 1119Neonatology Department, The Affiliated Hospital of Qingdao University, NO.16 Jiangsu Road, Shinan district, Qingdao, 266003 Shandong China
| | - Lulu Zhang
- grid.412521.10000 0004 1769 1119Neonatology Department, The Affiliated Hospital of Qingdao University, NO.16 Jiangsu Road, Shinan district, Qingdao, 266003 Shandong China
| |
Collapse
|
17
|
Gragnaniello V, Pijnappel PW, Burlina AP, In 't Groen SL, Gueraldi D, Cazzorla C, Maines E, Polo G, Salviati L, Di Salvo G, Burlina AB. Newborn screening for Pompe disease in Italy: Long-term results and future challenges. Mol Genet Metab Rep 2022; 33:100929. [PMID: 36310651 PMCID: PMC9597184 DOI: 10.1016/j.ymgmr.2022.100929] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Pompe disease (PD) is a progressive neuromuscular disorder caused by a lysosomal acid α-glucosidase (GAA) deficiency. Enzymatic replacement therapy is available, but early diagnosis by newborn screening (NBS) is essential for early treatment and better outcomes, especially with more severe forms. We present results from 7 years of NBS for PD and the management of infantile-onset (IOPD) and late-onset (LOPD) patients, during which we sought candidate predictive parameters of phenotype severity at baseline and during follow-up. We used a tandem mass spectrometry assay for α-glucosidase activity to screen 206,741 newborns and identified 39 positive neonates (0.019%). Eleven had two pathogenic variants of the GAA gene (3 IOPD, 8 LOPD); six carried variants of uncertain significance (VUS). IOPD patients were treated promptly and had good outcomes. LOPD and infants with VUS were followed; all were asymptomatic at the last visit (mean age 3.4 years, range 0.5–5.5). Urinary glucose tetrasaccharide was a useful and biomarker for rapidly differentiating IOPD from LOPD and monitoring response to therapy during follow-up. Our study, the largest reported to date in Europe, presents data from longstanding NBS for PD, revealing an incidence in North East Italy of 1/18,795 (IOPD 1/68,914; LOPD 1/25,843), and the absence of mortality in IOPD treated from birth. In LOPD, rigorous long-term follow-up is needed to evaluate the best time to start therapy. The high pseudodeficiency frequency, ethical issues with early LOPD diagnosis, and difficulty predicting phenotypes based on biochemical parameters and genotypes, especially in LOPD, need further study.
Collapse
Key Words
- Acid α-glucosidase
- CLIR, Collaborative Laboratory Integrated Reports
- CRIM, cross-reactive immunological material
- DBS, dried blood spot
- DMF, digital microfluidics
- ECG, electrocardiogram
- EF, ejection fraction
- EMG, electromyography
- ERT, enzyme replacement therapy
- Enzyme replacement therapy
- GAA, acid α-glucosidase
- GMFM-88, Gross Motor Function Measure
- Glc4, glucose tetrasaccharide
- IOPD, infantile-onset Pompe disease
- ITI, immunotolerance induction
- LOPD, late-onset Pompe disease
- LVMI, left ventricular max index
- MFM-20, motor function measurement
- MRC, Medical Research Council Scale
- MRI, magnetic resonance imaging
- MS/MS, tandem mass spectrometry
- NBS, newborn screening
- Newborn screening
- PBMC, peripheral blood mononuclear cells
- PD, Pompe disease
- PPV, positive predictive value
- Pompe disease
- RUSP, Recommended Uniform Screening Panel
- Tandem mass-spectrometry
- Urinary tetrasaccharide
- VUS, variants of uncertain significance.
- nv, normal values
- rhGAA, recombinant human GAA
Collapse
Affiliation(s)
- Vincenza Gragnaniello
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Pim W.W.M. Pijnappel
- Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Stijn L.M. In 't Groen
- Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Daniela Gueraldi
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Chiara Cazzorla
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Evelina Maines
- Division of Pediatrics, S. Chiara General Hospital, Trento, Italy
| | - Giulia Polo
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women's and Children's Health, and Myology Center, University of Padova, Padova, Italy
| | - Giovanni Di Salvo
- Division of Paediatric Cardiology, Department of Women's and Children's Health, University Hospital Padua, Padua, Italy
| | - Alberto B. Burlina
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
- Corresponding author at: Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, via Orus 2/c, 35129 Padua, Italy.
| |
Collapse
|
18
|
Goomber S, Huggins E, Rehder CW, Cohen JL, Bali DS, Kishnani PS. Development of a clinically validated in vitro functional assay to assess pathogenicity of novel GAA variants in patients with Pompe disease identified via newborn screening. Front Genet 2022; 13:1001154. [PMID: 36246652 PMCID: PMC9562992 DOI: 10.3389/fgene.2022.1001154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose: The addition of Pompe disease (Glycogen Storage Disease Type II) to the Recommended Uniform Screening Panel in the United States has led to an increase in the number of variants of uncertain significance (VUS) and novel variants identified in the GAA gene. This presents a diagnostic challenge, especially in the setting of late-onset Pompe disease when symptoms are rarely apparent at birth. There is an unmet need for validated functional studies to aid in classification of GAA variants. Methods: We developed an in vitro mammalian cell expression and functional analysis system based on guidelines established by the Clinical Genome Resource (ClinGen) Sequence Variant Interpretation Working Group for PS3/BS3. We validated the assay with 12 control variants and subsequently analyzed eight VUS or novel variants in GAA identified in patients with a positive newborn screen for Pompe disease without phenotypic evidence of infantile-onset disease. Results: The control variants were analyzed in our expression system and an activity range was established. The pathogenic controls had GAA activity between 0% and 11% of normal. The benign or likely benign controls had an activity range of 54%–100%. The pseudodeficiency variant had activity of 17%. These ranges were then applied to the variants selected for functional studies. Using the threshold of <11%, we were able to apply PS3_ supporting to classify two variants as likely pathogenic (c.316C > T and c.1103G > A) and provide further evidence to support the classification of likely pathogenic for two variants (c.1721T > C and c.1048G > A). One variant (c.1123C > T) was able to be reclassified based on other supporting evidence. We were unable to reclassify three variants (c.664G > A, c.2450A > G, and c.1378G > A) due to insufficient or conflicting evidence. Conclusion: We investigated eight GAA variants as proof of concept using our validated and reproducible in vitro expression and functional analysis system. While additional work is needed to further refine our system with additional controls and different variant types in order to apply the PS3/BS3 criteria at a higher level, this tool can be utilized for variant classification to meet the growing need for novel GAA variant classification in the era of newborn screening for Pompe disease.
Collapse
Affiliation(s)
- Shelly Goomber
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Erin Huggins
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Catherine W. Rehder
- Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Jennifer L. Cohen
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Deeksha S. Bali
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- Biochemical Genetics Laboratory, Duke University Medical Center, Durham, NC, United States
| | - Priya S. Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- *Correspondence: Priya S. Kishnani,
| |
Collapse
|
19
|
Yang CF, Liao TWE, Chu YL, Chen LZ, Huang LY, Yang TF, Ho HC, Kao SM, Niu DM. Long-term outcomes of very early treated infantile-onset Pompe disease with short-term steroid premedication: experiences from a nationwide newborn screening programme. J Med Genet 2022; 60:430-439. [PMID: 36137614 DOI: 10.1136/jmg-2022-108675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/02/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Starting enzyme replacement therapy (ERT) before severe irreversible muscular damage occurs is important in infantile-onset Pompe disease (IOPD). This long-term follow-up study demonstrates our diagnostic and treatment strategies for IOPD and compares our clinical outcomes with those of other medical centres. METHODS In this long-term follow-up study, we analysed the outcomes of very early ERT with premedication hydrocortisone in patients with IOPD. Out of 1 228 539 infants screened between 1 January 2010 and 28 February 2021, 33 newborns had confirmed IOPD in Taipei Veterans General Hospital. Twenty-six were regularly treated and monitored at Taipei Veterans General Hospital. Echocardiographic parameters, biomarkers, IgG antibodies against alglucosidase alpha, pulmonary function variables and developmental status were all assessed regularly over an average follow-up duration of 6.18±3.14 years. We compared the long-term treatment outcomes of our patients with those of other research groups. RESULTS The average age at ERT initiation was 9.75±3.17 days for patients with classic IOPD. The average of the latest antialglucosidase alpha IgG titre was 669.23±1159.23. All enrolled patients had normal heart sizes, motor milestones, cognitive function and pulmonary function that were near-normal to normal. Compared with patients in other studies, our patients had better outcomes in all aspects. CONCLUSION Very early ERT using our rapid diagnostic and treatment strategy enabled our patients with IOPD to have better outcomes than patients in other medical centres.
