1
|
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
|
2
|
Schoser B, Raben N, Varfaj F, Walzer M, Toscano A. Acid α-glucosidase (GAA) activity and glycogen content in muscle biopsy specimens of patients with Pompe disease: A systematic review. Mol Genet Metab Rep 2024; 39:101085. [PMID: 38698877 PMCID: PMC11064613 DOI: 10.1016/j.ymgmr.2024.101085] [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: 12/13/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024] Open
Abstract
Pompe disease is a rare genetic disorder characterized by a deficiency of acid α-glucosidase (GAA), leading to the accumulation of glycogen in various tissues, especially in skeletal muscles. The disease manifests as a large spectrum of phenotypes from infantile-onset Pompe disease (IOPD) to late-onset Pompe disease (LOPD), depending on the age of symptoms onset. Quantifying GAA activity and glycogen content in skeletal muscle provides important information about the disease severity. However, the distribution of GAA and glycogen levels in skeletal muscles from healthy individuals and those impacted by Pompe disease remains poorly understood, and there is currently no universally accepted standard assay for GAA activity measurement. This systematic literature review aims to provide an overview of the available information on GAA activity and glycogen content levels in skeletal muscle biopsies from patients with Pompe disease. A structured review of PubMed and Google Scholar literature (with the latter used to check that no additional publications were identified) was conducted to identify peer-reviewed publications on glycogen storage disease type II [MeSH term] + GAA, protein human (supplementary concept), Pompe, muscle; and muscle, acid alpha-glucosidase. A limit of English language was applied. Results were grouped by methodologies used to quantify GAA activity and glycogen content in skeletal muscle. The search and selection strategy were devised and carried out in line with Preferred Reporting of Items in Systematic Reviews and Meta-Analysis guidelines and documented using a flowchart. Bibliographies of papers included in the analysis were reviewed and applicable publications not already identified in the search were included. Of the 158 articles retrieved, 24 (comprising >100 muscle biopsies from >100 patients) were included in the analysis, with four different assays. Analysis revealed that patients with IOPD exhibited markedly lower GAA activity in skeletal muscles than those with LOPD, regardless of the measurement method employed. Additionally, patients with IOPD had notably higher glycogen content levels in skeletal muscles than those with LOPD. In general, however, it was difficult to fully characterize GAA activity because of the different methods used. The findings underscore the challenges in the interpretation and comparison of the results across studies because of the substantial methodological variations. There is a need to establish standardized reference ranges of GAA activity and glycogen content in healthy individuals and in Pompe disease patients based on globally standardized methods to improve comparability and reliability in assessing this rare disease.
Collapse
Affiliation(s)
- Benedikt Schoser
- Friedrich-Baur-Institute, Department of Neurology, LMU Klinikum, Ludwig-Maximilians University, Munich, Germany
| | | | | | - Mark Walzer
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Antonio Toscano
- ERN-NMD Center of Messina for Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
3
|
Budzynska K, Siemionow M, Stawarz K, Chambily L, Siemionow K. Chimeric Cell Therapies as a Novel Approach for Duchenne Muscular Dystrophy (DMD) and Muscle Regeneration. Biomolecules 2024; 14:575. [PMID: 38785982 PMCID: PMC11117592 DOI: 10.3390/biom14050575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Chimerism-based strategies represent a pioneering concept which has led to groundbreaking advancements in regenerative medicine and transplantation. This new approach offers therapeutic potential for the treatment of various diseases, including inherited disorders. The ongoing studies on chimeric cells prompted the development of Dystrophin-Expressing Chimeric (DEC) cells which were introduced as a potential therapy for Duchenne Muscular Dystrophy (DMD). DMD is a genetic condition that leads to premature death in adolescent boys and remains incurable with current methods. DEC therapy, created via the fusion of human myoblasts derived from normal and DMD-affected donors, has proven to be safe and efficacious when tested in experimental models of DMD after systemic-intraosseous administration. These studies confirmed increased dystrophin expression, which correlated with functional and morphological improvements in DMD-affected muscles, including cardiac, respiratory, and skeletal muscles. Furthermore, the application of DEC therapy in a clinical study confirmed its long-term safety and efficacy in DMD patients. This review summarizes the development of chimeric cell technology tested in preclinical models and clinical studies, highlighting the potential of DEC therapy in muscle regeneration and repair, and introduces chimeric cell-based therapies as a promising, novel approach for muscle regeneration and the treatment of DMD and other neuromuscular disorders.
Collapse
Affiliation(s)
- Katarzyna Budzynska
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60607, USA; (K.B.); (K.S.); (L.C.); (K.S.)
| | - Maria Siemionow
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60607, USA; (K.B.); (K.S.); (L.C.); (K.S.)
- Chair and Department of Traumatology, Orthopaedics, and Surgery of the Hand, Poznan University of Medical Sciences, 61-545 Poznan, Poland
| | - Katarzyna Stawarz
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60607, USA; (K.B.); (K.S.); (L.C.); (K.S.)
| | - Lucile Chambily
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60607, USA; (K.B.); (K.S.); (L.C.); (K.S.)
| | - Krzysztof Siemionow
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60607, USA; (K.B.); (K.S.); (L.C.); (K.S.)
| |
Collapse
|
4
|
Do H, Meena NK, Raben N. Failure of Autophagy in Pompe Disease. Biomolecules 2024; 14:573. [PMID: 38785980 PMCID: PMC11118179 DOI: 10.3390/biom14050573] [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: 04/08/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Autophagy is an evolutionarily conserved lysosome-dependent degradation of cytoplasmic constituents. The system operates as a critical cellular pro-survival mechanism in response to nutrient deprivation and a variety of stress conditions. On top of that, autophagy is involved in maintaining cellular homeostasis through selective elimination of worn-out or damaged proteins and organelles. The autophagic pathway is largely responsible for the delivery of cytosolic glycogen to the lysosome where it is degraded to glucose via acid α-glucosidase. Although the physiological role of lysosomal glycogenolysis is not fully understood, its significance is highlighted by the manifestations of Pompe disease, which is caused by a deficiency of this lysosomal enzyme. Pompe disease is a severe lysosomal glycogen storage disorder that affects skeletal and cardiac muscles most. In this review, we discuss the basics of autophagy and describe its involvement in the pathogenesis of muscle damage in Pompe disease. Finally, we outline how autophagic pathology in the diseased muscles can be used as a tool to fast track the efficacy of therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Nina Raben
- M6P Therapeutics, 20 S. Sarah Street, St. Louis, MO 63108, USA; (H.D.); (N.K.M.)
| |
Collapse
|
5
|
Smith EC, Hopkins S, Case LE, Xu M, Walters C, Dearmey S, Han SO, Spears TG, Chichester JA, Bossen EH, Hornik CP, Cohen JL, Bali D, Kishnani PS, Koeberl DD. Phase I study of liver depot gene therapy in late-onset Pompe disease. Mol Ther 2023; 31:1994-2004. [PMID: 36805083 PMCID: PMC10362382 DOI: 10.1016/j.ymthe.2023.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/03/2023] [Accepted: 02/16/2023] [Indexed: 02/21/2023] Open
Abstract
Gene therapy with an adeno-associated virus serotype 8 (AAV8) vector (AAV8-LSPhGAA) could eliminate the need for enzyme replacement therapy (ERT) by creating a liver depot for acid α-glucosidase (GAA) production. We report initial safety and bioactivity of the first dose (1.6 × 1012 vector genomes/kg) cohort (n = 3) in a 52-week open-label, single-dose, dose-escalation study (NCT03533673) in patients with late-onset Pompe disease (LOPD). Subjects discontinued biweekly ERT after week 26 based on the detection of elevated serum GAA activity and the absence of clinically significant declines per protocol. Prednisone (60 mg/day) was administered as immunoprophylaxis through week 4, followed by an 11-week taper. All subjects demonstrated sustained serum GAA activities from 101% to 235% of baseline trough activity 2 weeks following the preceding ERT dose. There were no treatment-related serious adverse events. No subject had anti-capsid T cell responses that decreased transgene expression. Muscle biopsy at week 24 revealed unchanged muscle glycogen content in two of three subjects. At week 52, muscle GAA activity for the cohort was significantly increased (p < 0.05). Overall, these initial data support the safety and bioactivity of AAV8-LSPhGAA, the safety of withdrawing ERT, successful immunoprophylaxis, and justify continued clinical development of AAV8-LSPhGAA therapy in Pompe disease.
Collapse
Affiliation(s)
- Edward C Smith
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Sam Hopkins
- Asklepios Biopharmaceutical, Inc. (Askbio), Durham, NC, USA
| | - Laura E Case
- Department of Orthopedics, Duke University School of Medicine, Durham, NC, USA
| | - Ming Xu
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Crista Walters
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Stephanie Dearmey
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Sang-Oh Han
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Tracy G Spears
- Clinical Trials Statistics, Duke Clinical Research Institute, Durham, NC, USA
| | - Jessica A Chichester
- Immunology Core, Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward H Bossen
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Christoph P Hornik
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Jennifer L Cohen
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Deeksha Bali
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Priya S Kishnani
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Dwight D Koeberl
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
6
|
Chen YH, Huang PW, Liu YJ, Tsai YJ. Blepharoptosis in infantile onset Pompe disease: Histological findings and surgical outcomes. Mol Genet Metab Rep 2023; 35:100969. [PMID: 36967722 PMCID: PMC10034147 DOI: 10.1016/j.ymgmr.2023.100969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
This retrospective observational case series is to evaluate the histopathological findings of drooping eyelids in patients with infantile-onset Pompe disease and assess the feasibility of levator muscle resection combined with conjoint fascial sheath suspension for ptosis correction. It included six patients from a single tertiary referral center with ptosis and infantile-onset Pompe disease between January 1, 2013, and December 31, 2021. They most suffered from recurrent ptosis after initial surgical correction (6/11 eyes, 54.55%). The recurrence rate was high in eyes with levator muscle resection alone (4/6 eyes, 66.67%). No recurrence of ptosis was observed in eyes with levator muscle resection combined with conjoint fascial sheath suspension. The follow-up period was approximately 16-94 months. Histopathological examination revealed that the levator muscle had the most abundant glycogen accumulation-related vacuolar changes, followed by Müller's muscle and extraocular muscles. No vacuolar changes were observed in the conjoint fascial sheath. For patients with infantile-onset Pompe disease-related ptosis, performing levator muscle resection alone is not sufficient, while utilizing conjoint fascial sheath suspension can achieve the desired long-term outcomes with minimal recurrence. These findings may have important implications for the management of ophthalmic complications in patients with infantile-onset Pompe disease.
Collapse
|
7
|
Scheffers LE, Kok R, van den Berg LE, Jmp H, Boersma E, van Capelle CI, Helbing WA, Ploeg AT, Koopman LP. Effects of enzyme replacement therapy on cardiac function in classic infantile Pompe disease. Int J Cardiol 2023; 380:65-71. [PMID: 36893858 DOI: 10.1016/j.ijcard.2023.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/17/2023] [Accepted: 03/05/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVE Patients with classic infantile Pompe disease are born with a hypertrophic cardiomyopathy, which resolves after treatment with Enzyme replacement therapy (ERT). We aimed to assess potential deterioration of cardiac function over time using myocardial deformation analysis. METHODS Twenty-seven patients treated with ERT were included. Cardiac function was assessed at regular time intervals (before and after start with ERT) using conventional echocardiography and myocardial deformation analysis. Separate linear mixed effect models were used to asses temporal changes within the first year and the long-term follow-up period. Echocardiograms of 103 healthy children served as controls. RESULTS A total of 192 echocardiograms were analyzed. Median follow-up was 9.9 years (IQR: 7.5-16.3). Mean LVMI before start of ERT was increased 292.3 g/m2 (95% CI: 202.8-381.8, mean Z-score + 7.6) and normalized after 1 year of ERT 87.3 g/m2 (CI: 67.5-107.1, mean Z-score + 0.8, p < 0.001). Mean shortening fraction was within normal limits before start of ERT, up to 22 years of follow-up. Cardiac function measured by RV/LV longitudinal, and circumferential strain was diminished before start of ERT, but normalized (<-16%) within 1 year after start of ERT, and all remained within normal limits during follow-up. Only LV circumferential strain gradually worsened in Pompe patients (+0.24%/year) during follow-up compared to controls. LV longitudinal strain was diminished in Pompe patients, but did not change significantly over time compared to controls. CONCLUSION Cardiac function, measured using myocardial deformation analysis, normalizes after start of ERT, and seems to remain stable over a median follow-up period of 9.9 years.
