1
|
Speziale R, Hocquemiller M, Mei X, Fabbrini D, Malancona S, Aiach K, Laufer R, Orsatti L. Tandem mass spectrometry-based assay for heparan-N-sulphatase in paediatric CSF: A potential pharmacodynamic biomarker for mucopolysaccharidosis type IIIA therapy. Clin Chim Acta 2024; 565:119987. [PMID: 39368684 DOI: 10.1016/j.cca.2024.119987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Mucopolysaccharidosis type IIIA is a lysosomal storage disorder caused by mutations in the gene coding for heparan-N-sulphatase, a crucial enzyme in the degradation of heparan sulfate. In mucopolysaccharidosis type IIIA, heparan sulfate accumulates in the lysosomes, predominantly affecting the central nervous system. It is the most common and most severe form of mucopolysaccharidosis type III, with onset typically before the age of ten years. There is an ongoing effort to develop therapies that aim at restoring enzyme function in the brain. This study introduces a novel tandem mass spectrometry method for assessing heparan-N-sulphatase activity in pediatric cerebrospinal fluid from healthy and disease individuals. Analysis of cerebrospinal fluid samples revealed marked differences in enzyme activity, with mucopolysaccharidosis type IIIA individuals exhibiting significantly reduced levels. This new method could serve as a valuable tool for evaluating the efficacy of future therapeutic interventions targeting sulphatase activity restoration in the brain.
Collapse
Affiliation(s)
- Roberto Speziale
- Experimental Pharmacology Department, IRBM SpA, Pomezia, Roma, Italy
| | | | - Xin Mei
- Lysogene, Neuilly-sur-Seine, France
| | - Danilo Fabbrini
- Medicinal Chemistry Department, IRBM SpA, Pomezia, Roma, Italy
| | | | | | | | - Laura Orsatti
- Experimental Pharmacology Department, IRBM SpA, Pomezia, Roma, Italy.
| |
Collapse
|
2
|
Polgreen LE, Chen AH, Pak Y, Luzzi A, Morales Garval A, Acevedo J, Bitan G, Iacovino M, O'Neill C, Eisengart JB. Anakinra in Sanfilippo syndrome: a phase 1/2 trial. Nat Med 2024; 30:2473-2479. [PMID: 38907160 PMCID: PMC11405265 DOI: 10.1038/s41591-024-03079-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 05/19/2024] [Indexed: 06/23/2024]
Abstract
Sanfilippo syndrome is a fatal childhood neurodegenerative disorder involving neuroinflammation among multiple pathologies. We hypothesized that anakinra, a recombinant interleukin-1 receptor antagonist, could improve neurobehavioral and functional symptoms owing to its capacity to treat neuroinflammation. This phase 1/2 trial aimed to test the safety, tolerability and effects of anakinra on neurobehavioral, functional and quality-of-life outcomes in patients and their caregivers. The primary outcome was the percent of participants requiring a dose increase at week 8 or week 16. Secondary efficacy outcomes included a multi-domain responder index (MDRI). Twenty-three participants (6-26 years of age) were enrolled. Twenty continued treatment to week 8, and 15 (75%) required an increased dose at week 8 or week 16. There was an improvement in at least one domain in the MDRI in 18 of 21 (86%) at week 8 and in 15 of 16 (94%) at week 36. Seven participants withdrew (intolerability of daily injections and lost to follow-up) before week 36. Adverse events occurred in 22 of 23 (96%) participants, most commonly mild injection site reactions. No serious adverse events were related to anakinra. In conclusion, anakinra was safe and associated with improved neurobehavioral and functional outcomes, supporting continued investigation of anakinra in Sanfilippo syndrome and other mucopolysaccharidoses. ClinicalTrials.gov identifier: NCT04018755 .
Collapse
Affiliation(s)
- Lynda E Polgreen
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA.
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Agnes H Chen
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Youngju Pak
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anna Luzzi
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Adolfo Morales Garval
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jonathan Acevedo
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine, Brain Research Institute and Molecular Biology Institute University of California, Los Angeles, Los Angeles, CA, USA
| | - Michelina Iacovino
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Julie B Eisengart
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
3
|
Alyazidi AS, Muthaffar OY, Baaishrah LS, Shawli MK, Jambi AT, Aljezani MA, Almaghrabi MA. Current Concepts in the Management of Sanfilippo Syndrome (MPS III): A Narrative Review. Cureus 2024; 16:e58023. [PMID: 38738088 PMCID: PMC11087936 DOI: 10.7759/cureus.58023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
Sanfilippo syndrome is a childhood-onset (1-4 years) autosomal recessive lysosomal storage disease that presents as a neurodegenerative disease by targeting the brain and spinal cord. It is also known as mucopolysaccharidosis III. Mucopolysaccharidosis III is divided into four subtypes (A, B, C, or D). It can cause delayed speech, behavior problems, and features of autism spectrum disorder. Sanfilippo syndrome is of a higher prevalence within consanguineous families that carry its gene alteration. If both parents have a nonfunctional copy of a gene linked to this condition, their children will have a 25% (1 in 4) chance of developing the disease. In Saudi Arabia, the incidence rate is estimated at 2 per 100,000 live births. Recent research focused on promising treatment approaches, such as gene therapy, modified enzyme replacement therapy, and stem cells. These approaches work by exogenous administration of the proper version of the mutant enzyme (enzyme replacement therapy), cleaning the defective enzyme in individuals with glycolipid storage disorders (substrate reduction therapy), or using a pharmacological chaperone to target improperly folded proteins. However, there is currently no approved curative medication for Sanfilippo syndrome that can effectively halt or reverse the disorder.