Collapse
Affiliation(s)
- Chia-Feng Yang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ting-Wei Ernie Liao
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen-Ling Chu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Li-Zhen Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ling-Yi Huang
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Internal Medicine, Division of Nephrology, Taipei City Hospital Heping Fuyou Branch, Taipei, Taiwan
| | - Tsui-Feng Yang
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hui-Chen Ho
- Taipei Institute of Pathology, Neonatal Screening Center, Taipei, Taiwan
| | - Shu-Min Kao
- Chinese Foundation of Health, Neonatal Screening Center, Taipei, Taiwan
| | - Dau-Ming Niu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan .,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
20
|
Improving Recognition of Treatable Rare Neuromuscular Disorders in Primary Care: A Pilot Feasibility Study. CHILDREN 2022; 9:children9071063. [PMID: 35884047 PMCID: PMC9317909 DOI: 10.3390/children9071063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/14/2022] [Accepted: 07/14/2022] [Indexed: 12/01/2022]
Abstract
Innovative targeted treatments for neuromuscular disorders (NMDs) can dramatically improve the course of illness. Diagnostic delay, however, is a major impediment. Here, we present a pilot project aimed at assessing the feasibility of a screening program to identify children at high risk for NMDs within the first 30 months of life. The Promoting Early Diagnosis for Neuromuscular Disorders (PEDINE) project implemented a three-step sequential screening in an area of about 300,000 people with (1) an assessment of the motor development milestones to identify “red flags” for NMDs by primary care pediatricians (PCPs) as part of the routine Health Status Check visits; (2) for the children who screened positive, a community neuropsychiatric assessment, with further referral of suspected NMD cases to (3) a hospital-based specialized tertiary care center. In the first-year feasibility study, a total of 10,032 PCP visits were conducted, and twenty children (0.2% of the total Health Status Check visits) screened positive and were referred to the community neuropsychiatrist. Of these, four had elevated creatine kinase (CK) serum levels. This pilot study shows that screening for NMDs in primary care settings is feasible and allows children at high risk for muscular disorder to be promptly identified.
Collapse
|
21
|
Chen YK, Teng CT, Yang CF, Niu DM, Huang WJ, Fan YH. Prevalence of lower urinary tract symptoms in children with early-treated infantile-onset Pompe disease: A single-centre cross-sectional study. Neurourol Urodyn 2022; 41:1177-1184. [PMID: 35481613 DOI: 10.1002/nau.24950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/12/2022] [Accepted: 04/20/2022] [Indexed: 11/06/2022]
Abstract
AIM To evaluate lower urinary tract symptoms (LUTS) in children with infantile-onset Pompe disease (IOPD) who received early treatment. METHODS Pompe disease (PD), or glycogen storage disease II is a rare autosomal recessive lysosomal storage disease that affects multiple organ systems. To our knowledge, only one study has focused on the relationship between LUTS and incontinence in children with PD. This cross-sectional study was conducted from August 2019 through March 2021 and children with IOPD, who had received early and regular enzyme replacement therapy, were enrolled. Participants or their parents completed the Dysfunctional Voiding Scoring System (DVSS) questionnaire. All children underwent uroflowmetry and postvoid residual urine measurements. Fourteen children (age, 4-9 years) with IOPD were enrolled. RESULTS Ten patients (71.4%) had abnormal uroflow curves. In addition, results of the DVSS revealed that approximately half (42.9%) of our IOPD patients had voiding dysfunction, with urinary incontinence as the most common symptom (64.3%, 9/14). No significant correlations were found between LUTS and uroflow curves in children with IOPD. CONCLUSIONS The frequency of LUTS and lower urinary tract dysfunction noted on uroflowmetry should encourage pediatricians to actively identify IOPD patients with LUTS, regardless of the timing and frequency of their treatments, and refer them to a urologist for further evaluation and appropriate treatment.
Collapse
Affiliation(s)
- Yu-Kuang Chen
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chao-Ting Teng
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Feng Yang
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Pediatrics, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pediatrics, College of Medicine, National Yang Ming University, Taipei, Taiwan
| | - Dau-Ming Niu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Pediatrics, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pediatrics, College of Medicine, National Yang Ming University, Taipei, Taiwan
| | - William J Huang
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Urology, College of Medicine and Shu-Tien Urological Science Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Hua Fan
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Urology, College of Medicine and Shu-Tien Urological Science Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
22
|
Huggins E, Holland M, Case LE, Blount J, Landstrom AP, Jones HN, Kishnani PS. Early clinical phenotype of late onset Pompe disease: Lessons learned from newborn screening. Mol Genet Metab 2022; 135:179-185. [PMID: 35123877 DOI: 10.1016/j.ymgme.2022.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 01/14/2023]
Abstract
PURPOSE Thoroughly phenotype children with late-onset Pompe disease (LOPD) diagnosed via newborn screening (NBS) to provide guidance for long-term follow up. METHODS Twenty infants ages 6-21 months with LOPD diagnosed by NBS underwent systematic clinical evaluation at Duke University including cardiac imaging, biomarker testing, physical therapy evaluation, and speech-language pathology evaluation. RESULTS Of the 20 infants, four were homozygous for the "late-onset" IVS1 splice site variant c.-32-13 T > G, fourteen were compound heterozygous, and two did not have any copies of this variant. None of the patients had evidence of cardiomyopathy or cardiac rhythm disturbances. Biomarker testing showed an increase in CK, AST, and ALT in 8 patients (40%) and increase in Glc4 in two patients (10%). All patients demonstrated postural and kinematic concerns. Three patients (17%) scored below the 10%ile on the Alberta Infant Motor Scale (AIMS) and 15 patients (83%) scored above the 10%ile. Speech-language pathology assessments were normal in all patients and mild feeding/swallowing abnormalities were noted in nine patients (45%). CONCLUSION Our data show high variability among children with LOPD diagnosed via NBS. Careful physical therapy evaluation is necessary to monitor for subtle musculoskeletal signs that may reflect early muscle involvement. Patients should be monitored closely for symptom progression.
Collapse
Affiliation(s)
- Erin Huggins
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Maggie Holland
- Department of Physical and Occupational Therapy, Duke University Health System, Durham, NC, USA
| | - Laura E Case
- Doctor of Physical Therapy Division, Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Janet Blount
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology and Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Harrison N Jones
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
23
|
Tocan V, Mushimoto Y, Kojima-Ishii K, Matsuda A, Toda N, Toyomura D, Hirata Y, Sanefuji M, Sawada T, Sakai Y, Nakamura K, Ohga S. The earliest enzyme replacement for infantile-onset Pompe disease in Japan. Pediatr Int 2022; 64:e15286. [PMID: 36074069 DOI: 10.1111/ped.15286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/15/2022] [Accepted: 06/18/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Infantile-onset Pompe disease (IOPD) is the most severe phenotype of a lysosomal storage disorder caused by acid alpha-glucosidase (GAA) deficiency. An enzymatic newborn screening (NBS) program started regionally in Japan in 2013 for early enzyme replacement therapy (ERT). We report the ERT responses of the first NBS-identified Japanese IOPD case and of another case diagnosed prior to NBS, to discuss the problems of promptly starting ERT in Japan. METHODS Acid alpha-glucosidase activity was measured by fluorometric assay in both patients. The diagnosis of IOPD was confirmed by next-generation followed by Sanger-method sequencing (patient 1) or direct sequencing of polymerase chain reaction (PCR)-amplified products (patient 2) of the GAA gene. RESULTS A female infant identified by NBS had a novel out-of-frame (p.F181Dfs*6) variant and a reported pathogenic (p.R600C) variant, along with two pseudodeficiency variants. Enzyme replacement therapy was started at age 58 days when the infant had increased serum levels of creatine kinase and slight myocardial hypertrophy. Clinical and biochemical markers improved promptly. She has been alive and well without delayed development at age 14 months. Patient 2, a Japanese male, received a diagnosis of IOPD at age 5 months before the NBS era. He had a homozygotic variant of GAA (p.R608X), later registered as a cross-reactive immunological material (CRIM)-negative genotype, and developed a high titer of anti-rhGAA antibodies. The patient has survived myocardial hypertrophy with continuous respiratory support for 12 years of ERT. CONCLUSIONS Enzyme replacement therapy should not be delayed over the age of 2 months for reversible cardiac function, although CRIM-negative cases may hamper turnaround time reduction.
Collapse
Affiliation(s)
- Vlad Tocan
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Yuichi Mushimoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Kanako Kojima-Ishii
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Akane Matsuda
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Naoko Toda
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Daisuke Toyomura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Yuichiro Hirata
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Masafumi Sanefuji
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan.,Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Takaaki Sawada
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| |
Collapse
|
24
|
Wang TH, Soong WJ, Niu DM, Chu YL, Chen LZ, Huang LY, Yang CF. Airway abnormalities and pulmonary complications in long-term treated late-onset Pompe disease: Diagnostic and interventional by flexible bronchoscopy. Pediatr Pulmonol 2022; 57:185-192. [PMID: 34647686 DOI: 10.1002/ppul.25725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/27/2021] [Accepted: 10/11/2021] [Indexed: 11/09/2022]
Abstract
This study evaluates the whole airway abnormalities of long-term treated late-onset Pompe disease (LOPD) patients, with interventions using the flexible bronchoscope (FB). As a retrospective study, we follow up with our five LOPD patients treated with Myozyme from 2012 to 2021 regularly, but with a focus on the whole airway abnormalities of these patients visualized through FB. The long-term clinical outcomes and relevant airway symptoms were assessed. Pulmonary function test and polysomnography were performed to evaluate the degree of respiratory compromise. All patients in the study had varying degrees of airway collapsibility, pulmonary complications, sleep apnea syndrome, and facial anomalies. Pulmonary function could preserve after Myozyme treatment, but potential deterioration thereafter. This is the first study that focuses on airway abnormalities and pulmonary complications in long-term treated LOPD patients using FB. Despite years of Myozyme treatment, we still observed airway abnormalities in these patients. In our series, the pulmonary complications seem more obvious than those observed in patients with infantile-onset Pompe disease, which might be related to the late diagnosis and treatment. We might recommend that FB could provide dynamic evaluation and interventions of airway abnormalities simultaneously. Early diagnosis of respiratory dysfunction is a critical prognostic factor of the long-term outcome of treated LOPD patients.
