51
|
Horgan C, Watts K, Ram D, Rust S, Hutton R, Jones S, Wynn R. A retrospective cohort study of Libmeldy (atidarsagene autotemcel) for MLD: What we have accomplished and what opportunities lie ahead. JIMD Rep 2023; 64:346-352. [PMID: 37701322 PMCID: PMC10494509 DOI: 10.1002/jmd2.12378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 09/14/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) results from ARSA gene mutations. Affected individuals meet early milestones before neurological deterioration and early death. Atidarsagene autotemcel (arsa-cel), an autologous haematopoietic stem cell gene therapy (HSC-GT) product, has demonstrated sustained clinical benefits in MLD. Arsa-cel was approved for NHS treatment in February 2022 for asymptomatic late infantile or early juvenile disease, or early symptomatic early juvenile MLD. We evaluate the impact of this approval in the largest real-world dataset of MLD HSC-GT. Hospital records were reviewed for all patients referred for NHS treatment following arsa-cel approval. Information was gathered about disease phenotype, presentation, eligibility, and affected siblings. In the year following NHS approval, 17 UK MLD patients were referred for treatment. Four patients met eligibility criteria and have been treated, including 1 infant who weighed 5 kg at leukapheresis. Eleven patients failed screening: 10 symptomatic patients with late infantile disease and 1 with early juvenile disease and cognitive decline. Two further patients with later onset subtypes did not meet the approval criteria. Three out of four treated patients were diagnosed by screening after MLD was diagnosed in a symptomatic older sibling. The success of HSC-GT for MLD has heralded a new era of hope for families affected by this devastating disease, yet currently, most patients are ineligible for treatment at diagnosis. The feasibility of apheresis in infants and the availability of a licenced, effective HSC-GT product highlights the urgent need for newborn screening to ensure that patients can be diagnosed and treated before symptom onset.
Collapse
Affiliation(s)
- Claire Horgan
- Royal Manchester Children's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Kelly Watts
- Royal Manchester Children's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Dipak Ram
- Royal Manchester Children's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Stewart Rust
- Royal Manchester Children's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Rebekah Hutton
- Royal Manchester Children's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Simon Jones
- Royal Manchester Children's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Rob Wynn
- Royal Manchester Children's HospitalManchester University NHS Foundation TrustManchesterUK
| |
Collapse
|
52
|
Mishra P, Silva A, Sharma J, Nguyen J, Pizzo DP, Hinz D, Sahoo D, Cherqui S. Rescue of Alzheimer's disease phenotype in a mouse model by transplantation of wild-type hematopoietic stem and progenitor cells. Cell Rep 2023; 42:112956. [PMID: 37561625 PMCID: PMC10617121 DOI: 10.1016/j.celrep.2023.112956] [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: 11/03/2022] [Revised: 05/19/2023] [Accepted: 07/22/2023] [Indexed: 08/12/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia; microglia have been implicated in AD pathogenesis, but their role is still matter of debate. Our study showed that single systemic wild-type (WT) hematopoietic stem and progenitor cell (HSPC) transplantation rescued the AD phenotype in 5xFAD mice and that transplantation may prevent microglia activation. Indeed, complete prevention of memory loss and neurocognitive impairment and decrease of β-amyloid plaques in the hippocampus and cortex were observed in the WT HSPC-transplanted 5xFAD mice compared with untreated 5xFAD mice and with mice transplanted with 5xFAD HSPCs. Neuroinflammation was also significantly reduced. Transcriptomic analysis revealed a significant decrease in gene expression related to "disease-associated microglia" in the cortex and "neurodegeneration-associated endothelial cells" in the hippocampus of the WT HSPC-transplanted 5xFAD mice compared with diseased controls. This work shows that HSPC transplant has the potential to prevent AD-associated complications and represents a promising therapeutic avenue for this disease.
Collapse
Affiliation(s)
- Priyanka Mishra
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Alexander Silva
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Jay Sharma
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Jacqueline Nguyen
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Donald P Pizzo
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Denise Hinz
- Flow Cytometry Core Facility, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Debashis Sahoo
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Department of Computer Science and Engineering, University of California, La Jolla, La Jolla, CA, USA; Moores Comprehensive Cancer Center, University of California, La Jolla, La Jolla, CA, USA
| | - Stephanie Cherqui
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
53
|
Wang Y, Shao W. Innate Immune Response to Viral Vectors in Gene Therapy. Viruses 2023; 15:1801. [PMID: 37766208 PMCID: PMC10536768 DOI: 10.3390/v15091801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Viral vectors play a pivotal role in the field of gene therapy, with several related drugs having already gained clinical approval from the EMA and FDA. However, numerous viral gene therapy vectors are currently undergoing pre-clinical research or participating in clinical trials. Despite advancements, the innate response remains a significant barrier impeding the clinical development of viral gene therapy. The innate immune response to viral gene therapy vectors and transgenes is still an important reason hindering its clinical development. Extensive studies have demonstrated that different DNA and RNA sensors can detect adenoviruses, adeno-associated viruses, and lentiviruses, thereby activating various innate immune pathways such as Toll-like receptor (TLR), cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING), and retinoic acid-inducible gene I-mitochondrial antiviral signaling protein (RLR-MAVS). This review focuses on elucidating the mechanisms underlying the innate immune response induced by three widely utilized viral vectors: adenovirus, adeno-associated virus, and lentivirus, as well as the strategies employed to circumvent innate immunity.
Collapse
Affiliation(s)
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China;
| |
Collapse
|
54
|
Trinidad M, Hong X, Froelich S, Daiker J, Sacco J, Nguyen HP, Campagna M, Suhr D, Suhr T, LeBowitz JH, Gelb MH, Clark WT. Predicting disease severity in metachromatic leukodystrophy using protein activity and a patient phenotype matrix. Genome Biol 2023; 24:172. [PMID: 37480112 PMCID: PMC10360315 DOI: 10.1186/s13059-023-03001-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 06/29/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Metachromatic leukodystrophy (MLD) is a lysosomal storage disorder caused by mutations in the arylsulfatase A gene (ARSA) and categorized into three subtypes according to age of onset. The functional effect of most ARSA mutants remains unknown; better understanding of the genotype-phenotype relationship is required to support newborn screening (NBS) and guide treatment. RESULTS We collected a patient data set from the literature that relates disease severity to ARSA genotype in 489 individuals with MLD. Patient-based data were used to develop a phenotype matrix that predicts MLD phenotype given ARSA alleles in a patient's genotype with 76% accuracy. We then employed a high-throughput enzyme activity assay using mass spectrometry to explore the function of ARSA variants from the curated patient data set and the Genome Aggregation Database (gnomAD). We observed evidence that 36% of variants of unknown significance (VUS) in ARSA may be pathogenic. By classifying functional effects for 251 VUS from gnomAD, we reduced the incidence of genotypes of unknown significance (GUS) by over 98.5% in the overall population. CONCLUSIONS These results provide an additional tool for clinicians to anticipate the disease course in MLD patients, identifying individuals at high risk of severe disease to support treatment access. Our results suggest that more than 1 in 3 VUS in ARSA may be pathogenic. We show that combining genetic and biochemical information increases diagnostic yield. Our strategy may apply to other recessive diseases, providing a tool to address the challenge of interpreting VUS within genotype-phenotype relationships and NBS.
Collapse
Affiliation(s)
- Marena Trinidad
- Translational Genomics Group, BioMarin Pharmaceutical Inc., Novato, CA, USA
| | - Xinying Hong
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Steven Froelich
- Translational Genomics Group, BioMarin Pharmaceutical Inc., Novato, CA, USA
| | - Jessica Daiker
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - James Sacco
- Translational Genomics Group, BioMarin Pharmaceutical Inc., Novato, CA, USA
| | - Hong Phuc Nguyen
- Translational Genomics Group, BioMarin Pharmaceutical Inc., Novato, CA, USA
| | - Madelynn Campagna
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | | | | | - Michael H Gelb
- Department of Chemistry, University of Washington, Seattle, WA, USA.
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
| | - Wyatt T Clark
- Translational Genomics Group, BioMarin Pharmaceutical Inc., Novato, CA, USA.
| |
Collapse
|
55
|
Martin S, Harris N, Romanus D. Evaluating meaningful changes in physical functioning and cognitive declines in metachromatic leukodystrophy: a caregiver interview study. J Patient Rep Outcomes 2023; 7:70. [PMID: 37458805 DOI: 10.1186/s41687-023-00595-7] [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: 10/10/2022] [Accepted: 05/23/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Metachromatic leukodystrophy (MLD) is a rare lysosomal storage disease caused by deficient activity of arylsulfatase A (ASA). Treatment options for patients are limited; gene therapy based on haematopoietic stem cell transplantation is the only approved treatment for some subtypes of MLD. Any therapeutic benefit of treatments must be meaningful for patients and their families. We evaluated the clinical meaningfulness of slowing the decline in gross motor function as measured by the Gross Motor Function Classification in MLD (GMFC-MLD) from the caregiver perspective via semi-structured telephone interviews with caregivers of children with late-infantile MLD. We also evaluated the perceived significance of declines in communication abilities measured by the Expressive Language Function Classification in MLD (ELFC-MLD). This work could help to inform the endpoints of a phase 2 clinical trial (NCT03771898) assessing the efficacy of intrathecal recombinant human ASA in MLD. RESULTS Twelve caregivers were recruited, reporting on 12 children with MLD. Children had a mean age of 6.1 years; mean age at symptom onset was 17.6 months. Most children (10/12) progressed from walking without support (categories 0-1) to a loss of locomotion (categories 5-6) in ≤ 2 years. Caregivers felt that GMFC-MLD and ELFC-MLD accurately described motor and language declines in their children, respectively. Most caregivers (10/12) reported that the idea of delaying disease progression would be meaningful. Further, a slowing of motor function decline in GMFC-MLD, from category 1 to category 3 or from category 2 to category 4 over 2 years, was seen as meaningful by all caregivers asked; however, only 3/12 caregivers reported that delayed decline would be meaningful if baseline category was ≥ 3. Caregivers also reported that delaying expressive language decline at any level that did not indicate a complete loss of expressive language (indicated by categories 1-3) would be meaningful. CONCLUSIONS Caregivers of children with MLD felt that a delayed decline in gross motor function, as assessed by the GMFC-MLD, would be meaningful, supporting the selection of primary and secondary endpoints for the phase 2 clinical trial. Communication abilities were another area of significance for consideration in future clinical trial design.
Collapse
Affiliation(s)
| | | | - Dorothy Romanus
- Takeda Development Center Americas, Inc., Lexington, MA, USA
| |
Collapse
|
56
|
Colella P, Meneghini V, Baldo G, Gomez-Ospina N. Editorial: Ex-vivo and in-vivo genome engineering for metabolic and neurometabolic diseases. Front Genome Ed 2023; 5:1248904. [PMID: 37484653 PMCID: PMC10359423 DOI: 10.3389/fgeed.2023.1248904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/25/2023] Open
Affiliation(s)
- Pasqualina Colella
- Department of Pediatrics, Stanford University, Stanford, CA, United States
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Guilherme Baldo
- Clinical Hospital of Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | | |
Collapse
|
57
|
Al‐Saady M, Beerepoot S, Plug BC, Breur M, Galabova H, Pouwels PJW, Boelens J, Lindemans C, van Hasselt PM, Matzner U, Vanderver A, Bugiani M, van der Knaap MS, Wolf NI. Neurodegenerative disease after hematopoietic stem cell transplantation in metachromatic leukodystrophy. Ann Clin Transl Neurol 2023; 10:1146-1159. [PMID: 37212343 PMCID: PMC10351661 DOI: 10.1002/acn3.51796] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/23/2023] Open
Abstract
OBJECTIVE Metachromatic leukodystrophy is a lysosomal storage disease caused by deficient arylsulfatase A. It is characterized by progressive demyelination and thus mainly affects the white matter. Hematopoietic stem cell transplantation may stabilize and improve white matter damage, yet some patients deteriorate despite successfully treated leukodystrophy. We hypothesized that post-treatment decline in metachromatic leukodystrophy might be caused by gray matter pathology. METHODS Three metachromatic leukodystrophy patients treated with hematopoietic stem cell transplantation with a progressive clinical course despite stable white matter pathology were clinically and radiologically analyzed. Longitudinal volumetric MRI was used to quantify atrophy. We also examined histopathology in three other patients deceased after treatment and compared them with six untreated patients. RESULTS The three clinically progressive patients developed cognitive and motor deterioration after transplantation, despite stable mild white matter abnormalities on MRI. Volumetric MRI identified cerebral and thalamus atrophy in these patients, and cerebellar atrophy in two. Histopathology showed that in brain tissue of transplanted patients, arylsulfatase A expressing macrophages were clearly present in the white matter, but absent in the cortex. Arylsulfatase A expression within patient thalamic neurons was lower than in controls, the same was found in transplanted patients. INTERPRETATION Neurological deterioration may occur after hematopoietic stem cell transplantation in metachromatic leukodystrophy despite successfully treated leukodystrophy. MRI shows gray matter atrophy, and histological data demonstrate absence of donor cells in gray matter structures. These findings point to a clinically relevant gray matter component of metachromatic leukodystrophy, which does not seem sufficiently affected by transplantation.
