1
|
van der Beek NAME, Theunissen MTM, van den Hout JMP, Pijnappel WWM, Schoser B, Laforêt P, Parenti G, van Doorn PA, van der Ploeg AT. Clinical insights in enzyme replacement therapy for metabolic storage disorders: lessons from Pompe disease. Lancet Neurol 2025; 24:230-245. [PMID: 39986311 DOI: 10.1016/s1474-4422(24)00518-0] [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: 09/27/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 02/24/2025]
Abstract
Metabolic storage disorders, including lysosomal storage disorders, pose complex challenges in management due to their progressive and life-threatening nature. Although enzyme replacement therapy has substantially improved outcomes for patients with lysosomal storage disorders, limitations of this therapy have become apparent throughout two decades of use. New clinical features of these diseases have emerged as patients live longer, leading to unresolved questions regarding ongoing treatment and long-term care. Innovative therapies are emerging that aim to improve targeting of tissues, particularly for previously inaccessible areas such as the CNS. These next-generation treatments hold promise for enhancing patient outcomes beyond what enzyme replacement therapy can do. Continued exploration of novel therapeutic strategies will be crucial for providing more effective and personalised care for these complex diseases.
Collapse
Affiliation(s)
- Nadine A M E van der Beek
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands.
| | - Maudy T M Theunissen
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands; Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands
| | - Johanna M P van den Hout
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands
| | - Wilhelmus W M Pijnappel
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands; Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands
| | - Benedikt Schoser
- Department of Neurology, Friedrich-Baur-Institute, Ludwig Maximilians University, Munich, Germany
| | - Pascal Laforêt
- Nord/Est/Île-de-France Neuromuscular Reference Center, FHU PHENIX, AP-HP, Raymond-Poincaré Hospital, Department of Neurology, Garches, France
| | - Giancarlo Parenti
- Federico II University, Department of Translational Medicine, Naples, Italy; Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Pieter A van Doorn
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands
| | - Ans T van der Ploeg
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands
| |
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
|
Christensen CL, Kan SH, Andrade-Heckman P, Rha AK, Harb JF, Wang RY. Base editing rescues acid α-glucosidase function in infantile-onset Pompe disease patient-derived cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102220. [PMID: 38948331 PMCID: PMC11214518 DOI: 10.1016/j.omtn.2024.102220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 05/16/2024] [Indexed: 07/02/2024]
Abstract
Infantile-onset Pompe disease (IOPD) results from pathogenic variants in the GAA gene, which encodes acid α-glucosidase. The correction of pathogenic variants through genome editing may be a valuable one-time therapy for PD and improve upon the current standard of care. We performed adenine base editing in human dermal fibroblasts harboring three transition nonsense variants, c.2227C>T (p.Q743∗; IOPD-1), c.2560C>T (p.R854∗; IOPD-2), and c.2608C>T (p.R870∗; IOPD-3). Up to 96% adenine deamination of target variants was observed, with minimal editing across >50 off-target sites. Post-base editing, expressed GAA protein was up to 0.66-fold normal (unaffected fibroblasts), an improvement over affected fibroblasts wherein GAA was undetectable. GAA enzyme activity was between 81.91 ± 13.51 and 129.98 ± 9.33 units/mg protein at 28 days post-transfection, which falls within the normal range (50-200 units/mg protein). LAMP2 protein was significantly decreased in the most robustly edited cell line, IOPD-3, indicating reduced lysosomal burden. Taken together, the findings reported herein demonstrate that base editing results in efficacious adenine deamination, restoration of GAA expression and activity, and reduction in lysosomal burden in the most robustly edited cells. Future work will assess base editing outcomes and the impact on Pompe pathology in two mouse models, Gaa c.2227C>T and Gaa c.2560C>T.
Collapse
Affiliation(s)
| | - Shih-Hsin Kan
- CHOC Children’s Research Institute, Orange, CA 92868, USA
| | | | | | - Jerry F. Harb
- CHOC Children’s Research Institute, Orange, CA 92868, USA
| | - Raymond Y. Wang
- Division of Metabolic Disorders, CHOC Children’s Specialists, Orange, CA 92868, USA
- Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| |
Collapse
|
4
|
İnci A, Ezgü FS, Tümer L. Advances in Immune Tolerance Induction in Enzyme Replacement Therapy. Paediatr Drugs 2024; 26:287-308. [PMID: 38664313 PMCID: PMC11074017 DOI: 10.1007/s40272-024-00627-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2024] [Indexed: 05/07/2024]
Abstract
Inborn errors of metabolism (IEMs) are a group of genetic diseases that occur due to the either deficiency of an enzyme involved in a metabolic/biochemical pathway or other disturbances in the metabolic pathway including transport protein or activator protein deficiencies, cofactor deficiencies, organelle biogenesis, maturation or trafficking problems. These disorders are collectively significant due to their substantial impact on both the well-being and survival of affected individuals. In the quest for effective treatments, enzyme replacement therapy (ERT) has emerged as a viable strategy for patients with many of the lysosomal storage disorders (LSD) and enzyme substitution therapy in the rare form of the other inborn errors of metabolism including phenylketonuria and hypophosphatasia. However, a major challenge associated with enzyme infusion in patients with these disorders, mainly LSD, is the development of high antibody titres. Strategies focusing on immunomodulation have shown promise in inducing immune tolerance to ERT, leading to improved overall survival rates. The implementation of immunomodulation concurrent with ERT administration has also resulted in a decreased occurrence of IgG antibody development compared with cases treated solely with ERT. By incorporating the knowledge gained from current approaches and analysing the outcomes of immune tolerance induction (ITI) modalities from clinical and preclinical trials have demonstrated significant improvement in the efficacy of ERT. In this comprehensive review, the progress in ITI modalities is assessed, drawing insights from both clinical and preclinical trials. The focus is on evaluating the advancements in ITI within the context of IEM, specifically addressing LSDs managed through ERT.
Collapse
Affiliation(s)
- Aslı İnci
- Department of Paediatric Metabolism and Nutrition, Gazi University School of Medicine, Emniyet Street, Yenimahalle, Ankara, Turkey.
| | - Fatih Süheyl Ezgü
- Department of Paediatric Metabolism and Nutrition, Gazi University School of Medicine, Emniyet Street, Yenimahalle, Ankara, Turkey
- Department of Paediatric Genetic, Gazi University School of Medicine, Ankara, Turkey
| | - Leyla Tümer
- Department of Paediatric Metabolism and Nutrition, Gazi University School of Medicine, Emniyet Street, Yenimahalle, Ankara, Turkey
| |
Collapse
|
5
|
Skorupan N, Peer CJ, Zhang X, Choo-Wosoba H, Ahmad MI, Lee MJ, Rastogi S, Sato N, Yu Y, Pegna GJ, Steinberg SM, Kalsi SS, Cao L, Figg WD, Trepel JB, Pastan I, FitzGerald D, Alewine C. Tofacitinib to prevent anti-drug antibody formation against LMB-100 immunotoxin in patients with advanced mesothelin-expressing cancers. Front Oncol 2024; 14:1386190. [PMID: 38706610 PMCID: PMC11066227 DOI: 10.3389/fonc.2024.1386190] [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] [Received: 02/14/2024] [Accepted: 03/22/2024] [Indexed: 05/07/2024] Open
Abstract
Background LMB-100 is a mesothelin (MSLN)-targeting recombinant immunotoxin (iTox) carrying a Pseudomonas exotoxin A payload that has shown promise against solid tumors, however, efficacy is limited by the development of neutralizing anti-drug antibodies (ADAs). Tofacitinib is an oral Janus Kinase (JAK) inhibitor that prevented ADA formation against iTox in preclinical studies. Methods A phase 1 trial testing LMB-100 and tofacitinib in patients with MSLN-expressing cancers (pancreatic adenocarcinoma, n=13; cholangiocarcinoma, n=1; appendiceal carcinoma, n=1; cystadenocarcinoma, n=1) was performed to assess safety and to determine if tofacitinib impacted ADA formation. Participants were treated for up to 3 cycles with LMB-100 as a 30-minute infusion on days 4, 6, and 8 at two dose levels (100 and 140 µg/kg) while oral tofacitinib was administered for the first 10 days of the cycle (10 mg BID). Peripheral blood was collected for analysis of ADA levels, serum cytokines and circulating immune subsets. Results The study was closed early due to occurrence of drug-induced pericarditis in 2 patients. Pericarditis with the combination was not reproducible in a transgenic murine model containing human MSLN. Two of 4 patients receiving all 3 cycles of treatment maintained effective LMB-100 levels, an unusual occurrence. Sustained increases in systemic IL-10 and TNF-α were seen, a phenomenon not observed in prior LMB-100 studies. A decrease in activated T cell subsets and an increase in circulating immunosuppressive myeloid populations occurred. No radiologic decreases in tumor volume were observed. Discussion Further testing of tofacitinib to prevent ADA formation is recommended in applicable non-malignant disease settings. Clinical trial registration https://www.clinicaltrials.gov/study/NCT04034238.
Collapse
Affiliation(s)
- Nebojsa Skorupan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Cody J. Peer
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xianyu Zhang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hyoyoung Choo-Wosoba
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mehwish I. Ahmad
- Office of Research Nursing, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shraddha Rastogi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Nahoko Sato
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yunkai Yu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Guillaume Joe Pegna
- Medical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shelley S. Kalsi
- Hematology Consult and Graduate Medical Section, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jane B. Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - David FitzGerald
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Christine Alewine
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
6
|
Desai AK, Shrivastava G, Grant CL, Wang RY, Burt TD, Kishnani PS. An updated management approach of Pompe disease patients with high-sustained anti-rhGAA IgG antibody titers: experience with bortezomib-based immunomodulation. Front Immunol 2024; 15:1360369. [PMID: 38524130 PMCID: PMC10959098 DOI: 10.3389/fimmu.2024.1360369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/26/2024] [Indexed: 03/26/2024] Open
Abstract
Introduction High sustained anti-rhGAA antibody titers (HSAT; ≥12,800) are directly linked to reduced efficacy of enzyme replacement therapy (ERT) and subsequent clinical deterioration in infantile-onset Pompe disease (IOPD). We have previously demonstrated the safety and effectiveness of a bortezomib-based immune-tolerance induction (ITI) regimen (bortezomib, rituximab, methotrexate, and IVIG) in eliminating HSAT. Methods Here, we describe two IOPD cases (patients 6 and 8) who developed HSAT at 8 and 10 weeks on ERT despite transient low-dose methotrexate ITI administration in the ERT-naïve setting and were treated with a bortezomib-based ITI regimen, and we compare their courses to a series of six historical patients (patients 1-5, and 7) with a similar presentation who exemplify our evolving approach to treatment. Results In total, patients 6 and 8 received 16 and 8 doses of bortezomib (4 doses=1 cycle) respectively reducing titers from 25,600 to seronegative, but differences in the course of their therapy were instructive regarding the optimal approach to initial treatment of HSAT; specifically, patient 6 was treated initially with only a single course of bortezomib rescue therapy, while patient 8 received two back-to-back courses. Patient 8 received IVIG therapy throughout the immunosuppression whereas patient 6 received IVIG therapy and was switched to subcutaneous IgG replacement. Patient 6 had a transient reduction in anti-rhGAA antibodies, after receiving a single initial cycle of bortezomib, but had a recurrence of high anti-rhGAA antibody titer after 160 weeks that required 3 additional cycles of bortezomib to ultimately achieve tolerance. In contrast, patient 8 achieved tolerance after being given two consecutive cycles of bortezomib during their initial treatment and had B cell recovery by week 54. Since the reduction in anti-rhGAA antibodies, both patients are doing well clinically, and have decreasing ALT, AST, and CK. No major infections leading to interruption of treatment were observed in either patient. The bortezomib-based ITI was safe and well-tolerated, and patients continue to receive ERT at 40 mg/kg/week. Discussion These case studies and our previous experience suggest that to achieve an effective reduction of anti-rhGAA antibodies in the setting of HSAT, bortezomib should be initiated at the earliest sign of high anti-rhGAA antibodies with a minimum of two consecutive cycles as shown in the case of patient 8. It is important to note that, despite initiation of ERT at age 2.3 weeks, patient 8 quickly developed HSAT. We recommend close monitoring of anti-rhGAA antibodies and early intervention with ITI as soon as significantly elevated anti-rhGAA antibody titers are noted.