Collapse
Affiliation(s)
- Ting-Hao Wang
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wen-Jue Soong
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Pediatric Pulmonology, China Medical University Children's Hospital, China Medical University, Taichung, Taiwan.,Department of Pediatrics, Tri-Service General Hospital, Taipei, Taiwan
| | - Dau-Ming Niu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | - Li-Zhen Chen
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ling-Yi Huang
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Taipei City Hospital-Heping Fuyou Branch, Taipei, Taiwan
| | - Chia-Feng Yang
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
25
|
Joyce Liao HC, Chen HJ. Multiplex Lysosomal Enzyme Activity Assay on Dried Blood Spots Using Tandem Mass Spectrometry. Methods Mol Biol 2022; 2546:261-269. [PMID: 36127596 DOI: 10.1007/978-1-0716-2565-1_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Deficiencies of the enzymes in lysosomes result in the accumulation of undegraded materials and subsequently cellular dysfunction. Early identification of deficiencies can lead to better clinical outcomes before irreversible organ and tissue damages occur. In this chapter, lysosomal enzymes are extracted from dried blood spots and incubated with the commercialized and multiplexed enzyme cocktail containing corresponding substrates and internal standards. After incubation, the enzymatic reactions are quenched, and the mixtures of the reaction products are prepared using liquid/liquid extractions. Multiple enzymes are quantified simultaneously using selected ion monitoring on liquid chromatography-mass spectrometry (LC-MS/MS) system.
Collapse
Affiliation(s)
- Hsuan-Chieh Joyce Liao
- Departments of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| | - Hsiao-Jan Chen
- The Chinese Foundation of Health, Newborn screening Center, Taipei, Taiwan
| |
Collapse
|
26
|
Importance of Timely Treatment Initiation in Infantile-Onset Pompe Disease, a Single-Centre Experience. CHILDREN (BASEL, SWITZERLAND) 2021; 8:children8111026. [PMID: 34828739 PMCID: PMC8620435 DOI: 10.3390/children8111026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 01/10/2023]
Abstract
Classic infantile Pompe disease (IPD) is a rare lysosomal storage disorder characterized by severe hypertrophic cardiomyopathy and profound muscle weakness. Without treatment, death occurs within the first 2 years of life. Although enzyme replacement therapy (ERT) with alglucosidase alfa has improved survival, treatment outcome is not good in many cases and is largely dependent on age at initiation. The objective of the study was (a) to analyse the different stages in the diagnosis and specific treatment initiation procedure in IPD patients, and (b) to compare clinical and biochemical outcomes depending on age at ERT initiation (<1 month of age vs. <3 months of age). Here, we show satisfactory clinical and biochemical outcomes in two IPD patients after early treatment initiation before 3 months of life with immunomodulatory therapy in the ERT-naïve setting, with a high ERT dose from the beginning. Despite the overall good evolution, the patient who initiated treatment <1 month of life presented even better outcomes than the patient who started treatment <3 months of life, with an earlier normalization of hypertrophic cardiomyopathy, along with CK normalization, highlighting the importance of early treatment initiation in this progressive disease before irreversible muscle damage has occurred.
Collapse
|
27
|
Dangouloff T, Boemer F, Servais L. Newborn screening of neuromuscular diseases. Neuromuscul Disord 2021; 31:1070-1080. [PMID: 34620514 DOI: 10.1016/j.nmd.2021.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 12/11/2022]
Abstract
Neuromuscular diseases represent an heterogenous group of more than 400 diseases, with a very broad phenotypic spectrum. Given their rarity and complexity, neuromuscular diseases are often diagnosed with a very significant delay after which irreversible muscle damage may limit the efficacy of treatments when available. In this context, neonatal screening could constitute a solution for early detection and treatment. A systematic review of the literature in PubMed up to May 1, 2021, was conducted according to PRISMA guidelines, including classical neuromuscular diseases and diseases with a clear peripheral nervous system involvement (including central nervous system disease with severe neuropathy). We found seven diseases for which newborn screening data were reported: spinal muscular atrophy (9), Duchenne muscular dystrophy (9), Pompe disease (8), X-linked adrenoleukodystrophy (5), Krabbe disease (4), myotonic dystrophy type 1 (1), metachromatic leukodystrophy (1). The future of newborn screening for neuromuscular disorders pass through a global technological switch, from a biochemical to a genetic-based approach. The rapid development of therapy also requires the possibility to quickly adapt the list of treated conditions, to allow innovative therapies to achieve their best efficacy.
Collapse
Affiliation(s)
- Tamara Dangouloff
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Pediatrics, University Hospital Liège & University of Liège, Belgium.
| | - François Boemer
- Biochemical Genetics Lab, Department of Human Genetics, CHU of Liège, University of Liège, Liège, Belgium
| | - Laurent Servais
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Pediatrics, University Hospital Liège & University of Liège, Belgium; MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford, UK.
| |
Collapse
|
28
|
Davids L, Sun Y, Moore RH, Lisi E, Wittenauer A, Wilcox WR, Ali N. Health care practitioners' experience-based opinions on providing care after a positive newborn screen for Pompe disease. Mol Genet Metab 2021; 134:20-28. [PMID: 34602357 DOI: 10.1016/j.ymgme.2021.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 01/14/2023]
Abstract
The addition of Pompe disease (PD) and other conditions with later-onset forms to newborn screening (NBS) in the United States (US) has been controversial. NBS technology cannot discern infantile-onset PD (IOPD) from later-onset PD (LOPD) without clinical follow-up. This study explores genetic health care practitioners' (HCPs) experiences and challenges providing NBS patient care throughout the US and their resultant opinions on NBS for PD. An online survey was distributed to genetic counselors, geneticists, NBS follow-up care coordinators, and nurse practitioners caring for patients with positive NBS results for PD. Analysis of 78 surveys revealed the majority of participating HCPs support inclusion of PD on NBS. Almost all HCPs (93.3%) feel their state has sufficient resources to provide follow-up medical care for IOPD; however, only three-fourths (74.6%) believed this for LOPD. Common barriers included time lag between NBS and confirmatory results, insurance difficulties for laboratory testing, and family difficulties in seeking medical care. HCPs more frequently encountered barriers providing care for LOPD than IOPD (53.9% LOPD identified ≥3 barriers, 31.1% IOPD). HCPs also believe creation of a population of presymptomatic individuals with LOPD creates a psychological burden on the family (87.3% agree/strongly agree), unnecessary medicalization of the child (63.5% agree/strongly agree), and parental hypervigilance (68.3% agree/strongly agree). Opinions were markedly divided on the use of reproductive benefit as a justification for NBS. Participants believe additional education for pediatricians and other specialists would be beneficial in providing care for patients with both IOPD and LOPD, in addition to the creation of evidence-based official guidelines for care and supportive resources for families with LOPD.
Collapse
Affiliation(s)
- Laura Davids
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States of America.
| | - Yuxian Sun
- Department of Biostatistics & Bioinformatics, Rollins School of Public Health and Biostatistics Collaboration Core, Emory University School of Medicine, Atlanta, GA, United States of America; Clinical Center on TB, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Reneé H Moore
- Department of Biostatistics & Bioinformatics, Rollins School of Public Health and Biostatistics Collaboration Core, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Emily Lisi
- Graduate School of Arts and Sciences, Biomedical Sciences Division, Wake Forest University, Winston-Salem, NC, United States of America
| | - Angela Wittenauer
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - William R Wilcox
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Nadia Ali
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States of America
| |
Collapse
|
29
|
Hearing characteristics of infantile-onset Pompe disease after early enzyme-replacement therapy. Orphanet J Rare Dis 2021; 16:348. [PMID: 34353347 PMCID: PMC8340467 DOI: 10.1186/s13023-021-01817-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 04/06/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Studies suggest that enzyme-replacement therapy (ERT) is crucial to the survival of patients with infantile-onset Pompe disease (IOPD). Hearing impairment (HI) is one of the clinical sequelae observed in long-term survivors. However, the benefits of early ERT for hearing outcomes have not yet been reported. This study aimed to investigate the impact of early ERT on IOPD patients. METHODS This retrospective longitudinal study recruited IOPD patients who were referred by newborn screening for confirmatory diagnosis based on our rapid diagnostic criteria and received early ERT treatment between January 1, 2010, and January 31, 2018. The hearing test battery included a tympanogram, otoacoustic emission, auditory brainstem evoked response (ABR), pure-tone audiometry or conditioned play audiometry. RESULTS Nineteen patients with IOPD were identified, 6 of whom had hearing impairment (HI); 1 had conductive HI, 2 had sensorineural HI (one had bilateral mild HI and one had mild HI in a single ear) and 1 had moderate mixed-type HI. Two patients failed the newborn screening test and had mild HI in the ABR. The mean age of the initial time to ERT was 11.05 ± 4.31 days, and the HI rate was 31.6% (6/19). CONCLUSION Our study is the largest cohort to show the characteristic hearing outcomes of IOPD patients after ERT. Early ERT within 2 weeks after birth may contribute to better hearing outcomes. Clinicians should be vigilant in testing for the hearing issues associated with IOPD and should intervene early if any HI is detected.