Collapse
Affiliation(s)
- Anas S Alyazidi
- Pediatrics, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Osama Y Muthaffar
- Pediatrics, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Layan S Baaishrah
- Faculty of Pharmacy, King Abdulaziz University Hospital, Jeddah, SAU
| | - Mohammed K Shawli
- Medicine, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Abdulaziz T Jambi
- Medicine, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Maram A Aljezani
- Pediatric Neurology, King Abdulaziz University Hospital, Jeddah, SAU
| | | |
Collapse
|
4
|
Shapiro EG, Eisengart JB, Whiteman D, Whitley CB. Ability change across multiple domains in mucopolysaccharidosis (Sanfilippo syndrome) type IIIA. Mol Genet Metab 2024; 141:108110. [PMID: 38151384 DOI: 10.1016/j.ymgme.2023.108110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/29/2023]
Abstract
The objective of this paper is 1) to expand the scope of the domains previously published in a natural history study of Mucopolysaccharidosis IIIA (Sanfilippo syndrome type A) (MPS IIIA) and 2) to present evidence regarding the capacity of a new metric, Growth Scale Values (GSVs), in comparison with traditional metrics, to show changes in skills as assessed by the Bayley Scales of Infant Development -III (BSID-III) and the Vineland Adaptive Behavior Scales, Second Edition (VABS-II). We re-analyzed a cohort of 25 children, 20 with rapid progressing disease and 5 with slow progression, who had been followed over two years using the BSID-III, and the VABS-II. Previously findings were reported using age equivalent scores; now we are also presenting findings with GSVs. For the re-analysis, Language and Motor scores were added to the Cognitive scale on the BSID-III, and Domain- and Subdomain-level scores added to the Total VABS-II score (i.e., ABC Composite). We evaluated raw scores, age equivalent scores, and GSVs (and standard scores for the VABS-II only). Individual patient data can be found in the appendices to this publication. Results indicate that 1) Cognition as measured by GSVs was the most sensitive to decline; 2) GSVs showed significant decline in the range of 4 to 6 years of age; 3) For children under 4 years of age, positive growth occurs on most scales and most metrics, with the exception of language which slows somewhat earlier; 4) Other than the Cognitive scale, Receptive Language on the BSID-III and Receptive Communication on the VABS-II showed the most sensitivity to change; 5) Gross Motor skills showed the least decline over time and appeared to lack sensitivity to MPS IIIA motor concerns; and 6) No evidence for sensitivity to change for any metric was found in time intervals less than one year. We conclude that GSVs are a precise measurement of change to detect decline in function, and they are a valuable method for future clinical trials in MPS IIIA. Evidence continues to support cognition as a primary endpoint. Additional work is needed to identify sensitive measures of meaningful endpoints to families.
Collapse
Affiliation(s)
- Elsa G Shapiro
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA; Shapiro Neuropsychology Consulting, LLC, Portland, OR, USA
| | - Julie B Eisengart
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.
| | | | - Chester B Whitley
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA; Advanced Therapies Program, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
5
|
Veraldi N, Quadri ID, van de Looij Y, Modernell LM, Sinquin C, Zykwinska A, Tournier BB, Dalonneau F, Li H, Li JP, Millet P, Vives R, Colliec-Jouault S, de Agostini A, Sanches EF, Sizonenko SV. Low-molecular weight sulfated marine polysaccharides: Promising molecules to prevent neurodegeneration in mucopolysaccharidosis IIIA? Carbohydr Polym 2023; 320:121214. [PMID: 37659814 DOI: 10.1016/j.carbpol.2023.121214] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 09/04/2023]
Abstract
Mucopolysaccharidosis IIIA is a hereditary disease caused by mutations in the sulfamidase enzyme that participates in catabolism of heparan sulfate (HS), leading to HS fragment accumulation and multisystemic failure. No cure exists and death occurs around the second decade of life. Two low molecular weight highly sulfated compounds derived from marine diabolican and infernan exopolysaccharides (A5_3 and A5_4, respectively) with heparanase inhibiting properties were tested in a MPSIIIA cell line model, resulting in limited degradation of intracellular HS. Next, we observed the effects of intraperitoneal injections of the diabolican derivative A5_3 from 4 to 12 weeks of age on MPSIIIA mice. Brain metabolism and microstructure, levels of proteins and genes involved in MPSIIIA brain pathophysiology were also investigated. 1H-Magnetic Resonance Spectroscopy (MRS) indicated deficits in energetic metabolism, tissue integrity and neurotransmission at both 4 and 12 weeks in MPSIIIA mice, with partial protective effects of A5_3. Ex-vivo Diffusion Tensor Imaging (DTI) showed white matter microstructural damage in MPSIIIA, with noticeable protective effects of A5_3. Protein and gene expression assessments displayed both pro-inflammatory and pro-apoptotic profiles in MPSIIIA mice, with benefits of A5_3 counteracting neuroinflammation. Overall, derivative A5_3 was well tolerated and was shown to be efficient in preventing brain metabolism failure and inflammation, resulting in preserved brain microstructure in the context of MPSIIIA.
Collapse
Affiliation(s)
- Noemi Veraldi
- Division of Clinical Pathology, Department of Diagnostics, Geneva University Hospitals, Geneva, Switzerland.
| | - Isabelle Dentand Quadri
- Department of Pathology and Immunology, Faculty of Medicine, Geneva University, Geneva, Switzerland.
| | - Yohan van de Looij
- Center for Biomedical Imaging, Animal Imaging Technology section, Federal Polytechnic School of Lausanne, Lausanne, Switzerland; Division of Development and Growth, Department of Pediatrics & Gynecology & Obstetrics, Children's Hospital, Geneva University Hospitals, Geneva, Switzerland.
| | - Laura Malaguti Modernell
- Division of Development and Growth, Department of Pediatrics & Gynecology & Obstetrics, Children's Hospital, Geneva University Hospitals, Geneva, Switzerland
| | | | | | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland.
| | | | - Honglian Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden.