Collapse
Affiliation(s)
- Ankit K. Desai
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Garima Shrivastava
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Christina L. Grant
- Division of Genetics and Metabolism, Children’s National Hospital, Washington, DC, United States
| | - Raymond Y. Wang
- Division of Metabolic Disorders, Children’s Hospital of Orange County, Orange, CA, United States
- Department of Pediatrics, University of California-Irvine School of Medicine, Orange, CA, United States
| | - Trevor D. Burt
- Division of Neonatology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- Children’s Health and Discovery Initiative, Duke University School of Medicine, Durham, NC, United States
| | - Priya S. Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
7
|
Liang Q, Vlaar EC, Pijnenburg JM, Rijkers E, Demmers JAA, Vulto AG, van der Ploeg AT, van Til NP, Pijnappel WWMP. Lentiviral gene therapy with IGF2-tagged GAA normalizes the skeletal muscle proteome in murine Pompe disease. J Proteomics 2024; 291:105037. [PMID: 38288553 DOI: 10.1016/j.jprot.2023.105037] [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: 05/16/2023] [Revised: 10/03/2023] [Accepted: 10/09/2023] [Indexed: 02/01/2024]
Abstract
Pompe disease is a lysosomal storage disorder caused by deficiency of acid alpha-glucosidase (GAA), resulting in glycogen accumulation with profound pathology in skeletal muscle. We recently developed an optimized form of lentiviral gene therapy for Pompe disease in which a codon-optimized version of the GAA transgene (LV-GAAco) was fused to an insulin-like growth factor 2 (IGF2) peptide (LV-IGF2.GAAco), to promote cellular uptake via the cation-independent mannose-6-phosphate/IGF2 receptor. Lentiviral gene therapy with LV-IGF2.GAAco showed superior efficacy in heart, skeletal muscle, and brain of Gaa -/- mice compared to gene therapy with untagged LV-GAAco. Here, we used quantitative mass spectrometry using TMT labeling to analyze the muscle proteome and the response to gene therapy in Gaa -/- mice. We found that muscle of Gaa -/- mice displayed altered levels of proteins including those with functions in the CLEAR signaling pathway, autophagy, cytoplasmic glycogen metabolism, calcium homeostasis, redox signaling, mitochondrial function, fatty acid transport, muscle contraction, cytoskeletal organization, phagosome maturation, and inflammation. Gene therapy with LV-GAAco resulted in partial correction of the muscle proteome, while gene therapy with LV-IGF2.GAAco resulted in a near-complete restoration to wild type levels without inducing extra proteomic changes, supporting clinical development of lentiviral gene therapy for Pompe disease. SIGNIFICANCE: Lysosomal glycogen accumulation is the primary cause of Pompe disease, and leads to a cascade of pathological events in cardiac and skeletal muscle and in the central nervous system. In this study, we identified the proteomic changes that are caused by Pompe disease in skeletal muscle of a mouse model. We showed that lentiviral gene therapy with LV-IGF2.GAAco nearly completely corrects disease-associated proteomic changes. This study supports the future clinical development of lentiviral gene therapy with LV-IGF2.GAAco as a new treatment option for Pompe disease.
Collapse
Affiliation(s)
- Qiushi Liang
- Department of Hematology and Research Laboratory of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands; Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands
| | - Eva C Vlaar
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands; Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands
| | - Joon M Pijnenburg
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands; Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands
| | - Erikjan Rijkers
- Proteomics Center, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands
| | - Jeroen A A Demmers
- Proteomics Center, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands
| | - Arnold G Vulto
- Hospital Pharmacy, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands
| | - Ans T van der Ploeg
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands
| | - Niek P van Til
- Department of Hematology, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands
| | - W W M Pim Pijnappel
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands; Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam 3015GE, the Netherlands.
| |
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
|
Pfrimmer C, Smitka M, Muschol N, Husain RA, Huemer M, Hennermann JB, Schuler R, Hahn A. Long-Term Outcome of Infantile Onset Pompe Disease Patients Treated with Enzyme Replacement Therapy - Data from a German-Austrian Cohort. J Neuromuscul Dis 2024; 11:167-177. [PMID: 38043017 PMCID: PMC10789365 DOI: 10.3233/jnd-230164] [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] [Accepted: 10/27/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND Enzyme replacement therapy (ERT) with recombinant human alglucosidase alfa (rhGAA) was approved in Europe in 2006. Nevertheless, data on the long-term outcome of infantile onset Pompe disease (IOPD) patients at school age is still limited. OBJECTIVE We analyzed in detail cardiac, respiratory, motor, and cognitive function of 15 German-speaking patients aged 7 and older who started ERT at a median age of 5 months. RESULTS Starting dose was 20 mg/kg biweekly in 12 patients, 20 mg/kg weekly in 2, and 40 mg/kg weekly in one patient. CRIM-status was positive in 13 patients (86.7%) and negative or unknown in one patient each (6.7%). Three patients (20%) received immunomodulation. Median age at last assessment was 9.1 (7.0-19.5) years. At last follow-up 1 patient (6.7%) had mild cardiac hypertrophy, 6 (42.9%) had cardiac arrhythmias, and 7 (46.7%) required assisted ventilation. Seven patients (46.7%) achieved the ability to walk independently and 5 (33.3%) were still ambulatory at last follow-up. Six patients (40%) were able to sit without support, while the remaining 4 (26.7%) were tetraplegic. Eleven patients underwent cognitive testing (Culture Fair Intelligence Test), while 4 were unable to meet the requirements for cognitive testing. Intelligence quotients (IQs) ranged from normal (IQ 117, 102, 96, 94) in 4 patients (36.4%) to mild developmental delay (IQ 81) in one patient (9.1%) to intellectual disability (IQ 69, 63, 61, 3x <55) in 6 patients (54.5%). White matter abnormalities were present in 10 out of 12 cerebral MRIs from 7 patients. CONCLUSION Substantial motor, cardiac, respiratory, and cognitive deficits are frequent in IOPD long-term survivors who started ERT before 2016. The findings of this study can be valuable as comparative data when evaluating the impact of newer treatment strategies including higher enzyme dosage, immunomodulation, modified enzymes, or early start of treatment following newborn screening.
Collapse
Affiliation(s)
- Charlotte Pfrimmer
- Department of Child Neurology, Justus-Liebig-University Gießen, Gießen, Germany
| | - Martin Smitka
- Children’s Hospital, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Nicole Muschol
- Department of Pediatrics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Ralf A. Husain
- Centre for Inborn Metabolic Disorders, Department of Neuropediatrics, Jena University Hospital, Jena, Germany
| | - Martina Huemer
- Department of Pediatrics, Landeskrankenhaus Bregenz, Bregenz, Austria and Division of Metabolism, Children’s Research Center and University Children’s Hospital Zurich, Zurich, Switzerland
| | - Julia B. Hennermann
- Villa Metabolica, Center for Pediatric and Adolescent Medicine, University Medical Center Mainz, Mainz, Germany
| | - Rahel Schuler
- Department of General Pediatrics and Neonatology, Justus-Liebig-University Gießen, Gießen, Germany
| | - Andreas Hahn
- Department of Child Neurology, Justus-Liebig-University Gießen, Gießen, Germany
| |
Collapse
|
10
|
Ditters IAM, van Kooten HA, van der Beek NAME, van der Ploeg AT, Huidekoper HH, van den Hout JMP. Are Anti-rhGAA Antibodies a Determinant of Treatment Outcome in Adults with Late-Onset Pompe Disease? A Systematic Review. Biomolecules 2023; 13:1414. [PMID: 37759814 PMCID: PMC10526476 DOI: 10.3390/biom13091414] [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: 07/14/2023] [Revised: 09/08/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Pompe disease is a lysosomal storage disease characterised by skeletal and respiratory muscle weakness. Since 2006, enzyme replacement therapy (ERT) with alglucosidase alfa has been available. ERT significantly improves the prognosis of patients with Pompe disease. The effect of high antibody titres on treatment response in adults with late-onset Pompe disease (LOPD) remains unclear but may contribute to interpatient variation. We therefore conducted a systematic review on this subject. METHODS A systematic search was performed in Embase, Medline Ovid, Web of Science, Psych Info Ovid, Cochrane (Clinical Trials only), and Google Scholar (random top-200). Articles were included if they involved adults with LOPD treated with alglucosidase alfa and mentioned anti-rhGAA antibodies or antibody titres. In addition, articles mentioning dosages different from the standard recommended dosage were included. RESULTS Our literature search retrieved 2562 publications, and 17 fulfilled our selection criteria, describing 443 cases. Seven publications reported on anti-rhGAA antibody titres on a group level, with the percentage of patients with a high titre as defined in the included articles ranging from 0-33%. Six publications reported on the effect of anti-rhGAA antibody titre on clinical course, and four found no correlation. Two studies reported a negative effect on treatment. The first study found a greater improvement in Medical Research Council (MRC) score in patients with no detectable antibody titre. In the second study, a patient discontinued ERT due to a declining neuromuscular state as a result of high anti-rhGAA antibody titres. Seven publications reported on 17 individual patients with a high antibody titre (range 1:12,800-1:3,906,250). In only two cases were high-sustained neutralising antibodies reported to interfere with treatment efficacy. CONCLUSIONS No clear effect of anti-rhGAA IgG antibodies on treatment response could be established for the majority of LOPD patients with a high antibody titre. In a minority of patients, a clinical decline related to (possible) interference of anti-rhGAA antibodies was described.
Collapse
Affiliation(s)
- Imke A. M. Ditters
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, Erasmus University Medical Center, P.O. Box 2060, 3000 CB Rotterdam, The Netherlands
| | - Harmke A. van Kooten
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC, Erasmus University Medical Center, P.O. Box 2060, 3000 CB Rotterdam, The Netherlands
| | - Nadine A. M. E. van der Beek
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC, Erasmus University Medical Center, P.O. Box 2060, 3000 CB Rotterdam, The Netherlands
| | - Ans T. van der Ploeg
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, Erasmus University Medical Center, P.O. Box 2060, 3000 CB Rotterdam, The Netherlands
| | - Hidde H. Huidekoper
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, Erasmus University Medical Center, P.O. Box 2060, 3000 CB Rotterdam, The Netherlands
| | - Johanna M. P. van den Hout
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, Erasmus University Medical Center, P.O. Box 2060, 3000 CB Rotterdam, The Netherlands
| |
Collapse
|
11
|
Tardieu M, Cudejko C, Cano A, Hoebeke C, Bernoux D, Goetz V, Pichard S, Brassier A, Schiff M, Feillet F, Rollier P, Mention K, Dobbelaere D, Fouilhoux A, Espil-Taris C, Eyer D, Huet F, Walther-Louvier U, Barth M, Chevret L, Kuster A, Lefranc J, Neveu J, Pitelet G, Ropars J, Rivier F, Roubertie A, Touati G, Vanhulle C, Tardieu E, Caillaud C, Froissart R, Champeaux M, Labarthe F, Chabrol B. Long-term follow-up of 64 children with classical infantile-onset Pompe disease since 2004: A French real-life observational study. Eur J Neurol 2023; 30:2828-2837. [PMID: 37235686 DOI: 10.1111/ene.15894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/21/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
BACKGROUND Classical infantile-onset Pompe disease (IOPD) is the most severe form of Pompe disease. Enzyme replacement therapy (ERT) has significantly increased survival but only a few studies have reported long-term outcomes. METHODS We retrospectively analyzed the outcomes of classical IOPD patients diagnosed in France between 2004 and 2020. RESULTS Sixty-four patients were identified. At diagnosis (median age 4 months) all patients had cardiomyopathy and most had severe hypotonia (57 of 62 patients, 92%). ERT was initiated in 50 (78%) patients and stopped later due to being ineffective in 10 (21%). Thirty-seven (58%) patients died during follow-up, including all untreated and discontinued ERT patients, and 13 additional patients. Mortality was higher during the first 3 years of life and after the age of 12 years. Persistence of cardiomyopathy during follow-up and/or the presence of heart failure were highly associated with an increased risk of death. In contrast, cross-reactive immunologic material (CRIM)-negative status (n = 16, 26%) was unrelated to increased mortality, presumably because immunomodulation protocols prevent the emergence of high antibody titers to ERT. Besides survival, decreased ERT efficacy appeared after the age of 6 years, with a progressive decline in motor and pulmonary functions for most survivors. CONCLUSIONS This study reports the long-term follow-up of one of the largest cohorts of classical IOPD patients and demonstrates high long-term mortality and morbidity rates with a secondary decline in muscular and respiratory functions. This decreased efficacy seems to be multifactorial, highlighting the importance of developing new therapeutic approaches targeting various aspects of pathogenesis.