Collapse
Affiliation(s)
- L E Scheffers
- Department of Pediatric Cardiology, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus MC- Sophia children's hospital, Rotterdam, the Netherlands.
| | - R Kok
- Department of Pediatric Cardiology, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - L E van den Berg
- Department of Pediatric Cardiology, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus MC- Sophia children's hospital, Rotterdam, the Netherlands; Department of Orthopedics and Sports Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Hout Jmp
- Center for Lysosomal and Metabolic Diseases, Erasmus MC- Sophia children's hospital, Rotterdam, the Netherlands
| | - E Boersma
- Department of Cardiology, Erasmus MC- Sophia children's hospital, Rotterdam, the Netherlands
| | - C I van Capelle
- Department of Pediatric Cardiology, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - W A Helbing
- Department of Pediatric Cardiology, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pediatrics, division of Cardiology, Radboud umc - Amalia Children's Hospital, Nijmegen, the Netherlands; Department of Radiology, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - A T Ploeg
- Center for Lysosomal and Metabolic Diseases, Erasmus MC- Sophia children's hospital, Rotterdam, the Netherlands
| | - L P Koopman
- Department of Pediatric Cardiology, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands
| |
Collapse
|
8
|
Nilsson MI, Crozier M, Di Carlo A, Xhuti D, Manta K, Roik LJ, Bujak AL, Nederveen JP, Tarnopolsky MG, Hettinga B, Meena NK, Raben N, Tarnopolsky MA. Nutritional co-therapy with 1,3-butanediol and multi-ingredient antioxidants enhances autophagic clearance in Pompe disease. Mol Genet Metab 2022; 137:228-240. [PMID: 35718712 DOI: 10.1016/j.ymgme.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/03/2022] [Accepted: 06/04/2022] [Indexed: 10/18/2022]
Abstract
Alglucosidase alpha is an orphan drug approved for enzyme replacement therapy (ERT) in Pompe disease (PD); however, its efficacy is limited in skeletal muscle because of a partial blockage of autophagic flux that hinders intracellular trafficking and enzyme delivery. Adjunctive therapies that enhance autophagic flux and protect mitochondrial integrity may alleviate autophagic blockage and oxidative stress and thereby improve ERT efficacy in PD. In this study, we compared the benefits of ERT combined with a ketogenic diet (ERT-KETO), daily administration of an oral ketone precursor (1,3-butanediol; ERT-BD), a multi-ingredient antioxidant diet (ERT-MITO; CoQ10, α-lipoic acid, vitamin E, beetroot extract, HMB, creatine, and citrulline), or co-therapy with the ketone precursor and multi-ingredient antioxidants (ERT-BD-MITO) on skeletal muscle pathology in GAA-KO mice. We found that two months of 1,3-BD administration raised circulatory ketone levels to ≥1.2 mM, attenuated autophagic buildup in type 2 muscle fibers, and preserved muscle strength and function in ERT-treated GAA-KO mice. Collectively, ERT-BD was more effective vs. standard ERT and ERT-KETO in terms of autophagic clearance, dampening of oxidative stress, and muscle maintenance. However, the addition of multi-ingredient antioxidants (ERT-BD-MITO) provided the most consistent benefits across all outcome measures and normalized mitochondrial protein expression in GAA-KO mice. We therefore conclude that nutritional co-therapy with 1,3-butanediol and multi-ingredient antioxidants may provide an alternative to ketogenic diets for inducing ketosis and enhancing autophagic flux in PD patients.
Collapse
Affiliation(s)
- Mats I Nilsson
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada; Exerkine Corporation, McMaster University, Hamilton, Ontario, Canada
| | - Michael Crozier
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Alessia Di Carlo
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Donald Xhuti
- Exerkine Corporation, McMaster University, Hamilton, Ontario, Canada
| | - Katherine Manta
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Liza J Roik
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Adam L Bujak
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Joshua P Nederveen
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | | | - Bart Hettinga
- Exerkine Corporation, McMaster University, Hamilton, Ontario, Canada
| | - Naresh K Meena
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Nina Raben
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada; Exerkine Corporation, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
9
|
Campkin DM, Shimadate Y, Bartholomew B, Bernhardt PV, Nash RJ, Sakoff JA, Kato A, Simone MI. Borylated 2,3,4,5-Tetrachlorophthalimide and Their 2,3,4,5-Tetrachlorobenzamide Analogues: Synthesis, Their Glycosidase Inhibition and Anticancer Properties in View to Boron Neutron Capture Therapy. Molecules 2022; 27:3447. [PMID: 35684388 PMCID: PMC9182199 DOI: 10.3390/molecules27113447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
Tetrachlorinated phthalimide analogues bearing a boron-pinacolate ester group were synthesised via two synthetic routes and evaluated in their glycosidase modulating and anticancer properties, with a view to use them in boron neutron capture therapy (BNCT), a promising radiation type for cancer, as this therapy does little damage to biological tissue. An unexpected decarbonylation/decarboxylation to five 2,3,4,5-tetrachlorobenzamides was observed and confirmed by X-ray crystallography studies, thus, giving access to a family of borylated 2,3,4,5-tetrachlorobenzamides. Biological evaluation showed the benzamide drugs to possess good to weak potencies (74.7-870 μM) in the inhibition of glycosidases, and to have good to moderate selectivity in the inhibition of a panel of 18 glycosidases. Furthermore, in the inhibition of selected glycosidases, there is a core subset of three animal glycosidases, which is always inhibited (rat intestinal maltase α-glucosidase, bovine liver β-glucosidase and β-galactosidase). This could indicate the involvement of the boron atom in the binding. These glycosidases are targeted for the management of diabetes, viral infections (via a broad-spectrum approach) and lysosomal storage disorders. Assays against cancer cell lines revealed potency in growth inhibition for three molecules, and selectivity for one of these molecules, with the growth of the normal cell line MCF10A not being affected by this compound. One of these molecules showed both potency and selectivity; thus, it is a candidate for further study in this area. This paper provides numerous novel aspects, including expedited access to borylated 2,3,4,5-tetrachlorophthalimides and to 2,3,4,5-tetrachlorobenzamides. The latter constitutes a novel family of glycosidase modulating drugs. Furthermore, a greener synthetic access to such structures is described.
Collapse
Affiliation(s)
- David M. Campkin
- Discipline of Chemistry, University of Newcastle, Callaghan, NSW 2308, Australia;
- Priority Research Centre for Drug Development, University of Newcastle, Callaghan, NSW 2308, Australia;
| | - Yuna Shimadate
- Department of Hospital Pharmacy, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.S.); (A.K.)
| | - Barbara Bartholomew
- Phytoquest Ltd., Plas Gogerddan, Aberystwyth, Ceredigion SY23 3EB, UK; (B.B.); (R.J.N.)
| | - Paul V. Bernhardt
- School of Chemistry & Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia;
| | - Robert J. Nash
- Phytoquest Ltd., Plas Gogerddan, Aberystwyth, Ceredigion SY23 3EB, UK; (B.B.); (R.J.N.)
| | - Jennette A. Sakoff
- Priority Research Centre for Drug Development, University of Newcastle, Callaghan, NSW 2308, Australia;
- Calvary Mater Newcastle Hospital, Edith Street, Waratah, NSW 2298, Australia
| | - Atsushi Kato
- Department of Hospital Pharmacy, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.S.); (A.K.)
| | - Michela I. Simone
- Discipline of Chemistry, University of Newcastle, Callaghan, NSW 2308, Australia;
- Priority Research Centre for Drug Development, University of Newcastle, Callaghan, NSW 2308, Australia;
| |
Collapse
|
10
|
Han SO, Gheorghiu D, Chang A, Mapatano SH, Li S, Brooks E, Koeberl D. Efficacious Androgen Hormone Administration in Combination with Adeno-Associated Virus Vector-Mediated Gene Therapy in Female Mice with Pompe Disease. Hum Gene Ther 2022; 33:479-491. [PMID: 35081735 PMCID: PMC9142766 DOI: 10.1089/hum.2021.218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/24/2022] [Indexed: 11/12/2022] Open
Abstract
Pompe disease is an autosomal recessive lysosomal storage disorder caused by deficiency of acid α-glucosidase (GAA), resulting in skeletal muscle weakness and cardiomyopathy that progresses despite currently available therapy in some patients. The development of gene therapy with adeno-associated virus (AAV) vectors revealed a sex-dependent decrease in efficacy in female mice with Pompe disease. This study evaluated the effect of testosterone on gene therapy with an AAV2/8 vector containing a liver-specific promoter to drive expression of GAA (AAV2/8-LSPhGAA) in female GAA-knockout (KO) mice that were implanted with pellets containing testosterone propionate before vector administration. Six weeks after treatment, neuromuscular function and muscle strength were improved as demonstrated by increased Rotarod and wirehang latency for female mice treated with testosterone and vector, in comparison with vector alone. Biochemical correction improved after the addition of testosterone as demonstrated by increased GAA activity and decreased glycogen content in the skeletal muscles of female mice treated with testosterone and vector, in comparison with vector alone. An alternative androgen, oxandrolone, was evaluated similarly to reveal increased GAA in the diaphragm and extensor digitorum longus of female GAA-KO mice after oxandrolone administration; however, glycogen content was unchanged by oxandrolone treatment. The efficacy of androgen hormone treatment in females correlated with increased mannose-6-phosphate receptor in skeletal muscle. These data confirmed the benefits of brief treatment with an androgen hormone in mice with Pompe disease during gene therapy.
Collapse
Affiliation(s)
- Sang-oh Han
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Dorothy Gheorghiu
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Alex Chang
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sweet Hope Mapatano
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Songtao Li
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Elizabeth Brooks
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Laboratory Animal Resources, Duke University, Durham, North Carolina, USA
| | - Dwight Koeberl
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
11
|
Han S, Gheorghiu DB, Li S, Kang HR, Koeberl D. Minimum Effective Dose to Achieve Biochemical Correction With AAV Vector-Mediated Gene Therapy in Mice With Pompe Disease. Hum Gene Ther 2022; 33:492-498. [PMID: 35102744 DOI: 10.1089/hum.2021.252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pompe disease is an autosomal recessive lysosomal storage disorder caused by deficiency of acid α-glucosidase (GAA), resulting in skeletal muscle weakness and cardiomyopathy. Muscle weakness progresses despite currently available therapy, which has prompted the development of gene therapy with adeno-associated virus (AAV) type 2 vectors cross-packaged as AAV8 (2/8). Preclinical studies of gene therapy demonstrated that the minimum effective dose for biochemical correction with AAV2/8-LSPhGAA was approximately 2 x 1011 vector genomes (vg)/kg body weight. The current study examined the transduction of AAV2/8-LSPeGFP vector in adult GAA-KO mice with Pompe disease, and correlated that degree of transduction with the biochemical correction achieved by the same dose of AAV2/8-LSPhGAA. The minimum effective dose was found to be approximately 2 x 1011 vg/kg, with all hepatocytes variably transducing at this dose. At this dose, liver GAA significantly increased, while liver glycogen significantly decreased. The 2 x 1011 vg/kg dose was sufficient to significantly decrease diaphragm glycogen. However, the heart, diaphragm, and quadriceps all required a four-fold higher dose to achieve correction of GAA deficiency in association with significant clearance of stored glycogen, which correlated with increased serum GAA activity. These data indicate that AAV2/8-LSPeGFP transduced all hepatocytes when the 2 x 1011 vg/kg dose was administered, which correlated with partial biochemical correction from the equivalent dose of AAV2/8-LSPhGAA. Together these data support the conclusion that substantial transduction of the liver is required to achieve biochemical correction from AAV2/8-LSPhGAA.