Collapse
|
30
|
Park KS. Carrier frequency and predicted genetic prevalence of Pompe disease based on a general population database. Mol Genet Metab Rep 2021; 27:100734. [PMID: 33717985 PMCID: PMC7933537 DOI: 10.1016/j.ymgmr.2021.100734] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 12/15/2022] Open
Abstract
Background The genetic prevalence of Pompe disease was estimated based on the proportion of individuals who have a causative genotype in a general population database. In addition, clinical severity for causative genotypes was assessed based on currently available locus-specific databases (LSDBs), which contain information on both genotype and clinical severity. Methods Genetic variants in the GAA gene in the Genome Aggregation Database (gnomAD) (v2.1.1) were analyzed in combination with LSDBs of ClinVar, ClinGen Evidence Repository, Pompe disease GAA variant database, and the Pompe Registry. Carrier frequency (CF) and predicted genetic prevalence (pGP) were estimated. Results Of 7 populations, East Asian and African showed higher proportions of pathogenic or likely pathogenic variants (PLPVs) associated with classic infantile-onset Pompe disease. Total CF and pGP in the overall population were 1.3% (1 in 77) and 1:23,232, respectively. The highest pGP was observed in the East Asian population at 1:12,125, followed by Non-Finnish European (1:13,756), Ashkenazi Jewish (1:22,851), African/African-American (1:26,560), Latino/Admixed American (1:57,620), South Asian (1:93,087), and Finnish (1:1,056,444). Conclusions Pompe disease has a higher pGP (1:23,232) than earlier accepted (1:40,000). The pGP for Pompe disease was expectedly wide by population and consistent with previous reports based on newborn screening programs (approximately 1:10,000-1:30,000).
Collapse
Affiliation(s)
- Kyung Sun Park
- Department of Laboratory Medicine, Kyung Hee University School of Medicine and Kyung Hee University Medical Center, Seoul, Republic of Korea
| |
Collapse
|
31
|
Li C, Desai AK, Gupta P, Dempsey K, Bhambhani V, Hopkin RJ, Ficicioglu C, Tanpaiboon P, Craigen WJ, Rosenberg AS, Kishnani PS. Transforming the clinical outcome in CRIM-negative infantile Pompe disease identified via newborn screening: the benefits of early treatment with enzyme replacement therapy and immune tolerance induction. Genet Med 2021; 23:845-855. [PMID: 33495531 PMCID: PMC8107133 DOI: 10.1038/s41436-020-01080-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/30/2022] Open
Abstract
Purpose: To assess the magnitude of benefit to early treatment initiation, enabled by newborn screening or prenatal diagnosis, in patients with cross-reactive immunological material (CRIM)-negative infantile Pompe disease (IPD), treated with enzyme replacement therapy (ERT) and prophylactic immune tolerance induction (ITI) with rituximab, methotrexate, and IVIG. Methods: A total of 41 CRIM-negative IPD patients were evaluated. Amongst patients who were treated with ERT+ITI (n=30), those who were invasive ventilator-free at baseline and had ≥6 months of follow-up were stratified based on age at treatment initiation: 1) early (≤4 weeks), 2) intermediate (>4 and ≤15 weeks), and 3) late (>15 weeks). A historical cohort of 11 CRIM-negative patients with IPD treated with ERT monotherapy served as an additional comparator group. Results: Twenty patients were included; five, seven, and eight in early, intermediate, and late treatment groups, respectively. Genotypes were similar across the three groups. Early-treated patients showed significant improvements in left ventricular mass index, motor and pulmonary outcomes, as well as biomarkers creatine kinase and urinary glucose tetrasaccharide, compared to those treated later. Conclusion: Our preliminary data suggest that early treatment with ERT+ITI can transform the long-term CRIM-negative IPD phenotype, which represents the most severe end of the Pompe disease spectrum.
Collapse
Affiliation(s)
- Cindy Li
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Ankit K Desai
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Punita Gupta
- St. Joseph's University Hospital, Paterson, NJ, USA
| | - Katherine Dempsey
- Center for Human Genetics and Department of Genetics and Genome Sciences, University Hospitals Cleveland Medical Center and Case Western Reserve University, Cleveland, OH, USA
| | - Vikas Bhambhani
- Children's Hospitals and Clinics of Minnesota, Minneapolis, MN, USA
| | - Robert J Hopkin
- Division of Medical Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Can Ficicioglu
- The Children's Hospital of Philadelphia, Division of Genetics and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Pranoot Tanpaiboon
- Division of Genetics and Metabolism, Children's National Hospital, Washington, DC, USA
| | - William J Craigen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Amy S Rosenberg
- Division of Biologics Review and Research 3, Office of Biotechnology Products, Center for Drug Evaluation and Research, US FDA, Bethesda, MD, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
32
|
Manta A, Spendiff S, Lochmüller H, Thompson R. Targeted Therapies for Metabolic Myopathies Related to Glycogen Storage and Lipid Metabolism: a Systematic Review and Steps Towards a 'Treatabolome'. J Neuromuscul Dis 2021; 8:401-417. [PMID: 33720849 PMCID: PMC8203237 DOI: 10.3233/jnd-200621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND Metabolic myopathies are a heterogenous group of muscle diseases typically characterized by exercise intolerance, myalgia and progressive muscle weakness. Effective treatments for some of these diseases are available, but while our understanding of the pathogenesis of metabolic myopathies related to glycogen storage, lipid metabolism and β-oxidation is well established, evidence linking treatments with the precise causative genetic defect is lacking. OBJECTIVE The objective of this study was to collate all published evidence on pharmacological therapies for the aforementioned metabolic myopathies and link this to the genetic mutation in a format amenable to databasing for further computational use in line with the principles of the "treatabolome" project. METHODS A systematic literature review was conducted to retrieve all levels of evidence examining the therapeutic efficacy of pharmacological treatments on metabolic myopathies related to glycogen storage and lipid metabolism. A key inclusion criterion was the availability of the genetic variant of the treated patients in order to link treatment outcome with the genetic defect. RESULTS Of the 1,085 articles initially identified, 268 full-text articles were assessed for eligibility, of which 87 were carried over into the final data extraction. The most studied metabolic myopathies were Pompe disease (45 articles), multiple acyl-CoA dehydrogenase deficiency related to mutations in the ETFDH gene (15 articles) and systemic primary carnitine deficiency (8 articles). The most studied therapeutic management strategies for these diseases were enzyme replacement therapy, riboflavin, and carnitine supplementation, respectively. CONCLUSIONS This systematic review provides evidence for treatments of metabolic myopathies linked with the genetic defect in a computationally accessible format suitable for databasing in the treatabolome system, which will enable clinicians to acquire evidence on appropriate therapeutic options for their patient at the time of diagnosis.
Collapse
Affiliation(s)
- A. Manta
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - S. Spendiff
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - H. Lochmüller
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Neuropediatrics and Muscle Disorders, Medical Center –University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
- Division of Neurology, Department of Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - R. Thompson
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| |
Collapse
|
33
|
Newborn Screening for Pompe Disease: Pennsylvania Experience. Int J Neonatal Screen 2020; 6:ijns6040089. [PMID: 33202836 PMCID: PMC7712483 DOI: 10.3390/ijns6040089] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Pennsylvania started newborn screening for Pompe disease in February 2016. Between February 2016 and December 2019, 531,139 newborns were screened. Alpha-Glucosidase (GAA) enzyme activity is measured by flow-injection tandem mass spectrometry (FIA/MS/MS) and full sequencing of the GAA gene is performed as a second-tier test in all newborns with low GAA enzyme activity [<2.10 micromole/L/h]. A total of 115 newborns had low GAA enzyme activity and abnormal genetic testing and were referred to metabolic centers. Two newborns were diagnosed with Infantile Onset Pompe Disease (IOPD), and 31 newborns were confirmed to have Late Onset Pompe Disease (LOPD). The incidence of IOPD + LOPD was 1:16,095. A total of 30 patients were compound heterozygous for one pathogenic and one variant of unknown significance (VUS) mutation or two VUS mutations and were defined as suspected LOPD. The incidence of IOPD + LOPD + suspected LOPD was 1: 8431 in PA. We also found 35 carriers, 15 pseudodeficiency carriers, and 2 false positive newborns.
Collapse
|
34
|
Gupta P, Shayota BJ, Desai AK, Kiblawi F, Myridakis D, Messina J, Tah P, Tambini-King L, Kishnani PS. A Race Against Time-Changing the Natural History of CRIM Negative Infantile Pompe Disease. Front Immunol 2020; 11:1929. [PMID: 33013846 PMCID: PMC7498628 DOI: 10.3389/fimmu.2020.01929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 07/17/2020] [Indexed: 01/19/2023] Open
Abstract
We report the clinical course of the first prenatally diagnosed cross-reactive immunologic material (CRIM)-negative infantile Pompe disease (IPD) patient [homozygous for c.2560C>T (p.Arg854X) variant in the GAA gene] to undergo prophylactic immune tolerance induction (ITI) and enzyme replacement therapy (ERT) within the first 2 days of life. Both parents were found to be carriers of the c.2560C>T (p.Arg854X) variant through prenatal carrier screening. Fetal echocardiogram at 31 weeks of gestation showed left ventricular hypertrophy. An echocardiogram on the 1st day of life revealed marked biventricular hypertrophy. Physical exam was significant for macroglossia and hypotonia. A short course of Prophylactic ITI with rituximab, methotrexate, and intravenous immunoglobulin (IVIG) in conjunction with ERT at a dose of 20 mg/kg every other week was started on day 2 of life. The patient completed the ITI protocol safely and complete B-cell recovery, based on CD19 count, was noted by 3 months of age. The patient never developed anti-rhGAA IgG antibodies to ERT. Vaccinations were initiated at 9 months of age, with adequate response noted. Complete recovery of cardiac function and left ventricular mass was seen by 11 weeks of age. At 8 months of age, the patient developmentally measured at 75–90% on the Alberta Infant Motor Scale, walked at 11 months and continues to develop age-appropriately at 50 months of age based on the Early Learning Accomplishment Profile. ERT dosing was increased to 40 mg/kg every 2 weeks at 32 months of age and frequency increased to 40 mg/kg every week at 47 months of age. Patient continues to have undetectable antibody titers, most recently at age 50 months and urine Hex4 has remained normal. To our knowledge, this is the first report of successful early ERT and ITI in a prenatally diagnosed CRIM-negative IPD patient and the youngest IPD patient to receive ITI safely. With the addition of Pompe disease to the Recommended Uniform Screening Panel(RUSP) and its addition to multiple state newborn screening programs, our case highlights the benefits of early diagnosis and timely initiation of treatment in babies with Pompe disease, who represent the most severe end of the disease spectrum.