Collapse
Affiliation(s)
- Murtadha Al‐Saady
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
| | - Shanice Beerepoot
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
- Center for Translational ImmunologyUniversity Medical Center UtrechtUtrechtthe Netherlands
- Nierkens and Lindemans GroupPrincess Máxima Center for Pediatric OncologyUtrechtthe Netherlands
| | - Bonnie C. Plug
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
- Department of Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical CentersVU University and Neuroscience Campus AmsterdamAmsterdamthe Netherlands
| | - Marjolein Breur
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
- Department of Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical CentersVU University and Neuroscience Campus AmsterdamAmsterdamthe Netherlands
| | - Hristina Galabova
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, Amsterdam University Medical CentersVU universityAmsterdamthe Netherlands
| | - Petra J. W. Pouwels
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, Amsterdam University Medical CentersVU universityAmsterdamthe Netherlands
| | - Jaap‐Jan Boelens
- Stem Cell Transplantation and Cellular Therapies Program, Department of PediatricsMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Caroline Lindemans
- Stem Cell Transplantation and Cellular Therapies Program, Department of PediatricsMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
- Pediatric Blood and Bone Marrow Transplantation, Princess Máxima Center for Pediatric OncologyUtrechtthe Netherlands
| | - Peter M. van Hasselt
- Stem Cell Transplantation and Cellular Therapies Program, Department of PediatricsMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Ulrich Matzner
- Institute of Biochemistry and Molecular Biology, Medical FacultyRheinische Friedrich‐Wilhelm UniversityBonnGermany
| | - Adeline Vanderver
- Division of Neurology, Department of Pediatrics, Children's Hospital of PhiladelphiaUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Marianna Bugiani
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
- Department of Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical CentersVU University and Neuroscience Campus AmsterdamAmsterdamthe Netherlands
| | - Marjo S. van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
- Department of Integrative NeurophysiologyVU UniversityAmsterdamthe Netherlands
| | - Nicole I. Wolf
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
| |
Collapse
|
58
|
Mullagulova A, Shaimardanova A, Solovyeva V, Mukhamedshina Y, Chulpanova D, Kostennikov A, Issa S, Rizvanov A. Safety and Efficacy of Intravenous and Intrathecal Delivery of AAV9-Mediated ARSA in Minipigs. Int J Mol Sci 2023; 24:ijms24119204. [PMID: 37298156 DOI: 10.3390/ijms24119204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a hereditary neurodegenerative disease characterized by demyelination and motor and cognitive impairments due to deficiencies of the lysosomal enzyme arylsulfatase A (ARSA) or the saposin B activator protein (SapB). Current treatments are limited; however, gene therapy using adeno-associated virus (AAV) vectors for ARSA delivery has shown promising results. The main challenges for MLD gene therapy include optimizing the AAV dosage, selecting the most effective serotype, and determining the best route of administration for ARSA delivery into the central nervous system. This study aims to evaluate the safety and efficacy of AAV serotype 9 encoding ARSA (AAV9-ARSA) gene therapy when administered intravenously or intrathecally in minipigs, a large animal model with anatomical and physiological similarities to humans. By comparing these two administration methods, this study contributes to the understanding of how to improve the effectiveness of MLD gene therapy and offers valuable insights for future clinical applications.
Collapse
Affiliation(s)
- Aysilu Mullagulova
- Institute for Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Alisa Shaimardanova
- Institute for Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Valeriya Solovyeva
- Institute for Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Yana Mukhamedshina
- Institute for Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Department of Histology, Cytology, and Embryology, Kazan State Medical University, 420012 Kazan, Russia
| | - Daria Chulpanova
- Institute for Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Alexander Kostennikov
- Institute for Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Shaza Issa
- Institute for Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Albert Rizvanov
- Institute for Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
59
|
Singh J, Goodman-Vincent E, Santosh P. Evidence Synthesis of Gene Therapy and Gene Editing from Different Disorders-Implications for Individuals with Rett Syndrome: A Systematic Review. Int J Mol Sci 2023; 24:ijms24109023. [PMID: 37240368 DOI: 10.3390/ijms24109023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/06/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
This systematic review and thematic analysis critically evaluated gene therapy trials in amyotrophic lateral sclerosis, haemoglobinopathies, immunodeficiencies, leukodystrophies, lysosomal storage disorders and retinal dystrophies and extrapolated the key clinical findings to individuals with Rett syndrome (RTT). The PRISMA guidelines were used to search six databases during the last decade, followed by a thematic analysis to identify the emerging themes. Thematic analysis across the different disorders revealed four themes: (I) Therapeutic time window of gene therapy; (II) Administration and dosing strategies for gene therapy; (III) Methods of gene therapeutics and (IV) Future areas of clinical interest. Our synthesis of information has further enriched the current clinical evidence base and can assist in optimising gene therapy and gene editing studies in individuals with RTT, but it would also benefit when applied to other disorders. The findings suggest that gene therapies have better outcomes when the brain is not the primary target. Across different disorders, early intervention appears to be more critical, and targeting the pre-symptomatic stage might prevent symptom pathology. Intervention at later stages of disease progression may benefit by helping to clinically stabilise patients and preventing disease-related symptoms from worsening. If gene therapy or editing has the desired outcome, older patients would need concerted rehabilitation efforts to reverse their impairments. The timing of intervention and the administration route would be critical parameters for successful outcomes of gene therapy/editing trials in individuals with RTT. Current approaches also need to overcome the challenges of MeCP2 dosing, genotoxicity, transduction efficiencies and biodistribution.
Collapse
Affiliation(s)
- Jatinder Singh
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London and South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
| | - Ella Goodman-Vincent
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London and South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
| | - Paramala Santosh
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London and South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
| |
Collapse
|
60
|
Ferrari S, Valeri E, Conti A, Scala S, Aprile A, Di Micco R, Kajaste-Rudnitski A, Montini E, Ferrari G, Aiuti A, Naldini L. Genetic engineering meets hematopoietic stem cell biology for next-generation gene therapy. Cell Stem Cell 2023; 30:549-570. [PMID: 37146580 DOI: 10.1016/j.stem.2023.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 05/07/2023]
Abstract
The growing clinical success of hematopoietic stem/progenitor cell (HSPC) gene therapy (GT) relies on the development of viral vectors as portable "Trojan horses" for safe and efficient gene transfer. The recent advent of novel technologies enabling site-specific gene editing is broadening the scope and means of GT, paving the way to more precise genetic engineering and expanding the spectrum of diseases amenable to HSPC-GT. Here, we provide an overview of state-of-the-art and prospective developments of the HSPC-GT field, highlighting how advances in biological characterization and manipulation of HSPCs will enable the design of the next generation of these transforming therapeutics.
Collapse
Affiliation(s)
- Samuele Ferrari
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Erika Valeri
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Anastasia Conti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Serena Scala
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Annamaria Aprile
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Raffaella Di Micco
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Giuliana Ferrari
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy.
| |
Collapse
|
61
|
Peviani M, Das S, Patel J, Jno‐Charles O, Kumar R, Zguro A, Mathews TD, Cabras P, Milazzo R, Cavalca E, Poletti V, Biffi A. An innovative hematopoietic stem cell gene therapy approach benefits CLN1 disease in the mouse model. EMBO Mol Med 2023; 15:e15968. [PMID: 36876653 PMCID: PMC10086581 DOI: 10.15252/emmm.202215968] [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: 03/04/2022] [Revised: 01/25/2023] [Accepted: 02/09/2023] [Indexed: 03/07/2023] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) can establish a long-lasting microglia-like progeny in the central nervous system of properly myeloablated hosts. We exploited this approach to treat the severe CLN1 neurodegenerative disorder, which is the most aggressive form of neuronal ceroid lipofuscinoses due to palmitoyl-protein thioesterase-1 (PPT1) deficiency. We here provide the first evidence that (i) transplantation of wild-type HSPCs exerts partial but long-lasting mitigation of CLN1 symptoms; (ii) transplantation of HSPCs over-expressing hPPT1 by lentiviral gene transfer enhances the therapeutic benefit of HSPCs transplant, with first demonstration of such a dose-effect benefit for a purely neurodegenerative condition like CLN1 disease; (iii) transplantation of hPPT1 over-expressing HSPCs by a novel intracerebroventricular (ICV) approach is sufficient to transiently ameliorate CLN1-symptoms in the absence of hematopoietic tissue engraftment of the transduced cells; and (iv) combinatorial transplantation of transduced HSPCs intravenously and ICV results in a robust therapeutic benefit, particularly on symptomatic animals. Overall, these findings provide first evidence of efficacy and feasibility of this novel approach to treat CLN1 disease and possibly other neurodegenerative conditions, paving the way for its future clinical application.
Collapse
Affiliation(s)
- Marco Peviani
- Gene Therapy ProgramDana‐Farber/Boston Children's Cancer and Blood Disorders CenterBostonMAUSA
- Harvard Medical SchoolBostonMAUSA
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), San Raffaele Scientific InstituteMilanItaly
- Department of Biology and Biotechnology “L. Spallanzani”University of PaviaPaviaItaly
| | - Sabyasachi Das
- Gene Therapy ProgramDana‐Farber/Boston Children's Cancer and Blood Disorders CenterBostonMAUSA
| | - Janki Patel
- Gene Therapy ProgramDana‐Farber/Boston Children's Cancer and Blood Disorders CenterBostonMAUSA
| | - Odella Jno‐Charles
- Gene Therapy ProgramDana‐Farber/Boston Children's Cancer and Blood Disorders CenterBostonMAUSA
| | - Rajesh Kumar
- Gene Therapy ProgramDana‐Farber/Boston Children's Cancer and Blood Disorders CenterBostonMAUSA
| | - Ana Zguro
- Gene Therapy ProgramDana‐Farber/Boston Children's Cancer and Blood Disorders CenterBostonMAUSA
| | - Tyler D Mathews
- Gene Therapy ProgramDana‐Farber/Boston Children's Cancer and Blood Disorders CenterBostonMAUSA
| | - Paolo Cabras
- Department of Biology and Biotechnology “L. Spallanzani”University of PaviaPaviaItaly
| | - Rita Milazzo
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), San Raffaele Scientific InstituteMilanItaly
| | - Eleonora Cavalca
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), San Raffaele Scientific InstituteMilanItaly
| | - Valentina Poletti
- Gene Therapy ProgramDana‐Farber/Boston Children's Cancer and Blood Disorders CenterBostonMAUSA
- Harvard Medical SchoolBostonMAUSA
| | - Alessandra Biffi
- Gene Therapy ProgramDana‐Farber/Boston Children's Cancer and Blood Disorders CenterBostonMAUSA
- Harvard Medical SchoolBostonMAUSA
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Woman's and Child Health DepartmentUniversity of PadovaPadovaItaly
| |
Collapse
|
62
|
Faccioli S, Sassi S, Pandarese D, Borghi C, Montemaggiori V, Sarzana M, Scarparo S, Butera C, Calbi V, Aiuti A, Fumagalli F. Preserving Ambulation in a Gene Therapy-Treated Girl Affected by Metachromatic Leukodystrophy: A Case Report. J Pers Med 2023; 13:jpm13040637. [PMID: 37109023 PMCID: PMC10144348 DOI: 10.3390/jpm13040637] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
(1) Background: Atidarsagene autotemcel is a hematopoietic stem and progenitor cell gene therapy (HSPC-GT) approved to treat early-onset metachromatic leukodystrophy (MLD). The purpose of this case report is to describe the long-term management of residual gait impairment of a child with late infantile MLD treated with HSPC-GT. (2) Methods: Assessment included Gross Motor Function Measure-88, nerve conduction study, body mass index (BMI), Modified Tardieu Scale, passive range of motion, modified Medical Research Council scale, and gait analysis. Interventions included orthoses, a walker, orthopedic surgery, physiotherapy, and botulinum. (3) Results: Orthoses and a walker were fundamental to maintaining ambulation. Orthopedic surgery positively influenced gait by reducing equinovarus. Nonetheless, unilateral recurrence of varo-supination was observed, attributable to spasticity and muscle imbalance. Botulinum improved foot alignment but induced transient overall weakness. A significant increase in BMI occurred. Finally, a shift to bilateral valgopronation was observed, more easily managed with orthoses. (4) Conclusions: HSPC-GT preserved survival and locomotor abilities. Rehabilitation was then considered fundamental as a complementary treatment. Muscle imbalance and increased BMI contributed to gait deterioration in the growing phase. Caution is recommended when considering botulinum in similar subjects, as the risk of inducing overall weakness can outweigh the benefits of spasticity reduction.