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden.
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland.
| | - Romain Vives
- University of Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France.
| | | | - Ariane de Agostini
- Division of Clinical Pathology, Department of Diagnostics, Geneva University Hospitals, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, Geneva University, Geneva, Switzerland.
| | - Eduardo Farias Sanches
- Division of Development and Growth, Department of Pediatrics & Gynecology & Obstetrics, Children's Hospital, Geneva University Hospitals, Geneva, Switzerland.
| | - Stéphane V Sizonenko
- Division of Development and Growth, Department of Pediatrics & Gynecology & Obstetrics, Children's Hospital, Geneva University Hospitals, Geneva, Switzerland.
| |
Collapse
|
6
|
Ashby F, Park H, Svensson M, Heldermon CD. Economic Burden of Sanfilippo Syndrome in the United States. RESEARCH SQUARE 2023:rs.3.rs-3001450. [PMID: 37398464 PMCID: PMC10312916 DOI: 10.21203/rs.3.rs-3001450/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Introduction Sanfilippo syndrome is a rare disease and fatal genetic disorder with no FDA-approved treatment in the United States (US), and no comprehensive assessment of economic disease burden is available. Objectives To develop a model to estimate the economic burden associated with Sanfilippo syndrome in the US using direct costs, indirect costs and valued intangibles (disability-adjusted life years, or DALYs) from 2023 onward. Design and Setting A multistage comorbidity model was generated based on Sanfilippo syndrome symptoms, and disability weights from the 2010 Global Burden of Disease Study. Attributable increase in caregiver mental health burden were estimated using data from the CDC National Comorbidity Survey and retrospective studies on caregiver burden. Direct costs were approximated from the 2019 EveryLife Foundation survey, and indirect costs were estimated from Federal income data. Monetary valuations were adjusted to USD 2023 and given a 3% discount rate from 2023 onward. Main Outcome Measures Incidence of Sanfilippo syndrome was calculated for each year, and year-over-year DALYs due to patient years lived with disability (YLDs) and years life lost (YLLs) were calculated by comparing to the health-adjusted life expectancy (HALE) in the US. Direct and indirect costs were calculated for each simulated patient from onset of symptoms to death. Results From 2023-2043, overall economic burden in the US attributable to Sanfilippo syndrome was estimated to be $2.04 billion USD present value (2023) with current standard of care. The burden to individual families exceeded $8 million present value from time of birth per child born with Sanfilippo syndrome. Conclusion Sanfilippo syndrome is a rare lysosomal storage disease, however the severe burden associated with the disease for individual families demonstrates a considerable cumulative impact. Our model represents the first disease burden value estimate associated with Sanfilippo syndrome, and underscores the substantial morbidity and mortality burden of Sanfilippo syndrome.
Collapse
Affiliation(s)
- Frederick Ashby
- College of Medicine, University of Florida - Gainesville, Florida, USA
| | - Haesuk Park
- College of Pharmacy, University of Florida - Gainesville, Florida, USA
| | - Mikael Svensson
- College of Pharmacy, University of Florida - Gainesville, Florida, USA
| | - Coy D Heldermon
- College of Medicine, University of Florida - Gainesville, Florida, USA
| |
Collapse
|
7
|
Pollock K, Noritake S, Imai DM, Pastenkos G, Olson M, Cary W, Yang S, Fierro FA, White J, Graham J, Dahlenburg H, Johe K, Nolta JA. An immune deficient mouse model for mucopolysaccharidosis IIIA (Sanfilippo syndrome). Sci Rep 2023; 13:18439. [PMID: 37891179 PMCID: PMC10611714 DOI: 10.1038/s41598-023-45178-0] [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: 02/23/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Mucopolysaccharidosis III (MPSIII, Sanfilippo syndrome) is a devastating lysosomal storage disease that primarily affects the central nervous system. MPSIIIA is caused by loss-of-function mutations in the gene coding for sulfamidase (N-sulfoglucosamine sulfohydrolase/SGSH) resulting in SGSH enzyme deficiency, a buildup of heparin sulfate and subsequent neurodegeneration. There is currently no cure or disease modifying treatment for MPSIIIA. A mouse model for MPSIIIA was characterized in 1999 and later backcrossed onto the C57BL/6 background. In the present study, a novel immune deficient MPSIIIA mouse model (MPSIIIA-TKO) was created by backcrossing the immune competent, C57BL/6 MPSIIIA mouse to an immune deficient mouse model lacking Rag2, CD47 and Il2rg genes. The resulting mouse model has undetectable SGSH activity, exhibits histological changes consistent with MPSIIIA and lacks T cells, B cells and NK cells. This new mouse model has the potential to be extremely useful in testing human cellular therapies in an animal model as it retains the MPSIIIA disease phenotype while tolerating xenotransplantation.
Collapse
Affiliation(s)
- Kari Pollock
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA.