Collapse
Affiliation(s)
- Marine Tardieu
- Centre de Référence des Maladies Héréditaires du Métabolisme ToTeM, Service de Médecine Pédiatrique, Hôpital Clocheville, Tours, France
| | - Céline Cudejko
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Neurométabolisme Pédiatrique, Hôpital Timone Enfants, AP-HM, Marseille, France
| | - Aline Cano
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Neurométabolisme Pédiatrique, Hôpital Timone Enfants, AP-HM, Marseille, France
| | - Célia Hoebeke
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Neurométabolisme Pédiatrique, Hôpital Timone Enfants, AP-HM, Marseille, France
| | - Delphine Bernoux
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Neurométabolisme Pédiatrique, Hôpital Timone Enfants, AP-HM, Marseille, France
| | - Violette Goetz
- Centre de Référence des Maladies Héréditaires du Métabolisme ToTeM, Service de Médecine Pédiatrique, Hôpital Clocheville, Tours, France
| | - Samia Pichard
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Métabolisme Pédiatrique, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Anaïs Brassier
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Métabolisme Pédiatrique, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Manuel Schiff
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Métabolisme Pédiatrique, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - François Feillet
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Médecine Infantile, Hôpital Brabois Enfants; Unité INSERM NGERE U 1256, Campus Babrois-Santé, Vandœuvre-lès-Nancy, France
| | - Paul Rollier
- Service de Génétique Clinique, Site Hôpital Sud, Rennes, France
| | - Karine Mention
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service Néphrologie, Endocrinologie, Maladies Métaboliques et Hématologie Pédiatrique, Hôpital Jeanne de Flandre, Lille, France
| | - Dries Dobbelaere
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service Néphrologie, Endocrinologie, Maladies Métaboliques et Hématologie Pédiatrique, Hôpital Jeanne de Flandre, Lille, France
| | - Alain Fouilhoux
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service d'Endocrinologie et de Diabétologie Pédiatriques et Maladies Héréditaires du Métabolisme, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron, France
| | - Caroline Espil-Taris
- Centre de Référence des Maladies Neuromusculaires AOC, Service de Neuropédiatrie, Hôpital des Enfants Pellegrin, Bordeaux, France
| | - Didier Eyer
- Service des Maladies Métaboliques, Hôpital de Hautepierre, Strasbourg, France
| | - Frédéric Huet
- Centre de Compétence des Maladies Héréditaires du Métabolisme, Service de Pédiatrie Multidisciplinaire, Hôpital d'Enfants, Dijon, France
| | - Ulrike Walther-Louvier
- Centre de Référence des Maladies Neuromusculaires AOC, Service de Neuropédiatrie, Hôpital Gui de Chauliac, Montpellier, France
| | - Magalie Barth
- Centre de Compétence des Maladies Héréditaires du Métabolisme, Service de Génétique, CHU Angers, Angers, France
| | - Laurent Chevret
- Service Pédiatrie et Urgences Pédiatriques, CH Saint-Brieuc, Saint-Brieuc, France
| | - Alice Kuster
- Centre de Compétence des Maladies Héréditaires du Métabolisme, Service de Réanimation Pédiatrique, CHU Nantes, Nantes, France
| | | | - Julien Neveu
- Service de Neuropédiatrie, Hôpitaux Pédiatriques de Nice, CHU Lenval, Nice, France
| | - Gaele Pitelet
- Service de Neuropédiatrie, Hôpitaux Pédiatriques de Nice, CHU Lenval, Nice, France
| | - Juliette Ropars
- Centre de Référence Maladies Neuromusculaires AOC, Service de Neuropédiatrie, Hôpital Morvan, Brest, France
| | - François Rivier
- Centre de Référence des Maladies Neuromusculaires AOC, Service de Neuropédiatrie, Hôpital Gui de Chauliac, Montpellier, France
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Agathe Roubertie
- Centre de Compétence des Maladies Héréditaires du Métabolisme, Service de Neurologie Pédiatrique, Hôpital Gui de Chauliac; INM, INSERM U 1298, Université de Montpellier, Montpellier, France
| | - Guy Touati
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Gastro-entérologie, Hépatologie, Nutrition et Maladies Héréditaires du Métabolisme Pédiatrique, Hôpital des Enfants, Toulouse, France
| | - Catherine Vanhulle
- Service de Néonatalogie et Réanimation Pédiatrique, Hôpital Charles Nicolle, Rouen, France
| | - Emilie Tardieu
- Service de Santé Universitaire, Université Lumière Lyon 2, Lyon, France
| | - Catherine Caillaud
- Service de Biochimie Métabolique, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Roseline Froissart
- Service de Biochimie et Biologie Moléculaire, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, Bron, France
| | - Murielle Champeaux
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Neurométabolisme Pédiatrique, Hôpital Timone Enfants, AP-HM, Marseille, France
| | - François Labarthe
- Centre de Référence des Maladies Héréditaires du Métabolisme ToTeM, Service de Médecine Pédiatrique, Hôpital Clocheville, Tours, France
- Inserm U1069, N2C, Université de Tours, Tours, France
| | - Brigitte Chabrol
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Neurométabolisme Pédiatrique, Hôpital Timone Enfants, AP-HM, Marseille, France
| |
Collapse
|
12
|
Ditters IAM, van Kooten HA, van der Beek NAME, Hardon JF, Ismailova G, Brusse E, Kruijshaar ME, van der Ploeg AT, van den Hout JMP, Huidekoper HH. Home-Based Infusion of Alglucosidase Alfa Can Safely be Implemented in Adults with Late-Onset Pompe Disease: Lessons Learned from 18,380 Infusions. BioDrugs 2023; 37:685-698. [PMID: 37326923 PMCID: PMC10432339 DOI: 10.1007/s40259-023-00609-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Enzyme replacement therapy (ERT) with alglucosidase alfa is the treatment for patients with Pompe disease, a hereditary metabolic myopathy. Home-based ERT is unavailable in many countries because of the boxed warning alglucosidase alfa received due to the risk of infusion-associated reactions (IARs). Since 2008, home infusions have been provided in The Netherlands. OBJECTIVES This study aimed to provide an overview of our experience with home-based infusions with alglucosidase alfa in adult Pompe patients, focusing on safety, including management of IARs. METHOD We analysed infusion data and IARs from adult patients starting ERT between 1999 and 2018. ERT was initially given in the hospital during the first year. Patients were eligible for home treatment if they were without IARs for multiple consecutive infusions and if a trained home nurse, with on-call back-up by a doctor, was available. The healthcare providers graded IARs. RESULTS We analysed data on 18,380 infusions with alglucosidase alfa in 121 adult patients; 4961 infusions (27.0%) were given in hospital and 13,419 (73.0%) were given at home. IARs occurred in 144 (2.9%) hospital infusions and 113 (0.8%) home infusions; 115 (79.9% of 144) IARs in hospital and 104 (92.0% of 113) IARs at home were mild, 25 IARs (17.4%) in hospital and 8 IARs (7.1%) at home were moderate, and very few severe IARs occurred (4 IARs in hospital [2.8%] and 1 IAR at home [0.9%]). Only one IAR in the home situation required immediate clinical evaluation in the hospital. CONCLUSION Given the small numbers of IARs that occurred with the home infusions, of which only one was severe, we conclude that alglucosidase alfa can be administered safely in the home situation, provided the appropriate infrastructure is present.
Collapse
Affiliation(s)
- Imke A M Ditters
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands
| | - Harmke A van Kooten
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Nadine A M E van der Beek
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Jacqueline F Hardon
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands
| | - Gamida Ismailova
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Esther Brusse
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Michelle E Kruijshaar
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands
| | - Ans T van der Ploeg
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands.
| | - Johanna M P van den Hout
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands
| | - Hidde H Huidekoper
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands
| |
Collapse
|
13
|
Burban A, Pucyło S, Sikora A, Opolski G, Grabowski M, Kołodzińska A. Hypertrophic Cardiomyopathy versus Storage Diseases with Myocardial Involvement. Int J Mol Sci 2023; 24:13239. [PMID: 37686045 PMCID: PMC10488064 DOI: 10.3390/ijms241713239] [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: 07/28/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
One of the main causes of heart failure is cardiomyopathies. Among them, the most common is hypertrophic cardiomyopathy (HCM), characterized by thickening of the left ventricular muscle. This article focuses on HCM and other cardiomyopathies with myocardial hypertrophy, including Fabry disease, Pompe disease, and Danon disease. The genetics and pathogenesis of these diseases are described, as well as current and experimental treatment options, such as pharmacological intervention and the potential of gene therapies. Although genetic approaches are promising and have the potential to become the best treatments for these diseases, further research is needed to evaluate their efficacy and safety. This article describes current knowledge and advances in the treatment of the aforementioned cardiomyopathies.
Collapse
Affiliation(s)
- Anna Burban
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
- Doctoral School, Medical University of Warsaw, 81 Żwirki i Wigury Street, 02-091 Warsaw, Poland
| | - Szymon Pucyło
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
| | - Aleksandra Sikora
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
| | - Grzegorz Opolski
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
| | - Marcin Grabowski
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
| | - Agnieszka Kołodzińska
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Erdem Ozdamar S, Koc AF, Durmus Tekce H, Kotan D, Ekmekci AH, Sengun IS, Yuceyar AN, Uluc K. Expert opinion on the diagnostic odyssey and management of late-onset Pompe disease: a neurologist's perspective. Front Neurol 2023; 14:1095134. [PMID: 37265469 PMCID: PMC10229878 DOI: 10.3389/fneur.2023.1095134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 04/14/2023] [Indexed: 06/03/2023] Open
Abstract
This consensus statement by a panel of neurology experts aimed to provide a practical and implementable guidance document to assist clinicians with the best clinical practice in terms of diagnosis, treatment, and monitoring of late-onset Pompe disease (LOPD). The participating experts consider the clinical suspicion of LOPD by the physician to be of utmost importance in the prevention of diagnostic and therapeutic delay in LOPD patients. A diagnostic algorithm is proposed to facilitate the diagnosis of LOPD in patients presenting with unexplained proximal/axial weakness (with or without respiratory symptoms) or restrictive respiratory insufficiency with hyperCKemia and/or exercise intolerance as the red flag symptoms/signs that raise the index of suspicion for LOPD diagnosis. The diagnosis is based on the subsequent use of dried blood spot (DBS) assay, and the DBS assay can be confirmed by acid alpha-glucosidase (GAA) tissue analysis in leukocytes, fibroblasts, or muscle fibers and/or genetic mutation analysis. Accordingly, experts consider increased awareness among physicians about potential presenting characteristics with a high index of suspicion for LOPD to be crucial to suspect and consider LOPD in the differential diagnosis, while strongly suggesting the use of a diagnostic algorithm combined with DBS assay and confirmatory tests in the timely diagnosis of LOPD and implementation of best practice patterns.
Collapse
Affiliation(s)
- Sevim Erdem Ozdamar
- Department of Neurology, Hacettepe University Faculty of Medicine, Ankara, Türkiye
| | - Ayse Filiz Koc
- Department of Neurology, Cukurova University Faculty of Medicine, Adana, Türkiye
| | - Hacer Durmus Tekce
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| | - Dilcan Kotan
- Department of Neurology, Sakarya University Faculty of Medicine, Sakarya, Türkiye
| | - Ahmet Hakan Ekmekci
- Department of Neurology, Selcuk University Faculty of Medicine, Konya, Türkiye
| | - Ihsan Sukru Sengun
- Department of Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Türkiye
| | - Ayse Nur Yuceyar
- Department of Neurology, Ege University Faculty of Medicine, Izmir, Türkiye
| | - Kayihan Uluc
- Department of Neurology, Marmara University School of Medicine, Istanbul, Türkiye
| |
Collapse
|
17
|
Dogan Y, Barese CN, Schindler JW, Yoon JK, Unnisa Z, Guda S, Jacobs ME, Oborski C, Maiwald T, Clarke DL, Schambach A, Pfeifer R, Harper C, Mason C, van Til NP. Screening chimeric GAA variants in preclinical study results in hematopoietic stem cell gene therapy candidate vectors for Pompe disease. Mol Ther Methods Clin Dev 2022; 27:464-487. [PMID: 36419467 PMCID: PMC9676529 DOI: 10.1016/j.omtm.2022.10.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022]
Abstract
Pompe disease is a rare genetic neuromuscular disorder caused by acid α-glucosidase (GAA) deficiency resulting in lysosomal glycogen accumulation and progressive myopathy. Enzyme replacement therapy, the current standard of care, penetrates poorly into the skeletal muscles and the peripheral and central nervous system (CNS), risks recombinant enzyme immunogenicity, and requires high doses and frequent infusions. Lentiviral vector-mediated hematopoietic stem and progenitor cell (HSPC) gene therapy was investigated in a Pompe mouse model using a clinically relevant promoter driving nine engineered GAA coding sequences incorporating distinct peptide tags and codon optimizations. Vectors solely including glycosylation-independent lysosomal targeting tags enhanced secretion and improved reduction of glycogen, myofiber, and CNS vacuolation in key tissues, although GAA enzyme activity and protein was consistently lower compared with native GAA. Genetically modified microglial cells in brains were detected at low levels but provided robust phenotypic correction. Furthermore, an amino acid substitution introduced in the tag reduced insulin receptor-mediated signaling with no evidence of an effect on blood glucose levels in Pompe mice. This study demonstrated the therapeutic potential of lentiviral HSPC gene therapy exploiting optimized GAA tagged coding sequences to reverse Pompe disease pathology in a preclinical mouse model, providing promising vector candidates for further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Chris Mason
- AVROBIO, Inc., Cambridge, MA 02139, USA
- Advanced Centre for Biochemical Engineering, University College London, London WC1E 6AE, UK
- Corresponding author: Chris Mason, Advanced Centre for Biochemical Engineering, University College London, London WC1E 6AE, UK
| | - Niek P. van Til
- AVROBIO, Inc., Cambridge, MA 02139, USA
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University, and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, 1081 HV Amsterdam, the Netherlands
- Corresponding author: Niek P. van Til, Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University, and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, 1081 HV Amsterdam, the Netherlands
| |
Collapse
|
18
|
Liang Q, Vlaar EC, Catalano F, Pijnenburg JM, Stok M, van Helsdingen Y, Vulto AG, Unger WW, van der Ploeg AT, Pijnappel WP, van Til NP. Lentiviral gene therapy prevents anti-human acid α-glucosidase antibody formation in murine Pompe disease. Mol Ther Methods Clin Dev 2022; 25:520-532. [PMID: 35662813 PMCID: PMC9127119 DOI: 10.1016/j.omtm.2022.04.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 04/29/2022] [Indexed: 01/20/2023]
Abstract
Enzyme replacement therapy (ERT) is the current standard treatment for Pompe disease, a lysosomal storage disorder caused by deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). ERT has shown to be lifesaving in patients with classic infantile Pompe disease. However, a major drawback is the development of neutralizing antibodies against ERT. Hematopoietic stem and progenitor cell-mediated lentiviral gene therapy (HSPC-LVGT) provides a novel, potential lifelong therapy with a single intervention and may induce immune tolerance. Here, we investigated whether ERT can be safely applied as additional or alternative therapy following HSPC-LVGT in a murine model of Pompe disease. We found that lentiviral expression at subtherapeutic dose was sufficient to induce tolerance to the transgene product, as well as to subsequently administered ERT. Immune tolerance was established within 4–6 weeks after gene therapy. The mice tolerated ERT doses up to 100 mg/kg, allowing ERT to eliminate glycogen accumulation in cardiac and skeletal muscle and normalizing locomotor function. The presence of HSPC-derived cells expressing GAA in the thymus suggested the establishment of central immune tolerance. These findings demonstrate that lentiviral gene therapy in murine Pompe disease induced robust and long-term immune tolerance to GAA either expressed by a transgene or supplied as ERT.