Collapse
Affiliation(s)
- Silvia Faccioli
- Children Rehabilitation Unit of S. M. Nuova Hospital, Azienda Unità Sanitaria Locale IRCCS di Reggio Emilia, 42121 Reggio Emilia, Italy
- PhD Program in Clinical and Experimental Medicine, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Silvia Sassi
- Children Rehabilitation Unit of S. M. Nuova Hospital, Azienda Unità Sanitaria Locale IRCCS di Reggio Emilia, 42121 Reggio Emilia, Italy
| | - Daniela Pandarese
- Children Rehabilitation Unit of S. M. Nuova Hospital, Azienda Unità Sanitaria Locale IRCCS di Reggio Emilia, 42121 Reggio Emilia, Italy
| | - Corrado Borghi
- Children Rehabilitation Unit of S. M. Nuova Hospital, Azienda Unità Sanitaria Locale IRCCS di Reggio Emilia, 42121 Reggio Emilia, Italy
| | - Valentina Montemaggiori
- Orthopaedic Unit of S. M. Nuova Hospital, Azienda Unità Sanitaria Locale IRCCS di Reggio Emilia, 42121 Reggio Emilia, Italy
| | - Marina Sarzana
- Pediatric Immunohematology Unit, IRCCS San Raffaele Scientific Institute, 20019 Milan, Italy
| | - Stefano Scarparo
- Pediatric Immunohematology Unit, IRCCS San Raffaele Scientific Institute, 20019 Milan, Italy
| | - Carla Butera
- Units of Neurology and Neurophysiology, IRCCS San Raffaele Scientific Institute, 20019 Milan, Italy
| | - Valeria Calbi
- Pediatric Immunohematology Unit, IRCCS San Raffaele Scientific Institute, 20019 Milan, Italy
| | - Alessandro Aiuti
- Pediatric Immunohematology Unit, IRCCS San Raffaele Scientific Institute, 20019 Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20019 Milan, Italy
- Vita-Salute San Raffaele University, 20019 Milan, Italy
| | - Francesca Fumagalli
- Pediatric Immunohematology Unit, IRCCS San Raffaele Scientific Institute, 20019 Milan, Italy
- Units of Neurology and Neurophysiology, IRCCS San Raffaele Scientific Institute, 20019 Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20019 Milan, Italy
| |
Collapse
|
63
|
Poletto E, Silva AO, Weinlich R, Martin PKM, Torres DC, Giugliani R, Baldo G. Ex vivo gene therapy for lysosomal storage disorders: future perspectives. Expert Opin Biol Ther 2023; 23:353-364. [PMID: 36920351 DOI: 10.1080/14712598.2023.2192348] [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] [Indexed: 03/16/2023]
Abstract
INTRODUCTION Lysosomal storage disorders (LSD) are a group of monogenic rare diseases caused by pathogenic variants in genes that encode proteins related to lysosomal function. These disorders are good candidates for gene therapy for different reasons: they are monogenic, most of lysosomal proteins are enzymes that can be secreted and cross-correct neighboring cells, and small quantities of these proteins are able to produce clinical benefits in many cases. Ex vivo gene therapy allows for autologous transplant of modified cells from different sources, including stem cells and hematopoietic precursors. AREAS COVERED Here, we summarize the main gene therapy and genome editing strategies that are currently being used as ex vivo gene therapy approaches for lysosomal disorders, highlighting important characteristics, such as vectors used, strategies, types of cells that are modified and main results in different disorders. EXPERT OPINION Clinical trials are already ongoing, and soon approved therapies for LSD based on ex vivo gene therapy approaches should reach the market.
Collapse
Affiliation(s)
- Edina Poletto
- Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto alegre, Brazil
- Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Andrew Oliveira Silva
- Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Ricardo Weinlich
- Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- Centro de Ensino e Pesquisa/Pesquisa Experimental, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Davi Coe Torres
- Centro de Ensino e Pesquisa/Pesquisa Experimental, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Roberto Giugliani
- Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto alegre, Brazil
- Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Guilherme Baldo
- Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto alegre, Brazil
- Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| |
Collapse
|
64
|
Critchley BJ, Gaspar HB, Benedetti S. Targeting the central nervous system in lysosomal storage diseases: Strategies to deliver therapeutics across the blood-brain barrier. Mol Ther 2023; 31:657-675. [PMID: 36457248 PMCID: PMC10014236 DOI: 10.1016/j.ymthe.2022.11.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/18/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are multisystem inherited metabolic disorders caused by dysfunctional lysosomal activity, resulting in the accumulation of undegraded macromolecules in a variety of organs/tissues, including the central nervous system (CNS). Treatments include enzyme replacement therapy, stem/progenitor cell transplantation, and in vivo gene therapy. However, these treatments are not fully effective in treating the CNS as neither enzymes, stem cells, nor viral vectors efficiently cross the blood-brain barrier. Here, we review the latest advancements in improving delivery of different therapeutic agents to the CNS and comment upon outstanding questions in the field of neurological LSDs.
Collapse
Affiliation(s)
- Bethan J Critchley
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK
| | - H Bobby Gaspar
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK; Orchard Therapeutics Ltd., London EC4N 6EU, UK
| | - Sara Benedetti
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
65
|
Ataei Z, Nouri Z, Tavakoli F, Pourreza MR, Narrei S, Tabatabaiefar MA. Novel in-frame duplication variant characterization in late infantile metachromatic leukodystrophy using whole-exome sequencing and molecular dynamics simulation. PLoS One 2023; 18:e0282304. [PMID: 36848337 PMCID: PMC9970088 DOI: 10.1371/journal.pone.0282304] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 02/12/2023] [Indexed: 03/01/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a neurodegenerative lysosomal storage disease caused by a deficiency in the arylsulfatase A (ARSA). ARSA deficiency leads to sulfatide accumulation, which involves progressive demyelination. The profound impact of early diagnosis on MLD treatment options necessitates the development of new or updated analysis tools and approaches. In this study, to identify the genetic etiology in a proband from a consanguineous family with MLD presentation and low ARSA activity, we employed Whole-Exome Sequencing (WES) followed by co-segregation analysis using Sanger sequencing. Also, MD simulation was utilized to study how the variant alters the structural behavior and function of the ARSA protein. GROMACS was applied and the data was analyzed by RMSD, RMSF, Rg, SASA, HB, atomic distance, PCA, and FEL. Variant interpretation was done based on the American College of Medical Genetics and Genomics (ACMG) guidelines. WES results showed a novel homozygous insertion mutation, c.109_126dup (p.Asp37_Gly42dup), in the ARSA gene. This variant is located in the first exon of ARSA, fulfilling the criteria of being categorized as likely pathogenic, according to the ACMG guidelines and it was also found to be co-segregating in the family. The MD simulation analysis revealed this mutation influenced the structure and the stabilization of ARSA and led to the protein function impairment. Here, we report a useful application of WES and MD to identify the causes of a neurometabolic disorder.
Collapse
Affiliation(s)
- Zahra Ataei
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Noncommunicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Nouri
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Noncommunicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farial Tavakoli
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | | | - Sina Narrei
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Research and Development, ERYTHROGEN Medical Genetics Lab, Isfahan, Iran
| | - Mohammad Amin Tabatabaiefar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Noncommunicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
- * E-mail: ,
| |
Collapse
|
66
|
Shaimardanova AA, Solovyeva VV, Issa SS, Rizvanov AA. Gene Therapy of Sphingolipid Metabolic Disorders. Int J Mol Sci 2023; 24:3627. [PMID: 36835039 PMCID: PMC9964151 DOI: 10.3390/ijms24043627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Sphingolipidoses are defined as a group of rare hereditary diseases resulting from mutations in the genes encoding lysosomal enzymes. This group of lysosomal storage diseases includes more than 10 genetic disorders, including GM1-gangliosidosis, Tay-Sachs disease, Sandhoff disease, the AB variant of GM2-gangliosidosis, Fabry disease, Gaucher disease, metachromatic leukodystrophy, Krabbe disease, Niemann-Pick disease, Farber disease, etc. Enzyme deficiency results in accumulation of sphingolipids in various cell types, and the nervous system is also usually affected. There are currently no known effective methods for the treatment of sphingolipidoses; however, gene therapy seems to be a promising therapeutic variant for this group of diseases. In this review, we discuss gene therapy approaches for sphingolipidoses that are currently being investigated in clinical trials, among which adeno-associated viral vector-based approaches and transplantation of hematopoietic stem cells genetically modified with lentiviral vectors seem to be the most effective.
Collapse
Affiliation(s)
- Alisa A. Shaimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Shaza S. Issa
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
67
|
Jubert C, De Berranger E, Castelle M, Dalle JH, Ouachee-Chardin M, Sevin C, Yakoub-Agha I, Brassier A. [Inborn error of metabolism and allogenic hematopoietic cell transplantation: Guidelines from the SFGM-TC]. Bull Cancer 2023; 110:S1-S12. [PMID: 36244825 DOI: 10.1016/j.bulcan.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/07/2022]
Abstract
Inherited Metabolic Diseases (IMD) are rare genetic diseases, including both lysosomal and peroxisomal diseases. Lysosomal diseases are related to the deficiency of one or more lysosomal enzymes or transporter. Lysosomal diseases are progressive and involve several tissues with most often neurological damage. Among peroxisomal diseases, X-linked adrenoleukodystrophy (ALD) is a neurodegenerative disease combining neurological and adrenal damage. For these diseases, enzyme replacement therapy (ERT), allogeneic hematopoietic cell transplantation (allo-HCT) and gene therapy represent various possible treatment options, used alone or in combination. The purpose of this workshop is to describe the indications, modalities, and follow-up of allo-HCT as well as the use of ERT peri-transplant. All indications for transplant in these rare diseases are associated with comorbidities and are subject to criteria that must be discussed in a dedicated national multidisciplinary consultation meeting. There are some consensual indications in type I-H mucopolysaccharidosis (MPS-IH) and in the cerebral form of ALD. For other IMDs, no clear benefit from the transplant has been demonstrated. The ideal donor is a non-heterozygous HLA-identical sibling. The recommended conditioning is myeloablative combining fludarabine and busulfan. In MPS-IH, ERT has to be started at diagnosis and continued until complete chimerism and normal enzyme assay are achieved. The pre-transplant assessment and post-transplant follow-up are made according to the published recommendations (PNDS). Standard follow-up is carried out jointly by the transplant and referral teams.