| | - Sabrina Noritake
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Denise M Imai
- Comparative Pathology Laboratory, University of California Davis, School of Veterinary Medicine, Davis, CA, USA
| | - Gabrielle Pastenkos
- Comparative Pathology Laboratory, University of California Davis, School of Veterinary Medicine, Davis, CA, USA
| | - Marykate Olson
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Whitney Cary
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Sheng Yang
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Fernando A Fierro
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Jeannine White
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Justin Graham
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Heather Dahlenburg
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Karl Johe
- ReMotor Therapeutics, Inc., San Diego, CA, USA
| | - Jan A Nolta
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| |
Collapse
|
8
|
Petrova R, Patil AR, Trinh V, McElroy KE, Bhakta M, Tien J, Wilson DS, Warren L, Stratton JR. Disease pathology signatures in a mouse model of Mucopolysaccharidosis type IIIB. Sci Rep 2023; 13:16699. [PMID: 37794029 PMCID: PMC10550979 DOI: 10.1038/s41598-023-42431-4] [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: 02/02/2023] [Accepted: 09/10/2023] [Indexed: 10/06/2023] Open
Abstract
Mucopolysaccharidosis type IIIB (MPS IIIB) is a rare and devastating childhood-onset lysosomal storage disease caused by complete loss of function of the lysosomal hydrolase α-N-acetylglucosaminidase. The lack of functional enzyme in MPS IIIB patients leads to the progressive accumulation of heparan sulfate throughout the body and triggers a cascade of neuroinflammatory and other biochemical processes ultimately resulting in severe mental impairment and early death in adolescence or young adulthood. The low prevalence and severity of the disease has necessitated the use of animal models to improve our knowledge of the pathophysiology and for the development of therapeutic treatments. In this study, we took a systematic approach to characterizing a classical mouse model of MPS IIIB. Using a series of histological, biochemical, proteomic and behavioral assays, we tested MPS IIIB mice at two stages: during the pre-symptomatic and early symptomatic phases of disease development, in order to validate previously described phenotypes, explore new mechanisms of disease pathology and uncover biomarkers for MPS IIIB. Along with previous findings, this study helps provide a deeper understanding of the pathology landscape of this rare disease with high unmet medical need and serves as an important resource to the scientific community.
Collapse
Affiliation(s)
- Ralitsa Petrova
- Biologics Discovery Science, Teva Pharmaceutical Industries Ltd, Redwood City, CA, USA.
| | - Abhijeet R Patil
- Genomics and Computational Biology, Teva Pharmaceutical Industries Ltd, West Chester, PA, USA
| | - Vivian Trinh
- Biologics Discovery Science, Teva Pharmaceutical Industries Ltd, Redwood City, CA, USA
| | - Kathryn E McElroy
- Biologics Discovery Science, Teva Pharmaceutical Industries Ltd, Redwood City, CA, USA
| | - Minoti Bhakta
- Biologics Discovery Science, Teva Pharmaceutical Industries Ltd, Redwood City, CA, USA
| | - Jason Tien
- Biologics Discovery Science, Teva Pharmaceutical Industries Ltd, Redwood City, CA, USA
| | - David S Wilson
- Biologics Discovery Science, Teva Pharmaceutical Industries Ltd, Redwood City, CA, USA
| | - Liling Warren
- Genomics and Computational Biology, Teva Pharmaceutical Industries Ltd, West Chester, PA, USA
| | - Jennifer R Stratton
- Biologics Discovery Science, Teva Pharmaceutical Industries Ltd, Redwood City, CA, USA.
| |
Collapse
|
9
|
De Pasquale V, Esposito A, Scerra G, Scarcella M, Ciampa M, Luongo A, D’Alonzo D, Guaragna A, D’Agostino M, Pavone LM. N-Substituted l-Iminosugars for the Treatment of Sanfilippo Type B Syndrome. J Med Chem 2023; 66:1790-1808. [PMID: 36696678 PMCID: PMC9923752 DOI: 10.1021/acs.jmedchem.2c01617] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Sanfilippo syndrome comprises a group of four genetic diseases due to the lack or decreased activity of enzymes involved in heparan sulfate (HS) catabolism. HS accumulation in lysosomes and other cellular compartments results in tissue and organ dysfunctions, leading to a wide range of clinical symptoms including severe neurodegeneration. To date, no approved treatments for Sanfilippo disease exist. Here, we report the ability of N-substituted l-iminosugars to significantly reduce substrate storage and lysosomal dysfunctions in Sanfilippo fibroblasts and in a neuronal cellular model of Sanfilippo B subtype. Particularly, we found that they increase the levels of defective α-N-acetylglucosaminidase and correct its proper sorting toward the lysosomal compartment. Furthermore, l-iminosugars reduce HS accumulation by downregulating protein levels of exostosin glycosyltransferases. These results highlight an interesting pharmacological potential of these glycomimetics in Sanfilippo syndrome, paving the way for the development of novel therapeutic approaches for the treatment of such incurable disease.
Collapse
Affiliation(s)
- Valeria De Pasquale
- Department
of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy
| | - Anna Esposito
- Department
of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy
| | - Gianluca Scerra
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Melania Scarcella
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Mariangela Ciampa
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Antonietta Luongo
- AORN
Sant’Anna e San Sebastiano, Via F. Palasciano, 81100 Caserta, Italy
| | - Daniele D’Alonzo
- Department
of Chemical Sciences, University of Naples
Federico II, Via Cintia, 80126 Napoli, Italy
| | - Annalisa Guaragna
- Department
of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy,
| | - Massimo D’Agostino
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy,
| | - Luigi Michele Pavone
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy,
| |
Collapse
|
10
|
Rintz E, Podlacha M, Cyske Z, Pierzynowska K, Węgrzyn G, Gaffke L. Activities of (Poly)phenolic Antioxidants and Other Natural Autophagy Modulators in the Treatment of Sanfilippo Disease: Remarkable Efficacy of Resveratrol in Cellular and Animal Models. Neurotherapeutics 2023; 20:254-271. [PMID: 36344724 PMCID: PMC10119361 DOI: 10.1007/s13311-022-01323-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2022] [Indexed: 11/09/2022] Open
Abstract
Sanfilippo disease, caused by mutations in the genes encoding heparan sulfate (HS) (a glycosaminoglycan; GAG) degradation enzymes, is a mucopolysaccharidosis (MPS), which is also known as MPS type III, and is characterized by subtypes A, B, C, and D, depending on identity of the dysfunctional enzyme. The lack of activity or low residual activity of an HS-degrading enzyme leads to excess HS in the cells, impairing the functions of different types of cells, including neurons. The disease usually leads to serious psychomotor dysfunction and death before adulthood. In this work, we show that the use of molecules known as dietary (poly)phenolic antioxidants and other natural compounds known as autophagy activators (genistein, capsaicin, curcumin, resveratrol, trehalose, and calcitriol) leads to accelerated degradation of accumulated HS in the fibroblasts of all subtypes of MPS III. Both the cytotoxicity tests we performed and the available literature data indicated that the use of selected autophagy inducers was safe. Since it showed the highest effectivity in cellular models, resveratrol efficacy was tested in experiments with a mouse model of MPS IIIB. Urinary GAG levels were normalized in MPS IIIB mice treated with 50 mg/kg/day resveratrol for 12 weeks or longer. Behavioral tests indicated complete correction of hyperactivity and anxiety in these animals. Biochemical analyses indicated that administration of resveratrol caused autophagy stimulation through an mTOR-independent pathway in the brains and livers of the MPS IIIB mice. These results indicate the potential use of resveratrol (and possibly other autophagy stimulators) in the treatment of Sanfilippo disease.