Collapse
Affiliation(s)
- Sangoh Han
- Duke University Department of Pediatrics, 200759, Pediatrics, 905 LaSalle St., GSRBI RM 4048, Durham, North Carolina, United States, 27710;
| | - Dorothy Brooke Gheorghiu
- Duke University Medical Center, 22957, Pediatric Medical Genetics, 905 S Lasalle St, Durham, North Carolina, United States, 27710-4699;
| | - Songtao Li
- Duke University School of Medicine, 12277, Pediatrics, Durham, North Carolina, United States;
| | - Hye Ri Kang
- UT Southwestern, 12334, Pediatrics, Dallas, Texas, United States;
| | - Dwight Koeberl
- Duke University School of Medicine, 12277, Pediatrics, DUMC 103856, Durham, North Carolina, United States, 27710;
| |
Collapse
|
12
|
Unnisa Z, Yoon JK, Schindler JW, Mason C, van Til NP. Gene Therapy Developments for Pompe Disease. Biomedicines 2022; 10:302. [PMID: 35203513 PMCID: PMC8869611 DOI: 10.3390/biomedicines10020302] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/05/2023] Open
Abstract
Pompe disease is an inherited neuromuscular disorder caused by deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). The most severe form is infantile-onset Pompe disease, presenting shortly after birth with symptoms of cardiomyopathy, respiratory failure and skeletal muscle weakness. Late-onset Pompe disease is characterized by a slower disease progression, primarily affecting skeletal muscles. Despite recent advancements in enzyme replacement therapy management several limitations remain using this therapeutic approach, including risks of immunogenicity complications, inability to penetrate CNS tissue, and the need for life-long therapy. The next wave of promising single therapy interventions involves gene therapies, which are entering into a clinical translational stage. Both adeno-associated virus (AAV) vectors and lentiviral vector (LV)-mediated hematopoietic stem and progenitor (HSPC) gene therapy have the potential to provide effective therapy for this multisystemic disorder. Optimization of viral vector designs, providing tissue-specific expression and GAA protein modifications to enhance secretion and uptake has resulted in improved preclinical efficacy and safety data. In this review, we highlight gene therapy developments, in particular, AAV and LV HSPC-mediated gene therapy technologies, to potentially address all components of the neuromuscular associated Pompe disease pathology.
Collapse
Affiliation(s)
- Zeenath Unnisa
- AVROBIO, Inc., Cambridge, MA 02139, USA; (Z.U.); (J.K.Y.); (J.W.S.); (C.M.)
| | - John K. Yoon
- AVROBIO, Inc., Cambridge, MA 02139, USA; (Z.U.); (J.K.Y.); (J.W.S.); (C.M.)
| | | | - Chris Mason
- AVROBIO, Inc., Cambridge, MA 02139, USA; (Z.U.); (J.K.Y.); (J.W.S.); (C.M.)
- Advanced Centre for Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - Niek P. van Til
- AVROBIO, Inc., Cambridge, MA 02139, USA; (Z.U.); (J.K.Y.); (J.W.S.); (C.M.)
- Child Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
13
|
Murray AK. The Release of a Soluble Glycosylated Protein from Glycogen by Recombinant Lysosomal α-Glucosidase (rhGAA) In Vitro and Its Presence in Serum In Vivo. Biomolecules 2020; 10:E1613. [PMID: 33260301 PMCID: PMC7761001 DOI: 10.3390/biom10121613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 01/19/2023] Open
Abstract
In studies on the degradation of glycogen by rhGAA, a glycosylated protein core material was found which consists of about 5-6% of the total starting glycogen. There was an additional 25% of the glycogen unaccounted for based on glucose released. After incubation of glycogen with rhGAA until no more glucose was released, no other carbohydrate was detected on HPAEC-PAD. Several oligosaccharides are then detectable if the medium is first boiled in 0.1 N HCl or incubated with trypsin. It is present in serum either in an HCl extract or in a trypsin digest. The characteristics of the in vivo serum material are identical to the material in the in vitro incubation medium. One oligosaccharide cannot be further degraded by rhGAA, from the incubation medium as well as from serum co-elute on HPAEC-PAD. Several masked oligosaccharides in serum contain m-inositol, e-inositol, and sorbitol as the major carbohydrates. The presence of this glycosylated protein in serum is a fraction of glycogen that is degraded outside the lysosome and the cell. The glycosylated protein in the serum is not present in the serum of Pompe mice not on ERT, but it is present in the serum of Pompe disease patients who are on ERT, so it is a biomarker of GAA degradation of lysosomal glycogen.
Collapse
Affiliation(s)
- Allen K. Murray
- HIBM Research Group, Inc., Chatsworth, CA 21053, USA; or ; Tel.: +1-949-689-9664
- Glycan Technologies, Inc., P.O. Box 17993, Irvine, CA 92623, USA
| |
Collapse
|
14
|
Kishnani PS, Sun B, Koeberl DD. Gene therapy for glycogen storage diseases. Hum Mol Genet 2019; 28:R31-R41. [PMID: 31227835 PMCID: PMC6796997 DOI: 10.1093/hmg/ddz133] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/02/2019] [Accepted: 06/07/2019] [Indexed: 12/17/2022] Open
Abstract
The focus of this review is the development of gene therapy for glycogen storage diseases (GSDs). GSD results from the deficiency of specific enzymes involved in the storage and retrieval of glucose in the body. Broadly, GSDs can be divided into types that affect liver or muscle or both tissues. For example, glucose-6-phosphatase (G6Pase) deficiency in GSD type Ia (GSD Ia) affects primarily the liver and kidney, while acid α-glucosidase (GAA) deficiency in GSD II causes primarily muscle disease. The lack of specific therapy for the GSDs has driven efforts to develop new therapies for these conditions. Gene therapy needs to replace deficient enzymes in target tissues, which has guided the planning of gene therapy experiments. Gene therapy with adeno-associated virus (AAV) vectors has demonstrated appropriate tropism for target tissues, including the liver, heart and skeletal muscle in animal models for GSD. AAV vectors transduced liver and kidney in GSD Ia and striated muscle in GSD II mice to replace the deficient enzyme in each disease. Gene therapy has been advanced to early phase clinical trials for the replacement of G6Pase in GSD Ia and GAA in GSD II (Pompe disease). Other GSDs have been treated in proof-of-concept studies, including GSD III, IV and V. The future of gene therapy appears promising for the GSDs, promising to provide more efficacious therapy for these disorders in the foreseeable future.
Collapse
Affiliation(s)
- Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Baodong Sun
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
15
|
Ginocchio VM, Ferla R, Auricchio A, Brunetti-Pierri N. Current Status on Clinical Development of Adeno-Associated Virus-Mediated Liver-Directed Gene Therapy for Inborn Errors of Metabolism. Hum Gene Ther 2019; 30:1204-1210. [PMID: 31517544 DOI: 10.1089/hum.2019.151] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Inborn errors of metabolism (IEM) are disorders affecting human biochemical pathways and represent attractive targets for gene therapy because of their severity, high overall prevalence, lack of effective treatments, and possibility of early diagnosis through newborn screening. The liver is a central organ involved in several metabolic reactions and is a favorite target for gene therapy in many IEM. Adeno-associated virus (AAV) vectors have emerged in the last years as the preferred vectors for in vivo gene delivery. Gene replacement strategies are aimed either at correcting liver disease or providing a source for production and secretion of the lacking enzyme for cross-correction of other tissues. A number of preclinical studies have been conducted in the last years and, for several diseases, gene therapy has reached the clinical stage, with a growing number of ongoing clinical trials. Moreover, recent applications of genome editing to the field of inherited metabolic diseases have further expanded potential therapeutic possibilities. This review describes relevant clinical gene therapy studies for IEM with particular attention to current obstacles and drawbacks.
Collapse
Affiliation(s)
- Virginia Maria Ginocchio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (Naples), Italy.,Department of Translational Medicine, "Federico II" University, Naples, Italy
| | - Rita Ferla
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (Naples), Italy.,Department of Translational Medicine, "Federico II" University, Naples, Italy
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (Naples), Italy.,Department of Advanced Biomedicine, "Federico II" University, Naples, Italy
| | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (Naples), Italy.,Department of Translational Medicine, "Federico II" University, Naples, Italy
| |
Collapse
|
16
|
Abstract
Pompe disease (PD) is caused by the deficiency of the lysosomal enzyme acid α-glucosidase (GAA), resulting in systemic pathological glycogen accumulation. PD can present with cardiac, skeletal muscle, and central nervous system manifestations, as a continuum of phenotypes among two main forms: classical infantile-onset PD (IOPD) and late-onset PD (LOPD). IOPD is caused by severe GAA deficiency and presents at birth with cardiac hypertrophy, muscle hypotonia, and severe respiratory impairment, leading to premature death, if not treated. LOPD is characterized by levels of residual GAA activity up to ∼20% of normal and presents both in children and adults with a varied severity of muscle weakness and motor and respiratory deficit. Enzyme replacement therapy (ERT), based on repeated intravenous (i.v.) infusions of recombinant human GAA (rhGAA), represents the only available treatment for PD. Upon more than 10 years from its launch, it is becoming evident that ERT can extend the life span of IOPD and stabilize disease progression in LOPD; however, it does not represent a cure for PD. The limited uptake of the enzyme in key affected tissues and the high immunogenicity of rhGAA are some of the hurdles that limit ERT efficacy. GAA gene transfer with adeno-associated virus (AAV) vectors has been shown to reduce glycogen storage and improve the PD phenotype in preclinical studies following different approaches. Here, we present an overview of the different gene therapy approaches for PD, focusing on in vivo gene transfer with AAV vectors and discussing the potential opportunities and challenges in developing safe and effective gene therapies for the disease. Based on emerging safety and efficacy data from clinical trials for other protein deficiencies, in vivo gene therapy with AAV vectors appears to have the potential to provide a therapeutically relevant, stable source of GAA enzyme, which could be highly beneficial in PD.
Collapse
Affiliation(s)
- Pasqualina Colella
- Genethon, Evry, France.,Department of Pediatrics, Stanford University, Stanford, California
| | - Federico Mingozzi
- Genethon, Evry, France.,Spark Therapeutics, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Kishnani PS, Koeberl DD. Liver depot gene therapy for Pompe disease. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:288. [PMID: 31392200 PMCID: PMC6642935 DOI: 10.21037/atm.2019.05.02] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 04/26/2019] [Indexed: 12/12/2022]
Abstract
Gene therapy for Pompe disease has advanced to early phase clinical trials, based upon proof-of-concept data indicating that gene therapy could surpass the benefits of the current standard of care, enzyme replacement therapy (ERT). ERT requires frequent infusions of large quantities of recombinant human acid α-glucosidase (GAA), whereas gene therapy involves a single infusion of a vector that stably transduces tissues to continuously produce GAA. Liver-specific expression of GAA with an adeno-associated virus (AAV) vector established stable GAA secretion from the liver accompanied by receptor-mediated uptake of GAA, which corrected the deficiency of GAA and cleared the majority of accumulated glycogen in the heart and skeletal muscle. Liver depot gene therapy was equivalent to ERT at a dose of the AAV vector that could be administered in an early phase clinical trial. Furthermore, high-level expression of GAA has decreased glycogen stored in the brain. A unique advantage of liver-specific expression stems from the induction of immune tolerance to GAA following AAV vector administration, thereby suppressing anti-GAA antibodies that otherwise interfere with efficacy. A Phase I clinical trial of AAV vector-mediated liver depot gene therapy has been initiated based upon promising preclinical data (NCT03533673). Overall, gene therapy promises to address limits of currently available ERT, if clinical translation currently underway is successful.