Collapse
Affiliation(s)
- Charlotte Jubert
- CHU de Bordeaux, groupe hospitalier Pellegrin, unité d'hématologie oncologie pédiatrique, place Améli-Raba-Léon, 33076 Bordeaux cedex, France.
| | - Eva De Berranger
- CHRU de Lille, service d'hématologie pédiatrique, avenue Eugène-Avinée, 59037 Lille, France
| | - Martin Castelle
- CHU de Necker-Enfants Malades, unité d'immuno-hématologie et rhumatologie pédiatrique, 149, rue de Sèvres, 75015 Paris, France
| | - Jean-Hugues Dalle
- Hôpital Robert-Debré, GHU Nord-Université de Paris, service d'immuno-hématologie pédiatrique, 48, boulevard Serurier, 75019 Paris, France
| | - Marie Ouachee-Chardin
- Institut d'hématologie et d'oncologie pédiatrique, 1, place Joseph-Renault, 69008 Lyon, France
| | - Caroline Sevin
- CHU de Kremlin-Bicêtre, neurologie pédiatrique, 78, rue du General-Leclerc, 94275 Le Kremlin-Bicêtre, France; ICM, 47, boulevard de l'Hôpital, 75013 Paris, France
| | - Ibrahim Yakoub-Agha
- Université de Lille, CHRU de Lille, Infinite, Inserm U1286, 59000 Lille, France
| | - Anais Brassier
- CHU de Necker, centre de référence des maladies héréditaires du métabolisme, 149, rue de Sèvres, 75015 Paris, France
| |
Collapse
|
68
|
Kido J, Sugawara K, Nakamura K. Gene therapy for lysosomal storage diseases: Current clinical trial prospects. Front Genet 2023; 14:1064924. [PMID: 36713078 PMCID: PMC9880060 DOI: 10.3389/fgene.2023.1064924] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of metabolic inborn errors caused by defective enzymes in the lysosome, resulting in the accumulation of undegraded substrates. LSDs are progressive diseases that exhibit variable rates of progression depending on the disease and the patient. The availability of effective treatment options, including substrate reduction therapy, pharmacological chaperone therapy, enzyme replacement therapy, and bone marrow transplantation, has increased survival time and improved the quality of life in many patients with LSDs. However, these therapies are not sufficiently effective, especially against central nerve system abnormalities and corresponding neurological and psychiatric symptoms because of the blood-brain barrier that prevents the entry of drugs into the brain or limiting features of specific treatments. Gene therapy is a promising tool for the treatment of neurological pathologies associated with LSDs. Here, we review the current state of gene therapy for several LSDs for which clinical trials have been conducted or are planned. Several clinical trials using gene therapy for LSDs are underway as phase 1/2 studies; no adverse events have not been reported in most of these studies. The administration of viral vectors has achieved good therapeutic outcomes in animal models of LSDs, and subsequent human clinical trials are expected to promote the practical application of gene therapy for LSDs.
Collapse
Affiliation(s)
- Jun Kido
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan,Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan,*Correspondence: Jun Kido,
| | - Keishin Sugawara
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan,Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
| |
Collapse
|
69
|
Crippa S, Conti A, Vavassori V, Ferrari S, Beretta S, Rivis S, Bosotti R, Scala S, Pirroni S, Jofra-Hernandez R, Santi L, Basso-Ricci L, Merelli I, Genovese P, Aiuti A, Naldini L, Di Micco R, Bernardo ME. Mesenchymal stromal cells improve the transplantation outcome of CRISPR-Cas9 gene-edited human HSPCs. Mol Ther 2023; 31:230-248. [PMID: 35982622 PMCID: PMC9840125 DOI: 10.1016/j.ymthe.2022.08.011] [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: 02/25/2022] [Revised: 07/29/2022] [Accepted: 08/12/2022] [Indexed: 01/26/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have been employed in vitro to support hematopoietic stem and progenitor cell (HSPC) expansion and in vivo to promote HSPC engraftment. Based on these studies, we developed an MSC-based co-culture system to optimize the transplantation outcome of clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 gene-edited (GE) human HSPCs. We show that bone marrow (BM)-MSCs produce several hematopoietic supportive and anti-inflammatory factors capable of alleviating the proliferation arrest and mitigating the apoptotic and inflammatory programs activated in GE-HSPCs, improving their expansion and clonogenic potential in vitro. The use of BM-MSCs resulted in superior human engraftment and increased clonal output of GE-HSPCs contributing to the early phase of hematological reconstitution in the peripheral blood of transplanted mice. In conclusion, our work poses the biological bases for a novel clinical use of BM-MSCs to promote engraftment of GE-HSPCs and improve their transplantation outcome.
Collapse
Affiliation(s)
- Stefania Crippa
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.
| | - Anastasia Conti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Valentina Vavassori
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Samuele Ferrari
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Stefano Beretta
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Silvia Rivis
- Laboratory of Tumor Inflammation and Angiogenesis, VIB-KULeuven, 3000 Leuven, Belgium
| | - Roberto Bosotti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Serena Scala
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Raisa Jofra-Hernandez
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ludovica Santi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Luca Basso-Ricci
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; National Research Council, Institute for Biomedical Technologies, 20132 Milan, Italy
| | - Pietro Genovese
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Department of Pediatric Oncology, Harvard Medical School, Boston, MA 02115, USA
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy; (")Vita Salute" San Raffaele University, 20132 Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (")Vita Salute" San Raffaele University, 20132 Milan, Italy
| | - Raffaella Di Micco
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Maria Ester Bernardo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy; (")Vita Salute" San Raffaele University, 20132 Milan, Italy.
| |
Collapse
|
70
|
Davison AS, Norman BP. Alkaptonuria – Past, present and future. Adv Clin Chem 2023. [DOI: 10.1016/bs.acc.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
71
|
Sharma A, Farnia S, Otegbeye F, Rinkle A, Shah J, Shah NN, Gill S, Maus MV. Nomenclature for Cellular and Genetic Therapies: A Need for Standardization. Transplant Cell Ther 2022; 28:795-801. [PMID: 36058548 PMCID: PMC9729423 DOI: 10.1016/j.jtct.2022.08.029] [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: 08/24/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 12/24/2022]
Abstract
As the field of cellular and genetic therapies transitions from a scientific concept to a clinical reality, it has become evident that there are several conflicting or imprecise nomenclatures to describe these novel therapeutic products. The lack of uniformity and accuracy in the terminology often creates regulatory, educational, administrative, and billing quagmires. Standardization of the nomenclature for these therapeutic products is essential for creation of a harmonized regulatory and developmental framework, development of training paradigms and educational programs, equitable and rational decisions about accessibility, and consistency in the billing and coding structures used for reimbursement. Here we propose an updated framework as a foundation for categorizing these cell-based and genetically modified therapies.
Collapse
Affiliation(s)
- Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee.
| | | | - Folashade Otegbeye
- Bone Marrow Transplantation and Immunotherapy Program, Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | | | | | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Saar Gill
- Cell Therapy and Transplantation program, Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Marcela V Maus
- Cellular Immunotherapy Program and Cancer Center, Massachusetts General Hospital, Charlestown, Massachusetts; Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
72
|
Olivé-Cirera G, Martínez-González MJ, Armangué T. Pearls & Oy-sters: Tumefactive Demyelinating Lesions With MOG Antibodies Preceding Late Infantile Metachromatic Leukodystrophy. Neurology 2022; 99:858-861. [PMID: 36041866 DOI: 10.1212/wnl.0000000000201230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022] Open
Abstract
The development of acute neurologic dysfunction associated with tumefactive demyelinating lesions and mild diffuse involvement of the corpus callosum has been described in children as a sentinel event that may allow diagnosis of juvenile metachromatic leukodystrophy (MLD) at an early and potentially treatable stage. We report a child who developed this clinico-radiologic pattern associated with myelin oligodendrocyte glycoprotein antibodies several months before the onset of progressive symptoms of late infantile MLD.
Collapse
Affiliation(s)
- Gemma Olivé-Cirera
- From the Neuroimmunology Program (G.O.-C., T.A.), IDIBAPS-Hospital Clínic, University of Barcelona; Pediatric Neurology Unit Hospital Parc Taulí de Sabadell (G.O.-C.); Pediatric Neurology Unit (M.J.M.-G.), Cruces University Hospital, Barakaldo; and Pediatric Neuroimmunology Unit (T.A.), Neurology Service, Sant Joan de Déu Children's Hospital, University of Barcelona, Spain
| | - Maria Jesús Martínez-González
- From the Neuroimmunology Program (G.O.-C., T.A.), IDIBAPS-Hospital Clínic, University of Barcelona; Pediatric Neurology Unit Hospital Parc Taulí de Sabadell (G.O.-C.); Pediatric Neurology Unit (M.J.M.-G.), Cruces University Hospital, Barakaldo; and Pediatric Neuroimmunology Unit (T.A.), Neurology Service, Sant Joan de Déu Children's Hospital, University of Barcelona, Spain
| | - Thaís Armangué
- From the Neuroimmunology Program (G.O.-C., T.A.), IDIBAPS-Hospital Clínic, University of Barcelona; Pediatric Neurology Unit Hospital Parc Taulí de Sabadell (G.O.-C.); Pediatric Neurology Unit (M.J.M.-G.), Cruces University Hospital, Barakaldo; and Pediatric Neuroimmunology Unit (T.A.), Neurology Service, Sant Joan de Déu Children's Hospital, University of Barcelona, Spain.
| |
Collapse
|
73
|
Schoenmakers DH, Beerepoot S, Krägeloh‐Mann I, Elgün S, Bender B, van der Knaap MS, Wolf NI, Groeschel S. Recognizing early MRI signs (or their absence) is crucial in diagnosing metachromatic leukodystrophy. Ann Clin Transl Neurol 2022; 9:1999-2009. [PMID: 36334091 PMCID: PMC9735365 DOI: 10.1002/acn3.51692] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVES Metachromatic leukodystrophy (MLD) has characteristic white matter (WM) changes on brain MRI, which often trigger biochemical and genetic confirmation of the diagnosis. In early or pre-symptomatic disease stages, these typical MRI changes might be absent, hampering early diagnosis. This study aims to describe the characteristics of MRI WM abnormalities at diagnosis, related to clinical presentation. METHODS We retrospectively reviewed brain MRIs of MLD patients followed in 2 centers at the time of diagnosis regarding MLD MRI score and presence of tigroid pattern. In addition, MLD subtype, symptom status, CNS/PNS phenotype, motor/cognitive/mixed phenotype, and the presence of CNS symptoms were evaluated. RESULTS We included 104 brain MRIs from patients with late-infantile (n = 43), early-juvenile (n = 24), late-juvenile (n = 20) and adult (n = 17) onset. Involvement of the corpus callosum was a characteristic early MRI sign and was present in 71% of the symptomatic late-infantile patients, 94% of the symptomatic early-juvenile patients and 100% of the symptomatic late-juvenile and adult patients. Symptomatic early-juvenile, late-juvenile and adult patients generally had WM abnormalities on MRI suggestive of MLD. By contrast, 47% of the early-symptomatic late-infantile patients had no or only mild WM abnormalities on MRI, even in the presence of CNS symptoms including pyramidal signs. INTERPRETATION Patients with late-infantile MLD may have no or only mild, nonspecific abnormalities at brain MRI, partly suggestive of 'delayed myelination', even with clear clinical symptoms. This may lead to significant diagnostic delay. Knowledge of these early MRI signs (or their absence) is important for fast diagnosis.