Collapse
Affiliation(s)
- Estera Rintz
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Magdalena Podlacha
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Zuzanna Cyske
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Lidia Gaffke
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland.
| |
Collapse
|
11
|
Harmatz P, Muenzer J, Ezgü F, Dalén P, Huledal G, Lindqvist D, Gelius SS, Wikén M, Önnestam K, Bröijersén A. Chemically modified recombinant human sulfamidase (SOBI003) in mucopolysaccharidosis IIIA patients: Results from an open, non-controlled, multicenter study. Mol Genet Metab 2022; 136:249-259. [PMID: 35835061 DOI: 10.1016/j.ymgme.2022.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Mucopolysaccharidosis IIIA (MPS IIIA) is an inherited lysosomal storage disorder caused by mutations in the N-sulfoglucosamine sulfohydrolase gene that result in deficient enzymatic degradation of heparan sulfate (HS), resulting in progressive neurodegeneration in early childhood and premature death. A chemically modified variant of recombinant human sulfamidase, SOBI003, has shown to cross the blood-brain barrier (BBB) in mice and achieve pharmacologically relevant levels in cerebrospinal fluid (CSF). We report on a phase 1/2, open-label, first-in-human (FIH) study (NCT03423186) and its extension study (NCT03811028) to evaluate the long-term safety, tolerability, pharmacokinetics/pharmacodynamics (PK/PD) and clinical efficacy of SOBI003 in patients with MPS IIIA for up to 104 weeks. METHODS Six patients aged 1-6 years with confirmed MPS IIIA with developmental age ≥ 12 months received weekly intravenous injections of SOBI003 at 3 mg/kg (Cohort 1, n = 3) or 10 mg/kg (Cohort 2, n = 3). During the extension study, the individual dose of SOBI003 could be adjusted up to 20 mg/kg at the discretion of the investigator. RESULTS SOBI003 was generally well tolerated. Serum concentrations of SOBI003 increased in proportion to dose, and presence in CSF confirmed that SOBI003 crosses the BBB. Anti-drug antibodies (ADA) were detected in serum and CSF in all patients, with subsequent reductions in serum SOBI003 exposure at high ADA titers. SOBI003 exerted a clear PD effect: a mean reduction in HS levels in CSF of 79% was recorded at the last assessment, together with reductions in HS levels in serum and urine. Neurocognitive development age-equivalent scores showed a stabilization of cognition for all patients, whereas no clear overall clinical effect was observed on adaptive behavior, sleep pattern or quality of life. CONCLUSION SOBI003 was well tolerated when administered as weekly intravenous infusions at doses of up to 20 mg/kg for up to 104 weeks. ADA development was common and likely affected both PK and PD parameters. SOBI003 crossed the BBB and showed pharmacological activity on HS in CSF.
Collapse
Affiliation(s)
- Paul Harmatz
- UCSF Benioff Children's Hospital Oakland, Oakland, CA, USA.
| | - Joseph Muenzer
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Fatih Ezgü
- Gazi University Hospital, Ankara, Turkey
| | - Per Dalén
- Swedish Orphan Biovitrum AB, SE-112 76 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
12
|
Ballantyne M, Chiu B, Sergi CM. Sanfilippo Syndrome Type A: Early Cardiac Involvement of Two patients with Cardiac Manifestations. Cardiovasc Pathol 2022; 60:107430. [DOI: 10.1016/j.carpath.2022.107430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 11/03/2022] Open
|
13
|
Splicing Modulation as a Promising Therapeutic Strategy for Lysosomal Storage Disorders: The Mucopolysaccharidoses Example. Life (Basel) 2022; 12:life12050608. [PMID: 35629276 PMCID: PMC9146820 DOI: 10.3390/life12050608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/07/2022] [Accepted: 04/15/2022] [Indexed: 11/17/2022] Open
Abstract
Over recent decades, the many functions of RNA have become more evident. This molecule has been recognized not only as a carrier of genetic information, but also as a specific and essential regulator of gene expression. Different RNA species have been identified and novel and exciting roles have been unveiled. Quite remarkably, this explosion of novel RNA classes has increased the possibility for new therapeutic strategies that tap into RNA biology. Most of these drugs use nucleic acid analogues and take advantage of complementary base pairing to either mimic or antagonize the function of RNAs. Among the most successful RNA-based drugs are those that act at the pre-mRNA level to modulate or correct aberrant splicing patterns, which are caused by specific pathogenic variants. This approach is particularly tempting for monogenic disorders with associated splicing defects, especially when they are highly frequent among affected patients worldwide or within a specific population. With more than 600 mutations that cause disease affecting the pre-mRNA splicing process, we consider lysosomal storage diseases (LSDs) to be perfect candidates for this type of approach. Here, we introduce the overall rationale and general mechanisms of splicing modulation approaches and highlight the currently marketed formulations, which have been developed for non-lysosomal genetic disorders. We also extensively reviewed the existing preclinical studies on the potential of this sort of therapeutic strategy to recover aberrant splicing and increase enzyme activity in our diseases of interest: the LSDs. Special attention was paid to a particular subgroup of LSDs: the mucopolysaccharidoses (MPSs). By doing this, we hoped to unveil the unique therapeutic potential of the use of this sort of approach for LSDs as a whole.