Collapse
Affiliation(s)
- Priya S. Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Dwight D. Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| |
Collapse
|
18
|
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
|
19
|
Do HV, Khanna R, Gotschall R. Challenges in treating Pompe disease: an industry perspective. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:291. [PMID: 31392203 DOI: 10.21037/atm.2019.04.15] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pompe disease is a rare inherited metabolic disorder of defective lysosomal glycogen catabolism due to a deficiency in acid alpha-glucosidase (GAA). Alglucosidase alfa enzyme replacement therapy (ERT) using recombinant human GAA (rhGAA ERT) is the only approved treatment for Pompe disease. Alglucosidase alfa has provided irrefutable clinical benefits, but has not been an optimal treatment primarily due to poor drug targeting of ERT to skeletal muscles. Several critical factors contribute to this inefficiency. Some are inherent to the anatomy of the body that cannot be altered, while others may be addressed with better drug design and engineering. The knowledge gained from alglucosidase alfa ERT over the past 2 decades has allowed us to better understand the challenges that hinder its effectiveness. In this review, we detail the problems which must be overcome for improving drug targeting and clinical efficacy. These same issues may also impact therapeutic enzymes derived from gene therapies, and thus, have important implications for the development of next generation therapies for Pompe.
Collapse
Affiliation(s)
- Hung V Do
- Amicus Therapeutics, Inc., Cranbury, NJ, USA
| | | | | |
Collapse
|
20
|
Desai AK, Kazi ZB, Bali DS, Kishnani PS. Characterization of immune response in Cross-Reactive Immunological Material (CRIM)-positive infantile Pompe disease patients treated with enzyme replacement therapy. Mol Genet Metab Rep 2019; 20:100475. [PMID: 31193175 PMCID: PMC6518314 DOI: 10.1016/j.ymgmr.2019.100475] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 10/27/2022] Open
Abstract
Enzyme replacement therapy (ERT) with rhGAA has improved clinical outcomes in infantile Pompe disease (IPD). A subset of CRIM-positive IPD patients develop high and sustained antibody titers (HSAT; ≥51,200) and/or sustained intermediate titer (SIT; ≥12,800 and <51,200), similar to CRIM-negative patients. To date there has been no systematic study to analyze the extent of IgG antibody response in CRIM-positive IPD. Such data would be critical and could serve as a comparator group for potential immune modulation approaches. A retrospective analysis of the dataset from the original rhGAA clinical trials final reports was conducted. CRIM-positive patients who received ERT monotherapy and had >6 months of antibody titer data available, were included in the study. Patients were classified based on their longitudinal antibody titers into HSAT, SIT, and low titer (LT; <12,800) groups. Of the 37 patients that met inclusion criteria, five (13%), seven (19%), and 25 (68%) developed HSAT, SIT, and LT, respectively. Median peak titers were 204,800 (51,200-409,600), 25,600 (12,800-51,200), and 800 (200-12,800) for HSAT, SIT, and LT groups, respectively. Median last titers were 102,400 (51,200-409,600), 1600 (200-25,600), and 400 (0-12,800) at median time since ERT initiation of 94 weeks (64-155 weeks), 104 weeks (86-144 weeks), and 130 weeks (38-182 weeks) for HSAT, SIT, and LT groups, respectively. 32% (12/37) of CRIM-positive IPD patients developed HSAT/SIT which may lead to limited ERT response and clinical decline. Further Studies are needed to identify CRIM-positive IPD patients at risk of developing HSAT/SIT, especially with the addition of Pompe disease to the newborn screening.
Collapse
Key Words
- AIMS, Alberta infant motor scale
- Anti-rhGAA Ig antibodies
- Antidrug antibodies
- CI-MPR, Cation-independent mannose 6-phosphate receptor
- CRIM, Cross-reactive immunological material
- EOW, Every other week
- ERT, Enzyme replacement therapy
- Enzyme replacement therapy
- GAA, Acid α-glucosidase
- GAA, Gene encoding acid α-glucosidase
- Glc4, Glucose tetrasaccharide
- Glycogen storage disease type II
- HLA, Human leukocyte antigen
- HSAT, High and sustained antibody titers
- IPD, Infantile Pompe disease
- IgG, Immunoglobulin G
- LT, Low titers
- LVMI, Left ventricular mass index
- MHC, Major histocompatibility complex
- Neuromuscular disease
- Pompe disease
- RUSP, Recommended universal screening panel
- SIT, Sustained intermediate titers
- iTEM, Individualized T-cell epitope measure
- rhGAA, Recombinant human acid α-glucosidase
Collapse
Affiliation(s)
- Ankit K Desai
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Zoheb B Kazi
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Deeksha S Bali
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
21
|
Nagree MS, Scalia S, McKillop WM, Medin JA. An update on gene therapy for lysosomal storage disorders. Expert Opin Biol Ther 2019; 19:655-670. [DOI: 10.1080/14712598.2019.1607837] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Murtaza S. Nagree
- Department of Medical Biophysics, University of Toronto, Toronto,
Ontario, Canada
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee,
WI, USA
| | - Simone Scalia
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee,
WI, USA
| | | | - Jeffrey A. Medin
- Department of Medical Biophysics, University of Toronto, Toronto,
Ontario, Canada
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee,
WI, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee,
WI, USA
| |
Collapse
|
22
|
Han SO, Li S, Everitt JI, Koeberl DD. Salmeterol with Liver Depot Gene Therapy Enhances the Skeletal Muscle Response in Murine Pompe Disease. Hum Gene Ther 2019; 30:855-864. [PMID: 30803275 DOI: 10.1089/hum.2018.197] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gene therapy for Pompe disease with adeno-associated virus (AAV) vectors has advanced into early phase clinical trials; however, the paucity of cation-independent mannose-6-phosphate receptor (CI-MPR) in skeletal muscle, where it is needed to take up acid α-glucosidase (GAA), has impeded the efficacy of Pompe disease gene therapy. Long-acting selective β2 receptor agonists previously enhanced the CI-MPR expression in muscle. In this study we have evaluated the selective β2 agonist salmeterol in GAA knockout mice in combination with an AAV vector expressing human GAA specifically in the liver. Quadriceps glycogen content was significantly decreased by administration of the AAV vector with salmeterol, in comparison with the AAV vector alone (p < 0.01). Importantly, glycogen content of the quadriceps was reduced to its lowest level by the combination of AAV vector and salmeterol administration. Rotarod testing revealed significant improvement following treatment, in comparison with untreated mice, and salmeterol improved wirehang performance. Salmeterol treatment decreased abnormalities of autophagy in the quadriceps, as shown be lower LC3 and p62. Vector administration reduced the abnormal vacuolization and accumulation of nuclei in skeletal muscle. Thus, salmeterol could be further developed as adjunctive therapy to improve the efficacy of liver depot gene therapy for Pompe disease.
Collapse
Affiliation(s)
- Sang-Oh Han
- 1Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina
| | - Songtao Li
- 1Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina
| | - Jeffrey I Everitt
- 2Department of Pathology, Duke University Medical School, Durham, North Carolina
| | - Dwight D Koeberl
- 1Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina.,3Department of Molecular Genetics and Metabolism, Duke University Medical School, Durham, North Carolina
| |
Collapse
|
23
|
Abstract
Metabolic myopathies are a heterogeneous group of disorders characterized by inherited defects of enzymatic pathways involved in muscle cellular energetics and adenosine triphosphate synthesis. Skeletal and respiratory muscles are most affected. There are multiple mechanisms of disease. The age of onset and prognosis vary. Metabolic myopathies cause exercise intolerance, myalgia, and increase in muscle breakdown products during exercise. Some affect smooth muscle like the diaphragm and cause respiratory failure. The pathophysiology is complex and the evidence in literature to guide diagnosis and management is sparse. Treatment is limited. This review discusses the pathophysiology and diagnostic evaluation of these disorders.
Collapse
Affiliation(s)
- Patrick Koo
- Department of Respiratory, Critical Care, and Sleep Medicine, University of Tennessee College of Medicine Chattanooga, Erlanger Health System, 975 East 3rd Street, C-735, Chattanooga, TN 37403, USA.
| | - Jigme M Sethi
- Department of Respiratory, Critical Care, and Sleep Medicine, University of Tennessee College of Medicine Chattanooga, Erlanger Health System, 975 East 3rd Street, C-735, Chattanooga, TN 37403, USA
| |
Collapse
|
24
|
Xu S, Lun Y, Frascella M, Garcia A, Soska R, Nair A, Ponery AS, Schilling A, Feng J, Tuske S, Valle MCD, Martina JA, Ralston E, Gotschall R, Valenzano KJ, Puertollano R, Do HV, Raben N, Khanna R. Improved efficacy of a next-generation ERT in murine Pompe disease. JCI Insight 2019; 4:125358. [PMID: 30843882 DOI: 10.1172/jci.insight.125358] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/17/2019] [Indexed: 01/14/2023] Open
Abstract
Pompe disease is a rare inherited disorder of lysosomal glycogen metabolism due to acid α-glucosidase (GAA) deficiency. Enzyme replacement therapy (ERT) using alglucosidase alfa, a recombinant human GAA (rhGAA), is the only approved treatment for Pompe disease. Although alglucosidase alfa has provided clinical benefits, its poor targeting to key disease-relevant skeletal muscles results in suboptimal efficacy. We are developing an rhGAA, ATB200 (Amicus proprietary rhGAA), with high levels of mannose-6-phosphate that are required for efficient cellular uptake and lysosomal trafficking. When administered in combination with the pharmacological chaperone AT2221 (miglustat), which stabilizes the enzyme and improves its pharmacokinetic properties, ATB200/AT2221 was substantially more potent than alglucosidase alfa in a mouse model of Pompe disease. The new investigational therapy is more effective at reversing the primary abnormality - intralysosomal glycogen accumulation - in multiple muscles. Furthermore, unlike the current standard of care, ATB200/AT2221 dramatically reduces autophagic buildup, a major secondary defect in the diseased muscles. The reversal of lysosomal and autophagic pathologies leads to improved muscle function. These data demonstrate the superiority of ATB200/AT2221 over the currently approved ERT in the murine model.
Collapse
Affiliation(s)
- Su Xu
- Amicus Therapeutics, Cranbury, New Jersey, USA
| | - Yi Lun
- Amicus Therapeutics, Cranbury, New Jersey, USA
| | | | | | | | - Anju Nair
- Amicus Therapeutics, Cranbury, New Jersey, USA
| | | | | | - Jessie Feng
- Amicus Therapeutics, Cranbury, New Jersey, USA
| | | | | | - José A Martina
- Laboratory of Protein Trafficking and Organelle Biology, Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Evelyn Ralston
- Light Imaging Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | | | | | - Rosa Puertollano
- Laboratory of Protein Trafficking and Organelle Biology, Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Hung V Do
- Amicus Therapeutics, Cranbury, New Jersey, USA
| | - Nina Raben
- Laboratory of Protein Trafficking and Organelle Biology, Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | | |
Collapse
|
25
|
Lagalice L, Pichon J, Gougeon E, Soussi S, Deniaud J, Ledevin M, Maurier V, Leroux I, Durand S, Ciron C, Franzoso F, Dubreil L, Larcher T, Rouger K, Colle MA. Satellite cells fail to contribute to muscle repair but are functional in Pompe disease (glycogenosis type II). Acta Neuropathol Commun 2018; 6:116. [PMID: 30382921 PMCID: PMC6211565 DOI: 10.1186/s40478-018-0609-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/29/2018] [Indexed: 12/26/2022] Open
Abstract
Pompe disease, which is due to acid alpha-glucosidase deficiency, is characterized by skeletal muscle dysfunction attributed to the accumulation of glycogen-filled lysosomes and autophagic buildup. Despite the extensive tissue damages, a failure of satellite cell (SC) activation and lack of muscle regeneration have been reported in patients. However, the origin of this defective program is unknown. Additionally, whether these deficits occur gradually over the disease course is unclear. Using a longitudinal pathophysiological study of two muscles in a Pompe mouse model, here, we report that the enzymatic defect results in a premature saturating glycogen overload and a high number of enlarged lysosomes. The muscles gradually display profound remodeling as the number of autophagic vesicles, centronucleated fibers, and split fibers increases and larger fibers are lost. Only a few regenerated fibers were observed regardless of age, although the SC pool was preserved. Except for the early age, during which higher numbers of activated SCs and myoblasts were observed, no myogenic commitment was observed in response to the damage. Following in vivo injury, we established that muscle retains regenerative potential, demonstrating that the failure of SC participation in repair is related to an activation signal defect. Altogether, our findings provide new insight into the pathophysiology of Pompe disease and highlight that the activation signal defect of SCs compromises muscle repair, which could be related to the abnormal energetic supply following autophagic flux impairment.