Collapse
Affiliation(s)
- Punita Gupta
- St. Joseph's University Hospital, Paterson, NJ, United States
| | - Brian J Shayota
- Texas Children's Hospital, Balor College of Medicine, Houston, TX, United States
| | - Ankit K Desai
- Duke University Medical Center, Durham, NC, United States
| | - Fuad Kiblawi
- St. Joseph's University Hospital, Paterson, NJ, United States
| | | | - John Messina
- St. Joseph's University Hospital, Paterson, NJ, United States
| | - Peter Tah
- St. Joseph's University Hospital, Paterson, NJ, United States
| | | | | |
Collapse
|
35
|
Ames EG, Fisher R, Kleyn M, Ahmad A. Current Practices for U.S. Newborn Screening of Pompe Disease and MPSI. Int J Neonatal Screen 2020; 6:ijns6030072. [PMID: 33239598 PMCID: PMC7569971 DOI: 10.3390/ijns6030072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 11/16/2022] Open
Abstract
Two lysosomal storage disorders (LSDs), Pompe disease and Mucopolysaccharidosis type I (MPSI) were added to the Recommended Uniform Screening Panel (RUSP) for newborn screening (NBS) in 2015 and 2016, respectively. These conditions are being screened with variable practice in terms of primary and reflex analytes (either biochemical or molecular testing) as well as collection of short- and long-term follow-up elements. The goal of this study is to evaluate practices of state health departments in regards to screening methods and follow-up data collected. We conducted online surveys and phone questionnaires to determine each U.S. state's practices for screening and follow-up of positive newborn screens. We report the first snapshot of practices for NBS for the LSDs included on the RUSP. All 50 U.S. states responded to our survey. The majority of U.S. states are not currently screening for Pompe disease and MPSI as of March 2020, but this number will increase to 38 states in the coming 1-3 years based on survey results. Our survey identifies data elements used by state health departments for short-and long-term follow-up that could serve as the basis of common elements for larger, public health-based analyses of the benefits and efficacy of screening for Pompe disease and MPSI.
Collapse
Affiliation(s)
- Elizabeth G. Ames
- Division of Pediatric Genetics, Metabolism and Genomic Medicine, Department of Pediatrics, University of Michigan Health System, D5240 Medical Professional Building, 1500 E. Medical Center Dr, Ann Arbor, MI 48109, USA; (R.F.); (A.A.)
- Correspondence: ; Tel.: +1-(734)-764-0579
| | - Rachel Fisher
- Division of Pediatric Genetics, Metabolism and Genomic Medicine, Department of Pediatrics, University of Michigan Health System, D5240 Medical Professional Building, 1500 E. Medical Center Dr, Ann Arbor, MI 48109, USA; (R.F.); (A.A.)
| | - Mary Kleyn
- Newborn Screening Follow-up Section, Michigan Department of Health and Human Services, 333 South Grand Avenue, Lansing, MI 48933, USA;
| | - Ayesha Ahmad
- Division of Pediatric Genetics, Metabolism and Genomic Medicine, Department of Pediatrics, University of Michigan Health System, D5240 Medical Professional Building, 1500 E. Medical Center Dr, Ann Arbor, MI 48109, USA; (R.F.); (A.A.)
| |
Collapse
|
36
|
Burton BK, Hickey R, Hitchins L. Newborn Screening for Mucopolysaccharidosis Type II in Illinois: An Update. Int J Neonatal Screen 2020; 6:73. [PMID: 33117908 PMCID: PMC7569764 DOI: 10.3390/ijns6030073] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/01/2020] [Indexed: 11/16/2022] Open
Abstract
Mucopolysaccharidosis type II (MPS II, Hunter syndrome) is a rare, progressive multisystemic lysosomal storage disorder with significant morbidity and premature mortality. Infants with MPS II develop signs and symptoms of the disorder in the early years of life, yet diagnostic delays are very common. Enzyme replacement therapy is an effective treatment option. It has been shown to prolong survival and improve or stabilize many somatic manifestations of the disorder. Our initial experience with newborn screening in 162,000 infants was previously reported. Here, we update that experience with the findings in 339,269 infants. Measurement of iduronate-2-sulfatase (I2S) activity was performed on dried blood spot samples submitted for other newborn screening disorders. A positive screen was defined as I2S activity less than or equal to 10% of the daily median. In this series, 28 infants had a positive screening test result, and four other infants had a borderline result. Three positive diagnoses of MPS II were established, and 25 were diagnosed as having I2S pseudodeficiency. The natural history and the clinical features of MPS II make it an ideal target for newborn screening. Newborn screening was effective in identifying affected infants in our population with an acceptable rate of false positive results.
Collapse
Affiliation(s)
- Barbara K Burton
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; (R.H.); (L.H.)
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rachel Hickey
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; (R.H.); (L.H.)
| | - Lauren Hitchins
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; (R.H.); (L.H.)
| |
Collapse
|
37
|
Desai AK, Baloh CH, Sleasman JW, Rosenberg AS, Kishnani PS. Benefits of Prophylactic Short-Course Immune Tolerance Induction in Patients With Infantile Pompe Disease: Demonstration of Long-Term Safety and Efficacy in an Expanded Cohort. Front Immunol 2020; 11:1727. [PMID: 32849613 PMCID: PMC7424004 DOI: 10.3389/fimmu.2020.01727] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/29/2020] [Indexed: 01/19/2023] Open
Abstract
Immune tolerance induction (ITI) with a short-course of rituximab, methotrexate, and/or IVIG in the enzyme replacement therapy (ERT)-naïve setting has prolonged survival and improved clinical outcomes in patients with infantile Pompe disease (IPD) lacking endogenous acid-alpha glucosidase (GAA), known as cross-reactive immunologic material (CRIM)-negative. In the context of cancer therapy, rituximab administration results in sustained B-cell depletion in 83% of patients for up to 26–39 weeks with B-cell reconstitution beginning at approximately 26 weeks post-treatment. The impact of rituximab on serum immunoglobulin levels is not well studied, available data suggest that rituximab can cause persistently low immunoglobulin levels and adversely impact vaccine responses. Data on a cohort of IPD patients who received a short-course of ITI with rituximab, methotrexate, and IVIG in the ERT-naïve setting and had ≥6 months of follow-up were retrospectively studied. B-cell quantitation, ANC, AST, ALT, immunization history, and vaccine titers after B-cell reconstitution were reviewed. Data were collected for 34 IPD patients (25 CRIM-negative and 9 CRIM-positive) with a median age at ERT initiation of 3.5 months (0.1–11.0 months). B-cell reconstitution, as measured by normalization of CD19%, was seen in all patients (n = 33) at a median time of 17 weeks range (11–55 weeks) post-rituximab. All maintained normal CD19% with the longest follow-up being 248 weeks post-rituximab. 30/34 (88%) maintained negative/low anti-rhGAA antibody titers, even with complete B-cell reconstitution. Infections during immunosuppression were reported in five CRIM-negative IPD patients, all resolved satisfactorily on antibiotics. There were no serious sequelae or deaths. Of the 31 evaluable patients, 27 were up to date on age-appropriate immunizations. Vaccine titers were available for 12 patients after B-cell reconstitution and adequate humoral response was observed in all except an inadequate response to the Pneumococcal vaccine (n = 2). These data show the benefits of short-course prophylactic ITI in IPD both in terms of safety and efficacy. Data presented here are from the youngest cohort of patients treated with rituximab and expands the evidence of its safety in the pediatric population.
Collapse
Affiliation(s)
- Ankit K Desai
- Division of Medical Genetics, Department of Pediatrics, Duke University Health System, Durham, NC, United States
| | - Carolyn H Baloh
- Division of Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Duke University Health System, Durham, NC, United States
| | - John W Sleasman
- Division of Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Duke University Health System, Durham, NC, United States
| | - Amy S Rosenberg
- Division of Biologics Review and Research 3, Office of Biotechnology Products, Center for Drug Evaluation and Research, US FDA, Bethesda, MD, United States
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Health System, Durham, NC, United States
| |
Collapse
|
38
|
Abstract
Glycogen storage disease type II (also known as Pompe disease (PD)) is an autosomal recessive disorder caused by defects in α-glucosidase (AαGlu), resulting in lysosomal glycogen accumulation in skeletal and heart muscles. Accumulation and tissue damage rates depend on residual enzyme activity. Enzyme replacement therapy (ERT) should be started before symptoms are apparent in order to achieve optimal outcomes. Early initiation of ERT in infantile-onset PD improves survival, reduces the need for ventilation, results in earlier independent walking, and enhances patient quality of life. Newborn screening (NBS) is the optimal approach for early diagnosis and treatment of PD. In NBS for PD, measurement of AαGlu enzyme activity in dried blood spots (DBSs) is conducted using fluorometry, tandem mass spectrometry, or digital microfluidic fluorometry. The presence of pseudodeficiency alleles, which are frequent in Asian populations, interferes with NBS for PD, and current NBS systems cannot discriminate between pseudodeficiency and cases with PD or potential PD. The combination of GAA gene analysis with NBS is essential for definitive diagnoses of PD. In this review, we introduce our experiences and discuss NBS programs for PD implemented in various countries.