Collapse
Affiliation(s)
- Daphne H. Schoenmakers
- Department of Child Neurology, Amsterdam Leukodystrophy CenterAmsterdam UMC location Vrije Universiteit Amsterdam, Emma's Children's HospitalBoelelaan 1117AmsterdamThe Netherlands,Amsterdam Neuroscience, Cellular & Molecular MechanismsAmsterdamThe Netherlands,Department of Endocrinology and MetabolismAmsterdam UMC location University of AmsterdamMeibergdreef 9AmsterdamThe Netherlands
| | - Shanice Beerepoot
- Department of Child Neurology, Amsterdam Leukodystrophy CenterAmsterdam UMC location Vrije Universiteit Amsterdam, Emma's Children's HospitalBoelelaan 1117AmsterdamThe Netherlands,Amsterdam Neuroscience, Cellular & Molecular MechanismsAmsterdamThe Netherlands,Center for Translational ImmunologyUniversity Medical Center UtrechtUtrechtThe Netherlands,Pediatric Transplant CenterPrincess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Ingeborg Krägeloh‐Mann
- Department of Child Neurology and Developmental MedicineUniversity Children's Hospital TübingenHoppe‐Seyler‐Straße 172076TübingenGermany
| | - Saskia Elgün
- Department of Child Neurology and Developmental MedicineUniversity Children's Hospital TübingenHoppe‐Seyler‐Straße 172076TübingenGermany
| | - Benjamin Bender
- Diagnostic and Interventional Neuroradiology, Department of RadiologyUniversity Hospital TübingenHoppe‐Seyler‐Straße 372076TübingenGermany
| | - Marjo S. van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy CenterAmsterdam UMC location Vrije Universiteit Amsterdam, Emma's Children's HospitalBoelelaan 1117AmsterdamThe Netherlands,Amsterdam Neuroscience, Cellular & Molecular MechanismsAmsterdamThe Netherlands,Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Nicole I. Wolf
- Department of Child Neurology, Amsterdam Leukodystrophy CenterAmsterdam UMC location Vrije Universiteit Amsterdam, Emma's Children's HospitalBoelelaan 1117AmsterdamThe Netherlands,Amsterdam Neuroscience, Cellular & Molecular MechanismsAmsterdamThe Netherlands
| | - Samuel Groeschel
- Department of Child Neurology and Developmental MedicineUniversity Children's Hospital TübingenHoppe‐Seyler‐Straße 172076TübingenGermany
| |
Collapse
|
74
|
Morton G, Thomas S, Roberts P, Clark V, Imrie J, Morrison A. The importance of early diagnosis and views on newborn screening in metachromatic leukodystrophy: results of a Caregiver Survey in the UK and Republic of Ireland. Orphanet J Rare Dis 2022; 17:403. [PMID: 36329444 PMCID: PMC9635117 DOI: 10.1186/s13023-022-02550-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/20/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Metachromatic Leukodystrophy (MLD) is a rare, autosomal recessive lysosomal storage disorder caused by a deficiency of the enzyme arylsulfatase A (ARSA). MLD causes progressive loss of motor function and severe decline in cognitive function, leading to premature death. Early diagnosis of MLD provides the opportunity to begin treatment before the disease progresses and causes severe disability. MLD is not currently included in newborn screening (NBS) in the UK. This study consisted of an online survey, and follow-up semi-structured interviews open to MLD patients or caregivers, aged 18 years and over. The aims of the study were to understand the importance of early diagnosis and to establish the views of families and caregivers of patients with MLD on NBS. A total of 24 patients took part in the survey, representing 20 families (two families had two children with MLD, one family had three children with MLD). Following on from the survey, six parents participated in the interviews. Our data showed diagnostic delay from first symptoms was between 0 and 3 years, with a median of 1 year (n = 18); during this time deterioration was rapid, especially in earlier onset MLD. In patients with late infantile MLD (n = 10), 50% were wheelchair dependent, 30% were unable to speak, and 50% were tube fed when a diagnosis of MLD was confirmed. In patients with early juvenile MLD (n = 5), over half used a wheelchair some of the time, had uncontrollable crying, and difficulty speaking (all 60%) before or at the time of diagnosis. A high degree of support was expressed for NBS among caregivers, 95% described it as very or extremely important and 86% believed detection of MLD at birth would have changed their child’s future. One parent expressed their gratitude for an early diagnosis as a result of familial MLD screening offered at birth and how it had changed their child’s future: “It did and it absolutely has I will be forever grateful for his early diagnosis thanks to his older sister.” The rapid rate of deterioration in MLD makes it an essential candidate for NBS, particularly now the first gene therapy (Libmeldy™) has been approved by the European Medicines Agency. Libmeldy™ has also been recommended as a treatment option in England and Wales by the National Institute for Health and Care Excellence (NICE) and is being made available to patients in Scotland via the Scottish Medicines Consortium’s ultra-orphan pathway.
Collapse
|
75
|
Cruz LJ, Rezaei S, Grosveld F, Philipsen S, Eich C. Nanoparticles targeting hematopoietic stem and progenitor cells: Multimodal carriers for the treatment of hematological diseases. Front Genome Ed 2022; 4:1030285. [PMID: 36407494 PMCID: PMC9666682 DOI: 10.3389/fgeed.2022.1030285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/10/2022] [Indexed: 10/03/2023] Open
Abstract
Modern-day hematopoietic stem cell (HSC) therapies, such as gene therapy, modify autologous HSCs prior to re-infusion into myelo-conditioned patients and hold great promise for treatment of hematological disorders. While this approach has been successful in numerous clinical trials, it relies on transplantation of ex vivo modified patient HSCs, which presents several limitations. It is a costly and time-consuming procedure, which includes only few patients so far, and ex vivo culturing negatively impacts on the viability and stem cell-properties of HSCs. If viral vectors are used, this carries the additional risk of insertional mutagenesis. A therapy delivered to HSCs in vivo, with minimal disturbance of the HSC niche, could offer great opportunities for novel treatments that aim to reverse disease symptoms for hematopoietic disorders and could bring safe, effective and affordable genetic therapies to all parts of the world. However, substantial unmet needs exist with respect to the in vivo delivery of therapeutics to HSCs. In the last decade, in particular with the development of gene editing technologies such as CRISPR/Cas9, nanoparticles (NPs) have become an emerging platform to facilitate the manipulation of cells and organs. By employing surface modification strategies, different types of NPs can be designed to target specific tissues and cell types in vivo. HSCs are particularly difficult to target due to the lack of unique cell surface markers that can be utilized for cell-specific delivery of therapeutics, and their shielded localization in the bone marrow (BM). Recent advances in NP technology and genetic engineering have resulted in the development of advanced nanocarriers that can deliver therapeutics and imaging agents to hematopoietic stem- and progenitor cells (HSPCs) in the BM niche. In this review we provide a comprehensive overview of NP-based approaches targeting HSPCs to control and monitor HSPC activity in vitro and in vivo, and we discuss the potential of NPs for the treatment of malignant and non-malignant hematological disorders, with a specific focus on the delivery of gene editing tools.
Collapse
Affiliation(s)
- Luis J. Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Somayeh Rezaei
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Frank Grosveld
- Erasmus University Medical Center, Department of Cell Biology, Rotterdam, Netherlands
| | - Sjaak Philipsen
- Erasmus University Medical Center, Department of Cell Biology, Rotterdam, Netherlands
| | - Christina Eich
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
76
|
Wolff JH, Mikkelsen JG. Delivering genes with human immunodeficiency virus-derived vehicles: still state-of-the-art after 25 years. J Biomed Sci 2022; 29:79. [PMID: 36209077 PMCID: PMC9548131 DOI: 10.1186/s12929-022-00865-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 09/29/2022] [Indexed: 11/10/2022] Open
Abstract
Viruses are naturally endowed with the capacity to transfer genetic material between cells. Following early skepticism, engineered viruses have been used to transfer genetic information into thousands of patients, and genetic therapies are currently attracting large investments. Despite challenges and severe adverse effects along the way, optimized technologies and improved manufacturing processes are driving gene therapy toward clinical translation. Fueled by the outbreak of AIDS in the 1980s and the accompanying focus on human immunodeficiency virus (HIV), lentiviral vectors derived from HIV have grown to become one of the most successful and widely used vector technologies. In 2022, this vector technology has been around for more than 25 years. Here, we celebrate the anniversary by portraying the vector system and its intriguing properties. We dive into the technology itself and recapitulate the use of lentiviral vectors for ex vivo gene transfer to hematopoietic stem cells and for production of CAR T-cells. Furthermore, we describe the adaptation of lentiviral vectors for in vivo gene delivery and cover the important contribution of lentiviral vectors to basic molecular research including their role as carriers of CRISPR genome editing technologies. Last, we dwell on the emerging capacity of lentiviral particles to package and transfer foreign proteins.
Collapse
Affiliation(s)
- Jonas Holst Wolff
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Jacob Giehm Mikkelsen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark.
| |
Collapse
|
77
|
Plasschaert RN, DeAndrade MP, Hull F, Nguyen Q, Peterson T, Yan A, Loperfido M, Baricordi C, Barbarossa L, Yoon JK, Dogan Y, Unnisa Z, Schindler JW, van Til NP, Biasco L, Mason C. High-throughput analysis of hematopoietic stem cell engraftment after intravenous and intracerebroventricular dosing. Mol Ther 2022; 30:3209-3225. [PMID: 35614857 PMCID: PMC9552809 DOI: 10.1016/j.ymthe.2022.05.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 04/15/2022] [Accepted: 05/21/2022] [Indexed: 11/27/2022] Open
Abstract
Hematopoietic stem/progenitor cell gene therapy (HSPC-GT) has shown clear neurological benefit in rare diseases, which is achieved through the engraftment of genetically modified microglia-like cells (MLCs) in the brain. Still, the engraftment dynamics and the nature of engineered MLCs, as well as their potential use in common neurogenerative diseases, have remained largely unexplored. Here, we comprehensively characterized how different routes of administration affect the biodistribution of genetically engineered MLCs and other HSPC derivatives in mice. We generated a high-resolution single-cell transcriptional map of MLCs and discovered that they could clearly be distinguished from macrophages as well as from resident microglia by the expression of a specific gene signature that is reflective of their HSPC ontogeny and irrespective of their long-term engraftment history. Lastly, using murine models of Parkinson's disease and frontotemporal dementia, we demonstrated that MLCs can deliver therapeutically relevant levels of transgenic protein to the brain, thereby opening avenues for the clinical translation of HSPC-GT to the treatment of major neurological diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Aimin Yan
- AVROBIO, Inc, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | - Niek P van Til
- AVROBIO, Inc, Cambridge, MA 02139, USA; Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, VU University, and Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Luca Biasco
- AVROBIO, Inc, Cambridge, MA 02139, USA; Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Chris Mason
- AVROBIO, Inc, Cambridge, MA 02139, USA; Advanced Centre for Biochemical Engineering, University College London, London, UK.
| |
Collapse
|
78
|
Eichler F, Sevin C, Barth M, Pang F, Howie K, Walz M, Wilds A, Calcagni C, Chanson C, Campbell L. Understanding caregiver descriptions of initial signs and symptoms to improve diagnosis of metachromatic leukodystrophy. Orphanet J Rare Dis 2022; 17:370. [PMID: 36195888 PMCID: PMC9531467 DOI: 10.1186/s13023-022-02518-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Metachromatic leukodystrophy (MLD), a relentlessly progressive and ultimately fatal condition, is a rare autosomal recessive lysosomal storage disorder caused by a deficiency of the enzyme arylsulfatase A (ARSA). Historically management has been palliative or supportive care. Hematopoietic stem cell transplantation is poorly effective in early-onset MLD and benefit in late-onset MLD remains controversial. Hematopoietic stem cell gene therapy, Libmeldy (atidarsagene autotemcel), was recently approved by the European Medicines Agency for early-onset MLD. Treatment benefit is mainly observed at an early disease stage, indicating the need for early diagnosis and intervention. This study contributes insights into the caregiver language used to describe initial MLD symptomatology, and thereby aims to improve communication between clinicians and families impacted by this condition and promote a faster path to diagnosis. RESULTS Data was collected through a moderator-assisted online 60-min survey and 30-min semi-structured follow-up telephone interview with 31 MLD caregivers in the United States (n = 10), France (n = 10), the United Kingdom (n = 5), and Germany (n = 6). All respondents were primary caregivers of a person with late infantile (n = 20), juvenile (n = 11) or borderline late infantile/juvenile (n = 1) MLD (one caregiver reported for 2 children leading to a sample of 32 individuals with MLD). Caregivers were asked questions related to their child's initial signs and symptoms, time to diagnosis and interactions with healthcare providers. These results highlight the caregiver language used to describe the most common initial symptoms of MLD and provide added context to help elevate the index of suspicion of disease. Distinctions between caregiver descriptions of late infantile and juvenile MLD in symptom onset and disease course were also identified. CONCLUSIONS This study captures the caregiver description of the physical, behavioral, and cognitive signs of MLD prior to diagnosis. The understanding of the caregiver language at symptom onset sheds light on a critical window of often missed opportunity for earlier diagnosis and therapeutic intervention in MLD.