Collapse
|
14
|
Kaczor-Kamińska M, Kamiński K, Wróbel M. Heparan Sulfate, Mucopolysaccharidosis IIIB and Sulfur Metabolism Disorders. Antioxidants (Basel) 2022; 11:antiox11040678. [PMID: 35453363 PMCID: PMC9026333 DOI: 10.3390/antiox11040678] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 02/01/2023] Open
Abstract
Mucopolysaccharidosis, type IIIB (MPS IIIB) is a rare disease caused by mutations in the N-alpha-acetylglucosaminidase (NAGLU) gene resulting in decreased or absent enzyme activity. On the cellular level, the disorder is characterized by the massive lysosomal storage of heparan sulfate (HS)—one species of glycosaminoglycans. HS is a sulfur-rich macromolecule, and its accumulation should affect the turnover of total sulfur in cells; according to the studies presented here, it, indeed, does. The lysosomal degradation of HS in cells produces monosaccharides and inorganic sulfate (SO42−). Sulfate is a product of L-cysteine metabolism, and any disruption of its levels affects the entire L-cysteine catabolism pathway, which was first reported in 2019. It is known that L-cysteine level is elevated in cells with the Naglu−/− gene mutation and in selected tissues of individuals with MPS IIIB. The level of glutathione and the Naglu−/− cells’ antioxidant potential are significantly reduced, as well as the activity of 3-mercaptopyruvate sulfurtransferase (MPST, EC 2.8.1.2) and the level of sulfane sulfur-containing compounds. The direct reason is not yet known. This paper attempts to identify some of cause-and-effect correlations that may lead to this condition and identifies research directions that should be explored.
Collapse
Affiliation(s)
- Marta Kaczor-Kamińska
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Krakow, Poland;
- Correspondence: ; Tel.: +48-12-422-7400
| | - Kamil Kamiński
- Department of Physical Chemistry, Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa St., 30-387 Krakow, Poland;
| | - Maria Wróbel
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Krakow, Poland;
| |
Collapse
|
15
|
Montenegro YHA, de Souza CFM, Kubaski F, Trapp FB, Burin MG, Michelin-Tirelli K, Leistner-Segal S, Facchin ACB, Medeiros FS, Giugliani L, Ribeiro EM, Lourenço CM, Cardoso-Dos-Santos AC, Ribeiro MG, Kim CA, Castro MAA, Embiruçu EK, Steiner CE, Moreira MLC, Montano HQ, Baldo G, Giugliani R. Sanfilippo syndrome type B: Analysis of patients diagnosed by the MPS Brazil Network. Am J Med Genet A 2021; 188:760-767. [PMID: 34806811 DOI: 10.1002/ajmg.a.62572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/15/2021] [Accepted: 10/29/2021] [Indexed: 11/08/2022]
Abstract
Mucopolysaccharidosis type IIIB is a rare autosomal recessive disorder characterized by deficiency of the enzyme N-acetyl-alpha-d-glucosaminidase (NAGLU), caused by biallelic pathogenic variants in the NAGLU gene, which leads to storage of heparan sulfate and a series of clinical consequences which hallmark is neurodegeneration. In this study clinical, epidemiological, and biochemical data were obtained from MPS IIIB patients diagnosed from 2004-2019 by the MPS Brazil Network ("Rede MPS Brasil"), which was created with the goal to provide an easily accessible and comprehensive investigation of all MPS types. One hundred and ten MPS IIIB patients were diagnosed during this period. Mean age at diagnosis was 10.9 years. Patients were from all over Brazil, with a few from abroad, with a possible cluster of MPS IIIB identified in Ecuador. All patients had increased urinary levels of glycosaminoglycans and low NAGLU activity in blood. Main clinical symptoms reported at diagnosis were coarse facies and neurocognitive regression. The most common variant was p.Leu496Pro (30% of alleles). MPS IIIB seems to be relatively frequent in Brazil, but patients are diagnosed later than in other countries, and reasons for that probably include the limited awareness about the disease by health professionals and the difficulties to access diagnostic tests, factors that the MPS Brazil Network is trying to mitigate.