Collapse
|
39
|
Sanders KA, Gavrilov DK, Oglesbee D, Raymond KM, Tortorelli S, Hopwood JJ, Lorey F, Majumdar R, Kroll CA, McDonald AM, Lacey JM, Turgeon CT, Tucker JN, Tang H, Currier R, Isaya G, Rinaldo P, Matern D. A Comparative Effectiveness Study of Newborn Screening Methods for Four Lysosomal Storage Disorders. Int J Neonatal Screen 2020; 6:44. [PMID: 32802993 PMCID: PMC7423013 DOI: 10.3390/ijns6020044] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/27/2020] [Indexed: 01/13/2023] Open
Abstract
Newborn screening for one or more lysosomal disorders has been implemented in several US states, Japan and Taiwan by multiplexed enzyme assays using either tandem mass spectrometry or digital microfluidics. Another multiplex assay making use of immunocapture technology has also been proposed. To investigate the potential variability in performance of these analytical approaches, we implemented three high-throughput screening assays for the simultaneous screening for four lysosomal disorders: Fabry disease, Gaucher disease, mucopolysaccharidosis type I, and Pompe disease. These assays were tested in a prospective comparative effectiveness study using nearly 100,000 residual newborn dried blood spot specimens. In addition, 2nd tier enzyme assays and confirmatory molecular genetic testing were employed. Post-analytical interpretive tools were created using the software Collaborative Laboratory Integrated Reports (CLIR) to determine its ability to improve the performance of each assay vs. the traditional result interpretation based on analyte-specific reference ranges and cutoffs. This study showed that all three platforms have high sensitivity, and the application of CLIR tools markedly improves the performance of each platform while reducing the need for 2nd tier testing by 66% to 95%. Moreover, the addition of disease-specific biochemical 2nd tier tests ensures the lowest false positive rates and the highest positive predictive values for any platform.
Collapse
Affiliation(s)
- Karen A. Sanders
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Dimitar K. Gavrilov
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Devin Oglesbee
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Kimiyo M. Raymond
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Silvia Tortorelli
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - John J. Hopwood
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (J.J.H.); (J.N.T.)
| | - Fred Lorey
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA 94804, USA; (F.L.); (H.T.); (R.C.)
| | - Ramanath Majumdar
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Charles A. Kroll
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Amber M. McDonald
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Jean M. Lacey
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Coleman T. Turgeon
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Justin N. Tucker
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (J.J.H.); (J.N.T.)
| | - Hao Tang
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA 94804, USA; (F.L.); (H.T.); (R.C.)
| | - Robert Currier
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA 94804, USA; (F.L.); (H.T.); (R.C.)
- Department of Pediatrics, University of California, San Francisco, CA 94143, USA
| | - Grazia Isaya
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Piero Rinaldo
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Dietrich Matern
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| |
Collapse
|
40
|
Cagin U, Puzzo F, Gomez MJ, Moya-Nilges M, Sellier P, Abad C, Van Wittenberghe L, Daniele N, Guerchet N, Gjata B, Collaud F, Charles S, Sola MS, Boyer O, Krijnse-Locker J, Ronzitti G, Colella P, Mingozzi F. Rescue of Advanced Pompe Disease in Mice with Hepatic Expression of Secretable Acid α-Glucosidase. Mol Ther 2020; 28:2056-2072. [PMID: 32526204 PMCID: PMC7474269 DOI: 10.1016/j.ymthe.2020.05.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/15/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Pompe disease is a neuromuscular disorder caused by disease-associated variants in the gene encoding for the lysosomal enzyme acid α-glucosidase (GAA), which converts lysosomal glycogen to glucose. We previously reported full rescue of Pompe disease in symptomatic 4-month-old Gaa knockout (Gaa−/−) mice by adeno-associated virus (AAV) vector-mediated liver gene transfer of an engineered secretable form of GAA (secGAA). Here, we showed that hepatic expression of secGAA rescues the phenotype of 4-month-old Gaa−/− mice at vector doses at which the native form of GAA has little to no therapeutic effect. Based on these results, we then treated severely affected 9-month-old Gaa−/− mice with an AAV vector expressing secGAA and followed the animals for 9 months thereafter. AAV-treated Gaa−/− mice showed complete reversal of the Pompe phenotype, with rescue of glycogen accumulation in most tissues, including the central nervous system, and normalization of muscle strength. Transcriptomic profiling of skeletal muscle showed rescue of most altered pathways, including those involved in mitochondrial defects, a finding supported by structural and biochemical analyses, which also showed restoration of lysosomal function. Together, these results provide insight into the reversibility of advanced Pompe disease in the Gaa−/− mouse model via liver gene transfer of secGAA.
Collapse
Affiliation(s)
- Umut Cagin
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Francesco Puzzo
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France; Sorbonne Université, Paris, France
| | - Manuel Jose Gomez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | | | - Pauline Sellier
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Catalina Abad
- Université de Rouen Normandie-IRIB, 76183 Rouen, France
| | | | - Nathalie Daniele
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Nicolas Guerchet
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Bernard Gjata
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Fanny Collaud
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Severine Charles
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Marcelo Simon Sola
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Olivier Boyer
- Université de Rouen Normandie-IRIB, 76183 Rouen, France
| | | | - Giuseppe Ronzitti
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Pasqualina Colella
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Federico Mingozzi
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France; Sorbonne Université, Paris, France; Spark Therapeutics, Philadelphia, PA 19103, USA.
| |
Collapse
|
41
|
Saich R, Brown R, Collicoat M, Jenner C, Primmer J, Clancy B, Holland T, Krinks S. Is Newborn Screening the Ultimate Strategy to Reduce Diagnostic Delays in Pompe Disease? The Parent and Patient Perspective. Int J Neonatal Screen 2020; 6:1. [PMID: 33073001 PMCID: PMC7422966 DOI: 10.3390/ijns6010001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023] Open
Abstract
Pompe disease (PD) is a rare, autosomal-recessively inherited deficiency in the enzyme acid α-glucosidase. It is a spectrum disorder; age at symptom onset and rate of deterioration can vary considerably. In affected infants prognosis is poor, such that without treatment most infants die within the first year of life. To lose a baby in their first year of life to a rare disease causes much regret, guilt, and loneliness to parents, family, and friends. To lose a baby needlessly when there is an effective treatment amplifies this sadness. With so little experience of rare disease in the community, once a baby transfers to their home they are subject to a very uncertain and unyielding diagnostic journey while their symptomology progresses and their health deteriorates. With a rare disease like PD, the best opportunity to diagnose a baby is at birth. PD is not yet included in the current newborn screening (NBS) panel in Australia. Should it be? In late 2018 the Australian Pompe Association applied to the Australian Standing committee on Newborn Screening to have PD included. The application was not upheld. Here we provide an overview of the rationale for NBS, drawing on the scientific literature and perspectives from The Australian Pompe Association, its patients and their families. In doing so, we hope to bring a new voice to this very important debate.
Collapse
Affiliation(s)
- Raymond Saich
- Australian Pompe Association Inc., Kellyville, NSW 2155, Australia
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Klug TL, Swartz LB, Washburn J, Brannen C, Kiesling JL. Lessons Learned from Pompe Disease Newborn Screening and Follow-up. Int J Neonatal Screen 2020; 6:11. [PMID: 33073009 PMCID: PMC7422965 DOI: 10.3390/ijns6010011] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/11/2020] [Indexed: 11/24/2022] Open
Abstract
In 2015, Pompe disease became the first lysosomal storage disorder to be recommended for universal newborn screening by the Secretary of the U.S. Department of Health and Human Services. Newborn screening for Pompe has been implemented in 20 states and several countries across the world. The rates of later-onset disease phenotypes for Pompe and pseudodeficiency alleles are higher than initially anticipated, and these factors must be considered during Pompe disease newborn screening. This report presents an overview of six years of data from the Missouri State Public Health Laboratory for Pompe disease newborn screening and follow-up.
Collapse
Affiliation(s)
- Tracy L. Klug
- Missouri Department of Health and Senior Services, P.O. Box 570, Jefferson City, MO 65102-0570, USA
| | - Lori B. Swartz
- Missouri Department of Health and Senior Services, P.O. Box 570, Jefferson City, MO 65102-0570, USA
| | - Jon Washburn
- Baebies, Inc., P.O. Box 14403, Durham, NC 27709, USA
| | | | - Jami L. Kiesling
- Missouri Department of Health and Senior Services, P.O. Box 570, Jefferson City, MO 65102-0570, USA
| |
Collapse
|
43
|
Burton BK, Charrow J, Hoganson GE, Fleischer J, Grange DK, Braddock SR, Hitchins L, Hickey R, Christensen KM, Groepper D, Shryock H, Smith P, Shao R, Basheeruddin K. Newborn Screening for Pompe Disease in Illinois: Experience with 684,290 Infants. Int J Neonatal Screen 2020; 6:4. [PMID: 33073003 PMCID: PMC7422983 DOI: 10.3390/ijns6010004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/18/2020] [Indexed: 11/16/2022] Open
Abstract
Statewide newborn screening for Pompe disease began in Illinois in 2015. As of 30 September 2019, a total of 684,290 infants had been screened and 395 infants (0.06%) were screen positive. A total of 29 cases of Pompe disease were identified (3 infantile, 26 late-onset). While many of the remainder were found to have normal alpha-glucosidase activity on the follow-up testing (234 of 395), other findings included 62 carriers, 39 infants with pseudodeficiency, and eight infants who could not be given a definitive diagnosis due to inconclusive follow-up testing.