Collapse
Affiliation(s)
- F Eichler
- Center for Rare Neurological Diseases, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Caroline Sevin
- Service de Neuropédiatrie, centre de reference des leucodystrophies et leucoencephalopathies genetiques de cause rare, CHU Paris-Sud-Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
| | - M Barth
- Service de Génétique, Hôpital Universitaire d'Angers, Angers, France
| | - F Pang
- Orchard Therapeutics, 245 Hammersmith Road, London, W6 8PW, UK.
| | - K Howie
- Magnolia Innovation, Hoboken, NJ, USA
| | - M Walz
- Magnolia Innovation, Hoboken, NJ, USA
| | - A Wilds
- Magnolia Innovation, Hoboken, NJ, USA
| | | | - C Chanson
- Orchard Therapeutics, 245 Hammersmith Road, London, W6 8PW, UK
| | - L Campbell
- Orchard Therapeutics, 245 Hammersmith Road, London, W6 8PW, UK
| |
Collapse
|
79
|
Gupta AO, Raymond G, Pierpont RI, Kemp S, McIvor RS, Rayannavar A, Miller B, Lund TC, Orchard PJ. Treatment of cerebral adrenoleukodystrophy: allogeneic transplantation and lentiviral gene therapy. Expert Opin Biol Ther 2022; 22:1151-1162. [DOI: 10.1080/14712598.2022.2124857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Ashish O Gupta
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapies, University of Minnesota
| | - Gerald Raymond
- Division of Neurogenetics and The Moser Center for Leukodystrophies, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | - Rene I Pierpont
- Division of Clinical Behavioral Neuroscience, Department of Pediatrics, University of Minnesota
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC - University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Neuroscience, 1105 AZ Amsterdam, The Netherlands
| | - R Scott McIvor
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota
| | | | - Bradley Miller
- Division of Pediatric Endocrinology, University of Minnesota
| | - Troy C Lund
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapies, University of Minnesota
| | - Paul J Orchard
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapies, University of Minnesota
| |
Collapse
|
80
|
Tsakiri M, Zivko C, Demetzos C, Mahairaki V. Lipid-based nanoparticles and RNA as innovative neuro-therapeutics. Front Pharmacol 2022; 13:900610. [PMID: 36016560 PMCID: PMC9395673 DOI: 10.3389/fphar.2022.900610] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
RNA-delivery is a promising tool to develop therapies for difficult to treat diseases such as neurological disorders, by silencing pathological genes or expressing therapeutic proteins. However, in many cases RNA delivery requires a vesicle that could effectively protect the molecule from bio-degradation, bypass barriers i.e., the blood brain barrier, transfer it to a targeted tissue and efficiently release the RNA inside the cells. Many vesicles such as viral vectors, and polymeric nanoparticles have been mentioned in literature. In this review, we focus in the discussion of lipid-based advanced RNA-delivery platforms. Liposomes and lipoplexes, solid lipid nanoparticles and lipid nanoparticles are the main categories of lipidic platforms for RNA-delivery to the central nervous systems (CNS). A variety of surface particles' modifications and routes of administration have been studied to target CNS providing encouraging results in vivo. It is concluded that lipid-based nanoplatforms will play a key role in the development of RNA neuro-therapies.
Collapse
Affiliation(s)
- Maria Tsakiri
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Cristina Zivko
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasiliki Mahairaki
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
81
|
Adang L. Leukodystrophies. Continuum (Minneap Minn) 2022; 28:1194-1216. [PMID: 35938662 PMCID: PMC11320896 DOI: 10.1212/con.0000000000001130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE OF REVIEW This article reviews the most common leukodystrophies and is focused on diagnosis, clinical features, and emerging therapeutic options. RECENT FINDINGS In the past decade, the recognition of leukodystrophies has exponentially increased, and now this class includes more than 30 distinct disorders. Classically recognized as progressive and fatal disorders affecting young children, it is now understood that leukodystrophies are associated with an increasing spectrum of neurologic trajectories and can affect all ages. Next-generation sequencing and newborn screening allow the opportunity for the recognition of presymptomatic and atypical cases. These new testing opportunities, in combination with growing numbers of natural history studies and clinical consensus guidelines, have helped improve diagnosis and clinical care. Additionally, a more granular understanding of disease outcomes informs clinical trial design and has led to several recent therapeutic advances. This review summarizes the current understanding of the clinical manifestations of disease and treatment options for the most common leukodystrophies. SUMMARY As early testing becomes more readily available through next-generation sequencing and newborn screening, neurologists will better understand the true incidence of the leukodystrophies and be able to diagnose children within the therapeutic window. As targeted therapies are developed, it becomes increasingly imperative that this broad spectrum of disorders is recognized and diagnosed. This work summarizes key advances in the leukodystrophy field.
Collapse
|
82
|
Mashima R, Nakanishi M. Mammalian Sulfatases: Biochemistry, Disease Manifestation, and Therapy. Int J Mol Sci 2022; 23:ijms23158153. [PMID: 35897729 PMCID: PMC9330403 DOI: 10.3390/ijms23158153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/11/2022] [Accepted: 07/21/2022] [Indexed: 02/01/2023] Open
Abstract
Sulfatases are enzymes that catalyze the removal of sulfate from biological substances, an essential process for the homeostasis of the body. They are commonly activated by the unusual amino acid formylglycine, which is formed from cysteine at the catalytic center, mediated by a formylglycine-generating enzyme as a post-translational modification. Sulfatases are expressed in various cellular compartments such as the lysosome, the endoplasmic reticulum, and the Golgi apparatus. The substrates of mammalian sulfatases are sulfolipids, glycosaminoglycans, and steroid hormones. These enzymes maintain neuronal function in both the central and the peripheral nervous system, chondrogenesis and cartilage in the connective tissue, detoxification from xenobiotics and pharmacological compounds in the liver, steroid hormone inactivation in the placenta, and the proper regulation of skin humidification. Human sulfatases comprise 17 genes, 10 of which are involved in congenital disorders, including lysosomal storage disorders, while the function of the remaining seven is still unclear. As for the genes responsible for pathogenesis, therapeutic strategies have been developed. Enzyme replacement therapy with recombinant enzyme agents and gene therapy with therapeutic transgenes delivered by viral vectors are administered to patients. In this review, the biochemical substrates, disease manifestation, and therapy for sulfatases are summarized.
Collapse
Affiliation(s)
- Ryuichi Mashima
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
- Correspondence: ; Fax: +81-3-3417-2238
| | | |
Collapse
|
83
|
Aigrot MS, Barthelemy C, Moyon S, Dufayet-Chaffaud G, Izagirre-Urizar L, Gillet-Legrand B, Tada S, Bayón-Cordero L, Chara JC, Matute C, Cartier N, Lubetzki C, Tepavčević V. Genetically modified macrophages accelerate myelin repair. EMBO Mol Med 2022; 14:e14759. [PMID: 35822550 PMCID: PMC9358396 DOI: 10.15252/emmm.202114759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 11/09/2022] Open
Abstract
Preventing neurodegeneration‐associated disability progression in patients with multiple sclerosis (MS) remains an unmet therapeutic need. As remyelination prevents axonal degeneration, promoting this process in patients might enhance neuroprotection. In demyelinating mouse lesions, local overexpression of semaphorin 3F (Sema3F), an oligodendrocyte progenitor cell (OPC) attractant, increases remyelination. However, molecular targeting to MS lesions is a challenge. A clinically relevant paradigm for delivering Sema3F to demyelinating lesions could be to use blood‐derived macrophages as vehicles. Thus, we chose transplantation of genetically modified hematopoietic stem cells (HSCs) as means of obtaining chimeric mice with circulating Sema3F‐overexpressing monocytes. We demonstrated that Sema3F‐transduced HSCs stimulate OPC migration in a neuropilin 2 (Nrp2, Sema3F receptor)‐dependent fashion, which was conserved in middle‐aged OPCs. While demyelinating lesions induced in mice with Sema3F‐expressing blood cells showed no changes in inflammation and OPC survival, OPC recruitment was enhanced which accelerated the onset of remyelination. Our results provide a proof of concept that blood cells, particularly monocytes/macrophages, can be used to deliver pro‐remyelinating agents “at the right time and place,” suggesting novel means for remyelination‐promoting strategies in MS.
Collapse
Affiliation(s)
| | - Clara Barthelemy
- INSERM UMR1127 Sorbonne Université, Paris Brain Institute (ICM), Paris, France
| | - Sarah Moyon
- NYU Langone Health, Neuroscience Institute, New York City, NY, USA
| | | | - Leire Izagirre-Urizar
- Achucarro Basque Center for Neuroscience/Department of Neuroscience, School of Medicine University of the Basque Country, Leioa, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Satoru Tada
- INSERM UMR1127 Sorbonne Université, Paris Brain Institute (ICM), Paris, France
| | - Laura Bayón-Cordero
- Achucarro Basque Center for Neuroscience/Department of Neuroscience, School of Medicine University of the Basque Country, Leioa, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan-Carlos Chara
- Achucarro Basque Center for Neuroscience/Department of Neuroscience, School of Medicine University of the Basque Country, Leioa, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience/Department of Neuroscience, School of Medicine University of the Basque Country, Leioa, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Nathalie Cartier
- Asklepios Biopharmaceutical, Inc., Institut du Cerveau (ICM), Paris, France
| | - Catherine Lubetzki
- INSERM UMR1127 Sorbonne Université, Paris Brain Institute (ICM), Paris, France.,AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - Vanja Tepavčević
- Achucarro Basque Center for Neuroscience/Department of Neuroscience, School of Medicine University of the Basque Country, Leioa, Spain
| |
Collapse
|
84
|
Clonal reconstruction from co-occurrence of vector integration sites accurately quantifies expanding clones in vivo. Nat Commun 2022; 13:3712. [PMID: 35764632 PMCID: PMC9240075 DOI: 10.1038/s41467-022-31292-6] [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: 02/07/2022] [Accepted: 06/13/2022] [Indexed: 11/08/2022] Open
Abstract
High transduction rates of viral vectors in gene therapies (GT) and experimental hematopoiesis ensure a high frequency of gene delivery, although multiple integration events can occur in the same cell. Therefore, tracing of integration sites (IS) leads to mis-quantification of the true clonal spectrum and limits safety considerations in GT. Hence, we use correlations between repeated measurements of IS abundances to estimate their mutual similarity and identify clusters of co-occurring IS, for which we assume a clonal origin. We evaluate the performance, robustness and specificity of our methodology using clonal simulations. The reconstruction methods, implemented and provided as an R-package, are further applied to experimental clonal mixes and preclinical models of hematopoietic GT. Our results demonstrate that clonal reconstruction from IS data allows to overcome systematic biases in the clonal quantification as an essential prerequisite for the assessment of safety and long-term efficacy of GT involving integrative vectors. High transduction rates of viral vectors ensure good gene delivery; however multiple integration events can occur in the same cell. Here the authors use correlations between repeated measurements of integration site abundances to estimate their mutual similarity and identify clusters of co-occurring sites.
Collapse
|
85
|
Jimenez-Kurlander L, Duncan CN. Gene Therapy for Pediatric Neurologic Disease. Hematol Oncol Clin North Am 2022; 36:853-864. [PMID: 35760664 DOI: 10.1016/j.hoc.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Pediatric lysosomal and peroxisomal storage disorders, leukodystrophies, and motor neuron diseases can have devastating neurologic manifestations. Despite efforts to exploit cross-correction to treat these monogenic disorders for several decades, definitive treatment has yet to be identified. This review explores recent attempts to transduce autologous hematopoietic stem cells with functional gene or provide therapeutic gene in vivo. Specifically, we discuss the rationale behind efforts to treat pediatric neurologic disorders with gene therapy, outline the specific disorders that have been targeted at this time, and review recent and current clinical investigations with attention to the future direction of therapy efforts.
Collapse
Affiliation(s)
- Lauren Jimenez-Kurlander
- Department of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Christine N Duncan
- Department of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
86
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
87
|
Hematopoietic stem cell gene therapy ameliorates CNS involvement in murine model of GM1-gangliosidosis. Mol Ther Methods Clin Dev 2022; 25:448-460. [PMID: 35615711 PMCID: PMC9118356 DOI: 10.1016/j.omtm.2022.04.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 04/25/2022] [Indexed: 11/23/2022]
Abstract
GM1-gangliosidosis is a progressive neurodegenerative glycosphingolipidosis resulting from a GLB1 gene mutation causing a deficiency of the lysosomal enzyme β-galactosidase, which leads to the abnormal accumulation of GM1 ganglioside in the central nervous system. In the most severe early infantile phenotype, excessive ganglioside accumulation results in a rapid decline in neurological and psychomotor functions, and death occurs within 2 years of age. Currently, there is no effective therapy for GM1-gangliosidosis. In this study, we evaluated the therapeutic efficacy of ex vivo gene therapy targeting hematopoietic stem cells using a lentiviral vector to increase enzyme activity, reduce substrate accumulation, and improve astrocytosis and motor function. Transplanting GLB1-transduced hematopoietic stem cells in mice increased β-galactosidase enzyme activity in the central nervous system and visceral organs. Specifically, this gene therapy significantly decreased GM1 ganglioside levels in the brain, especially in the cerebrum. More important, this gene therapy rectified astrocytosis in the cerebrum and improved motor function deficits. Furthermore, the elevation of serum β-galactosidase activity in secondary-transplanted mice suggested the ability of transduced hematopoietic stem cells to repopulate long term. These data indicate that ex vivo gene therapy with lentiviral vectors is a promising approach for the treatment of brain deficits in GM1 gangliosidosis.