Collapse
Affiliation(s)
- Qiushi Liang
- Department of Hematology and Research Laboratory of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Eva C. Vlaar
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Fabio Catalano
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Joon M. Pijnenburg
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Merel Stok
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Hematology, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Yvette van Helsdingen
- Department of Hematology, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Arnold G. Vulto
- Hospital Pharmacy, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Wendy W.J. Unger
- Laboratory of Pediatrics, Erasmus MC University Medical Center-Sophia Children’s Hospital, 3015GE Rotterdam, the Netherlands
| | - Ans T. van der Ploeg
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - W.W.M. Pim Pijnappel
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Corresponding author W.W.M. Pim Pijnappel, PhD, Erasmus University Medical Center, 3015GE Rotterdam, the Netherlands.
| | - Niek P. van Til
- Department of Hematology, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| |
Collapse
|
19
|
Gragnaniello V, Deodato F, Gasperini S, Donati MA, Canessa C, Fecarotta S, Pascarella A, Spadaro G, Concolino D, Burlina A, Parenti G, Strisciuglio P, Fiumara A, Casa RD. Immune responses to alglucosidase in infantile Pompe disease: recommendations from an Italian pediatric expert panel. Ital J Pediatr 2022; 48:41. [PMID: 35248118 PMCID: PMC8898438 DOI: 10.1186/s13052-022-01219-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 01/22/2022] [Indexed: 11/29/2022] Open
Abstract
Background Classic infantile onset of Pompe disease (c-IOPD) leads to hypotonia and hypertrophic cardiomyopathy within the first days to weeks of life and, without treatment, patients die of cardiorespiratory failure in their first 1–2 years of life. Enzymatic replacement therapy (ERT) with alglucosidase alfa is the only available treatment, but adverse immune reactions can reduce ERT’s effectiveness and safety. It is therefore very important to identify strategies to prevent and manage these complications. Several articles have been written on this disease over the last 10 years, but no univocal indications have been established. Methods Our study presents a review of the current literature on management of immune responses to ERT in c-IOPD as considered by an Italian study group of pediatric metabolists and immunologists in light of our shared patient experience. Results We summarize the protocols for the management of adverse reactions to ERT, analyzing their advantages and disadvantages, and provide expert recommendations for their optimal management, to the best of current knowledge. However, further studies are needed to improve actual management protocols, which still have several limitations.
Collapse
|
20
|
van Kooten HA, Ditters IAM, Hoogeveen-Westerveld M, Jacobs EH, van den Hout JMP, van Doorn PA, Pijnappel WWMP, van der Ploeg AT, van der Beek NAME. Antibodies against recombinant human alpha-glucosidase do not seem to affect clinical outcome in childhood onset Pompe disease. Orphanet J Rare Dis 2022; 17:31. [PMID: 35109913 PMCID: PMC8812154 DOI: 10.1186/s13023-022-02175-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/16/2022] [Indexed: 01/16/2023] Open
Abstract
Background Enzyme replacement therapy (ERT) with recombinant human alpha-glucosidase (rhGAA, alglucosidase alfa) has improved survival, motor outcomes, daily life activity and quality of life in Pompe patients. However, ERT in Pompe disease often induces formation of antibodies, which may reduce the efficacy of treatment and can lead to adverse events. In this study antibody formation and their effect on clinical outcome in patients with childhood onset Pompe disease treated with enzyme replacement therapy (ERT) with recombinant human alpha-glucosidase (rhGAA) are analyzed. Methods Enzyme-linked immunosorbent assay (ELISA) was used to determine anti-rhGAA antibody titers at predefined time points. The effect of antibodies on rhGAA activity (neutralizing effects) was measured in vitro. Clinical effects were evaluated by assessing muscle strength (MRC score) and function (QMFT-score), pulmonary function and infusion associated reactions (IARs). Results Twenty-two patients were included (age at start ERT 1.1–16.4 years, median treatment duration 12.4 years). Peak antibody titers were low (< 1:1250) in 9%, intermediate (1:1250–1:31,250) in 68% and high (≥ 1:31250) in 23% of patients; three patients (14%) had more than one titer of ≥ 1:31,250. Four patients (18%) experienced IARs; two patients from the high titer group had 86% of all IARs. Inhibition of intracellular GAA activity (58%) in vitro was found in one sample. The clinical course did not appear to be influenced by antibody titers. Conclusions Ninety-one percent of childhood onset Pompe patients developed anti-rhGAA antibodies (above background level), a minority of whom had high antibody titers at repeated time points, which do not seem to interfere with clinical outcome. High antibody titers may be associated with the occurrence of IARs. Although the majority of patients does not develop high titers; antibody titers should be determined in case of clinical deterioration. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02175-2.
Collapse
Affiliation(s)
- Harmke A van Kooten
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Imke A M Ditters
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marianne Hoogeveen-Westerveld
- Department of Pediatrics, Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Edwin H Jacobs
- Department of Pediatrics, Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Johanna M P van den Hout
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Pieter A van Doorn
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - W W M Pim Pijnappel
- Department of Pediatrics, Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Ans T van der Ploeg
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nadine A M E van der Beek
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands. .,Department of Neurology, Erasmus University Medical Center, Mailbox 2040, 3000 CA, Rotterdam, the Netherlands.
| |
Collapse
|
21
|
Ditters IAM, Huidekoper HH, Kruijshaar ME, Rizopoulos D, Hahn A, Mongini TE, Labarthe F, Tardieu M, Chabrol B, Brassier A, Parini R, Parenti G, van der Beek NAME, van der Ploeg AT, van den Hout JMP, Mengel E, Hennermann J, Smitka M, Muschol N, Marquardt T, Marquardt M, Thiels C, Spada M, Pagliardini V, Menni F, della Casa R, Deodato F, Gasperini S, Burlina A, Donati A, Pichard S, Feillet F, Huet F, Mention K, Eyer D, Kuster A, Espil Taris C, Lefranc J, Barth M, Bruel H, Chevret L, Pitelet G, Pitelet C, Rivier F, Dobbelaere D. Effect of alglucosidase alfa dosage on survival and walking ability in patients with classic infantile Pompe disease: a multicentre observational cohort study from the European Pompe Consortium. THE LANCET CHILD & ADOLESCENT HEALTH 2022; 6:28-37. [DOI: 10.1016/s2352-4642(21)00308-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 12/29/2022]
|
22
|
Pharmacological Chaperone Therapy for Pompe Disease. Molecules 2021; 26:molecules26237223. [PMID: 34885805 PMCID: PMC8659197 DOI: 10.3390/molecules26237223] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 11/17/2022] Open
Abstract
Pompe disease (PD), a lysosomal storage disease, is caused by mutations of the GAA gene, inducing deficiency in the acid alpha-glucosidase (GAA). This enzymatic impairment causes glycogen burden in lysosomes and triggers cell malfunctions, especially in cardiac, smooth and skeletal muscle cells and motor neurons. To date, the only approved treatment available for PD is enzyme replacement therapy (ERT) consisting of intravenous administration of rhGAA. The limitations of ERT have motivated the investigation of new therapies. Pharmacological chaperone (PC) therapy aims at restoring enzymatic activity through protein stabilization by ligand binding. PCs are divided into two classes: active site-specific chaperones (ASSCs) and the non-inhibitory PCs. In this review, we summarize the different pharmacological chaperones reported against PD by specifying their PC class and activity. An emphasis is placed on the recent use of these chaperones in combination with ERT.
Collapse
|
23
|
Tarallo A, Damiano C, Strollo S, Minopoli N, Indrieri A, Polishchuk E, Zappa F, Nusco E, Fecarotta S, Porto C, Coletta M, Iacono R, Moracci M, Polishchuk R, Medina DL, Imbimbo P, Monti DM, De Matteis MA, Parenti G. Correction of oxidative stress enhances enzyme replacement therapy in Pompe disease. EMBO Mol Med 2021; 13:e14434. [PMID: 34606154 PMCID: PMC8573602 DOI: 10.15252/emmm.202114434] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pompe disease is a metabolic myopathy due to acid alpha-glucosidase deficiency. In addition to glycogen storage, secondary dysregulation of cellular functions, such as autophagy and oxidative stress, contributes to the disease pathophysiology. We have tested whether oxidative stress impacts on enzyme replacement therapy with recombinant human alpha-glucosidase (rhGAA), currently the standard of care for Pompe disease patients, and whether correction of oxidative stress may be beneficial for rhGAA therapy. We found elevated oxidative stress levels in tissues from the Pompe disease murine model and in patients' cells. In cells, stress levels inversely correlated with the ability of rhGAA to correct the enzymatic deficiency. Antioxidants (N-acetylcysteine, idebenone, resveratrol, edaravone) improved alpha-glucosidase activity in rhGAA-treated cells, enhanced enzyme processing, and improved mannose-6-phosphate receptor localization. When co-administered with rhGAA, antioxidants improved alpha-glucosidase activity in tissues from the Pompe disease mouse model. These results indicate that oxidative stress impacts on the efficacy of enzyme replacement therapy in Pompe disease and that manipulation of secondary abnormalities may represent a strategy to improve the efficacy of therapies for this disorder.