Collapse
Affiliation(s)
- Yorran Hardman Araújo Montenegro
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Post-graduate Program in Genetics and Molecular Biology, Department of Genetics/UFRGS, Porto Alegre, Brazil.,INAGEMP, Porto Alegre, Brazil.,MPS Brazil Network, Medical Genetics Service, HCPA, Porto Alegre, Brazil
| | | | - Francyne Kubaski
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Franciele Barbosa Trapp
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,INAGEMP, Porto Alegre, Brazil.,MPS Brazil Network, Medical Genetics Service, HCPA, Porto Alegre, Brazil
| | - Maira Graeff Burin
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,MPS Brazil Network, Medical Genetics Service, HCPA, Porto Alegre, Brazil
| | - Kristiane Michelin-Tirelli
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,MPS Brazil Network, Medical Genetics Service, HCPA, Porto Alegre, Brazil
| | - Sandra Leistner-Segal
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,MPS Brazil Network, Medical Genetics Service, HCPA, Porto Alegre, Brazil
| | - Ana Carolina Brusius Facchin
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,MPS Brazil Network, Medical Genetics Service, HCPA, Porto Alegre, Brazil
| | - Fernanda S Medeiros
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,MPS Brazil Network, Medical Genetics Service, HCPA, Porto Alegre, Brazil
| | | | | | - Charles Marques Lourenço
- Centro Universitário Estácio, Ribeirão Preto, Brazil.,Faculdade de Medicina de São José do Rio Preto, FAMERP, São José do Rio Preto, Brazil
| | - Augusto César Cardoso-Dos-Santos
- Post-graduate Program in Genetics and Molecular Biology, Department of Genetics/UFRGS, Porto Alegre, Brazil.,INAGEMP, Porto Alegre, Brazil
| | - Márcia Gonçalves Ribeiro
- Medical Genetics Service, Instituto de Puericultura e Pediatria Martagão Gesteira/UFRJ, Rio de Janeiro, Brazil
| | - Chong Ae Kim
- Genetic Unity, Instituto da Criança HC FMUSP, São Paulo, Brazil
| | | | | | - Carlos Eduardo Steiner
- Department of Medical Genetics and Genomic Medicine, Faculdade de Ciências Médicas/UNICAMP, São Paulo, Brazil
| | | | | | - Guilherme Baldo
- Post-graduate Program in Genetics and Molecular Biology, Department of Genetics/UFRGS, Porto Alegre, Brazil
| | - Roberto Giugliani
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Post-graduate Program in Genetics and Molecular Biology, Department of Genetics/UFRGS, Porto Alegre, Brazil.,INAGEMP, Porto Alegre, Brazil.,MPS Brazil Network, Medical Genetics Service, HCPA, Porto Alegre, Brazil
| |
Collapse
|
16
|
Jensen TL, Gøtzsche CR, Woldbye DPD. Current and Future Prospects for Gene Therapy for Rare Genetic Diseases Affecting the Brain and Spinal Cord. Front Mol Neurosci 2021; 14:695937. [PMID: 34690692 PMCID: PMC8527017 DOI: 10.3389/fnmol.2021.695937] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, gene therapy has been raising hopes toward viable treatment strategies for rare genetic diseases for which there has been almost exclusively supportive treatment. We here review this progress at the pre-clinical and clinical trial levels as well as market approvals within diseases that specifically affect the brain and spinal cord, including degenerative, developmental, lysosomal storage, and metabolic disorders. The field reached an unprecedented milestone when Zolgensma® (onasemnogene abeparvovec) was approved by the FDA and EMA for in vivo adeno-associated virus-mediated gene replacement therapy for spinal muscular atrophy. Shortly after EMA approved Libmeldy®, an ex vivo gene therapy with lentivirus vector-transduced autologous CD34-positive stem cells, for treatment of metachromatic leukodystrophy. These successes could be the first of many more new gene therapies in development that mostly target loss-of-function mutation diseases with gene replacement (e.g., Batten disease, mucopolysaccharidoses, gangliosidoses) or, less frequently, gain-of-toxic-function mutation diseases by gene therapeutic silencing of pathologic genes (e.g., amyotrophic lateral sclerosis, Huntington's disease). In addition, the use of genome editing as a gene therapy is being explored for some diseases, but this has so far only reached clinical testing in the treatment of mucopolysaccharidoses. Based on the large number of planned, ongoing, and completed clinical trials for rare genetic central nervous system diseases, it can be expected that several novel gene therapies will be approved and become available within the near future. Essential for this to happen is the in depth characterization of short- and long-term effects, safety aspects, and pharmacodynamics of the applied gene therapy platforms.
Collapse
Affiliation(s)
- Thomas Leth Jensen
- Department of Neurology, Rigshospitalet University Hospital, Copenhagen, Denmark
| | | | | |
Collapse
|
17
|
Huang W, Cheng YS, Yang S, Swaroop M, Xu M, Huang W, Zheng W. Disease modeling for Mucopolysaccharidosis type IIIB using patient derived induced pluripotent stem cells. Exp Cell Res 2021; 407:112785. [PMID: 34411609 DOI: 10.1016/j.yexcr.2021.112785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/28/2021] [Accepted: 08/13/2021] [Indexed: 12/22/2022]
Abstract
Mucopolysaccharidosis type IIIB (MPS IIIB) is a lysosomal disease caused by mutations in the NAGLU gene encoding α-N-acetylglucosaminidase (NAGLU) which degrades heparan sulfate in lysosomes. Deficiency in NAGLU results in lysosomal accumulation of glycosaminoglycans (GAGs) and neurological symptoms. Currently, there is no effective treatment or cure for this disease. In this study, induced pluripotent stem cell lines were established from two MPS IIIB patient fibroblast lines and differentiated into neural stem cells and neurons. MPS IIIB neural stem cells exhibited NAGLU deficiency accompanied with GAG accumulation, as well as lysosomal enlargement and secondary lipid accumulation. Treatments with recombinant NAGLU, δ-tocopherol, and 2-hydroxypropyl-b-cyclodextrin significantly reduced the disease phenotypes in these cells. These results indicate the MPS IIIB neural stem cells and neurons have the disease relevant phenotype and can be used as a cell-based disease model system for evaluation of drug efficacy and compound screening for drug development.