Collapse
|
88
|
Meng J, Moore M, Counsell J, Muntoni F, Popplewell L, Morgan J. Optimized lentiviral vector to restore full-length dystrophin via a cell-mediated approach in a mouse model of Duchenne muscular dystrophy. Mol Ther Methods Clin Dev 2022; 25:491-507. [PMID: 35615709 PMCID: PMC9121076 DOI: 10.1016/j.omtm.2022.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 04/28/2022] [Indexed: 11/16/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a muscle wasting disorder caused by mutations in the DMD gene. Restoration of full-length dystrophin protein in skeletal muscle would have therapeutic benefit, but lentivirally mediated delivery of such a large gene in vivo has been hindered by lack of tissue specificity, limited transduction, and insufficient transgene expression. To address these problems, we developed a lentiviral vector, which contains a muscle-specific promoter and sequence-optimized full-length dystrophin, to constrain dystrophin expression to differentiated myotubes/myofibers and enhance the transgene expression. We further explored the efficiency of restoration of full-length dystrophin in vivo, by grafting DMD myoblasts that had been corrected by this optimized lentiviral vector intramuscularly into an immunodeficient DMD mouse model. We show that these lentivirally corrected DMD myoblasts effectively reconstituted full-length dystrophin expression in 93.58% ± 2.17% of the myotubes in vitro. Moreover, dystrophin was restored in 64.4% ± 2.87% of the donor-derived regenerated muscle fibers in vivo, which were able to recruit members of the dystrophin-glycoprotein complex at the sarcolemma. This study represents a significant advance over existing cell-mediated gene therapy strategies for DMD that aim to restore full-length dystrophin expression in skeletal muscle.
Collapse
Affiliation(s)
- Jinhong Meng
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Marc Moore
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham TW20 0EX, UK
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - John Counsell
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- UCL Division of Surgery and Interventional Science, Charles Bell House, 43-45 Foley Street, London W1W 7TY, UK
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Linda Popplewell
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham TW20 0EX, UK
| | - Jennifer Morgan
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| |
Collapse
|
89
|
Liang Q, Vlaar EC, Catalano F, Pijnenburg JM, Stok M, van Helsdingen Y, Vulto AG, Unger WW, van der Ploeg AT, Pijnappel WP, van Til NP. Lentiviral gene therapy prevents anti-human acid α-glucosidase antibody formation in murine Pompe disease. Mol Ther Methods Clin Dev 2022; 25:520-532. [PMID: 35662813 PMCID: PMC9127119 DOI: 10.1016/j.omtm.2022.04.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 04/29/2022] [Indexed: 01/20/2023]
Abstract
Enzyme replacement therapy (ERT) is the current standard treatment for Pompe disease, a lysosomal storage disorder caused by deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). ERT has shown to be lifesaving in patients with classic infantile Pompe disease. However, a major drawback is the development of neutralizing antibodies against ERT. Hematopoietic stem and progenitor cell-mediated lentiviral gene therapy (HSPC-LVGT) provides a novel, potential lifelong therapy with a single intervention and may induce immune tolerance. Here, we investigated whether ERT can be safely applied as additional or alternative therapy following HSPC-LVGT in a murine model of Pompe disease. We found that lentiviral expression at subtherapeutic dose was sufficient to induce tolerance to the transgene product, as well as to subsequently administered ERT. Immune tolerance was established within 4–6 weeks after gene therapy. The mice tolerated ERT doses up to 100 mg/kg, allowing ERT to eliminate glycogen accumulation in cardiac and skeletal muscle and normalizing locomotor function. The presence of HSPC-derived cells expressing GAA in the thymus suggested the establishment of central immune tolerance. These findings demonstrate that lentiviral gene therapy in murine Pompe disease induced robust and long-term immune tolerance to GAA either expressed by a transgene or supplied as ERT.
Collapse
Affiliation(s)
- Qiushi Liang
- Department of Hematology and Research Laboratory of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Eva C. Vlaar
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Fabio Catalano
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Joon M. Pijnenburg
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Merel Stok
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Hematology, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Yvette van Helsdingen
- Department of Hematology, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Arnold G. Vulto
- Hospital Pharmacy, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - Wendy W.J. Unger
- Laboratory of Pediatrics, Erasmus MC University Medical Center-Sophia Children’s Hospital, 3015GE Rotterdam, the Netherlands
| | - Ans T. van der Ploeg
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| | - W.W.M. Pim Pijnappel
- Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
- Corresponding author W.W.M. Pim Pijnappel, PhD, Erasmus University Medical Center, 3015GE Rotterdam, the Netherlands.
| | - Niek P. van Til
- Department of Hematology, Erasmus MC University Medical Center, 3015GE Rotterdam, the Netherlands
| |
Collapse
|
90
|
Sala D, Ornaghi F, Morena F, Argentati C, Valsecchi M, Alberizzi V, Di Guardo R, Bolino A, Aureli M, Martino S, Gritti A. Therapeutic advantages of combined gene/cell therapy strategies in a murine model of GM2 gangliosidosis. Mol Ther Methods Clin Dev 2022; 25:170-189. [PMID: 35434178 PMCID: PMC8983315 DOI: 10.1016/j.omtm.2022.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/13/2022] [Indexed: 11/28/2022]
Abstract
Genetic deficiency of β-N-acetylhexosaminidase (Hex) functionality leads to accumulation of GM2 ganglioside in Tay-Sachs disease and Sandhoff disease (SD), which presently lack approved therapies. Current experimental gene therapy (GT) approaches with adeno-associated viral vectors (AAVs) still pose safety and efficacy issues, supporting the search for alternative therapeutic strategies. Here we leveraged the lentiviral vector (LV)-mediated intracerebral (IC) GT platform to deliver Hex genes to the CNS and combined this strategy with bone marrow transplantation (BMT) to provide a timely, pervasive, and long-lasting source of the Hex enzyme in the CNS and periphery of SD mice. Combined therapy outperformed individual treatments in terms of lifespan extension and normalization of the neuroinflammatory/neurodegenerative phenotypes of SD mice. These benefits correlated with a time-dependent increase in Hex activity and a remarkable reduction in GM2 storage in brain tissues that single treatments failed to achieve. Our results highlight the synergic mode of action of LV-mediated IC GT and BMT, clarify the contribution of treatments to the therapeutic outcome, and inform on the realistic threshold of corrective enzymatic activity. These results have important implications for interpretation of ongoing experimental therapies and for design of more effective treatment strategies for GM2 gangliosidosis.
Collapse
Affiliation(s)
- Davide Sala
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Francesca Ornaghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Francesco Morena
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Chiara Argentati
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Manuela Valsecchi
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Via Fratelli Cervi 93, 20090 Segrate, MI, Italy
| | - Valeria Alberizzi
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, 20132 Milan, Italy
| | - Roberta Di Guardo
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, 20132 Milan, Italy
| | - Alessandra Bolino
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, 20132 Milan, Italy
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Via Fratelli Cervi 93, 20090 Segrate, MI, Italy
| | - Sabata Martino
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| |
Collapse
|
91
|
Russo F, Ruggiero E, Curto R, Passeri L, Sanvito F, Bortolomai I, Villa A, Gregori S, Annoni A. Editing T cell repertoire by thymic epithelial cell-directed gene transfer abrogates risk of type 1 diabetes development. Mol Ther Methods Clin Dev 2022; 25:508-519. [PMID: 35615710 PMCID: PMC9121074 DOI: 10.1016/j.omtm.2022.04.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/29/2022] [Indexed: 11/29/2022]
Abstract
Insulin is the primary autoantigen (Ag) targeted by T cells in type 1 diabetes (T1D). Although biomarkers precisely identifying subjects at high risk of T1D are available, successful prophylaxis is still an unmet need. Leaky central tolerance to insulin may be partially ascribed to the instability of the MHC-InsB9-23 complex, which lowers TCR avidity, thus resulting in defective negative selection of autoreactive clones and inadequate insulin-specific T regulatory cell (Treg) induction. We developed a lentiviral vector (LV)-based strategy to engineer thymic epithelial cells (TECs) to correct diabetogenic T cell repertoire. Intrathymic (it) LV injection established stable transgene expression in EpCAM+ TECs, by virtue of transduction of TEC precursors. it-LV-driven presentation of the immunodominant portion of ovalbumin allowed persistent and complete negative selection of responsive T cells in OT-II chimeric mice. We successfully applied this strategy to correct the diabetogenic repertoire of young non-obese diabetic mice, imposing the presentation by TECs of the stronger agonist InsulinB9-23R22E and partially depleting the existing T cell compartment. We further circumscribed LV-driven presentation of InsulinB9-23R22E by micro-RNA regulation to CD45− TECs without loss of efficacy in protection from diabetes, associated with expanded insulin-specific Tregs. Overall, our gene transfer-based prophylaxis fine-tuned the central tolerance processes of negative selection and Treg induction, correcting an autoimmune prone T cell repertoire.
Collapse
Affiliation(s)
- Fabio Russo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Rosalia Curto
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Laura Passeri
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Francesca Sanvito
- Pathology Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ileana Bortolomai
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy.,Milan Unit, Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), 20090 Milan, Italy
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Andrea Annoni
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| |
Collapse
|
92
|
Aguilar-González A, González-Correa JE, Barriocanal-Casado E, Ramos-Hernández I, Lerma-Juárez MA, Greco S, Rodríguez-Sevilla JJ, Molina-Estévez FJ, Montalvo-Romeral V, Ronzitti G, Sánchez-Martín RM, Martín F, Muñoz P. Isogenic GAA-KO Murine Muscle Cell Lines Mimicking Severe Pompe Mutations as Preclinical Models for the Screening of Potential Gene Therapy Strategies. Int J Mol Sci 2022; 23:6298. [PMID: 35682977 PMCID: PMC9181599 DOI: 10.3390/ijms23116298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Pompe disease (PD) is a rare disorder caused by mutations in the acid alpha-glucosidase (GAA) gene. Most gene therapies (GT) partially rely on the cross-correction of unmodified cells through the uptake of the GAA enzyme secreted by corrected cells. In the present study, we generated isogenic murine GAA-KO cell lines resembling severe mutations from Pompe patients. All of the generated GAA-KO cells lacked GAA activity and presented an increased autophagy and increased glycogen content by means of myotube differentiation as well as the downregulation of mannose 6-phosphate receptors (CI-MPRs), validating them as models for PD. Additionally, different chimeric murine GAA proteins (IFG, IFLG and 2G) were designed with the aim to improve their therapeutic activity. Phenotypic rescue analyses using lentiviral vectors point to IFG chimera as the best candidate in restoring GAA activity, normalising the autophagic marker p62 and surface levels of CI-MPRs. Interestingly, in vivo administration of liver-directed AAVs expressing the chimeras further confirmed the good behaviour of IFG, achieving cross-correction in heart tissue. In summary, we generated different isogenic murine muscle cell lines mimicking the severe PD phenotype, as well as validating their applicability as preclinical models in order to reduce animal experimentation.