Collapse
Affiliation(s)
- Barbara K Burton
- Department of Pediatrics, Feinberg School of Medicine of Northwestern University, Chicago, IL 60611, USA;
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; (L.H.); (R.H.)
| | - Joel Charrow
- Department of Pediatrics, Feinberg School of Medicine of Northwestern University, Chicago, IL 60611, USA;
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; (L.H.); (R.H.)
| | - George E Hoganson
- Department of Pediatrics, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| | - Julie Fleischer
- Department of Pediatrics, Southern Illinois University School of Medicine, Springfield, IL 62701, USA; (J.F.); (D.G.)
| | - Dorothy K Grange
- Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, St. Louis, MO 63110, USA;
| | - Stephen R Braddock
- Department of Pediatrics, Saint Louis University, St. Louis, MO 63104, USA; (S.R.B.); (K.M.C.)
| | - Lauren Hitchins
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; (L.H.); (R.H.)
| | - Rachel Hickey
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; (L.H.); (R.H.)
| | - Katherine M Christensen
- Department of Pediatrics, Saint Louis University, St. Louis, MO 63104, USA; (S.R.B.); (K.M.C.)
| | - Daniel Groepper
- Department of Pediatrics, Southern Illinois University School of Medicine, Springfield, IL 62701, USA; (J.F.); (D.G.)
| | - Heather Shryock
- Office of Health Promotion, Illinois Department of Public Health, Springfield, IL 62761, USA; (H.S.); (P.S.)
| | - Pamela Smith
- Office of Health Promotion, Illinois Department of Public Health, Springfield, IL 62761, USA; (H.S.); (P.S.)
| | - Rong Shao
- Newborn Screening Laboratory, Illinois Department of Public Health, Chicago, IL 60603, USA; (R.S.); (K.B.)
| | - Khaja Basheeruddin
- Newborn Screening Laboratory, Illinois Department of Public Health, Chicago, IL 60603, USA; (R.S.); (K.B.)
| |
Collapse
|
44
|
Tang H, Feuchtbaum L, Sciortino S, Matteson J, Mathur D, Bishop T, Olney RS. The First Year Experience of Newborn Screening for Pompe Disease in California. Int J Neonatal Screen 2020; 6:9. [PMID: 33073007 PMCID: PMC7422988 DOI: 10.3390/ijns6010009] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/05/2020] [Indexed: 12/22/2022] Open
Abstract
The California Department of Public Health started universal newborn screening for Pompe disease in August 2018 with a two-tier process including: (1) acid alpha-glucosidase (GAA) enzyme activity assay followed by, (2) GAA gene sequencing analysis. This study examines results from the first year of screening in a large and diverse screening population. With 453,152 screened newborns, the birth prevalence and GAA enzyme activity associated with various types of Pompe disease classifications are described. The frequency of GAA gene mutations and allele variants are reported. Of 88 screen positives, 18 newborns were resolved as Pompe disease, including 2 classic infantile-onset and 16 suspected late-onset form. The c.-32-13T>G variant was the most common pathogenic mutation reported. African American and Asian/Pacific Islander newborns had higher allele frequencies for both pathogenic and pseudodeficiency variants. After the first year of Pompe disease screening in California, the disease distribution in the population is now better understood. With the ongoing long-term follow-up system currently in place, our understanding of the complex genotype-phenotype relationships will become more evident in the future, and this should help us better understand the clinical significance of identified cases.
Collapse
Affiliation(s)
- Hao Tang
- Genetic Disease Screening Program, California Department of Public Health, 850 Marina Bay Parkway, MS 8200, USA; (L.F.); (S.S.); (J.M.); (D.M.); (T.B.); (R.S.O.)
| | - Lisa Feuchtbaum
- Genetic Disease Screening Program, California Department of Public Health, 850 Marina Bay Parkway, MS 8200, USA; (L.F.); (S.S.); (J.M.); (D.M.); (T.B.); (R.S.O.)
| | - Stanley Sciortino
- Genetic Disease Screening Program, California Department of Public Health, 850 Marina Bay Parkway, MS 8200, USA; (L.F.); (S.S.); (J.M.); (D.M.); (T.B.); (R.S.O.)
| | - Jamie Matteson
- Genetic Disease Screening Program, California Department of Public Health, 850 Marina Bay Parkway, MS 8200, USA; (L.F.); (S.S.); (J.M.); (D.M.); (T.B.); (R.S.O.)
| | - Deepika Mathur
- Genetic Disease Screening Program, California Department of Public Health, 850 Marina Bay Parkway, MS 8200, USA; (L.F.); (S.S.); (J.M.); (D.M.); (T.B.); (R.S.O.)
| | - Tracey Bishop
- Genetic Disease Screening Program, California Department of Public Health, 850 Marina Bay Parkway, MS 8200, USA; (L.F.); (S.S.); (J.M.); (D.M.); (T.B.); (R.S.O.)
| | - Richard S Olney
- Genetic Disease Screening Program, California Department of Public Health, 850 Marina Bay Parkway, MS 8200, USA; (L.F.); (S.S.); (J.M.); (D.M.); (T.B.); (R.S.O.)
| |
Collapse
|
45
|
Yang CF, Niu DM, Tai SK, Wang TH, Su HT, Huang LY, Soong WJ. Airway abnormalities in very early treated infantile-onset Pompe disease: A large-scale survey by flexible bronchoscopy. Am J Med Genet A 2020; 182:721-729. [PMID: 31953985 DOI: 10.1002/ajmg.a.61481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 12/08/2019] [Accepted: 12/16/2019] [Indexed: 11/08/2022]
Abstract
Early enzyme replacement therapy (ERT) improve long-term outcomes in patients with infantile-onset Pompe disease (IOPD). Our cohort of patients with IOPD at Taipei Veterans General Hospital (TVGH) joined Taiwan Pompe newborn screening program from 2008, testing more than one million newborns until 2018. By 2010, we had established rapid diagnostic strategies. Now, the average age of ERT initiation starts at an average age of <10 days-old, the earliest group in the world. However, they still presented some airway problems. We present a retrospective study focused on airway abnormalities in these patients along 8 years of observation. Fifteen patients with IOPD, who received very early treatment at a mean age of 8.94 ± 3.75 days, underwent flexible bronchoscopy (FB) for dynamic assessment of the whole airway. Long-term clinical outcomes and relevant symptoms of the upper airway were assessed. All patients in the study had varying degrees of severity of upper airway abnormalities and speech disorders. The three oldest children (Age 94, 93, and 88 months, respectively) had poor movement of the vocal cords with reduced abduction and adduction and had silent aspiration of saliva through the glottis during respiration. This is the largest cohort study presented to date about airway abnormalities in very early treated patients with IOPD patients by FB. Despite very early treatment, we observed upper airway abnormalities in these IOPD patients. In IOPD, upper airway abnormalities seem inevitable over time. We suggest early and continuous monitoring for all IOPD patients, even with early and regular treatment.
Collapse
Affiliation(s)
- Chia-Feng Yang
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Dau-Ming Niu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shyh-Kuan Tai
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Otolaryngology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ting-Hao Wang
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hsiao-Ting Su
- Department of Audiology and Speech Language Pathology, Mackay Medical College, Taipei, Taiwan
| | - Ling-Yi Huang
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen-Jue Soong
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Children's Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
46
|
Burton BK, Hoganson GE, Fleischer J, Grange DK, Braddock SR, Hickey R, Hitchins L, Groepper D, Christensen KM, Kirby A, Moody C, Shryock H, Ashbaugh L, Shao R, Basheeruddin K. Population-Based Newborn Screening for Mucopolysaccharidosis Type II in Illinois: The First Year Experience. J Pediatr 2019; 214:165-167.e1. [PMID: 31477379 DOI: 10.1016/j.jpeds.2019.07.053] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/28/2019] [Accepted: 07/23/2019] [Indexed: 01/24/2023]
Abstract
OBJECTIVES To assess the outcome of population-based newborn screening for mucopolysaccharidosis type II (MPS II) during the first year of screening in Illinois. STUDY DESIGN Tandem mass spectrometry was used to measure iduronate-2-sulfatase (I2S) activity in dried blood spot specimens obtained from 162 000 infant samples sent to the Newborn Screening Laboratory of the Illinois Department of Public Health in Chicago. RESULTS One case of MPS II and 14 infants with pseudodeficiency for I2S were identified. CONCLUSIONS Newborn screening for MPS II by measurement of I2S enzyme activity was successfully integrated into the statewide newborn screening program in Illinois.