Collapse
Affiliation(s)
- Lydie Lagalice
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Julien Pichon
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
- INSERM UMR1089, Université de Nantes, Centre Hospitalier Universitaire, Nantes, France
| | - Eliot Gougeon
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Salwa Soussi
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Johan Deniaud
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Mireille Ledevin
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Virginie Maurier
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Isabelle Leroux
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Sylvie Durand
- BIA, INRA, Centre INRA Pays de la Loire, Nantes, F-44300 France
| | - Carine Ciron
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Francesca Franzoso
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Laurence Dubreil
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Thibaut Larcher
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Karl Rouger
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Marie-Anne Colle
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| |
Collapse
|
26
|
Koeberl DD, Case LE, Smith EC, Walters C, Han SO, Li Y, Chen W, Hornik CP, Huffman KM, Kraus WE, Thurberg BL, Corcoran DL, Bali D, Bursac N, Kishnani PS. Correction of Biochemical Abnormalities and Improved Muscle Function in a Phase I/II Clinical Trial of Clenbuterol in Pompe Disease. Mol Ther 2018; 26:2304-2314. [PMID: 30025991 PMCID: PMC6127508 DOI: 10.1016/j.ymthe.2018.06.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/22/2018] [Accepted: 06/25/2018] [Indexed: 01/10/2023] Open
Abstract
This 52-week, phase I/II double-blind, randomized, placebo-controlled study investigated the novel use of clenbuterol in late-onset Pompe disease (LOPD) stably treated with ERT. Eleven of thirteen participants completed the study. No serious adverse events were related to clenbuterol, and transient minor adverse events included mild elevations of creatine kinase, muscle spasms, and tremors. At week 52, the 6-min walk test distance increased by a mean of 16 m (p = 0.08), or a mean of 3% of predicted performance (p = 0.03), and the maximum inspiratory pressure increased 8% (p = 0.003) for the clenbuterol group. The quick motor function test score improved by a mean of seven points (p = 0.007); and the gait, stairs, gower, chair test improved by a mean of two points (p = 0.004). Clenbuterol decreased glycogen content in the vastus lateralis by 50% at week 52. Transcriptome analysis revealed more normal muscle gene expression for 38 of 44 genes related to Pompe disease following clenbuterol. The placebo group demonstrated no significant changes over the course of the study. This study provides initial evidence for safety and efficacy of adjunctive clenbuterol in patients with LOPD (NCT01942590).
Collapse
Affiliation(s)
- Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Laura E Case
- Department of Physical and Occupational Therapy, Duke University School of Medicine, Durham, NC 27710, USA
| | - Edward C Smith
- Division of Neurology, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Crista Walters
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sang-Oh Han
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yanzhen Li
- Department of Biomedical Engineering, Duke University School of Medicine, Durham, NC 27710, USA
| | - Wei Chen
- Duke Center for Genomic and Computational Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Christoph P Hornik
- Division of Critical Care Medicine, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kim M Huffman
- Division of Rheumatology and Immunology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - William E Kraus
- Division of Cardiology, Department of Medicine; Duke University School of Medicine, Durham, NC 27710, USA
| | | | - David L Corcoran
- Duke Center for Genomic and Computational Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Deeksha Bali
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University School of Medicine, Durham, NC 27710, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
27
|
Khodabukus A, Prabhu N, Wang J, Bursac N. In Vitro Tissue-Engineered Skeletal Muscle Models for Studying Muscle Physiology and Disease. Adv Healthc Mater 2018; 7:e1701498. [PMID: 29696831 PMCID: PMC6105407 DOI: 10.1002/adhm.201701498] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 02/18/2018] [Indexed: 12/18/2022]
Abstract
Healthy skeletal muscle possesses the extraordinary ability to regenerate in response to small-scale injuries; however, this self-repair capacity becomes overwhelmed with aging, genetic myopathies, and large muscle loss. The failure of small animal models to accurately replicate human muscle disease, injury and to predict clinically-relevant drug responses has driven the development of high fidelity in vitro skeletal muscle models. Herein, the progress made and challenges ahead in engineering biomimetic human skeletal muscle tissues that can recapitulate muscle development, genetic diseases, regeneration, and drug response is discussed. Bioengineering approaches used to improve engineered muscle structure and function as well as the functionality of satellite cells to allow modeling muscle regeneration in vitro are also highlighted. Next, a historical overview on the generation of skeletal muscle cells and tissues from human pluripotent stem cells, and a discussion on the potential of these approaches to model and treat genetic diseases such as Duchenne muscular dystrophy, is provided. Finally, the need to integrate multiorgan microphysiological systems to generate improved drug discovery technologies with the potential to complement or supersede current preclinical animal models of muscle disease is described.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Neel Prabhu
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Jason Wang
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Nenad Bursac
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| |
Collapse
|
28
|
Puzzo F, Colella P, Biferi MG, Bali D, Paulk NK, Vidal P, Collaud F, Simon-Sola M, Charles S, Hardet R, Leborgne C, Meliani A, Cohen-Tannoudji M, Astord S, Gjata B, Sellier P, van Wittenberghe L, Vignaud A, Boisgerault F, Barkats M, Laforet P, Kay MA, Koeberl DD, Ronzitti G, Mingozzi F. Rescue of Pompe disease in mice by AAV-mediated liver delivery of secretable acid α-glucosidase. Sci Transl Med 2018; 9:9/418/eaam6375. [PMID: 29187643 DOI: 10.1126/scitranslmed.aam6375] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 06/13/2017] [Indexed: 12/26/2022]
Abstract
Glycogen storage disease type II or Pompe disease is a severe neuromuscular disorder caused by mutations in the lysosomal enzyme, acid α-glucosidase (GAA), which result in pathological accumulation of glycogen throughout the body. Enzyme replacement therapy is available for Pompe disease; however, it has limited efficacy, has high immunogenicity, and fails to correct pathological glycogen accumulation in nervous tissue and skeletal muscle. Using bioinformatics analysis and protein engineering, we developed transgenes encoding GAA that could be expressed and secreted by hepatocytes. Then, we used adeno-associated virus (AAV) vectors optimized for hepatic expression to deliver the GAA transgenes to Gaa knockout (Gaa-/-) mice, a model of Pompe disease. Therapeutic gene transfer to the liver rescued glycogen accumulation in muscle and the central nervous system, and ameliorated cardiac hypertrophy as well as muscle and respiratory dysfunction in the Gaa-/- mice; mouse survival was also increased. Secretable GAA showed improved therapeutic efficacy and lower immunogenicity compared to nonengineered GAA. Scale-up to nonhuman primates, and modeling of GAA expression in primary human hepatocytes using hepatotropic AAV vectors, demonstrated the therapeutic potential of AAV vector-mediated liver expression of secretable GAA for treating pathological glycogen accumulation in multiple tissues in Pompe disease.
Collapse
Affiliation(s)
- Francesco Puzzo
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France.,Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Pasqualina Colella
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| | - Maria G Biferi
- University Pierre and Marie Curie Paris 6 and INSERM U974, Paris, France
| | - Deeksha Bali
- Biochemical Genetics Laboratory, Duke University Health System, Durham, NC 27710, USA
| | - Nicole K Paulk
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Patrice Vidal
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France.,University Pierre and Marie Curie Paris 6 and INSERM U974, Paris, France
| | - Fanny Collaud
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| | - Marcelo Simon-Sola
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France.,University Pierre and Marie Curie Paris 6 and INSERM U974, Paris, France
| | - Severine Charles
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| | - Romain Hardet
- University Pierre and Marie Curie Paris 6 and INSERM U974, Paris, France
| | - Christian Leborgne
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| | - Amine Meliani
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France.,University Pierre and Marie Curie Paris 6 and INSERM U974, Paris, France
| | | | - Stephanie Astord
- University Pierre and Marie Curie Paris 6 and INSERM U974, Paris, France
| | - Bernard Gjata
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| | - Pauline Sellier
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France.,University Pierre and Marie Curie Paris 6 and INSERM U974, Paris, France
| | | | - Alban Vignaud
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| | - Florence Boisgerault
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| | - Martine Barkats
- University Pierre and Marie Curie Paris 6 and INSERM U974, Paris, France
| | - Pascal Laforet
- Paris-Est Neuromuscular Center, Pitié-Salpêtrière Hospital and Raymond Poincaré Teaching Hospital, Garches, APHP, Paris, France
| | - Mark A Kay
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics and Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Giuseppe Ronzitti
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France.
| | - Federico Mingozzi
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, 91002 Evry, France. .,University Pierre and Marie Curie Paris 6 and INSERM U974, Paris, France
| |
Collapse
|
29
|
Efficient therapy for refractory Pompe disease by mannose 6-phosphate analogue grafting on acid α-glucosidase. J Control Release 2018; 269:15-23. [DOI: 10.1016/j.jconrel.2017.10.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 01/30/2023]
|
30
|
Bond JE, Kishnani PS, Koeberl DD. Immunomodulatory, liver depot gene therapy for Pompe disease. Cell Immunol 2017; 342:103737. [PMID: 29295737 DOI: 10.1016/j.cellimm.2017.12.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/15/2017] [Accepted: 12/27/2017] [Indexed: 01/09/2023]
Abstract
Pompe disease is caused by mutations in acid alpha glucosidase (GAA) that causes accumulation of lysosomal glycogen affecting the heart and skeletal muscles, and can be fatal. Enzyme replacement therapy (ERT) with recombinant human GAA (rhGAA) improves muscle function by reducing glycogen accumulation. Limitations of ERT include a short half-life and the formation of antibodies that result in reduced efficacy. By harnessing the immune tolerance induction properties of the liver, liver-targeted gene delivery (with an adeno-associated virus vector containing a liver specific promoter), suppresses immunity against the GAA introduced by gene therapy. This induces immune tolerance to rhGAA by activating regulatory T cells and simultaneously, corrects GAA deficiency. Potentially, liver-targeted gene therapy can be performed once with lasting effects, by administering a relatively low dose of an adeno-associated virus type 8 vector to replace and induce immune tolerance to GAA.
Collapse
Affiliation(s)
- J E Bond
- Clinical and Translational Science Institute, Duke University, Durham, NC, USA
| | - P S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, NC, USA
| | - D D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, NC, USA; Molecular Genetics and Microbiology, Duke University, Durham, NC, USA.
| |
Collapse
|
31
|
Ripolone M, Violano R, Ronchi D, Mondello S, Nascimbeni A, Colombo I, Fagiolari G, Bordoni A, Fortunato F, Lucchini V, Saredi S, Filosto M, Musumeci O, Tonin P, Mongini T, Previtali S, Morandi L, Angelini C, Mora M, Sandri M, Sciacco M, Toscano A, Comi GP, Moggio M. Effects of short-to-long term enzyme replacement therapy (ERT) on skeletal muscle tissue in late onset Pompe disease (LOPD). Neuropathol Appl Neurobiol 2017; 44:449-462. [PMID: 28574618 DOI: 10.1111/nan.12414] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/26/2017] [Accepted: 06/02/2017] [Indexed: 12/31/2022]
Abstract
AIMS Pompe disease is an autosomal recessive lysosomal storage disorder resulting from deficiency of acid α-glucosidase (GAA) enzyme. Histopathological hallmarks in skeletal muscle tissue are fibre vacuolization and autophagy. Since 2006, enzyme replacement therapy (ERT) is the only approved treatment with human recombinant GAA alglucosidase alfa. We designed a study to examine ERT-related skeletal muscle changes in 18 modestly to moderately affected late onset Pompe disease (LOPD) patients along with the relationship between morphological/biochemical changes and clinical outcomes. Treatment duration was short-to-long term. METHODS We examined muscle biopsies from 18 LOPD patients at both histopathological and biochemical level. All patients underwent two muscle biopsies, before and after ERT administration respectively. The study is partially retrospective because the first biopsies were taken before the study was designed, whereas the second biopsy was always performed after at least 6 months of ERT administration. RESULTS After ERT, 15 out of 18 patients showed improved 6-min walking test (6MWT; P = 0.0007) and most of them achieved respiratory stabilization. Pretreatment muscle biopsies disclosed marked histopathological variability, ranging from an almost normal pattern to a severe vacuolar myopathy. After treatment, we detected morphological improvement in 15 patients and worsening in three patients. Post-ERT GAA enzymatic activity was mildly increased compared with pretreatment levels in all patients. Protein levels of the mature enzyme increased in 14 of the 18 patients (mean increase = +35%; P < 0.05). Additional studies demonstrated an improved autophagic flux after ERT in some patients. CONCLUSIONS ERT positively modified skeletal muscle pathology as well as motor and respiratory outcomes in the majority of LOPD patients.