Collapse
Affiliation(s)
- Antonietta Tarallo
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Carla Damiano
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Sandra Strollo
- Telethon Institute of Genetics and MedicinePozzuoliItaly
| | - Nadia Minopoli
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Alessia Indrieri
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Institute for Genetic and Biomedical Research (IRGB)National Research Council (CNR)MilanItaly
| | | | - Francesca Zappa
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Present address:
Department of Molecular, Cellular, and Developmental BiologyUniversity of CaliforniaSanta BarbaraCAUSA
| | - Edoardo Nusco
- Telethon Institute of Genetics and MedicinePozzuoliItaly
| | - Simona Fecarotta
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Caterina Porto
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Marcella Coletta
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
- Present address:
IInd Division of NeurologyMultiple Sclerosis CenterUniversity of Campania "Luigi Vanvitelli"NaplesItaly
| | - Roberta Iacono
- Department of BiologyUniversity of Naples "Federico II", Complesso Universitario di Monte S. AngeloNaplesItaly
- Institute of Biosciences and BioResources ‐ National Research Council of ItalyNaplesItaly
| | - Marco Moracci
- Department of BiologyUniversity of Naples "Federico II", Complesso Universitario di Monte S. AngeloNaplesItaly
- Institute of Biosciences and BioResources ‐ National Research Council of ItalyNaplesItaly
| | | | - Diego Luis Medina
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Paola Imbimbo
- Department of Chemical SciencesFederico II UniversityNaplesItaly
| | | | - Maria Antonietta De Matteis
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Molecular Medicine and Medical BiotechnologiesFederico II UniversityNaplesItaly
| | - Giancarlo Parenti
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| |
Collapse
|
24
|
A Systematic Review and Meta-Analysis of Enzyme Replacement Therapy in Late-Onset Pompe Disease. J Clin Med 2021; 10:jcm10214828. [PMID: 34768348 PMCID: PMC8584814 DOI: 10.3390/jcm10214828] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
Pompe disease (PD) is a glycogen storage disorder caused by deficient activity of acid alpha-glucosidase (GAA). We sought to review the latest available evidence on the safety and efficacy of recombinant human GAA enzyme replacement therapy (ERT) for late-onset PD (LOPD). METHODS We systematically searched the MEDLINE (via PubMed), Embase, and Cochrane databases for prospective clinical studies evaluating ERT for LOPD on pre-specified outcomes. A meta-analysis was also performed. RESULTS Of 1601 articles identified, 22 were included. Studies were heterogeneous and with very low certainty of evidence for most outcomes. The following outcomes showed improvements associated with GAA ERT, over a mean follow-up of 32.5 months: distance walked in the 6-min walking test (6MWT) (mean change 35.7 m (95% confidence interval [CI] 7.78, 63.75)), physical domain of the SF-36 quality of life (QOL) questionnaire (mean change 1.96 (95% CI 0.33, 3.59)), and time on ventilation (TOV) (mean change -2.64 h (95% CI -5.28, 0.00)). There were no differences between the pre- and post-ERT period for functional vital capacity (FVC), Walton and Gardner-Medwin Scale score, upper-limb strength, or total SF-36 QOL score. Adverse events (AEs) after ERT were mild in most cases. CONCLUSION Considering the limitations imposed by the rarity of PD, our data suggest that GAA ERT improves 6MWT, physical QOL, and TOV in LOPD patients. ERT was safe in the studied population. PROSPERO register: 135102.
Collapse
|
25
|
Correlation of GAA Genotype and Acid-α-Glucosidase Enzyme Activity in Hungarian Patients with Pompe Disease. Life (Basel) 2021; 11:life11060507. [PMID: 34072668 PMCID: PMC8228169 DOI: 10.3390/life11060507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 01/20/2023] Open
Abstract
Pompe disease is caused by the accumulation of glycogen in the lysosomes due to a deficiency of the lysosomal acid-α-glucosidase (GAA) enzyme. Depending on residual enzyme activity, the disease manifests two distinct phenotypes. In this study, we assess an enzymatic and genetic analysis of Hungarian patients with Pompe disease. Twenty-four patients diagnosed with Pompe disease were included. Enzyme activity of acid-α-glucosidase was measured by mass spectrometry. Sanger sequencing and an MLPA of the GAA gene were performed in all patients. Twenty (83.33%) patients were classified as having late-onset Pompe disease and four (16.66%) had infantile-onset Pompe disease. Fifteen different pathogenic GAA variants were detected. The most common finding was the c.-32-13 T > G splice site alteration. Comparing the α-glucosidase enzyme activity of homozygous cases to the compound heterozygous cases of the c.-32-13 T > G disease-causing variant, the mean GAA activity in homozygous cases was significantly higher. The lowest enzyme activity was found in cases where the c.-32-13 T > G variant was not present. The localization of the identified sequence variations in regions encoding the crucial protein domains of GAA correlates with severe effects on enzyme activity. A better understanding of the impact of pathogenic gene variations may help earlier initiation of enzyme replacement therapy (ERT) if subtle symptoms occur. Further information on the effect of GAA gene variation on the efficacy of treatment and the extent of immune response to ERT would be of importance for optimal disease management and designing effective treatment plans.
Collapse
|
26
|
Hernández-Arévalo P, Santotoribio JD, Delarosa-Rodríguez R, González-Meneses A, García-Morillo S, Jiménez-Arriscado P, Guerrero JM, Macher HC. Genotype-phenotype correlation of 17 cases of Pompe disease in Spanish patients and identification of 4 novel GAA variants. Orphanet J Rare Dis 2021; 16:233. [PMID: 34020684 PMCID: PMC8139113 DOI: 10.1186/s13023-021-01864-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/10/2021] [Indexed: 11/15/2022] Open
Abstract
Background Pompe disease (PD) is an autosomal recessive metabolic disorder caused by pathogenic variants in the acid -glucosidase gene (GAA) that produces defects in the lysosomal acid -1,4-glucosidase. We aimed to identify genetic variations and clinical features in Spanish subjects to establish genotypephenotype correlation. Methods A total of 2637 samples of patients who showed symptoms or susceptible signs of PD were enrolled in this observational study. Enzymatic activity was detected by fluorometric techniques and the genetic study was carried out using Next-Generation Sequencing. Results Fourteen different variants from 17 diagnosed patients were identified, seven males and nine females with LOPD (mean age 36.07, SD 20.57, range 764) and a 2-day-old boy with IOPD, four genetic variants had not been described in the literature previously, including a homozygous variant. In all of them -glucosidase activity was decreased. Muscle weakness, respiratory distress, exercise intolerance, hypotonia, dysphagia and myalgia were commonly observed in patients. Conclusions This study report four new genetic variants that contribute to the pathogenic variants spectrum of the GAA gene. We confirm that patients in Spain have a characteristic profile of a European population, with c.-32-13T>G being the most prevalent variant. Furthermore, it was confirmed that the c.236_246delCCACACAGTGC pathogenic variant in homozygosity is associated with early disease and a worse prognosis.
Collapse
Affiliation(s)
- Paula Hernández-Arévalo
- Fundación Pública Andaluza para la Gestión de la Investigación en Salud de Sevilla (FISEVI), Molecular Diagnosis and Rare Diseases Laboratory, Department of Clinical Biochemistry, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - José D Santotoribio
- Molecular Diagnosis and Rare Diseases Laboratory, Department of Clinical Biochemistry, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Rocío Delarosa-Rodríguez
- Fundación Pública Andaluza para la Gestión de la Investigación en Salud de Sevilla (FISEVI), Molecular Diagnosis and Rare Diseases Laboratory, Department of Clinical Biochemistry, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Antonio González-Meneses
- Dysmorphology Unit, Department of Pediatrics, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Salvador García-Morillo
- Collagenosis and Minority Diseases Unit, Experimental Cardiovascular Risk Unit, Department of Internal Medicine, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Pilar Jiménez-Arriscado
- Molecular Diagnosis and Rare Diseases Laboratory, Department of Clinical Biochemistry, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Juan M Guerrero
- Department of Clinical Biochemistry and Molecular Biology Hospital Universitario Virgen del Rocío, Institute of Biomedicine of Seville (Ibis),, Seville University, Seville, Spain
| | - Hada C Macher
- Molecular Diagnosis and Rare Diseases Laboratory, Department of Clinical Biochemistry, Hospital Universitario Virgen del Rocío, Seville, Spain.
| |
Collapse
|
27
|
Sheth J, Nair A. Treatment for Lysosomal Storage Disorders. Curr Pharm Des 2021; 26:5110-5118. [PMID: 33059565 DOI: 10.2174/1381612826666201015154932] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/22/2020] [Indexed: 12/31/2022]
Abstract
Lysosomal storage disorders comprise a group of approximately 70 types of inherited diseases resulting due to lysosomal gene defects. The outcome of the defect is a deficiency in either of the three: namely, lysosomal enzymes, activator protein, or transmembrane protein, as a result of which there is an unwanted accumulation of biomolecules inside the lysosomes. The pathophysiology of these conditions is complex affecting several organ systems and nervous system involvement in a majority of cases. Several research studies have well elucidated the mechanism underlying the disease condition leading to the development in devising the treatment strategies for the same. Currently, these approaches aim to reduce the severity of symptoms or delay the disease progression but do not provide a complete cure. The main treatment methods include Enzyme replacement therapy, Bone marrow transplantation, Substrate reduction therapy, use of molecular chaperones, and Gene therapy. This review article presents an elaborate description of these strategies and discusses the ongoing studies for the same.
Collapse
Affiliation(s)
- Jayesh Sheth
- Foundation for Research in Genetics and Endocrinology, Institute of Human Genetics, FRIGE House, Jodhpur Gam Road, Satellite, Ahmedabad, Gujarat, India
| | - Aadhira Nair
- Foundation for Research in Genetics and Endocrinology, Institute of Human Genetics, FRIGE House, Jodhpur Gam Road, Satellite, Ahmedabad, Gujarat, India
| |
Collapse
|
28
|
Sarah B, Giovanna B, Emanuela K, Nadi N, Josè V, Alberto P. Clinical efficacy of the enzyme replacement therapy in patients with late-onset Pompe disease: a systematic review and a meta-analysis. J Neurol 2021; 269:733-741. [PMID: 33851281 PMCID: PMC8782782 DOI: 10.1007/s00415-021-10526-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 01/10/2023]
Abstract
In patients with late-onset Pompe disease (LOPD), the efficacy of the enzyme replacement therapy (ERT) with recombinant human alpha-glucosidase (rhGAA) is difficult to evaluate, due to the clinical heterogeneity and the small sample sizes in published studies. Therefore, we conduct a systematic literature review and meta-analysis of the literature to evaluate the efficacy of ERT in LOPD patients considering the walking distance, respiratory function and muscle strength. Particularly, six-minute walk test (6MWT), forced vital capacity (FVC), medical research council (MRC) grading, quantitative muscle testing (QMT), and quick motor function test (QMFT) were outcomes of interest. Overall, 619 studies were identified in PubMed, EMBASE and by manual search on July 18th, 2020. After an initial assessment, 16 studies were included in the meta-analysis, containing clinical data from 589 patients with LOPD. For the 6MWT, 419 patients were analyzed. Walking distance improved on average, 32.2 m greater during the observed period (p = 0.0003), compared to the distance at the baseline. The meta-analysis did not show any improvement in FVC and only a tendency towards better muscle strength after treatment with ERT, but the difference was not statistically significant. In conclusion, the available data showed that ERT has a significant beneficial efficacy in the improvement of walking distance in LOPD patients and a non-significant improvement of muscle strength. No improvement in respiratory capacity was found. More prospective and controlled trials are needed to demonstrate a clear clinical benefit of ERT.
Collapse
Affiliation(s)
- Berli Sarah
- Institute for Intensive Care Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Brandi Giovanna
- Institute for Intensive Care Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland.
| | - Keller Emanuela
- Institute for Intensive Care Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland.,Department of Neurosurgery and Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Najia Nadi
- Institute for Intensive Care Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Vitale Josè
- Intensive Care Unit, Regional Hospital Mendrisio, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,Centro Medico, Mendrisio, Switzerland
| | - Pagnamenta Alberto
- Intensive Care Unit, Regional Hospital Mendrisio, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,Unit of Biostatistics, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,Division of Pneumology, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
29
|
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
|
30
|
de Faria DOS, 't Groen SLMI, Hoogeveen-Westerveld M, Nino MY, van der Ploeg AT, Bergsma AJ, Pijnappel WWMP. Update of the Pompe variant database for the prediction of clinical phenotypes: Novel disease-associated variants, common sequence variants, and results from newborn screening. Hum Mutat 2020; 42:119-134. [PMID: 33560568 PMCID: PMC7898817 DOI: 10.1002/humu.24148] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/06/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022]
Abstract
Pompe disease is an inherited disorder caused by disease-associated variants in the acid α-glucosidase gene (GAA). The Pompe disease GAA variant database (http://www.pompevariantdatabase.nl) is a curated, open-source, disease-specific database, and lists disease-associated GAA variants, in silico predictions, and clinical phenotypes reported until 2016. Here, we provide an update to include 226 disease-associated variants that were published until 2020. We also listed 148 common GAA sequence variants that do not cause Pompe disease. GAA variants with unknown severity that were identified only in newborn screening programs were listed as a new feature to indicate the reason why phenotypes were still unknown. Expression studies were performed for common missense variants to predict their severity. The updated Pompe disease GAA variant database now includes 648 disease-associated variants, 26 variants from newborn screening, and 237 variants with unknown severity. Regular updates of the Pompe disease GAA variant database will be required to improve genetic counseling and the study of genotype-phenotype relationships.