Collapse
Affiliation(s)
- Barbara K Burton
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL; Northwestern University Feinberg School of Medicine, Chicago, IL.
| | | | | | | | - Stephen R Braddock
- Saint Louis University, St. Louis, MO; SSM Health Cardinal Glennon Children's Medical Center, St. Louis, MO
| | - Rachel Hickey
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - Lauren Hitchins
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL; Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Katherine M Christensen
- Saint Louis University, St. Louis, MO; SSM Health Cardinal Glennon Children's Medical Center, St. Louis, MO
| | - Amelia Kirby
- Saint Louis University, St. Louis, MO; SSM Health Cardinal Glennon Children's Medical Center, St. Louis, MO
| | - Conny Moody
- Office of Health Promotion, Illinois Department of Public Health, Springfield, IL
| | - Heather Shryock
- Office of Health Promotion, Illinois Department of Public Health, Springfield, IL
| | - Laura Ashbaugh
- Office of Health Promotion, Illinois Department of Public Health, Springfield, IL
| | - Rong Shao
- Newborn Screening Laboratory, Illinois Department of Public Health, Chicago, IL
| | - Khaja Basheeruddin
- Newborn Screening Laboratory, Illinois Department of Public Health, Chicago, IL
| |
Collapse
|
47
|
Ngiwsara L, Wattanasirichaigoon D, Tim-Aroon T, Rojnueangnit K, Noojaroen S, Khongkraparn A, Sawangareetrakul P, Ketudat-Cairns JR, Charoenwattanasatien R, Champattanachai V, Kuptanon C, Pangkanon S, Svasti J. Clinical course, mutations and its functional characteristics of infantile-onset Pompe disease in Thailand. BMC MEDICAL GENETICS 2019; 20:156. [PMID: 31510962 PMCID: PMC6737665 DOI: 10.1186/s12881-019-0878-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 08/21/2019] [Indexed: 11/18/2022]
Abstract
Background Pompe disease is a lysosomal storage disorder caused by the deficiency of acid alpha-glucosidase (EC. 3.2.1.20) due to mutations in human GAA gene. The objective of the present study was to examine clinical and molecular characteristics of infantile-onset Pompe disease (IOPD) in Thailand. Methods Twelve patients with infantile-onset Pompe disease (IOPD) including 10 Thai and two other Asian ethnicities were enrolled. To examine the molecular characteristics of Pompe patients, GAA gene was analyzed by PCR amplification and direct Sanger-sequencing of 20 exons coding region. The novel mutations were transiently transfected in COS-7 cells for functional verification. The severity of the mutation was rated by study of the GAA enzyme activity detected in transfected cells and culture media, as well as the quantity and quality of the proper sized GAA protein demonstrated by western blot analysis. The GAA three dimensional structures were visualized by PyMol software tool. Results All patients had hypertrophic cardiomyopathy, generalized muscle weakness, and undetectable or < 1% of GAA normal activity. Three patients received enzyme replacement therapy with variable outcome depending on the age of the start of enzyme replacement therapy (ERT). Seventeen pathogenic mutations including four novel variants: c.876C > G (p.Tyr292X), c.1226insG (p.Asp409GlyfsX95), c.1538G > A (p.Asp513Gly), c.1895 T > G (p.Leu632Arg), and a previously reported rare allele of unknown significance: c.781G > A (p.Ala261Thr) were identified. The rating system ranked p.Tyr292X, p. Asp513Gly and p. Leu632Arg as class “B” and p. Ala261Thr as class “D” or “E”. These novel mutations were located in the N-terminal beta-sheet domain and the catalytic domain. Conclusions The present study provides useful information on the mutations of GAA gene in the underrepresented population of Asia which are more diverse than previously described and showing the hotspots in exons 14 and 5, accounting for 62% of mutant alleles. Almost all mutations identified are in class A/B. These data can benefit rapid molecular diagnosis of IOPD and severity rating of the mutations can serve as a partial substitute for cross reactive immunological material (CRIM) study.
Collapse
Affiliation(s)
- Lukana Ngiwsara
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, Thailand
| | - Duangrurdee Wattanasirichaigoon
- Division of Medical Genetics, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| | - Thipwimol Tim-Aroon
- Division of Medical Genetics, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kitiwan Rojnueangnit
- Pediatrics Department, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Saisuda Noojaroen
- Division of Medical Genetics, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Arthaporn Khongkraparn
- Division of Medical Genetics, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - James R Ketudat-Cairns
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, Thailand.,School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Ratana Charoenwattanasatien
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, Thailand.,Current address: Synchrotron Light Research Institute, Nakhon Ratchasima, Thailand
| | | | | | | | - Jisnuson Svasti
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, Thailand
| |
Collapse
|
48
|
Abstract
Pompe disease (PD) is caused by the deficiency of the lysosomal enzyme acid α-glucosidase (GAA), resulting in systemic pathological glycogen accumulation. PD can present with cardiac, skeletal muscle, and central nervous system manifestations, as a continuum of phenotypes among two main forms: classical infantile-onset PD (IOPD) and late-onset PD (LOPD). IOPD is caused by severe GAA deficiency and presents at birth with cardiac hypertrophy, muscle hypotonia, and severe respiratory impairment, leading to premature death, if not treated. LOPD is characterized by levels of residual GAA activity up to ∼20% of normal and presents both in children and adults with a varied severity of muscle weakness and motor and respiratory deficit. Enzyme replacement therapy (ERT), based on repeated intravenous (i.v.) infusions of recombinant human GAA (rhGAA), represents the only available treatment for PD. Upon more than 10 years from its launch, it is becoming evident that ERT can extend the life span of IOPD and stabilize disease progression in LOPD; however, it does not represent a cure for PD. The limited uptake of the enzyme in key affected tissues and the high immunogenicity of rhGAA are some of the hurdles that limit ERT efficacy. GAA gene transfer with adeno-associated virus (AAV) vectors has been shown to reduce glycogen storage and improve the PD phenotype in preclinical studies following different approaches. Here, we present an overview of the different gene therapy approaches for PD, focusing on in vivo gene transfer with AAV vectors and discussing the potential opportunities and challenges in developing safe and effective gene therapies for the disease. Based on emerging safety and efficacy data from clinical trials for other protein deficiencies, in vivo gene therapy with AAV vectors appears to have the potential to provide a therapeutically relevant, stable source of GAA enzyme, which could be highly beneficial in PD.
Collapse
Affiliation(s)
- Pasqualina Colella
- Genethon, Evry, France.,Department of Pediatrics, Stanford University, Stanford, California
| | - Federico Mingozzi
- Genethon, Evry, France.,Spark Therapeutics, Philadelphia, Pennsylvania
| |
Collapse
|
49
|
Infantile onset Pompe disease presenting with non-immune hydrops fetalis. Mol Genet Metab Rep 2019; 21:100503. [PMID: 31467850 PMCID: PMC6713818 DOI: 10.1016/j.ymgmr.2019.100503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 08/15/2019] [Indexed: 11/23/2022] Open
|
50
|
Xu L, Ba H, Pei Y, Huang X, Liang Y, Zhang L, Huang H, Zhang C, Tang W. Comprehensive approach to weaning in difficult-to-wean infantile and juvenile-onset glycogen-storage disease type II patients: a case series. Ital J Pediatr 2019; 45:106. [PMID: 31439017 PMCID: PMC6704633 DOI: 10.1186/s13052-019-0692-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 07/30/2019] [Indexed: 12/05/2022] Open
Abstract
Background Glycogen storage disease type II (GSD II) is caused by acid alpha-glucosidase (GAA) deficiency. Both infantile-onset and juvenile-onset GSD II lead to proximal muscle weakness and respiratory insufficiency and require mechanical ventilation. However, GSD II is also independently associated with delayed weaning from mechanical ventilation. This study aimed to describe a comprehensive approach including sequential invasive-noninvasive mechanical ventilation weaning and enzyme replacement therapy (ERT) in patients with weaning difficulties. Case presentation We studied six difficult-to-wean GSD II (three juvenile-onset, three infantile-onset) patients at the First Affiliated Hospital, Sun Yat-sen University from October 2015 to December 2017. Difficulty in weaning was defined as follows: the need for more than three spontaneous breathing trials or more than 1 week to achieve successful weaning. All patients received comprehensive treatment including sequential invasive-noninvasive mechanical ventilation weaning, ERT and general treatment. Recombinant human acid alpha-glucosidase enzyme therapy (20 mg/kg every 14 days) was used after diagnosis, and Patients 1–6 received ERT for 15.5, 4.5, 2, 2.5, 17, and 2 months, respectively. The therapeutic effect of the comprehensive treatment was observed. The patients’ respiratory function and limb muscle strength improved after each ERT session. Patients who successfully completed a spontaneous breathing trial could proceed to extubation, and then start non-invasive ventilation. The patients’ age range at initial mechanical ventilation was 3–47 (median 26.5) months, duration of invasive ventilation was 1–36 (median 2.75) months, and duration of noninvasive ventilation was 0–0.6 (median 0.05) month. The patients’ nutritional status improved after enhanced nutritional support. Patients 2, 3, and 5 were successfully weaned off the ventilator. Patient 1 underwent tracheal intubation after six weaning failures, and Patients 4 and 6 died after therapy was abandoned by their parents. Discussion and conclusions Male sex, GSD II type, and the presence of malnutrition and neurological impairment may predict poor respiratory outcomes. The above-described comprehensive sequential invasive-noninvasive mechanical ventilation weaning strategy may increase the success rate of weaning from mechanical ventilation.
Collapse
Affiliation(s)
- Lingling Xu
- Department of PICU, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Second Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Hongjun Ba
- Department of Cardiovascular pediatrics, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Second Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Yuxin Pei
- Department of PICU, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Second Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Xueqiong Huang
- Department of PICU, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Second Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Yujian Liang
- Department of PICU, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Second Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Lidan Zhang
- Department of PICU, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Second Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Huimin Huang
- Department of PICU, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Second Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Cheng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Second Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Wen Tang
- Department of PICU, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Second Road, Guangzhou, Guangdong, 510080, People's Republic of China.
| |
Collapse
|