Collapse
Affiliation(s)
- M Ripolone
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - R Violano
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - D Ronchi
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - S Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - A Nascimbeni
- Department of Neurosciences, University of Padova, Padova, Italy
| | - I Colombo
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - G Fagiolari
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - A Bordoni
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - F Fortunato
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - V Lucchini
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - S Saredi
- Neuromuscular Diseases and Neuroimmunology, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - M Filosto
- Unit of Neurology, Center for Neuromuscular Diseases and Neuropathies, University Hospital "Spedali Civili", Brescia, Italy
| | - O Musumeci
- Department of Clinical and Experimental Medicine, Centro di Riferimento Regionale per le Malattie Neuromuscolari rare, University of Messina, Messina, Italy
| | - P Tonin
- Section of Clinical Neurology, Department of Neurological, Biomedical and Movement Sciences, University of Verona, Verona, Italy
| | - T Mongini
- Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - S Previtali
- Division of Neuroscience, Inspe, San Raffaele, Milan, Italy
| | - L Morandi
- Neuromuscular Diseases and Neuroimmunology, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - C Angelini
- Fondazione San Camillo Hospital IRCCS, Venice, Italy
| | - M Mora
- Neuromuscular Diseases and Neuroimmunology, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - M Sandri
- Department of Biomedical Science, University of Padova, Padova, Italy.,Dulbecco Telethon Institute at Venetian Institute of Molecular Medicine, Padova, Italy
| | - M Sciacco
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - A Toscano
- Department of Clinical and Experimental Medicine, Centro di Riferimento Regionale per le Malattie Neuromuscolari rare, University of Messina, Messina, Italy
| | - G P Comi
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - M Moggio
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
32
|
Han SO, Ronzitti G, Arnson B, Leborgne C, Li S, Mingozzi F, Koeberl D. Low-Dose Liver-Targeted Gene Therapy for Pompe Disease Enhances Therapeutic Efficacy of ERT via Immune Tolerance Induction. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 4:126-136. [PMID: 28344998 PMCID: PMC5363303 DOI: 10.1016/j.omtm.2016.12.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/30/2016] [Indexed: 11/18/2022]
Abstract
Pompe disease results from acid α-glucosidase (GAA) deficiency, and enzyme replacement therapy (ERT) with recombinant human (rh) GAA has clinical benefits, although its limitations include the short half-life of GAA and the formation of antibody responses. The present study compared the efficacy of ERT against gene transfer with an adeno-associated viral (AAV) vector containing a liver-specific promoter. GAA knockout (KO) mice were administered either a weekly injection of rhGAA (20 mg/kg) or a single injection of AAV2/8-LSPhGAA (8 × 1011 vector genomes [vg]/kg). Both treatments significantly reduced glycogen content of the heart and diaphragm. Although ERT triggered anti-GAA antibody formation, there was no detectable antibody response following AAV vector administration. The efficacy of three lower dosages of AAV2/8-LSPhGAA was evaluated in GAA-KO mice, either alone or in combination with ERT. The minimum effective dose (MED) identified was 8 × 1010 vg/kg to reduce glycogen content in the heart and diaphragm of GAA-KO mice. A 3-fold higher dose was required to suppress antibody responses to ERT. Efficacy from liver gene therapy was slightly greater in male mice than in female mice. Vector dose correlated inversely with anti-GAA antibody formation, whereas higher vector doses suppressed previously formed anti-GAA antibodies as late as 25 weeks after the start of ERT and achieved biochemical correction of glycogen accumulation. In conclusion, we identified the MED for effective AAV2/8-LSPhGAA-mediated tolerogenic gene therapy in Pompe disease mice.
Collapse
Affiliation(s)
- Sang-oh Han
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - Benjamin Arnson
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - Songtao Li
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Federico Mingozzi
- Genethon and INSERM U951, 91002 Evry, France
- University Pierre and Marie Curie – Paris 6, 75005 Paris, France
| | - Dwight Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
- Corresponding author: Dwight Koeberl, Duke University Medical Center, Box 103856, Durham, NC 27710, USA.
| |
Collapse
|
33
|
Alglucosidase alfa treatment alleviates liver disease in a mouse model of glycogen storage disease type IV. Mol Genet Metab Rep 2016; 9:31-33. [PMID: 27747161 PMCID: PMC5053031 DOI: 10.1016/j.ymgmr.2016.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 09/20/2016] [Accepted: 09/20/2016] [Indexed: 01/21/2023] Open
Abstract
Patients with progressive hepatic form of GSD IV often die of liver failure in early childhood. We tested the feasibility of using recombinant human acid-α glucosidase (rhGAA) for treating GSD IV. Weekly intravenously injection of rhGAA at 40 mg/kg for 4 weeks significantly reduced hepatic glycogen accumulation, lowered liver/body weight ratio, and reduced plasma ALP and ALT activities in GSD IV mice. Our data suggests that rhGAA is a potential therapy for GSD IV. An FDA approved therapy is proposed as a new therapeutic approach for GSD IV. A short-term rhGAA treatment significantly reduced liver glycogen content in GSD IV mice. rhGAA treatment alleviated liver disease progression in GSD IV mice. Our data suggests that rhGAA is a potential therapy for hepatic form of GSD IV.
Collapse
|
34
|
Bursac N, Juhas M, Rando TA. Synergizing Engineering and Biology to Treat and Model Skeletal Muscle Injury and Disease. Annu Rev Biomed Eng 2016; 17:217-42. [PMID: 26643021 DOI: 10.1146/annurev-bioeng-071114-040640] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although skeletal muscle is one of the most regenerative organs in our body, various genetic defects, alterations in extrinsic signaling, or substantial tissue damage can impair muscle function and the capacity for self-repair. The diversity and complexity of muscle disorders have attracted much interest from both cell biologists and, more recently, bioengineers, leading to concentrated efforts to better understand muscle pathology and develop more efficient therapies. This review describes the biological underpinnings of muscle development, repair, and disease, and discusses recent bioengineering efforts to design and control myomimetic environments, both to study muscle biology and function and to aid in the development of new drug, cell, and gene therapies for muscle disorders. The synergy between engineering-aided biological discovery and biology-inspired engineering solutions will be the path forward for translating laboratory results into clinical practice.
Collapse
Affiliation(s)
- Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708;
| | - Mark Juhas
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708;
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305.,Rehabilitation Research & Development Service, VA Palo Alto Health Care System, Palo Alto, California 94304
| |
Collapse
|
35
|
Talbot J, Maves L. Skeletal muscle fiber type: using insights from muscle developmental biology to dissect targets for susceptibility and resistance to muscle disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2016; 5:518-34. [PMID: 27199166 DOI: 10.1002/wdev.230] [Citation(s) in RCA: 247] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 01/14/2016] [Accepted: 01/16/2016] [Indexed: 12/13/2022]
Abstract
Skeletal muscle fibers are classified into fiber types, in particular, slow twitch versus fast twitch. Muscle fiber types are generally defined by the particular myosin heavy chain isoforms that they express, but many other components contribute to a fiber's physiological characteristics. Skeletal muscle fiber type can have a profound impact on muscle diseases, including certain muscular dystrophies and sarcopenia, the aging-induced loss of muscle mass and strength. These findings suggest that some muscle diseases may be treated by shifting fiber type characteristics either from slow to fast, or fast to slow phenotypes, depending on the disease. Recent studies have begun to address which components of muscle fiber types mediate their susceptibility or resistance to muscle disease. However, for many diseases it remains largely unclear why certain fiber types are affected. A substantial body of work has revealed molecular pathways that regulate muscle fiber type plasticity and early developmental muscle fiber identity. For instance, recent studies have revealed many factors that regulate muscle fiber type through modulating the activity of the muscle regulatory transcription factor MYOD1. Future studies of muscle fiber type development in animal models will continue to enhance our understanding of factors and pathways that may provide therapeutic targets to treat muscle diseases. WIREs Dev Biol 2016, 5:518-534. doi: 10.1002/wdev.230 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Jared Talbot
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
36
|
Han SO, Li S, Koeberl DD. Salmeterol enhances the cardiac response to gene therapy in Pompe disease. Mol Genet Metab 2016; 118:35-40. [PMID: 27017193 PMCID: PMC4833676 DOI: 10.1016/j.ymgme.2016.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/16/2016] [Accepted: 03/16/2016] [Indexed: 12/21/2022]
Abstract
Enzyme replacement therapy (ERT) with recombinant human (rh) acid α-glucosidase (GAA) has prolonged the survival of patients. However, the paucity of cation-independent mannose-6-phosphate receptor (CI-MPR) in skeletal muscle, where it is needed to take up rhGAA, correlated with a poor response to ERT by muscle in Pompe disease. Clenbuterol, a selective β2 receptor agonist, enhanced the CI-MPR expression in striated muscle through Igf-1 mediated muscle hypertrophy, which correlated with increased CI-MPR (also the Igf-2 receptor) expression. In this study we have evaluated 4 new drugs in GAA knockout (KO) mice in combination with an adeno-associated virus (AAV) vector encoding human GAA, 3 alternative β2 agonists and dehydroepiandrosterone (DHEA). Mice were injected with AAV2/9-CBhGAA (1E+11 vector particles) at a dose that was not effective at clearing glycogen storage from the heart. Heart GAA activity was significantly increased by either salmeterol (p<0.01) or DHEA (p<0.05), in comparison with untreated mice. Furthermore, glycogen content was reduced in the heart by treatment with DHEA (p<0.001), salmeterol (p<0.05), formoterol (p<0.01), or clenbuterol (p<0.01) in combination with the AAV vector, in comparison with untreated GAA-KO mice. Wirehang testing revealed that salmeterol and the AAV vector significantly increased performance, in comparison with the AAV vector alone (p<0.001). Similarly, salmeterol with the vector increased performance significantly more than any of the other drugs. The most effective individual drugs had no significant effect in absence of vector, in comparison with untreated mice. Thus, salmeterol should be further developed as adjunctive therapy in combination with either ERT or gene therapy for Pompe disease.