Collapse
Affiliation(s)
- Douglas O S de Faria
- 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
| | | | - Monica Y Nino
- 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
| | - Ans T van der Ploeg
- Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Atze J Bergsma
- 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
| | - W W M Pim 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
| |
Collapse
|
31
|
Poelman E, van den Dorpel JJA, Hoogeveen‐Westerveld M, van den Hout JMP, van der Giessen LJ, van der Beek NAME, Pijnappel WWMP, van der Ploeg AT. Effects of higher and more frequent dosing of alglucosidase alfa and immunomodulation on long-term clinical outcome of classic infantile Pompe patients. J Inherit Metab Dis 2020; 43:1243-1253. [PMID: 32506446 PMCID: PMC7689828 DOI: 10.1002/jimd.12268] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/28/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022]
Abstract
The aim of this study was to compare the long-term outcome of classic infantile Pompe patients treated with 20 mg/kg alglucosidase alfa every other week (eow) to those treated with 40 mg/kg/week, and to study the additional effect of immunomodulation. Six patients received 20 mg/kg eow and twelve 40 mg/kg/week. Five patients were cross-reactive immunologic material (CRIM)-negative, two in the 20 mg, three in the 40 mg group. We compared (ventilator-free) survival, motor outcome, infusion associated reactions (IARs), and antibody formation. From 2012 on patients >2 months in the 40 mg group also received immunomodulation with rituximab, methotrexate, and intravenous immunoglobulin (IVIG) in an enzyme replacement therapy (ERT)-naïve setting. Survival was 66% in the 20 mg group and 92% in the 40 mg group. Ventilator-free survival was 50% and 92%. Both CRIM-negative patients in the 20 mg group died, whereas all three are alive in the 40 mg group. In the 20 mg group, 67% learned to walk compared with 92% in the 40 mg group. At the age of 3 years, 33% and 92% were able to walk. Peak antibody titers ranged from 1:1250 to 1:31 250 in the 20 mg group and from 1:250 to 1:800 000 in the 40 mg group. Five patients of the 40 mg group of whom two CRIM-negative also received immunomodulation. B-cell recovery was observed between 5.7 and 7.9 months after the last dose of rituximab. After B-cell recovery titers of patients with and without immunomodulation were similar (ranges 1:6 250-1:800 000 and 1:250-1:781 250). This study shows that classic infantile patients treated with 40 mg/kg/week from the start to end have a better (ventilator-free) survival and motor outcome. Immunomodulation did not prevent antibody formation in our study.
Collapse
Affiliation(s)
- Esther Poelman
- Center for Lysosomal and Metabolic Diseases, Department of PediatricsErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Jan J. A. van den Dorpel
- Center for Lysosomal and Metabolic Diseases, Department of PediatricsErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Marianne Hoogeveen‐Westerveld
- Center for Lysosomal and Metabolic Diseases, Department of Clinical GeneticsErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Johanna M. P. van den Hout
- Center for Lysosomal and Metabolic Diseases, Department of PediatricsErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Lianne J. van der Giessen
- Center for Lysosomal and Metabolic Diseases, Department of Pediatric PhysiotherapyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Nadine A. M. E. van der Beek
- Center for Lysosomal and Metabolic Diseases, Department of PediatricsErasmus MC University Medical CenterRotterdamThe Netherlands
- Center for Lysosomal and Metabolic Diseases, Department of NeurologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - W. W. M. Pim Pijnappel
- Center for Lysosomal and Metabolic Diseases, Department of Clinical GeneticsErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Ans T. van der Ploeg
- Center for Lysosomal and Metabolic Diseases, Department of PediatricsErasmus MC University Medical CenterRotterdamThe Netherlands
| |
Collapse
|
32
|
van den Dorpel JJA, Poelman E, Harlaar L, van Kooten HA, van der Giessen LJ, van Doorn PA, van der Ploeg AT, van den Hout JMP, van der Beek NAME. Distal muscle weakness is a common and early feature in long-term enzyme-treated classic infantile Pompe patients. Orphanet J Rare Dis 2020; 15:247. [PMID: 32928284 PMCID: PMC7488760 DOI: 10.1186/s13023-020-01482-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/26/2020] [Indexed: 12/25/2022] Open
Abstract
Background Enzyme replacement therapy (ERT; alglucosidase alfa) has improved the prospects for patients with classic infantile Pompe disease considerably. However, over time we noticed that many of these children exhibit distal muscle weakness at an early age, which is in contrast to the primarily proximal and axial muscle weakness in patients with late-onset Pompe disease. This was reason to study the prevalence and severity of distal muscle weakness, and the sequence of muscle involvement over time in patients that had learned to walk under ERT. Methods In this prospective, single-center cohort study, we studied 16 classic infantile patients. We used video recordings that were made during regular standardized assessments to investigate distal muscle function (active dorsiflexion of the feet during walking; ability to use a pincer grasp/actively extend the fingers) and proximal muscle function (standing up from a supine position; raising the arms above the head). Results Median age at start of ERT was 3.2 months (0.1–5.8 months), median age at study end was 5.6 years (2.9–18.2 years). Six patients (6/16, 38%) initially had no evident signs of distal muscle weakness and developed a gait with active dorsiflexion of the feet. The other 10 patients never exhibited active dorsiflexion of the feet during walking. At study-end two patients showed no loss of distal muscle function. A subset of five patients (5/16, 31%) developed also weakness of the hands, particularly of the extensors of the 3rd and 4th digit. Conclusions We found that the majority (14/16, 88%) of patients who had learned to walk exhibited distal muscle weakness of the lower extremities, while a subset (5/16, 31%) also developed weakness of the hands. The distal muscle weakness was often more serious than, and preceded the development of, the proximal muscle weakness.
Collapse
Affiliation(s)
- J J A van den Dorpel
- Center for Lysosomal and Metabolic Diseases, Department of Pediatrics, Erasmus MC University Medical Center, P.O. Box 2060, Rotterdam, 3000, CB, The Netherlands
| | - E Poelman
- Center for Lysosomal and Metabolic Diseases, Department of Pediatrics, Erasmus MC University Medical Center, P.O. Box 2060, Rotterdam, 3000, CB, The Netherlands
| | - L Harlaar
- Center for Lysosomal and Metabolic Diseases, Department of Neurology, Erasmus MC University Medical Center, P.O. Box 2060, Rotterdam, 3000, CB, The Netherlands
| | - H A van Kooten
- Center for Lysosomal and Metabolic Diseases, Department of Neurology, Erasmus MC University Medical Center, P.O. Box 2060, Rotterdam, 3000, CB, The Netherlands
| | - L J van der Giessen
- Center for Lysosomal and Metabolic Diseases, Department of Pediatric Physiotherapy, Erasmus MC University Medical Center, P.O. Box 2060, Rotterdam, 3000, CB, The Netherlands
| | - P A van Doorn
- Center for Lysosomal and Metabolic Diseases, Department of Neurology, Erasmus MC University Medical Center, P.O. Box 2060, Rotterdam, 3000, CB, The Netherlands
| | - A T van der Ploeg
- Center for Lysosomal and Metabolic Diseases, Department of Pediatrics, Erasmus MC University Medical Center, P.O. Box 2060, Rotterdam, 3000, CB, The Netherlands
| | - J M P van den Hout
- Center for Lysosomal and Metabolic Diseases, Department of Pediatrics, Erasmus MC University Medical Center, P.O. Box 2060, Rotterdam, 3000, CB, The Netherlands
| | - N A M E van der Beek
- Center for Lysosomal and Metabolic Diseases, Department of Pediatrics, Erasmus MC University Medical Center, P.O. Box 2060, Rotterdam, 3000, CB, The Netherlands. .,Center for Lysosomal and Metabolic Diseases, Department of Neurology, Erasmus MC University Medical Center, P.O. Box 2060, Rotterdam, 3000, CB, The Netherlands.
| |
Collapse
|
33
|
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
|
34
|
Predicting the Development of Anti-Drug Antibodies against Recombinant alpha-Galactosidase A in Male Patients with Classical Fabry Disease. Int J Mol Sci 2020; 21:ijms21165784. [PMID: 32806627 PMCID: PMC7460974 DOI: 10.3390/ijms21165784] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022] Open
Abstract
Fabry Disease (FD) is a rare, X-linked, lysosomal storage disease that mainly causes renal, cardiac and cerebral complications. Enzyme replacement therapy (ERT) with recombinant alpha-galactosidase A is available, but approximately 50% of male patients with classical FD develop inhibiting anti-drug antibodies (iADAs) that lead to reduced biochemical responses and an accelerated loss of renal function. Once immunization has occurred, iADAs tend to persist and tolerization is hard to achieve. Here we developed a pre-treatment prediction model for iADA development in FD using existing data from 120 classical male FD patients from three European centers, treated with ERT. We found that nonsense and frameshift mutations in the α-galactosidase A gene (p = 0.05), higher plasma lysoGb3 at baseline (p < 0.001) and agalsidase beta as first treatment (p = 0.006) were significantly associated with iADA development. Prediction performance of a Random Forest model, using multiple variables (AUC-ROC: 0.77) was compared to a logistic regression (LR) model using the three significantly associated variables (AUC-ROC: 0.77). The LR model can be used to determine iADA risk in individual FD patients prior to treatment initiation. This helps to determine in which patients adjusted treatment and/or immunomodulatory regimes may be considered to minimize iADA development risk.
Collapse
|
35
|
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
|
36
|
Ying S, Zhihua Z, Yucan Z, Yu J, Qian L, Bixia Z, Weixia C, Zhifeng L. Molecular Diagnosis of Panel-Based Next-Generation Sequencing Approach and Clinical Symptoms in Patients With Glycogen Storage Disease: A Single Center Retrospective Study. Front Pediatr 2020; 8:600446. [PMID: 33344388 PMCID: PMC7744419 DOI: 10.3389/fped.2020.600446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 11/05/2020] [Indexed: 01/01/2023] Open
Abstract
Aim: The aim of this study was to investigate the clinical utility of panel-based next-generation sequencing (NGS) in the diagnostic approach of glycogen storage disease (GSD). Methods: We performed a retrospective review of the 32 cases with suspected GSDs between April 2013 and November 2019 through panel-based NGS, clinical and biochemical data and long-term complications. Results: Of the 32 clinical cases, we identified 41 different variants, including 24 missense (58.5%), one synonymous (2.4%), three nonsense (8%), one splice (2.4%), four frameshift (9.8%), one deletion (2.4%), four insertions (9.8%), two deletion-insertion (4.9%) and one duplication(2.4%), of which 13(31.7%) were previously unreported in the literature. In addition, patients with different types of GSDs showed important differences in biochemical parameters (i.e., CK, rGGT, TG, and UA). Conclusions: The panel-based NGS played an important diagnostic role in the suspicious GSDs patients, especially in the mild phenotype and ruled out detectable pathologic conditions. Besides, differences between our GSDs patients reflect biochemical heterogeneity.
Collapse
Affiliation(s)
- Shen Ying
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhang Zhihua
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Yucan
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Yu
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Qian
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Bixia
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Cheng Weixia
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Liu Zhifeng
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
37
|
Harlaar L, Hogrel JY, Perniconi B, Kruijshaar ME, Rizopoulos D, Taouagh N, Canal A, Brusse E, van Doorn PA, van der Ploeg AT, Laforêt P, van der Beek NAME. Large variation in effects during 10 years of enzyme therapy in adults with Pompe disease. Neurology 2019; 93:e1756-e1767. [PMID: 31619483 PMCID: PMC6946483 DOI: 10.1212/wnl.0000000000008441] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022] Open
Abstract
Objective To determine the effects of 10 years of enzyme replacement therapy (ERT) in adult patients with Pompe disease, focusing on individual variability in treatment response. Methods In this prospective, multicenter cohort study, we studied 30 patients from the Netherlands and France who had started ERT during the only randomized placebo-controlled clinical trial with ERT in late-onset Pompe disease (NCT00158600) or its extension (NCT00455195) in 2005 to 2008. Main outcomes were walking ability (6-minute walk test [6MWT]), muscle strength (manual muscle testing using Medical Research Council [MRC] grading), and pulmonary function (forced vital capacity [FVC] in the upright and supine positions), assessed at 3- to 6-month intervals before and after the start of ERT. Data were analyzed with linear mixed-effects models for repeated measurements. Results Median follow-up duration on ERT was 9.8 years (interquartile range [IQR] 8.3–10.2 years). At the group level, baseline 6MWT was 49% of predicted (IQR 41%–60%) and had deteriorated by 22.2 percentage points (pp) at the 10-year treatment point (p < 0.001). Baseline FVC upright was 54% of predicted (IQR 47%–68%) and decreased by 11 pp over 10 years (p < 0.001). Effects of ERT on MRC sum score and FVC supine were similar. At the individual level, 93% of patients had initial benefit of ERT. Depending on the outcome measured, 35% to 63% of patients had a secondary decline after ≈3 to 5 years. Still, at 10 years of ERT, 52% had equal or better 6MWT and/or FVC upright compared to baseline. Conclusions The majority of patients with Pompe disease benefit from long-term ERT, but many patients experience some secondary decline after ≈3 to 5 years. Individual variation, however, is considerable. Classification of evidence This study provides Class IV evidence that for the majority of adults with Pompe disease, long-term ERT positively affects, or slows deterioration in, muscle strength, walking ability, and/or pulmonary function.