Collapse
Affiliation(s)
- Wei Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA; Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Yu-Shan Cheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Shu Yang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Manju Swaroop
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Wenwei Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
18
|
De Pasquale V, Scerra G, Scarcella M, D'Agostino M, Pavone LM. Competitive binding of extracellular accumulated heparan sulfate reduces lysosomal storage defects and triggers neuronal differentiation in a model of Mucopolysaccharidosis IIIB. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119113. [PMID: 34329663 DOI: 10.1016/j.bbamcr.2021.119113] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 12/26/2022]
Abstract
Mucopolysaccharidoses (MPSs) are a group of inherited lysosomal storage disorders associated with the deficiency of lysosomal enzymes involved in glycosaminoglycan (GAG) degradation. The resulting cellular accumulation of GAGs is responsible for widespread tissue and organ dysfunctions. The MPS III, caused by mutations in the genes responsible for the degradation of heparan sulfate (HS), includes four subtypes (A, B, C, and D) that present significant neurological manifestations such as progressive cognitive decline and behavioral disorders. The established treatments for the MPS III do not cure the disease but only ameliorate non-neurological clinical symptoms. We previously demonstrated that the natural variant of the hepatocyte growth factor NK1 reduces the lysosomal pathology and reactivates impaired growth factor signaling in fibroblasts from MPS IIIB patients. Here, we show that the recombinant NK1 is effective in rescuing the morphological and functional dysfunctions of lysosomes in a neuronal cellular model of the MPS IIIB. More importantly, NK1 treatment is able to stimulate neuronal differentiation of neuroblastoma SK-NBE cells stable silenced for the NAGLU gene causative of the MPS IIIB. These results provide the basis for the development of a novel approach to possibly correct the neurological phenotypes of the MPS IIIB as well as of other MPSs characterized by the accumulation of HS and progressive neurodegeneration.
Collapse
Affiliation(s)
- Valeria De Pasquale
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via F. Delpino 1, 80127 Naples, Italy
| | - Gianluca Scerra
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Melania Scarcella
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Massimo D'Agostino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
19
|
He X, Urip BA, Zhang Z, Ngan CC, Feng B. Evolving AAV-delivered therapeutics towards ultimate cures. J Mol Med (Berl) 2021; 99:593-617. [PMID: 33594520 PMCID: PMC7885987 DOI: 10.1007/s00109-020-02034-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022]
Abstract
Gene therapy has entered a new era after decades-long efforts, where the recombinant adeno-associated virus (AAV) has stood out as the most potent vector for in vivo gene transfer and demonstrated excellent efficacy and safety profiles in numerous preclinical and clinical studies. Since the first AAV-derived therapeutics Glybera was approved by the European Medicines Agency (EMA) in 2012, there is an increasing number of AAV-based gene augmentation therapies that have been developed and tested for treating incurable genetic diseases. In the subsequent years, the United States Food and Drug Administration (FDA) approved two additional AAV gene therapy products, Luxturna and Zolgensma, to be launched into the market. Recent breakthroughs in genome editing tools and the combined use with AAV vectors have introduced new therapeutic modalities using somatic gene editing strategies. The promising outcomes from preclinical studies have prompted the continuous evolution of AAV-delivered therapeutics and broadened the scope of treatment options for untreatable diseases. Here, we describe the clinical updates of AAV gene therapies and the latest development using AAV to deliver the CRISPR components as gene editing therapeutics. We also discuss the major challenges and safety concerns associated with AAV delivery and CRISPR therapeutics, and highlight the recent achievement and toxicity issues reported from clinical applications.
Collapse
Affiliation(s)
- Xiangjun He
- School of Biomedical Sciences, Faculty of Medicine; Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Brian Anugerah Urip
- School of Biomedical Sciences, Faculty of Medicine; Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Zhenjie Zhang
- School of Biomedical Sciences, Faculty of Medicine; Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Chun Christopher Ngan
- School of Biomedical Sciences, Faculty of Medicine; Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Shatin N.T., Hong Kong SAR, China
| | - Bo Feng
- School of Biomedical Sciences, Faculty of Medicine; Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Shatin N.T., Hong Kong SAR, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510320, China.
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
20
|
Schuchman EH, Ledesma MD, Simonaro CM. New paradigms for the treatment of lysosomal storage diseases: targeting the endocannabinoid system as a therapeutic strategy. Orphanet J Rare Dis 2021; 16:151. [PMID: 33766102 PMCID: PMC7992818 DOI: 10.1186/s13023-021-01779-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/16/2021] [Indexed: 01/10/2023] Open
Abstract
Over the past three decades the lysosomal storage diseases have served as model for rare disease treatment development. While these efforts have led to considerable success, important challenges remain. For example, no treatments are currently approved for nearly two thirds of all lysosomal diseases, and there is limited impact of the existing drugs on the central nervous system. In addition, the costs of these therapies are extremely high, in part due to the fact that drug development has focused on a "single hit" approach - i.e., one drug for one disease. To overcome these obstacles researchers have begun to focus on defining common disease mechanisms in the lysosomal diseases, particularly in the central nervous system, with the hope of identifying drugs that might be used in several lysosomal diseases rather than an individual disease. With this concept in mind, herein we review a new potential treatment approach for the lysosomal storage diseases that focuses on modulation of the endocannabinoid system. We provide a short introduction to lysosomal storage diseases and the endocannabinoid system, followed by a brief review of data supporting this concept.
Collapse
Affiliation(s)
- Edward H Schuchman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine At Mount Sinai, 1425 Madison Avenue, Room 14-20A, New York, NY, 10029, USA.
| | - Maria D Ledesma
- Centro Biologia Molecular Severo Ochoa, 28049, Madrid, Spain
| | - Calogera M Simonaro
- Department of Genetics and Genomic Sciences, Icahn School of Medicine At Mount Sinai, 1425 Madison Avenue, Room 14-20A, New York, NY, 10029, USA
| |
Collapse
|
21
|
Kong W, Yao Y, Zhang J, Lu C, Ding Y, Meng Y. Update of treatment for mucopolysaccharidosis type III (sanfilippo syndrome). Eur J Pharmacol 2020; 888:173562. [DOI: 10.1016/j.ejphar.2020.173562] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 12/26/2022]
|