Collapse
Affiliation(s)
- Sang-Oh Han
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Songtao Li
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
37
|
Han SO, Pope R, Li S, Kishnani PS, Steet R, Koeberl DD. A beta-blocker, propranolol, decreases the efficacy from enzyme replacement therapy in Pompe disease. Mol Genet Metab 2016; 117:114-9. [PMID: 26454691 PMCID: PMC4755835 DOI: 10.1016/j.ymgme.2015.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 01/13/2023]
Abstract
UNLABELLED Enzyme replacement therapy (ERT) with recombinant human acid α-glucosidase (rhGAA) fails to completely reverse muscle weakness in Pompe disease. β2-agonists enhanced ERT by increasing receptor-mediated uptake of rhGAA in skeletal muscles. PURPOSE To test the hypothesis that a β-blocker might reduce the efficacy of ERT, because the action of β-blockers opposes those of β2-agonists. METHODS Mice with Pompe disease were treated with propranolol (a β-blocker) or clenbuterol in combination with ERT, or with ERT alone. RESULTS Propranolol-treated mice had decreased weight gain (p<0.01), in comparison with clenbuterol-treated mice. Left ventricular mass was decreased (and comparable to wild-type) in ERT only and clenbuterol-treated groups of mice, and unchanged in propranolol-treated mice. GAA activity increased following either clenbuterol or propranolol in skeletal muscles. However, muscle glycogen was reduced only in clenbuterol-treated mice, not in propranolol-treated mice. Cell-based experiments confirmed that propranolol reduces uptake of rhGAA into Pompe fibroblasts and also demonstrated that the drug induces intracellular accumulation of glycoproteins at higher doses. CONCLUSION Propranolol, a commonly prescribed β-blocker, reduced weight, increased left ventricular mass and decreased glycogen clearance in skeletal muscle following ERT. β-Blockers might therefore decrease the efficacy from ERT in patients with Pompe disease.
Collapse
Affiliation(s)
- Sang-Oh Han
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Rand Pope
- Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Songtao Li
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Richard Steet
- Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
38
|
Han SO, Li S, Bird A, Koeberl D. Synergistic Efficacy from Gene Therapy with Coreceptor Blockade and a β2-Agonist in Murine Pompe Disease. Hum Gene Ther 2015; 26:743-50. [PMID: 26417913 DOI: 10.1089/hum.2015.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pompe disease (glycogen storage disease type II; acid maltase deficiency) is a devastating myopathy resulting from acid α-glucosidase (GAA) deficiency in striated and smooth muscle. Despite the availability of enzyme replacement therapy (ERT) with recombinant human GAA (rhGAA), the limitations of ERT have prompted the preclinical development of gene therapy. Gene therapy has the advantage of continuously producing GAA, in contrast to ERT, which requires frequent injections of rhGAA. An adeno-associated viral (AAV) vector containing a muscle-specific promoter, AAV-MHCK7hGAApA, achieved high GAA expression in heart and skeletal muscle in mice with Pompe disease. However, elevated GAA activity was not sufficient to completely clear accumulated glycogen in skeletal muscle. The process of glycogen clearance from lysosomes might require improved trafficking of GAA to the lysosomes in skeletal muscle, previously achieved with the β(2)-agonist clenbuterol that enhanced glycogen clearance in skeletal muscle without increasing GAA activity. Glycogen clearance was clearly enhanced by treatment with a nondepleting anti-CD4 monoclonal antibody (anti-CD4 mAb) along with muscle-specific GAA expression in cardiac muscle, but that treatment was not effective in skeletal muscle. Furthermore, anti-CD4 mAb treatment along with clenbuterol achieved synergistic therapeutic efficacy in both cardiac and skeletal muscle. This triple therapy increased both muscle strength and weight gain. Overall, triple therapy to enhance GAA trafficking and to suppress immune responses significantly improved the efficacy of muscle-targeted gene therapy in murine Pompe disease.
Collapse
Affiliation(s)
- Sang-oh Han
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center , Durham, North Carolina
| | - Songtao Li
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center , Durham, North Carolina
| | - Andrew Bird
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center , Durham, North Carolina
| | - Dwight Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
39
|
Vill K, Schessl J, Teusch V, Schroeder S, Blaschek A, Schoser B, Müller-Felber W. Muscle ultrasound in classic infantile and adult Pompe disease: A useful screening tool in adults but not in infants. Neuromuscul Disord 2015; 25:120-6. [DOI: 10.1016/j.nmd.2014.09.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 09/29/2014] [Indexed: 12/14/2022]
|
40
|
Valayannopoulos V. Enzyme Replacement Therapy in Lysosomal Storage Diseases. Rare Dis 2015. [DOI: 10.1007/978-94-017-9214-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
41
|
Raval KK, Tao R, White BE, De Lange WJ, Koonce CH, Yu J, Kishnani PS, Thomson JA, Mosher DF, Ralphe JC, Kamp TJ. Pompe disease results in a Golgi-based glycosylation deficit in human induced pluripotent stem cell-derived cardiomyocytes. J Biol Chem 2014; 290:3121-36. [PMID: 25488666 DOI: 10.1074/jbc.m114.628628] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Infantile-onset Pompe disease is an autosomal recessive disorder caused by the complete loss of lysosomal glycogen-hydrolyzing enzyme acid α-glucosidase (GAA) activity, which results in lysosomal glycogen accumulation and prominent cardiac and skeletal muscle pathology. The mechanism by which loss of GAA activity causes cardiomyopathy is poorly understood. We reprogrammed fibroblasts from patients with infantile-onset Pompe disease to generate induced pluripotent stem (iPS) cells that were differentiated to cardiomyocytes (iPSC-CM). Pompe iPSC-CMs had undetectable GAA activity and pathognomonic glycogen-filled lysosomes. Nonetheless, Pompe and control iPSC-CMs exhibited comparable contractile properties in engineered cardiac tissue. Impaired autophagy has been implicated in Pompe skeletal muscle; however, control and Pompe iPSC-CMs had comparable clearance rates of LC3-II-detected autophagosomes. Unexpectedly, the lysosome-associated membrane proteins, LAMP1 and LAMP2, from Pompe iPSC-CMs demonstrated higher electrophoretic mobility compared with control iPSC-CMs. Brefeldin A induced disruption of the Golgi in control iPSC-CMs reproduced the higher mobility forms of the LAMPs, suggesting that Pompe iPSC-CMs produce LAMPs lacking appropriate glycosylation. Isoelectric focusing studies revealed that LAMP2 has a more alkaline pI in Pompe compared with control iPSC-CMs due largely to hyposialylation. MALDI-TOF-MS analysis of N-linked glycans demonstrated reduced diversity of multiantennary structures and the major presence of a trimannose complex glycan precursor in Pompe iPSC-CMs. These data suggest that Pompe cardiomyopathy has a glycan processing abnormality and thus shares features with hypertrophic cardiomyopathies observed in the congenital disorders of glycosylation.
Collapse
Affiliation(s)
- Kunil K Raval
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705, the WiCell Institute, Madison, Wisconsin 53719
| | - Ran Tao
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705
| | - Brent E White
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705
| | - Willem J De Lange
- the Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792
| | - Chad H Koonce
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705
| | - Junying Yu
- Cellular Dynamics International, Madison, Wisconsin 53711
| | - Priya S Kishnani
- the Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina 27710
| | - James A Thomson
- the Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53706, the Genome Center of Wisconsin, University of Wisconsin, Madison, Wisconsin 53706, the Morgridge Institute for Research, Madison, Wisconsin 53715
| | - Deane F Mosher
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705, the Department of Biomolecular Chemistry, University of Wisconsin, Madison, Wisconsin 53706, and
| | - John C Ralphe
- the Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792
| | - Timothy J Kamp
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705, the Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53706, the WiCell Institute, Madison, Wisconsin 53719,
| |
Collapse
|
42
|
Lim JA, Li L, Raben N. Pompe disease: from pathophysiology to therapy and back again. Front Aging Neurosci 2014; 6:177. [PMID: 25183957 PMCID: PMC4135233 DOI: 10.3389/fnagi.2014.00177] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 07/04/2014] [Indexed: 11/13/2022] Open
Abstract
Pompe disease is a lysosomal storage disorder in which acid alpha-glucosidase (GAA) is deficient or absent. Deficiency of this lysosomal enzyme results in progressive expansion of glycogen-filled lysosomes in multiple tissues, with cardiac and skeletal muscle being the most severely affected. The clinical spectrum ranges from fatal hypertrophic cardiomyopathy and skeletal muscle myopathy in infants to relatively attenuated forms, which manifest as a progressive myopathy without cardiac involvement. The currently available enzyme replacement therapy (ERT) proved to be successful in reversing cardiac but not skeletal muscle abnormalities. Although the overall understanding of the disease has progressed, the pathophysiology of muscle damage remains poorly understood. Lysosomal enlargement/rupture has long been considered a mechanism of relentless muscle damage in Pompe disease. In past years, it became clear that this simple view of the pathology is inadequate; the pathological cascade involves dysfunctional autophagy, a major lysosome-dependent intracellular degradative pathway. The autophagic process in Pompe skeletal muscle is affected at the termination stage—impaired autophagosomal-lysosomal fusion. Yet another abnormality in the diseased muscle is the accelerated production of large, unrelated to ageing, lipofuscin deposits—a marker of cellular oxidative damage and a sign of mitochondrial dysfunction. The massive autophagic buildup and lipofuscin inclusions appear to cause a greater effect on muscle architecture than the enlarged lysosomes outside the autophagic regions. Furthermore, the dysfunctional autophagy affects the trafficking of the replacement enzyme and interferes with its delivery to the lysosomes. Several new therapeutic approaches have been tested in Pompe mouse models: substrate reduction therapy, lysosomal exocytosis following the overexpression of transcription factor EB and a closely related but distinct factor E3, and genetic manipulation of autophagy.
Collapse
Affiliation(s)
- Jeong-A Lim
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health Bethesda, MD, USA
| | - Lishu Li
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health Bethesda, MD, USA
| | - Nina Raben
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health Bethesda, MD, USA
| |
Collapse
|
43
|
Chien YH, Lee NC, Tsai YJ, Thurberg BL, Tsai FJ, Hwu WL. Prominent vacuolation of the eyelid levator muscle in an early-treated child with infantile-onset Pompe disease. Muscle Nerve 2014; 50:301-2. [DOI: 10.1002/mus.24257] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/24/2014] [Accepted: 03/28/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Yin-Hsiu Chien
- Department of Pediatrics and Medical Genetics; National Taiwan University Hospital and National Taiwan University School of Medicine; Taipei Taiwan
| | - Ni-Chung Lee
- Department of Pediatrics and Medical Genetics; National Taiwan University Hospital and National Taiwan University School of Medicine; Taipei Taiwan
| | - Yueh-Ju Tsai
- Department of Ophthalmology; Chang Gung Memorial Hospital, Linkou, and College of Medicine, Chang Gung University; Taoyuan Taiwan
| | - Beth L. Thurberg
- Department of Pathology; Genzyme, a Sanofi Company; Framingham MA USA
| | - Fuu-Jen Tsai
- Department of Pediatrics and Medical Genetics; China Medical University Hospital; Taichung Taiwan
| | - Wuh-Liang Hwu
- Department of Pediatrics and Medical Genetics; National Taiwan University Hospital and National Taiwan University School of Medicine; Taipei Taiwan
| |
Collapse
|
44
|
Andreassen CS, Schlütter JM, Vissing J, Andersen H. Effect of enzyme replacement therapy on isokinetic strength for all major muscle groups in four patients with Pompe disease-a long-term follow-up. Mol Genet Metab 2014; 112:40-3. [PMID: 24685124 DOI: 10.1016/j.ymgme.2014.02.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 02/19/2014] [Indexed: 10/25/2022]
Abstract
Pompe disease is a rare, inherited metabolic myopathy characterized by progressive weakness of the proximal limb and respiratory muscles. We report the findings from four patients with late-onset Pompe disease treated with α-glucosidase (Myozyme) for 2 (n=2) and 6 (n=2) years, and monitored with isokinetic dynamometry, 6-minute walking test (6MWT), and vital capacity. Patients were evaluated after 6, 12, 24, 36, 48, 60, and 72months. In two patients, muscle size estimated by MRI and DXA scanning was also performed prior to and following 6months of treatment. After 2years of α-glucosidase treatment, maximal isokinetic muscle strength increased by 11% (0%-50%) [median (range)] and 6MWT improved by 18% (2%-40%). In the two patients treated for 6years, the increase in muscle strength stabilized at 40% and 6MWT stabilized at 32%. The improvements primarily occurred during the first 6months of treatment. Interestingly, the weakest muscle groups seemed to benefit more than those less affected, and greater improvements occurred for flexor muscles compared to extensor muscles. Vital capacity did not improve on treatment.