Collapse
Affiliation(s)
- Laurike Harlaar
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Jean-Yves Hogrel
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Barbara Perniconi
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Michelle E Kruijshaar
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Dimitris Rizopoulos
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Nadjib Taouagh
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Aurélie Canal
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Esther Brusse
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Pieter A van Doorn
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Ans T van der Ploeg
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Pascal Laforêt
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Nadine A M E van der Beek
- From the Departments of Neurology (L.H., E.B., P.A.v.D., N.A.M.E.v.d.B.) and Pediatrics (M.E.K., A.T.v.d.P.), Center for Lysosomal and Metabolic Diseases Erasmus MC, and Department of Biostatistics (D.R.), University Medical Center Rotterdam, Netherlands; Institute of Myology (J.-Y.H., B.P., N.T., A.C.), Pitié-Salpêtrière Hospital, Paris; Department of Neurology (P.L.), Nord/Est/Ile de France Neuromuscular Center, Raymond Poincaré Teaching Hospital, AP-HP, Garches; and INSERM U1179 (P.L.), END-ICAP, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France.
| |
Collapse
|
38
|
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] [MESH Headings] [Grants] [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
|
39
|
Peruzzo P, Pavan E, Dardis A. Molecular genetics of Pompe disease: a comprehensive overview. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:278. [PMID: 31392190 PMCID: PMC6642931 DOI: 10.21037/atm.2019.04.13] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/03/2019] [Indexed: 12/27/2022]
Abstract
Pompe disease (PD) is an autosomal recessive lysosomal disorder caused by the deficient activity of acid alpha-glucosidase (GAA) enzyme due to mutations in the GAA gene. The enzymatic deficiency leads to the accumulation of glycogen within the lysosomes. Clinically, the disease has been classically classified in infantile and childhood/adult forms. The GAA gene has been localized to chromosome 17q25.2-q25.3 and to date, 582 mutations distributed throughout the whole gene have been reported (HGMD: http://www.hgmd.cf.ac.uk/ac/). All types of mutations have been described; missense variants are the most frequent type followed by small deletions. Most GAA mutations are private or found in a small number of families. However, an exception is represented by the c.-32-13T>G splice mutation that is very common in patients of Caucasian origin affected by the childhood/adult form of the disease, with an allelic frequency ranging from 40% to 70%. In this article, we review the spectrum of GAA mutations, their distribution in different populations, and their classification according to their impact on GAA splicing process, protein expression and activity. In addition, whenever possible, we discuss the phenotype/genotype correlation. The information collected in this review provides an overview of the molecular genetics of PD and can be used to facilitate diagnosis and genetic counseling of families affected by this disorder.
Collapse
Affiliation(s)
- Paolo Peruzzo
- Regional Coordinator Centre for Rare Diseases, University Hospital Santa Maria della Misericordia, Udine, Italy
| | - Eleonora Pavan
- Regional Coordinator Centre for Rare Diseases, University Hospital Santa Maria della Misericordia, Udine, Italy
| | - Andrea Dardis
- Regional Coordinator Centre for Rare Diseases, University Hospital Santa Maria della Misericordia, Udine, Italy
| |
Collapse
|
40
|
Ronzitti G, Collaud F, Laforet P, Mingozzi F. Progress and challenges of gene therapy for Pompe disease. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:287. [PMID: 31392199 DOI: 10.21037/atm.2019.04.67] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pompe disease (PD) is a monogenic disorder caused by mutations in the acid alpha-glucosidase gene (Gaa). GAA is a lysosomal enzyme essential for the degradation of glycogen. Deficiency of GAA results in a severe, systemic disorder that, in its most severe form, can be fatal. About a decade ago, the prognosis of PD has changed dramatically with the marketing authorization of an enzyme replacement therapy (ERT) based on recombinant GAA. Despite the breakthrough nature of ERT, long-term follow-up of both infantile and late-onset Pompe disease patients (IOPD and LOPD, respectively), revealed several limitations of the approach. In recent years several investigational therapies for PD have entered preclinical and clinical development, with a few next generation ERTs entering late-stage clinical development. Gene therapy holds the potential to change dramatically the way we treat PD, based on the ability to express the Gaa gene long-term, ideally driving enhanced therapeutic efficacy compared to ERT. Several gene therapy approaches to PD have been tested in preclinical animal models, with a handful of early phase clinical trials started or about to start. The complexity of PD and of the endpoints used to measure efficacy of investigational treatments remains a challenge, however the hope is for a future with more therapeutic options for both IOPD and LOPD patients.
Collapse
Affiliation(s)
| | | | - Pascal Laforet
- Raymond Poincaré Teaching Hospital, APHP, Garches, France.,Nord/Est/Ile de France Neuromuscular Center, France
| | | |
Collapse
|
41
|
Hahn A, Schänzer A. Long-term outcome and unmet needs in infantile-onset Pompe disease. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:283. [PMID: 31392195 DOI: 10.21037/atm.2019.04.70] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Infantile-onset Pompe disease (IOPD) is characterized by virtually complete absence of acid alpha-glucosidase (GAA)-activity, resulting in rapidly progressive hypertrophic cardiomyopathy (HCM), profound skeletal muscle weakness, and death usually within the first 12 months of life. Enzyme replacement therapy (ERT) with recombinant GAA in humans started in 1999, and pivotal studies demonstrated that the treatment ameliorated HCM, improved motor function in some patients, and prolonged overall and ventilator-free survival. These outcomes led to the approval of ERT in 2006. Implementation of ERT has uncovered multisystemic character of IOPD, not known in the pre-ERT era. Although ERT has substantially improved the prognosis of IOPD, mortality is still considerable, and decline of motor function with time is frequent in long-term survivors. This review details the new complex IOPD phenotype, outlines problems related to ERT, and highlights unmet needs.
Collapse
Affiliation(s)
- Andreas Hahn
- Department of Child Neurology, Justus-Liebig-University, Giessen, Germany
| | - Anne Schänzer
- Institute of Neuropathology, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
42
|
Poswar FDO, Vairo F, Burin M, Michelin-Tirelli K, Brusius-Facchin AC, Kubaski F, Souza CFMD, Baldo G, Giugliani R. Lysosomal diseases: Overview on current diagnosis and treatment. Genet Mol Biol 2019; 42:165-177. [PMID: 31067291 PMCID: PMC6687355 DOI: 10.1590/1678-4685-gmb-2018-0159] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/30/2018] [Indexed: 12/13/2022] Open
Abstract
Lysosomal diseases (LDs), also known as lysosomal storage diseases (LSDs), are a heterogeneous group of conditions caused by defects in lysosomal function. LDs may result from deficiency of lysosomal hydrolases, membrane-associated transporters or other non-enzymatic proteins. Interest in the LD field is growing each year, as more conditions are, or will soon be treatable. In this article, we review the diagnosis of LDs, from clinical suspicion and screening tests to the identification of enzyme or protein deficiencies and molecular genetic diagnosis. We also cover the treatment approaches that are currently available or in development, including hematopoietic stem cell transplantation, enzyme replacement therapy, small molecules, and gene therapy.
Collapse
Affiliation(s)
- Fabiano de Oliveira Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Filippo Vairo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Maira Burin
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | | | | | - Francyne Kubaski
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | | | - Guilherme Baldo
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Postgraduate Program in Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Physiology and Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Roberto Giugliani
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
43
|
Poelman E, Hoogeveen-Westerveld M, van den Hout JMP, Bredius RGM, Lankester AC, Driessen GJA, Kamphuis SSM, Pijnappel WWM, van der Ploeg AT. Effects of immunomodulation in classic infantile Pompe patients with high antibody titers. Orphanet J Rare Dis 2019; 14:71. [PMID: 30902109 PMCID: PMC6431009 DOI: 10.1186/s13023-019-1039-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/28/2019] [Indexed: 08/26/2023] Open
Abstract
Purpose To evaluate whether immunomodulation can eliminate high sustained antibody levels, and thereby improve clinical outcome in classic infantile Pompe patients receiving enzyme replacement therapy (ERT) with recombinant human alpha-glucosidase (rhGAA). Methods Three patients (two cross-reactive immunologic material (CRIM) negative) with high sustained antibodies received a three-week treatment protocol with Rituximab and Bortezomib, followed by daily Rapamycin and monthly IVIG. Patients received 40 mg/kg/week rhGAA. Antibody titers were measured using ELISA. Neutralizing effects on cellular uptake were determined. Clinical efficacy was measured in terms of (ventilator-free) survival, reduction in left ventricular mass index (LVMI) and improvement in motor function. Results Before immunomodulation anti-rhGAA antibody titers ranged from 1:156,250 to 1:781,250 and at last assessment from 1:31,250 to 1:156,250. Neutralizing effects of anti-rhGAA antibody titers (observed in two patients) disappeared. Infusion-associated reactions were no longer present. Immunomodulation resulted in substantial increases of aspartate transaminase, alanine transaminase, and creatine kinase levels. The two CRIM-negative patients who could walk at start of immunomodulation maintained their ability to walk; the patient who had lost this ability did not regain it. Conclusions To some extent, the immunomodulation protocol used in our study reduced antibody titers, but it did not eliminate them. Overall, there have been few reports on secondary immunomodulation, and various protocols have been applied. Future research should seek to identify the most successful immunomodulation protocol in patients with high sustained titers. Electronic supplementary material The online version of this article (10.1186/s13023-019-1039-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- E Poelman
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, P.O. BOX 2060, 3000, CB, Rotterdam, The Netherlands
| | - M Hoogeveen-Westerveld
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, P.O. BOX 2060, 3000, CB, Rotterdam, The Netherlands
| | - J M P van den Hout
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, P.O. BOX 2060, 3000, CB, Rotterdam, The Netherlands
| | - R G M Bredius
- Department of Pediatrics, Leiden Medical University Center, Leiden, The Netherlands
| | - A C Lankester
- Department of Pediatrics, Leiden Medical University Center, Leiden, The Netherlands
| | - G J A Driessen
- Department of Pediatrics, Juliana Children's Hospital, The Hague, The Netherlands
| | - S S M Kamphuis
- Department of Pediatric Rheumatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - W W M Pijnappel
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, P.O. BOX 2060, 3000, CB, Rotterdam, The Netherlands
| | - A T van der Ploeg
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, P.O. BOX 2060, 3000, CB, Rotterdam, The Netherlands.
| |
Collapse
|
44
|
Lim JA, Yi H, Gao F, Raben N, Kishnani PS, Sun B. Intravenous Injection of an AAV-PHP.B Vector Encoding Human Acid α-Glucosidase Rescues Both Muscle and CNS Defects in Murine Pompe Disease. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 12:233-245. [PMID: 30809555 PMCID: PMC6376130 DOI: 10.1016/j.omtm.2019.01.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/21/2019] [Indexed: 01/01/2023]
Abstract
Pompe disease, a severe and often fatal neuromuscular disorder, is caused by a deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). The disease is characterized by the accumulation of excess glycogen in the heart, skeletal muscle, and CNS. Currently approved enzyme replacement therapy or experimental adeno-associated virus (AAV)-mediated gene therapy has little effect on CNS correction. Here we demonstrate that a newly developed AAV-PHP.B vector can robustly transduce both the CNS and skeletal muscles in GAA-knockout (GAAKO) mice. A single intravenous injection of an AAV-PHP.B vector expressing human GAA under the control of cytomegalovirus (CMV) enhancer-chicken β-actin (CB) promoter into 2-week-old GAAKO mice resulted in widespread GAA expression in the affected tissues. Glycogen contents were reduced to wild-type levels in the brain and heart, and they were significantly decreased in skeletal muscle by the AAV treatment. The histological assay showed no visible glycogen in any region of the brain and spinal cord of AAV-treated mice. In this study, we describe a set of behavioral tests that can detect early neurological deficits linked to extensive lysosomal glycogen accumulation in the CNS of untreated GAAKO mice. Furthermore, we demonstrate that the therapy can help prevent the development of these abnormalities.