Collapse
Affiliation(s)
- Araceli Aguilar-González
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Juan Elías González-Correa
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Eliana Barriocanal-Casado
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Iris Ramos-Hernández
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Miguel A. Lerma-Juárez
- Instituto de Investigación del Hospital Universitario La Paz, IdiPAZ, 28029 Madrid, Spain;
| | - Sara Greco
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Juan José Rodríguez-Sevilla
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Francisco Javier Molina-Estévez
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Fundación para la Investigación Biosanitaria de Andalucía Oriental-Alejandro Otero (FIBAO), 18012 Granada, Spain
| | - Valle Montalvo-Romeral
- Généthon, Integrare Research Unit UMR_S951, INSERM, Université Paris-Saclay, Univ Evry, 91002 Evry, France; (V.M.-R.); (G.R.)
| | - Giuseppe Ronzitti
- Généthon, Integrare Research Unit UMR_S951, INSERM, Université Paris-Saclay, Univ Evry, 91002 Evry, France; (V.M.-R.); (G.R.)
| | - Rosario María Sánchez-Martín
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Francisco Martín
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Departamento de Bioquímica y Biología Molecular 3 e Inmunología, Facultad de Medicina, Universidad de Granada, Avda. de la Investigación 11, 18071 Granada, Spain
| | - Pilar Muñoz
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Departmento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Campus Fuentenueva, 18071 Granada, Spain
| |
Collapse
|
93
|
Consiglieri G, Bernardo ME, Brunetti-Pierri N, Aiuti A. Ex Vivo and In Vivo Gene Therapy for Mucopolysaccharidoses: State of the Art. Hematol Oncol Clin North Am 2022; 36:865-878. [DOI: 10.1016/j.hoc.2022.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
94
|
Genetics and gene therapy in Dravet syndrome. Epilepsy Behav 2022; 131:108043. [PMID: 34053869 DOI: 10.1016/j.yebeh.2021.108043] [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: 11/17/2020] [Revised: 05/02/2021] [Accepted: 05/02/2021] [Indexed: 12/17/2022]
Abstract
Dravet syndrome is a well-established electro-clinical condition first described in 1978. A main genetic cause was identified with the discovery of a loss-of-function SCN1A variant in 2001. Mechanisms underlying the phenotypic variations have subsequently been a main topic of research. Various genetic modifiers of clinical severities have been elucidated through many rigorous studies on genotype-phenotype correlations and the recent advances in next generation sequencing technology. Furthermore, a deeper understanding of the regulation of gene expression and remarkable progress on genome-editing technology using the CRISPR-Cas9 system provide significant opportunities to overcome hurdles of gene therapy, such as enhancing NaV1.1 expression. This article reviews the current understanding of genetic pathology and the status of research toward the development of gene therapy for Dravet syndrome. This article is part of the Special Issue "Severe Infantile Epilepsies".
Collapse
|
95
|
Abstract
The earliest conceptual history of gene therapy began with the recognition of DNA as the transforming substance capable of changing the phenotypic character of a bacterium and then as the carrier of the genomic code. Early studies of oncogenic viruses that could insert into the mammalian genome led to the concept that these same viruses might be engineered to carry new genetic material into mammalian cells, including human hematopoietic stem cells (HSC). In addition to properly engineered vectors capable of efficient safe transduction of HSC, successful gene therapy required the development of efficient materials, methods, and equipment to procure, purify, and culture HSC. Increased understanding of the preparative conditioning of patients was needed to optimize the engraftment of genetically modified HSC. Testing concepts in pivotal clinical trials to assess the efficacy and determine the cause of adverse events has advanced the efficiency and safety of gene therapy. This article is a historical overview of the separate threads of discovery that joined together to comprise our current state of gene therapy targeting HSC.
Collapse
|
96
|
Rossini L, Durante C, Marzollo A, Biffi A. New Indications for Hematopoietic Stem Cell Gene Therapy in Lysosomal Storage Disorders. Front Oncol 2022; 12:885639. [PMID: 35646708 PMCID: PMC9136164 DOI: 10.3389/fonc.2022.885639] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 12/04/2022] Open
Abstract
Lysosomal storage disorders (LSDs) are a heterogenous group of disorders due to genetically determined deficits of lysosomal enzymes. The specific molecular mechanism and disease phenotype depends on the type of storage material. Several disorders affect the brain resulting in severe clinical manifestations that substantially impact the expectancy and quality of life. Current treatment modalities for LSDs include enzyme replacement therapy (ERT) and hematopoietic cell transplantation (HCT) from allogeneic healthy donors, but are available for a limited number of disorders and lack efficacy on several clinical manifestations. Hematopoietic stem cell gene therapy (HSC GT) based on integrating lentiviral vectors resulted in robust clinical benefit when administered to patients affected by Metachromatic Leukodystrophy, for whom it is now available as a registered medicinal product. More recently, HSC GT has also shown promising results in Hurler syndrome patients. Here, we discuss possible novel HSC GT indications that are currently under development. If these novel drugs will prove effective, they might represent a new standard of care for these disorders, but several challenges will need to be addresses, including defining and possibly expanding the patient population for whom HSC GT could be efficacious.
Collapse
Affiliation(s)
- Linda Rossini
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
| | - Caterina Durante
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
| | - Antonio Marzollo
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
- Fondazione Citta’ della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | - Alessandra Biffi
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
- Maternal and Child Health Department, Padua University, Padua, Italy
- *Correspondence: Alessandra Biffi,
| |
Collapse
|
97
|
Chiesa R, Bernardo ME. Haematopoietic stem cell gene therapy in inborn errors of metabolism. Br J Haematol 2022; 198:227-243. [PMID: 35535965 DOI: 10.1111/bjh.18179] [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: 05/10/2021] [Revised: 03/06/2022] [Accepted: 03/21/2022] [Indexed: 11/30/2022]
Abstract
Over the last 30 years, allogeneic haematopoietic stem cell transplantation (allo-HSCT) has been adopted as a therapeutic strategy for many inborn errors of metabolism (IEM), due to the ability of donor-derived cells to provide life-long enzyme delivery to deficient tissues and organs. However, (a) the clinical benefit of allo-HSCT is limited to a small number of IEM, (b) patients are left with a substantial residual disease burden and (c) allo-HSCT is still associated with significant short- and long-term toxicities and transplant-related mortality. Haematopoietic stem/progenitor cell gene therapy (HSPC-GT) was established in the 1990s for the treatment of selected monogenic primary immunodeficiencies and over the past few years, its use has been extended to a number of IEM. HSPC-GT is particularly attractive in neurodegenerative IEM, as gene corrected haematopoietic progenitors can deliver supra-physiological enzyme levels to difficult-to-reach areas, such as the brain and the skeleton, with potential increased clinical benefit. Moreover, HSPC-GT is associated with reduced morbidity and mortality compared to allo-HSCT, although this needs to be balanced against the potential risk of insertional mutagenesis. The number of clinical trials in the IEM field is rapidly increasing and some HSPC-GT products recently received market approval. This review describes the development of ex vivo HSPC-GT in a number of IEM, with a focus on recent results from GT clinical trials and risks versus benefits considerations, when compared to established therapeutic strategies, such as allo-HSCT.
Collapse
Affiliation(s)
- Robert Chiesa
- Bone Marrow Transplantation Department, Great Ormond Street Hospital for Sick Children NHS Foundation Trust, London, UK
| | - Maria Ester Bernardo
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy.,"Vita Salute" San Raffaele University, Milan, Italy
| |
Collapse
|
98
|
Ryan MM. Gene therapy for neuromuscular disorders: prospects and ethics. Arch Dis Child 2022; 107:421-426. [PMID: 34462265 DOI: 10.1136/archdischild-2020-320908] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 08/11/2021] [Indexed: 11/04/2022]
Abstract
Most childhood neuromuscular disorders are caused by mutations causing abnormal expression or regulation of single genes or genetic pathways. The potential for gene therapy, gene editing and genetic therapies to ameliorate the course of these conditions is extraordinarily exciting, but there are significant challenges associated with their use, particularly with respect to safety, efficacy, cost and equity. Engagement with these novel technologies mandates careful assessment of the benefits and burdens of treatment for the patient, their family and their society. The examples provided by spinal muscular atrophy and Duchenne muscular dystrophy illustrate the potential value and challenges of gene and genetic therapies for paediatric neurological conditions. The cost and complexity of administration of these agents is a challenge for all countries. Jurisdictional variations in availability of newborn screening, genetic diagnostics, drug approval and reimbursement pathways, treatment and rehabilitation will affect equity of access, nationally and internationally. These challenges will best be addressed by collaboration by governments, pharma, clinicians and patient groups to establish frameworks for safe and cost-effective use of these exciting new therapies.
Collapse
Affiliation(s)
- Monique M Ryan
- Children's Neurosciences Centre, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia .,Murdoch Children's Research Institute, Parkville, Victoria, Australia
| |
Collapse
|
99
|
Horgan C, Jones SA, Bigger BW, Wynn R. Current and Future Treatment of Mucopolysaccharidosis (MPS) Type II: Is Brain-Targeted Stem Cell Gene Therapy the Solution for This Devastating Disorder? Int J Mol Sci 2022; 23:4854. [PMID: 35563245 PMCID: PMC9105950 DOI: 10.3390/ijms23094854] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 02/04/2023] Open
Abstract
Mucopolysaccharidosis type II (Hunter Syndrome) is a rare, x-linked recessive, progressive, multi-system, lysosomal storage disease caused by the deficiency of iduronate-2-sulfatase (IDS), which leads to the pathological storage of glycosaminoglycans in nearly all cell types, tissues and organs. The condition is clinically heterogeneous, and most patients present with a progressive, multi-system disease in their early years. This article outlines the pathology of the disorder and current treatment strategies, including a detailed review of haematopoietic stem cell transplant outcomes for MPSII. We then discuss haematopoietic stem cell gene therapy and how this can be employed for treatment of the disorder. We consider how preclinical innovations, including novel brain-targeted techniques, can be incorporated into stem cell gene therapy approaches to mitigate the neuropathological consequences of the condition.
Collapse
Affiliation(s)
- Claire Horgan
- Blood and Marrow Transplant Unit, Department of Paediatric Haematology, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK;
| | - Simon A. Jones
- Willink Unit, Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK;
| | - Brian W. Bigger
- Stem Cell and Neuropathies, Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester M13 9PT, UK;
| | - Robert Wynn
- Blood and Marrow Transplant Unit, Department of Paediatric Haematology, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK;
| |
Collapse
|
100
|
Costei C, Barbarosie M, Bernard G, Brais B, La Piana R. Adult Hereditary White Matter Diseases With Psychiatric Presentation: Clinical Pointers and MRI Algorithm to Guide the Diagnostic Process. J Neuropsychiatry Clin Neurosci 2022; 33:180-193. [PMID: 33951919 DOI: 10.1176/appi.neuropsych.20110294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The investigators aimed to provide clinical and MRI guidelines for determining when genetic workup should be considered in order to exclude hereditary leukoencephalopathies in affected patients with a psychiatric presentation. METHODS A systematic literature review was conducted, and clinical cases are provided. Given the central role of MRI pattern recognition in the diagnosis of white matter disorders, the investigators adapted an MRI algorithm that guides the interpretation of MRI findings and thus directs further investigations, such as genetic testing. RESULTS Twelve genetic leukoencephalopathies that can present with psychiatric symptoms were identified. As examples of presentations that can occur in clinical practice, five clinical vignettes from patients assessed at a referral center for adult genetic leukoencephalopathies are provided. CONCLUSIONS Features such as drug-resistant symptoms, presence of long-standing somatic features, trigger events, consanguinity, and positive family history should orient the clinician toward diagnostic workup to exclude the presence of a genetic white matter disorder. The identification of MRI white matter abnormalities, especially when presenting a specific pattern of involvement, should prompt genetic testing for known forms of genetic leukoencephalopathies.
Collapse
Affiliation(s)
- Catalina Costei
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal (Costei, Brais, La Piana); Department of Psychiatry, McGill University (Barbarosie); Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University (Bernard); Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal (Bernard); Child Health and Human Development Program, Research Institute of the McGill University Health Center (Bernard); and Department of Diagnostic Radiology, McGill University (La Piana)
| | - Michaela Barbarosie
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal (Costei, Brais, La Piana); Department of Psychiatry, McGill University (Barbarosie); Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University (Bernard); Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal (Bernard); Child Health and Human Development Program, Research Institute of the McGill University Health Center (Bernard); and Department of Diagnostic Radiology, McGill University (La Piana)
| | - Geneviève Bernard
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal (Costei, Brais, La Piana); Department of Psychiatry, McGill University (Barbarosie); Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University (Bernard); Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal (Bernard); Child Health and Human Development Program, Research Institute of the McGill University Health Center (Bernard); and Department of Diagnostic Radiology, McGill University (La Piana)
| | - Bernard Brais
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal (Costei, Brais, La Piana); Department of Psychiatry, McGill University (Barbarosie); Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University (Bernard); Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal (Bernard); Child Health and Human Development Program, Research Institute of the McGill University Health Center (Bernard); and Department of Diagnostic Radiology, McGill University (La Piana)
| | - Roberta La Piana
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal (Costei, Brais, La Piana); Department of Psychiatry, McGill University (Barbarosie); Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University (Bernard); Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal (Bernard); Child Health and Human Development Program, Research Institute of the McGill University Health Center (Bernard); and Department of Diagnostic Radiology, McGill University (La Piana)
| |
Collapse
|