Collapse
Affiliation(s)
- Christer Swan Andreassen
- Clinical Neurology Research Group, Department of Neurology, Aarhus University Hospital, Nørrebrogade 44, 8000 Aarhus C., Denmark; Institute of Clinical Medicine, Aarhus University, Brendstrupgårdsvej 100, 8200 Aarhus N., Denmark.
| | - Jacob Mørup Schlütter
- Clinical Neurology Research Group, Department of Neurology, Aarhus University Hospital, Nørrebrogade 44, 8000 Aarhus C., Denmark.
| | - John Vissing
- Neuromuscular Research Unit, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Henning Andersen
- Clinical Neurology Research Group, Department of Neurology, Aarhus University Hospital, Nørrebrogade 44, 8000 Aarhus C., Denmark.
| |
Collapse
|
45
|
Farah BL, Madden L, Li S, Nance S, Bird A, Bursac N, Yen PM, Young SP, Koeberl DD. Adjunctive β2-agonist treatment reduces glycogen independently of receptor-mediated acid α-glucosidase uptake in the limb muscles of mice with Pompe disease. FASEB J 2014; 28:2272-80. [PMID: 24448824 DOI: 10.1096/fj.13-244202] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Enzyme or gene replacement therapy with acid α-glucosidase (GAA) has achieved only partial efficacy in Pompe disease. We evaluated the effect of adjunctive clenbuterol treatment on cation-independent mannose-6-phosphate receptor (CI-MPR)-mediated uptake and intracellular trafficking of GAA during muscle-specific GAA expression with an adeno-associated virus (AAV) vector in GAA-knockout (KO) mice. Clenbuterol, which increases expression of CI-MPR in muscle, was administered with the AAV vector. This combination therapy increased latency during rotarod and wirehang testing at 12 wk, in comparison with vector alone. The mean urinary glucose tetrasaccharide (Glc4), a urinary biomarker, was lower in GAA-KO mice following combination therapy, compared with vector alone. Similarly, glycogen content was lower in cardiac and skeletal muscle following 12 wk of combination therapy in heart, quadriceps, diaphragm, and soleus, compared with vector alone. These data suggested that clenbuterol treatment enhanced trafficking of GAA to lysosomes, given that GAA was expressed within myofibers. The integral role of CI-MPR was demonstrated by the lack of effectiveness from clenbuterol in GAA-KO mice that lacked CI-MPR in muscle, where it failed to reverse the high glycogen content of the heart and diaphragm or impaired wirehang performance. However, the glycogen content of skeletal muscle was reduced by the addition of clenbuterol in the absence of CI-MPR, as was lysosomal vacuolation, which correlated with increased AKT signaling. In summary, β2-agonist treatment enhanced CI-MPR-mediated uptake and trafficking of GAA in mice with Pompe disease, and a similarly enhanced benefit might be expected in other lysosomal storage disorders.
Collapse
Affiliation(s)
- Benjamin L Farah
- 2Duke University Medical Center, Box 103856, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Koeberl DD, Austin S, Case LE, Smith EC, Buckley AF, Young SP, Bali D, Kishnani PS. Adjunctive albuterol enhances the response to enzyme replacement therapy in late-onset Pompe disease. FASEB J 2014; 28:2171-6. [PMID: 24443373 DOI: 10.1096/fj.13-241893] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Effective dosages for enzyme replacement therapy (ERT) in Pompe disease are much higher than for other lysosomal storage disorders, which has been attributed to low cation-independent mannose-6-phosphate receptor (CI-MPR) in skeletal muscle. We have previously demonstrated the benefit of increased CI-MPR-mediated uptake of recombinant human acid-α-glucosidase during ERT in mice with Pompe disease following addition of albuterol therapy. Currently we have completed a pilot study of albuterol in patients with late-onset Pompe disease already on ERT for >2 yr, who were not improving further. The 6-min walk test (6MWT) distance increased in all 7 subjects at wk 6 (30±13 m; P=0.002), wk 12 (34±14 m; P=0.004), and wk 24 (42±37 m; P=0.02), in comparison with baseline. Grip strength was improved significantly for both hands at wk 12. Furthermore, individual subjects reported benefits; e.g., a female patient could stand up from sitting on the floor much more easily (time for supine to standing position decreased from 30 to 11 s), and a male patient could readily swing his legs out of his van seat (hip abduction increased from 1 to 2+ on manual muscle testing). Finally, analysis of the quadriceps biopsies suggested increased CI-MPR at wk 12 (P=0.08), compared with baseline. With the exception of 1 patient who succumbed to respiratory complications of Pompe disease in the first week, only mild adverse events have been reported, including tremor, transient difficulty falling asleep, and mild urinary retention (requiring early morning voiding). Therefore, this pilot study revealed initial safety and efficacy in an open label study of adjunctive albuterol therapy in patients with late-onset Pompe disease who had been stable on ERT with no improvements noted over the previous several years.
Collapse
Affiliation(s)
- Dwight D Koeberl
- 1Duke University Medical Center, Box 103856, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
van den Berg LEM, Drost MR, Schaart G, de Laat J, van Doorn PA, van der Ploeg AT, Reuser AJJ. Muscle fiber-type distribution, fiber-type-specific damage, and the Pompe disease phenotype. J Inherit Metab Dis 2013; 36:787-94. [PMID: 23053471 DOI: 10.1007/s10545-012-9541-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 09/03/2012] [Accepted: 09/11/2012] [Indexed: 10/27/2022]
Abstract
Pompe disease is a lysosomal storage disorder caused by acid α-glucosidase deficiency and characterized by progressive muscle weakness. Enzyme replacement therapy (ERT) has ameliorated patients' perspectives, but reversal of skeletal muscle pathology remains a challenge. We studied pretreatment biopsies of 22 patients with different phenotypes to investigate to what extent fiber-type distribution and fiber-type-specific damage contribute to clinical diversity. Pompe patients have the same fiber-type distribution as healthy persons, but among nonclassic patients with the same GAA mutation (c.-32-13T>G), those with early onset of symptoms tend to have more type 2 muscle fibers than those with late-onset disease. Further, it seemed that the older, more severely affected classic infantile patients and the wheelchair-bound and ventilated nonclassic patients had a greater proportion of type 2x muscle fibers. However, as in other diseases, this may be caused by physical inactivity of those patients.
Collapse
Affiliation(s)
- L E M van den Berg
- Department of Pediatrics, Division of Metabolic Diseases and Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
48
|
Pazopanib and sunitinib trigger autophagic and non-autophagic death of bladder tumour cells. Br J Cancer 2013; 109:1040-50. [PMID: 23887605 PMCID: PMC3749583 DOI: 10.1038/bjc.2013.420] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 06/18/2013] [Accepted: 07/03/2013] [Indexed: 12/21/2022] Open
Abstract
Background: Tyrosine kinase inhibitors (TKI) such as sunitinib and pazopanib display their efficacy in a variety of solid tumours. However, their use in therapy is limited by the lack of evidence about the ability to induce cell death in cancer cells. Our aim was to evaluate cytotoxic effects induced by sunitinib and pazopanib in 5637 and J82 bladder cancer cell lines. Methods: Cell viability was tested by MTT assay. Autophagy was evaluated by western blot using anti-LC3 and anti-p62 antibodies, acridine orange staining and FACS analysis. Oxygen radical generation and necrosis were determined by FACS analysis using DCFDA and PI staining. Cathepsin B activation was evaluated by western blot and fluorogenic Z-Arg-Arg-AMC peptide. Finally, gene expression was performed using RT–PCR Profiler array. Results: We found that sunitinib treatment for 24 h triggers incomplete autophagy, impairs cathepsin B activation and stimulates a lysosomal-dependent necrosis. By contrast, treatment for 48 h with pazopanib induces cathepsin B activation and autophagic cell death, markedly reversed by CA074-Me and 3-MA, cathepsin B and autophagic inhibitors, respectively. Finally, pazopanib upregulates the α-glucosidase and downregulates the TP73 mRNA expression. Conclusion: Our results showing distinct cell death mechanisms activated by different TKIs, provide the biological basis for novel molecularly targeted approaches.
Collapse
|
49
|
Maga JA, Zhou J, Kambampati R, Peng S, Wang X, Bohnsack RN, Thomm A, Golata S, Tom P, Dahms NM, Byrne BJ, LeBowitz JH. Glycosylation-independent lysosomal targeting of acid α-glucosidase enhances muscle glycogen clearance in pompe mice. J Biol Chem 2012. [PMID: 23188827 PMCID: PMC3548456 DOI: 10.1074/jbc.m112.438663] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We have used a peptide-based targeting system to improve lysosomal delivery of acid α-glucosidase (GAA), the enzyme deficient in patients with Pompe disease. Human GAA was fused to the glycosylation-independent lysosomal targeting (GILT) tag, which contains a portion of insulin-like growth factor II, to create an active, chimeric enzyme with high affinity for the cation-independent mannose 6-phosphate receptor. GILT-tagged GAA was taken up by L6 myoblasts about 25-fold more efficiently than was recombinant human GAA (rhGAA). Once delivered to the lysosome, the mature form of GILT-tagged GAA was indistinguishable from rhGAA and persisted with a half-life indistinguishable from rhGAA. GILT-tagged GAA was significantly more effective than rhGAA in clearing glycogen from numerous skeletal muscle tissues in the Pompe mouse model. The GILT-tagged GAA enzyme may provide an improved enzyme replacement therapy for Pompe disease patients.
Collapse
Affiliation(s)
- John A Maga
- ZyStor Therapeutics, Milwaukee, Wisconsin 53226-4838, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Nilsson MI, Samjoo IA, Hettinga BP, Koeberl DD, Zhang H, Hawke TJ, Nissar AA, Ali T, Brandt L, Ansari MU, Hazari H, Patel N, Amon J, Tarnopolsky MA. Aerobic training as an adjunctive therapy to enzyme replacement in Pompe disease. Mol Genet Metab 2012; 107:469-79. [PMID: 23041258 DOI: 10.1016/j.ymgme.2012.09.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 09/11/2012] [Accepted: 09/11/2012] [Indexed: 01/06/2023]
Abstract
BACKGROUND Aerobic exercise may be used in conjunction with enzyme replacement therapy (ERT) to attenuate cardiovascular deconditioning, skeletal muscle wasting, and loss of motor function in Pompe disease (glycogen storage disease type II; GSDII), but the effects on lysosomal glycogen content and macroautophagy have not been defined to date. PURPOSE The main objectives of this study were to determine if acute aerobic exercise enhances 24-h uptake of recombinant human enzyme (rhGAA; Myozyme® [aim 1]) and if endurance training improves disease pathology when combined with ERT [aim 2] in Pompe mice. METHODS For the first aim in our study, Pompe mutant mice (6(neo)/6(neo)) were grouped into ERT (Myozyme® injection only [40 mg/kg]) and ERT+EX (Myozyme® injection followed by 90 min treadmill exercise) cohorts, and enzyme uptake was assessed in the heart and quadriceps 24h post injection. For the second aim of our study, mutant mice were randomized into control, endurance-trained, enzyme-treated, or combination therapy groups. Exercised animals underwent 14 weeks of progressive treadmill training with or without biweekly Myozyme® injections (40 mg/kg) and tissues were harvested 1 week post last treatment. RESULTS Myozyme® uptake (GAA activity) was not improved in ERT+EX over ERT alone at 24-h post injection. Endurance exercise training, with or without ERT, improved aerobic capacity and normalized grip strength, motor function, and lean mass (P<0.05), but did not reduce glycogen content or normalize macroautophagy beyond traditional enzyme replacement therapy. CONCLUSIONS Endurance training is beneficial as an adjunctive therapy to ERT in Pompe disease, although it works by mechanisms independent of a reduction in glycogen content.
Collapse
Affiliation(s)
- Mats I Nilsson
- Department of Pediatrics and Medicine, Neuromuscular Clinic, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|