Collapse
Affiliation(s)
- Jeong-A Lim
- Department of Pediatrics, Division of Medical Genetics, Duke University School of Medicine, Durham, NC, USA
| | - Haiqing Yi
- Department of Pediatrics, Division of Medical Genetics, Duke University School of Medicine, Durham, NC, USA
| | - Fengqin Gao
- Department of Pediatrics, Division of Medical Genetics, Duke University School of Medicine, Durham, NC, USA
| | - Nina Raben
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Priya S Kishnani
- Department of Pediatrics, Division of Medical Genetics, Duke University School of Medicine, Durham, NC, USA
| | - Baodong Sun
- Department of Pediatrics, Division of Medical Genetics, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
45
|
Cherukuri A, Cahan H, de Hart G, Van Tuyl A, Slasor P, Bray L, Henshaw J, Ajayi T, Jacoby D, O'Neill CA, Schweighardt B. Immunogenicity to cerliponase alfa intracerebroventricular enzyme replacement therapy for CLN2 disease: Results from a Phase 1/2 study. Clin Immunol 2018; 197:68-76. [DOI: 10.1016/j.clim.2018.09.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/23/2018] [Accepted: 09/07/2018] [Indexed: 10/28/2022]
|
46
|
Owens P, Wong M, Bhattacharya K, Ellaway C. Infantile-onset Pompe disease: A case series highlighting early clinical features, spectrum of disease severity and treatment response. J Paediatr Child Health 2018; 54:1255-1261. [PMID: 29889338 DOI: 10.1111/jpc.14070] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/26/2018] [Accepted: 05/02/2018] [Indexed: 02/03/2023]
Abstract
AIM Pompe disease is a rare, autosomal, recessive disorder. Alterations in the gene encoding lysosomal acid alpha-glucosidase cause impaired glycogen degradation and resultant lysosomal glycogen accumulation. Classic infantile-onset Pompe disease (IPD) manifests soon after birth, severe cases have complete/near complete enzyme deficiency. IPD is associated with a broad spectrum of non-specific clinical features, and diagnostic delays are common. Without treatment, death typically occurs within the first 2 years of life. We present case experiences to help expand paediatricians' understanding of factors contributing to diagnostic delay, clinical decline and to highlight the need for timely therapy. METHODS Data were extracted from IPD cases managed at our hospital. Key aspects of clinical presentation, diagnosis, genetic variations, management and overall outcomes were collated then compared with what is already known in the literature. RESULTS We report four IPD cases (three female). Two patients were cross-reactive immunological material negative. Age at symptom onset was 3-9 months, presenting clinical features were varied, and confirmatory diagnosis was significantly delayed in one patient. In concert with the literature, cardiomegaly, ventricular hypertrophy and delayed developmental milestones were seen in all four cases. Our cases demonstrate a range of disease severity, response to enzyme replacement therapy and antibody development. Significant immune responses were seen in two cases (one cross-reactive immunological material positive); despite immunomodulation therapy, both were associated with fatal outcomes. CONCLUSION Timely diagnosis and initiation of enzyme replacement therapy is critical to patient outcomes as IPD progresses rapidly and irreversible changes in clinical status may occur during the delay.
Collapse
Affiliation(s)
- Penny Owens
- Genetic Metabolic Disorders Service, Western Sydney Genetics Program, Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Melanie Wong
- Department of Immunology, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Child and Adolescent Health, University of Sydney, Sydney, New South Wales, Australia
| | - Kaustuv Bhattacharya
- Genetic Metabolic Disorders Service, Western Sydney Genetics Program, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Child and Adolescent Health, University of Sydney, Sydney, New South Wales, Australia.,Discipline of Genetic Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Carolyn Ellaway
- Genetic Metabolic Disorders Service, Western Sydney Genetics Program, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Child and Adolescent Health, University of Sydney, Sydney, New South Wales, Australia.,Discipline of Genetic Medicine, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
47
|
Abstract
Pompe disease is a rare and deadly muscle disorder. As a clinical entity, the disease has been known for over 75 years. While an optimist might be excited about the advances made during this time, a pessimist would note that we have yet to find a cure. However, both sides would agree that many findings in basic science-such as the Nobel prize-winning discoveries of glycogen metabolism, the lysosome, and autophagy-have become the foundation of our understanding of Pompe disease. The disease is a glycogen storage disorder, a lysosomal disorder, and an autophagic myopathy. In this review, we will discuss how these past discoveries have guided Pompe research and impacted recent therapeutic developments.
Collapse
Affiliation(s)
- Lara Kohler
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Nina Raben
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
48
|
Poelman E, Hoogeveen-Westerveld M, Kroos-de Haan MA, van den Hout JMP, Bronsema KJ, van de Merbel NC, van der Ploeg AT, Pijnappel WWMP. High Sustained Antibody Titers in Patients with Classic Infantile Pompe Disease Following Immunomodulation at Start of Enzyme Replacement Therapy. J Pediatr 2018; 195:236-243.e3. [PMID: 29428273 DOI: 10.1016/j.jpeds.2017.11.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/10/2017] [Accepted: 11/20/2017] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To evaluate whether immunomodulation at start of enzyme replacement therapy induces immune tolerance to recombinant human acid alpha-glucosidase (rhGAA) in patients with classic infantile Pompe disease. STUDY DESIGN Three patients (1 cross reactive immunologic material negative, 2 cross reactive immunologic material positive) were treated with 4 weekly doses of rituximab, weekly methotrexate, and monthly intravenous immunoglobulin and enzyme replacement therapy at 40 mg/kg/week. Antibody titers were measured using enzyme-linked immunosorbent assay. Neutralizing effects on rhGAA activity and cellular uptake were determined and combined with pharmacokinetic analysis. Clinical efficacy was evaluated by (ventilator-free) survival, reduction in left ventricular mass index, and improvement of motor function. RESULTS Immunomodulation induced B cell depletion that was accompanied by absence of antibody formation in all 3 patients. Upon cessation of rituximab treatment, all 3 patients showed B cell recovery, which was accompanied by formation of very high sustained antibody titers in 2 patients. Neutralizing effects on infused rhGAA were low to mild/moderate. All patients were alive at study end, learned to walk, and showed (near) normalization of left ventricular mass index. CONCLUSIONS Immunomodulation as recommended in the literature prevented formation of rhGAA antibodies only during B cell depletion but failed to induce immune tolerance in 2 out of 3 patients.
Collapse
Affiliation(s)
- Esther Poelman
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pediatrics, Division of Metabolic Diseases and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Marianne Hoogeveen-Westerveld
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marian A Kroos-de Haan
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Johanna M P van den Hout
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pediatrics, Division of Metabolic Diseases and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Kees J Bronsema
- Bioanalytical Laboratory, PRA Health Sciences, Early Development Services, Assen, The Netherlands; Analytical Biochemistry, Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Nico C van de Merbel
- Bioanalytical Laboratory, PRA Health Sciences, Early Development Services, Assen, The Netherlands; Analytical Biochemistry, Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Ans T van der Ploeg
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pediatrics, Division of Metabolic Diseases and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - W W M Pim Pijnappel
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pediatrics, Division of Metabolic Diseases and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
49
|
Parini R, De Lorenzo P, Dardis A, Burlina A, Cassio A, Cavarzere P, Concolino D, Della Casa R, Deodato F, Donati MA, Fiumara A, Gasperini S, Menni F, Pagliardini V, Sacchini M, Spada M, Taurisano R, Valsecchi MG, Di Rocco M, Bembi B. Long term clinical history of an Italian cohort of infantile onset Pompe disease treated with enzyme replacement therapy. Orphanet J Rare Dis 2018; 13:32. [PMID: 29422078 PMCID: PMC5806382 DOI: 10.1186/s13023-018-0771-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/25/2018] [Indexed: 01/12/2023] Open
Abstract
Background Enzyme replacement therapy (ERT) has deeply modified the clinical history of Infantile Onset Pompe Disease (IOPD). However, its long-term effectiveness is still not completely defined. Available data shows a close relationship between clinical outcome and patients’ cross-reactive immunological status (CRIM), being CRIM-negative status a negative prognostic factor. At the same time limited data are available on the long-term treatment in CRIM-positive infants. Methods A retrospective multicentre observational study was designed to analyse the long-term effectiveness of ERT in IOPD. Thirteen Italian centres spread throughout the country were involved and a cohort of 28 patients (15 females, 13 males, born in the period: February 2002–January 2013) was enrolled. IOPD diagnosis was based on clinical symptoms, enzymatic and molecular analysis. All patients received ERT within the first year of life. Clinical, laboratory, and functional data (motor, cardiac and respiratory) were collected and followed for a median period of 71 months (5 years 11 months). Results Median age at onset, diagnosis and start of ERT were 2, 3 and 4 months, respectively. CRIM status was available for 24/28 patients: 17/24 (71%) were CRIM-positive. Nineteen patients (67%) survived > 2 years: 4 were CRIM-negative, 14 CRIM-positive and one unknown. Six patients (5 CRIM-positive and one unknown) never needed ventilation support (21,4%) and seven (6 CRIM-positive and one unknown: 25%) developed independent ambulation although one subsequently lost this function. Brain imaging study was performed in 6 patients and showed peri-ventricular white matter abnormalities in all of them. Clinical follow-up confirmed the better prognosis for CRIM-positive patients, though a slow, progressive worsening of motor and/or respiratory functions was detected in 8 patients. Conclusions These data are the result of the longest independent retrospective study on ERT in IOPD reported so far outside clinical trials. The data obtained confirmed the better outcome of the CRIM-positive patients but at the same time, showed the inability of the current therapeutic approach to reverse or stabilize the disease progression. The results also evidenced the involvement of central nervous system in Pompe disease. To better understand the disease clinical history and to improve treatment efficacy larger multicentre studies are needed as well as the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Rossella Parini
- Pediatric Rare Diseases Unit, Department of Pediatrics, MBBM Foundation, ATS Monza e Brianza, Via Pergolesi 33, 20900, Monza, Italy.
| | - Paola De Lorenzo
- Centre of Biostatistics for Clinical Epidemiology, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Andrea Dardis
- Centre for Rare Diseases, University Hospital Santa Maria della Misericordia, Udine, Italy
| | - Alberto Burlina
- Department for Women and Children's Health, U.O.C. Inborn Metabolic Diseases, University Hospital, Padova, Italy
| | | | - Paolo Cavarzere
- Department of Pediatrics, University Magna Graecia, Catanzaro, Italy
| | - Daniela Concolino
- Department of Pediatrics, University Magna Graecia, Catanzaro, Italy
| | - Roberto Della Casa
- Department of Translational Sciences, Pediatrics, University Federico II, Naples, Italy
| | - Federica Deodato
- Division of Metabolism Bambino Gesù Children's Hospital, Rome, Italy
| | - Maria Alice Donati
- Department of Pediatrics, Meyer Children's Hospital, Metabolic and Muscular Unit, University of Firenze, Florence, Italy
| | - Agata Fiumara
- Department of Clinical and Experimental Medicine, Metabolic Diseases, Pediatric Clinic, University of Catania, Catania, Italy
| | - Serena Gasperini
- Pediatric Rare Diseases Unit, Department of Pediatrics, MBBM Foundation, ATS Monza e Brianza, Via Pergolesi 33, 20900, Monza, Italy
| | - Francesca Menni
- Department of Pathophysiology and Transplantation, Pediatric Highly Intensive Care Unit, University of Milano, IRCCS Ca' Granda Ospedale Maggiore Policlinico Foundation, Milan, Italy
| | | | - Michele Sacchini
- Department of Pediatrics, Meyer Children's Hospital, Metabolic and Muscular Unit, University of Firenze, Florence, Italy
| | - Marco Spada
- Department of Pediatrics, University of Torino, Torino, Italy
| | - Roberta Taurisano
- Division of Metabolism Bambino Gesù Children's Hospital, Rome, Italy
| | - Maria Grazia Valsecchi
- Centre of Biostatistics for Clinical Epidemiology, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Maja Di Rocco
- Rare Diseases Unit, Pediatric Hospital Istituto Giannina Gaslini, Genoa, Italy
| | - Bruno Bembi
- Centre for Rare Diseases, University Hospital Santa Maria della Misericordia, Udine, Italy
| |
Collapse
|
50
|
Nelson BC, Hashem SI, Adler ED. Human-Induced Pluripotent Stem Cell-Based Modeling of Cardiac Storage Disorders. Curr Cardiol Rep 2017; 19:26. [PMID: 28251514 DOI: 10.1007/s11886-017-0829-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW The aim of this study is to review the published human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) models of cardiac storage disorders and to evaluate the limitations and future applications of this technology. RECENT FINDINGS Several cardiac storage disorders (CSDs) have been modeled using patient-specific hiPSC-CMs, including Anderson-Fabry disease, Danon disease, and Pompe disease. These models have shown that patient-specific hiPSC-CMs faithfully recapitulate key phenotypic features of CSDs and respond predictably to pharmacologic manipulation. hiPSC-CMs generated from patients with CSDs are representative models of the patient disease state and can be used as an in vitro system for the study of human cardiomyocytes. While these models suffer from several limitations, they are likely to play an important role in future mechanistic studies of cardiac storage disorders and the development of targeted therapeutics for these diseases.
Collapse
Affiliation(s)
- Bradley C Nelson
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, Biomedical Research Facility, Room 1217 AA, La Jolla, CA, 92093, USA
| | - Sherin I Hashem
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, Biomedical Research Facility, Room 1217 AA, La Jolla, CA, 92093, USA
| | - Eric D Adler
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, Biomedical Research Facility, Room 1217 AA, La Jolla, CA, 92093, USA.
| |
Collapse
|